1
|
Szilagyi IA, Schiphof D, Chaker L, Boer CG, Aribas E, Kavousi M, Ikram MA, Bierma-Zeinstra SMA, van Meurs JBJ. Associations between testosterone and knee and hand osteoarthritis among males and females from the general population. Osteoarthritis Cartilage 2025:S1063-4584(25)00878-7. [PMID: 40221126 DOI: 10.1016/j.joca.2025.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/15/2025] [Accepted: 03/22/2025] [Indexed: 04/14/2025]
Abstract
PURPOSE The aim of this study was to examine the relationship between testosterone levels (total and free) and the prevalence, incidence and progression of radiographic osteoarthritis (OA) and chronic pain outcomes in the knee and hand joints. METHODS This study is embedded in the Rotterdam Study cohort, a population-based prospective study with participants aged 45 and older. The association between testosterone levels and OA was tested in more than 8600 individuals, using generalized linear models and generalized estimating equations models, adjusting for age, body mass index (BMI), lifestyle factors and co-morbidities. RESULTS Higher total testosterone (Relative Risk [RR] 0.84, 95% Confidence Interval [CI] 0.69-1.00) and higher free testosterone (RR 0.85, 95% CI 0.70-1.00) were significantly associated with a lower prevalence of radiographic knee OA in females, but not significant in males. Higher free testosterone was significantly associated with lower incidence of chronic knee pain (CKP) in females (RR 0.85, 95% CI 0.71-0.98), but not in males. In contrast, higher free testosterone (RR 1.07, 95% CI 1.02-1.11) and total testosterone (RR 1.08, 95% CI 1.03-1.13) were significantly associated with higher severity of hand OA in males, but not in females. These associations were independent of BMI, other lifestyle factors, co-morbidities and years since menopause. CONCLUSIONS Our findings suggest a protective effect of free testosterone for CKP only in females. Future replication of our findings is needed in prospective cohort studies.
Collapse
Affiliation(s)
- Ingrid A Szilagyi
- Dept. of General Practice, Erasmus MC University Medical Center Rotterdam, the Netherlands; Dept. of Internal Medicine, Erasmus MC University Medical Center Rotterdam, the Netherlands
| | - Dieuwke Schiphof
- Dept. of General Practice, Erasmus MC University Medical Center Rotterdam, the Netherlands
| | - Layal Chaker
- Dept. of Internal Medicine, Erasmus MC University Medical Center Rotterdam, the Netherlands; Dept. of Epidemiology, Erasmus MC University Medical Center Rotterdam, the Netherlands
| | - Cindy G Boer
- Dept. of Internal Medicine, Erasmus MC University Medical Center Rotterdam, the Netherlands
| | - Elif Aribas
- Dept. of Epidemiology, Erasmus MC University Medical Center Rotterdam, the Netherlands
| | - Maryam Kavousi
- Dept. of Epidemiology, Erasmus MC University Medical Center Rotterdam, the Netherlands
| | - M Arfan Ikram
- Dept. of Epidemiology, Erasmus MC University Medical Center Rotterdam, the Netherlands
| | - Sita M A Bierma-Zeinstra
- Dept. of General Practice, Erasmus MC University Medical Center Rotterdam, the Netherlands; Dept. of Orthopedics and Sportsmedicine, Erasmus MC University Medical Center Rotterdam, the Netherlands
| | - Joyce B J van Meurs
- Dept. of Internal Medicine, Erasmus MC University Medical Center Rotterdam, the Netherlands; Dept. of Orthopedics and Sportsmedicine, Erasmus MC University Medical Center Rotterdam, the Netherlands.
| |
Collapse
|
2
|
Rissardo JP, Caprara ALF. A Narrative Review on Biochemical Markers and Emerging Treatments in Prodromal Synucleinopathies. Clin Pract 2025; 15:65. [PMID: 40136601 PMCID: PMC11941140 DOI: 10.3390/clinpract15030065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/08/2025] [Accepted: 03/11/2025] [Indexed: 03/27/2025] Open
Abstract
Alpha-synuclein has been associated with neurodegeneration, especially in Parkinson's disease (PD). This study aimed to review clinical, biochemical, and neuroimaging markers and management of prodromal synucleinopathies. The prodromal state of synucleinopathies can be better understood with PD pathophysiology, and it can be separated into premotor and pre-diagnostic phases. The incidence of PD in patients with prodromal phase symptoms ranges from 0.07 to 14.30, and the most frequently studied pathology is the REM behavioral disorder (RBD). Neuroimaging markers are related to dopamine denervation, brain perfusion changes, gross anatomy changes, and peripheral abnormalities. α-synuclein assays (SAA) in CSF revealed high sensitivity (up to 97%) and high specificity (up to 92%); in the last decade, there was the development of other matrices (blood, skin, and olfactory mucosa) for obtaining quantitative and qualitative α-synuclein. Other biomarkers are neurofilament light chain, DOPA decarboxylase, and multiplexed mass spectrometry assay. Regarding genetic counseling in α-synucleinopathies, it is an important topic in clinical practice to discuss with patients with high-risk individuals and should involve basic principles of autonomy, beneficence, and non-maleficence. Some of the themes that should be reviewed are the involvement of physical activity, diet (including alcohol, coffee, and vitamin supplementation), smoking, sleep, and stress in the pathophysiology of synucleinopathies. The number of trials related to prodromal symptoms is still scarce, and the number of studies evaluating intervention is even lower.
Collapse
|
3
|
Ortega N, Mueller NJ, Dehghan A, de Crom TOE, von Gunten A, Preisig M, Marques-Vidal P, Vinceti M, Voortman T, Rodondi N, Chocano-Bedoya PO. Dairy intake and cognitive function in older adults in three cohorts: a mendelian randomization study. Nutr J 2025; 24:20. [PMID: 39891239 PMCID: PMC11784005 DOI: 10.1186/s12937-025-01083-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/20/2025] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND Meta-analyses of observational studies on the effect of dairy on cognitive function have yielded inconclusive results, potentially due to unmeasured confounding. To avoid the no-unmeasured confounding assumption, we used lactase persistence genetic variant as an instrumental variable, for which the CC genotype is associated with lower lactase production and, consequently, lower dairy consumption. We used it to assess the effect of long-term consumption of total and non-fermented dairy on cognitive function. METHODS We included 43,836 individuals over 55 years old with genotyping, dietary data, and cognitive function measurements from three population-based studies: CoLaus|PsyCoLaus (Switzerland), the Rotterdam Study (the Netherlands) and the Canadian Longitudinal Study on Aging (CLSA - Canada). We performed a one-sample Mendelian randomization using two-stage least-squares regression. First, we estimated total and non-fermented dairy consumption by T-allele frequency. Second, we used the estimated dairy consumption in linear regression models on general cognition, assessed by the Mini-Mental State Examination (MMSE) and the Mental Alternation Test, executive function, verbal fluency, verbal learning, and memory. RESULTS Per T-allele, total dairy intake and non-fermented was 24.8 and 15.3 g/day higher in PsyCoLaus, 57.9 and 49.8 g/day in the Rotterdam Study, and 0.31 and 0.29 times/day in CLSA, respectively. We found no association between the genetically predicted difference and the MMSE in PsyCoLaus and the Rotterdam Study. However, lactase persistent individuals scored 3.4 (95% CI 2.1- 4.7) and 3.5 (95% CI 2.3-4.7) points more in the Mental Alternation Test for total and fermented dairy, respectively, in CLSA. Similarly, lactase persistent participants in CLSA had higher verbal fluency, verbal learning and executive function, but no differences were found in the other cohorts. Such inconsistencies might stem from different FFQs across cohorts and consumption ranges. Nonetheless, the generally small magnitude of effect sizes may suggest that there is no real effect between total or non-fermented dairy intake and cognitive function. CONCLUSION The evidence for a causal effect of dairy consumption on general cognitive function is weak, consistent with previous results from classic analysis from observational studies. Interventions targeting dairy are unlikely to have a relevant effect on cognitive function.
Collapse
Affiliation(s)
- Natalia Ortega
- Institute for Primary Health Care (BIHAM), University of Bern, Bern, Switzerland.
- Population Health Laboratory (#PopHealthLab), University of Fribourg, Fribourg, Switzerland.
- Graduate School for Health Sciences, University of Bern, Bern, Switzerland.
- , Mittelstrasse 43, Bern, 3012, Switzerland.
| | - Nick J Mueller
- Institute for Primary Health Care (BIHAM), University of Bern, Bern, Switzerland
| | - Abbas Dehghan
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Dementia Research Institute, Imperial College London, London, UK
- MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
| | - Tosca O E de Crom
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Armin von Gunten
- Center for Primary Care and Public Health, University Center of General Medicine and Public Health, Lausanne, Vaud, Switzerland
| | - Martin Preisig
- Service of Old-Age Psychiatry, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
- Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Pedro Marques-Vidal
- Department of Medicine, Internal Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Marco Vinceti
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Trudy Voortman
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
- Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, CA, USA
| | - Nicolas Rodondi
- Institute for Primary Health Care (BIHAM), University of Bern, Bern, Switzerland
- Department of General Internal Medicine, Inselspital University Hospital Bern, Bern, Switzerland
| | | |
Collapse
|
4
|
Szilagyi IA, Nguyen NL, Boer CG, Schiphof D, Ahmadizar F, Kavousi M, Bierma-Zeinstra SMA, van Meurs JBJ. Metabolic syndrome, radiographic osteoarthritis progression and chronic pain of the knee among men and women from the general population: The Rotterdam study. Semin Arthritis Rheum 2024; 69:152544. [PMID: 39288696 DOI: 10.1016/j.semarthrit.2024.152544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 08/20/2024] [Accepted: 08/23/2024] [Indexed: 09/19/2024]
Abstract
OBJECTIVE Although a relationship between osteoarthritis and components of metabolic syndrome (MetS) has been suggested, most of the results have been cross-sectional. We, therefore, aimed to investigate the sex-specific longitudinal association of (components of) MetS with progression of radiographic osteoarthritis and chronic pain in the knee joints in a large prospective cohort. METHOD In the large population-based Rotterdam study of up to 6,138 individuals, median follow-up time 5.7 (IQR 5.5) years, we examined the relation between MetS and its components (abdominal obesity, high triglycerides, low high-density lipoprotein, elevated blood pressure, and type 2 diabetes) with the progression of osteoarthritis using generalized estimating equations, generalized linear models and competing risk analysis. Analyses were stratified for sex. Covariates adjusted for: age, smoking, alcohol use, education, sub-cohort, baseline K/L grade, months between radiographs and BMI. RESULTS The presence of MetS (37.6 % in men, 39 % in women) and elevated blood pressure was associated with an increased risk of knee osteoarthritis progression in both men and women. MetS was associated with an increased risk of incident chronic knee pain (CKP) in men. In addition, abdominal obesity and high triglycerides showed higher riskfor incidence of CKP in men,but not in women. The associations were attenuated and no longer significant after BMI-adjustment, except for the association of MetS and high triglycerides with incidence of CKP in men that stayed significant (OR 1.04, 95 %CI 1.00-1.07 for MetS and OR 1.04, 95 %CI 1.01-1.07 for high triglycerides). CONCLUSION Metabolic syndrome and individual metabolic components, such as abdominal obesity and elevated blood pressure, were associated with radiographic progression of knee OA in both men and women, but not independent of BMI. Metabolic syndrome and high triglycerides were associated with incidence of CKP only in men.
Collapse
Affiliation(s)
- I A Szilagyi
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands; Department of General Practice, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - N L Nguyen
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - C G Boer
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - D Schiphof
- Department of General Practice, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - F Ahmadizar
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands; Julius Global Health, University Utrecht Medical Center, Utrecht, the Netherlands
| | - M Kavousi
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - S M A Bierma-Zeinstra
- Department of General Practice, Erasmus MC, University Medical Center Rotterdam, the Netherlands; Department of Orthopedics & Sports Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - J B J van Meurs
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands; Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands; Department of Orthopedics & Sports Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands.
| |
Collapse
|
5
|
Magalhães TA, Carneiro ACDC, Moreira VDM, Trad HS, Lopes MMU, Cerci RJ, Nacif MS, Schvartzman PR, Chagas ACP, Costa IBSDS, Schmidt A, Shiozaki AA, Montenegro ST, Piegas LS, Zapparoli M, Nicolau JC, Fernandes F, Hadlich MS, Ghorayeb N, Mesquita ET, Gonçalves LFG, Ramires FJA, Fernandes JDL, Schwartzmann PV, Rassi S, Torreão JA, Mateos JCP, Beck-da-Silva L, Silva MC, Liberato G, Oliveira GMMD, Feitosa Filho GS, Carvalho HDSMD, Markman Filho B, Rocha RPDS, Azevedo Filho CFD, Taratsoutchi F, Coelho-Filho OR, Kalil Filho R, Hajjar LA, Ishikawa WY, Melo CA, Jatene IB, Albuquerque ASD, Rimkus CDM, Silva PSDD, Vieira TDR, Jatene FB, Azevedo GSAAD, Santos RD, Monte GU, Ramires JAF, Bittencourt MS, Avezum A, Silva LSD, Abizaid A, Gottlieb I, Precoma DB, Szarf G, Sousa ACS, Pinto IMF, Medeiros FDM, Caramelli B, Parga Filho JR, Santos TSGD, Prazeres CEED, Lopes MACQ, Avila LFRD, Scanavacca MI, Gowdak LHW, Barberato SH, Nomura CH, Rochitte CE. Cardiovascular Computed Tomography and Magnetic Resonance Imaging Guideline of the Brazilian Society of Cardiology and the Brazilian College of Radiology - 2024. Arq Bras Cardiol 2024; 121:e20240608. [PMID: 39475988 DOI: 10.36660/abc.20240608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025] Open
Affiliation(s)
- Tiago Augusto Magalhães
- Complexo Hospital de Clínicas da Universidade Federal do Paraná (CHC-UFPR), Curitiba, PR - Brasil
- Hospital do Coração (HCOR), São Paulo, SP - Brasil
- Hospital Sírio Libanês, SP, São Paulo, SP - Brasil
| | | | - Valéria de Melo Moreira
- Instituto do Coração (Incor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo SP - Brasil
| | | | - Marly Maria Uellendahl Lopes
- Universidade Federal de São Paulo (UNIFESP), São Paulo, SP - Brasil
- DASA - Diagnósticos da América S/A, São Paulo, SP - Brasil
| | | | - Marcelo Souto Nacif
- Universidade Federal Fluminense, Niterói, RJ - Brasil
- Hospital Universitário Antonio Pedro, Niterói, RJ - Brasil
| | | | - Antônio Carlos Palandrini Chagas
- Instituto do Coração (Incor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo SP - Brasil
- Faculdade de Medicina do ABC, Santo André, SP - Brasil
| | | | - André Schmidt
- Universidade de São Paulo (USP), Ribeirão Preto, SP - Brasil
| | - Afonso Akio Shiozaki
- ND Núcleo Diagnóstico, Maringá, PR - Brasil
- Ômega Diagnóstico, Maringá, PR - Brasil
- Hospital Paraná, Maringá, PR - Brasil
| | | | | | - Marcelo Zapparoli
- Quanta Diagnóstico por Imagem, Curitiba, PR - Brasil
- DAPI, Curitiba, PR - Brasil
| | - José Carlos Nicolau
- Instituto do Coração (Incor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo SP - Brasil
| | - Fabio Fernandes
- Instituto do Coração (Incor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo SP - Brasil
| | - Marcelo Souza Hadlich
- Fleury Medicina e Saúde, Rio de Janeiro, RJ - Brasil
- Rede D'Or RJ, Rio de Janeiro, RJ - Brasil
- Unimed, Rio de Janeiro, RJ - Brasil
- Instituto Nacional de Cardiologia (INC), Rio de Janeiro, RJ - Brasil
| | - Nabil Ghorayeb
- Instituto Dante Pazzanese de Cardiologia, São Paulo, SP - Brasil
- Inspirali Educação, São Paulo, SP - Brasil
- Anhanguera Educacional, São Paulo, SP - Brasil
| | | | - Luiz Flávio Galvão Gonçalves
- Hospital São Lucas, Rede D'Or SE, Aracaju, SE - Brasil
- Hospital Universitário da Universidade Federal de Sergipe, Aracaju, SE - Brasil
- Clínica Climedi, Aracaju, SE - Brasil
| | - Felix José Alvarez Ramires
- Instituto do Coração (Incor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo SP - Brasil
| | | | - Pedro Vellosa Schwartzmann
- Hospital Unimed Ribeirão Preto, Ribeirão Preto, SP - Brasil
- Centro Avançado de Pesquisa, Ensino e Diagnóstico (CAPED), Ribeirão Preto, SP - Brasil
| | | | | | - José Carlos Pachón Mateos
- Hospital do Coração (HCOR), São Paulo, SP - Brasil
- Hospital Sírio Libanês, SP, São Paulo, SP - Brasil
| | - Luiz Beck-da-Silva
- Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS - Brasil
| | | | - Gabriela Liberato
- Hospital Sírio Libanês, SP, São Paulo, SP - Brasil
- Instituto do Coração (Incor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo SP - Brasil
| | | | | | - Hilka Dos Santos Moraes de Carvalho
- PROCAPE - Universidade de Pernambuco, Recife, PE - Brasil
- Hospital das Clínicas de Pernambuco da Universidade Federal de Pernambuco (UFPE), Recife, PE - Brasil
- Real Hospital Português de Pernambuco, Recife, PE - Brasil
| | - Brivaldo Markman Filho
- Hospital das Clínicas de Pernambuco da Universidade Federal de Pernambuco (UFPE), Recife, PE - Brasil
| | | | | | - Flávio Taratsoutchi
- Instituto do Coração (Incor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo SP - Brasil
| | | | - Roberto Kalil Filho
- Instituto do Coração (Incor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo SP - Brasil
| | | | - Walther Yoshiharu Ishikawa
- Instituto do Coração (Incor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo SP - Brasil
| | - Cíntia Acosta Melo
- Hospital Beneficência Portuguesa de São Paulo, São Paulo, SP - Brasil
- Hospital Infantil Sabará, São Paulo, SP - Brasil
| | | | | | - Carolina de Medeiros Rimkus
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo SP - Brasil
- Instituto D'Or de Pesquisa e Ensino (IDOR), São Paulo SP - Brasil
| | - Paulo Savoia Dias da Silva
- Fleury Medicina e Saúde, Rio de Janeiro, RJ - Brasil
- University of Iowa Hospitals and Clinics, Iowa City - EUA
| | - Thiago Dieb Ristum Vieira
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo SP - Brasil
| | - Fabio Biscegli Jatene
- Instituto do Coração (Incor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo SP - Brasil
| | - Guilherme Sant Anna Antunes de Azevedo
- ECOMAX, Blumenau, SC - Brasil
- Hospital Unimed Blumenau, Blumenau, SC - Brasil
- Hospital São José de Jaraguá do Sul, Blumenau, SC - Brasil
- Cliniimagem Criciúma, Blumenau, SC - Brasil
| | - Raul D Santos
- Hospital Sírio Libanês, SP, São Paulo, SP - Brasil
- Universidade de São Paulo (USP), Ribeirão Preto, SP - Brasil
| | | | - José Antonio Franchini Ramires
- Instituto do Coração (Incor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo SP - Brasil
| | | | - Alvaro Avezum
- Hospital Alemão Oswaldo Cruz, São Paulo, SP - Brasil
| | | | | | - Ilan Gottlieb
- Fonte Imagem Medicina Diagnostica, Rio de Janeiro, RJ - Brasil
| | | | - Gilberto Szarf
- Universidade Federal de São Paulo (UNIFESP), São Paulo, SP - Brasil
| | - Antônio Carlos Sobral Sousa
- Universidade Federal de Sergipe, Aracaju, SE - Brasil
- Hospital São Lucas, Aracaju, SE - Brasil
- Rede D'Or de Aracaju, Aracaju, SE - Brasil
| | | | | | - Bruno Caramelli
- Instituto do Coração (Incor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo SP - Brasil
| | - José Rodrigues Parga Filho
- Hospital Sírio Libanês, SP, São Paulo, SP - Brasil
- Instituto do Coração (Incor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo SP - Brasil
| | | | | | | | | | - Mauricio Ibrahim Scanavacca
- Instituto do Coração (Incor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo SP - Brasil
| | - Luis Henrique Wolff Gowdak
- Instituto do Coração (Incor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo SP - Brasil
- Universidade de São Paulo (USP), Ribeirão Preto, SP - Brasil
| | - Silvio Henrique Barberato
- Quanta Diagnóstico por Imagem, Curitiba, PR - Brasil
- Cardioeco, Centro de Diagnóstico Cardiovascular, Curitiba, PR - Brasil
| | | | - Carlos Eduardo Rochitte
- Hospital do Coração (HCOR), São Paulo, SP - Brasil
- Instituto do Coração (Incor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo SP - Brasil
- DASA - Diagnósticos da América S/A, São Paulo, SP - Brasil
| |
Collapse
|
6
|
Ortega N, Schütte L, de Crom TOE, Voortman T, Okereke OI, Vinceti M, von Gunten A, Marques-Vidal P, Rodondi N, Chiolero A, Chocano-Bedoya PO. Dietary patterns, inflammatory biomarkers and cognition in older adults: An analysis of three population-based cohorts. Clin Nutr 2024; 43:2336-2343. [PMID: 39236406 DOI: 10.1016/j.clnu.2024.08.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 07/22/2024] [Accepted: 08/21/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND Targeting effective strategies to prevent cognitive decline is key in the aging population. Some diets have been linked to a slower cognitive decline, potentially through reducing inflammation. We aimed at determining the effect of inflammatory dietary patterns (IDPs) on cognitive function in three population-based cohorts. METHODS In this longitudinal study, we analyzed data from the Canadian Longitudinal Study of Aging, CoLaus|PsyCoLaus and Rotterdam Study. Our analytical sample included participants over 55 years old with baseline data on cognition, dietary intake, and inflammatory markers. IDPs were derived for each cohort using reduced rank regression to reflect maximal variation in three inflammatory markers. We calculated scores of consumption of the IDPs, higher scores indicating more IDP consumption. We used inverse probability of treatment and censoring weights in the marginal structural models to estimate associations of higher versus lower quarters of consumption of an IDP on general cognition (Mini-Mental State Evaluation) and four cognitive domains (memory, verbal fluency, verbal learning and processing speed and executive function) during at least 3 years of follow-up. RESULTS We included 10,366 participants (mean age 68) followed-up for a mean of 5 years. Diet explained between 1 and 2% of the variation of the inflammatory markers. There were no differences in general cognition when comparing the highest to the lowest quarter of consumption of IDPs among the three cohorts. Mean differences for the four cognitive domains were of small magnitude across cohorts and not clinically relevant. CONCLUSION Diet explained low variation in inflammatory markers. Consuming IDPs was not associated with mean differences in general or domain-specific cognitive function.
Collapse
Affiliation(s)
- Natalia Ortega
- Institute for Primary Health Care (BIHAM), University of Bern, Bern, Switzerland; Population Health Laboratory (#PopHealthLab), University of Fribourg, Fribourg, Switzerland; Graduate School for Health Sciences, University of Bern, Bern, Switzerland.
| | - Leona Schütte
- Institute for Primary Health Care (BIHAM), University of Bern, Bern, Switzerland
| | - Tosca O E de Crom
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Trudy Voortman
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands; Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, CA, USA
| | - Olivia I Okereke
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Marco Vinceti
- Environmental, Genetic and Nutritional Epidemiology Research Center, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Italy; Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Armin von Gunten
- Service of Old-Age Psychiatry, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Pedro Marques-Vidal
- Department of Medicine, Internal Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Nicolas Rodondi
- Institute for Primary Health Care (BIHAM), University of Bern, Bern, Switzerland; Department of General Internal Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Arnaud Chiolero
- Institute for Primary Health Care (BIHAM), University of Bern, Bern, Switzerland; Population Health Laboratory (#PopHealthLab), University of Fribourg, Fribourg, Switzerland; School of Population and Global Health, McGill University, Montreal, Canada
| | | |
Collapse
|
7
|
Al-Hassany L, Acarsoy C, Ikram MK, Bos D, MaassenVanDenBrink A. Sex-Specific Association of Cardiovascular Risk Factors With Migraine: The Population-Based Rotterdam Study. Neurology 2024; 103:e209700. [PMID: 39083723 PMCID: PMC11319068 DOI: 10.1212/wnl.0000000000209700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 06/20/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Although several lines of evidence suggest a link between migraine and cardiovascular events, less is known about the association between cardiovascular risk factors (CVRFs) and migraine. This knowledge is clinically important to provide directions on mitigating the cardiovascular risk in patients with migraine. We hypothesized that CVRFs are associated with a higher migraine prevalence. Therefore, our primary objective was to investigate sex-specific associations between CVRFs and lifetime prevalence of migraine. METHODS We performed cross-sectional analyses within an ongoing population-based cohort study (Rotterdam Study), including middle-aged and elderly individuals. By means of (structured) interviews, physical examinations, and blood sampling, we obtained information on the lifetime prevalence of migraine and the following traditional CVRFs: current smoking, obesity, hypercholesterolemia, hypertension, and diabetes mellitus. Similarly, we obtained information on quantitative component data on these CVRFs, including pack-years of smoking, lipid levels, systolic and diastolic blood pressure (BP), body mass index, and fasting glucose levels. Patients with migraine were age-matched to individuals without migraine, and we performed conditional logistic regression analyses to investigate the sex-stratified association of CVRFs with migraine. RESULTS In total, 7,266 community-dwelling middle-aged and elderly persons were included (median age 66.6 [IQR 56.4-74.8] years, 57.5% females). The lifetime prevalence of migraine was 14.9%. In females, current smoking (odds ratio (OR) 0.72, 95% CI 0.58-0.90), more pack-years (OR per SD increase 0.91, 95% CI 0.84-1.00), diabetes mellitus (OR 0.74, 95% CI 0.56-0.98), and higher fasting glucose levels (OR per SD increase in glucose 0.90, 95% CI 0.82 - 0.98) were all related to a lower migraine prevalence while a higher diastolic BP related to a higher migraine prevalence (OR per SD increase 1.16, 95% CI 1.04-1.29). In males, no significant associations between CVRFs and migraine were observed. DISCUSSION Traditional CVRFs were either unrelated or inversely related to migraine in middle-aged and elderly individuals, but only in females. In males, we did not find any association between CVRFs and migraine. Because only an increased diastolic BP was related to a higher migraine prevalence in females, our study contributes to the hypothesis that migraine is not directly associated with traditional CVRFs. Future studies are warranted to extrapolate these findings to younger populations.
Collapse
Affiliation(s)
- Linda Al-Hassany
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.A-H., A.M.), and Departments of Epidemiology (C.A., M.K.I., D.B.), Neurology (M.K.I.), Radiology and Nuclear Medicine (D.B.), Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Cevdet Acarsoy
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.A-H., A.M.), and Departments of Epidemiology (C.A., M.K.I., D.B.), Neurology (M.K.I.), Radiology and Nuclear Medicine (D.B.), Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - M Kamran Ikram
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.A-H., A.M.), and Departments of Epidemiology (C.A., M.K.I., D.B.), Neurology (M.K.I.), Radiology and Nuclear Medicine (D.B.), Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Daniel Bos
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.A-H., A.M.), and Departments of Epidemiology (C.A., M.K.I., D.B.), Neurology (M.K.I.), Radiology and Nuclear Medicine (D.B.), Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Antoinette MaassenVanDenBrink
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.A-H., A.M.), and Departments of Epidemiology (C.A., M.K.I., D.B.), Neurology (M.K.I.), Radiology and Nuclear Medicine (D.B.), Erasmus MC University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
8
|
Pan KY, van Tuijl L, Basten M, Rijnhart JJM, de Graeff A, Dekker J, Geerlings MI, Hoogendoorn A, Ranchor AV, Vermeulen R, Portengen L, Voogd AC, Abell J, Awadalla P, Beekman ATF, Bjerkeset O, Boyd A, Cui Y, Frank P, Galenkamp H, Garssen B, Hellingman S, Hollander M, Huisman M, Huss A, Keats MR, Kok AAL, Krokstad S, van Leeuwen FE, Luik AI, Noisel N, Payette Y, Penninx BWJH, Picavet S, Rissanen I, Roest AM, Rosmalen JGM, Ruiter R, Schoevers RA, Soave D, Spaan M, Steptoe A, Stronks K, Sund ER, Sweeney E, Teyhan A, Twait EL, van der Willik KD, Lamers F. The mediating role of health behaviors in the association between depression, anxiety and cancer incidence: an individual participant data meta-analysis. Psychol Med 2024; 54:2744-2757. [PMID: 38680088 DOI: 10.1017/s0033291724000850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
BACKGROUND Although behavioral mechanisms in the association among depression, anxiety, and cancer are plausible, few studies have empirically studied mediation by health behaviors. We aimed to examine the mediating role of several health behaviors in the associations among depression, anxiety, and the incidence of various cancer types (overall, breast, prostate, lung, colorectal, smoking-related, and alcohol-related cancers). METHODS Two-stage individual participant data meta-analyses were performed based on 18 cohorts within the Psychosocial Factors and Cancer Incidence consortium that had a measure of depression or anxiety (N = 319 613, cancer incidence = 25 803). Health behaviors included smoking, physical inactivity, alcohol use, body mass index (BMI), sedentary behavior, and sleep duration and quality. In stage one, path-specific regression estimates were obtained in each cohort. In stage two, cohort-specific estimates were pooled using random-effects multivariate meta-analysis, and natural indirect effects (i.e. mediating effects) were calculated as hazard ratios (HRs). RESULTS Smoking (HRs range 1.04-1.10) and physical inactivity (HRs range 1.01-1.02) significantly mediated the associations among depression, anxiety, and lung cancer. Smoking was also a mediator for smoking-related cancers (HRs range 1.03-1.06). There was mediation by health behaviors, especially smoking, physical inactivity, alcohol use, and a higher BMI, in the associations among depression, anxiety, and overall cancer or other types of cancer, but effects were small (HRs generally below 1.01). CONCLUSIONS Smoking constitutes a mediating pathway linking depression and anxiety to lung cancer and smoking-related cancers. Our findings underline the importance of smoking cessation interventions for persons with depression or anxiety.
Collapse
Affiliation(s)
- Kuan-Yu Pan
- Department of Psychiatry, Amsterdam UMC, location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Public Health, Mental Health program, Amsterdam, the Netherlands
- Unit of Occupational Medicine, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lonneke van Tuijl
- Health Psychology Section, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Department of Clinical Psychology, Utrecht University, Utrecht, the Netherlands
| | - Maartje Basten
- Amsterdam Public Health, Mental Health program, Amsterdam, the Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
- Department of Health Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Public Health, Health Behaviors and Chronic Diseases program, Amsterdam, the Netherlands
| | | | - Alexander de Graeff
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Joost Dekker
- Department of Psychiatry, Amsterdam UMC, location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Public Health, Mental Health program, Amsterdam, the Netherlands
| | - Mirjam I Geerlings
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
- Department of General Practice, Amsterdam UMC, location UvA, Amsterdam, the Netherlands
- Amsterdam Public Health, Aging & Later Life, and Personalized Medicine, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, and Mood, Anxiety, Psychosis, Stress, and Sleep, Amsterdam, the Netherlands
| | - Adriaan Hoogendoorn
- Department of Psychiatry, Amsterdam UMC, location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Adelita V Ranchor
- Health Psychology Section, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Roel Vermeulen
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, the Netherlands
| | - Lützen Portengen
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, the Netherlands
| | - Adri C Voogd
- Department of Epidemiology, Maastricht University, Maastricht, the Netherlands
- Department of Research and Development, Netherlands Comprehensive Cancer Organization (IKNL), Utrecht, the Netherlands
| | - Jessica Abell
- Department of Behavioral Science and Health, University College London, London, UK
| | - Philip Awadalla
- Ontario Institute for Cancer Research, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Aartjan T F Beekman
- Department of Psychiatry, Amsterdam UMC, location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Ottar Bjerkeset
- Faculty of Nursing and Health Sciences, Nord University, Levanger, Norway
| | - Andy Boyd
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Yunsong Cui
- Atlantic Partnership for Tomorrow's Health, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Philipp Frank
- Department of Behavioral Science and Health, University College London, London, UK
| | - Henrike Galenkamp
- Amsterdam Public Health, Health Behaviors and Chronic Diseases program, Amsterdam, the Netherlands
- Department of Public and Occupational Health, location University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Bert Garssen
- Health Psychology Section, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Sean Hellingman
- Department of Mathematics, Wilfrid Laurier University, Waterloo, Canada
| | - Monika Hollander
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | - Martijn Huisman
- Department of Epidemiology & Data Science, Amsterdam UMC, location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Department of Sociology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Anke Huss
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, the Netherlands
| | - Melanie R Keats
- Faculty of Health, School of Health and Human Performance, Dalhousie University, Halifax, NS, Canada
| | - Almar A L Kok
- Department of Psychiatry, Amsterdam UMC, location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Department of Epidemiology & Data Science, Amsterdam UMC, location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Steinar Krokstad
- Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, HUNT Research Centre, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - Flora E van Leeuwen
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Annemarie I Luik
- Department of Epidemiology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Nolwenn Noisel
- CARTaGENE, CHU Sainte-Justine, 3175, Chemin de la Côte-Sainte-Catherine, Montréal, QC, Canada
| | - Yves Payette
- CARTaGENE, CHU Sainte-Justine, 3175, Chemin de la Côte-Sainte-Catherine, Montréal, QC, Canada
| | - Brenda W J H Penninx
- Department of Psychiatry, Amsterdam UMC, location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Public Health, Mental Health program, Amsterdam, the Netherlands
| | - Susan Picavet
- Centre for Nutrition, Prevention and Health Services, National Institute for Public Health and the Environment, Utrecht Bilthoven, the Netherlands
| | - Ina Rissanen
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | - Annelieke M Roest
- Department of Developmental Psychology, University of Groningen, Groningen, the Netherlands
| | - Judith G M Rosmalen
- Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Department of Psychiatry, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Rikje Ruiter
- Department of Epidemiology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
- Department of Internal Medicine, Maasstad Hospital, Rotterdam, the Netherlands
| | - Robert A Schoevers
- Department of Psychiatry, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - David Soave
- Ontario Institute for Cancer Research, Toronto, ON, Canada
- Department of Mathematics, Wilfrid Laurier University, Waterloo, Canada
| | - Mandy Spaan
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Andrew Steptoe
- Department of Behavioral Science and Health, University College London, London, UK
| | - Karien Stronks
- Department of Public and Occupational Health, location University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- Center for Urban Mental Health, University of Amsterdam, Amsterdam, the Netherlands
| | - Erik R Sund
- Faculty of Nursing and Health Sciences, Nord University, Levanger, Norway
- Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, HUNT Research Centre, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - Ellen Sweeney
- Atlantic Partnership for Tomorrow's Health, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Alison Teyhan
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Emma L Twait
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
- Amsterdam Public Health, Aging & Later Life, and Personalized Medicine, Amsterdam, the Netherlands
- Department of General Practice, Amsterdam UMC, location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Kimberly D van der Willik
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Epidemiology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Femke Lamers
- Department of Psychiatry, Amsterdam UMC, location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Public Health, Mental Health program, Amsterdam, the Netherlands
| |
Collapse
|
9
|
Jaye S, Sandau US, Saugstad JA. Clathrin mediated endocytosis in Alzheimer's disease: cell type specific involvement in amyloid beta pathology. Front Aging Neurosci 2024; 16:1378576. [PMID: 38694257 PMCID: PMC11061891 DOI: 10.3389/fnagi.2024.1378576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/03/2024] [Indexed: 05/04/2024] Open
Abstract
This review provides a comprehensive examination of the role of clathrin-mediated endocytosis (CME) in Alzheimer's disease (AD) pathogenesis, emphasizing its impact across various cellular contexts beyond neuronal dysfunction. In neurons, dysregulated CME contributes to synaptic dysfunction, amyloid beta (Aβ) processing, and Tau pathology, highlighting its involvement in early AD pathogenesis. Furthermore, CME alterations extend to non-neuronal cell types, including astrocytes and microglia, which play crucial roles in Aβ clearance and neuroinflammation. Dysregulated CME in these cells underscores its broader implications in AD pathophysiology. Despite significant progress, further research is needed to elucidate the precise mechanisms underlying CME dysregulation in AD and its therapeutic implications. Overall, understanding the complex interplay between CME and AD across diverse cell types holds promise for identifying novel therapeutic targets and interventions.
Collapse
Affiliation(s)
| | | | - Julie A. Saugstad
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
10
|
Joshi A, Li H, Parikh NA, He L. A systematic review of automated methods to perform white matter tract segmentation. Front Neurosci 2024; 18:1376570. [PMID: 38567281 PMCID: PMC10985163 DOI: 10.3389/fnins.2024.1376570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 03/04/2024] [Indexed: 04/04/2024] Open
Abstract
White matter tract segmentation is a pivotal research area that leverages diffusion-weighted magnetic resonance imaging (dMRI) for the identification and mapping of individual white matter tracts and their trajectories. This study aims to provide a comprehensive systematic literature review on automated methods for white matter tract segmentation in brain dMRI scans. Articles on PubMed, ScienceDirect [NeuroImage, NeuroImage (Clinical), Medical Image Analysis], Scopus and IEEEXplore databases and Conference proceedings of Medical Imaging Computing and Computer Assisted Intervention Society (MICCAI) and International Symposium on Biomedical Imaging (ISBI), were searched in the range from January 2013 until September 2023. This systematic search and review identified 619 articles. Adhering to the specified search criteria using the query, "white matter tract segmentation OR fiber tract identification OR fiber bundle segmentation OR tractography dissection OR white matter parcellation OR tract segmentation," 59 published studies were selected. Among these, 27% employed direct voxel-based methods, 25% applied streamline-based clustering methods, 20% used streamline-based classification methods, 14% implemented atlas-based methods, and 14% utilized hybrid approaches. The paper delves into the research gaps and challenges associated with each of these categories. Additionally, this review paper illuminates the most frequently utilized public datasets for tract segmentation along with their specific characteristics. Furthermore, it presents evaluation strategies and their key attributes. The review concludes with a detailed discussion of the challenges and future directions in this field.
Collapse
Affiliation(s)
- Ankita Joshi
- Imaging Research Center, Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
- Neurodevelopmental Disorders Prevention Center, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Hailong Li
- Imaging Research Center, Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
- Neurodevelopmental Disorders Prevention Center, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
- Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Nehal A. Parikh
- Neurodevelopmental Disorders Prevention Center, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Lili He
- Imaging Research Center, Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
- Neurodevelopmental Disorders Prevention Center, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
- Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Computer Science, Biomedical Informatics, and Biomedical Engineering, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
11
|
Marzlin N, Chapel A, Adefisoye J, Garg K, Zlochiver V, Walczak S, Plautz D, Peterson M, Muthukumar L, Harland DR, Jain R, Port S, Galazka P. Differences and Disparities Among Self-Referred and Physician-Referred Populations Undergoing Coronary Artery Calcium Scanning. Circ Cardiovasc Imaging 2024; 17:e015712. [PMID: 38377241 DOI: 10.1161/circimaging.123.015712] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 01/12/2024] [Indexed: 02/22/2024]
Abstract
BACKGROUND Coronary artery calcium computed tomography (CAC) is an important tool for identifying subclinical atherosclerosis and cardiovascular risk stratification. Despite robust evidence and inclusion in current guidelines, CAC is considered investigational by some US insurance carriers and requires out-of-pocket expenses. CAC can be obtained via self-referral (SR) or physician referral (PR). We aimed to examine differences in patient, socioeconomic, and CAC characteristics between referral groups. METHODS We evaluated demographic, medical history, and CAC results of consecutive patients with a CAC completed at one of multiple Wisconsin sites from March 1, 2019, to June 30, 2021. We separated patients into SR and PR groups. Through census data, we analyzed socioeconomic variables at the block level including race and ethnicity, median income, average household size, and high school completion in the areas where patients resided at the time of CAC. RESULTS The final analysis included 19 726 patients: 13 835 (70.1%) PR and 5891 (29.9%) SR. Most patients in both groups were White (95.2% versus 95.1%), with the Black/African American population representing 2.7% (SR) and 2.3% (PR). The PR group had a higher prevalence of cardiovascular risk factors. SR patients were more likely to have a score of 0 (41.2% versus 38.1%; P<0.001); PR patients had a higher prevalence of CAC >300 (16.8% versus 14.8%; P<0.001). SR patients were more likely to be women (55.1% versus 48.9%; P<0.001) and were found to live in higher income areas (19.5% versus 16.4%; P<0.001). Patients from low-income areas comprised the smallest proportion in both groups (7.5%). CONCLUSIONS Patients who obtain out-of-pocket CAC live predominantly in medium- and high-income areas, and patients from lower income locations are less likely to obtain CAC despite having more cardiovascular disease risk factors. Consideration should be made from a policy perspective to promote health equity and improve utilization of CAC testing among underrepresented groups.
Collapse
Affiliation(s)
- Nathan Marzlin
- Aurora Cardiovascular and Thoracic Services, Aurora Sinai/Aurora St. Luke's Medical Centers, Advocate Aurora Health, University of Wisconsin School of Medicine and Public Health, Milwaukee, WI (N.M, A.C, J.A, S.W., D.P, M.P, L.M., D.H., R.J., S.P., P.G.)
| | - Andrew Chapel
- Aurora Cardiovascular and Thoracic Services, Aurora Sinai/Aurora St. Luke's Medical Centers, Advocate Aurora Health, University of Wisconsin School of Medicine and Public Health, Milwaukee, WI (N.M, A.C, J.A, S.W., D.P, M.P, L.M., D.H., R.J., S.P., P.G.)
| | - James Adefisoye
- Aurora Cardiovascular and Thoracic Services, Aurora Sinai/Aurora St. Luke's Medical Centers, Advocate Aurora Health, University of Wisconsin School of Medicine and Public Health, Milwaukee, WI (N.M, A.C, J.A, S.W., D.P, M.P, L.M., D.H., R.J., S.P., P.G.)
| | - Kritika Garg
- Academic Affairs, Cardiovascular Research, Aurora Sinai/Aurora St. Luke's Medical Center, Milwaukee, WI (K.G., V.Z.)
| | - Viviana Zlochiver
- Academic Affairs, Cardiovascular Research, Aurora Sinai/Aurora St. Luke's Medical Center, Milwaukee, WI (K.G., V.Z.)
| | - Sara Walczak
- Aurora Cardiovascular and Thoracic Services, Aurora Sinai/Aurora St. Luke's Medical Centers, Advocate Aurora Health, University of Wisconsin School of Medicine and Public Health, Milwaukee, WI (N.M, A.C, J.A, S.W., D.P, M.P, L.M., D.H., R.J., S.P., P.G.)
| | - Doreen Plautz
- Aurora Cardiovascular and Thoracic Services, Aurora Sinai/Aurora St. Luke's Medical Centers, Advocate Aurora Health, University of Wisconsin School of Medicine and Public Health, Milwaukee, WI (N.M, A.C, J.A, S.W., D.P, M.P, L.M., D.H., R.J., S.P., P.G.)
| | - Michael Peterson
- Aurora Cardiovascular and Thoracic Services, Aurora Sinai/Aurora St. Luke's Medical Centers, Advocate Aurora Health, University of Wisconsin School of Medicine and Public Health, Milwaukee, WI (N.M, A.C, J.A, S.W., D.P, M.P, L.M., D.H., R.J., S.P., P.G.)
| | - Lakshmi Muthukumar
- Aurora Cardiovascular and Thoracic Services, Aurora Sinai/Aurora St. Luke's Medical Centers, Advocate Aurora Health, University of Wisconsin School of Medicine and Public Health, Milwaukee, WI (N.M, A.C, J.A, S.W., D.P, M.P, L.M., D.H., R.J., S.P., P.G.)
| | - Daniel R Harland
- Aurora Cardiovascular and Thoracic Services, Aurora Sinai/Aurora St. Luke's Medical Centers, Advocate Aurora Health, University of Wisconsin School of Medicine and Public Health, Milwaukee, WI (N.M, A.C, J.A, S.W., D.P, M.P, L.M., D.H., R.J., S.P., P.G.)
| | - Renuka Jain
- Aurora Cardiovascular and Thoracic Services, Aurora Sinai/Aurora St. Luke's Medical Centers, Advocate Aurora Health, University of Wisconsin School of Medicine and Public Health, Milwaukee, WI (N.M, A.C, J.A, S.W., D.P, M.P, L.M., D.H., R.J., S.P., P.G.)
| | - Steven Port
- Aurora Cardiovascular and Thoracic Services, Aurora Sinai/Aurora St. Luke's Medical Centers, Advocate Aurora Health, University of Wisconsin School of Medicine and Public Health, Milwaukee, WI (N.M, A.C, J.A, S.W., D.P, M.P, L.M., D.H., R.J., S.P., P.G.)
| | - Patrycja Galazka
- Aurora Cardiovascular and Thoracic Services, Aurora Sinai/Aurora St. Luke's Medical Centers, Advocate Aurora Health, University of Wisconsin School of Medicine and Public Health, Milwaukee, WI (N.M, A.C, J.A, S.W., D.P, M.P, L.M., D.H., R.J., S.P., P.G.)
| |
Collapse
|
12
|
Zheng E, Madura P, Grandos J, Broncel M, Pawlos A, Woźniak E, Gorzelak-Pabiś P. When the same treatment has different response: The role of pharmacogenomics in statin therapy. Biomed Pharmacother 2024; 170:115966. [PMID: 38061135 DOI: 10.1016/j.biopha.2023.115966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 11/22/2023] [Accepted: 11/27/2023] [Indexed: 01/10/2024] Open
Abstract
Statins, also known as HMG-CoA reductase inhibitors, are one of the most potently prescribed and thoroughly researched medications, predominantly utilized for managing cardiovascular diseases by modulating serum cholesterol levels. Despite the well-documented efficacy of statins in reducing overall mortality via attenuating the risk of cardiovascular diseases, notable interindividual variability in therapeutic responses persists as such variability could compromise the lipid-lowering efficacy of the drug, potentially increasing susceptibility to adverse effects or attenuating therapeutic outcomes.This phenomenon has catalysed a growing interest in the scientific community to explore common genetic polymorphisms within genes that encode for pivotal enzymes within the pharmacokinetic pathways of statins. In our review, we focus to provide insight into potentially clinically relevant polymorphisms associated with statins' pharmacokinetic participants and assess their consequent implications on modulating the therapeutic outcomes of statins among distinct genetic carrier.
Collapse
Affiliation(s)
- Edward Zheng
- Dept. of Internal Diseases and Clinical Pharmacology, The Laboratory of Tissue Immunopharmacology, Medical University of Lodz, Poland
| | - Paulina Madura
- Dept. of Internal Diseases and Clinical Pharmacology, The Laboratory of Tissue Immunopharmacology, Medical University of Lodz, Poland
| | - Jakub Grandos
- Dept. of Internal Diseases and Clinical Pharmacology, The Laboratory of Tissue Immunopharmacology, Medical University of Lodz, Poland
| | - Marlena Broncel
- Dept. of Internal Diseases and Clinical Pharmacology, The Laboratory of Tissue Immunopharmacology, Medical University of Lodz, Poland
| | - Agnieszka Pawlos
- Dept. of Internal Diseases and Clinical Pharmacology, The Laboratory of Tissue Immunopharmacology, Medical University of Lodz, Poland
| | - Ewelina Woźniak
- Dept. of Internal Diseases and Clinical Pharmacology, The Laboratory of Tissue Immunopharmacology, Medical University of Lodz, Poland
| | - Paulina Gorzelak-Pabiś
- Dept. of Internal Diseases and Clinical Pharmacology, The Laboratory of Tissue Immunopharmacology, Medical University of Lodz, Poland.
| |
Collapse
|
13
|
Tiwari R, Verma S, Verma N, Verma D, Narayan J. Correlation of serum uric acid levels with certain anthropometric parameters in prediabetic and drug-naive diabetic subjects. Ann Afr Med 2024; 23:13-18. [PMID: 38358165 PMCID: PMC10922179 DOI: 10.4103/aam.aam_40_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/22/2023] [Accepted: 06/13/2023] [Indexed: 02/16/2024] Open
Abstract
Introduction Uric acid is produced during the metabolism of nucleotide and adenosine triphosphate and contains the final product of human purine metabolism. It acts both as an antioxidant and pro-inflammatory marker and has a positive association with visceral fat in overweight subjects. The aim of the present study is to find an association of uric acid level with certain anthropometric parameters in subjects having type 2 diabetes. Materials and Methods The study included 124 urban drug-naive diabetic Indian subjects above 18 years of age from the general population of the city of North India. Uric acid concentrations were estimated by the uricase method. Fasting plasma glucose (FPG) concentrations were estimated by the glucose oxidase-peroxidase method. Anthropometric measurements and information on lifestyle factors and disease history were collected through in-person meeting. Results All participants of the study subjects had a body mass index (BMI) of more than 23.5. BMI, waist-to-hip ratio (WHR), waist-to-height ratio, waist circumference, neck circumference, weight, age, sagittal abdominal diameter (SAD), skinfold thickness, and body roundness index were positively correlated with the serum uric acid level. The correlation of weight, BMI, SAD, and WHR was statistically significant. Conclusion We found that serum uric acid level increases as body fat content increases. Statistical data show remarkable results for a significant correlation of uric acid level with BMI, WHR, SAD, and FPG. Hypertrophy occurs as a result of inflammatory processes and oxidative stress when the supply of energy starts to exceed the storage capacity of adipocytes, as a result, adipokines such as interleukin (IL)-1, IL-6, and tumor-necrosis factor-alpha are released more frequently which lead to low-grade chronic inflammation. Uric acid levels are much lean toward visceral obesity than overall body fat content.
Collapse
Affiliation(s)
- Ritu Tiwari
- Department of Physiology, King George Medical University, Lucknow, Uttar Pradesh, India
| | - Shivam Verma
- Department of Physiology, King George Medical University, Lucknow, Uttar Pradesh, India
| | - Narsingh Verma
- Department of Physiology, King George Medical University, Lucknow, Uttar Pradesh, India
| | - Dileep Verma
- Department of Physiology, King George Medical University, Lucknow, Uttar Pradesh, India
| | - Jagdish Narayan
- Department of Physiology, King George Medical University, Lucknow, Uttar Pradesh, India
| |
Collapse
|
14
|
Grant JK, Orringer CE. Coronary and Extra-coronary Subclinical Atherosclerosis to Guide Lipid-Lowering Therapy. Curr Atheroscler Rep 2023; 25:911-920. [PMID: 37971683 DOI: 10.1007/s11883-023-01161-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2023] [Indexed: 11/19/2023]
Abstract
PURPOSE OF REVIEW To discuss and review the technical considerations, fundamentals, and guideline-based indications for coronary artery calcium scoring, and the use of other non-invasive imaging modalities, such as extra-coronary calcification in cardiovascular risk prediction. RECENT FINDINGS The most robust evidence for the use of CAC scoring is in select individuals, 40-75 years of age, at borderline to intermediate 10-year ASCVD risk. Recent US recommendations support the use of CAC scoring in varying clinical scenarios. First, in adults with very high CAC scores (CAC ≥ 1000), the use of high-intensity statin therapy and, if necessary, guideline-based add-on LDL-C lowering therapies (ezetimibe, PCSK9-inhibitors) to achieve a ≥ 50% reduction in LDL-C and optimally an LDL-C < 70 mg/dL is recommended. In patients with a CAC score ≥ 100 at low risk of bleeding, the benefits of aspirin use may outweigh the risk of bleeding. Other applications of CAC scoring include risk estimation on non-contrast CT scans of the chest, risk prediction in younger patients (< 40 years of age), its value as a gatekeeper for the decision to perform nuclear stress testing, and to aid in risk stratification in patients presenting with low-risk chest pain. There is a correlation between extra-coronary calcification (e.g., breast arterial calcification, aortic calcification, and aortic valve calcification) and incident ASCVD events. However, its role in informing lipid management remains unclear. Identification of coronary calcium in selected patients is the single best non-invasive imaging modality to identify future ASCVD risk and inform lipid-lowering therapy decision-making.
Collapse
Affiliation(s)
- Jelani K Grant
- Johns Hopkins Ciccarone Center for the Prevention of Heart Disease, Baltimore, MD, USA
| | - Carl E Orringer
- NCH Rooney Heart Institute, 399 9th Street North, Suite 300, Naples, FL, 34102, USA.
| |
Collapse
|
15
|
Touilloux B, Bongard C, Lechartier B, Truong MK, Marques-Vidal P, Vollenweider P, Vaucher J, Casutt A, von Garnier C. Prevalence of small airway dysfunction in the Swiss PneumoLaus Cohort. ERJ Open Res 2023; 9:00381-2023. [PMID: 37701366 PMCID: PMC10493711 DOI: 10.1183/23120541.00381-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 07/03/2023] [Indexed: 09/14/2023] Open
Abstract
Background Recent evidence identified exposure to particulate matter of size ≤2.5 µm (PM2.5) as a risk factor for high prevalence of small airway dysfunction (SAD). We assessed the prevalence of SAD in a European region with low air pollution levels. Methods SAD was defined as a maximum mid-expiratory flow (MMEF) <65% of predicted value (PV) or MMEF Results Among 3351 participants (97.6% Caucasian, 55.7% female sex, mean age 62.7 years), we observed MMEF <65% PV in 425 (12.7%) and MMEF 65 years only. In an area where ambient PM2.5 concentration was <15 µg·m-3 during the observation period (2010 and 2020), ≥72% of participants with SAD were ever-smokers. Conclusions The observed low prevalence of SAD of 5.0-12.7% depending on criteria employed may be related to lower PM2.5 exposure. Smoking was the main factor associated with SAD in an area with low PM2.5 exposure. Employing a MMEF threshold <65% PV carries a risk of SAD overdiagnosis in elderly individuals.
Collapse
Affiliation(s)
- Brice Touilloux
- Division of Pulmonology, Department of Medicine, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
- Division of Pulmonology, Department of Medicine and Specialties, Fribourg Hospital, Fribourg, Switzerland
| | - Cedric Bongard
- Division of Pulmonology, Department of Medicine, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Benoit Lechartier
- Division of Pulmonology, Department of Medicine, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Minh Khoa Truong
- Division of Pulmonology, Department of Medicine, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Pedro Marques-Vidal
- Division of Internal Medicine, Department of Medicine, CHUV and UNIL, Lausanne, Switzerland
| | - Peter Vollenweider
- Division of Internal Medicine, Department of Medicine, CHUV and UNIL, Lausanne, Switzerland
| | - Julien Vaucher
- Division of Internal Medicine, Department of Medicine, CHUV and UNIL, Lausanne, Switzerland
- Division of Internal Medicine, Department of Medicine and Specialties, Fribourg Hospital, Fribourg, Switzerland
- University of Fribourg, Fribourg, Switzerland
| | - Alessio Casutt
- Division of Pulmonology, Department of Medicine, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
- Division of Pneumology, Ospedale Regionale di Lugano, Ente Ospedaliero Cantonale, Lugano, Switzerland
- These authors contributed equally
| | - Christophe von Garnier
- Division of Pulmonology, Department of Medicine, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
- These authors contributed equally
| |
Collapse
|
16
|
Hangartner K, Bajka A, Wiest MRJ, Sidhu S, Toro MD, Maloca PM, Zweifel SA. Assessment of Retinal Vessel Tortuosity Index in Patients with Fabry Disease Using Optical Coherence Tomography Angiography (OCTA). Diagnostics (Basel) 2023; 13:2496. [PMID: 37568859 PMCID: PMC10417007 DOI: 10.3390/diagnostics13152496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 07/16/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Vessel tortuosity (VT) is a parameter used to assess retinal involvement in patients affected by systemic diseases such as Fabry disease (FD). In this study, we assessed a retinal VT index (VTI) using optical coherence tomography angiography (OCTA) in a group of patients with FD (FD cohort) compared to a healthy control group (HC cohort). This is a single-center, retrospective study analysis of all consecutive patients with genetically tested and confirmed FD who underwent regular ophthalmological visits from December 2017 to January 2020 at the Department of Ophthalmology at the University Hospital of Zurich, Switzerland. VTI was calculated for each OCTA image and the results were compared between FD and HC cohort. A total of 56 participants, 32 (male:female ratio 12:20) in the FD cohort and 24 (male:female ratio 13:11) in the HC cohort. Classic onset was determined in 18 patients. Overall, mean VTI (±SD) was 0.21 (±0.07). Male patients with classic-onset FD had a significantly higher mean VTI (0.33, SD ± 0.35) compared to all other subgroups (p-value < 0.05). Further investigations of retinal VTI in patients with FD could be helpful to use OCTA as a noninvasive screening and follow-up modality to assess disease progression in affected patients.
Collapse
Affiliation(s)
- Kevin Hangartner
- Faculty of Human Medicine, University of Zurich, 8032 Zurich, Switzerland
| | - Anahita Bajka
- Faculty of Human Medicine, University of Zurich, 8032 Zurich, Switzerland
- Department of Ophthalmology, University Hospital of Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Maximilian R. J. Wiest
- Faculty of Human Medicine, University of Zurich, 8032 Zurich, Switzerland
- Department of Ophthalmology, University Hospital of Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Sophia Sidhu
- Faculty of Medicine, University of California San Diego, 5998 Alcala Park, San Diego, CA 92110, USA
| | - Mario D. Toro
- Department of Ophthalmology, University Hospital of Zurich, University of Zurich, 8091 Zurich, Switzerland
- Chair and Department of General and Pediatric Ophthalmology, Medical University of Lublin, 20079 Lublin, Poland
- Eye Clinic, Department of Public Health, University Federico II, 80131 Naples, Italy
| | - Peter M. Maloca
- Institute of Molecular and Clinical Ophthalmology Basel (IOB), 4031 Basel, Switzerland
- Department of Ophthalmology, University Hospital Basel, 4031 Basel, Switzerland
- Moorfields Eye Hospital NHS Foundation Trust, London EC1V 2PD, UK
| | - Sandrine A. Zweifel
- Faculty of Human Medicine, University of Zurich, 8032 Zurich, Switzerland
- Department of Ophthalmology, University Hospital of Zurich, University of Zurich, 8091 Zurich, Switzerland
| |
Collapse
|
17
|
Ghazi N, Aarabi MH, Soltanian-Zadeh H. Deep Learning Methods for Identification of White Matter Fiber Tracts: Review of State-of-the-Art and Future Prospective. Neuroinformatics 2023; 21:517-548. [PMID: 37328715 DOI: 10.1007/s12021-023-09636-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2023] [Indexed: 06/18/2023]
Abstract
Quantitative analysis of white matter fiber tracts from diffusion Magnetic Resonance Imaging (dMRI) data is of great significance in health and disease. For example, analysis of fiber tracts related to anatomically meaningful fiber bundles is highly demanded in pre-surgical and treatment planning, and the surgery outcome depends on accurate segmentation of the desired tracts. Currently, this process is mainly done through time-consuming manual identification performed by neuro-anatomical experts. However, there is a broad interest in automating the pipeline such that it is fast, accurate, and easy to apply in clinical settings and also eliminates the intra-reader variabilities. Following the advancements in medical image analysis using deep learning techniques, there has been a growing interest in using these techniques for the task of tract identification as well. Recent reports on this application show that deep learning-based tract identification approaches outperform existing state-of-the-art methods. This paper presents a review of current tract identification approaches based on deep neural networks. First, we review the recent deep learning methods for tract identification. Next, we compare them with respect to their performance, training process, and network properties. Finally, we end with a critical discussion of open challenges and possible directions for future works.
Collapse
Affiliation(s)
- Nayereh Ghazi
- Control and Intelligent Processing Center of Excellence (CIPCE), School of Electrical and Computer Engineering, College of Engineering, University of Tehran, Tehran, 14399, Iran
| | - Mohammad Hadi Aarabi
- Department of Neuroscience, University of Padova, Padova, Italy
- Padova Neuroscience Center (PNC), University of Padova, Padova, Italy
| | - Hamid Soltanian-Zadeh
- Control and Intelligent Processing Center of Excellence (CIPCE), School of Electrical and Computer Engineering, College of Engineering, University of Tehran, Tehran, 14399, Iran.
- Medical Image Analysis Laboratory, Departments of Radiology and Research Administration, Henry Ford Health System, Detroit, MI, 48202, USA.
| |
Collapse
|
18
|
Vosberg DE, Pausova Z, Paus T. The genetics of a "femaleness/maleness" score in cardiometabolic traits in the UK biobank. Sci Rep 2023; 13:9109. [PMID: 37277458 DOI: 10.1038/s41598-023-36132-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 05/30/2023] [Indexed: 06/07/2023] Open
Abstract
We recently devised continuous "sex-scores" that sum up multiple quantitative traits, weighted by their respective sex-difference effect sizes, as an approach to estimating polyphenotypic "maleness/femaleness" within each binary sex. To identify the genetic architecture underlying these sex-scores, we conducted sex-specific genome-wide association studies (GWASs) in the UK Biobank cohort (females: n = 161,906; males: n = 141,980). As a control, we also conducted GWASs of sex-specific "sum-scores", simply aggregating the same traits, without weighting by sex differences. Among GWAS-identified genes, while sum-score genes were enriched for genes differentially expressed in the liver in both sexes, sex-score genes were enriched for genes differentially expressed in the cervix and across brain tissues, particularly for females. We then considered single nucleotide polymorphisms with significantly different effects (sdSNPs) between the sexes for sex-scores and sum-scores, mapping to male-dominant and female-dominant genes. Here, we identified brain-related enrichment for sex-scores, especially for male-dominant genes; these findings were present but weaker for sum-scores. Genetic correlation analyses of sex-biased diseases indicated that both sex-scores and sum-scores were associated with cardiometabolic, immune, and psychiatric disorders.
Collapse
Affiliation(s)
- Daniel E Vosberg
- Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Montreal, QC, Canada
- Department of Neuroscience, Faculty of Medicine, University of Montreal, Montreal, QC, Canada
- Research Institute of the Hospital for Sick Children, Toronto, ON, Canada
| | - Zdenka Pausova
- Research Institute of the Hospital for Sick Children, Toronto, ON, Canada
- Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| | - Tomáš Paus
- Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Montreal, QC, Canada.
- Department of Neuroscience, Faculty of Medicine, University of Montreal, Montreal, QC, Canada.
- Departments of Psychology and Psychiatry, University of Toronto, Toronto, ON, Canada.
- ECOGENE-21, Chicoutimi, QC, Canada.
- Department of Psychiatry and Addictology, Faculty of Medicine, University of Montreal, Montreal, QC, Canada.
| |
Collapse
|
19
|
Zhou W, van Rooij JGJ, van de Laarschot DM, Zervou Z, Bruggenwirth H, Appelman‐Dijkstra NM, Ebeling PR, Demirdas S, Verkerk AJMH, Zillikens MC. Prevalence of Monogenic Bone Disorders in a Dutch Cohort of Atypical Femur Fracture Patients. J Bone Miner Res 2023; 38:896-906. [PMID: 37076969 PMCID: PMC10946469 DOI: 10.1002/jbmr.4801] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 02/12/2023] [Accepted: 03/07/2023] [Indexed: 04/21/2023]
Abstract
Atypical femur fractures (AFFs), considered rare associations of bisphosphonates, have also been reported in patients with monogenic bone disorders without bisphosphonate use. The exact association between AFFs and monogenic bone disorders remains unknown. Our aim was to determine the prevalence of monogenic bone disorders in a Dutch AFF cohort. AFF patients were recruited from two specialist bone centers in the Netherlands. Medical records of the AFF patients were reviewed for clinical features of monogenic bone disorders. Genetic variants identified by whole-exome sequencing in 37 candidate genes involved in monogenic bone disorders were classified based on the American College of Medical Genetics and Genomics (ACMG) classification guidelines. Copy number variations overlapping the candidate genes were also evaluated using DNA array genotyping data. The cohort comprises 60 AFF patients (including a pair of siblings), with 95% having received bisphosphonates. Fifteen AFF patients (25%) had clinical features of monogenic bone disorders. Eight of them (54%), including the pair of siblings, had a (likely) pathogenic variant in either PLS3, COL1A2, LRP5, or ALPL. One patient carried a likely pathogenic variant in TCIRG1 among patients not suspected of monogenic bone disorders (2%). In total, nine patients in this AFF cohort (15%) had a (likely) pathogenic variant. In one patient, we identified a 12.7 Mb deletion in chromosome 6, encompassing TENT5A. The findings indicate a strong relationship between AFFs and monogenic bone disorders, particularly osteogenesis imperfecta and hypophosphatasia, but mainly in individuals with symptoms of these disorders. The high yield of (likely) pathogenic variants in AFF patients with a clinical suspicion of these disorders stresses the importance of careful clinical evaluation of AFF patients. Although the relevance of bisphosphonate use in this relationship is currently unclear, clinicians should consider these findings in medical management of these patients. © 2023 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Wei Zhou
- Department of Internal MedicineErasmus MCRotterdamThe Netherlands
| | | | | | - Zografia Zervou
- Department of Internal MedicineErasmus MCRotterdamThe Netherlands
| | | | - Natasha M Appelman‐Dijkstra
- Department of Internal Medicine, Division of EndocrinologyLeiden University Medical CenterLeidenThe Netherlands
| | - Peter R Ebeling
- Department of MedicineSchool of Clinical Sciences, Monash UniversityClaytonAustralia
| | - Serwet Demirdas
- Department of Clinical GeneticsErasmus MCRotterdamThe Netherlands
| | | | | |
Collapse
|
20
|
Bosman A, Koek WNH, Campos-Obando N, van der Eerden BCJ, Ikram MA, Uitterlinden AG, van Leeuwen JPTM, Zillikens MC. Sexual dimorphisms in serum calcium and phosphate concentrations in the Rotterdam Study. Sci Rep 2023; 13:8310. [PMID: 37221192 DOI: 10.1038/s41598-023-34800-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 05/08/2023] [Indexed: 05/25/2023] Open
Abstract
Sex differences in serum phosphate and calcium have been reported but the exact nature and underlying regulatory mechanisms remain unclear. We aimed to compare calcium and phosphate concentrations between sexes, and explore potential covariates to elucidate underlying mechanisms of sex differences in a prospective, population-based cohort study. Pooled data of subjects > 45 years from three independent cohorts of the Rotterdam Study (RS) were used: RS-I-3 (n = 3623), RS-II-1 (n = 2394), RS-III-1 (n = 3241), with separate analyses from an additional time point of the first cohort RS-I-1 (n = 2688). Compared to men, women had significantly higher total serum calcium and phosphate concentrations which was not explained by BMI, kidney function nor smoking. Adjustment for serum estradiol diminished sex differences in serum calcium while adjustment for serum testosterone diminished sex differences in serum phosphate. Adjustment for vitamin D and alkaline phosphatase did not change the association between sex and calcium or phosphate in RS-I-1. In the sex-combined group, both serum calcium and phosphate decreased with age with a significant interaction for sex differences for serum calcium but not phosphate. In sex-stratified analyses, serum estradiol but not testosterone was inversely associated with serum calcium in both sexes. Serum estradiol was inversely associated with serum phosphate in both sexes to a similar degree, while serum testosterone was inversely associated with serum phosphate in both sexes with an apparent stronger effect in men than in women. Premenopausal women had lower serum phosphate compared to postmenopausal women. Serum testosterone was inversely associated with serum phosphate in postmenopausal women only. In conclusion, women > 45 years have higher serum calcium and phosphate concentrations compared to men of similar age, not explained by vitamin D or alkaline phosphatase concentrations. Serum estradiol but not testosterone was inversely associated with serum calcium while serum testosterone was inversely associated with serum phosphate in both sexes. Serum testosterone may in part explain sex differences in serum phosphate while estradiol could partly explain sex differences in serum calcium.
Collapse
Affiliation(s)
- Ariadne Bosman
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - W Nadia H Koek
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Natalia Campos-Obando
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Bram C J van der Eerden
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - M A Ikram
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - André G Uitterlinden
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Johannes P T M van Leeuwen
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - M C Zillikens
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands.
| |
Collapse
|
21
|
Khan SS, Post WS, Guo X, Tan J, Zhu F, Bos D, Sedaghati-Khayat B, van Rooij J, Aday A, Allen NB, Bos MM, Uitterlinden AG, Budoff MJ, Lloyd-Jones DM, Mosley JD, Rotter JI, Greenland P, Kavousi M. Coronary Artery Calcium Score and Polygenic Risk Score for the Prediction of Coronary Heart Disease Events. JAMA 2023; 329:1768-1777. [PMID: 37219552 PMCID: PMC10208141 DOI: 10.1001/jama.2023.7575] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 04/19/2023] [Indexed: 05/24/2023]
Abstract
Importance Coronary artery calcium score and polygenic risk score have each separately been proposed as novel markers to identify risk of coronary heart disease (CHD), but no prior studies have directly compared these markers in the same cohorts. Objective To evaluate change in CHD risk prediction when a coronary artery calcium score, a polygenic risk score, or both are added to a traditional risk factor-based model. Design, Setting, and Participants Two observational population-based studies involving individuals aged 45 years through 79 years of European ancestry and free of clinical CHD at baseline: the Multi-Ethnic Study of Atherosclerosis (MESA) study involved 1991 participants at 6 US centers and the Rotterdam Study (RS) involved 1217 in Rotterdam, the Netherlands. Exposure Traditional risk factors were used to calculate CHD risk (eg, pooled cohort equations [PCEs]), computed tomography for the coronary artery calcium score, and genotyped samples for a validated polygenic risk score. Main Outcomes and Measures Model discrimination, calibration, and net reclassification improvement (at the recommended risk threshold of 7.5%) for prediction of incident CHD events were assessed. Results The median age was 61 years in MESA and 67 years in RS. Both log (coronary artery calcium+1) and polygenic risk score were significantly associated with 10-year risk of incident CHD (hazards ratio per SD, 2.60; 95% CI, 2.08-3.26 and 1.43; 95% CI, 1.20-1.71, respectively), in MESA. The C statistic for the coronary artery calcium score was 0.76 (95% CI, 0.71-0.79) and for the polygenic risk score, 0.69 (95% CI, 0.63-0.71). The change in the C statistic when each was added to the PCEs was 0.09 (95% CI, 0.06-0.13) for the coronary artery calcium score, 0.02 (95% CI, 0.00-0.04) for the polygenic risk score, and 0.10 (95% CI, 0.07-0.14) for both. Overall categorical net reclassification improvement was significant when the coronary artery calcium score (0.19; 95% CI, 0.06-0.28) but was not significant when the polygenic risk score (0.04; 95% CI, -0.05 to 0.10) was added to the PCEs. Calibration of the PCEs and models with coronary artery calcium and/or polygenic risk scores was adequate (all χ2<20). Subgroup analysis stratified by the median age demonstrated similar findings. Similar findings were observed for 10-year risk in RS and in longer-term follow-up in MESA (median, 16.0 years). Conclusions and Relevance In 2 cohorts of middle-aged to older adults from the US and the Netherlands, the coronary artery calcium score had better discrimination than the polygenic risk score for risk prediction of CHD. In addition, the coronary artery calcium score but not the polygenic risk score significantly improved risk discrimination and risk reclassification for CHD when added to traditional risk factors.
Collapse
Affiliation(s)
- Sadiya S. Khan
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Wendy S. Post
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California
| | - Jingyi Tan
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California
| | - Fang Zhu
- Department of Epidemiology Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Daniel Bos
- Department of Epidemiology Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Bahar Sedaghati-Khayat
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Jeroen van Rooij
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Aaron Aday
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Norrina B. Allen
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Maxime M. Bos
- Department of Epidemiology Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - André G. Uitterlinden
- Department of Epidemiology Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Matthew J. Budoff
- Department of Medicine, Lundquist Research Institute at Harbor-UCLA Medical Center, Torrance, California
| | - Donald M. Lloyd-Jones
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Jonathan D. Mosley
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California
| | - Philip Greenland
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Senior Editor, JAMA
| | - Maryam Kavousi
- Department of Epidemiology Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
22
|
Herbreteau L, Couturaud F, Hoffmann C, Bressollette L, Pan-Petesch B, Rio L, Lippert E, Ianotto JC. Atrial fibrillation and peripheral arterial disease define MPN patients with very high risk of thrombosis. Thromb Res 2023; 226:93-99. [PMID: 37141796 DOI: 10.1016/j.thromres.2023.04.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 03/20/2023] [Accepted: 04/25/2023] [Indexed: 05/06/2023]
Abstract
Myeloproliferative neoplasms (MPNs) are associated with a high risk of thrombotic and hemorrhagic complications, especially in elderly patients. Atrial fibrillation (AF) and peripheral arterial disease (PAD), also frequently discovered in aging patients, are associated with similar complications. We analysed the incidence and complication rates of AF and PAD in a large cohort of MPN patients. In total, 289/1113 patients (26 %) suffered at least one of these diseases as follows: 179 (16.1 %) with AF alone, 81 with PAD alone (7.3 %) and 29 (2.6 %) with both conditions. Postdiagnosis thrombotic events were observed in 31.3 % of AF patients (p = 0.002, OR = 1.80 [1.23;2.61]), 35.8 % of PAD patients (p = 0.002, OR = 2.21[1.31;3.67]) and 62.1 % of AF/PAD patients (p < 0.0001, OR = 6.47 [2.83;15.46]) compared to 20.1 % of no-AF/no-PAD patients. Postdiagnosis hemorrhagic events were also identified in 17.9 %, 16 %, 24.1 % and 10.1 % of AF, PAD, AF/PAD, and no-AF/no-PAD patients, respectively (p = 0.003). This significantly higher risk of thrombosis/bleeding was also observed in patients <60 years old. AF and PAD were significant risk factors for both thrombotic and hemorrhagic risks in multivariate analysis. We identified AF and PAD as criteria for high risk of thrombosis, hemorrhage, and death, emphasizing the interest in early detection and efficient treatment of these conditions.
Collapse
Affiliation(s)
- Laura Herbreteau
- Service d'Hématologie et d'Hémostase Clinique, Institut de Cancéro-Hématologie, CHU de Brest, Brest, France
| | - Francis Couturaud
- Département de Médecine Interne et Pneumologie, CHU de Brest, Brest, France; INSERM, Univ Brest, CHU de Brest, UMR 1304, GETBO, Brest, France
| | - Clément Hoffmann
- Service de Médecine vasculaire, CHU de Brest, Brest, France; INSERM, Univ Brest, CHU de Brest, UMR 1304, GETBO, Brest, France
| | - Luc Bressollette
- Service de Médecine vasculaire, CHU de Brest, Brest, France; INSERM, Univ Brest, CHU de Brest, UMR 1304, GETBO, Brest, France
| | - Brigitte Pan-Petesch
- Service d'Hématologie et d'Hémostase Clinique, Institut de Cancéro-Hématologie, CHU de Brest, Brest, France; INSERM, Univ Brest, CHU de Brest, UMR 1304, GETBO, Brest, France
| | - Laetitia Rio
- Service d'Hématologie et d'Hémostase Clinique, Institut de Cancéro-Hématologie, CHU de Brest, Brest, France
| | - Eric Lippert
- Laboratoire d'Hématologie, CHU de Brest, Brest, France; France Intergroupe des syndromes Myéloprolifératifs (FIM), Paris, France
| | - Jean-Christophe Ianotto
- Service d'Hématologie et d'Hémostase Clinique, Institut de Cancéro-Hématologie, CHU de Brest, Brest, France; INSERM, Univ Brest, CHU de Brest, UMR 1304, GETBO, Brest, France; France Intergroupe des syndromes Myéloprolifératifs (FIM), Paris, France.
| |
Collapse
|
23
|
Conundrum for Psoriasis and Thyroid Involvement. Int J Mol Sci 2023; 24:ijms24054894. [PMID: 36902323 PMCID: PMC10003398 DOI: 10.3390/ijms24054894] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 02/21/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Strategies concerning thyroid anomalies in patients confirmed with psoriasis, either on clinical level or molecular levels, and their genetic findings remain an open issue. Identification of the exact subgroup of individuals that are candidates to endocrine assessments is also controversial. Our purpose in this work was to overview clinical and pathogenic data concerning psoriasis and thyroid comorbidities from a dual perspective (dermatologic and endocrine). This was a narrative review of English literature between January 2016 and January 2023. We included clinically relevant, original articles with different levels of statistical evidence published on PubMed. We followed four clusters of conditions: thyroid dysfunction, autoimmunity, thyroid cancer, and subacute thyroiditis. A new piece of information in this field was the fact that psoriasis and autoimmune thyroid diseases (ATD) have been shown to be related to the immune-based side effects of modern anticancer drugs-namely, immune checkpoint inhibitors (ICP). Overall, we identified 16 confirmatory studies, but with heterogeneous data. Psoriatic arthritis had a higher risk of positive antithyroperoxidase antibodies (TPOAb) (25%) compared to cutaneous psoriasis or control. There was an increased risk of thyroid dysfunction versus control, and hypothyroidism was the most frequent type of dysfunction (subclinical rather than clinical), among thyroid anomalies correlated with >2-year disease duration, peripheral > axial and polyarticular involvement. With a few exceptions, there was a female predominance. Hormonal imbalance included, most frequently, low thyroxine (T4) and/or triiodothyronine (T3) with normal thyroid stimulating hormone (TSH), followed by high TSH (only one study had higher total T3). The highest ratio of thyroid involvement concerning dermatologic subtypes was 59% for erythrodermic psoriasis. Most studies found no correlation between thyroid anomalies and psoriasis severity. Statistically significant odds ratios were as follows: hypothyroidism: 1.34-1.38; hyperthyroidism: 1.17-1.32 (fewer studies than hypo); ATD: 1.42-2.05; Hashimoto's thyroiditis (HT): 1.47-2.09; Graves' disease: 1.26-1.38 (fewer studies than HT). A total of 8 studies had inconsistent or no correlations, while the lowest rate of thyroid involvement was 8% (uncontrolled studies). Other data included 3 studies on patients with ATD looking for psoriasis, as well as 1 study on psoriasis and thyroid cancer. ICP was shown to potentially exacerbate prior ATD and psoriasis or to induce them both de novo (5 studies). At the case report level, data showed subacute thyroiditis due to biological medication (ustekinumab, adalimumab, infliximab). Thyroid involvement in patients with psoriasis thus remained puzzling. We observed significant data that confirmed a higher risk of identifying positive antibodies and/or thyroid dysfunction, especially hypothyroidism, in these subjects. Awareness will be necessary to improve overall outcomes. The exact profile of individuals diagnosed with psoriasis who should be screened by the endocrinology team is still a matter of debate, in terms of dermatological subtype, disease duration, activity, and other synchronous (especially autoimmune) conditions.
Collapse
|
24
|
Weihs A, Chaker L, Martin TC, Braun KV, Campbell PJ, Cox SR, Fornage M, Gieger C, Grabe HJ, Grallert H, Harris SE, Kühnel B, Marioni RE, Martin NG, McCartney DL, McRae AF, Meisinger C, van Meurs JB, Nano J, Nauck M, Peters A, Prokisch H, Roden M, Selvin E, Beekman M, van Heemst D, Slagboom EP, Swenson BR, Tin A, Tsai PC, Uitterlinden A, Visser WE, Völzke H, Waldenberger M, Walsh JP, Köttgen A, Wilson SG, Peeters RP, Bell JT, Medici M, Teumer A. Epigenome-Wide Association Study Reveals CpG Sites Associated with Thyroid Function and Regulatory Effects on KLF9. Thyroid 2023; 33:301-311. [PMID: 36719767 PMCID: PMC10024591 DOI: 10.1089/thy.2022.0373] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Background: Thyroid hormones play a key role in differentiation and metabolism and are known regulators of gene expression through both genomic and epigenetic processes including DNA methylation. The aim of this study was to examine associations between thyroid hormones and DNA methylation. Methods: We carried out a fixed-effect meta-analysis of epigenome-wide association study (EWAS) of blood DNA methylation sites from 8 cohorts from the ThyroidOmics Consortium, incorporating up to 7073 participants of both European and African ancestry, implementing a discovery and replication stage. Statistical analyses were conducted using normalized beta CpG values as dependent and log-transformed thyrotropin (TSH), free thyroxine, and free triiodothyronine levels, respectively, as independent variable in a linear model. The replicated findings were correlated with gene expression levels in whole blood and tested for causal influence of TSH and free thyroxine by two-sample Mendelian randomization (MR). Results: Epigenome-wide significant associations (p-value <1.1E-7) of three CpGs for free thyroxine, five for free triiodothyronine, and two for TSH concentrations were discovered and replicated (combined p-values = 1.5E-9 to 4.3E-28). The associations included CpG sites annotated to KLF9 (cg00049440) and DOT1L (cg04173586) that overlap with all three traits, consistent with hypothalamic-pituitary-thyroid axis physiology. Significant associations were also found for CpGs in FKBP5 for free thyroxine, and at CSNK1D/LINCO1970 and LRRC8D for free triiodothyronine. MR analyses supported a causal effect of thyroid status on DNA methylation of KLF9. DNA methylation of cg00049440 in KLF9 was inversely correlated with KLF9 gene expression in blood. The CpG at CSNK1D/LINC01970 overlapped with thyroid hormone receptor alpha binding peaks in liver cells. The total additive heritability of the methylation levels of the six significant CpG sites was between 25% and 57%. Significant methylation QTLs were identified for CpGs at KLF9, FKBP5, LRRC8D, and CSNK1D/LINC01970. Conclusions: We report novel associations between TSH, thyroid hormones, and blood-based DNA methylation. This study advances our understanding of thyroid hormone action particularly related to KLF9 and serves as a proof-of-concept that integrations of EWAS with other -omics data can provide a valuable tool for unraveling thyroid hormone signaling in humans by complementing and feeding classical in vitro and animal studies.
Collapse
Affiliation(s)
- Antoine Weihs
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Layal Chaker
- Erasmus MC Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
- Erasmus MC Academic Center for Thyroid Diseases, Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Tiphaine C. Martin
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Twin Research and Genetic Epidemiology, St Thomas' Hospital Campus, King's College London, London, United Kingdom
| | - Kim V.E. Braun
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Purdey J. Campbell
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, Australia
| | - Simon R. Cox
- Lothian Birth Cohorts, Department of Psychology; Institute of Genetics and Cancer; University of Edinburgh, Edinburgh, United Kingdom
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, Houston, Texas, USA
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Christian Gieger
- Research Unit Molecular Epidemiology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
- Institute of Epidemiology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Hans J. Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- German Centre for Neurodegenerative Diseases (DZNE), Site Rostock, Greifswald, Germany
| | - Harald Grallert
- Research Unit Molecular Epidemiology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
- Institute of Epidemiology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
| | - Sarah E. Harris
- Lothian Birth Cohorts, Department of Psychology; Institute of Genetics and Cancer; University of Edinburgh, Edinburgh, United Kingdom
| | - Brigitte Kühnel
- Research Unit Molecular Epidemiology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
- Institute of Epidemiology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
| | - Riccardo E. Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer; University of Edinburgh, Edinburgh, United Kingdom
| | | | - Daniel L. McCartney
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer; University of Edinburgh, Edinburgh, United Kingdom
| | - Allan F. McRae
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Australia
| | - Christa Meisinger
- Epidemiology, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Joyce B.J. van Meurs
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Orthopeadics and Sports Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jana Nano
- Institute of Epidemiology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
- Institute for Medical Informatics, Biometrics and Epidemiology, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Annette Peters
- Research Unit Molecular Epidemiology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
- Institute of Epidemiology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Institute for Medical Informatics, Biometrics and Epidemiology, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Holger Prokisch
- Institute of Neurogenomics, Computational Health Center; Helmholtz Munich, Neuherberg, Germany
- Institute of Human Genetics, School of Medicine, Technical University Munich, Munich, Germany
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Medical Faculty; Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Division of Endocrinology and Diabetology, Medical Faculty; Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | - Elizabeth Selvin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Marian Beekman
- Section of Molecular Epidemiology, Department of Biomedical Data Sciences, Department of Internal Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Diana van Heemst
- Section of Gerontology and Geriatrics, Department of Internal Medicine; Leiden University Medical Center, Leiden, Netherlands
| | - Eline P. Slagboom
- Section of Molecular Epidemiology, Department of Biomedical Data Sciences, Department of Internal Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Brenton R. Swenson
- Cardiovascular Health Research Unit, University of Washington, Seattle, Washington, USA
| | - Adrienne Tin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Pei-Chien Tsai
- Department of Twin Research and Genetic Epidemiology, St Thomas' Hospital Campus, King's College London, London, United Kingdom
- Department of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Andre Uitterlinden
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - W. Edward Visser
- Erasmus MC Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Henry Völzke
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Institute for Community Medicine; University Medicine Greifswald, Greifswald, Germany
| | - Melanie Waldenberger
- Research Unit Molecular Epidemiology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
- Institute of Epidemiology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - John P. Walsh
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, Australia
- Medical School, University of Western Australia, Crawley, Australia
| | - Anna Köttgen
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center—University of Freiburg, Freiburg, Germany
| | - Scott G. Wilson
- Department of Twin Research and Genetic Epidemiology, St Thomas' Hospital Campus, King's College London, London, United Kingdom
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, Australia
- School of Biomedical Sciences, University of Western Australia, Perth, Australia
| | - Robin P. Peeters
- Erasmus MC Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jordana T. Bell
- Department of Twin Research and Genetic Epidemiology, St Thomas' Hospital Campus, King's College London, London, United Kingdom
| | - Marco Medici
- Erasmus MC Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Alexander Teumer
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Institute for Community Medicine; University Medicine Greifswald, Greifswald, Germany
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
25
|
Lipid-induced transcriptomic changes in blood link to lipid metabolism and allergic response. Nat Commun 2023; 14:544. [PMID: 36725846 PMCID: PMC9892529 DOI: 10.1038/s41467-022-35663-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 12/16/2022] [Indexed: 02/03/2023] Open
Abstract
Immune cell function can be altered by lipids in circulation, a process potentially relevant to lipid-associated inflammatory diseases including atherosclerosis and rheumatoid arthritis. To gain further insight in the molecular changes involved, we here perform a transcriptome-wide association analysis of blood triglycerides, HDL cholesterol, and LDL cholesterol in 3229 individuals, followed by a systematic bidirectional Mendelian randomization analysis to assess the direction of effects and control for pleiotropy. Triglycerides are found to induce transcriptional changes in 55 genes and HDL cholesterol in 5 genes. The function and cell-specific expression pattern of these genes implies that triglycerides downregulate both cellular lipid metabolism and, unexpectedly, allergic response. Indeed, a Mendelian randomization approach based on GWAS summary statistics indicates that several of these genes, including interleukin-4 (IL4) and IgE receptors (FCER1A, MS4A2), affect the incidence of allergic diseases. Our findings highlight the interplay between triglycerides and immune cells in allergic disease.
Collapse
|
26
|
Wang L, Zhao X, Zheng H, Zhu C, Liu Y. AIF-1, a potential biomarker of aggressive tumor behavior in patients with non-small cell lung cancer. PLoS One 2022; 17:e0279211. [PMID: 36520870 PMCID: PMC9754194 DOI: 10.1371/journal.pone.0279211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 12/02/2022] [Indexed: 12/23/2022] Open
Abstract
Allogeneic inflammatory factor-1 (AIF-1) overexpression has been reported to be associated with tumorigenesis and tumor metastasis. This study aimed to investigate the role of AIF-1 in the development and progression of non-small cell lung cancer (NSCLC). AIF-1, IL-6, and VEGF expressions in human NSCLC tissue were examined by immunofluorescence staining. Bioinformatics analyses were performed to identify AIF-1-related molecules and pathways in NSCLC. Human lung cancer A549 cell proliferation was assessed by CCK-8 assay, and cell migration was evaluated with wound-healing assay. IL-6 and VEGF secretions in A549 cell culture supernatants were quantified using the Elecsys IL-6 immunoassay kit and Vascular Endothelial Growth Factor Assay Kit. RT-PCR and western blot were performed to quantify the expressions of AIF-1, IL-6, and VEGF mRNAs and proteins involved in p38-MAPK and JAK/STAT3 signaling such as p-p38 and p-STAT3. The effects of AIF-1 on A549 cell proliferation and the expressions of IL-6 and VEGF were assessed using SB203580 and ruxolitinib. The results showed that AIF-1 expression was higher in human NSCLC tissue than that in paracancer tissue. High AIF-1 expression was associated with metastasis, higher TNM stage, and poorer survival. Bioinformatics connected AIF-1 to JAK/STAT signaling in NSCLC. AIF-1 increased A549 cell proliferation, migration, IL-6 secretion and, VEGF secretion, and these effects were attenuated by inhibition of p38-MAPK or JAK/STAT3 signaling. In conclusion, AIF-1 may promote aggressive NSCLC behavior via activation of p38-MAPK and JAK/STAT signaling.
Collapse
Affiliation(s)
- Lingling Wang
- Department of Laboratory Diagnosis, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Laboratory Diagnosis, The Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Xing Zhao
- Department of Pathology, The Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Huachuan Zheng
- Department of Oncology and Experimental Center, The Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Cuimin Zhu
- Department of Oncology, The Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Yanhong Liu
- Department of Laboratory Diagnosis, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- * E-mail:
| |
Collapse
|
27
|
Waqas K, Chen J, Lu T, van der Eerden BCJ, Rivadeneira F, Uitterlinden AG, Voortman T, Zillikens MC. Dietary advanced glycation end-products (dAGEs) intake and its relation to sarcopenia and frailty - The Rotterdam Study. Bone 2022; 165:116564. [PMID: 36150657 DOI: 10.1016/j.bone.2022.116564] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 07/18/2022] [Accepted: 09/19/2022] [Indexed: 11/02/2022]
Abstract
Studies on mice have shown a relationship between dietary intake of advanced glycation end-products (dAGEs) and deterioration of musculoskeletal health, but human studies are absent. We investigated the relationship between dietary intake of carboxymethyllysine (dCML) - an AGE prototype - and risk of sarcopenia at baseline and after 5 years of follow-up and a single evaluation of physical frailty in participants from the population-based Rotterdam Study. Appendicular lean mass (ALM) was obtained using insight dual-energy X-ray absorptiometry and hand grip strength (HGS) using a hydraulic hand dynamometer. Subjects with both low ALM and weak HGS were classified as having sarcopenia. Frailty (yes/no) was defined by presence of ≥3 and pre-frailty by presence of 1 or 2 components namely, exhaustion, weakness, slowness, weight loss or low physical activity. dCML was calculated using a food frequency questionnaire and dAGE databases. Logistic regression analysis was used to evaluate the odds of physical frailty and prevalent sarcopenia at baseline and follow-up and incident sarcopenia. 2782 participants with an age 66.4 ± 9.9 years and dCML intake 3.3 ± 1.3 mg/day, had data on sarcopenia at both time points. Of whom 84 had sarcopenia at baseline and 73 developed sarcopenia at follow-up. We observed an association of one SD increase in dCML intake with prevalent sarcopenia at baseline [odds ratio, OR = 1.27 (1.01-1.59)] and no association of dCML with incident sarcopenia at 5-year follow-up [OR = 1.12 (0.86-1.44)]. For frailty we analyzed 3577 participants, of whom 1972 were pre-frail and 158 were frail. We observed no association of dCML with either pre-frailty [OR = 0.99 (0.91-1.07)] or frailty [OR = 1.01 (0.83-1.22)] when non-frail subjects were used as reference. Our results show an association of dAGEs with sarcopenia cross-sectionally but not longitudinally where inconclusive findings are observed possibly due to a very low incidence of sarcopenia. There was no association with frailty cross-sectionally.
Collapse
Affiliation(s)
- Komal Waqas
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Jinluan Chen
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands; Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - T Lu
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands; Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - B C J van der Eerden
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands; Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - André G Uitterlinden
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands; Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Trudy Voortman
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands; Division of Human Nutrition & Health, Wageningen University & Research, Wageningen, the Netherlands
| | - M Carola Zillikens
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands.
| |
Collapse
|
28
|
Waqas K, Szilagyi IA, Schiphof D, Boer CG, Bierma-Zeinstra S, van Meurs JBJ, Zillikens MC. Skin autofluorescence, a non-invasive biomarker of advanced glycation end products, and its relation to radiographic and MRI based osteoarthritis. Osteoarthritis Cartilage 2022; 30:1631-1639. [PMID: 36087928 DOI: 10.1016/j.joca.2022.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 08/11/2022] [Accepted: 08/24/2022] [Indexed: 02/02/2023]
Abstract
OBJECTIVES Accumulation of advanced glycation end products (AGEs) in articular cartilage during aging has been proposed as a mechanism involved in the development of osteoarthritis (OA). Therefore, we investigated a cross-sectional relationship between skin AGEs, a biomarker for systemic AGEs accumulation, and OA. METHODS Skin AGEs were estimated with the AGE Reader™ as skin autofluorescence (SAF). Knee and hip X-rays were scored according to Kellgren and Lawrence (KL) system. KL-sum score of all four joints was calculated per participant to assess severity of overall radiographic OA (ROA) including or excluding those with prosthesis. Knee MRI of tibiofemoral joint (TFMRI) was assessed for cartilage loss. Sex-stratified regression models were performed after testing interaction with SAF. RESULTS 2,153 participants were included for this cross-sectional analysis. In women (n = 1,206) for one unit increase in SAF, the KL-sum score increased by 1.15 (95% confidence interval = 1.00-1.33) but excluding women with prosthesis, there was no KL-sum score increase [0.96 (0.83-1.11)]. SAF was associated with higher prevalence of prosthesis [Odds ratio, OR = 1.67 (1.10-2.54)] but not with ROA [OR = 0.83 (0.61-1.14)] when compared to women with no ROA. In men (n = 947), there was inconclusive association between SAF and KL sum score or prosthesis. For TFMRI (n = 103 women), SAF was associated with higher prevalence of cartilage loss, full-thickness [OR = 5.44 (1.27-23.38)] and partial-thickness [OR = 1.45 (0.38-5.54)], when compared to participants with no cartilage loss. CONCLUSION Higher SAF in women was associated with higher prosthesis prevalence and a trend towards higher cartilage loss on MRI. Our data presents inconclusive results between SAF and ROA in both sexes.
Collapse
Affiliation(s)
- K Waqas
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands.
| | - I A Szilagyi
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of General Practice, Erasmus University Medical Center, Rotterdam, the Netherlands.
| | - D Schiphof
- Department of General Practice, Erasmus University Medical Center, Rotterdam, the Netherlands.
| | - C G Boer
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands.
| | - S Bierma-Zeinstra
- Department of General Practice, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Orthopaedics & Sports Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands.
| | - J B J van Meurs
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Department of Orthopaedics & Sports Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands.
| | - M C Zillikens
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
29
|
Milić J. Aging and Neuropsychiatric Disease: A General Overview of Prevalence and Trends. Physiology (Bethesda) 2022. [DOI: 10.5772/intechopen.103102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The increasing trend of life-expectancy is becoming a significant demographic, societal and economic challenge. Currently, global number of people above sixty years of age is 900 million, while United Nations expect this number to rise to over 1.4 billion in 2030 and over 2.5 billion by 2050. Concordant to this trend, numerous physiological changes are associated with aging and brain-related ones are associated with neuropsychiatric diseases. The main goal of this chapter is to identify the most important neuropsychiatric diseases to assess in older patients to help to promote health and prevent diseases and complications associated with chronic illness, as these changes are progressive and require important psychological and setting-related social adjustments. Findings identify several health-aspects highly present in elderly: stroke, white matter lesions, dementia rise with age, changes in levels of neurotransmitters and hormones, depression as well as the bereavement following loss of the loved one, and the most common neurodegenerative disease—Alzheimer’s disease and Parkinson’s. In conclusion, studying the aging process should include all developmental, circumstantial, and individual aspects of aging. This offers opportunities to improve the health of elderly by using a wide range of skills and knowledge. Thus, further studies are necessary to elucidate what can be done do to improve the aging process and health of elderly in the future.
Collapse
|
30
|
Molsberry SA, Hughes KC, Schwarzschild MA, Ascherio A. Who to Enroll in Parkinson Disease Prevention Trials? The Case for Composite Prodromal Cohorts. Neurology 2022; 99:26-33. [PMID: 35970591 DOI: 10.1212/wnl.0000000000200788] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 04/11/2022] [Indexed: 11/15/2022] Open
Abstract
Significant progress has been made in expanding our understanding of prodromal Parkinson disease (PD), particularly for recognition of early motor and nonmotor signs and symptoms. Although identification of these prodromal features may improve our understanding of the earliest stages of PD, they are individually insufficient for early disease detection and enrollment of participants in prevention trials in most cases because of low sensitivity, specificity, and positive predictive value. Composite cohorts, composed of individuals with multiple co-occurring prodromal features, are an important resource for conducting prodromal PD research and eventual prevention trials because they are more representative of the population at risk for PD, allow investigators to evaluate the efficacy of an intervention across individuals with varying prodromal feature patterns, are able to produce larger sample sizes, and capture individuals at different stages of prodromal PD. A key challenge in identifying individuals with prodromal disease for composite cohorts and prevention trial participation is that we know little about the natural history of prodromal PD. To move toward prevention trials, it is critical that we better understand common prodromal feature patterns and be able to predict the probability of progression and phenoconversion. Ongoing research in cohort studies and administrative databases is beginning to address these questions, but further longitudinal analyses in a large population-based sample are necessary to provide a convincing and definitive strategy for identifying individuals to be enrolled in a prevention trial.
Collapse
Affiliation(s)
- Samantha A Molsberry
- From the Department of Nutrition (S.A.M., A.A.), Harvard T.H. Chan School of Public Health; Epidemiology (K.C.H.), Optum; Department of Neurology (M.A.S.), and MassGeneral Institute for Neurodegenerative Disease (M.A.S.), Massachusetts General Hospital; Department of Epidemiology (A.A.), Harvard T.H. Chan School of Public Health; and Channing Division of Network Medicine (A.A.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA.
| | - Katherine C Hughes
- From the Department of Nutrition (S.A.M., A.A.), Harvard T.H. Chan School of Public Health; Epidemiology (K.C.H.), Optum; Department of Neurology (M.A.S.), and MassGeneral Institute for Neurodegenerative Disease (M.A.S.), Massachusetts General Hospital; Department of Epidemiology (A.A.), Harvard T.H. Chan School of Public Health; and Channing Division of Network Medicine (A.A.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Michael A Schwarzschild
- From the Department of Nutrition (S.A.M., A.A.), Harvard T.H. Chan School of Public Health; Epidemiology (K.C.H.), Optum; Department of Neurology (M.A.S.), and MassGeneral Institute for Neurodegenerative Disease (M.A.S.), Massachusetts General Hospital; Department of Epidemiology (A.A.), Harvard T.H. Chan School of Public Health; and Channing Division of Network Medicine (A.A.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Alberto Ascherio
- From the Department of Nutrition (S.A.M., A.A.), Harvard T.H. Chan School of Public Health; Epidemiology (K.C.H.), Optum; Department of Neurology (M.A.S.), and MassGeneral Institute for Neurodegenerative Disease (M.A.S.), Massachusetts General Hospital; Department of Epidemiology (A.A.), Harvard T.H. Chan School of Public Health; and Channing Division of Network Medicine (A.A.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
31
|
Porcu E, Claringbould A, Weihs A, Lepik K, Richardson TG, Völker U, Santoni FA, Teumer A, Franke L, Reymond A, Kutalik Z. Limited evidence for blood eQTLs in human sexual dimorphism. Genome Med 2022; 14:89. [PMID: 35953856 PMCID: PMC9373355 DOI: 10.1186/s13073-022-01088-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 07/14/2022] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND The genetic underpinning of sexual dimorphism is very poorly understood. The prevalence of many diseases differs between men and women, which could be in part caused by sex-specific genetic effects. Nevertheless, only a few published genome-wide association studies (GWAS) were performed separately in each sex. The reported enrichment of expression quantitative trait loci (eQTLs) among GWAS-associated SNPs suggests a potential role of sex-specific eQTLs in the sex-specific genetic mechanism underlying complex traits. METHODS To explore this scenario, we combined sex-specific whole blood RNA-seq eQTL data from 3447 European individuals included in BIOS Consortium and GWAS data from UK Biobank. Next, to test the presence of sex-biased causal effect of gene expression on complex traits, we performed sex-specific transcriptome-wide Mendelian randomization (TWMR) analyses on the two most sexually dimorphic traits, waist-to-hip ratio (WHR) and testosterone levels. Finally, we performed power analysis to calculate the GWAS sample size needed to observe sex-specific trait associations driven by sex-biased eQTLs. RESULTS Among 9 million SNP-gene pairs showing sex-combined associations, we found 18 genes with significant sex-biased cis-eQTLs (FDR 5%). Our phenome-wide association study of the 18 top sex-biased eQTLs on >700 traits unraveled that these eQTLs do not systematically translate into detectable sex-biased trait-associations. In addition, we observed that sex-specific causal effects of gene expression on complex traits are not driven by sex-specific eQTLs. Power analyses using real eQTL- and causal-effect sizes showed that millions of samples would be necessary to observe sex-biased trait associations that are fully driven by sex-biased cis-eQTLs. Compensatory effects may further hamper their detection. CONCLUSIONS Our results suggest that sex-specific eQTLs in whole blood do not translate to detectable sex-specific trait associations of complex diseases, and vice versa that the observed sex-specific trait associations cannot be explained by sex-specific eQTLs.
Collapse
Affiliation(s)
- Eleonora Porcu
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland. .,Swiss Institute of Bioinformatics, Lausanne, Switzerland. .,University Center for Primary Care and Public Health, Lausanne, Switzerland.
| | - Annique Claringbould
- University Medical Centre Groningen, Groningen, the Netherlands.,Structural and Computational Biology Unit, European Molecular Biology Laboratories (EMBL), Heidelberg, Germany
| | - Antoine Weihs
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Kaido Lepik
- Institute of Computer Science, University of Tartu, Tartu, Estonia.,Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | | | - Tom G Richardson
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK.,Novo Nordisk Research Centre Oxford, Old Road Campus, Oxford, OX3 7DQ, UK
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
| | - Federico A Santoni
- Endocrine, Diabetes, and Metabolism Service, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland.,Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Alexander Teumer
- DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany.,Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Lude Franke
- University Medical Centre Groningen, Groningen, the Netherlands
| | - Alexandre Reymond
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland.
| | - Zoltán Kutalik
- Swiss Institute of Bioinformatics, Lausanne, Switzerland. .,University Center for Primary Care and Public Health, Lausanne, Switzerland. .,Department of Computational Biology, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
32
|
Campos-Obando N, Bosman A, Kavousi M, Medina-Gomez C, van der Eerden BCJ, Bos D, Franco OH, Uitterlinden AG, Zillikens MC. Genetic Evidence for a Causal Role of Serum Phosphate in Coronary Artery Calcification: The Rotterdam Study. J Am Heart Assoc 2022; 11:e023024. [PMID: 35904204 PMCID: PMC9375490 DOI: 10.1161/jaha.121.023024] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Background Hyperphosphatemia has been associated with coronary artery calcification (CAC) mostly in chronic kidney disease, but the association between phosphate levels within the normal phosphate range and CAC is unclear. Our objectives were to evaluate associations between phosphate levels and CAC among men and women from the general population and assess causality through Mendelian randomization. Methods and Results CAC, measured by electron‐beam computed tomography, and serum phosphate levels were assessed in 1889 individuals from the RS (Rotterdam Study). Phenotypic associations were tested through linear models adjusted for age, body mass index, blood pressure, smoking, prevalent cardiovascular disease and diabetes, 25‐hydroxyvitamin D, total calcium, C‐reactive protein, glucose, and total cholesterol : high‐density lipoprotein cholesterol ratio. Mendelian randomization was implemented through an allele score including 8 phosphate‐related single‐nucleotide polymorphisms. In phenotypic analyses, serum phosphate (per 1 SD) was associated with CAC with evidence for sex interaction (Pinteraction=0.003) (men β, 0.44 [95% CI, 0.30–0.59]; P=3×10−9; n=878; women β, 0.24 [95% CI, 0.08–0.40]; P=0.003; n=1011). Exclusion of hyperphosphatemia, chronic kidney disease (estimated glomerular filtration rate <60 mL/min per 1.73 m2) and prevalent cardiovascular disease yielded similar results. In Mendelian randomization analyses, instrumented phosphate was associated with CAC (total population β, 0.93 [95% CI: 0.07–1.79]; P=0.034; n=1693), even after exclusion of hyperphosphatemia, chronic kidney disease and prevalent cardiovascular disease (total population β, 1.23 [95% CI, 0.17–2.28]; P=0.023; n=1224). Conclusions Serum phosphate was associated with CAC in the general population with stronger effects in men. Mendelian randomization findings support a causal relation, also for serum phosphate and CAC in subjects without hyperphosphatemia, chronic kidney disease, and cardiovascular disease. Further research into underlying mechanisms of this association and sex differences is needed.
Collapse
Affiliation(s)
- Natalia Campos-Obando
- Department of Internal Medicine Erasmus MC, University Medical Center Rotterdam Rotterdam the Netherlands
| | - Ariadne Bosman
- Department of Internal Medicine Erasmus MC, University Medical Center Rotterdam Rotterdam the Netherlands
| | - Maryam Kavousi
- Department of Epidemiology Erasmus MC, University Medical Center Rotterdam Rotterdam the Netherlands
| | - Carolina Medina-Gomez
- Department of Internal Medicine Erasmus MC, University Medical Center Rotterdam Rotterdam the Netherlands.,Department of Epidemiology Erasmus MC, University Medical Center Rotterdam Rotterdam the Netherlands
| | - Bram C J van der Eerden
- Department of Internal Medicine Erasmus MC, University Medical Center Rotterdam Rotterdam the Netherlands
| | - Daniel Bos
- Department of Epidemiology Erasmus MC, University Medical Center Rotterdam Rotterdam the Netherlands.,Department of Radiology and Nuclear Medicine Erasmus MC, University Medical Center Rotterdam Rotterdam the Netherlands
| | - Oscar H Franco
- Department of Epidemiology Erasmus MC, University Medical Center Rotterdam Rotterdam the Netherlands.,Institute of Social and Preventive Medicine (ISPM) University of Bern Switzerland
| | - André G Uitterlinden
- Department of Internal Medicine Erasmus MC, University Medical Center Rotterdam Rotterdam the Netherlands.,Department of Epidemiology Erasmus MC, University Medical Center Rotterdam Rotterdam the Netherlands
| | - M Carola Zillikens
- Department of Internal Medicine Erasmus MC, University Medical Center Rotterdam Rotterdam the Netherlands.,Department of Epidemiology Erasmus MC, University Medical Center Rotterdam Rotterdam the Netherlands
| |
Collapse
|
33
|
Lommatzsch C, Rothaus K, Schopmeyer L, Feldmann M, Bauer D, Grisanti S, Heinz C, Kasper M. Elevated endothelin-1 levels as risk factor for an impaired ocular blood flow measured by OCT-A in glaucoma. Sci Rep 2022; 12:11801. [PMID: 35821224 PMCID: PMC9276731 DOI: 10.1038/s41598-022-15401-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 06/23/2022] [Indexed: 11/28/2022] Open
Abstract
The purpose of this study was to ascertain whether a correlation exists between glaucoma-associated alteration of ocular vascular haemodynamics and endothelin-1 (ET-1) levels exist. Eyes of patients with cataract (n = 30) or glaucoma (n = 68) were examined with optical coherence tomography (OCT) and OCT-angiography (OCT-A; AngioVue™-RTVue-XR; Optovue, Fremont, California, USA). The peripapillary and the macular vessel density (VD) values were measured. Inferior and superior retinal nerve fibre layer (RNFL) thickness loss was used for further OCT staging. Aqueous humour of the examined eye and plasma were sampled during cataract or glaucoma surgery and analysed by means of ELISA to determine their ET-1 level. Glaucoma eyes are characterised by reductions in RNFL thickness and VD that correlate significantly with the OCT GSS score. Peripheral and ocular ET-1 level were significantly elevated in patients with glaucoma and correlate positively with the OCT-GSS score of the entire study population. Peripapillary and macula VD of glaucoma patients correlates negatively with plasma ET-1 levels. Multivariable analysis showed a subordinate role of intraocular pressure predictive factor for impaired retinal blood flow compared with plasma ET-1 level in glaucoma. Peripheral ET-1 level serves as risk factor for detection of ocular blood flow changes in the optic nerve head region of glaucomatous eyes.
Collapse
Affiliation(s)
- Claudia Lommatzsch
- Department of Ophthalmology and Ophtha Lab at St. Franziskus Hospital, Hohenzollernring 74, 48145, Muenster, Germany. .,Department of Ophthalmology, University of Luebeck, Luebeck, Germany.
| | - Kai Rothaus
- Department of Ophthalmology and Ophtha Lab at St. Franziskus Hospital, Hohenzollernring 74, 48145, Muenster, Germany
| | | | - Maria Feldmann
- Department of Ophthalmology, Braunschweig Hospital, Braunschweig, Germany
| | - Dirk Bauer
- Department of Ophthalmology and Ophtha Lab at St. Franziskus Hospital, Hohenzollernring 74, 48145, Muenster, Germany
| | - Swaantje Grisanti
- Department of Ophthalmology, University of Luebeck, Luebeck, Germany
| | - Carsten Heinz
- Department of Ophthalmology and Ophtha Lab at St. Franziskus Hospital, Hohenzollernring 74, 48145, Muenster, Germany.,Department of Ophthalmology, University of Essen, Essen, Germany
| | - Maren Kasper
- Department of Ophthalmology and Ophtha Lab at St. Franziskus Hospital, Hohenzollernring 74, 48145, Muenster, Germany
| |
Collapse
|
34
|
Sedaghati-Khayat B, Boer CG, Runhaar J, Bierma-Zeinstra SMA, Broer L, Ikram MA, Zeggini E, Uitterlinden AG, van Rooij JGJ, van Meurs JBJ. Risk assessment for hip and knee osteoarthritis using polygenic risk scores. Arthritis Rheumatol 2022; 74:1488-1496. [PMID: 35644035 PMCID: PMC9541521 DOI: 10.1002/art.42246] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 04/24/2022] [Accepted: 05/24/2022] [Indexed: 11/25/2022]
Abstract
Objective Polygenic risk scores (PRS) allow risk stratification using common single‐nucleotide polymorphisms (SNPs), and clinical applications are currently explored for several diseases. This study was undertaken to assess the risk of hip and knee osteoarthritis (OA) using PRS. Methods We analyzed 12,732 individuals from a population‐based cohort from the Rotterdam Study (n = 11,496), a clinical cohort (Cohort Hip and Cohort Knee [CHECK] study; n = 908), and a high‐risk cohort of overweight women (Prevention of Knee OA in Overweight Females [PROOF] study; n = 328), for the association of the PRS with prevalence/incidence of radiographic OA, of clinical OA, and of total hip replacement (THR) or total knee replacement (TKR). The hip PRS and knee PRS contained 44 and 24 independent SNPs, respectively, and were derived from a recent genome‐wide association study meta‐analysis. Standardized PRS (with Z transformation) were used in all analyses. Results We found a stronger association of the PRS for clinically defined OA compared to radiographic OA phenotypes, and we observed the highest PRS risk stratification for TKR/THR. The odds ratio (OR) per SD was 1.3 for incident THR (95% confidence interval [95% CI] 1.1–1.5) and 1.6 (95% CI 1.3–1.9) for incident TKR in the Rotterdam Study. The knee PRS was associated with incident clinical knee OA in the CHECK study (OR 1.3 [95% CI 1.1–1.5]), but not for the PROOF study (OR 1.2 [95% CI 0.8–1.7]). The OR for OA increased gradually across the PRS distribution, up to 2.1 (95% CI 1.4–3.2) for individuals with the 10% highest PRS compared to the middle 50% of the PRS distribution. Conclusion Our findings validated the association of PRS across OA definitions. Since OA is becoming frequent and primary prevention is not commonly applicable, PRS‐based risk assessment could play a role in OA prevention. However, the utility of PRS is dependent on the setting. Further studies are needed to test the integration of genetic risk assessment in diverse health care settings.
Collapse
Affiliation(s)
- Bahar Sedaghati-Khayat
- Department of Internal medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Cindy G Boer
- Department of Internal medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jos Runhaar
- Department of General Practice, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Sita M A Bierma-Zeinstra
- Department of General Practice, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Orthopaedics & Sports Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Linda Broer
- Department of Internal medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Eleftheria Zeggini
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.,Technical University of Munich (TUM) and Klinikum Rechts der Isar, TUM School of Medicine, Munich, Germany
| | - André G Uitterlinden
- Department of Internal medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jeroen G J van Rooij
- Department of Internal medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Joyce B J van Meurs
- Department of Internal medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
35
|
Castaneda AB, Petty LE, Scholz M, Jansen R, Weiss S, Zhang X, Schramm K, Beutner F, Kirsten H, Schminke U, Hwang SJ, Marzi C, Dhana K, Seldenrijk A, Krohn K, Homuth G, Wolf P, Peters MJ, Dörr M, Peters A, van Meurs JBJ, Uitterlinden AG, Kavousi M, Levy D, Herder C, van Grootheest G, Waldenberger M, Meisinger C, Rathmann W, Thiery J, Polak J, Koenig W, Seissler J, Bis JC, Franceshini N, Giambartolomei C, Hofman A, Franco OH, Penninx BWJH, Prokisch H, Völzke H, Loeffler M, O'Donnell CJ, Below JE, Dehghan A, de Vries PS. Associations of carotid intima media thickness with gene expression in whole blood and genetically predicted gene expression across 48 tissues. Hum Mol Genet 2022; 31:1171-1182. [PMID: 34788810 PMCID: PMC8976428 DOI: 10.1093/hmg/ddab236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 06/11/2021] [Accepted: 08/11/2021] [Indexed: 11/13/2022] Open
Abstract
Carotid intima media thickness (cIMT) is a biomarker of subclinical atherosclerosis and a predictor of future cardiovascular events. Identifying associations between gene expression levels and cIMT may provide insight to atherosclerosis etiology. Here, we use two approaches to identify associations between mRNA levels and cIMT: differential gene expression analysis in whole blood and S-PrediXcan. We used microarrays to measure genome-wide whole blood mRNA levels of 5647 European individuals from four studies. We examined the association of mRNA levels with cIMT adjusted for various potential confounders. Significant associations were tested for replication in three studies totaling 3943 participants. Next, we applied S-PrediXcan to summary statistics from a cIMT genome-wide association study (GWAS) of 71 128 individuals to estimate the association between genetically determined mRNA levels and cIMT and replicated these analyses using S-PrediXcan on an independent GWAS on cIMT that included 22 179 individuals from the UK Biobank. mRNA levels of TNFAIP3, CEBPD and METRNL were inversely associated with cIMT, but these associations were not significant in the replication analysis. S-PrediXcan identified associations between cIMT and genetically determined mRNA levels for 36 genes, of which six were significant in the replication analysis, including TLN2, which had not been previously reported for cIMT. There was weak correlation between our results using differential gene expression analysis and S-PrediXcan. Differential expression analysis and S-PrediXcan represent complementary approaches for the discovery of associations between phenotypes and gene expression. Using these approaches, we prioritize TNFAIP3, CEBPD, METRNL and TLN2 as new candidate genes whose differential expression might modulate cIMT.
Collapse
Affiliation(s)
- Andy B Castaneda
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Lauren E Petty
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany.,LIFE Research Center of Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Rick Jansen
- Department of Psychiatry, VU University Medical Center, Amsterdam, the Netherlands
| | - Stefan Weiss
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
| | - Xiaoling Zhang
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA.,Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA.,The Framingham Heart Study, Framingham, MA, USA
| | - Katharina Schramm
- Institute of Neurogenomics, Helmholz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.,Institute of Human Genetics, Technical University Munich, Munich, Germany
| | | | - Holger Kirsten
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany.,LIFE Research Center of Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Ulf Schminke
- Department of Neurology, University Medicine Greifswald, Greifswald, Germany
| | - Shih-Jen Hwang
- The Framingham Heart Study, Framingham, MA, USA.,Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, Bethesda, MD, USA
| | - Carola Marzi
- Institute of Epidemiology, Helmholz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Klodian Dhana
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Adrie Seldenrijk
- Department of Psychiatry, VU University Medical Center, Amsterdam, the Netherlands
| | - Knut Krohn
- Interdisciplinary Center of Clinical Research, University of Leipzig, Leipzig, Germany
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Petra Wolf
- Institute of Neurogenomics, Helmholz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.,Institute of Human Genetics, Technical University Munich, Munich, Germany
| | - Marjolein J Peters
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Marcus Dörr
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany.,Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Joyce B J van Meurs
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - André G Uitterlinden
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Daniel Levy
- The Framingham Heart Study, Framingham, MA, USA.,Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, Bethesda, MD, USA
| | - Christian Herder
- Institute of Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany.,German Center for Diabetes Research (DZD e.V.), München-Neuherberg, Germany.,Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | | | - Melanie Waldenberger
- Institute of Epidemiology, Helmholz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Christa Meisinger
- Institute of Epidemiology, Helmholz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.,Chair of Epidemiology, Ludwig-Maximilians-Universität München, UNIKA-T Augsburg, Augsburg, Germany
| | - Wolfgang Rathmann
- Institute of Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Joachim Thiery
- LIFE Research Center of Civilization Diseases, University of Leipzig, Leipzig, Germany.,Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig, Leipzig, Germany
| | - Joseph Polak
- Tufts University School of Medicine, Boston, MA, USA
| | - Wolfgang Koenig
- Deutsches Herzzentrum München, Technische Universität München, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.,Department of Internal Medicine II-Cardiology, University of Ulm Medical Center, Ulm, Germany
| | - Jochen Seissler
- Diabetes Center, Diabetes Research Group, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität, Munich, Germany
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Nora Franceshini
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | | | | | - Albert Hofman
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Oscar H Franco
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands.,Institute of Social and Preventive Medicine, University of Bern, Switzerland
| | - Brenda W J H Penninx
- Department of Psychiatry, VU University Medical Center, Amsterdam, the Netherlands
| | - Holger Prokisch
- Institute of Neurogenomics, Helmholz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.,Institute of Human Genetics, Technical University Munich, Munich, Germany
| | - Henry Völzke
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany.,Institute of Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Markus Loeffler
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany.,LIFE Research Center of Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Christopher J O'Donnell
- The Framingham Heart Study, Framingham, MA, USA.,Cardiology Section, Department of Medicine, Boston Veteran's Administration Healthcare and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jennifer E Below
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Abbas Dehghan
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Epidemiology and Biostatistics, Imperial College London, London, UK.,MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, Norfolk Place, London, UK.,UK Dementia Research Institute at Imperial College London, Burlington Danes Building, Hammersmith Hospital, Du Cane Road, London W12 0NN UK
| | - Paul S de Vries
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA.,Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
36
|
Liepelt-Scarfone I, Ophey A, Kalbe E. Cognition in prodromal Parkinson's disease. PROGRESS IN BRAIN RESEARCH 2022; 269:93-111. [PMID: 35248208 DOI: 10.1016/bs.pbr.2022.01.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
One characteristic of Parkinson's disease (PD) is a prodromal phase, lasting many years during which both pre-clinical motor and non-motor symptoms occur. Around one-fifth of patients with PD manifest mild cognitive impairment at time of clinical diagnosis. Thus, important challenges are to define the time of onset of cognitive dysfunction in the prodromal phase of PD, and to define its co-occurrence with other specific characteristics. Evidence for cognitive change in prodromal PD comes from various study designs, including both longitudinal and cross-sectional approaches with different target groups. These studies support the concept that changes in global cognitive function and alterations in executive functions occur, and that these changes may be present up to 6 years before clinical PD diagnosis. Notably, this evidence led to including global cognitive impairment as an independent prodromal marker in the recently updated research criteria of the Movement Disorder Society for prodromal PD. Knowledge in this field, however, is still at its beginning, and evidence is sparse about many aspects of this topic. Further longitudinal studies including standardized assessments of global and domain-specific cognitive functions are needed to gain further knowledge about the first appearance, the course, and the interaction of cognitive deficits with other non-motor symptoms in prodromal stage PD. Treatment approaches, including non-pharmacological interventions, in individuals with prodromal PD might help to prevent or delay cognitive dysfunction in early PD.
Collapse
Affiliation(s)
- Inga Liepelt-Scarfone
- German Center for Neurodegenerative Diseases (DZNE) and Hertie Institute for Clinical Brain Research, Department of Neurodegenerative Diseases, University of Tübingen, Tübingen, Germany; IB-Hochschule, Stuttgart, Germany.
| | - Anja Ophey
- Medical Psychology, Neuropsychology and Gender Studies, Center for Neuropsychological Diagnostics and Intervention (CeNDI), University Hospital Cologne and Medical Faculty of the University of Cologne, Cologne, Germany
| | - Elke Kalbe
- Medical Psychology, Neuropsychology and Gender Studies, Center for Neuropsychological Diagnostics and Intervention (CeNDI), University Hospital Cologne and Medical Faculty of the University of Cologne, Cologne, Germany
| |
Collapse
|
37
|
Azhar AZ, Rai D, Bandyopadhyay D, Rzechorzek W, Akhtar T, Aronow WS, Ranjan P. Use of coronary artery calcium and coronary tomography angiography in the evaluation of ischemic heart disease. Hosp Pract (1995) 2022; 50:9-16. [PMID: 35037541 DOI: 10.1080/21548331.2022.2030630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 01/14/2022] [Indexed: 10/19/2022]
Abstract
Over the years, significant technological advances have been made in the field of cardiac CT imaging which has led to the widespread use of the modality in the evaluation of ischemic and structural heart disease. The advent of newer scanning techniques has led to a reduction in scanning time as well as a reduction in the radiation and contrast media dose required - making these scans both convenient and safer to perform. Research has shown that coronary CT angiography has a high negative predictive value in the evaluation of patients with coronary artery disease. There is more recent evidence that coronary CTA has a positive impact on clinical outcomes as well. In this review article, we discuss the clinical applications of coronary CTA in the evaluation of patients with stable ischemic heart disease, the most recent studies evaluating the efficacy and limitations of the modality, the role of coronary calcium in cardiovascular risk prediction in asymptomatic patients and the future applications of the modality.
Collapse
Affiliation(s)
| | - Devesh Rai
- Department of Cardiology, Rochester General Hospital, Rochester, NY, USA
| | | | - Wojciech Rzechorzek
- Department of Cardiology, Westchester Medical Center at New York Medical College, Valhalla, NY, USA
| | - Tauseef Akhtar
- Medicine, John's Hopkins University School of Medicine, Baltimore, MD, USA
| | - Wilbert S Aronow
- Department of Cardiology, Westchester Medical Center at New York Medical College, Valhalla, NY, USA
| | - Pragya Ranjan
- Department of Cardiology, Westchester Medical Center at New York Medical College, Valhalla, NY, USA
| |
Collapse
|
38
|
Waqas K, Chen J, Rivadeneira F, Uitterlinden AG, Voortman T, Zillikens MC. Skin autofluorescence, a non-invasive biomarker of advanced glycation end-products (AGEs), is associated with frailty: The Rotterdam study. J Gerontol A Biol Sci Med Sci 2022; 77:2032-2039. [PMID: 35099530 PMCID: PMC9536452 DOI: 10.1093/gerona/glac025] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Indexed: 11/19/2022] Open
Abstract
Background Accumulation of advanced glycation end-products (AGEs) in tissues has been linked to various age-related disease phenotypes. Therefore, we investigated the potential relationship between skin AGE accumulation and frailty. Methods A cross-sectional analysis was performed on 2 521 participants from the Rotterdam Study. Skin AGEs were assessed as skin autofluorescence (SAF) using the AGE reader™. We used 2 approaches to define frailty. Fried’s criteria, including weight loss, weakness, slow gait speed, exhaustion, and low physical activity, were used to define physical frailty (presence of ≥3 components) and prefrailty (presence of ≤2 components). Rockwood’s concept, including 38 deficits from physical and psychosocial health domains, was used to calculate the frailty index (score 0–1). Multinomial logistic and multivariate linear regression were used with SAF as exposure and physical frailty (ordinal) and frailty index (continuous) as outcome adjusting for age, sex, diabetes, renal function, socioeconomic status, and smoking status. Results The mean SAF was 2.39 ± 0.49 arbitrary units and the median age was 74.2 (14.0) years. Regarding physical frailty, 96 persons (4%) were frail and 1 221 (48%) were prefrail. Skin autofluorescence was associated with both being prefrail (odds ratio [95% confidence interval] = 1.29 [1.07–1.56]) and frail (1.87 [1.20–2.90]) compared with nonfrail. Regarding the frailty index, the median value was 0.14 (0.10–0.19) and higher SAF was also associated with a higher frailty index (coefficient, B = 0.017 (0.011–0.023]). Conclusions Higher skin AGEs are associated with both physical frailty and frailty index. Longitudinal studies are needed to evaluate the causality and the potential of SAF as a biomarker to screen frailty.
Collapse
Affiliation(s)
- Komal Waqas
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jinluan Chen
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - André G Uitterlinden
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands.,Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Trudy Voortman
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands.,Division of Human Nutrition & Health, Wageningen University & Research, Wageningen, the Netherlands
| | - M Carola Zillikens
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
39
|
Waqas K, Chen J, Trajanoska K, Ikram MA, Uitterlinden AG, Rivadeneira F, Zillikens MC. Skin Autofluorescence, a Noninvasive Biomarker for Advanced Glycation End-products, Is Associated With Sarcopenia. J Clin Endocrinol Metab 2022; 107:e793-e803. [PMID: 34453164 PMCID: PMC8764216 DOI: 10.1210/clinem/dgab632] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Indexed: 01/23/2023]
Abstract
BACKGROUND Accumulation of advanced glycation end-products (AGEs) in skeletal muscle has been implicated in development of sarcopenia. AIM To obtain further insight in the pathophysiology of sarcopenia, we studied its relationship with skin AGEs in the general population. METHODS In a cross-sectional analysis, 2744 participants of northern European background, mean age 74.1 years, were included from the Rotterdam Study. Skin AGEs were measured as skin autofluorescence (SAF) using AGE ReaderTM, appendicular skeletal mass index (ASMI) using insight dual-energy X-ray absorptiometry, hand grip strength (HGS) using a hydraulic hand dynamometer, and, in a subgroup, gait speed (GS) measured on an electronic walkway (n = 2080). We defined probable sarcopenia (low HGS) and confirmed sarcopenia (low HGS and low ASMI) based on the European Working Group on Sarcopenia in Older People (EWGSOP2) revised criteria cutoffs. Multivariate linear and logistic regression were performed adjusting for age, sex, body fat percentage, height, renal function, diabetes, and smoking status. RESULTS The prevalence of low ASMI was 7.7%; probable sarcopenia, 24%, slow GS, 3%; and confirmed sarcopenia, 3.5%. SAF was inversely associated with ASMI [β -0.062 (95% CI -0.092, -0.032)], HGS [β -0.051 (95% CI -0.075, -0.026)], and GS [β -0.074 (95% CI -0.116, -0.033)]. A 1-unit increase in SAF was associated with higher odds of probable sarcopenia [odds ratio (OR) 1.36 (95% CI 1.09, 1.68)] and confirmed sarcopenia [OR 2.01 (95% CI 1.33, 3.06)]. CONCLUSION Higher skin AGEs are associated with higher sarcopenia prevalence. We call for future longitudinal studies to explore the role of SAF as a potential biomarker of sarcopenia.
Collapse
Affiliation(s)
- Komal Waqas
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jinluan Chen
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Katerina Trajanoska
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - André G Uitterlinden
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - M Carola Zillikens
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
- Correspondence: M.C. Zillikens, MD, PhD, Department of Internal Medicine, Erasmus University Medical Center, ‘s-Gravendijkwal 230, 3015CE, Rotterdam, The Netherlands.
| |
Collapse
|
40
|
Jonkman TH, Dekkers KF, Slieker RC, Grant CD, Ikram MA, van Greevenbroek MMJ, Franke L, Veldink JH, Boomsma DI, Slagboom PE, Consortium BIOS, Heijmans BT. Functional genomics analysis identifies T and NK cell activation as a driver of epigenetic clock progression. Genome Biol 2022; 23:24. [PMID: 35031073 PMCID: PMC8759260 DOI: 10.1186/s13059-021-02585-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 12/20/2021] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Epigenetic clocks use DNA methylation (DNAm) levels of specific sets of CpG dinucleotides to accurately predict individual chronological age. A popular application of these clocks is to explore whether the deviation of predicted age from chronological age is associated with disease phenotypes, where this deviation is interpreted as a potential biomarker of biological age. This wide application, however, contrasts with the limited insight in the processes that may drive the running of epigenetic clocks. RESULTS We perform a functional genomics analysis on four epigenetic clocks, including Hannum's blood predictor and Horvath's multi-tissue predictor, using blood DNA methylome and transcriptome data from 3132 individuals. The four clocks result in similar predictions of individual chronological age, and their constituting CpGs are correlated in DNAm level and are enriched for similar histone modifications and chromatin states. Interestingly, DNAm levels of CpGs from the clocks are commonly associated with gene expression in trans. The gene sets involved are highly overlapping and enriched for T cell processes. Further analysis of the transcriptome and methylome of sorted blood cell types identifies differences in DNAm between naive and activated T and NK cells as a probable contributor to the clocks. Indeed, within the same donor, the four epigenetic clocks predict naive cells to be up to 40 years younger than activated cells. CONCLUSIONS The ability of epigenetic clocks to predict chronological age involves their ability to detect changes in proportions of naive and activated immune blood cells, an established feature of immuno-senescence. This finding may contribute to the interpretation of associations between clock-derived measures and age-related health outcomes.
Collapse
Affiliation(s)
- Thomas H Jonkman
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Einthovenweg 20, 2333 ZC, Leiden, The Netherlands
| | - Koen F Dekkers
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Einthovenweg 20, 2333 ZC, Leiden, The Netherlands
| | - Roderick C Slieker
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC, Leiden, The Netherlands
- Department of Epidemiology and Biostatistics, Amsterdam Public Health Institute, Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Crystal D Grant
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Einthovenweg 20, 2333 ZC, Leiden, The Netherlands
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus Medical Center, Doctor Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Marleen M J van Greevenbroek
- Department of Internal Medicine and School for Cardiovascular Diseases, Maastricht University Medical Center, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| | - Lude Franke
- Department of Genetics, University Medical Centre Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Jan H Veldink
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | - Dorret I Boomsma
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Van der Boechorststraat 1, 1081 BT, Amsterdam, The Netherlands
| | - P Eline Slagboom
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Einthovenweg 20, 2333 ZC, Leiden, The Netherlands
| | | | - Bastiaan T Heijmans
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Einthovenweg 20, 2333 ZC, Leiden, The Netherlands.
| |
Collapse
|
41
|
Ligthart S, Hasbani NR, Ahmadizar F, van Herpt TTW, Leening MJG, Uitterlinden AG, Sijbrands EJG, Morrison AC, Boerwinkle E, Pankow JS, Selvin E, Ikram MA, Kavousi M, de Vries PS, Dehghan A. Genetic susceptibility, obesity and lifetime risk of type 2 diabetes: The ARIC study and Rotterdam Study. Diabet Med 2021; 38:e14639. [PMID: 34245042 PMCID: PMC8429251 DOI: 10.1111/dme.14639] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/02/2021] [Accepted: 05/17/2021] [Indexed: 12/26/2022]
Abstract
AIMS Both lifestyle factors and genetic background contribute to the development of type 2 diabetes. Estimation of the lifetime risk of diabetes based on genetic information has not been presented, and the extent to which a normal body weight can offset a high lifetime genetic risk is unknown. METHODS We used data from 15,671 diabetes-free participants of European ancestry aged 45 years and older from the prospective population-based ARIC study and Rotterdam Study (RS). We quantified the remaining lifetime risk of diabetes stratified by genetic risk and quantified the effect of normal weight in terms of relative and lifetime risks in low, intermediate and high genetic risk. RESULTS At age 45 years, the lifetime risk of type 2 diabetes in ARIC in the low, intermediate and high genetic risk category was 33.2%, 41.3% and 47.2%, and in RS 22.8%, 30.6% and 35.5% respectively. The absolute lifetime risk for individuals with normal weight compared to individuals with obesity was 24% lower in ARIC and 8.6% lower in RS in the low genetic risk group, 36.3% lower in ARIC and 31.3% lower in RS in the intermediate genetic risk group, and 25.0% lower in ARIC and 29.4% lower in RS in the high genetic risk group. CONCLUSIONS Genetic variants for type 2 diabetes have value in estimating the lifetime risk of type 2 diabetes. Normal weight mitigates partly the deleterious effect of high genetic risk.
Collapse
Affiliation(s)
- Symen Ligthart
- Department of EpidemiologyErasmus MC ‐ University Medical Center RotterdamRotterdamthe Netherlands
- Department of Adult Intensive CareErasmus MC ‐ University Medical Center RotterdamRotterdamthe Netherlands
| | - Natalie R. Hasbani
- Human Genetics CenterDepartment of EpidemiologyHuman Genetics, and Environmental SciencesSchool of Public HealthThe University of Texas Health Science Center at HoustonHoustonTXUSA
| | - Fariba Ahmadizar
- Department of EpidemiologyErasmus MC ‐ University Medical Center RotterdamRotterdamthe Netherlands
| | - Thijs T. W. van Herpt
- Department of EpidemiologyErasmus MC ‐ University Medical Center RotterdamRotterdamthe Netherlands
- Department of Internal MedicineErasmus MC ‐ University Medical Center RotterdamRotterdamthe Netherlands
| | - Maarten J. G. Leening
- Department of EpidemiologyErasmus MC ‐ University Medical Center RotterdamRotterdamthe Netherlands
- Department of CardiologyErasmus MC ‐ University Medical Center RotterdamRotterdamthe Netherlands
- Department of EpidemiologyHarvard T.H. Chan School of Public HealthBostonMAUSA
| | - André G. Uitterlinden
- Department of Internal MedicineErasmus MC ‐ University Medical Center RotterdamRotterdamthe Netherlands
| | - Eric J. G. Sijbrands
- Department of Internal MedicineErasmus MC ‐ University Medical Center RotterdamRotterdamthe Netherlands
| | - Alanna C. Morrison
- Human Genetics CenterDepartment of EpidemiologyHuman Genetics, and Environmental SciencesSchool of Public HealthThe University of Texas Health Science Center at HoustonHoustonTXUSA
| | - Eric Boerwinkle
- Human Genetics CenterDepartment of EpidemiologyHuman Genetics, and Environmental SciencesSchool of Public HealthThe University of Texas Health Science Center at HoustonHoustonTXUSA
- Human Genome Sequencing CenterBaylor College of MedicineHoustonTXUSA
| | - James S. Pankow
- Division of Epidemiology and Community HealthSchool of Public HealthUniversity of MinnesotaMinneapolisMNUSA
| | - Elizabeth Selvin
- Department of EpidemiologyBloomberg School of Public HealthJohns Hopkins UniversityBaltimoreMDUSA
- Welch Center for Prevention, Epidemiology and Clinical ResearchJohns Hopkins UniversityBaltimoreMDUSA
| | - M. Arfan Ikram
- Department of EpidemiologyErasmus MC ‐ University Medical Center RotterdamRotterdamthe Netherlands
| | - Maryam Kavousi
- Department of EpidemiologyErasmus MC ‐ University Medical Center RotterdamRotterdamthe Netherlands
| | - Paul S. de Vries
- Human Genetics CenterDepartment of EpidemiologyHuman Genetics, and Environmental SciencesSchool of Public HealthThe University of Texas Health Science Center at HoustonHoustonTXUSA
| | - Abbas Dehghan
- Department of Biostatistics and EpidemiologyMRC‐PHE Centre for Environment and HealthSchool of Public HealthImperial College LondonLondonUK
| |
Collapse
|
42
|
van Tuijl LA, Voogd AC, de Graeff A, Hoogendoorn AW, Ranchor AV, Pan KY, Basten M, Lamers F, Geerlings MI, Abell JG, Awadalla P, Bakker MF, Beekman ATF, Bjerkeset O, Boyd A, Cui Y, Galenkamp H, Garssen B, Hellingman S, Huisman M, Huss A, Keats MR, Kok AAL, Luik AI, Noisel N, Onland-Moret NC, Payette Y, Penninx BWJH, Portengen L, Rissanen I, Roest AM, Rosmalen JGM, Ruiter R, Schoevers RA, Soave DM, Spaan M, Steptoe A, Stronks K, Sund ER, Sweeney E, Teyhan A, Vaartjes I, van der Willik KD, van Leeuwen FE, van Petersen R, Verschuren WMM, Visseren F, Vermeulen R, Dekker J. Psychosocial factors and cancer incidence (PSY-CA): Protocol for individual participant data meta-analyses. Brain Behav 2021; 11:e2340. [PMID: 34473425 PMCID: PMC8553309 DOI: 10.1002/brb3.2340] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 08/12/2021] [Indexed: 01/12/2023] Open
Abstract
OBJECTIVES Psychosocial factors have been hypothesized to increase the risk of cancer. This study aims (1) to test whether psychosocial factors (depression, anxiety, recent loss events, subjective social support, relationship status, general distress, and neuroticism) are associated with the incidence of any cancer (any, breast, lung, prostate, colorectal, smoking-related, and alcohol-related); (2) to test the interaction between psychosocial factors and factors related to cancer risk (smoking, alcohol use, weight, physical activity, sedentary behavior, sleep, age, sex, education, hormone replacement therapy, and menopausal status) with regard to the incidence of cancer; and (3) to test the mediating role of health behaviors (smoking, alcohol use, weight, physical activity, sedentary behavior, and sleep) in the relationship between psychosocial factors and the incidence of cancer. METHODS The psychosocial factors and cancer incidence (PSY-CA) consortium was established involving experts in the field of (psycho-)oncology, methodology, and epidemiology. Using data collected in 18 cohorts (N = 617,355), a preplanned two-stage individual participant data (IPD) meta-analysis is proposed. Standardized analyses will be conducted on harmonized datasets for each cohort (stage 1), and meta-analyses will be performed on the risk estimates (stage 2). CONCLUSION PSY-CA aims to elucidate the relationship between psychosocial factors and cancer risk by addressing several shortcomings of prior meta-analyses.
Collapse
Affiliation(s)
- Lonneke A van Tuijl
- Department of Internal Medicine, Maasstad Hospital, Rotterdam, The Netherlands
| | - Adri C Voogd
- Department of Internal Medicine, Division of Medical Oncology, GROW, Maastricht University Medical Centre, Maastricht, The Netherlands.,Department of Epidemiology, GROW, Maastricht University, Maastricht, The Netherlands.,Department of Research, Netherlands Comprehensive Cancer Organization (IKNL), Utrecht, The Netherlands
| | - Alexander de Graeff
- Department of Medical Oncology, Cancer Center University Medical Center, University of Utrecht, Utrecht, The Netherlands
| | - Adriaan W Hoogendoorn
- Amsterdam UMC, Department of Psychiatry, Amsterdam Public Health Research Institute, Vrije Universiteit, Amsterdam, The Netherlands.,GGZ inGeest Specialized Mental Health Care, Amsterdam, The Netherlands
| | - Adelita V Ranchor
- Department of Internal Medicine, Maasstad Hospital, Rotterdam, The Netherlands
| | - Kuan-Yu Pan
- Amsterdam UMC, Department of Psychiatry, Amsterdam Public Health Research Institute, Vrije Universiteit, Amsterdam, The Netherlands
| | - Maartje Basten
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | - Femke Lamers
- Amsterdam UMC, Department of Psychiatry, Amsterdam Public Health Research Institute, Vrije Universiteit, Amsterdam, The Netherlands
| | - Mirjam I Geerlings
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | - Jessica G Abell
- Department of Behavioural Science and Health, University College London, London, UK
| | - Philip Awadalla
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada.,Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Marije F Bakker
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | - Aartjan T F Beekman
- Amsterdam UMC, Department of Psychiatry, Amsterdam Public Health Research Institute, Vrije Universiteit, Amsterdam, The Netherlands
| | - Ottar Bjerkeset
- Faculty of Nursing and Health Sciences, Nord University, Bodø, Norway.,Faculty of Medicine and Health Sciences, Department of Mental Health, Norwegian University of Science and Technology, Trondheim, Norway
| | - Andy Boyd
- Bristol Medical School, Population Health Sciences, University of Bristol, Bristol, UK
| | - Yunsong Cui
- Atlantic Partnership for Tomorrow's Health, Faculty of Health, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Henrike Galenkamp
- Department of Public and Occupational Health, Amsterdam UMC, and Amsterdam Public Health Research Institute, University of Amsterdam, Amsterdam, Netherlands
| | - Bert Garssen
- Department of Internal Medicine, Maasstad Hospital, Rotterdam, The Netherlands
| | - Sean Hellingman
- Department of Mathematics, Wilfrid Laurier University, Waterloo, Canada
| | - Martijn Huisman
- Amsterdam UMC, Department of Epidemiology & Data Science, Amsterdam Public Health institute, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Department of Sociology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Anke Huss
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands
| | - Melanie R Keats
- School of Health and Human Performance, Faculty of Health, Dalhousie University, Halifax, Canada
| | - Almar A L Kok
- Amsterdam UMC, Department of Psychiatry, Amsterdam Public Health Research Institute, Vrije Universiteit, Amsterdam, The Netherlands.,Amsterdam UMC, Department of Epidemiology & Data Science, Amsterdam Public Health institute, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Annemarie I Luik
- Department of Epidemiology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands.,Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Nolwenn Noisel
- CARTaGENE, CHU Sainte-Justine, 3175, Chemin de la Côte-Sainte-Catherine, Montréal, Québec, Canada
| | - N Charlotte Onland-Moret
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | - Yves Payette
- CARTaGENE, CHU Sainte-Justine, 3175, Chemin de la Côte-Sainte-Catherine, Montréal, Québec, Canada
| | - Brenda W J H Penninx
- Amsterdam UMC, Department of Psychiatry, Amsterdam Public Health Research Institute, Vrije Universiteit, Amsterdam, The Netherlands
| | - Lützen Portengen
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands
| | - Ina Rissanen
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | - Annelieke M Roest
- Department of Developmental Psychology, University of Groningen, Groningen, The Netherlands
| | - Judith G M Rosmalen
- Departments of Psychiatry and Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Rikje Ruiter
- Department of Epidemiology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands.,Department of Internal Medicine, Maasstad, Rotterdam, The Netherlands
| | - Robert A Schoevers
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - David M Soave
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada.,Department of Mathematics, Wilfrid Laurier University, Waterloo, Canada
| | - Mandy Spaan
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Andrew Steptoe
- Department of Behavioural Science and Health, University College London, London, UK
| | - Karien Stronks
- Department of Public and Occupational Health, Amsterdam UMC, and Amsterdam Public Health Research Institute, University of Amsterdam, Amsterdam, Netherlands
| | - Erik R Sund
- Faculty of Nursing and Health Sciences, Nord University, Bodø, Norway.,Department of Public Health and Nursing, HUNT Research Centre, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Levanger hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - Ellen Sweeney
- Atlantic Partnership for Tomorrow's Health, Faculty of Health, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Alison Teyhan
- Bristol Medical School, Population Health Sciences, University of Bristol, Bristol, UK
| | - Ilonca Vaartjes
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | - Kimberly D van der Willik
- Department of Epidemiology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands.,Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Flora E van Leeuwen
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Rutger van Petersen
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | - W M Monique Verschuren
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands.,Centre for Nutrition, Prevention and Health Services, National Institute for Public Health and the Environment, Utrecht, the Netherlands
| | - Frank Visseren
- Department of Vascular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Roel Vermeulen
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands
| | - Joost Dekker
- Amsterdam Public Health Research Institute, Amsterdam, Noord-Holland, The Netherlands.,Department of Rehabilitation Medicine and Department of Psychiatry, Amsterdam UMC - VUMC, Amsterdam, Noord-Holland, The Netherlands
| |
Collapse
|
43
|
Allouche J, Rachmin I, Adhikari K, Pardo LM, Lee JH, McConnell AM, Kato S, Fan S, Kawakami A, Suita Y, Wakamatsu K, Igras V, Zhang J, Navarro PP, Lugo CM, Noonan HR, Christie KA, Itin K, Mujahid N, Lo JA, Won CH, Evans CL, Weng QY, Wang H, Osseiran S, Lovas A, Németh I, Cozzio A, Navarini AA, Hsiao JJ, Nguyen N, Kemény LV, Iliopoulos O, Berking C, Ruzicka T, Gonzalez-José R, Bortolini MC, Canizales-Quinteros S, Acuna-Alonso V, Gallo C, Poletti G, Bedoya G, Rothhammer F, Ito S, Schiaffino MV, Chao LH, Kleinstiver BP, Tishkoff S, Zon LI, Nijsten T, Ruiz-Linares A, Fisher DE, Roider E. NNT mediates redox-dependent pigmentation via a UVB- and MITF-independent mechanism. Cell 2021; 184:4268-4283.e20. [PMID: 34233163 PMCID: PMC8349839 DOI: 10.1016/j.cell.2021.06.022] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 03/09/2021] [Accepted: 06/15/2021] [Indexed: 12/26/2022]
Abstract
Ultraviolet (UV) light and incompletely understood genetic and epigenetic variations determine skin color. Here we describe an UV- and microphthalmia-associated transcription factor (MITF)-independent mechanism of skin pigmentation. Targeting the mitochondrial redox-regulating enzyme nicotinamide nucleotide transhydrogenase (NNT) resulted in cellular redox changes that affect tyrosinase degradation. These changes regulate melanosome maturation and, consequently, eumelanin levels and pigmentation. Topical application of small-molecule inhibitors yielded skin darkening in human skin, and mice with decreased NNT function displayed increased pigmentation. Additionally, genetic modification of NNT in zebrafish alters melanocytic pigmentation. Analysis of four diverse human cohorts revealed significant associations of skin color, tanning, and sun protection use with various single-nucleotide polymorphisms within NNT. NNT levels were independent of UVB irradiation and redox modulation. Individuals with postinflammatory hyperpigmentation or lentigines displayed decreased skin NNT levels, suggesting an NNT-driven, redox-dependent pigmentation mechanism that can be targeted with NNT-modifying topical drugs for medical and cosmetic purposes.
Collapse
Affiliation(s)
- Jennifer Allouche
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Inbal Rachmin
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Kaustubh Adhikari
- School of Mathematics and Statistics, The Open University, Milton Keynes, MK7 6AA, UK; Department of Genetics, Evolution and Environment and UCL Genetics Institute, University College London, London WC1E 6BT, UK
| | - Luba M Pardo
- Department of Dermatology, Erasmus Medical Center, 3015 Rotterdam, the Netherlands
| | - Ju Hee Lee
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, 03722 Seoul, Korea
| | - Alicia M McConnell
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and the Howard Hughes Medical Institute, Boston, MA 02115, USA
| | - Shinichiro Kato
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; Department of Immunology, Center for 5D Cell Dynamics, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Shaohua Fan
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, School of Life Sciences, Fudan University, 200438 Shanghai, China
| | - Akinori Kawakami
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Yusuke Suita
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Kazumasa Wakamatsu
- Institute for Melanin Chemistry, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Vivien Igras
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Jianming Zhang
- National Research Center for Translational Medicine (Shanghai), State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China
| | - Paula P Navarro
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Camila Makhlouta Lugo
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Haley R Noonan
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and the Howard Hughes Medical Institute, Boston, MA 02115, USA
| | - Kathleen A Christie
- Center for Genomic Medicine and Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| | - Kaspar Itin
- Department of Dermatology, University Hospital of Basel, 4031 Basel, Switzerland
| | - Nisma Mujahid
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; Boston University School of Medicine, Boston, MA 02118, USA; University of Utah, Department of Dermatology, Salt Lake City, UT 84132, USA
| | - Jennifer A Lo
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Chong Hyun Won
- Department of Dermatology, Asan Medical Center, Ulsan University College of Medicine, 05505 Seoul, Korea
| | - Conor L Evans
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Qing Yu Weng
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Hequn Wang
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Sam Osseiran
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Alyssa Lovas
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - István Németh
- Department of Dermatology and Allergology, University of Szeged, 6720 Szeged, Hungary
| | - Antonio Cozzio
- Department of Dermatology, Venerology, and Allergology, Kantonsspital St. Gallen, 9007 St. Gallen, Switzerland
| | - Alexander A Navarini
- Department of Dermatology, University Hospital of Basel, 4031 Basel, Switzerland
| | - Jennifer J Hsiao
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Nhu Nguyen
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Lajos V Kemény
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; Department of Dermatology, Venereology, and Dermatooncology, Semmelweis University, 1085 Budapest, Hungary
| | - Othon Iliopoulos
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA
| | - Carola Berking
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich Alexander University Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Thomas Ruzicka
- Department of Dermatology and Allergy, University Hospital Munich, Ludwig Maximilian University, 80337 Munich, Germany
| | - Rolando Gonzalez-José
- Instituto Patagónico de Ciencias Sociales y Humanas-Centro Nacional Patagónico, CONICET, Puerto Madryn U912OACD, Argentina
| | - Maria-Cátira Bortolini
- Departamento de Genética, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, Brazil
| | - Samuel Canizales-Quinteros
- Unidad de Genómica de Poblaciones Aplicada a la Salud, Facultad de Química, Universidad Nacional Autónoma de México e Instituto Nacional de Medicina Genómica, Mexico City 04510, Mexico
| | | | - Carla Gallo
- Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima 15102, Peru
| | - Giovanni Poletti
- Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima 15102, Peru
| | - Gabriel Bedoya
- Genética Molecular (GENMOL), Universidad de Antioquia, Medellín 5001000, Colombia
| | - Francisco Rothhammer
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000009, Chile; Programa de Genetica Humana, ICBM, Facultad de Medicina, Universidad de Chile, Santiago 1027, Chile
| | - Shosuke Ito
- Institute for Melanin Chemistry, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Maria Vittoria Schiaffino
- Internal Medicine, Diabetes and Endocrinology Unit, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Luke H Chao
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Benjamin P Kleinstiver
- Center for Genomic Medicine and Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| | - Sarah Tishkoff
- Departments of Genetics and Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Leonard I Zon
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and the Howard Hughes Medical Institute, Boston, MA 02115, USA
| | - Tamar Nijsten
- Department of Dermatology, Erasmus Medical Center, 3015 Rotterdam, the Netherlands
| | - Andrés Ruiz-Linares
- Ministry of Education Key Laboratory of Contemporary Anthropology and Collaborative Innovation Center of Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, Shanghai 200433, China; UMR 7268, CNRS-EFS-ADES, Aix-Marseille University, Marseille 13005, France
| | - David E Fisher
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA.
| | - Elisabeth Roider
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; Department of Dermatology, University Hospital of Basel, 4031 Basel, Switzerland; Department of Dermatology and Allergology, University of Szeged, 6720 Szeged, Hungary.
| |
Collapse
|
44
|
de Roos EW, Lahousse L, Verhamme KMC, Braunstahl GJ, In 't Veen JJCCM, Stricker BH, Brusselle GGO. Incidence and predictors of asthma exacerbations in middle-aged and older adults: the Rotterdam Study. ERJ Open Res 2021; 7:00126-2021. [PMID: 34262968 PMCID: PMC8273296 DOI: 10.1183/23120541.00126-2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 04/09/2021] [Indexed: 11/05/2022] Open
Abstract
Aim The aim of this study was to investigate occurrence and determinants of asthma exacerbations in an ageing general population. Methods Subjects aged 45 years or above with physician-diagnosed asthma in the Rotterdam Study, a population-based prospective cohort from January 1991 to May 2018, were assessed for asthma exacerbations. Exacerbations were defined as acute episodes of worsening asthma treated with oral corticosteroids. Cox proportional hazards analysis was used to investigate risk factors for a future exacerbation. Results Out of 763 participants with asthma (mean age 61.3 years, 69.2% female), 427 (56.0%) experienced at least one exacerbation, in a mean follow-up time of 13.9 years. The mean annual exacerbation rate was 0.22. Most exacerbations occurred during winter months. Risk factors for exacerbations were a history of previous exacerbations (HR 4.25; 95% CI 3.07–5.90, p<0.001)), respiratory complaints (HR 2.18; 95% CI 1.48–3.21, p<0.001), airflow obstruction (HR 1.52; 95% CI 1.07–2.15, p=0.019), obesity (HR 1.38; 95% CI 1.01–1.87, p=0.040) and depressive symptoms (HR 1.55; 95% CI 1.05–2.29, p=0.027). Compared to those not using respiratory medication, we observed higher hazard ratios for those on short-acting β2-agonists (SABA, i.e. rescue medication) only (HR 3.08, 95% CI 1.61–5.90, p=0.001) than those on controller medication (HR 2.50, 95% CI 1.59–3.92, p<0.001). Conclusion Many older adults with asthma suffer from at least one severe exacerbation. Previous exacerbations, use of SABA without concomitant controller medication, respiratory complaints, obesity, airway obstruction and depression are independent risk factors for exacerbations. Most middle-aged and older adults with asthma suffer from at least one severe exacerbation. Risk factors are previous exacerbations, use of SABA without concomitant controller medication, respiratory complaints, obesity, airway obstruction and depression.https://bit.ly/3gcTO6t
Collapse
Affiliation(s)
- Emmely W de Roos
- Dept of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium.,Dept of Epidemiology, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Lies Lahousse
- Dept of Epidemiology, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands.,Dept of Bioanalysis, Ghent University, Ghent, Belgium
| | - Katia M C Verhamme
- Dept of Bioanalysis, Ghent University, Ghent, Belgium.,Dept of Medical Informatics, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Gert-Jan Braunstahl
- Dept of Respiratory Medicine, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands.,Dept of Respiratory Medicine, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Johannes J C C M In 't Veen
- Dept of Respiratory Medicine, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands.,Dept of Respiratory Medicine, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Bruno H Stricker
- Dept of Epidemiology, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands.,Dept of Internal Medicine, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Guy G O Brusselle
- Dept of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium.,Dept of Epidemiology, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands.,Dept of Respiratory Medicine, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
45
|
Letarouilly JG, Flipo RM, Cortet B, Tournadre A, Paccou J. Body composition in patients with rheumatoid arthritis: a narrative literature review. Ther Adv Musculoskelet Dis 2021; 13:1759720X211015006. [PMID: 34221129 PMCID: PMC8221676 DOI: 10.1177/1759720x211015006] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 04/13/2021] [Indexed: 01/09/2023] Open
Abstract
There is growing interest in the alterations in body composition (BC) that accompany rheumatoid arthritis (RA). The purpose of this review is to (i) investigate how BC is currently measured in RA patients, (ii) describe alterations in body composition in RA patients and (iii) evaluate the effect on nutrition, physical training, and treatments; that is, corticosteroids and biologic Disease Modifying Anti-Rheumatic Disease (bDMARDs), on BC in RA patients. The primary-source literature for this review was acquired using PubMed, Scopus and Cochrane database searches for articles published up to March 2021. The Medical Subject Headings (MeSH) terms used were 'Arthritis, Rheumatoid', 'body composition', 'sarcopenia', 'obesity', 'cachexia', 'Absorptiometry, Photon' and 'Electric Impedance'. The titles and abstracts of all articles were reviewed for relevant subjects. Whole-BC measurements were usually performed using dual energy x-ray absorptiometry (DXA) to quantify lean- and fat-mass parameters. In RA patients, lean mass is lower and adiposity is higher than in healthy controls, both in men and women. The prevalence of abnormal BC conditions such as overfat, sarcopenia and sarcopenic obesity is significantly higher in RA patients than in healthy controls; these alterations in BC are observed even at an early stage of the disease. Data on the effect treatments on BC in RA patients are scarce. In the few studies published, (a) creatine supplementation and progressive resistance training induce a slight and temporary increase in lean mass, (b) exposure to corticosteroids induces a gain in fat mass and (c) tumour necrosis factor alpha (TNFα) inhibitors might be associated with a gain in fat mass, while tocilizumab might be associated with a gain in lean mass. The available data clearly demonstrate that alterations in BC occur in RA patients, but data on the effect of treatments, especially bDMARDs, are inconsistent and further studies are needed in this area.
Collapse
Affiliation(s)
- Jean-Guillaume Letarouilly
- University of Lille F-59000 Lille, CHU Lille F-59000 Lille, France; University of Littoral Côte d'Opale F-62200 Boulogne-sur-Mer, France; Marrow Adiposity and Bone Lab - MABLab ULR4490Lille, France
| | - René-Marc Flipo
- Department of Rheumatology, University of Lille, CHU Lille, F-59000 Lille, France
| | - Bernard Cortet
- University of Lille F-59000 Lille, CHU Lille F-59000 Lille, France; University of Littoral Côte d'Opale F-62200 Boulogne-sur-Mer, France; Marrow Adiposity and Bone Lab - MABLab ULR4490Lille, France
| | - Anne Tournadre
- University of Clermont Auvergne, CHU Clermont-Ferrand, UNH-UMR 1019, INRA Department of Rheumatology, F-63003 Clermont-Ferrand, France
| | - Julien Paccou
- MABlab ULR 4490, Department of Rheumatology, CHU Lille, 2, Avenue Oscar Lambret - 59037 Lille Cedex
| |
Collapse
|
46
|
Juul Rasmussen I, Rasmussen KL, Nordestgaard BG, Tybjærg-Hansen A, Frikke-Schmidt R. Impact of cardiovascular risk factors and genetics on 10-year absolute risk of dementia: risk charts for targeted prevention. Eur Heart J 2021; 41:4024-4033. [PMID: 33022702 PMCID: PMC7672536 DOI: 10.1093/eurheartj/ehaa695] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 05/07/2020] [Accepted: 08/12/2020] [Indexed: 12/18/2022] Open
Abstract
Aims Dementia is a major global challenge for health and social care in aging populations. A third of all dementia may be preventable due to cardiovascular risk factors. Intensive multi-domain intervention trials targeting primarily cardiovascular risk factors show improved cognitive function in people at risk. Such interventions will, however, be expensive to implement in all individuals at risk and will represent unrealistic economic tasks for most societies. Therefore, a risk score identifying high-risk individuals is warranted. Methods and results In 61 664 individuals from two prospective cohorts of the Danish general population, we generated 10-year absolute risk scores for all-cause dementia from cardiovascular risk factors and genetics. In both sexes, 10-year absolute risk of all-cause dementia increased with increasing age, number of apolipoprotein E (APOE) ɛ4 alleles, number of genome-wide association studies (GWAS) risk alleles, and cardiovascular risk factors. The highest 10-year absolute risks of all-cause dementia seen in smoking women with diabetes, low education, APOE ɛ44 genotype, and 22–31 GWAS risk alleles were 6%, 23%, 48%, and 66% in those aged 50–59, 60–69, 70–79, and 80–100, respectively. Corresponding values for men were 5%, 19%, 42%, and 60%, respectively. Conclusion Ten-year absolute risk of all-cause dementia increased with age, APOE ɛ4 alleles, GWAS risk alleles, diabetes, low education, and smoking in both women and men. Ten-year absolute risk charts for dementia will facilitate identification of high-risk individuals, those who likely will benefit the most from an early intervention against cardiovascular risk factors. ![]()
Collapse
Affiliation(s)
- Ida Juul Rasmussen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, DK-2100 Copenhagen, Denmark.,The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 1, DK-2730 Herlev, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen, Denmark
| | - Katrine Laura Rasmussen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, DK-2100 Copenhagen, Denmark.,The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 1, DK-2730 Herlev, Denmark
| | - Børge G Nordestgaard
- The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 1, DK-2730 Herlev, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen, Denmark.,Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 1, DK-2730 Herlev, Denmark.,The Copenhagen City Heart Study, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nordre Fasanvej 57, DK-2000 Frederiksberg, Denmark
| | - Anne Tybjærg-Hansen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, DK-2100 Copenhagen, Denmark.,The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 1, DK-2730 Herlev, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen, Denmark.,The Copenhagen City Heart Study, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nordre Fasanvej 57, DK-2000 Frederiksberg, Denmark
| | - Ruth Frikke-Schmidt
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, DK-2100 Copenhagen, Denmark.,The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 1, DK-2730 Herlev, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen, Denmark
| |
Collapse
|
47
|
Currant H, Hysi P, Fitzgerald TW, Gharahkhani P, Bonnemaijer PWM, Senabouth A, Hewitt AW, UK Biobank Eye and Vision Consortium, International Glaucoma Genetics Consortium, Atan D, Aung T, Charng J, Choquet H, Craig J, Khaw PT, Klaver CCW, Kubo M, Ong JS, Pasquale LR, Reisman CA, Daniszewski M, Powell JE, Pébay A, Simcoe MJ, Thiadens AAHJ, van Duijn CM, Yazar S, Jorgenson E, MacGregor S, Hammond CJ, Mackey DA, Wiggs JL, Foster PJ, Patel PJ, Birney E, Khawaja AP. Genetic variation affects morphological retinal phenotypes extracted from UK Biobank optical coherence tomography images. PLoS Genet 2021; 17:e1009497. [PMID: 33979322 PMCID: PMC8143408 DOI: 10.1371/journal.pgen.1009497] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 05/24/2021] [Accepted: 03/18/2021] [Indexed: 12/15/2022] Open
Abstract
Optical Coherence Tomography (OCT) enables non-invasive imaging of the retina and is used to diagnose and manage ophthalmic diseases including glaucoma. We present the first large-scale genome-wide association study of inner retinal morphology using phenotypes derived from OCT images of 31,434 UK Biobank participants. We identify 46 loci associated with thickness of the retinal nerve fibre layer or ganglion cell inner plexiform layer. Only one of these loci has been associated with glaucoma, and despite its clear role as a biomarker for the disease, Mendelian randomisation does not support inner retinal thickness being on the same genetic causal pathway as glaucoma. We extracted overall retinal thickness at the fovea, representative of foveal hypoplasia, with which three of the 46 SNPs were associated. We additionally associate these three loci with visual acuity. In contrast to the Mendelian causes of severe foveal hypoplasia, our results suggest a spectrum of foveal hypoplasia, in part genetically determined, with consequences on visual function.
Collapse
Affiliation(s)
- Hannah Currant
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Pirro Hysi
- School of Life Course Sciences, Section of Ophthalmology, King’s College London, London, United Kingdom
- Department of Twin Research and Genetic Epidemiology, King’s College London, London, United Kingdom
| | - Tomas W. Fitzgerald
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Puya Gharahkhani
- Statistical Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Pieter W. M. Bonnemaijer
- Department of Ophthalmology, Erasmus MC, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
- The Rotterdam Eye Hospital, Rotterdam, The Netherlands
| | - Anne Senabouth
- Garvan Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, The Kinghorn Cancer Centre, Darlinghurst, Australia
| | - Alex W. Hewitt
- Menzies Institute for Medical Research, School of Medicine, University of Tasmania, Tasmania, Australia
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia
| | | | | | - Denize Atan
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Bristol Eye Hospital, University Hospitals Bristol & Weston NHS Foundation Trust, Bristol, United Kingdom
| | - Tin Aung
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jason Charng
- Centre for Ophthalmology and Visual Science, Lions Eye Institute, The University of Western Australia, Perth, Australia
| | - Hélène Choquet
- Kaiser Permanente Northern California Division of Research, Oakland, California, United States of America
| | - Jamie Craig
- Department of Ophthalmology, Flinders University, Flinders Medical Centre, Bedford Park, Australia
| | - Peng T. Khaw
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, United Kingdom
| | - Caroline C. W. Klaver
- Department of Ophthalmology, Erasmus MC, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
- Department of Ophthalmology Radboud University Medical Center, Nijmegen, The Netherlands
- Institute of Molecular and Clinical Ophthalmology, Basel, Switzerland
| | - Michiaki Kubo
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Jue-Sheng Ong
- Statistical Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Louis R. Pasquale
- Eye and Vision Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Charles A. Reisman
- Topcon Healthcare Solutions R&D, Oakland, New Jersey, United States of America
| | - Maciej Daniszewski
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, Australia
| | - Joseph E. Powell
- Garvan Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, The Kinghorn Cancer Centre, Darlinghurst, Australia
- UNSW Cellular Genomics Futures Institute, University of New South Wales, Sydney, Australia
| | - Alice Pébay
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, Australia
- Department of Surgery, The University of Melbourne, Parkville, Australia
| | - Mark J. Simcoe
- Department of Ophthalmology, Kings College London, London, United Kingdom
- Institute of Ophthalmology, University College London, London, United Kingdom
| | | | - Cornelia M. van Duijn
- Nuffield Department Of Population Health, University of Oxford, Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, Oxford, United Kingdom
| | - Seyhan Yazar
- Garvan-Weizmann Centre for Single Cell Genomics, Garvan Institute of Medical Research, Sydney, Australia
| | - Eric Jorgenson
- Kaiser Permanente Northern California Division of Research, Oakland, California, United States of America
| | - Stuart MacGregor
- Statistical Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Chris J. Hammond
- School of Life Course Sciences, Section of Ophthalmology, King’s College London, London, United Kingdom
| | - David A. Mackey
- Centre for Ophthalmology and Visual Science, Lions Eye Institute, The University of Western Australia, Perth, Australia
| | - Janey L. Wiggs
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear, Boston, Massachusetts, United States of America
| | - Paul J. Foster
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, United Kingdom
| | - Praveen J. Patel
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, United Kingdom
| | - Ewan Birney
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Anthony P. Khawaja
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, United Kingdom
| |
Collapse
|
48
|
Simcoe M, Valdes A, Liu F, Furlotte NA, Evans DM, Hemani G, Ring SM, Smith GD, Duffy DL, Zhu G, Gordon SD, Medland SE, Vuckovic D, Girotto G, Sala C, Catamo E, Concas MP, Brumat M, Gasparini P, Toniolo D, Cocca M, Robino A, Yazar S, Hewitt A, Wu W, Kraft P, Hammond CJ, Shi Y, Chen Y, Zeng C, Klaver CCW, Uitterlinden AG, Ikram MA, Hamer MA, van Duijn CM, Nijsten T, Han J, Mackey DA, Martin NG, Cheng CY, Hinds DA, Spector TD, Kayser M, Hysi PG. Genome-wide association study in almost 195,000 individuals identifies 50 previously unidentified genetic loci for eye color. SCIENCE ADVANCES 2021; 7:eabd1239. [PMID: 33692100 PMCID: PMC7946369 DOI: 10.1126/sciadv.abd1239] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 01/25/2021] [Indexed: 05/03/2023]
Abstract
Human eye color is highly heritable, but its genetic architecture is not yet fully understood. We report the results of the largest genome-wide association study for eye color to date, involving up to 192,986 European participants from 10 populations. We identify 124 independent associations arising from 61 discrete genomic regions, including 50 previously unidentified. We find evidence for genes involved in melanin pigmentation, but we also find associations with genes involved in iris morphology and structure. Further analyses in 1636 Asian participants from two populations suggest that iris pigmentation variation in Asians is genetically similar to Europeans, albeit with smaller effect sizes. Our findings collectively explain 53.2% (95% confidence interval, 45.4 to 61.0%) of eye color variation using common single-nucleotide polymorphisms. Overall, our study outcomes demonstrate that the genetic complexity of human eye color considerably exceeds previous knowledge and expectations, highlighting eye color as a genetically highly complex human trait.
Collapse
Affiliation(s)
- Mark Simcoe
- Department of Twins Research and Genetic Epidemiology, King's College London, London, UK
- Department of Ophthalmology, King's College London, London, UK
| | - Ana Valdes
- Department of Twins Research and Genetic Epidemiology, King's College London, London, UK
- Division of Rheumatology, Orthopaedics and Dermatology, School of Medicine, University of Nottingham, Nottingham, UK
| | - Fan Liu
- Department of Genetic Identification, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | | | - David M Evans
- University of Queensland Diamantina Institute, University of Queensland, Brisbane, Queensland, Australia
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Gibran Hemani
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences Bristol Medical School University of Bristol, Bristol, UK
| | - Susan M Ring
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences Bristol Medical School University of Bristol, Bristol, UK
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences Bristol Medical School University of Bristol, Bristol, UK
| | - David L Duffy
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Gu Zhu
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Scott D Gordon
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Sarah E Medland
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Dragana Vuckovic
- Department of Medical Sciences, University of Trieste, Trieste, Italy
- Institute for Maternal and Child Health IRCCS "Burlo Garofolo", Trieste, Italy
- Epidemiology and Biostatistics Department, Faculty of Medicine, School of Public Health, Imperial College London, London, UK
| | - Giorgia Girotto
- Department of Medical Sciences, University of Trieste, Trieste, Italy
- Institute for Maternal and Child Health IRCCS "Burlo Garofolo", Trieste, Italy
| | - Cinzia Sala
- Division of Genetics of Common Disorders, S. Raffaele Scientific Institute, Milan, Italy
| | - Eulalia Catamo
- Department of Medical Sciences, University of Trieste, Trieste, Italy
| | - Maria Pina Concas
- Institute for Maternal and Child Health IRCCS "Burlo Garofolo", Trieste, Italy
| | - Marco Brumat
- Department of Medical Sciences, University of Trieste, Trieste, Italy
| | - Paolo Gasparini
- Department of Medical Sciences, University of Trieste, Trieste, Italy
- Institute for Maternal and Child Health IRCCS "Burlo Garofolo", Trieste, Italy
| | - Daniela Toniolo
- Division of Genetics of Common Disorders, S. Raffaele Scientific Institute, Milan, Italy
| | - Massimiliano Cocca
- Institute for Maternal and Child Health IRCCS "Burlo Garofolo", Trieste, Italy
| | - Antonietta Robino
- Institute for Maternal and Child Health IRCCS "Burlo Garofolo", Trieste, Italy
| | - Seyhan Yazar
- Centre for Ophthalmology and Visual Science, University of Western Australia, Lions Eye Institute, Perth, Australia
| | - Alex Hewitt
- Centre for Ophthalmology and Visual Science, University of Western Australia, Lions Eye Institute, Perth, Australia
- Centre for Eye Research Australia, University of Melbourne, Department of Ophthalmology, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
- School of Medicine, Menzies Research Institute Tasmania, University of Tasmania, Hobart, Australia
| | - Wenting Wu
- Department of Epidemiology, Fairbanks School of Public Health, Indiana University, and Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN, USA
| | - Peter Kraft
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Christopher J Hammond
- Department of Twins Research and Genetic Epidemiology, King's College London, London, UK
- Department of Ophthalmology, King's College London, London, UK
| | - Yuan Shi
- Singapore Eye Research Institute, Singapore National Eye Center, Singapore
| | - Yan Chen
- Department of Genetic Identification, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Changqing Zeng
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Caroline C W Klaver
- Department of Ophthalmology, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Ophthalmology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Andre G Uitterlinden
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Internal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Merel A Hamer
- Department of Dermatology, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Tamar Nijsten
- Department of Dermatology, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Jiali Han
- Department of Epidemiology, Fairbanks School of Public Health, Indiana University, and Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN, USA
| | - David A Mackey
- Centre for Ophthalmology and Visual Science, University of Western Australia, Lions Eye Institute, Perth, Australia
| | - Nicholas G Martin
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Ching-Yu Cheng
- Singapore Eye Research Institute, Singapore National Eye Center, Singapore
- Duke-NUS Medical School, Singapore
| | | | - Timothy D Spector
- Department of Twins Research and Genetic Epidemiology, King's College London, London, UK
| | - Manfred Kayser
- Department of Genetic Identification, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands.
| | - Pirro G Hysi
- Department of Twins Research and Genetic Epidemiology, King's College London, London, UK.
- Department of Ophthalmology, King's College London, London, UK
| |
Collapse
|
49
|
van der Graaf A, Zorro MM, Claringbould A, Võsa U, Aguirre-Gamboa R, Li C, Mooiweer J, Ricaño-Ponce I, Borek Z, Koning F, Kooy-Winkelaar Y, Sollid LM, Qiao SW, Kumar V, Li Y, Franke L, Withoff S, Wijmenga C, Sanna S, Jonkers I, BIOS Consortium. Systematic Prioritization of Candidate Genes in Disease Loci Identifies TRAFD1 as a Master Regulator of IFNγ Signaling in Celiac Disease. Front Genet 2021; 11:562434. [PMID: 33569077 PMCID: PMC7868554 DOI: 10.3389/fgene.2020.562434] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 12/16/2020] [Indexed: 12/17/2022] Open
Abstract
Celiac disease (CeD) is a complex T cell-mediated enteropathy induced by gluten. Although genome-wide association studies have identified numerous genomic regions associated with CeD, it is difficult to accurately pinpoint which genes in these loci are most likely to cause CeD. We used four different in silico approaches-Mendelian randomization inverse variance weighting, COLOC, LD overlap, and DEPICT-to integrate information gathered from a large transcriptomics dataset. This identified 118 prioritized genes across 50 CeD-associated regions. Co-expression and pathway analysis of these genes indicated an association with adaptive and innate cytokine signaling and T cell activation pathways. Fifty-one of these genes are targets of known drug compounds or likely druggable genes, suggesting that our methods can be used to pinpoint potential therapeutic targets. In addition, we detected 172 gene combinations that were affected by our CeD-prioritized genes in trans. Notably, 41 of these trans-mediated genes appear to be under control of one master regulator, TRAF-type zinc finger domain containing 1 (TRAFD1), and were found to be involved in interferon (IFN)γ signaling and MHC I antigen processing/presentation. Finally, we performed in vitro experiments in a human monocytic cell line that validated the role of TRAFD1 as an immune regulator acting in trans. Our strategy confirmed the role of adaptive immunity in CeD and revealed a genetic link between CeD and IFNγ signaling as well as with MHC I antigen processing, both major players of immune activation and CeD pathogenesis.
Collapse
Affiliation(s)
- Adriaan van der Graaf
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Maria M. Zorro
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Annique Claringbould
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Urmo Võsa
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Raúl Aguirre-Gamboa
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Chan Li
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Immunology, K. G. Jebsen Coeliac Disease Research Centre, University of Oslo, Oslo, Norway
| | - Joram Mooiweer
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Isis Ricaño-Ponce
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Zuzanna Borek
- Deutsches Rheumaforschungszentrum Berlin (DRFZ), An Institute of the Leibniz Association, Berlin, Germany
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Gastroenterology, Infectious Diseases and Rheumatology, Berlin, Germany
| | - Frits Koning
- Department of Immunology, Leiden University, Leiden, Netherlands
| | | | - Ludvig M. Sollid
- Department of Immunology, K. G. Jebsen Coeliac Disease Research Centre, University of Oslo, Oslo, Norway
| | - Shuo-Wang Qiao
- Department of Immunology, K. G. Jebsen Coeliac Disease Research Centre, University of Oslo, Oslo, Norway
| | - Vinod Kumar
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands
| | - Yang Li
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands
- Department of Computational Biology for Individualised Infection Medicine, Centre for Individualised Infection Medicine, Helmholtz Centre for Infection Research, Hannover Medical School, Hanover, Germany
| | - Lude Franke
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Sebo Withoff
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Cisca Wijmenga
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Immunology, K. G. Jebsen Coeliac Disease Research Centre, University of Oslo, Oslo, Norway
| | - Serena Sanna
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Istituto di Ricerca Genetica e Biomedica (IRGB) del Consiglio Nazionale delle Ricerche (CNR), Monserrato, Italy
| | - Iris Jonkers
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Immunology, K. G. Jebsen Coeliac Disease Research Centre, University of Oslo, Oslo, Norway
| | | |
Collapse
|
50
|
Boenniger MM, Diers K, Herholz SC, Shahid M, Stöcker T, Breteler MMB, Huijbers W. A Functional MRI Paradigm for Efficient Mapping of Memory Encoding Across Sensory Conditions. Front Hum Neurosci 2021; 14:591721. [PMID: 33551773 PMCID: PMC7859438 DOI: 10.3389/fnhum.2020.591721] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 12/02/2020] [Indexed: 11/13/2022] Open
Abstract
We introduce a new and time-efficient memory-encoding paradigm for functional magnetic resonance imaging (fMRI). This paradigm is optimized for mapping multiple contrasts using a mixed design, using auditory (environmental/vocal) and visual (scene/face) stimuli. We demonstrate that the paradigm evokes robust neuronal activity in typical sensory and memory networks. We were able to detect auditory and visual sensory-specific encoding activities in auditory and visual cortices. Also, we detected stimulus-selective activation in environmental-, voice-, scene-, and face-selective brain regions (parahippocampal place and fusiform face area). A subsequent recognition task allowed the detection of sensory-specific encoding success activity (ESA) in both auditory and visual cortices, as well as sensory-unspecific positive ESA in the hippocampus. Further, sensory-unspecific negative ESA was observed in the precuneus. Among others, the parallel mixed design enabled sustained and transient activity comparison in contrast to rest blocks. Sustained and transient activations showed great overlap in most sensory brain regions, whereas several regions, typically associated with the default-mode network, showed transient rather than sustained deactivation. We also show that the use of a parallel mixed model had relatively little influence on positive or negative ESA. Together, these results demonstrate a feasible, versatile, and brief memory-encoding task, which includes multiple sensory stimuli to guarantee a comprehensive measurement. This task is especially suitable for large-scale clinical or population studies, which aim to test task-evoked sensory-specific and sensory-unspecific memory-encoding performance as well as broad sensory activity across the life span within a very limited time frame.
Collapse
Affiliation(s)
- Meta M. Boenniger
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Kersten Diers
- Image Analysis Group, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Sibylle C. Herholz
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Mohammad Shahid
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Tony Stöcker
- MR Physics, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Monique M. B. Breteler
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Institute for Medical Biometry, Informatics and Epidemiology (IMBIE), Faculty of Medicine, University of Bonn, Bonn, Germany
| | - Willem Huijbers
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| |
Collapse
|