1
|
Mirzaei H, Patrova J, Mannheimer B, Lindh JD, Falhammar H. Prevalence and Incidence of Dementia in Patients With Non-Overtly Functional Adrenal Tumours. Clin Endocrinol (Oxf) 2025; 102:371-379. [PMID: 39722568 PMCID: PMC11874157 DOI: 10.1111/cen.15186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 12/12/2024] [Accepted: 12/14/2024] [Indexed: 12/28/2024]
Abstract
OBJECTIVE To investigate the prevalence and incidence of dementia in patients with non-overtly functional adrenal tumours (NOFATs). DESIGN A national retrospective register-based study was conducted on patients diagnosed with NOFAT and controls diagnosed between 2005 and 2019, in Sweden. Individuals diagnosed with overt adrenal hormone excess or previous malignancies were excluded. Sensitivity analyses were performed in subgroups with a combination of gallbladder/biliary tract/pancreatic disease. and acute appendicitis, as well as 3- and 12-months of malignancy-free survival. MEASUREMENTS Prevalence and incidence of dementia. The secondary outcomes were Alzheimer's disease and vascular dementia. RESULTS Among 20,390 cases, 12,120 (59.4%) were women, and the median (IQR) age was 66 (57-73) years. Among the 125,392 controls, 69,994 (55.8%) were women and the median (IQR) age was 66 (57-73) years. Patients with NOFATs had a lower prevalence of dementia compared to controls (odds ratio [OR] 0.58, 95% CI 0.50-0.68, adjusted OR [aOR] 0.47, 95% CI 0.40-0.56). During the follow-up period (median 4.9 years, IQR 2.2-8.2), incidence of dementia was similar in NOFATs and controls (hazard ratio [HR] 1.05, 95% CI 0.97-1.15, adjusted HR [aHR] 1.06, 95% CI 0.97-1.15). Similar results obtained for Alzheimer's dementia (aOR 0.44, 95% CI 0.34-0.57; aHR 0.94, 95% CI 0.80-1.10) and vascular dementia (OR 0.71, 95% CI 0.52-0.94, aOR 0.48, 95% CI 0.35-0.64; HR 1.29, 95% CI 1.08-1.53, aHR 1.13, 95% CI 0.95-1.35) as well as in the sensitivity analyses. Adrenalectomy did not change the results. CONCLUSION NOFAT was not associated with an increased risk of dementia.
Collapse
Affiliation(s)
- Hadis Mirzaei
- Department of EndocrinologySödersjukhusetStockholmSweden
- Department of Clinical Science ad EducationSödersjukhuset, Karolinska InstituteStockholmSweden
| | - Jekaterina Patrova
- Department of EndocrinologySödersjukhusetStockholmSweden
- Department of Clinical Science ad EducationSödersjukhuset, Karolinska InstituteStockholmSweden
| | - Buster Mannheimer
- Department of EndocrinologySödersjukhusetStockholmSweden
- Department of Clinical Science ad EducationSödersjukhuset, Karolinska InstituteStockholmSweden
| | - Jonatan D. Lindh
- Laboratory Medicine, Division of Clinical PharmacologyKarolinska InstituteStockholmSweden
| | - Henrik Falhammar
- Department of Molecular Medicine and SurgeryKarolinska InstituteStockholmSweden
- Department of EndocrinologyKarolinska University HospitalStockholmSweden
| |
Collapse
|
2
|
McLennan S, Meyer L, Wangmo T, Gaab J, Elger B, Seaward H. Psychological stressors of imprisonment and coping of older incarcerated persons: a qualitative interview study. BMC Public Health 2025; 25:328. [PMID: 39865221 PMCID: PMC11770940 DOI: 10.1186/s12889-025-21452-w] [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: 09/10/2024] [Accepted: 01/14/2025] [Indexed: 01/28/2025] Open
Abstract
BACKGROUND Imprisonment has a major impact on a person's psychological well-being. The proportion of older imprisoned persons is dramatically increasing worldwide, and they are likely to have greater physical and mental health needs compared to younger persons in prisons. However, there is currently a lack of research on the psychological stressors and the coping strategies of older imprisoned persons. This study therefore aims to explore the key psychological stressors experienced by older imprisoned persons and their coping strategies. METHODS Individual semi-structured qualitative interviews were conducted between April 2017 and December 2018 with a purposive sample of 79 participants from 2 different groups in Switzerland: older imprisoned persons (n = 50) and mental health professionals (n = 29) with experience working in prisons. Transcripts were analysed using conventional content analysis. RESULTS This study has identified various ways in which the prison environment not only undermines older incarcerated persons´ psychological well-being, but also their ability to manage the stress they are experiencing. Two overarching psychological stressors identified were a lack of physical and emotional closeness in social relationships (with prison staff, with other incarcerated persons, and outside of prison), and the loss of autonomy in prison. Participants reported five main ways that older incarcerated persons coped with the psychological stressors in prison: 1) recognising a lack of control over situation, 2) withdrawing and isolating, 3) self-improvement, 4) staying connected to the outside world, and 5) self-expression. CONCLUSIONS To improve the psychological well-being of older incarcerated persons, there is a need for: specialised training of prison staff regarding hardships that elderly persons can face in prison relationships, encouragement and enablement of social contacts with the outside world, and increasing possibilities for autonomy, new challenges, and thus stimuli within a limited setting.
Collapse
Affiliation(s)
- Stuart McLennan
- Institute for Biomedical Ethics, University of Basel, Bernoullistrasse 28, 4056, Basel, Switzerland
- Institute of History and Ethics in Medicine, Department of Preclinical Medicine, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Leila Meyer
- Institute for Biomedical Ethics, University of Basel, Bernoullistrasse 28, 4056, Basel, Switzerland
| | - Tenzin Wangmo
- Institute for Biomedical Ethics, University of Basel, Bernoullistrasse 28, 4056, Basel, Switzerland
| | - Jens Gaab
- Division of Clinical Psychology and Psychotherapy, Faculty of Psychology, University of Basel, Basel, Switzerland
| | - Bernice Elger
- Institute for Biomedical Ethics, University of Basel, Bernoullistrasse 28, 4056, Basel, Switzerland
- Center for Legal Medicine (CURML), Medical Faculty, University of Geneva, Geneva, Switzerland
| | - Helene Seaward
- Institute for Biomedical Ethics, University of Basel, Bernoullistrasse 28, 4056, Basel, Switzerland.
| |
Collapse
|
3
|
Granov R, Vedad S, Wang SH, Durham A, Shah D, Pasinetti GM. The Role of the Neural Exposome as a Novel Strategy to Identify and Mitigate Health Inequities in Alzheimer's Disease and Related Dementias. Mol Neurobiol 2025; 62:1205-1224. [PMID: 38967905 PMCID: PMC11711138 DOI: 10.1007/s12035-024-04339-6] [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: 12/26/2023] [Accepted: 06/28/2024] [Indexed: 07/06/2024]
Abstract
With the continuous increase of the elderly population, there is an urgency to understand and develop relevant treatments for Alzheimer's disease and related dementias (ADRD). In tandem with this, the prevalence of health inequities continues to rise as disadvantaged communities fail to be included in mainstream research. The neural exposome poses as a relevant mechanistic approach and tool for investigating ADRD onset, progression, and pathology as it accounts for several different factors: exogenous, endogenous, and behavioral. Consequently, through the neural exposome, health inequities can be addressed in ADRD research. In this paper, we address how the neural exposome relates to ADRD by contributing to the discourse through defining how the neural exposome can be developed as a tool in accordance with machine learning. Through this, machine learning can allow for developing a greater insight into the application of transferring and making sense of experimental mouse models exposed to health inequities and potentially relate it to humans. The overall goal moving beyond this paper is to define a multitude of potential factors that can increase the risk of ADRD onset and integrate them to create an interdisciplinary approach to the study of ADRD and subsequently translate the findings to clinical research.
Collapse
Affiliation(s)
- Ravid Granov
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10019, USA
| | - Skyler Vedad
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10019, USA
| | - Shu-Han Wang
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10019, USA
| | - Andrea Durham
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10019, USA
| | - Divyash Shah
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10019, USA
| | - Giulio Maria Pasinetti
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10019, USA.
- Geriatrics Research, Education and Clinical Center, JJ Peters VA Medical Center, Bronx, NY, 10468, USA.
| |
Collapse
|
4
|
Martocchia A, Stefanelli M, Gallucci M, Noale M, Maggi S, Cassol M, Postacchini D, Proietti A, Barbagallo M, Dominguez LJ, Ferri C, Desideri G, Toussan L, Pastore F, Falaschi GM, Paolisso G, Falaschi P. Increased nocturnal urinary cortisol levels in the elderly patients with depression, coexisting major geriatric syndromes and combined pathogenetic mechanisms. Aging Clin Exp Res 2024; 36:196. [PMID: 39331197 PMCID: PMC11436436 DOI: 10.1007/s40520-024-02849-w] [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: 08/08/2024] [Accepted: 09/09/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND The mechanisms at the basis of depression are still matter of debate, but several studies in the literature suggest common pathways with dementia (genetic predispositions, metabolic and inflammatory mechanisms, neuropathological changes) and other geriatric syndromes. AIMS To evaluate the role of cortisol (as marker of the HPA, hypothalamus-pituitary-adrenal axis hyperactivity) in elderly subjects with depressive symptoms (by the means of the AGICO, AGIng and COrtisol, study), in relationship to the presence of the major geriatric syndromes. METHODS The AGICO study enrolled patients from ten Geriatric Units in Italy. Every subject received a comprehensive geriatric assessment or CGA (including the Mini Mental State Examination or MMSE, Geriatric Depression Scale or GDS and Cornell Scale for Depression in Dementia or CSDD), the neurological examination (with a computed tomography scan or magnetic resonance imaging of the brain), the assessment of the metabolic syndrome (MetS), the evaluation of the cortisol activity by two consecutive urine collections (diurnal and nocturnal), a CGA-derived frailty index (FI) and a modified measure of allostatic load (AL). RESULTS The MMSE scores were significantly and inversely related to the values of GDS (p < 0.001) and CSDD (p < 0.05), respectively. The patients with depressive symptoms (GDS/CSDD > 8) showed significantly increased disability, MetS, inflammation, FI and AL and significantly reduced MMSE and renal function. The diurnal and nocturnal urinary cortisol levels in the patients with depressive symptoms (GDS/CSDD > 8) were higher with respects to controls (p < 0.05 for nocturnal difference). DISCUSSION The AGICO study showed that the stress response is activated in the patients with depression. CONCLUSION The depression in elderly patient should be reconsidered as a systemic disease, with coexisting major geriatric syndromes (disability, dementia, frailty) and combined pathogenetic mechanisms (metabolic syndrome, impaired renal function, low-grade inflammation, and allostatic load). Cortisol confirmed its role as principal mediator of the aging process in both dementia and metabolic syndrome.
Collapse
Affiliation(s)
- Antonio Martocchia
- S. Andrea Hospital, Sapienza University of Rome, Via Di Grottarossa 1035, 00189, Rome, Italy.
| | - Manuela Stefanelli
- S. Andrea Hospital, Sapienza University of Rome, Via Di Grottarossa 1035, 00189, Rome, Italy
- Casa Di Cura Villa Domelia, Rome, Italy
| | - Maurizio Gallucci
- Cognitive Impairment Centre, Local Unit of Health and Social Services N.2, Marca Trevigiana, Treviso, Italy
| | - Marianna Noale
- Aging Branch, Neuroscience Institute, National Research Council (CNR), Padua, Italy
| | - Stefania Maggi
- Aging Branch, Neuroscience Institute, National Research Council (CNR), Padua, Italy
| | | | - Demetrio Postacchini
- Geriatrics Operative Unit, Italian National Research Centre On Aging (IRCCS INRCA), Fermo, Italy
| | - Antonella Proietti
- S. Andrea Hospital, Sapienza University of Rome, Via Di Grottarossa 1035, 00189, Rome, Italy
| | - Mario Barbagallo
- Department of Medicine, Geriatric Unit, University of Palermo, Palermo, Italy
| | - Ligia J Dominguez
- Department of Medicine, Geriatric Unit, University of Palermo, Palermo, Italy
| | - Claudio Ferri
- Department of Clinical Medicine, Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Giovambattista Desideri
- Department of Clinical, Internal Medicine, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Lavinia Toussan
- S. Andrea Hospital, Sapienza University of Rome, Via Di Grottarossa 1035, 00189, Rome, Italy
- RSA Anni Azzurri Parco Di Veio, Rome, Italy
| | | | - Giulia M Falaschi
- S. Andrea Hospital, Sapienza University of Rome, Via Di Grottarossa 1035, 00189, Rome, Italy
| | - Giuseppe Paolisso
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Paolo Falaschi
- S. Andrea Hospital, Sapienza University of Rome, Via Di Grottarossa 1035, 00189, Rome, Italy
| |
Collapse
|
5
|
Peng T, Yang Y, Ma J, Xu P, Xie X, Hu N, Yan Y. Dementia and metabolic syndrome: a bibliometric analysis. Front Aging Neurosci 2024; 16:1400589. [PMID: 38934020 PMCID: PMC11199533 DOI: 10.3389/fnagi.2024.1400589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
Background Dementia is a progressive neurodegenerative condition, while metabolic syndrome (MetS) is characterized by a combination of metabolic abnormalities such as hypertension, high blood sugar, and obesity. There exists a connection and overlap between the two conditions in certain aspects, and both are influenced to varying degrees by the process of aging. This study presents an overview of the current research landscape regarding dementia and MetS through bibliometric analysis. Methods A systematic search was conducted to retrieve relevant literature on dementia and MetS published between 1 January 2000, and 30 November 2023, from the Web of Science Core Collection database. Various bibliometric tools, including VOSviewer, CiteSpace, and the R software package "bibliometrix," were utilized for analysis. Results A total of 717 articles were identified, showing an upward trend in annual publications. Leading contributors included the United States, Italy, and China, with institutions such as the University of California System at the forefront. The Journal of Alzheimer's Disease emerged as the top publisher, while research published in Neurology garnered significant citations. Noteworthy authors encompassed Panza, Francesco; Frisardi, Vincenza; and Feldman, Eva L, with Kristine Yaffe being the most cited author (280 citations). Recent studies have focused on themes like "gut microbiota," "neuroinflammation," "fatty acids," and "microglia." Conclusion This bibliometric analysis summarizes the foundational knowledge structure in the realm of dementia and MetS from 2000 to 2023. By highlighting current research frontiers and trending topics, this analysis serves as a valuable reference for researchers in the field.
Collapse
Affiliation(s)
- Tao Peng
- First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Yifan Yang
- Department of Encephalopathy, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Jingying Ma
- First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Peili Xu
- First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Xinchun Xie
- First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Nan Hu
- First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Yongmei Yan
- Department of Encephalopathy, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| |
Collapse
|
6
|
Hawley AL, Baum JI. Nutrition as the foundation for successful aging: a focus on dietary protein and omega-3 polyunsaturated fatty acids. Nutr Rev 2024; 82:389-406. [PMID: 37319363 DOI: 10.1093/nutrit/nuad061] [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] [Indexed: 06/17/2023] Open
Abstract
Skeletal muscle plays a critical role throughout the aging process. People living with sarcopenia, a progressive and generalized loss of skeletal muscle mass and function, often experience diminished quality of life, which can be attributed to a long period of decline and disability. Therefore, it is important to identify modifiable factors that preserve skeletal muscle and promote successful aging (SA). In this review, SA was defined as (1) low cardiometabolic risk, (2) preservation of physical function, and (3) positive state of wellbeing, with nutrition as an integral component. Several studies identify nutrition, specifically high-quality protein (eg, containing all essential amino acids), and long-chain omega-3 polyunsaturated fatty acids (n-3 PUFAs), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA), as positive regulators of SA. Recently, an additive anabolic effect of protein and n-3 PUFAs has been identified in skeletal muscle of older adults. Evidence further suggests that the additive effect of protein and n-3 PUFAs may project beyond skeletal muscle anabolism and promote SA. The key mechanism(s) behind the enhanced effects of intake of protein and n-3 PUFAs needs to be defined. The first objective of this review is to evaluate skeletal muscle as a driver of cardiometabolic health, physical function, and wellbeing to promote SA. The second objective is to examine observational and interventional evidence of protein and n-3 PUFAs on skeletal muscle to promote SA. The final objective is to propose mechanisms by which combined optimal intake of high-quality protein and n-3 PUFAs likely play a key role in SA. Current evidence suggests that increased intake of protein above the Recommended Dietary Allowance and n-3 PUFAs above the Dietary Guidelines for Americans recommendations for late middle-aged and older adults is required to maintain skeletal muscle mass and to promote SA, potentially through the mechanistical target of rapamycin complex 1 (mTORC1).
Collapse
Affiliation(s)
- Aubree L Hawley
- School of Human and Environmental Sciences, University of Arkansas, Fayetteville, AR, USA
| | - Jamie I Baum
- Center for Human Nutrition, Department of Food Science, University of Arkansas System Division of Agriculture, Fayetteville, AR, USA
| |
Collapse
|
7
|
Lakhani A, Kang DH, Kang YE, Park JO. Toward Systems-Level Metabolic Analysis in Endocrine Disorders and Cancer. Endocrinol Metab (Seoul) 2023; 38:619-630. [PMID: 37989266 PMCID: PMC10764991 DOI: 10.3803/enm.2023.1814] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/27/2023] [Accepted: 11/01/2023] [Indexed: 11/23/2023] Open
Abstract
Metabolism is a dynamic network of biochemical reactions that support systemic homeostasis amidst changing nutritional, environmental, and physical activity factors. The circulatory system facilitates metabolite exchange among organs, while the endocrine system finely tunes metabolism through hormone release. Endocrine disorders like obesity, diabetes, and Cushing's syndrome disrupt this balance, contributing to systemic inflammation and global health burdens. They accompany metabolic changes on multiple levels from molecular interactions to individual organs to the whole body. Understanding how metabolic fluxes relate to endocrine disorders illuminates the underlying dysregulation. Cancer is increasingly considered a systemic disorder because it not only affects cells in localized tumors but also the whole body, especially in metastasis. In tumorigenesis, cancer-specific mutations and nutrient availability in the tumor microenvironment reprogram cellular metabolism to meet increased energy and biosynthesis needs. Cancer cachexia results in metabolic changes to other organs like muscle, adipose tissue, and liver. This review explores the interplay between the endocrine system and systems-level metabolism in health and disease. We highlight metabolic fluxes in conditions like obesity, diabetes, Cushing's syndrome, and cancers. Recent advances in metabolomics, fluxomics, and systems biology promise new insights into dynamic metabolism, offering potential biomarkers, therapeutic targets, and personalized medicine.
Collapse
Affiliation(s)
- Aliya Lakhani
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Da Hyun Kang
- Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Yea Eun Kang
- Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Junyoung O. Park
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
8
|
Burns DM, He C, Li YL, Zhuo J, Qian DQ, Chen L, Jalluri R, Diamond S, Covington MB, Li Y, Wynn R, Scherle P, Yeleswaram S, Hollis G, Friedman S, Metcalf B, Yao W. Discovery of a novel 2-spiroproline steroid mimetic scaffold for the potent inhibition of 11β-HSD1. Bioorg Med Chem Lett 2022; 73:128884. [PMID: 35835377 DOI: 10.1016/j.bmcl.2022.128884] [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: 03/09/2022] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 11/30/2022]
Abstract
11β-hydroxysteroid dehydrogenase 1 (11β-HSD1) has been identified as the primary enzyme responsible for the activation of hepatic cortisone to cortisol in specific peripheral tissues, resulting in the concomitant antagonism of insulin action within these tissues. Dysregulation of 11β-HSD1, particularly in adipose tissues, has been associated with a variety of ailments including metabolic syndrome and type 2 diabetes mellitus. Therefore, inhibition of 11β-HSD1 with a small nonsteroidal molecule is therapeutically desirable. Implementation of a scaffold-hopping approach revealed a 3-point pharmacophore for 11β-HSD1 that was utilized to design a 2-spiroproline derivative as a steroid mimetic scaffold. Reiterative optimization provided valuable insight into the bioactive conformation of our novel scaffold and led to the discovery of several leads, such as compounds 39 and 51. Importantly, deleterious hERG inhibition and pregnane X receptor induction were mitigated by the introduction of a 4-hydroxyl group to the proline ring system.
Collapse
Affiliation(s)
- David M Burns
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA.
| | - Chunhong He
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | | | - Jincong Zhuo
- Prelude Therapeutics, 200 Powder Mill Road, Wilmington, DE 19803, USA
| | - Ding-Quan Qian
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | | | | | - Sharon Diamond
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | | | - Yanlong Li
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | | | - Peggy Scherle
- Prelude Therapeutics, 200 Powder Mill Road, Wilmington, DE 19803, USA
| | - Swamy Yeleswaram
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | | | | | | | | |
Collapse
|
9
|
Souza-Teodoro LH, Andrade LHS, Carvalho LA. Could be dehydroepiandrosterone (DHEA) a novel target for depression? JOURNAL OF AFFECTIVE DISORDERS REPORTS 2022. [DOI: 10.1016/j.jadr.2022.100340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
10
|
Tian R, Han K, Geng Y, Yang C, Shi C, Thomas PB, Pearce C, Moffatt K, Ma S, Xu S, Yang G, Zhou X, Gladyshev VN, Liu X, Fisher DO, Chopin LK, Leiner NO, Baker AM, Fan G, Seim I. A chromosome-level genome of Antechinus flavipes provides a reference for an Australian marsupial genus with male death after mating. Mol Ecol Resour 2022; 22:740-754. [PMID: 34486812 PMCID: PMC9290055 DOI: 10.1111/1755-0998.13501] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 08/16/2021] [Accepted: 08/31/2021] [Indexed: 11/30/2022]
Abstract
The 15 species of small carnivorous marsupials that comprise the genus Antechinus exhibit semelparity, a rare life-history strategy in mammals where synchronized death occurs after one breeding season. Antechinus males, but not females, age rapidly (demonstrate organismal senescence) during the breeding season and show promise as new animal models of ageing. Some antechinus species are also threatened or endangered. Here, we report a chromosome-level genome of a male yellow-footed antechinus Antechinus flavipes. The genome assembly has a total length of 3.2 Gb with a contig N50 of 51.8 Mb and a scaffold N50 of 636.7 Mb. We anchored and oriented 99.7% of the assembly on seven pseudochromosomes and found that repetitive DNA sequences occupy 51.8% of the genome. Draft genome assemblies of three related species in the subfamily Phascogalinae, two additional antechinus species (Antechinus argentus and A. arktos) and the iteroparous sister species Murexia melanurus, were also generated. Preliminary demographic analysis supports the hypothesis that climate change during the Pleistocene isolated species in Phascogalinae and shaped their population size. A transcriptomic profile across the A. flavipes breeding season allowed us to identify genes associated with aspects of the male die-off. The chromosome-level A. flavipes genome provides a steppingstone to understanding an enigmatic life-history strategy and a resource to assist the conservation of antechinuses.
Collapse
Affiliation(s)
- Ran Tian
- Integrative Biology LaboratoryCollege of Life SciencesNanjing Normal UniversityNanjingChina
- Jiangsu Key Laboratory for Biodiversity and BiotechnologyCollege of Life SciencesNanjing Normal UniversityNanjingChina
| | - Kai Han
- BGI‐QingdaoBGI‐ShenzhenQingdaoChina
| | - Yuepan Geng
- Integrative Biology LaboratoryCollege of Life SciencesNanjing Normal UniversityNanjingChina
| | - Chen Yang
- Integrative Biology LaboratoryCollege of Life SciencesNanjing Normal UniversityNanjingChina
| | | | - Patrick B. Thomas
- Ghrelin Research GroupTranslational Research Institute‐Institute of Health and Biomedical InnovationSchool of Biomedical SciencesQueensland University of TechnologyBrisbaneQLDAustralia
- Australian Prostate Cancer Research Centre‐QueenslandTranslational Research Institute – Institute of Health and Biomedical InnovationQueensland University of TechnologyBrisbaneQLDAustralia
- Queensland Bladder Cancer InitiativeTranslational Research Institute‐Institute of Health and Biomedical InnovationSchool of Biomedical SciencesQueensland University of TechnologyWoolloongabbaQLDAustralia
| | - Coral Pearce
- School of Biology and Environmental ScienceQueensland University of TechnologyBrisbaneQLDAustralia
| | - Kate Moffatt
- School of Biology and Environmental ScienceQueensland University of TechnologyBrisbaneQLDAustralia
| | - Siming Ma
- Genome Institute of SingaporeAgency for Science, Technology and Research (A*STAR)SingaporeSingapore
| | - Shixia Xu
- Jiangsu Key Laboratory for Biodiversity and BiotechnologyCollege of Life SciencesNanjing Normal UniversityNanjingChina
| | - Guang Yang
- Jiangsu Key Laboratory for Biodiversity and BiotechnologyCollege of Life SciencesNanjing Normal UniversityNanjingChina
| | - Xuming Zhou
- Key Laboratory of Animal Ecology and Conservation BiologyInstitute of ZoologyChinese Academy of SciencesBeijingChina
| | - Vadim N. Gladyshev
- Division of GeneticsDepartment of MedicineBrigham and Women’s Hospital and Harvard Medical SchoolBostonMAUSA
| | - Xin Liu
- BGI‐QingdaoBGI‐ShenzhenQingdaoChina
| | - Diana O. Fisher
- School of Biological SciencesUniversity of QueenslandBrisbaneQLDAustralia
| | - Lisa K. Chopin
- Ghrelin Research GroupTranslational Research Institute‐Institute of Health and Biomedical InnovationSchool of Biomedical SciencesQueensland University of TechnologyBrisbaneQLDAustralia
- Australian Prostate Cancer Research Centre‐QueenslandTranslational Research Institute – Institute of Health and Biomedical InnovationQueensland University of TechnologyBrisbaneQLDAustralia
| | - Natália O. Leiner
- Laboratório de Ecologia de MamíferosInstituto de BiologiaUniversidade Federal de UberlândiaUberlândiaBrazil
| | - Andrew M. Baker
- School of Biology and Environmental ScienceQueensland University of TechnologyBrisbaneQLDAustralia
- Natural Environments ProgramQueensland MuseumSouth BrisbaneQLDAustralia
| | - Guangyi Fan
- BGI‐QingdaoBGI‐ShenzhenQingdaoChina
- State Key Laboratory of Quality Research in Chinese MedicineInstitute of Chinese Medical SciencesUniversity of MacauMacauChina
- State Key Laboratory of Agricultural GenomicsBGI‐ShenzhenShenzhenChina
| | - Inge Seim
- Integrative Biology LaboratoryCollege of Life SciencesNanjing Normal UniversityNanjingChina
- Ghrelin Research GroupTranslational Research Institute‐Institute of Health and Biomedical InnovationSchool of Biomedical SciencesQueensland University of TechnologyBrisbaneQLDAustralia
- Australian Prostate Cancer Research Centre‐QueenslandTranslational Research Institute – Institute of Health and Biomedical InnovationQueensland University of TechnologyBrisbaneQLDAustralia
- School of Biology and Environmental ScienceQueensland University of TechnologyBrisbaneQLDAustralia
| |
Collapse
|
11
|
Exploring New Drug Targets for Type 2 Diabetes: Success, Challenges and Opportunities. Biomedicines 2022; 10:biomedicines10020331. [PMID: 35203540 PMCID: PMC8869656 DOI: 10.3390/biomedicines10020331] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 01/02/2023] Open
Abstract
There are substantial shortcomings in the drugs currently available for treatment of type 2 diabetes mellitus. The global diabetic crisis has not abated despite the introduction of new types of drugs and targets. Persistent unaddressed patient needs remain a significant factor in the quest for new leads in routine studies. Drug discovery methods in this area have followed developments in the market, contributing to a recent rise in the number of molecules. Nevertheless, troubling developments and fresh challenges are still evident. Recently, metformin, the most widely used first-line drug for diabetes, was found to contain a carcinogenic contaminant known as N-nitroso dimethylamine (NDMA). Therefore, purity and toxicity are also a big challenge for drug discovery and development. Moreover, newer drug classes against SGLT-2 illustrate both progress and difficulties. The same was true previously in the case of glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors. Furthermore, researchers must study the importance of mechanistic characteristics of novel compounds, as well as exposure-related hazardous aspects of current and newly identified protein targets, in order to identify new pharmacological molecules with improved selectivity and specificity.
Collapse
|
12
|
Cellular Senescence in Adrenocortical Biology and Its Disorders. Cells 2021; 10:cells10123474. [PMID: 34943980 PMCID: PMC8699888 DOI: 10.3390/cells10123474] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/26/2021] [Accepted: 12/06/2021] [Indexed: 01/10/2023] Open
Abstract
Cellular senescence is considered a physiological process along with aging and has recently been reported to be involved in the pathogenesis of many age-related disorders. Cellular senescence was first found in human fibroblasts and gradually explored in many other organs, including endocrine organs. The adrenal cortex is essential for the maintenance of blood volume, carbohydrate metabolism, reaction to stress and the development of sexual characteristics. Recently, the adrenal cortex was reported to harbor some obvious age-dependent features. For instance, the circulating levels of aldosterone and adrenal androgen gradually descend, whereas those of cortisol increase with aging. The detailed mechanisms have remained unknown, but cellular senescence was considered to play an essential role in age-related changes of the adrenal cortex. Recent studies have demonstrated that the senescent phenotype of zona glomerulosa (ZG) acts in association with reduced aldosterone production in both physiological and pathological aldosterone-producing cells, whereas senescent cortical-producing cells seemed not to have a suppressed cortisol-producing ability. In addition, accumulated lipofuscin formation, telomere shortening and cellular atrophy in zona reticularis cells during aging may account for the age-dependent decline in adrenal androgen levels. In adrenocortical disorders, including both aldosterone-producing adenoma (APA) and cortisol-producing adenoma (CPA), different cellular subtypes of tumor cells presented divergent senescent phenotypes, whereby compact cells in both APA and CPA harbored more senescent phenotypes than clear cells. Autonomous cortisol production from CPA reinforced a local cellular senescence that was more severe than that in APA. Adrenocortical carcinoma (ACC) was also reported to harbor oncogene-induced senescence, which compensatorily follows carcinogenesis and tumor progress. Adrenocortical steroids can induce not only a local senescence but also a periphery senescence in many other tissues. Therefore, herein, we systemically review the recent advances related to cellular senescence in adrenocortical biology and its associated disorders.
Collapse
|
13
|
Stebbins RC, Edwards JK, Plassman BL, Yang YC, Noppert GA, Haan M, Aiello AE. Immune function, cortisol, and cognitive decline & dementia in an aging latino population. Psychoneuroendocrinology 2021; 133:105414. [PMID: 34563836 PMCID: PMC8600484 DOI: 10.1016/j.psyneuen.2021.105414] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/06/2021] [Accepted: 09/05/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND The etiology of dementias and cognitive decline remain largely unknown. It is widely accepted that inflammation in the central nervous system plays a critical role in the pathogenesis of dementia. However, less is known about the role of the peripheral immune system and interactions with cortisol, though evidence suggests that these, too, may play a role. METHODS Using data from 1337 participants aged 60+ years from the Sacramento Area Latino Study of Aging (observational cohort) we investigated variation in trajectories of cognitive decline by pathogen IgG and cytokine levels. Linear mixed effects models were used to examine the association between baseline Interleukin (IL)-6, C-reactive protein, tumor necrosis factor (TNF)-α, and five persistent pathogens' IgG response and trajectories of cognition over 10 years, and to examine interactions between immune biomarkers and cortisol. Stratified cumulative incidence functions were used to assess the relation between biomarkers and incident dementia. Inverse probability weights accounted for loss-to-follow-up and confounding. RESULTS IL-6, TNF-α, and CMV IgG were statistically significantly associated with a higher log of Modified Mini-Mental State Examination errors (IL-6, β=0.0935 (95%CI: 0.055, 0.13), TNF-alpha β= 0.0944 (95%CI: 0.032, 0.157), and CMV, β= 0.0409 (95%CI: 0.013, 0.069)). Furthermore, cortisol interacted with HSV-1 and IL-6, and CRP for both cross-sectional cognitive function and rate of decline. No statistically significant relationship was detected between biomarkers and incidence of dementia. CONCLUSIONS These findings support the theory that the peripheral immune system may play a role in cognitive decline but not incident dementia. Furthermore, they identify specific markers amenable for intervention for slowing decline.
Collapse
Affiliation(s)
- Rebecca C Stebbins
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States; Social, Genetic, & Developmental Psychiatry CentreInstitute of Psychiatry, Psychology, and Neuroscience King's College London, London, United Kingdom.
| | - Jessie K Edwards
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Brenda L Plassman
- Departments of Psychiatry and Neurology, Duke University School of Medicine, Durham, NC, United States
| | - Y Claire Yang
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States; Department of Sociology, Lineberger Cancer Center University of North Carolina at Chapel Hill, United States
| | - Grace A Noppert
- Social Environment and Health, Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, United States
| | - Mary Haan
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, United States
| | - Allison E Aiello
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States; Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
14
|
Lopresti AL, Smith SJ, Drummond PD. Modulation of the hypothalamic-pituitary-adrenal (HPA) axis by plants and phytonutrients: a systematic review of human trials. Nutr Neurosci 2021; 25:1704-1730. [PMID: 33650944 DOI: 10.1080/1028415x.2021.1892253] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION The hypothalamic-pituitary-adrenal (HPA) axis plays a central role in the stress response. Plants, herbs, spices, and plant-based nutrients may influence HPA-axis activity. OBJECTIVE To evaluate randomised controlled, human trials assessing the effects of single plants or phytonutrients on HPA-axis related hormones. METHODS A systematic review of PubMed, Cochrane library, and the Cumulative Index to Nursing and Allied Health Literature was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Inclusion criteria comprised of human, randomised controlled studies with a control intervention examining the effects of a single herb, spice, plant, or extract on pre- and post-changes in blood, saliva, urine, or hair concentrations of cortisol, cortisone, corticotrophin-releasing hormone, or adrenocorticotropic hormone. Databases were searched from inception until October 2020. RESULTS Fifty-two studies were identified examining the effects of ashwagandha, Korean ginseng, St John's Wort, cannabidiol, Rhodiola rosea, curcumin, cherry juice, asparagus, Jiaogulan, Black cohosh, Siberian ginseng, Bacopa monnieri, blueberries, green tea, Caralluma fimbriata, cashew apple juice, melon, American ginseng, Ginkgo biloba, grape juice, grapefruit juice, rosella, hops, mangosteen, holy basil, and pomegranate juice. Due to significant variability in study designs, the effect of phytonutrients on HPA-axis activity in humans was unclear. The most consistent finding was a morning, cortisol-lowering effect from ashwagandha supplementation. CONCLUSION For most phytonutrients, the effects of supplementation on HPA-axis activity in humans is unclear. Before more definitive conclusions about the effects of phytonutrients on the HPA-axis can be made, further research is required.
Collapse
Affiliation(s)
- Adrian L Lopresti
- Clinical Research Australia, Perth, Australia.,College of Science, Health, Engineering and Education, Murdoch University, Perth, Australia
| | - Stephen J Smith
- Clinical Research Australia, Perth, Australia.,College of Science, Health, Engineering and Education, Murdoch University, Perth, Australia
| | - Peter D Drummond
- College of Science, Health, Engineering and Education, Murdoch University, Perth, Australia
| |
Collapse
|
15
|
Triolo F, Harber-Aschan L, Belvederi Murri M, Calderón-Larrañaga A, Vetrano DL, Sjöberg L, Marengoni A, Dekhtyar S. The complex interplay between depression and multimorbidity in late life: risks and pathways. Mech Ageing Dev 2020; 192:111383. [DOI: 10.1016/j.mad.2020.111383] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 09/14/2020] [Accepted: 10/05/2020] [Indexed: 12/20/2022]
|
16
|
Martocchia A, Gallucci M, Noale M, Maggi S, Cassol M, Stefanelli M, Postacchini D, Proietti A, Barbagallo M, Dominguez LJ, Ferri C, Desideri G, Toussan L, Pastore F, Falaschi GM, Paolisso G, Falaschi P. The cortisol burden in elderly subjects with metabolic syndrome and its association with low-grade inflammation. Aging Clin Exp Res 2020; 32:1309-1315. [PMID: 31471891 DOI: 10.1007/s40520-019-01322-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 08/13/2019] [Indexed: 02/05/2023]
Abstract
BACKGROUND Elderly people are exposed to an increased load of stressful events and neuro-hormonal stimulation is a key finding in metabolic syndrome and its related disorders. AIMS To determine the role of cortisol in elderly subjects, with or without metabolic syndrome (MetS), by means of a national multicentre observational study, AGICO (AGIng and Cortisol). METHODS From 2012 to 2017, the AGICO study enrolled n.339 subjects (aged > 65), after obtaining their informed consent. The investigators assessed a cardio-metabolic panel (including electrocardiogram, carotid ultrasonography and echocardiography), the presence of MetS (on Adult Treatment Panel III criteria), a neurological examination (including brain imaging), and cortisol activity (using a consecutive collection of diurnal and nocturnal urine). RESULTS In the patients presenting with MetS, the standardized diurnal and nocturnal cortisol excretion rates were 210.7 ± 145.5 and 173.7 ± 118.1 (mean ± standard deviation) μg/g creatinine/12 h; in those without MetS, the standardized diurnal and nocturnal cortisol excretion rates were 188.7 ± 92.7 and 144.1 ± 82.3 μg/g creatinine/12 h, respectively (nocturnal urinary cortisol in patients with MetS versus those without MetS p = 0.05, female patients with MetS vs female patients without MetS, p < 0.025). A significant positive correlation was found between the CRP levels and both the diurnal and nocturnal urinary cortisol levels with r = 0.187 (p < 0.025) and r = 0.411 (p < 0.00000001), respectively. DISCUSSION The elderly patients with MetS showed a trend towards increased standardized nocturnal cortisol excretions, with particular regard to the female subjects. CONCLUSION The positive correlation between cortisol excretion and low-grade inflammation suggests a common mechanism driving both hormonal and inflammatory changes.
Collapse
Affiliation(s)
- A Martocchia
- S. Andrea Hospital, Sapienza University of Rome, Via di Grottarossa 1035, 00189, Rome, Italy.
| | - M Gallucci
- Cognitive Impairment Centre, Local Health Authority n.2, Marca Trevigiana, Treviso, Italy
| | - M Noale
- National Research Council, Neuroscience Institute, Padua, Italy
| | - S Maggi
- National Research Council, Neuroscience Institute, Padua, Italy
| | - M Cassol
- S. Pietro Fatebenefratelli Hospital, Rome, Italy
| | - M Stefanelli
- S. Andrea Hospital, Sapienza University of Rome, Via di Grottarossa 1035, 00189, Rome, Italy
| | - D Postacchini
- Italian National Research Centre on Aging IRCCS-INRCA, Fermo, Italy
| | - A Proietti
- S. Andrea Hospital, Sapienza University of Rome, Via di Grottarossa 1035, 00189, Rome, Italy
| | - M Barbagallo
- Department of Internal Medicine and Geriatrics, University of Palermo, Palermo, Italy
| | - L J Dominguez
- Department of Internal Medicine and Geriatrics, University of Palermo, Palermo, Italy
| | - C Ferri
- University of L'Aquila, L'Aquila, Italy
| | | | - L Toussan
- S. Andrea Hospital, Sapienza University of Rome, Via di Grottarossa 1035, 00189, Rome, Italy
| | - F Pastore
- S. Andrea Hospital, Sapienza University of Rome, Via di Grottarossa 1035, 00189, Rome, Italy
| | - G M Falaschi
- S. Andrea Hospital, Sapienza University of Rome, Via di Grottarossa 1035, 00189, Rome, Italy
| | - G Paolisso
- Department of Advanced Medical and Surgical Sciences, University of Campania "L. Vanvitelli", Naples, Italy
| | - P Falaschi
- S. Andrea Hospital, Sapienza University of Rome, Via di Grottarossa 1035, 00189, Rome, Italy
| |
Collapse
|
17
|
Huang Y, Xiao Y, Liu Y, Guo M, Guo Q, Zhou F, Liu T, Su T, Xiao Y, Luo X. MicroRNA-188 regulates aging-associated metabolic phenotype. Aging Cell 2020; 19:e13077. [PMID: 31762181 PMCID: PMC6974730 DOI: 10.1111/acel.13077] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 10/09/2019] [Accepted: 10/25/2019] [Indexed: 12/24/2022] Open
Abstract
With the increasing aging population, aging-associated diseases are becoming epidemic worldwide, including aging-associated metabolic dysfunction. However, the underlying mechanisms are poorly understood. In the present study, we aimed to investigate the role of microRNA miR-188 in the aging-associated metabolic phenotype. The results showed that the expression of miR-188 increased gradually in brown adipose tissue (BAT) and inguinal white adipose tissue (iWAT) of mice during aging. MiR-188 knockout mice were resistant to the aging-associated metabolic phenotype and had higher energy expenditure. Meanwhile, adipose tissue-specific miR-188 transgenic mice displayed the opposite phenotype. Mechanistically, we identified the thermogenic-related gene Prdm16 (encoding PR domain containing 16) as the direct target of miR-188. Notably, inhibition of miR-188 expression in BAT and iWAT of aged mice by tail vein injection of antagomiR-188 ameliorated aging-associated metabolic dysfunction significantly. Taken together, our findings suggested that miR-188 plays an important role in the regulation of the aging-associated metabolic phenotype, and targeting miR-188 could be an effective strategy to prevent aging-associated metabolic dysfunction.
Collapse
Affiliation(s)
- Yan Huang
- Department of Endocrinology, Endocrinology Research CenterXiangya Hospital of Central South UniversityChangshaChina
| | - Ye Xiao
- Department of Endocrinology, Endocrinology Research CenterXiangya Hospital of Central South UniversityChangshaChina
| | - Ya Liu
- Department of Endocrinology, Endocrinology Research CenterXiangya Hospital of Central South UniversityChangshaChina
| | - Min Guo
- Department of Endocrinology, Endocrinology Research CenterXiangya Hospital of Central South UniversityChangshaChina
| | - Qi Guo
- Department of Endocrinology, Endocrinology Research CenterXiangya Hospital of Central South UniversityChangshaChina
| | - Fangliang Zhou
- Department of Biochemistry and Molecular BiologyHunan University of Chinese MedicineChangshaChina
| | - Ting Liu
- Department of EndocrinologyChangsha Central HospitalChangshaChina
| | - Tian Su
- Department of Endocrinology, Endocrinology Research CenterXiangya Hospital of Central South UniversityChangshaChina
| | - Yuzhong Xiao
- Department of Endocrinology, Endocrinology Research CenterXiangya Hospital of Central South UniversityChangshaChina
| | - Xiang‐Hang Luo
- Department of Endocrinology, Endocrinology Research CenterXiangya Hospital of Central South UniversityChangshaChina
| |
Collapse
|
18
|
Qian YX, Liu JH, Ma QH, Sun HP, Xu Y, Pan CW. Associations of sleep durations and sleep-related parameters with metabolic syndrome among older Chinese adults. Endocrine 2019; 66:240-248. [PMID: 31473919 DOI: 10.1007/s12020-019-02064-y] [Citation(s) in RCA: 12] [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: 11/14/2018] [Accepted: 08/20/2019] [Indexed: 12/15/2022]
Abstract
PURPOSE There is a lack of data on the role of sleep in the development of metabolic syndrome (MetS) in older adults. We aimed to examine the associations of sleep durations at night and other sleep-related parameters with the presence of MetS in older Chinese adults. METHODS Data of 4579 individuals aged 60 years or older from the Weitang Geriatric Disease Study were analyzed. MetS was diagnosed based on the Adult Treatment Panel III (ATP III) criteria. Information regarding sleep durations and other sleep-related parameters was collected by questionnaires. RESULTS Compared with those with daytime napping, individuals without daytime napping were at an increased risk of MetS (odds ratio [OR] = 1.23; 95% confidence interval [CI] = 1.03-1.47). Each hour increase in daytime napping increased the risk of MetS by 33% (OR = 1.33; 95% CI = 1.15-1.52). After adjusting for the effect of age and gender, a J-shaped association between sleep durations at night and the presence of MetS was observed. The risk of MetS was lowest among those who slept 7-8 h at night. Gender-stratified analysis indicated that the J-shaped relationship between sleep durations at night and MetS remained significant in men but not in women. CONCLUSIONS In older Chinese adults, sleep durations at night might have a dual effect on the risk of MetS and this effect was particularly pronounced in men. The results indicated that circadian rhythm might play an important role in the development of MetS in older populations.
Collapse
Affiliation(s)
- Yu-Xi Qian
- School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Jing-Hong Liu
- School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Qing-Hua Ma
- The 3rd People's Hospital of Xiangcheng District, Suzhou, China
| | - Hong-Peng Sun
- School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Yong Xu
- School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Chen-Wei Pan
- School of Public Health, Medical College of Soochow University, Suzhou, China.
| |
Collapse
|
19
|
Ouanes S, Popp J. High Cortisol and the Risk of Dementia and Alzheimer's Disease: A Review of the Literature. Front Aging Neurosci 2019; 11:43. [PMID: 30881301 PMCID: PMC6405479 DOI: 10.3389/fnagi.2019.00043] [Citation(s) in RCA: 265] [Impact Index Per Article: 44.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 02/13/2019] [Indexed: 01/09/2023] Open
Abstract
Introduction: Cortisol effects on the brain are exerted through two distinct receptors, inducing complex and even opposite effects on the cerebral structures implicated in the various cognitive functions. High cortisol may also have deleterious effects on the brain structures and contribute to neurodegeneration, in particular Alzheimer’s disease (AD), via different mechanisms. Objective: To examine the interrelationships between cortisol, cognitive impairment and AD. Methods: Review of the literature. Results: Clinical studies found that elevated cortisol was associated with poorer overall cognitive functioning, as well as with poorer episodic memory, executive functioning, language, spatial memory, processing speed, and social cognition; while in animals, glucocorticoid administration resulted in cognitive impairment and abnormal behavior. In cognitively healthy subjects, higher cortisol levels have been associated with an increased risk of cognitive decline and AD. Subjects with dementia and Mild Cognitive Impairment (MCI) due to AD have been found to have higher CSF cortisol levels than cognitively healthy controls. Elevated CSF cortisol may also be associated with a more rapid cognitive decline in MCI due to AD. Elevated cortisol levels have been also found in delirium. High cortisol may mediate the impact of stressful life events, high neuroticism, depression, sleep disturbances, as well as cardiovascular risk factors on cognitive performance, neurodegeneration, and cognitive decline. High cortisol may also exert neurotoxic effects on the hippocampus, and promote oxidative stress and amyloid β peptide toxicity. Further possible underlying mechanisms include the interactions of cortisol with inflammatory mediators, neurotransmitters, and growth factors. Conclusion: Elevated cortisol levels may exert detrimental effects on cognition and contribute to AD pathology. Further studies are needed to investigate cortisol-reducing and glucocorticoidreceptor modulating interventions to prevent cognitive decline.
Collapse
Affiliation(s)
- Sami Ouanes
- Department of Psychiatry, Hospital of Cery, University Hospital of Lausanne, Lausanne, Switzerland.,Department of Psychiatry, Hamad Medical Corporation, Doha, Qatar
| | - Julius Popp
- Department of Psychiatry, Hospital of Cery, University Hospital of Lausanne, Lausanne, Switzerland.,Geriatric Psychiatry, Department of Mental Health and Psychiatry, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
20
|
Increase serum cortisol in young guinea pig offspring in response to maternal iron deficiency. Nutr Res 2018; 54:69-79. [PMID: 29914669 DOI: 10.1016/j.nutres.2018.03.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 03/20/2018] [Accepted: 03/30/2018] [Indexed: 11/23/2022]
Abstract
Iron deficiency (ID) has been reported as a risk factor in the pathology of attention-deficit/hyperactivity disorder, although the mechanisms seem unclear. Previous results from our research group showed that guinea pig offspring born from ID dams were significantly more active in the Open Field Test than the controls. This behavior could potentially be associated to stress. We therefore hypothesized that maternal iron deficiency (MID) elevates the offspring serum cortisol, a biomarker of stress, during childhood and possibly at mature age. Twenty-four female guinea pigs were fed an iron-sufficient (IS) diet (114 mg/kg) or ID diet (11.7 mg/kg) during the gestation and lactation. Pups in both groups were weaned at postnatal day (PNd) 9 and given an IS diet. Hematocrit and serum cortisol levels were measured in dams at every trimester of gestation and in pups at PNd24 and 84. We found no impact of MID on dam's cortisol values. However, our findings indicate that MID increased cortisol secretion in the offspring during childhood, cortisol values being significantly elevated in ID than IS pups at PNd24 (P < .05). During adulthood (PNd84), both groups showed comparable cortisol levels. The elevated cortisol secretion observed in the offspring born from ID mothers during childhood may indicate increased stress reactivity which may have contributed to the higher level of activity when tested in a novel open environment. These findings suggest that MID can potentially act as internal stressor affecting the early development conceivably leading to increased stress levels in the children.
Collapse
|
21
|
Abstract
OBJECTIVE Childhood trauma has been associated with greater psychological and physical morbidity, including a greater risk of developing cardiovascular disease (CVD). This may partially reflect trauma-induced disturbances in how stress is later perceived and regulated. This study evaluated the associations of childhood trauma with perceived stress and hair cortisol concentrations (HCC) in a large sample of adults with coronary artery disease (CAD) and in non-CVD patients experiencing other nonfatal illnesses. Whether sex, age, or CVD status influenced these associations was also examined. METHODS A total of 1124 men and women (aged 65.2 [6.9] years) recruited from a hospital cohort completed the Childhood Trauma and Perceived Stress Questionnaires, whereas hair samples were obtained from 598 participants. Health status was confirmed via medical records. RESULTS Moderate to severe childhood trauma was experienced by 359 participants. Childhood trauma was associated with greater perceived stress levels for the past 2 years (r = .308, p = .01; β = 0.263, p < .001), but not 3-month cortisol secretion in hair. Perceived stress correlated negatively with age (r = -.241, p < .001). In secondary analyses, age moderated the relation between sexual abuse and perceived stress (β = -0.067, p = .016). Although sexual abuse was associated with greater levels of perceived stress among all participants, this relation was strongest in younger individuals. CONCLUSIONS Participants who experienced trauma in their youth reported greater levels of perceived stress, but not HCC, in late adulthood. Whether this suggests intact hypothalamic-pituitary-adrenal regulation in those exposed to childhood trauma or whether this reflects the characteristics of our sample requires further investigation.
Collapse
|
22
|
Cala MP, Agulló‐Ortuño MT, Prieto‐García E, González‐Riano C, Parrilla‐Rubio L, Barbas C, Díaz‐García CV, García A, Pernaut C, Adeva J, Riesco MC, Rupérez FJ, Lopez‐Martin JA. Multiplatform plasma fingerprinting in cancer cachexia: a pilot observational and translational study. J Cachexia Sarcopenia Muscle 2018; 9:348-357. [PMID: 29464940 PMCID: PMC5879957 DOI: 10.1002/jcsm.12270] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 10/04/2017] [Accepted: 10/24/2017] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Cachexia is a metabolic syndrome that affects up to 50-80% of cancer patients. The pathophysiology is characterized by a variable combination of reduced food intake and abnormal metabolism, including systemic inflammation and negative protein and energy balance. Despite its high clinical significance, defined diagnostic criteria and established therapeutic strategies are lacking. The 'omics' technologies provide a global view of biological systems. We hypothesize that blood-based metabolomics might identify findings in cachectic patients that could provide clues to gain knowledge on its pathophysiology, and eventually postulate new therapeutic strategies. METHODS This is a cross-sectional observational study in two cohorts of cancer patients, with and without cachexia. Patients were consecutively recruited from routine clinical practice of a General Oncology Department at '12 de Octubre' University Hospital. Selected clinical and biochemical features were collected. Blood metabolite fingerprinting was performed using three analytical platforms, gas chromatography coupled to mass spectrometry (GC-MS), capillary electrophoresis coupled to mass spectrometry (CE-MS), and liquid chromatography coupled to mass spectrometry (LC-MS). Besides, we performed pathway-based metabolite analyses to obtain more information on biological functions. RESULTS A total of 15 subjects were included in this study, 8 cachectic and 7 non-cachectic patients. Metabolomic analyses were able to correctly classify their samples in 80% (GC-MS), 97% (CE-MS), 96% [LC-MS (positive mode)], and 89% [LC-MS (negative mode)] of the cases. The most prominent metabolic alteration in plasma of cachectic patients was the decrease of amino acids and derivatives [especially arginine, tryptophan, indolelactic acid, and threonine, with 0.4-fold change (FC) compared with non-cachectic patients], along with the reduction of glycerophospholipids [mainly lysophosphatidylcholines(O-16:0) and lysophosphatidylcholines(20:3) sn-1, FC = 0.1] and sphingolipids [SM(d30:0), FC = 0.5]. The metabolite with the highest increase was cortisol (FC = 1.6). Such alterations suggest a role of the following metabolic pathways in the pathophysiology of cancer cachexia: arginine and proline metabolism; alanine, aspartate, and glutamate metabolism; phenylalanine metabolism; lysine degradation; aminoacyl-tRNA biosynthesis; fatty acid elongation in mitochondria; tricarboxylic acids cycle; among others. CONCLUSIONS These findings suggest that plasma amino acids and lipids profiling has great potential to find the mechanisms involved in the pathogenesis of cachexia. Metabolic profiling of plasma from cancer patients show differences between cachexia and non-cachexia in amino acids and lipids that might be related to mechanisms involved in its pathophysiology. A better understanding of these mechanisms might identify novel therapeutic approaches to palliate this unmet medical condition.
Collapse
Affiliation(s)
- Mónica Patricia Cala
- Centre for Metabolomic and Bioanalysis (CEMBIO), Facultad de FarmaciaUniversidad San Pablo CEUUrbanización Montepríncipe, M‐501 km 028660Boadilla del Monte, MadridSpain
- Grupo de Investigación en Química Analítica y Bioanalítica (GABIO), Department of Chemistry, Faculty of SciencesUniversidad de los AndesCra. 1 No. 18a‐10111710BogotáColombia
| | - María Teresa Agulló‐Ortuño
- Clinical & Translational Cancer Research GroupInstituto de Investigación Sanitaria Hospital 12 de Octubre (i+12)Av Córdoba s/n28041MadridSpain
| | - Elena Prieto‐García
- Clinical & Translational Cancer Research GroupInstituto de Investigación Sanitaria Hospital 12 de Octubre (i+12)Av Córdoba s/n28041MadridSpain
| | - Carolina González‐Riano
- Centre for Metabolomic and Bioanalysis (CEMBIO), Facultad de FarmaciaUniversidad San Pablo CEUUrbanización Montepríncipe, M‐501 km 028660Boadilla del Monte, MadridSpain
| | - Lucía Parrilla‐Rubio
- Medical Oncology DepartmentHospital Universitario 12 de OctubreAv de Córdoba s/n28041MadridSpain
| | - Coral Barbas
- Centre for Metabolomic and Bioanalysis (CEMBIO), Facultad de FarmaciaUniversidad San Pablo CEUUrbanización Montepríncipe, M‐501 km 028660Boadilla del Monte, MadridSpain
| | - Carmen Vanesa Díaz‐García
- Clinical & Translational Cancer Research GroupInstituto de Investigación Sanitaria Hospital 12 de Octubre (i+12)Av Córdoba s/n28041MadridSpain
| | - Antonia García
- Centre for Metabolomic and Bioanalysis (CEMBIO), Facultad de FarmaciaUniversidad San Pablo CEUUrbanización Montepríncipe, M‐501 km 028660Boadilla del Monte, MadridSpain
| | - Cristina Pernaut
- Medical Oncology DepartmentHospital Universitario 12 de OctubreAv de Córdoba s/n28041MadridSpain
| | - Jorge Adeva
- Medical Oncology DepartmentHospital Universitario 12 de OctubreAv de Córdoba s/n28041MadridSpain
| | - María Carmen Riesco
- Medical Oncology DepartmentHospital Universitario 12 de OctubreAv de Córdoba s/n28041MadridSpain
| | - Francisco Javier Rupérez
- Centre for Metabolomic and Bioanalysis (CEMBIO), Facultad de FarmaciaUniversidad San Pablo CEUUrbanización Montepríncipe, M‐501 km 028660Boadilla del Monte, MadridSpain
| | - Jose Antonio Lopez‐Martin
- Clinical & Translational Cancer Research GroupInstituto de Investigación Sanitaria Hospital 12 de Octubre (i+12)Av Córdoba s/n28041MadridSpain
- Medical Oncology DepartmentHospital Universitario 12 de OctubreAv de Córdoba s/n28041MadridSpain
| |
Collapse
|
23
|
Justice NJ. The relationship between stress and Alzheimer's disease. Neurobiol Stress 2018; 8:127-133. [PMID: 29888308 PMCID: PMC5991350 DOI: 10.1016/j.ynstr.2018.04.002] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 04/03/2018] [Accepted: 04/19/2018] [Indexed: 12/04/2022] Open
Abstract
Stress is critically involved in the development and progression of disease. From the stress of undergoing treatments to facing your own mortality, the physiological processes that stress drives have a serious detrimental effect on the ability to heal, cope and maintain a positive quality of life. This is becoming increasingly clear in the case of neurodegenerative diseases. Neurodegenerative diseases involve the devastating loss of cognitive and motor function which is stressful in itself, but can also disrupt neural circuits that mediate stress responses. Disrupting these circuits produces aberrant emotional and aggressive behavior that causes long-term care to be especially difficult. In addition, added stress drives progression of the disease and can exacerbate symptoms. In this review, I describe how neural and endocrine pathways activated by stress interact with ongoing neurodegenerative disease from both a clinical and experimental perspective.
Collapse
Affiliation(s)
- Nicholas J. Justice
- Institute of Molecular Medicine, University of Texas Health Sciences Center, Houston, TX, 77030, USA
| |
Collapse
|
24
|
Leehey M, Luo S, Sharma S, Wills AMA, Bainbridge JL, Wong PS, Simon DK, Schneider J, Zhang Y, Pérez A, Dhall R, Christine CW, Singer C, Cambi F, Boyd JT. Association of metabolic syndrome and change in Unified Parkinson's Disease Rating Scale scores. Neurology 2017; 89:1789-1794. [PMID: 28972194 DOI: 10.1212/wnl.0000000000004572] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 07/28/2017] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE To explore the association between metabolic syndrome and the Unified Parkinson's Disease Rating Scale (UPDRS) scores and, secondarily, the Symbol Digit Modalities Test (SDMT). METHODS This is a secondary analysis of data from 1,022 of 1,741 participants of the National Institute of Neurological Disorders and Stroke Exploratory Clinical Trials in Parkinson Disease Long-Term Study 1, a randomized, placebo-controlled trial of creatine. Participants were categorized as having or not having metabolic syndrome on the basis of modified criteria from the National Cholesterol Education Program Adult Treatment Panel III. Those who had the same metabolic syndrome status at consecutive annual visits were included. The change in UPDRS and SDMT scores from randomization to 3 years was compared in participants with and without metabolic syndrome. RESULTS Participants with metabolic syndrome (n = 396) compared to those without (n = 626) were older (mean [SD] 63.9 [8.1] vs 59.9 [9.4] years; p < 0.0001), were more likely to be male (75.3% vs 57.0%; p < 0.0001), and had a higher mean uric acid level (men 5.7 [1.3] vs 5.3 [1.1] mg/dL, women 4.9 [1.3] vs 3.9 [0.9] mg/dL, p < 0.0001). Participants with metabolic syndrome experienced an additional 0.6- (0.2) unit annual increase in total UPDRS (p = 0.02) and 0.5- (0.2) unit increase in motor UPDRS (p = 0.01) scores compared with participants without metabolic syndrome. There was no difference in the change in SDMT scores. CONCLUSIONS Persons with Parkinson disease meeting modified criteria for metabolic syndrome experienced a greater increase in total UPDRS scores over time, mainly as a result of increases in motor scores, compared to those who did not. Further studies are needed to confirm this finding. CLINICALTRIALSGOV IDENTIFIER NCT00449865.
Collapse
Affiliation(s)
- Maureen Leehey
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC.
| | - Sheng Luo
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - Saloni Sharma
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - Anne-Marie A Wills
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - Jacquelyn L Bainbridge
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - Pei Shieen Wong
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - David K Simon
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - Jay Schneider
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - Yunxi Zhang
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - Adriana Pérez
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - Rohit Dhall
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - Chadwick W Christine
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - Carlos Singer
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - Franca Cambi
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - James T Boyd
- From the Department of Neurology (M.L.) and Department of Clinical Pharmacy (J.L.B.), Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora; Department of Biostatistics (S.L., Y.Z.), University of Texas Health Science Center at Houston; Center for Human Experimental Therapeutics (S.S.), University of Rochester, NY; Department of Neurology (A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Pharmacy (P.S.W.), Singapore General Hospital; Department of Neurology (D.K.S.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; Department of Pathology, Anatomy, & Cell Biology (J.S.), Thomas Jefferson University, Philadelphia, PA; Department of Biostatistics (Y.Z.), School of Public Health, University of Texas Health Science Center, Houston; Department of Biostatistics (A.P.), School of Public Health, University of Texas Health Science Center at Houston-UTHealth, Austin; Department of Neurology (R.D.), University of Arkansas for Medical Sciences, Little Rock; Department of Neurology (C.W.C.), University of California San Francisco; Department of Neurology (C.S.), Leonard M. Miller School of Medicine, University of Miami, FL; Department of Neurology (F.C.), University of Pittsburgh, PA; and Department of Neurological Sciences (J.T.B.), Larner College of Medicine, University of Vermont, Burlington. Dr. Luo is currently with the Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| |
Collapse
|
25
|
Holm H, Nägga K, Nilsson ED, Ricci F, Cinosi E, Melander O, Hansson O, Bachus E, Magnusson M, Fedorowski A. N-Terminal Prosomatostatin and Risk of Vascular Dementia. Cerebrovasc Dis 2017; 44:259-265. [PMID: 28854435 DOI: 10.1159/000479940] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Accepted: 07/27/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Increased somatostatin plasma concentration has been found in patients with vascular dementia. However, it is unknown whether or not somatostatin levels may predict dementia development in the general population. To this end, we sought to assess the association of circulating N-terminal prosomatostatin (NT-proSST) with incident dementia among community-dwelling older adults. METHODS In the prospective population-based Malmö Preventive Project, 5,347 study participants (mean age: 69 ± 6years; 70% men) provided plasma for the determination of NT-proSST concentration. Of these, 373 participants (7%) were diagnosed with dementia (120 Alzheimer's disease, 83 vascular, 102 mixed, and 68 other aetiology) during a follow-up period of 4.6 ± 1.3 years. The association of NT-proSST with the risk of dementia and its subtypes was studied using multivariable-adjusted Cox regression models controlling for age, gender, body mass index, systolic blood pressure, antihypertensive treatment, smoking, diabetes, lipid levels and prevalent stroke. RESULTS Higher levels of NT-proSST were significantly associated with an increased risk of vascular dementia (hazard ratio [HR] per 1 SD: 1.29; 95% CI 1.05-1.59; p = 0.016), whereas no association was observed with Alzheimer's disease (HR per 1 SD: 0.99; 95% CI 0.81-1.20; p = 0.91), all-cause dementia (HR per 1 SD: 1.04; 95% CI 0.94-1.16; p = 0.44), and mixed dementia (HR per 1 SD: 0.98; 95% CI 0.79-1.21; p = 0.84). Levels of NT-proSST above 563 pmol/L (highest quartile) conferred distinctly increased risk of vascular dementia (HR 1.66; 95% CI 1.05-2.63; p = 0.029) compared with lower values. CONCLUSIONS Higher levels of circulating N-terminal-prosomatostatin are associated with increased incidence of vascular dementia. Our findings might be of importance for the understanding of dementia development in older adults.
Collapse
Affiliation(s)
- Hannes Holm
- Department of Clinical Sciences, Lund University, Clinical Research Center, Malmö, Sweden
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Age-dependent alterations in serum cytokines, peripheral blood mononuclear cell cytokine production, natural killer cell activity, and prostaglandin F2α. Immunol Res 2017; 65:1009-1016. [DOI: 10.1007/s12026-017-8940-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
27
|
Oh JS, Seo HS, Kim KH, Pyo H, Chung BC, Lee J. Urinary profiling of tryptophan and its related metabolites in patients with metabolic syndrome by liquid chromatography-electrospray ionization/mass spectrometry. Anal Bioanal Chem 2017; 409:5501-5512. [PMID: 28710517 DOI: 10.1007/s00216-017-0486-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 05/24/2017] [Accepted: 06/22/2017] [Indexed: 01/09/2023]
Abstract
Tryptophan (Trp) is an essential amino acid that plays an important role in protein synthesis and is a precursor of various substances related to diverse biological functions. An imbalance in Trp metabolites is associated with inflammatory diseases. The accurate and precise measurement of these compounds in biological specimens would provide meaningful information for understanding the biochemical states of various metabolic syndrome-related diseases, such as hyperlipidemia, hypertension, diabetes, and obesity. In this study, we developed a rapid, accurate, and sensitive liquid chromatography-tandem mass spectrometry-based method for the simultaneous targeted analysis of Trp and its related metabolites of the kynurenine (Kyn), serotonin, and tryptamine pathways in urine. The application of the developed method was tested using urine samples after protein precipitation. The detection limits of Trp and its metabolites were in the range of 0.01 to 0.1 μg/mL. The method was successfully validated and applied to urine samples from controls and patients with metabolic syndrome. Our results revealed high concentrations of Kyn, kynurenic acid, xanthurenic acid, and quinolinic acid as well as a high Kyn-to-Trp ratio (KTR) in patients with metabolic syndromes. The levels of urine Kyn and KTR were significantly increased in patients under 60 years old. The profiling of urinary Trp metabolites could be a useful indicator for age-related diseases including metabolic syndrome. ᅟ.
Collapse
Affiliation(s)
- Ji Sun Oh
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea.,Department of Biotechnology, Graduate School, Korea University, Seoul, Republic of Korea
| | - Hong Seong Seo
- Cardiovascular Center, Korea University Guro Hospital, Seoul, Republic of Korea
| | - Kyoung Heon Kim
- Department of Biotechnology, Graduate School, Korea University, Seoul, Republic of Korea
| | - Heesoo Pyo
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Bong Chul Chung
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Jeongae Lee
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea.
| |
Collapse
|
28
|
The ACE Gene Is Associated with Late-Life Major Depression and Age at Dementia Onset in a Population-Based Cohort. Am J Geriatr Psychiatry 2017; 25:170-177. [PMID: 27639288 DOI: 10.1016/j.jagp.2016.06.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/31/2016] [Accepted: 06/20/2016] [Indexed: 11/21/2022]
Abstract
OBJECTIVE Depression and dementia in the elderly have been suggested to share similar risk factors and pathogenetic background, and recently the authors reported that the APOEɛ4 allele is a risk factor for both disorders in the general population. The aim of the present study was to examine the influence of the well-known polymorphisms rs1799752 in the angiotensin-converting enzyme (ACE) and rs5186 in the angiotensin receptor II type 1 (AGTR1) on late-life depression and dementia in a population-based Swedish cohort of older individuals followed over 12 years. METHODS In 2000-2001, 900 individuals underwent neuropsychiatric and neuropsychological examinations. Follow-up evaluations were performed in 2005-2006 and 2009-2010, and register data on dementia to 2012 were included. Cross-sectional associations between genotypes/alleles and depression and dementia at baseline and between genotypes/alleles and depression on at least one occasion during the study period and dementia onset to 2012 were investigated. RESULTS As previously found for rs1799752 in ACE, rs5186 in AGTR1 was associated with dementia at baseline (OR: 3.25 [CI: 1.42-7.06], z = 2.90, p = 0.004). These associations became substantially weaker, or disappeared, when dementia onset to 2012 was included. For rs1799752 this could be explained by a significant association with age at onset (mean: 79.5 [SD: 6.45] years for risk-genotype carriers and 81.7 [SD: 7.12] years for carriers of other genotypes, b = -2.43, t = -2.38, df = 192, p = 0.02). When individuals with major depression on at least one occasion were analyzed, a significant association (OR: 2.14 [95% CI: 1.13-4.20], z = 2.28, p = 0.02), remaining after exclusion of dementia, with rs1799752 in ACE was found. CONCLUSION In this population-based sample of older individuals, genetic variations in ACE seem to be important both for late-life major depression and dementia.
Collapse
|
29
|
Carlesso LC, Sturgeon JA, Zautra AJ. Exploring the relationship between disease-related pain and cortisol levels in women with osteoarthritis. Osteoarthritis Cartilage 2016; 24:2048-2054. [PMID: 27374879 PMCID: PMC5406207 DOI: 10.1016/j.joca.2016.06.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 05/23/2016] [Accepted: 06/21/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVES To determine if (1) Osteoarthritis (OA)-related pain is associated with the diurnal cortisol pattern and cortisol levels; (2) the diurnal pattern of cortisol varies with severity of OA pain and (3) the association between OA pain and cortisol is mediated by daily experience variables (DEV). DESIGN In a community-based study of changes in regional and widespread pain among women with OA, participants (n = 31) completed daily diaries and collected three saliva samples daily for 7 days. Severity of OA-related pain was assessed by the validated Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) pain subscale. Multilevel regression analyses estimated associations between OA pain and diurnal cortisol levels and slopes, controlling for body mass index (BMI), medication use, time and day. Mediation analyses examined DEV as potential mediators of the association between OA pain and cortisol. RESULTS The mean age was 57 years and average BMI 31 kg/m2. Mean WOMAC pain subscale score was 8.8. Women with higher WOMAC pain scores had higher cortisol throughout the day. The estimated association of WOMAC with cortisol [β 0.083(0.02, 0.15) P = 0.009] represents a ∼9% increase in cortisol for every unit increase in WOMAC pain score. Women with WOMAC pain scores ≥9 had higher cortisol levels than those with scores <9. Examination of DEV revealed no significant mediated associations between these relationships at the daily level. CONCLUSION In women with OA, disease-related pain is positively associated with cortisol production, particularly with greater pain severity. Future studies should explore biologic mediating variables between OA pain and cortisol.
Collapse
Affiliation(s)
- Lisa C. Carlesso
- Assistant Professor, School of Rehabilitation, Faculty of Medicine, Université de Montréal, Maisonneuve-Rosemont Hospital Research Centre, Montréal, Quebec, Canada
| | - John A. Sturgeon
- Stanford University School of Medicine, Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford Systems Neuroscience and Pain Laboratory, Palo Alto, CA, USA
| | - Alex J. Zautra
- Professor, Department of Psychology, Arizona State University, Tempe, Arizona
| |
Collapse
|
30
|
Forkhead box A3 mediates glucocorticoid receptor function in adipose tissue. Proc Natl Acad Sci U S A 2016; 113:3377-82. [PMID: 26957608 DOI: 10.1073/pnas.1601281113] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Glucocorticoids (GCs) are widely prescribed anti-inflammatory agents, but their chronic use leads to undesirable side effects such as excessive expansion of adipose tissue. We have recently shown that the forkhead box protein A3 (Foxa3) is a calorie-hoarding factor that regulates the selective enlargement of epididymal fat depots and suppresses energy expenditure in a nutritional- and age-dependent manner. It has been demonstrated that Foxa3 levels are elevated in adipose depots in response to high-fat diet regimens and during the aging process; however no studies to date have elucidated the mechanisms that control Foxa3's expression in fat. Given the established effects of GCs in increasing visceral adiposity and in reducing thermogenesis, we assessed the existence of a possible link between GCs and Foxa3. Computational prediction analysis combined with molecular studies revealed that Foxa3 is regulated by the glucocorticoid receptor (GR) in preadipocytes, adipocytes, and adipose tissues and is required to facilitate the binding of the GR to its target gene promoters in fat depots. Analysis of the long-term effects of dexamethasone treatment in mice revealed that Foxa3 ablation protects mice specifically against fat accretion but not against other pathological side effects elicited by this synthetic GC in tissues such as liver, muscle, and spleen. In conclusion our studies provide the first demonstration, to our knowledge, that Foxa3 is a direct target of GC action in adipose tissues and point to a role of Foxa3 as a mediator of the side effects induced in fat tissues by chronic treatment with synthetic steroids.
Collapse
|
31
|
Pearson-Leary J, Osborne DM, McNay EC. Role of Glia in Stress-Induced Enhancement and Impairment of Memory. Front Integr Neurosci 2016; 9:63. [PMID: 26793072 PMCID: PMC4707238 DOI: 10.3389/fnint.2015.00063] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 12/05/2015] [Indexed: 12/20/2022] Open
Abstract
Both acute and chronic stress profoundly affect hippocampally-dependent learning and memory: moderate stress generally enhances, while chronic or extreme stress can impair, neural and cognitive processes. Within the brain, stress elevates both norepinephrine and glucocorticoids, and both affect several genomic and signaling cascades responsible for modulating memory strength. Memories formed at times of stress can be extremely strong, yet stress can also impair memory to the point of amnesia. Often overlooked in consideration of the impact of stress on cognitive processes, and specifically memory, is the important contribution of glia as a target for stress-induced changes. Astrocytes, microglia, and oligodendrocytes all have unique contributions to learning and memory. Furthermore, these three types of glia express receptors for both norepinephrine and glucocorticoids and are hence immediate targets of stress hormone actions. It is becoming increasingly clear that inflammatory cytokines and immunomodulatory molecules released by glia during stress may promote many of the behavioral effects of acute and chronic stress. In this review, the role of traditional genomic and rapid hormonal mechanisms working in concert with glia to affect stress-induced learning and memory will be emphasized.
Collapse
Affiliation(s)
- Jiah Pearson-Leary
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia Philadelphia, PA, USA
| | | | - Ewan C McNay
- Behavioral Neuroscience and Biology, University at Albany Albany, NY, USA
| |
Collapse
|