1
|
Reed RM, Zelek WM, Morgan BP, Whelehan G, Lockhart S, O'Rahilly S, Witard OC, Whyte MB, Goff LM. Plasma complement system markers and their association with cardiometabolic risk factors: an ethnic comparison of White European and Black African men. Am J Physiol Endocrinol Metab 2025; 328:E611-E619. [PMID: 40047170 DOI: 10.1152/ajpendo.00419.2024] [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: 10/23/2024] [Revised: 01/27/2025] [Accepted: 02/27/2025] [Indexed: 04/08/2025]
Abstract
Populations of Black African (BA) ancestry are disproportionately affected by cardiometabolic diseases, possibly due to dysregulation of the complement system. This study aimed to determine relationships between fasting complement markers and cardiometabolic risk in BA and White European (WE) men, and whether postprandial complement response differs by ethnicity. Eighty-eight BA and 97 WE men [age = 44.4 (42.0-47.6) yr, body mass index (BMI) = 29.2 ± 4.5 kg·m-2] were assessed for fasting plasma complement markers and cardiometabolic risk factors. A second cohort (n = 20 men, 10 BA) [age = 31.0 ± 1.1 yr, BMI = 27.1 (26.0-28.6) kg·m-2] men underwent a moderate-to-high-fat feeding protocol to measure postprandial plasma complement, serum insulin, plasma glucose, triacylglycerol (TAG), and nonesterified fatty acids. C4 and Factor D were lower, and iC3b was higher in BA compared with WE men. C3 and C4 were strongly associated with all adiposity markers in both ethnicities, but the WE cohort showed stronger associations between C3 and subcutaneous adipose tissue, C5 and WC, and iC3b and visceral adipose tissue compared with BA. C3 was associated with all cardiometabolic risk factors in both ethnicities. Associations between C5 and cholesterol, C4 and TAG, and terminal complement complex and (both total and LDL)-cholesterol were only observed in the WE cohort. There was a trend toward ethnic differences in postprandial Factor D (P = 0.097) and iC3b (P = 0.085). The weaker associations between the complement system markers with adiposity and lipid profiles in BA compared with WE men suggest ethnic differences in the determinants of complement production and activation, whereby adipose tissue may play a less important role in BA men.NEW & NOTEWORTHY The present study found that markers of the complement system were less strongly associated with adiposity and lipid profiles in Black African men compared with White European men, suggesting ethnic differences in the determinants of complement production and activation. In Black African men, adipose tissue may play a less important role in complement production and activation and also in the link with traditional cardiometabolic risk factors.
Collapse
Affiliation(s)
- Reuben M Reed
- Department of Nutritional Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Wioleta M Zelek
- Dementia Research Institute Cardiff, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - B Paul Morgan
- Dementia Research Institute Cardiff, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Gráinne Whelehan
- Diabetes Research Centre, University of Leicester, Leicester, United Kingdom
- NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, United Kingdom
| | - Sam Lockhart
- MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Stephen O'Rahilly
- MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Oliver C Witard
- Centre for Human & Applied Physiological Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Martin B Whyte
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Louise M Goff
- Diabetes Research Centre, University of Leicester, Leicester, United Kingdom
- NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
2
|
Zhuo Y, Fu S, Qiu Y. Regulation of the immune microenvironment by SUMO in diabetes mellitus. Front Immunol 2025; 16:1506500. [PMID: 40078991 PMCID: PMC11896877 DOI: 10.3389/fimmu.2025.1506500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
Post-translational modifications such as SUMOylation are crucial for the functionality and signal transduction of a diverse array of proteins. Analogous to ubiquitination, SUMOylation has garnered significant attention from researchers and has been implicated in the pathogenesis of various human diseases in recent years, such as cancer, neurological lesions, cardiovascular diseases, diabetes mellitus, and so on. The pathogenesis of diabetes, particularly type 1 and type 2 diabetes, has been closely associated with immune dysfunction, which constitutes the primary focus of this review. This review will elucidate the process of SUMOylation and its impact on diabetes mellitus development and associated complications, focusing on its regulatory effects on the immune microenvironment. This article summarizes various signaling pathways at both cellular and molecular levels that are implicated in these processes. Furthermore, it proposes potential new targets for drug development aimed at the prevention and treatment of diabetes mellitus based on insights gained from the SUMOylation process.
Collapse
Affiliation(s)
- Yuting Zhuo
- Department of Endocrinology and Metabolism, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, China
- The Second School of Clinical Medicine, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Shangui Fu
- The Second School of Clinical Medicine, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yue Qiu
- Department of Endocrinology and Metabolism, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, China
| |
Collapse
|
3
|
Lin J, Liang Z, Liang Y, Cao X, Tang X, Zhuang H, Yin X, Zhao D, Shen L. A systematically investigation of plasma complement and coagulation-related proteins and adiponectin in gestational diabetes mellitus by multiple reaction monitoring technology. Acta Diabetol 2025:10.1007/s00592-025-02451-0. [PMID: 39821309 DOI: 10.1007/s00592-025-02451-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 01/05/2025] [Indexed: 01/19/2025]
Abstract
BACKGROUND Gestational diabetes mellitus (GDM) is defined as a glucose intolerance resulting in hyperglycaemia of variable severity with onset during pregnancy, and is prevalent worldwide. The study of diagnostic markers of GDM in early pregnancy is important for early diagnosis and early intervention of GDM. The aim of this study was to search for biomarkers of GDM in early and mid-pregnancy using a targeted proteomics approach. METHODS Through multiple response monitoring (MRM) technology and bioinformatics analysis including machine learning, 44 proteins associated with complement and coagulation cascades, and one protein, adiponectin, which is frequently reported to be associated with GDM, were targeted for quantitative analysis, and potential biomarkers were screened. RESULTS The results showed that 7 and 6 proteins were identified as differentially expressed proteins (DEPs) between pregnant women subsequently diagnosed with GDM and controls during the first trimester, as well as between GDM cases and controls during the second trimester, respectively. Among them, C1QC and CFHR1 may serve as early predictive markers, and C1QC and adiponectin may serve as mid-term diagnostic markers. DISCUSSION Complement and coagulation-related proteins and adiponectin, have been implicated in the pathogenesis of GDM, and some of these proteins have the potential to serve as markers for the prediction or diagnosis of GDM.
Collapse
Affiliation(s)
- Jing Lin
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, P. R. China
| | - Zhiyuan Liang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, P. R. China
| | - Yi Liang
- Department of Clinical Nutrition, Affiliated Hospital of Guizhou Medical University, Guiyang, P.R. China
| | - Xueshan Cao
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, P. R. China
| | - Xiaoxiao Tang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, P. R. China
| | - Hongbin Zhuang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, P. R. China
| | - Xiaoping Yin
- Department of Obstetrics and Gynecology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, P. R. China
| | - Danqing Zhao
- Department of Obstetrics and Gynecology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, P. R. China.
| | - Liming Shen
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, P. R. China.
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, P. R. China.
| |
Collapse
|
4
|
Alic L, Dendinovic K, Papac-Milicevic N. The complement system in lipid-mediated pathologies. Front Immunol 2024; 15:1511886. [PMID: 39635529 PMCID: PMC11614835 DOI: 10.3389/fimmu.2024.1511886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 10/31/2024] [Indexed: 12/07/2024] Open
Abstract
The complement system, a coordinator and facilitator of the innate immune response, plays an essential role in maintaining host homeostasis. It promotes clearance of pathogen- and danger-associated molecular patterns, regulates adaptive immunity, and can modify various metabolic processes such as energy expenditure, lipid metabolism, and glucose homeostasis. In this review, we will focus on the intricate interplay between complement components and lipid metabolism. More precisely, we will display how alterations in the activation and regulation of the complement system affect pathological outcome in lipid-associated diseases, such as atherosclerosis, obesity, metabolic syndrome, age-related macular degeneration, and metabolic dysfunction-associated steatotic liver disease. In addition to that, we will present and evaluate underlying complement-mediated physiological mechanisms, observed both in vitro and in vivo. Our manuscript will demonstrate the clinical significance of the complement system as a bridging figure between innate immunity and lipid homeostasis.
Collapse
Affiliation(s)
- Lejla Alic
- Department of Medical Biochemistry, Faculty of Medicine, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Kristina Dendinovic
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Nikolina Papac-Milicevic
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
5
|
Rodríguez-González D, García-González M, Gómez-Bernal F, Quevedo-Abeledo JC, González-Rivero AF, González-López E, Ocejo-Vinyals JG, González-Gay MÁ, Ferraz-Amaro I. Understanding the Role of the Complement System in Insulin Resistance and Metabolic Syndrome in Patients With Rheumatoid Arthritis. J Rheumatol 2024; 51:1069-1076. [PMID: 39089839 DOI: 10.3899/jrheum.2024-0437] [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] [Accepted: 07/11/2024] [Indexed: 08/04/2024]
Abstract
OBJECTIVE The complement system has been associated with the etiopathogenesis of rheumatoid arthritis (RA). Insulin resistance (IR) and metabolic syndrome (MetS) are prevalent among patients with RA. The aim of this study was to explore the relationship between a comprehensive evaluation of the complement system and IR, as well as MetS, in patients with RA. METHODS A total of 339 nondiabetic patients with RA were recruited. Functional assays of the 3 complement pathways were assessed. Additionally, serum levels of the following individual components of the complement system were measured: C1q (classical); lectin (lectin); C2, C4, and C4b (classical lectin); factor D and properdin (alternative); C3 and C3a (common); C5, C5a, and C9 (terminal); as well as the factor I and C1 inhibitor regulators. IR and β cell function indices were calculated using the homeostatic model assessment. Criteria for MetS were applied. Multivariable linear regression analysis was performed to investigate the association between the complement system and IR in patients with RA. RESULTS Many elements of the upstream and common complement pathways, but not the functional tests of the 3 routes, correlated positively with higher levels of IR and β cell function. However, after multivariable adjustment for factors associated with IR, these relationships were lost. Conversely, the presence of MetS in patients with RA maintained a relationship with higher levels of C1q, C4, C3, properdin, and factor I after adjusting for confounders. CONCLUSION There is a positive correlation between the complement system and MetS among nondiabetic patients with RA. This association is independent of traditional IR factors.
Collapse
Affiliation(s)
- Dara Rodríguez-González
- D. Rodríguez-González, MD, F. Gómez-Bernal, MD, A.F. González-Rivero, MD, Division of Central Laboratory, Hospital Universitario de Canarias, Tenerife
| | - María García-González
- M. García-González, MD, Division of Rheumatology, Hospital Universitario de Canarias, Tenerife
| | - Fuensanta Gómez-Bernal
- D. Rodríguez-González, MD, F. Gómez-Bernal, MD, A.F. González-Rivero, MD, Division of Central Laboratory, Hospital Universitario de Canarias, Tenerife
| | - Juan C Quevedo-Abeledo
- J.C. Quevedo-Abeledo, MD, Division of Rheumatology, Hospital Doctor Negrín, Las Palmas de Gran Canaria
| | - Agustín F González-Rivero
- D. Rodríguez-González, MD, F. Gómez-Bernal, MD, A.F. González-Rivero, MD, Division of Central Laboratory, Hospital Universitario de Canarias, Tenerife
| | - Elena González-López
- E. González-López, MD, J.G. Ocejo-Vinyals, MD, Division of Immunology, Hospital Universitario Marqués de Valdecilla, IDIVAL, Santander
| | - J Gonzalo Ocejo-Vinyals
- E. González-López, MD, J.G. Ocejo-Vinyals, MD, Division of Immunology, Hospital Universitario Marqués de Valdecilla, IDIVAL, Santander
| | - Miguel Á González-Gay
- M.Á. González-Gay, MD, Division of Rheumatology, IIS-Fundación Jiménez Díaz, Madrid, and Department of Medicine and Psychiatry, University of Cantabria, Santander;
| | - Iván Ferraz-Amaro
- I. Ferraz-Amaro, MD, Division of Rheumatology, Hospital Universitario de Canarias, Tenerife, and Department of Internal Medicine, University of La Laguna (ULL), Tenerife, Spain.
| |
Collapse
|
6
|
Nadal E, Benito E, Ródenas-Navarro AM, Palanca A, Martinez-Hervas S, Civera M, Ortega J, Alabadi B, Piqueras L, Ródenas JJ, Real JT. Machine Learning Model in Obesity to Predict Weight Loss One Year after Bariatric Surgery: A Pilot Study. Biomedicines 2024; 12:1175. [PMID: 38927382 PMCID: PMC11200726 DOI: 10.3390/biomedicines12061175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/13/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024] Open
Abstract
Roux-en-Y gastric bypass (RYGB) is a treatment for severe obesity. However, many patients have insufficient total weight loss (TWL) after RYGB. Although multiple factors have been involved, their influence is incompletely known. The aim of this exploratory study was to evaluate the feasibility and reliability of the use of machine learning (ML) techniques to estimate the success in weight loss after RYGP, based on clinical, anthropometric and biochemical data, in order to identify morbidly obese patients with poor weight responses. We retrospectively analyzed 118 patients, who underwent RYGB at the Hospital Clínico Universitario of Valencia (Spain) between 2013 and 2017. We applied a ML approach using local linear embedding (LLE) as a tool for the evaluation and classification of the main parameters in conjunction with evolutionary algorithms for the optimization and adjustment of the parameter model. The variables associated with one-year postoperative %TWL were obstructive sleep apnea, osteoarthritis, insulin treatment, preoperative weight, insulin resistance index, apolipoprotein A, uric acid, complement component 3, and vitamin B12. The model correctly classified 71.4% of subjects with TWL < 30% although 36.4% with TWL ≥ 30% were incorrectly classified as "unsuccessful procedures". The ML-model processed moderate discriminatory precision in the validation set. Thus, in severe obesity, ML-models can be useful to assist in the selection of patients before bariatric surgery.
Collapse
Affiliation(s)
- Enrique Nadal
- Instituto Universitario de Ingeniería Mecánica y Biomecánica (I2MB), Universitat Politècnica de València, 46022 Valencia, Spain;
| | - Esther Benito
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28040 Madrid, Spain; (E.B.); (B.A.); (L.P.); (J.T.R.)
| | - Ana María Ródenas-Navarro
- Endocrinology and Nutrition Service, Clinical University Hospital of Valencia, 46010 Valencia, Spain; (A.M.R.-N.); (A.P.); (M.C.)
| | - Ana Palanca
- Endocrinology and Nutrition Service, Clinical University Hospital of Valencia, 46010 Valencia, Spain; (A.M.R.-N.); (A.P.); (M.C.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain;
| | - Sergio Martinez-Hervas
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28040 Madrid, Spain; (E.B.); (B.A.); (L.P.); (J.T.R.)
- Endocrinology and Nutrition Service, Clinical University Hospital of Valencia, 46010 Valencia, Spain; (A.M.R.-N.); (A.P.); (M.C.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain;
- Department of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Miguel Civera
- Endocrinology and Nutrition Service, Clinical University Hospital of Valencia, 46010 Valencia, Spain; (A.M.R.-N.); (A.P.); (M.C.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain;
| | - Joaquín Ortega
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain;
- General Surgery Service, University Hospital of Valencia, 46010 Valencia, Spain
- Department of Surgery, University of Valencia, 46010 Valencia, Spain
| | - Blanca Alabadi
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28040 Madrid, Spain; (E.B.); (B.A.); (L.P.); (J.T.R.)
- Endocrinology and Nutrition Service, Clinical University Hospital of Valencia, 46010 Valencia, Spain; (A.M.R.-N.); (A.P.); (M.C.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain;
| | - Laura Piqueras
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28040 Madrid, Spain; (E.B.); (B.A.); (L.P.); (J.T.R.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain;
- Department of Pharmacology, University of Valencia, 46010 Valencia, Spain
| | - Juan José Ródenas
- Instituto Universitario de Ingeniería Mecánica y Biomecánica (I2MB), Universitat Politècnica de València, 46022 Valencia, Spain;
| | - José T. Real
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28040 Madrid, Spain; (E.B.); (B.A.); (L.P.); (J.T.R.)
- Endocrinology and Nutrition Service, Clinical University Hospital of Valencia, 46010 Valencia, Spain; (A.M.R.-N.); (A.P.); (M.C.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain;
- Department of Medicine, University of Valencia, 46010 Valencia, Spain
| |
Collapse
|
7
|
Rajamanickam A, Dasan B, Munisankar S, Nott S, Menon PA, Ahamed Shaik F, Chinnaiyan P, Nutman TB, Babu S. Impact of Strongyloides stercoralis infection on complement activation in Type 2 diabetes mellitus: Insights from a clinical and anthelmintic intervention study. PLoS Negl Trop Dis 2024; 18:e0012048. [PMID: 38564496 PMCID: PMC10986927 DOI: 10.1371/journal.pntd.0012048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 03/05/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Numerous studies indicate a potential protective role of helminths in diabetes mellitus (DM) progression. The complement system, vital for host defense, plays a crucial role in tissue homeostasis and immune surveillance. Dysregulated complement activation is implicated in diabetic complications. We aimed to investigate the influence of the helminth, Strongyloides stercoralis (Ss) on complement activation in individuals with type 2 DM (T2D). METHODOLOGY We assessed circulating levels of complement proteins (C1q, C2, C3, C4, C4b, C5, C5a, and MBL (Lectin)) and their regulatory components (Factor B, Factor D, Factor H, and Factor I) in individuals with T2D with (n = 60) or without concomitant Ss infection (n = 58). Additionally, we evaluated the impact of anthelmintic therapy on these parameters after 6 months in Ss-infected individuals (n = 60). RESULTS Ss+DM+ individuals demonstrated reduced levels of complement proteins (C1q, C4b, MBL (Lectin), C3, C5a, and C3b/iC3b) and complement regulatory proteins (Factor B and Factor D) compared to Ss-DM+ individuals. Following anthelmintic therapy, there was a partial reversal of these levels in Ss+DM+ individuals. CONCLUSION Our findings indicate that Ss infection reduces complement activation, potentially mitigating inflammatory processes in individuals with T2D. The study underscores the complex interplay between helminth infections, complement regulation, and diabetes mellitus, offering insights into potential therapeutic avenues.
Collapse
Affiliation(s)
| | - Bindu Dasan
- NIH-NIAID-International Center for Excellence in Research, Chennai, India
| | | | - Sujatha Nott
- Infectious Diseases, Dignity Health, Chandler, Arizona, United States of America
| | | | - Fayaz Ahamed Shaik
- NIH-NIAID-International Center for Excellence in Research, Chennai, India
| | | | - Thomas B. Nutman
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Subash Babu
- NIH-NIAID-International Center for Excellence in Research, Chennai, India
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
8
|
Kulak K, Kuska K, Colineau L, Mckay M, Maziarz K, Slaby J, Blom AM, King BC. Intracellular C3 protects β-cells from IL-1β-driven cytotoxicity via interaction with Fyn-related kinase. Proc Natl Acad Sci U S A 2024; 121:e2312621121. [PMID: 38346191 PMCID: PMC10895342 DOI: 10.1073/pnas.2312621121] [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: 07/31/2023] [Accepted: 01/03/2024] [Indexed: 02/15/2024] Open
Abstract
One of the hallmarks of type 1 but also type 2 diabetes is pancreatic islet inflammation, associated with altered pancreatic islet function and structure, if unresolved. IL-1β is a proinflammatory cytokine which detrimentally affects β-cell function. In the course of diabetes, complement components, including the central complement protein C3, are deregulated. Previously, we reported high C3 expression in human pancreatic islets, with upregulation after IL-1β treatment. In the current investigation, using primary human and rodent material and CRISPR/Cas9 gene-edited β-cells deficient in C3, or producing only cytosolic C3 from a noncanonical in-frame start codon, we report a protective effect of C3 against IL-1β-induced β-cell death, that is attributed to the cytosolic fraction of C3. Further investigation revealed that intracellular C3 alleviates IL-1β-induced β-cell death, by interaction with and inhibition of Fyn-related kinase (FRK), which is involved in the response of β-cells to cytokines. Furthermore, these data were supported by increased β-cell death in vivo in a β-cell-specific C3 knockout mouse. Our data indicate that a functional, cytoprotective association exists between FRK and cytosolic C3.
Collapse
Affiliation(s)
- Klaudia Kulak
- Section of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö 214-28, Sweden
| | - Katarzyna Kuska
- Section of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö 214-28, Sweden
| | - Lucie Colineau
- Section of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö 214-28, Sweden
| | - Marina Mckay
- Section of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö 214-28, Sweden
| | - Karolina Maziarz
- Section of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö 214-28, Sweden
| | - Julia Slaby
- Section of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö 214-28, Sweden
| | - Anna M Blom
- Section of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö 214-28, Sweden
| | - Ben C King
- Section of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö 214-28, Sweden
| |
Collapse
|
9
|
Kietsiriroje N, Scott GE, Ajjan RA, Brôz J, Schroeder V, Campbell MD. Plasma levels of mannan-binding lectin-associated serine proteases are increased in type 1 diabetes patients with insulin resistance. Clin Exp Immunol 2024; 215:58-64. [PMID: 37832142 PMCID: PMC10776244 DOI: 10.1093/cei/uxad113] [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: 05/06/2023] [Revised: 07/07/2023] [Accepted: 10/12/2023] [Indexed: 10/15/2023] Open
Abstract
Activation of the lectin pathway of the complement system, as demonstrated by elevated levels of mannan-binding lectin proteins (MBL), contributes to vascular pathology in type 1 diabetes (T1D). Vascular complications are greatest in T1D individuals with concomitant insulin resistance (IR), however, whether IR amplifies activiation of the lectin pathway in T1D is unknown. We pooled pretreatment data from two RCTs and performed a cross-sectional analysis on 46 T1D individuals. We employed estimated glucose disposal rate (eGDR), a validated IR surrogate with cut-points of: <5.1, 5.1-8.7, and > 8.7 mg/kg/min to determine IR status, with lower eGDR values conferring higher degrees of IR. Plasma levels of MBL-associated proteases (MASP-1, MASP-2, and MASP-3) and their regulatory protein MAp44 were compared among eGDR classifications. In a subset of 14 individuals, we assessed change in MASPs and MAp44 following improvement in IR. We found that MASP-1, MASP-2, MASP-3, and MAp44 levels increased in a stepwise fashion across eGDR thresholds with elevated MASPs and MAp44 levels conferring greater degrees of IR. In a subset of 14 patients, improvement in IR was associated with significant reductions in MASPs, but not MAp44, levels. In conclusion, IR in T1D amplifies levels of MASP-1/2/3 and their regulator MAp44, and improvement of IR normalizes MASP-1/2/3 levels. Given that elevated levels of these proteins contribute to vascular pathology, amplification of the lectin pathway of the complement system may offer mechanistic insight into the relationship between IR and vascular complications in T1D.
Collapse
Affiliation(s)
- Noppadol Kietsiriroje
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
- Endocrinology and Metabolism Unit, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Georgia E Scott
- School of Nursing and Health Sciences, University of Sunderland, Sunderland, UK
| | - Ramzi A Ajjan
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Jan Brôz
- Department of Internal Medicine, Charles University, Prague, Czech Republic
| | - Verena Schroeder
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Matthew D Campbell
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
- School of Nursing and Health Sciences, University of Sunderland, Sunderland, UK
| |
Collapse
|
10
|
Reed RM, Whyte MB, Goff LM. Cardiometabolic disease in Black African and Caribbean populations: an ethnic divergence in pathophysiology? Proc Nutr Soc 2023:1-11. [PMID: 38230432 DOI: 10.1017/s0029665123004895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
In the UK, populations of Black African and Caribbean (BAC) ethnicity suffer higher rates of cardiometabolic disease than White Europeans (WE). Obesity, leading to increased visceral adipose tissue (VAT) and intrahepatic lipid (IHL), has long been associated with cardiometabolic risk, driving insulin resistance and defective fatty acid/lipoprotein metabolism. These defects are compounded by a state of chronic low-grade inflammation, driven by dysfunctional adipose tissue. Emerging evidence has highlighted associations between central complement system components and adipose tissue, fatty acid metabolism and inflammation; it may therefore sit at the intersection of various cardiometabolic disease risk factors. However, increasing evidence suggests an ethnic divergence in pathophysiology, whereby current theories fail to explain the high rates of cardiometabolic disease in BAC populations. Lower fasting and postprandial TAG has been reported in BAC, alongside lower VAT and IHL deposition, which are paradoxical to the high rates of cardiometabolic disease exhibited by this ethnic group. Furthermore, BAC have been shown to exhibit a more anti-inflammatory profile, with lower TNF-α and greater IL-10. In contrast, recent evidence has revealed greater complement activation in BAC compared to WE, suggesting its dysregulation may play a greater role in the high rates of cardiometabolic disease experienced by this population. This review outlines the current theories of how obesity is proposed to drive cardiometabolic disease, before discussing evidence for ethnic differences in disease pathophysiology between BAC and WE populations.
Collapse
Affiliation(s)
- Reuben M Reed
- Department of Nutritional Sciences, Faculty of Life Sciences & Medicine, King's College London, London SE1 9NH, UK
| | - Martin B Whyte
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey GU2 7WG, UK
| | - Louise M Goff
- Leicester Diabetes Research Centre, University of Leicester, Leicester, UK
| |
Collapse
|
11
|
Jin S, Eussen SJPM, Schalkwijk CG, Stehouwer CDA, van Greevenbroek MMJ. Plasma factor D is cross-sectionally associated with low-grade inflammation, endothelial dysfunction and cardiovascular disease: The Maastricht study. Atherosclerosis 2023; 377:60-67. [PMID: 37406499 DOI: 10.1016/j.atherosclerosis.2023.06.079] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 06/15/2023] [Accepted: 06/15/2023] [Indexed: 07/07/2023]
Abstract
BACKGROUND AND AIMS The complement system, particularly the alternative complement pathway, may contribute to vascular damage and development of cardiovascular disease (CVD). We investigated the association of factor D, the rate-limiting protease in alternative pathway activation, with adverse cardiovascular outcomes. METHODS In 2947 participants (50.6% men, 59.9 ± 8.2 years, 26.5% type 2 diabetes [T2D], oversampled) we measured markers of low-grade inflammation (LGI, composite score, in SD) and, endothelial dysfunction (ED, composite score, in SD), carotid intima-media thickness (cIMT, μm), ankle-brachial index (ABI), CVD (yes/no) and plasma concentrations of factor D (in SD). Associations were estimated using multiple linear and logistic regression, adjusting for demographic, lifestyle, and dietary factors. RESULTS Factor D (per SD) significantly associated with LGI (0.171 SD [0.137; 0.205]), ED (0.158 SD [0.123; 0.194]) and CVD (OR 1.15 [1.04; 1.27]) but not significantly with cIMT (-6.62 μm [-13.51; 0.27]) or ABI (-0.003 [-0.007; 0.001]). Interaction analyses show that factor D more strongly associated with ED in non-diabetes (0.237 SD [0.189; 0.285] than in T2D (0.095 SD [0.034; 0.157]), pinteraction <0.05. These results were largely corroborated by additional analyses with C3 and C3a. In contrast, factor D inversely associated with cIMT in non-diabetes (-13.37 μm [-21.84; -4.90]), but not in T2D (4.49 [-7.91; 16.89]), pinteraction <0.05. CONCLUSIONS Plasma factor D is independently associated with LGI, ED, and prevalent CVD but not with ABI or cIMT. Hence, greater plasma factor D concentration in CVD may potentially induce complement activation which, in turn, might contribute to further disease progression via a process that may involve inflammation and endothelial dysfunction but was not directly related to atherosclerosis or arterial injury. The observation that, in participants without diabetes, factor D associated with worse ED but smaller cIMT warrants further investigation.
Collapse
Affiliation(s)
- Shunxin Jin
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, the Netherlands
| | - Simone J P M Eussen
- Department of Epidemiology, CARIM School for Cardiovascular Diseases, the Netherlands; CAPHRI School for Public Health and Primary Care, Maastricht University and Maastricht University Medical Centre, the Netherlands
| | - Casper G Schalkwijk
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, the Netherlands
| | - Coen D A Stehouwer
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, the Netherlands
| | | |
Collapse
|
12
|
Jiang T, Li Y, He S, Huang N, Du M, Zhai Q, Pu K, Wu M, Yan C, Ma Z, Wang Q. Reprogramming astrocytic NDRG2/NF-κB/C3 signaling restores the diabetes-associated cognitive dysfunction. EBioMedicine 2023; 93:104653. [PMID: 37329577 DOI: 10.1016/j.ebiom.2023.104653] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 04/14/2023] [Accepted: 05/31/2023] [Indexed: 06/19/2023] Open
Abstract
BACKGROUND Dementia is a serious complication in patients with diabetes-associated cognitive dysfunction (DACD). In this study, we aim to explore the protective effect of exercise on DACD in diabetic mice, and the role of NDRG2 as a potential guarder for reversing the pathological structure of neuronal synapses. METHODS Seven weeks of standardized exercise at moderate intensity was carried out using an animal treadmill in the vehicle + Run and STZ + Run groups. Based on quantitative transcriptome and tandem mass tag (TMT) proteome sequencing, weighted gene co-expression analysis (WGCNA) and gene set enrichment analysis (GSEA) were used to investigate the activation of complement cascades to injury neuronal synaptic plasticity. Golgi staining, Western blotting, immunofluorescence staining, and electrophysiology were used to verify the reliability of sequencing data. The role of NDRG2 was assessed by overexpressing or inhibiting the NDRG2 gene in vivo. Moreover, we estimated the cognitive function in diabetic or normal patients using DSST scores. FINDINGS Exercise reversed the injury of neuronal synaptic plasticity and the downregulation of astrocytic NDRG2 in diabetic mice, which succeeded in attenuating DACD. The deficiency of NDRG2 aggravated the activation of complement C3 by accelerating the phosphorylation of NF-κB, ultimately leading to synaptic injury and cognitive dysfunction. Conversely, the overexpression of NDRG2 promoted astrocytic remodeling by inhibiting complement C3, thus attenuating synaptic injury and cognitive dysfunction. Meanwhile, C3aR blockade rescued dendritic spines loss and cognitive deficits in diabetic mice. Moreover, the average DSST score of diabetic patients was significantly lower than that of non-diabetic peers. Levels of complement C3 in human serum were elevated in diabetic patients compared to those in non-diabetic patients. INTERPRETATION Our findings illustrate the effectiveness and integrative mechanism of NDRG2-induced improvement of cognition from a multi-omics perspective. Additionally, they confirm that the expression of NDRG2 is closely related to cognitive function in diabetic mice and the activation of complement cascades accelerated impairment of neuronal synaptic plasticity. NDRG2 acts as a regulator of astrocytic-neuronal interaction via NF-κB/C3/C3aR signaling to restore synaptic function in diabetic mice. FUNDING This study was supported by the National Natural Science Foundation of China (No. 81974540, 81801899, 81971290), the Key Research and Development Program of Shaanxi (Program No. 2022ZDLSF02-09) and Fundamental Research Funds for the Central Universities (Grant No. xzy022019020).
Collapse
Affiliation(s)
- Tao Jiang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China; Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi, China
| | - Yansong Li
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Shuxuan He
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Ning Huang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China; Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, China; Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, China
| | - Mengyu Du
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Qian Zhai
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Kairui Pu
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Meiyan Wu
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Chaoying Yan
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Zhi Ma
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Qiang Wang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China.
| |
Collapse
|
13
|
Jiang J, Wang H, Liu K, He S, Li Z, Yuan Y, Yu K, Long P, Wang J, Diao T, Zhang X, He M, Guo H, Wu T. Association of Complement C3 With Incident Type 2 Diabetes and the Mediating Role of BMI: A 10-Year Follow-Up Study. J Clin Endocrinol Metab 2023; 108:736-744. [PMID: 36205019 DOI: 10.1210/clinem/dgac586] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 10/01/2022] [Indexed: 11/19/2022]
Abstract
CONTEXT Impairment of immune and inflammatory homeostasis is reported to be one of the causal factors of diabetes. However, the association of complement C3 levels with incident diabetes in humans remains unclear. OBJECTIVE This study aimed to examine the association between C3 levels and incident type 2 diabetes mellitus (T2DM), and further explore the potential mediating role of body mass index (BMI) in C3-T2DM associations. METHODS We determined serum C3 levels of 2662 nondiabetic middle-aged and elderly (64.62 ± 7.25 years) individuals from the Dongfeng-Tongji cohort at baseline. Cox regression was employed to examine the incidence of T2DM in relationship to C3 levels during 10 years of follow-up. Mediation analysis was further applied to assess potential effect of BMI on the C3-T2DM associations. RESULTS Overall, 711 (26.7%) participants developed T2DM during 23 067 person-years of follow-up. Higher serum C3 was significantly associated with higher risk of incident T2DM after full adjustment (HR [95% CI] = 1.16 [1.05, 1.27]; per SD higher). Compared with the first quartile of C3 levels, the HR in the fourth quartile was 1.52 (95% CI = [1.14, 2.02]; Ptrend = 0.029). Robust significant linear dose-response relationship was observed between C3 levels and BMI (Poverall < 0.001, Pnonlinear = 0.96). Mediation analyses indicated that BMI might mediate 41.0% of the associations between C3 and T2DM. CONCLUSION The present prospective study revealed that C3 could be an early biomarker for incident T2DM, and that BMI might play a potential mediating role in the C3-T2DM associations, which provided clues for the pathogenesis of diabetes.
Collapse
Affiliation(s)
- Jing Jiang
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hao Wang
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Kang Liu
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- School of Public Health, Guangzhou Medical University, Guangzhou 511436, China
| | - Shiqi He
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhaoyang Li
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yu Yuan
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Kuai Yu
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Pinpin Long
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jing Wang
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tingyue Diao
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaomin Zhang
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Meian He
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Huan Guo
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tangchun Wu
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
14
|
Jin S, Reesink KD, Kroon AA, de Galan B, van der Kallen CJH, Wesselius A, Schalkwijk CG, Stehouwer CDA, van Greevenbroek MMJ. Complement factors D and C3 cross-sectionally associate with arterial stiffness, but not independently of metabolic risk factors: The Maastricht Study. J Hypertens 2022; 40:2161-2170. [PMID: 35881455 DOI: 10.1097/hjh.0000000000003237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Arterial stiffness predicts cardiovascular outcomes. The complement system, particularly the alternative complement pathway, has been implicated in cardiovascular diseases. We herein investigated the associations of factor D, the rate-limiting protease of the alternative pathway, and C3, the central complement component, with arterial stiffness. METHODS In 3019 population-based participants (51.9% men, 60.1 ± 8.2 years, 27.7% type 2 diabetes [T2D], oversampled]), we measured carotid-femoral pulse wave velocity (cfPWV), carotid distensibility coefficient (DC) and carotid Young's elastic modulus (YEM), and plasma concentrations of factors D and C3. We conducted multiple linear regression to investigate the association of factors D and C3 (main independent variables, standardized) with cfPWV (primary outcome) and DC and YEM (secondary outcomes), adjusted for potential confounders. RESULTS Per SD higher factors D and C3, cfPWV was 0.41 m/s [95% confidence interval: 0.34; 0.49] and 0.33 m/s [0.25; 0.41] greater, respectively. These associations were substantially attenuated when adjusted for age, sex, education, mean arterial pressure, and heart rate (0.08 m/s [0.02; 0.15] and 0.11 m/s [0.05; 0.18], respectively), and were not significant when additionally adjusted for T2D, waist circumference and additional cardiovascular risk factors (0.06 m/s [-0.01; 0.13] and 0.01 m/s [-0.06; 0.09], respectively). Results were comparable for carotid YEM and DC. In persons with T2D, but not in those without, the association between factors D and cfPWV was significant in the fully adjusted model (0.14 m/s, [0.01; 0.27], P = 0.038, Pinteraction < 0.05). CONCLUSION The strong association of plasma factors D and C3 with arterial stiffness in this population-based cohort was not independent of T2D and other metabolic risk factors. Our data suggest that a possible causal pathway starting from alternative complement activation may via hypertension and T2D contribute to greater arterial stiffness.
Collapse
Affiliation(s)
- Shunxin Jin
- CARIM School for Cardiovascular Diseases
- Department of Internal Medicine
| | - Koen D Reesink
- CARIM School for Cardiovascular Diseases
- Department of Biomedical Technology
| | - Abraham A Kroon
- CARIM School for Cardiovascular Diseases
- Department of Internal Medicine
| | | | | | - Anke Wesselius
- Department of Genetics
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University and Maastricht University Medical Centre, Maastricht, The Netherlands
| | | | | | | |
Collapse
|
15
|
Roy B, Runa SA. SARS-CoV-2 infection and diabetes: Pathophysiological mechanism of multi-system organ failure. World J Virol 2022; 11:252-274. [PMID: 36188734 PMCID: PMC9523319 DOI: 10.5501/wjv.v11.i5.252] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/25/2022] [Accepted: 08/01/2022] [Indexed: 02/05/2023] Open
Abstract
Since the discovery of the coronavirus disease 2019 outbreak, a vast majority of studies have been carried out that confirmed the worst outcome of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in people with preexisting health conditions, including diabetes, obesity, hypertension, cancer, and cardiovascular diseases. Likewise, diabetes itself is one of the leading causes of global public health concerns that impose a heavy global burden on public health as well as socio-economic development. Both diabetes and SARS-CoV-2 infection have their independent ability to induce the pathogenesis and severity of multi-system organ failure, while the co-existence of these two culprits can accelerate the rate of disease progression and magnify the severity of the disease. However, the exact pathophysiology of multi-system organ failure in diabetic patients after SARS-CoV-2 infection is still obscure. This review summarized the organ-specific possible molecular mechanisms of SARS-CoV-2 and diabetes-induced pathophysiology of several diseases of multiple organs, including the lungs, heart, kidneys, brain, eyes, gastrointestinal system, and bones, and sub-sequent manifestation of multi-system organ failure.
Collapse
Affiliation(s)
- Bipradas Roy
- Department of Physiology, Wayne State University, Detroit, MI 48201, United States
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health System, Detroit, MI 48202, United States
| | - Sadia Afrin Runa
- Department of Biotechnology and Genetic Engineering, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| |
Collapse
|
16
|
The landscape of immune cell infiltration in the glomerulus of diabetic nephropathy: evidence based on bioinformatics. BMC Nephrol 2022; 23:303. [PMID: 36064366 PMCID: PMC9442983 DOI: 10.1186/s12882-022-02906-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 07/29/2022] [Indexed: 11/10/2022] Open
Abstract
Background Increasing evidence suggests that immune cell infiltration contributes to the pathogenesis and progression of diabetic nephropathy (DN). We aim to unveil the immune infiltration pattern in the glomerulus of DN and provide potential targets for immunotherapy. Methods Infiltrating percentage of 22 types of immune cell in the glomerulus tissues were estimated by the CIBERSORT algorithm based on three transcriptome datasets mined from the GEO database. Differentially expressed genes (DEGs) were identified by the “limma” package. Then immune-related DEGs were identified by intersecting DEGs with immune-related genes (downloaded from Immport database). The protein–protein interactions of Immune-related DEGs were explored using the STRING database and visualized by Cytoscape. The enrichment analyses for KEGG pathways and GO terms were carried out by the gene set enrichment analysis (GSEA) method. Results 11 types of immune cell were revealed to be significantly altered in the glomerulus tissues of DN (Up: B cells memory, T cells gamma delta, NK cells activated, Macrophages.M1, Macrophages M2, Dendritic cells resting, Mast cells resting; Down: B cells naive, NK cells resting, Mast cells activated, Neutrophils). Several pathways related to immune, autophagy and metabolic process were significantly activated. Moreover, 6 hub genes with a medium to strong correlation with renal function (eGFR) were identified (SERPINA3, LTF, C3, PTGDS, EGF and ALB). Conclusion In the glomerulus of DN, the immune infiltration pattern changed significantly. A complicated and tightly regulated network of immune cells exists in the pathological of DN. The hub genes identified here will facilitate the development of immunotherapy. Supplementary Information The online version contains supplementary material available at 10.1186/s12882-022-02906-4.
Collapse
|
17
|
Bernea EG, Suica VI, Uyy E, Cerveanu-Hogas A, Boteanu RM, Ivan L, Ceausu I, Mihai DA, Ionescu-Tîrgoviște C, Antohe F. Exosome Proteomics Reveals the Deregulation of Coagulation, Complement and Lipid Metabolism Proteins in Gestational Diabetes Mellitus. Molecules 2022; 27:molecules27175502. [PMID: 36080270 PMCID: PMC9457917 DOI: 10.3390/molecules27175502] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/22/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022] Open
Abstract
Exosomes are small extracellular vesicles with a variable protein cargo in consonance with cell origin and pathophysiological conditions. Gestational diabetes mellitus (GDM) is characterized by different levels of chronic low-grade inflammation and vascular dysfunction; however, there are few data characterizing the serum exosomal protein cargo of GDM patients and associated signaling pathways. Eighteen pregnant women were enrolled in the study: 8 controls (CG) and 10 patients with GDM. Blood samples were collected from patients, for exosomes’ concentration. Protein abundance alterations were demonstrated by relative mass spectrometric analysis and their association with clinical parameters in GDM patients was performed using Pearson’s correlation analysis. The proteomics analysis revealed 78 significantly altered proteins when comparing GDM to CG, related to complement and coagulation cascades, platelet activation, prothrombotic factors and cholesterol metabolism. Down-regulation of Complement C3 (C3), Complement C5 (C5), C4-B (C4B), C4b-binding protein beta chain (C4BPB) and C4b-binding protein alpha chain (C4BPA), and up-regulation of C7, C9 and F12 were found in GDM. Our data indicated significant correlations between factors involved in the pathogenesis of GDM and clinical parameters that may improve the understanding of GDM pathophysiology. Data are available via ProteomeXchange with identifier PXD035673.
Collapse
Affiliation(s)
- Elena G. Bernea
- “Prof. N. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 020474 Bucharest, Romania
| | - Viorel I. Suica
- Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania
| | - Elena Uyy
- Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania
| | - Aurel Cerveanu-Hogas
- Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania
| | - Raluca M. Boteanu
- Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania
| | - Luminita Ivan
- Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania
| | - Iuliana Ceausu
- University of Medicine and Pharmacy “Carol Davila”, 020021 Bucharest, Romania
- “Dr. I. Cantacuzino” Hospital, 020475 Bucharest, Romania
| | - Doina A. Mihai
- “Prof. N. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 020474 Bucharest, Romania
- University of Medicine and Pharmacy “Carol Davila”, 020021 Bucharest, Romania
| | - Constantin Ionescu-Tîrgoviște
- “Prof. N. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 020474 Bucharest, Romania
- University of Medicine and Pharmacy “Carol Davila”, 020021 Bucharest, Romania
| | - Felicia Antohe
- Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania
- Correspondence: ; Tel.: +40213194518
| |
Collapse
|
18
|
Stanimirovic J, Radovanovic J, Banjac K, Obradovic M, Essack M, Zafirovic S, Gluvic Z, Gojobori T, Isenovic ER. Role of C-Reactive Protein in Diabetic Inflammation. Mediators Inflamm 2022; 2022:3706508. [PMID: 35620114 PMCID: PMC9129992 DOI: 10.1155/2022/3706508] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/20/2022] [Accepted: 04/29/2022] [Indexed: 01/08/2023] Open
Abstract
Even though type 2 diabetes mellitus (T2DM) represents a worldwide chronic health issue that affects about 462 million people, specific underlying determinants of insulin resistance (IR) and impaired insulin secretion are still unknown. There is growing evidence that chronic subclinical inflammation is a triggering factor in the origin of T2DM. Increased C-reactive protein (CRP) levels have been linked to excess body weight since adipocytes produce tumor necrosis factor α (TNF-α) and interleukin 6 (IL-6), which are pivotal factors for CRP stimulation. Furthermore, it is known that hepatocytes produce relatively low rates of CRP in physiological conditions compared to T2DM patients, in which elevated levels of inflammatory markers are reported, including CRP. CRP also participates in endothelial dysfunction, the production of vasodilators, and vascular remodeling, and increased CRP level is closely associated with vascular system pathology and metabolic syndrome. In addition, insulin-based therapies may alter CRP levels in T2DM. Therefore, determining and clarifying the underlying CRP mechanism of T2DM is imperative for novel preventive and diagnostic procedures. Overall, CRP is one of the possible targets for T2DM progression and understanding the connection between insulin and inflammation may be helpful in clinical treatment and prevention approaches.
Collapse
Affiliation(s)
- Julijana Stanimirovic
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Jelena Radovanovic
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Katarina Banjac
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Milan Obradovic
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Magbubah Essack
- King Abdullah University of Science and Technology (KAUST), Computer, Electrical, and Mathematical Sciences and Engineering (CEMSE) Division, Computational Bioscience Research Center (CBRC), Thuwal, Saudi Arabia
| | - Sonja Zafirovic
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Zoran Gluvic
- University Clinical-Hospital Centre Zemun-Belgrade, Clinic of Internal Medicine, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Takashi Gojobori
- King Abdullah University of Science and Technology (KAUST), Computer, Electrical, and Mathematical Sciences and Engineering (CEMSE) Division, Computational Bioscience Research Center (CBRC), Thuwal, Saudi Arabia
| | - Esma R. Isenovic
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
19
|
Mills EL, Harmon C, Jedrychowski MP, Xiao H, Gruszczyk AV, Bradshaw GA, Tran N, Garrity R, Laznik-Bogoslavski D, Szpyt J, Prendeville H, Lynch L, Murphy MP, Gygi SP, Spiegelman BM, Chouchani ET. Cysteine 253 of UCP1 regulates energy expenditure and sex-dependent adipose tissue inflammation. Cell Metab 2022; 34:140-157.e8. [PMID: 34861155 PMCID: PMC8732317 DOI: 10.1016/j.cmet.2021.11.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 09/15/2021] [Accepted: 11/08/2021] [Indexed: 01/07/2023]
Abstract
Uncoupling protein 1 (UCP1) is a major regulator of brown and beige adipocyte energy expenditure and metabolic homeostasis. However, the widely employed UCP1 loss-of-function model has recently been shown to have a severe deficiency in the entire electron transport chain of thermogenic fat. As such, the role of UCP1 in metabolic regulation in vivo remains unclear. We recently identified cysteine-253 as a regulatory site on UCP1 that elevates protein activity upon covalent modification. Here, we examine the physiological importance of this site through the generation of a UCP1 cysteine-253-null (UCP1 C253A) mouse, a precise genetic model for selective disruption of UCP1 in vivo. UCP1 C253A mice exhibit significantly compromised thermogenic responses in both males and females but display no measurable effect on fat accumulation in an obesogenic environment. Unexpectedly, we find that a lack of C253 results in adipose tissue redox stress, which drives substantial immune cell infiltration and systemic inflammatory pathology in adipose tissues and liver of male, but not female, mice. Elevation of systemic estrogen reverses this male-specific pathology, providing a basis for protection from inflammation due to loss of UCP1 C253 in females. Together, our results establish the UCP1 C253 activation site as a regulator of acute thermogenesis and sex-dependent tissue inflammation.
Collapse
Affiliation(s)
- Evanna L Mills
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Cathal Harmon
- Department of Immunology, Harvard Medical School, Boston, MA, USA; Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, USA
| | - Mark P Jedrychowski
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Haopeng Xiao
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Anja V Gruszczyk
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK
| | - Gary A Bradshaw
- Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA
| | - Nhien Tran
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ryan Garrity
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - John Szpyt
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Hannah Prendeville
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Lydia Lynch
- Department of Immunology, Harvard Medical School, Boston, MA, USA; Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, USA; School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Michael P Murphy
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK; Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Bruce M Spiegelman
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Edward T Chouchani
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
20
|
Babu SN, Govindarajan S, Noor A. Aloe vera and its two bioactive constituents in alleviation of diabetes -proteomic & mechanistic insights. JOURNAL OF ETHNOPHARMACOLOGY 2021; 280:114445. [PMID: 34303804 DOI: 10.1016/j.jep.2021.114445] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 07/14/2021] [Accepted: 07/20/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Aloe barbadensis Miller, commonly known as Aloe vera has been used since time immemorial for treatment of various diseases such as cancer, inflammatory disorders, diabetes, wound healing etc. AIM: Diabetes mellitus is a complex disorder and understanding the molecular mechanisms involved is a key to identify different markers for early diagnosis of the disease. The proteomic approach offers a plethora of opportunities to identify markers and targets involved in pathogenesis of diabetes. The present study was undertaken to understand the mechanism of action of Aloe vera and its two constituents (Carbohydrates and Polypeptides) in the alleviation of diabetes in streptozotocin-induced diabetic rats through a proteomics approach. METHODS Different groups of rats were fed with Aloe vera extract, carbohydrate fraction and peptide/polypeptide fraction for three weeks. The diabetic rats fed with Aloe vera and its two fractions restored the glucose and insulin levels to normal. The plasma of the rats was depleted with IgG and albumin and proteomic analysis was carried out. Apolipoproteins (dyslipidemia), complement factors (inflammatory pathways), zonulin (intestinal permeability), anti-oxidant related proteins were selected in this study as these are involved in the progression of diabetes. RESULTS It was observed that Aloe vera extract is involved in the alleviation of diabetes through these pathways while the carbohydrate fraction alleviates diabetes through an anti-oxidant mechanism and glucose uptake while the polypeptide fraction alleviates diabetes through the restoration of intestinal permeability by reduced zonulin levels. CONCLUSION The constituents of Aloe vera works different pathways involved in diabetes and the synergistic effect of these constituents make Aloe vera extract a prospective candidate, which can alleviate diabetes through regulation of the pathways involved in the progression of diabetes.
Collapse
Affiliation(s)
- Spoorthy N Babu
- Centre for Bio Separation Technology (CBST), Vellore Institute of Technology (VIT), Vellore-632 014, Tamil Nadu, India
| | - S Govindarajan
- Centre for Bio Separation Technology (CBST), Vellore Institute of Technology (VIT), Vellore-632 014, Tamil Nadu, India
| | - Ayesha Noor
- Centre for Bio Separation Technology (CBST), Vellore Institute of Technology (VIT), Vellore-632 014, Tamil Nadu, India.
| |
Collapse
|
21
|
King BC, Blom AM. Complement in metabolic disease: metaflammation and a two-edged sword. Semin Immunopathol 2021; 43:829-841. [PMID: 34159399 PMCID: PMC8613079 DOI: 10.1007/s00281-021-00873-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 05/23/2021] [Indexed: 01/28/2023]
Abstract
We are currently experiencing an enduring global epidemic of obesity and diabetes. It is now understood that chronic low-grade tissue inflammation plays an important role in metabolic disease, brought upon by increased uptake of a so-called Western diet, and a more sedentary lifestyle. Many evolutionarily conserved links exist between metabolism and the immune system, and an imbalance in this system induced by chronic over-nutrition has been termed 'metaflammation'. The complement system is an important and evolutionarily ancient part of innate immunity, but recent work has revealed that complement not only is involved in the recognition of pathogens and induction of inflammation, but also plays important roles in cellular and tissue homeostasis. Complement can therefore contribute both positively and negatively to metabolic control, depending on the nature and anatomical site of its activity. This review will therefore focus on the interactions of complement with mechanisms and tissues relevant for metabolic control, obesity and diabetes.
Collapse
Affiliation(s)
- B C King
- Department of Translational Medicine, Lund University, Lund, Sweden.
| | - A M Blom
- Department of Translational Medicine, Lund University, Lund, Sweden
| |
Collapse
|
22
|
Xin Y, Hertle E, van der Kallen CJH, Vogelzangs N, Arts ICW, Schalkwijk CG, Stehouwer CDA, van Greevenbroek MMJ. C3 and alternative pathway components are associated with an adverse lipoprotein subclass profile: The CODAM study. J Clin Lipidol 2021; 15:311-319. [PMID: 33612457 DOI: 10.1016/j.jacl.2021.01.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 12/04/2020] [Accepted: 01/31/2021] [Indexed: 11/23/2022]
Abstract
BACKGROUND Plasma lipoproteins contain heterogeneous subclasses. Previous studies on the associations of the complement system with lipids and lipoproteins are mainly limited to the major lipid classes, and associations of complement with lipoprotein subclass characteristics remain unknown. OBJECTIVE We investigated the associations of C3 and other components of the alternative complement pathway with plasma lipoprotein subclass profile. METHODS Plasma complement concentrations (complement component 3 [C3], properdin, factor H, factor D, MASP-3, C3a, Bb), and lipoprotein subclass profile (as measured by nuclear magnetic resonance spectroscopy) were obtained in 523 participants (59.6 ± 6.9 years, 60.8% men) of the Cohort on Diabetes and Atherosclerosis Maastricht (CODAM) study. Multiple linear regression was used to investigate the associations of C3 (primary determinant) and other alternative pathway components (secondary determinants) with characteristics (particle concentration and size [main outcomes], and lipid contents [secondary outcomes]) of 14 lipoprotein subclasses, ranging from extremely large VLDL to small HDL (all standardized [std] values). RESULTS Participants with higher C3 concentrations had more circulating VLDL (stdβs ranging from 0.27 to 0.36), IDL and LDL (stdβs ranging from 0.14 to 0.17), and small HDL (stdβ = 0.21). In contrast, they had fewer very large and large HDL particles (stdβs = -0.36). In persons with higher C3 concentrations, all lipoprotein subclasses were enriched in triglycerides. Similar but weaker associations were observed for properdin, factor H, factor D, and MASP-3, but not for C3a and Bb. CONCLUSIONS The alternative complement pathway, and most prominently C3, is associated with an adverse lipoprotein subclass profile that is characterized by more triglyceride-enriched lipoproteins but fewer large HDL.
Collapse
Affiliation(s)
- Ying Xin
- Department of Internal Medicine, Maastricht University Medical Centre and CARIM School for Cardiovascular Diseases, Maastricht University, the Netherlands; Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Elisabeth Hertle
- Department of Internal Medicine, Maastricht University Medical Centre and CARIM School for Cardiovascular Diseases, Maastricht University, the Netherlands
| | - Carla J H van der Kallen
- Department of Internal Medicine, Maastricht University Medical Centre and CARIM School for Cardiovascular Diseases, Maastricht University, the Netherlands
| | - Nicole Vogelzangs
- Department of Epidemiology, CARIM School for Cardiovascular Diseases, Maastricht University, the Netherlands; Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, the Netherlands
| | - Ilja C W Arts
- Department of Epidemiology, CARIM School for Cardiovascular Diseases, Maastricht University, the Netherlands; Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, the Netherlands
| | - Casper G Schalkwijk
- Department of Internal Medicine, Maastricht University Medical Centre and CARIM School for Cardiovascular Diseases, Maastricht University, the Netherlands
| | - Coen D A Stehouwer
- Department of Internal Medicine, Maastricht University Medical Centre and CARIM School for Cardiovascular Diseases, Maastricht University, the Netherlands
| | - Marleen M J van Greevenbroek
- Department of Internal Medicine, Maastricht University Medical Centre and CARIM School for Cardiovascular Diseases, Maastricht University, the Netherlands.
| |
Collapse
|
23
|
Abstract
A growing body of evidence indicates that obesity is strongly and independently associated with adverse outcomes of COVID-19, including death. By combining emerging knowledge of the pathological processes involved in COVID-19 with insights into the mechanisms underlying the adverse health consequences of obesity, we present some hypotheses regarding the deleterious impact of obesity on the course of COVID-19. These hypotheses are testable and could guide therapeutic and preventive interventions. As obesity is now almost ubiquitous and no vaccine for COVID-19 is currently available, even a modest reduction in the impact of obesity on mortality and morbidity from this viral infection could have profound consequences for public health.
Collapse
Affiliation(s)
- Sam M Lockhart
- MRC Metabolic Diseases Unit, Wellcome Trust-Medical Research Council Institute of Metabolic Science and NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Stephen O'Rahilly
- MRC Metabolic Diseases Unit, Wellcome Trust-Medical Research Council Institute of Metabolic Science and NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge CB2 0QQ, UK
| |
Collapse
|
24
|
Craddock JC, Neale EP, Peoples GE, Probst YC. Plant‐based eating patterns and endurance performance: A focus on inflammation, oxidative stress and immune responses. NUTR BULL 2020. [DOI: 10.1111/nbu.12427] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- J. C. Craddock
- Faculty of Science Medicine and Health School of Medicine University of Wollongong Wollongong NSW Australia
- Illawarra Health and Medical Research Institute Wollongong NSW Australia
| | - E. P. Neale
- Faculty of Science Medicine and Health School of Medicine University of Wollongong Wollongong NSW Australia
| | - G. E. Peoples
- Faculty of Science Medicine and Health School of Medicine University of Wollongong Wollongong NSW Australia
| | - Y. C. Probst
- Faculty of Science Medicine and Health School of Medicine University of Wollongong Wollongong NSW Australia
- Illawarra Health and Medical Research Institute Wollongong NSW Australia
| |
Collapse
|
25
|
Su JB, Wu YY, Xu F, Wang X, Cai HL, Zhao LH, Zhang XL, Chen T, Huang HY, Wang XQ. Serum complement C3 and islet β-cell function in patients with type 2 diabetes: A 4.6-year prospective follow-up study. Endocrine 2020; 67:321-330. [PMID: 31786774 DOI: 10.1007/s12020-019-02144-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Accepted: 11/18/2019] [Indexed: 12/16/2022]
Abstract
PURPOSE Serum complement C3 has been shown to contribute to the incidence of type 2 diabetes (T2D), but how serum complement C3 affects islet β-cell function throughout the course of T2D is unclear. This study explored whether serum complement C3 is independently associated with changes in islet β-cell function over time in patients with T2D. METHODS Serum complement C3 was measured, and endogenous β-cell function was evaluated by area under the C-peptide curve (AUCcp) during an oral glucose tolerance test (OGTT) in 411 patients with T2D at baseline from 2011 to 2015. Next, 347 of those patients with available data were pooled for a final follow-up analysis from 2014 to 2018. Changes in islet β-cell function at follow-up were evaluated by AUCcp percentage changes (ΔAUCcp%). In addition, other possible clinical risks for diabetes were also examined. RESULTS The 347 patients included in the analysis had a diabetes duration of 4.84 ± 3.63 years at baseline. Baseline serum complement C3 (baseline C3) levels were positively correlated with baseline natural logarithm of AUCcp (lnAUCcp) (n = 347, r = 0.288, p < 0.001), and baseline C3 was independently associated with baseline lnAUCcp (β = 0.17, t = 3.52, p < 0.001) after adjustment for baseline glycemic status and other clinical confounders by multivariate liner regression analysis. Compared with the baseline values, complement C3 changes (ΔC3) and ΔAUCcp% was -0.15 ± 0.28 mg/ml and -17.2 ± 18.4%, respectively, at a follow-up visit 4.57 ± 0.78 years later. Moreover, ΔC3 was positively correlated with ΔAUCcp% (n = 347, r = 0.302, p < 0.001). Furthermore, each 0.1 mg/ml increase in ΔC3 was associated with a higher ΔAUCcp% (1.41% [95% CI, 0.82-2.00%]) after adjusting for changes in glycemic status and other clinical confounders at follow-up. CONCLUSIONS In addition to serum complement C3 being independently associated with islet β-cell function at baseline, its changes were also independently associated with changes in islet β-cell function over time in patients with T2D.
Collapse
Affiliation(s)
- Jian-Bin Su
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University, and First People's Hospital of Nantong City, No. 6, Haierxiang North Road, Nantong, 226001, China.
| | - Yun-Yu Wu
- Medical School of Nantong University, No. 19, Qixiu Road, Nantong, 226001, China
| | - Feng Xu
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University, and First People's Hospital of Nantong City, No. 6, Haierxiang North Road, Nantong, 226001, China
| | - Xing Wang
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University, and First People's Hospital of Nantong City, No. 6, Haierxiang North Road, Nantong, 226001, China
| | - Hong-Li Cai
- Department of Geriatrics, Affiliated Hospital 2 of Nantong University, and First People's Hospital of Nantong City, No. 6 North Haierxiang Road, Nantong, 226001, China
| | - Li-Hua Zhao
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University, and First People's Hospital of Nantong City, No. 6, Haierxiang North Road, Nantong, 226001, China
| | - Xiu-Lin Zhang
- Department of Clinical Laboratory, Affiliated Hospital 2 of Nantong University, and First People's Hospital of Nantong City, No. 6, Haierxiang North Road, Nantong, 226001, China
| | - Tong Chen
- Department of Clinical Laboratory, Affiliated Hospital 2 of Nantong University, and First People's Hospital of Nantong City, No. 6, Haierxiang North Road, Nantong, 226001, China
| | - Hai-Yan Huang
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University, and First People's Hospital of Nantong City, No. 6, Haierxiang North Road, Nantong, 226001, China
| | - Xue-Qin Wang
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University, and First People's Hospital of Nantong City, No. 6, Haierxiang North Road, Nantong, 226001, China.
| |
Collapse
|
26
|
Shim K, Begum R, Yang C, Wang H. Complement activation in obesity, insulin resistance, and type 2 diabetes mellitus. World J Diabetes 2020; 11:1-12. [PMID: 31938469 PMCID: PMC6927818 DOI: 10.4239/wjd.v11.i1.1] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 11/07/2019] [Accepted: 11/26/2019] [Indexed: 02/05/2023] Open
Abstract
Amplified inflammatory reaction has been observed to be involved in cardiometabolic diseases such as obesity, insulin resistance, diabetes, dyslipidemia, and atherosclerosis. The complement system was originally viewed as a supportive first line of defense against microbial invaders, and research over the past decade has come to appreciate that the functions of the complement system extend beyond the defense and elimination of microbes, involving in such diverse processes as clearance of the immune complexes, complementing T and B cell immune functions, tissue regeneration, and metabolism. The focus of this review is to summarize the role of the activation of complement system and the initiation and progression of metabolic disorders including obesity, insulin resistance and diabetes mellitus. In addition, we briefly describe the interaction of the activation of the complement system with diabetic complications such as diabetic retinopathy, nephropathy and neuropathy, highlighting that targeting complement system therapeutics could be one of possible routes to slow down those aforementioned diabetic complications.
Collapse
Affiliation(s)
- Kyumin Shim
- Department of Basic Science, California Northstate University College of Medicine, Elk Grove, CA 95757, United States
| | - Rayhana Begum
- Department of Pharmacy, Primeasia University, Dhaka 1213, Bangladesh
| | - Catherine Yang
- Department of Basic Science, California Northstate University College of Medicine, Elk Grove, CA 95757, United States
- California Northstate University College of Graduate Studies, Elk Grove, CA 95757, United States
| | - Hongbin Wang
- Department of Basic Science, California Northstate University College of Medicine, Elk Grove, CA 95757, United States
- California Northstate University College of Graduate Studies, Elk Grove, CA 95757, United States
- Department of Pharmaceutical and Biomedical Sciences, California Northstate University College of Pharmacy, Elk Grove, CA 95757, United States
| |
Collapse
|
27
|
Copenhaver MM, Yu CY, Zhou D, Hoffman RP. Relationships of complement components C3 and C4 and their genetics to cardiometabolic risk in healthy, non-Hispanic white adolescents. Pediatr Res 2020; 87:88-94. [PMID: 31404919 PMCID: PMC6962538 DOI: 10.1038/s41390-019-0534-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 07/18/2019] [Accepted: 07/25/2019] [Indexed: 12/01/2022]
Abstract
BACKGROUND Complement promotes inflammatory and immune responses and may affect cardiometabolic risk. This study was designed to investigate the effect of complement components C3 and C4 on cardiometabolic risk in healthy non-Hispanic white adolescents. METHODS Body mass index (BMI), BMI percentile, waist circumference, and percent body fat were assessed in 75 adolescents. Arterial stiffness was assessed using arterial tomography and endothelial function using reactive hyperemia. Fasting lipids, inflammatory markers, and complement levels were measured and oral glucose tolerance test was performed. A single C3 polymorphism and C4 gene copy number variations were assessed. RESULTS C3 plasma levels increased with measures of obesity. Endothelial function worsened with increased C3 and C4 levels. Triglycerides and low-density lipoprotein increased and high-density lipoprotein (HDL) and insulin sensitivity decreased with increasing C3 levels, but the relationships were lost when body habitus was included in the model. C4 negatively related to HDL and positively to inflammatory markers. Subjects with at least one C3F allele had increased BMI and fat mass index. HDL was significantly related to C4L, C4S, C4A, and C4B gene copy number variation. CONCLUSIONS C3 levels increase with increasing body mass and increased C4 levels and copy number are associated with increased cardiometabolic risk in healthy adolescents.
Collapse
Affiliation(s)
- Melanie M Copenhaver
- Department of Pediatrics, Division of Emergency Medicine, Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Chack-Yung Yu
- Department of Pediatrics, Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Danlei Zhou
- Department of Pediatrics, Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Robert P Hoffman
- Department of Pediatrics, Division of Endocrinology, Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
28
|
van der Kolk BW, Kalafati M, Adriaens M, van Greevenbroek MMJ, Vogelzangs N, Saris WHM, Astrup A, Valsesia A, Langin D, van der Kallen CJH, Eussen SJPM, Schalkwijk CG, Stehouwer CDA, Goossens GH, Arts ICW, Jocken JWE, Evelo CT, Blaak EE. Subcutaneous Adipose Tissue and Systemic Inflammation Are Associated With Peripheral but Not Hepatic Insulin Resistance in Humans. Diabetes 2019; 68:2247-2258. [PMID: 31492661 DOI: 10.2337/db19-0560] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 08/27/2019] [Indexed: 11/13/2022]
Abstract
Obesity-related insulin resistance (IR) may develop in multiple organs, representing various etiologies for cardiometabolic diseases. We identified abdominal subcutaneous adipose tissue (ScAT) transcriptome profiles in liver or muscle IR by means of RNA sequencing in overweight or obese participants of the Diet, Obesity, and Genes (DiOGenes) (NCT00390637, ClinicalTrials.gov) cohort (n = 368). Tissue-specific IR phenotypes were derived from a 5-point oral glucose tolerance test. Hepatic and muscle IR were characterized by distinct abdominal ScAT transcriptome profiles. Genes related to extracellular remodeling were upregulated in individuals with primarily hepatic IR, while genes related to inflammation were upregulated in individuals with primarily muscle IR. In line with this, in two independent cohorts, the Cohort on Diabetes and Atherosclerosis Maastricht (CODAM) (n = 325) and the Maastricht Study (n = 685), an increased systemic low-grade inflammation profile was specifically related to muscle IR but not to liver IR. We propose that increased ScAT inflammatory gene expression may translate into an increased systemic inflammatory profile, linking ScAT inflammation to the muscle IR phenotype. These distinct IR phenotypes may provide leads for more personalized prevention of cardiometabolic diseases.
Collapse
Affiliation(s)
- Birgitta W van der Kolk
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| | - Marianthi Kalafati
- Department of Bioinformatics - BiGCaT, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, the Netherlands
| | - Michiel Adriaens
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, the Netherlands
| | - Marleen M J van Greevenbroek
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Nicole Vogelzangs
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, the Netherlands
- Department of Epidemiology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Wim H M Saris
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| | - Arne Astrup
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | | | - Dominique Langin
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
- Paul Sabatier University, Toulouse, France
- Laboratory of Clinical Biochemistry, Toulouse University Hospitals, Toulouse, France
| | - Carla J H van der Kallen
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Simone J P M Eussen
- Department of Epidemiology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Casper G Schalkwijk
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Coen D A Stehouwer
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Gijs H Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| | - Ilja C W Arts
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, the Netherlands
- Department of Epidemiology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Johan W E Jocken
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| | - Chris T Evelo
- Department of Bioinformatics - BiGCaT, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, the Netherlands
| | - Ellen E Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
29
|
Wildberg C, Masuch A, Budde K, Kastenmüller G, Artati A, Rathmann W, Adamski J, Kocher T, Völzke H, Nauck M, Friedrich N, Pietzner M. Plasma Metabolomics to Identify and Stratify Patients With Impaired Glucose Tolerance. J Clin Endocrinol Metab 2019; 104:6357-6370. [PMID: 31390012 DOI: 10.1210/jc.2019-01104] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 08/01/2019] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Impaired glucose tolerance (IGT) is one of the presymptomatic states of type 2 diabetes mellitus and requires an oral glucose tolerance test (OGTT) for diagnosis. Our aims were twofold: (i) characterize signatures of small molecules predicting the OGTT response and (ii) identify metabolic subgroups of participants with IGT. METHODS Plasma samples from 827 participants of the Study of Health in Pomerania free of diabetes were measured using mass spectrometry and proton-nuclear magnetic resonance spectroscopy. Linear regression analyses were used to screen for metabolites significantly associated with the OGTT response after 2 hours, adjusting for baseline glucose and insulin levels as well as important confounders. A signature predictive for IGT was established using regularized logistic regression. All cases with IGT (N = 159) were selected and subjected to unsupervised clustering using a k-means approach. RESULTS AND CONCLUSION In total, 99 metabolites and 22 lipoprotein measures were significantly associated with either 2-hour glucose or 2-hour insulin levels. Those comprised variations in baseline concentrations of branched-chain amino ketoacids, acylcarnitines, lysophospholipids, or phosphatidylcholines, largely confirming previous studies. By the use of these metabolites, subjects with IGT segregated into two distinct groups. Our IGT prediction model combining both clinical and metabolomics traits achieved an area under the curve of 0.84, slightly improving the prediction based on established clinical measures. The present metabolomics approach revealed molecular signatures associated directly to the response of the OGTT and to IGT in line with previous studies. However, clustering of subjects with IGT revealed distinct metabolic signatures of otherwise similar individuals, pointing toward the possibility of metabolomics for patient stratification.
Collapse
Affiliation(s)
- Charlotte Wildberg
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Annette Masuch
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Kathrin Budde
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- German Centre for Cardiovascular Research, partner site Greifswald, Greifswald, Germany
| | - Gabi Kastenmüller
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Anna Artati
- Institute of Experimental Genetics, Genome Analysis Center, Helmholtz Zentrum München, Neuherberg, Germany
| | - Wolfgang Rathmann
- Institute of Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Jerzy Adamski
- Institute of Experimental Genetics, Genome Analysis Center, Helmholtz Zentrum München, Neuherberg, Germany
- Lehrstuhl für Experimentelle Genetik, Technische Universität München, Freising-Weihenstephan, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Thomas Kocher
- Unit of Periodontology, Department of Restorative Dentistry, Periodontology, Endodontology, and Pediatric and Preventive Dentistry, Dental School, University Medicine Greifswald, Greifswald, Germany
| | - Henry Völzke
- German Centre for Cardiovascular Research, partner site Greifswald, Greifswald, Germany
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- German Center for Diabetes Research, site Greifswald, Greifswald, Germany
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- German Centre for Cardiovascular Research, partner site Greifswald, Greifswald, Germany
| | - Nele Friedrich
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- German Centre for Cardiovascular Research, partner site Greifswald, Greifswald, Germany
| | - Maik Pietzner
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- German Centre for Cardiovascular Research, partner site Greifswald, Greifswald, Germany
| |
Collapse
|
30
|
Tong Y, Pei L, Luo K, Zhao M, Xu J, Li A, Li R, Yang M, Xu Q. The mediated role of complement C3 in PM 2.5 exposure and type 2 diabetes mellitus: an elderly panel study in Beijing, China. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:34479-34486. [PMID: 31642019 DOI: 10.1007/s11356-019-06487-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 09/09/2019] [Indexed: 06/10/2023]
Abstract
Diabetes mellitus (DM) is a common chronic disease worldwide. Ambient air pollution has long been proven to be associated with type 2 diabetes mellitus (T2DM) progression, but the underlying mechanism is not clear yet. In addition, previous studies mainly focused on the prevention of healthy people against the incidence of T2DM. We designed a panel study including two follow-ups and enrolled 39 patients with T2DM living in Beijing. Linear mixed model was fitted to assess the association between two pairs of variables (ambient air pollution exposure and C3 levels, ambient air pollution exposures and T2DM index). Mediation analysis of C3 between ambient air pollution exposure and indicators of T2DM progression was conducted. We found that PM2.5 exposures is are negatively associated with serum complement C3. Given that C3 might act as a protector of pancreas β cell, PM2.5 exposures could accelerate disease in T2DM populations. No mediation effects were found. This study reveals that exposures to PM2.5 can cause progression of diseases among T2DM populations.
Collapse
Affiliation(s)
- Yuanren Tong
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
- Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100005, China
| | - Lu Pei
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
- Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100005, China
| | - Kai Luo
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
- Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100005, China
| | - Meiduo Zhao
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
- Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100005, China
| | - Jing Xu
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
- Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100005, China
| | - Ang Li
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
- Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100005, China
| | - Runkui Li
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Resources and Environmental Information System, Institute of Geographic Sciences and Natural Resources Research, Chinese Academy of Sciences, Beijing, 100101, China
| | - Mingan Yang
- Division of Biostatistics and Epidemiology, Graduate School of Public Health, San Diego State University, San Diego, CA, 92182, USA
| | - Qun Xu
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China.
| |
Collapse
|
31
|
Ding L, Fan L, Xu X, Fu J, Xue Y. Identification of core genes and pathways in type 2 diabetes mellitus by bioinformatics analysis. Mol Med Rep 2019; 20:2597-2608. [PMID: 31524257 PMCID: PMC6691242 DOI: 10.3892/mmr.2019.10522] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 05/07/2019] [Indexed: 02/07/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a metabolic disorder. Numerous proteins have been identified that are associated with the occurrence and development of T2DM. This study aimed to identify potential core genes and pathways involved in T2DM, through exhaustive bioinformatic analyses using GSE20966 microarray profiles of pancreatic β‑cells obtained from healthy controls and patients with T2DM. The original microarray data were downloaded from the Gene Expression Omnibus database. Data were processed by the limma package in R software and the differentially expressed genes (DEGs) were identified. Gene Ontology functional analysis and Kyoto Encyclopedia of Genes and Genomes pathway analysis were carried out to identify potential biological functions and pathways of the DEGs. Key transcription factors were identified using the WEB‑based GEne SeT AnaLysis Toolkit (WebGestalt) and Enrichr. The Search Tool for the Retrieval of Interacting Genes (STRING) database was used to establish a protein‑protein interaction (PPI) network for the DEGs. In total, 329 DEGs were involved in T2DM, with 208 upregulated genes enriched in pancreatic secretion and the complement and coagulation cascades, and 121 downregulated genes enriched in insulin secretion, carbohydrate digestion and absorption, and the Toll‑like receptor pathway. Furthermore, hepatocyte nuclear factor 1‑alpha (HNF1A), signal transducer and activator of transcription 3 (STAT3) and glucocorticoid receptor (GR) were key transcription factors in T2DM. Twenty important nodes were detected in the PPI network. Finally, two core genes, serpin family G member 1 (SERPING1) and alanyl aminopeptidase, membrane (ANPEP), were shown to be associated with the development of T2DM. On the whole, the findings of this study enhance our understanding of the potential molecular mechanisms of T2DM and provide potential targets for further research.
Collapse
Affiliation(s)
- Linchao Ding
- Department of Scientific Research, Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, P.R. China
| | - Lei Fan
- Department of Pharmacy, Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, P.R. China
| | - Xiaodong Xu
- Department of Endocrinology, Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, P.R. China
| | - Jianfei Fu
- Department of Scientific Research, Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, P.R. China
| | - Yadong Xue
- Department of Scientific Research, Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, P.R. China
| |
Collapse
|
32
|
Role of complement in diabetes. Mol Immunol 2019; 114:270-277. [PMID: 31400630 DOI: 10.1016/j.molimm.2019.07.031] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/29/2019] [Accepted: 07/30/2019] [Indexed: 02/07/2023]
Abstract
Accumulating evidence suggests a role for the complement system in the pathogenesis of diabetes and the vascular complications that characterise this condition. Complement proteins contribute to the development of type 1 diabetes (T1D) by enhancing the underlying organ-specific autoimmune processes. Complement upregulation and activation is also an important feature of insulin resistance and the development of type 2 diabetes (T2D). Moreover, animal and human studies indicate that complement proteins are involved in the pathogenic mechanisms leading to diabetic microvascular and macrovascular complications. The adverse vascular effects of complement appear to be related to enhancement of the inflammatory process and the predisposition to a thrombotic environment, eventually leading to vascular occlusion. Complement proteins have been considered as therapeutic targets to prevent or treat vascular disease but studies have been mainly conducted in animal models, while human work has been both limited and inconclusive so far. Further studies are needed to understand the potential role of complement proteins as therapeutic targets for reversal of the pathological processes leading to T1D and T2D and for the prevention/treatment of diabetic vascular complications.
Collapse
|
33
|
Shen L, Zhao D, Chen Y, Zhang K, Chen X, Lin J, Li C, Iqbal J, Zhao Y, Liang Y, Wei Y, Feng C. Comparative Proteomics Analysis of Serum Proteins in Gestational Diabetes during Early and Middle Stages of Pregnancy. Proteomics Clin Appl 2019; 13:e1800060. [DOI: 10.1002/prca.201800060] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/26/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Liming Shen
- College of Life Science and OceanographyShenzhen University Shenzhen 518060 P. R. China
| | - Danqing Zhao
- Department of Obstetrics and GynecologyAffiliated Hospital of Guizhou Medical University Guiyang 550004 P. R. China
| | - Youjiao Chen
- College of Life Science and OceanographyShenzhen University Shenzhen 518060 P. R. China
| | - Kaoyuan Zhang
- College of Life Science and OceanographyShenzhen University Shenzhen 518060 P. R. China
| | - Xinqian Chen
- College of Life Science and OceanographyShenzhen University Shenzhen 518060 P. R. China
| | - Jing Lin
- College of Life Science and OceanographyShenzhen University Shenzhen 518060 P. R. China
| | - Cuihua Li
- College of Life Science and OceanographyShenzhen University Shenzhen 518060 P. R. China
| | - Javed Iqbal
- College of Life Science and OceanographyShenzhen University Shenzhen 518060 P. R. China
| | - Yuxi Zhao
- College of Life Science and OceanographyShenzhen University Shenzhen 518060 P. R. China
| | - Yi Liang
- School of Public HealthGuizhou Medical University Guiyang 550025 P. R. China
| | - Yan Wei
- School of Public HealthGuizhou Medical University Guiyang 550025 P. R. China
| | - Chengyun Feng
- Maternal and Child Health Hospital of Baoan Shenzhen 518100 P. R. China
| |
Collapse
|
34
|
Bao X, Borné Y, Muhammad IF, Schulz CA, Persson M, Orho-Melander M, Niu K, Christensson A, Engström G. Complement C3 and incident hospitalization due to chronic kidney disease: a population-based cohort study. BMC Nephrol 2019; 20:61. [PMID: 30791918 PMCID: PMC6383246 DOI: 10.1186/s12882-019-1248-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 02/06/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Circulating C3 has been associated with diabetes and hypertension, which are the leading causes of chronic kidney disease (CKD). C3 activation is considered to contribute to several renal diseases. Here we examined whether elevated C3 concentration is associated with hospitalization due to CKD in the general population, and whether this relationship is mediated by factors such as diabetes and hypertension. METHODS Baseline plasma C3 was quantified in 4552 participants, without previous hospital admission due to CKD, from the Malmö Diet and Cancer cohort study. Incidence of first hospitalization due to CKD (main diagnosis) was investigated in relation to C3 levels using Cox proportional hazards regression models after a mean follow-up of 19.2 ± 4.16 years. Traditional risk factors of CKD including diabetes, blood pressure, C-reactive protein and baseline renal function were considered in adjustments and sensitivity analyses. RESULTS During the follow-up period, 94 subjects were admitted to hospital due to CKD. After multivariate adjustment, the hazard ratios (95% confidence interval) for hospitalization from CKD across quartiles of C3 were 1.00 (reference), 1.68 (0.69, 4.13), 2.71 (1.15, 6.39), and 3.16 (1.36, 7.34) (p for trend = 0.003). Results were generally consistent across different sensitivity analyses. CONCLUSIONS These findings indicate that C3 is associated with incidence of first hospitalization due to CKD in the general population. The observed relationship cannot be entirely attributed to hyperglycemia and hypertension.
Collapse
Affiliation(s)
- Xue Bao
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
- Department of Clinical Sciences, Lund University, Malmö, CRC 60:13, Jan Waldenströms gata 35, S-20502 Malmö, Sweden
| | - Yan Borné
- Department of Clinical Sciences, Lund University, Malmö, CRC 60:13, Jan Waldenströms gata 35, S-20502 Malmö, Sweden
| | - Iram Faqir Muhammad
- Department of Clinical Sciences, Lund University, Malmö, CRC 60:13, Jan Waldenströms gata 35, S-20502 Malmö, Sweden
| | - Christina-Alexandra Schulz
- Department of Clinical Sciences, Lund University, Malmö, CRC 60:13, Jan Waldenströms gata 35, S-20502 Malmö, Sweden
| | - Margaretha Persson
- Department of Clinical Sciences, Lund University, Malmö, CRC 60:13, Jan Waldenströms gata 35, S-20502 Malmö, Sweden
| | - Marju Orho-Melander
- Department of Clinical Sciences, Lund University, Malmö, CRC 60:13, Jan Waldenströms gata 35, S-20502 Malmö, Sweden
| | - Kaijun Niu
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Anders Christensson
- Department of Clinical Sciences, Lund University, Malmö, CRC 60:13, Jan Waldenströms gata 35, S-20502 Malmö, Sweden
| | - Gunnar Engström
- Department of Clinical Sciences, Lund University, Malmö, CRC 60:13, Jan Waldenströms gata 35, S-20502 Malmö, Sweden
| |
Collapse
|
35
|
King BC, Kulak K, Krus U, Rosberg R, Golec E, Wozniak K, Gomez MF, Zhang E, O'Connell DJ, Renström E, Blom AM. Complement Component C3 Is Highly Expressed in Human Pancreatic Islets and Prevents β Cell Death via ATG16L1 Interaction and Autophagy Regulation. Cell Metab 2019; 29:202-210.e6. [PMID: 30293775 DOI: 10.1016/j.cmet.2018.09.009] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 07/23/2018] [Accepted: 09/07/2018] [Indexed: 01/25/2023]
Abstract
We show here that human pancreatic islets highly express C3, which is both secreted and present in the cytosol. Within isolated human islets, C3 expression correlates with type 2 diabetes (T2D) donor status, HbA1c, and inflammation. Islet C3 expression is also upregulated in several rodent diabetes models. C3 interacts with ATG16L1, which is essential for autophagy. Autophagy relieves cellular stresses faced by β cells during T2D and maintains cellular homeostasis. C3 knockout in clonal β cells impaired autophagy and led to increased apoptosis after exposure of cells to palmitic acid and IAPP. In the absence of C3, autophagosomes do not undergo fusion with lysosomes. Thus, C3 may be upregulated in islets during T2D as a cytoprotective factor against β cell dysfunction caused by impaired autophagy. Therefore, we revealed a previously undescribed intracellular function for C3, connecting the complement system directly to autophagy, with a broad potential importance in other diseases and cell types.
Collapse
Affiliation(s)
- Ben C King
- Section of Medical Protein Chemistry, Department of Translational Medicine, Lund University, 214-28 Malmö, Sweden
| | - Klaudia Kulak
- Section of Medical Protein Chemistry, Department of Translational Medicine, Lund University, 214-28 Malmö, Sweden
| | - Ulrika Krus
- Lund University Diabetes Centre, Department of Clinical Sciences Malmö, Lund University, 214-28 Malmö, Sweden
| | - Rebecca Rosberg
- Section of Medical Protein Chemistry, Department of Translational Medicine, Lund University, 214-28 Malmö, Sweden
| | - Ewelina Golec
- Section of Medical Protein Chemistry, Department of Translational Medicine, Lund University, 214-28 Malmö, Sweden
| | - Katarzyna Wozniak
- Section of Medical Protein Chemistry, Department of Translational Medicine, Lund University, 214-28 Malmö, Sweden
| | - Maria F Gomez
- Lund University Diabetes Centre, Department of Clinical Sciences Malmö, Lund University, 214-28 Malmö, Sweden
| | - Enming Zhang
- Lund University Diabetes Centre, Department of Clinical Sciences Malmö, Lund University, 214-28 Malmö, Sweden
| | - David J O'Connell
- School of Biomolecular & Biomedical Science, Conway Institute, University College Dublin, Dublin, Ireland
| | - Erik Renström
- Lund University Diabetes Centre, Department of Clinical Sciences Malmö, Lund University, 214-28 Malmö, Sweden
| | - Anna M Blom
- Section of Medical Protein Chemistry, Department of Translational Medicine, Lund University, 214-28 Malmö, Sweden.
| |
Collapse
|
36
|
Xin Y, Hertle E, van der Kallen CJH, Schalkwijk CG, Stehouwer CDA, van Greevenbroek MMJ. Complement C3 and C4, but not their regulators or activated products, are associated with incident metabolic syndrome: the CODAM study. Endocrine 2018; 62:617-627. [PMID: 30132263 PMCID: PMC6244913 DOI: 10.1007/s12020-018-1712-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 08/07/2018] [Indexed: 12/13/2022]
Abstract
PURPOSE We investigated the associations of components of the alternative (C3, C3a, Bb, factor D [FD], factor H [FH], properdin) and the classical complement pathway (C4, C1q, C1-inhibitor [C1-INH]) with prevalent and incident metabolic syndrome in a cohort with a moderately increased risk of cardiometabolic disease. METHODS The study cohort was comprised of 574 participants (61% men, age 59.6 ± 7.0 years) at baseline and 489 participants after 7-year follow-up. Multiple logistic regression analyses were done to investigate the associations of concentrations of baseline plasma complement (standardized values) with prevalent and incident (in those without metabolic syndrome at baseline, n = 189) metabolic syndrome. RESULTS C3 (odds ratio (OR) = 1.48 [95% confidence interval: 1.02; 2.14]) and C4 (OR = 1.95 [1.32; 2.88]), but none of the other complement components were associated with incident metabolic syndrome (n = 40 cases). Notably, in the cross-sectional analyses, we did observe higher levels of C3a (OR = 1.25 [1.03; 1.52]), FH (OR = 2.93 [2.24; 3.83]), and properdin (OR = 1.88 [1.50; 2.34]), in addition to C3 (OR = 3.60 [2.73; 4.75]) and C4 (OR = 1.39 [1.13; 1.69]), in those with the metabolic syndrome compared to those without, while no association was observed for FD, Bb, C1q, or C1-INH. CONCLUSIONS In the cross-sectional analyses, the effects sizes (standardized regression coefficients) for C3 and C4 were similar to those of (some of) the regulators and activators, yet only C3 and C4 were associated with incident disease. These findings suggest a role for C3 and C4, but not their regulators or activated products, in the development of the metabolic syndrome.
Collapse
Affiliation(s)
- Ying Xin
- Department of Internal Medicine, Maastricht University Medical Centre and CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | - Elisabeth Hertle
- Department of Internal Medicine, Maastricht University Medical Centre and CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | - Carla J H van der Kallen
- Department of Internal Medicine, Maastricht University Medical Centre and CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | - Casper G Schalkwijk
- Department of Internal Medicine, Maastricht University Medical Centre and CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | - Coen D A Stehouwer
- Department of Internal Medicine, Maastricht University Medical Centre and CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | - Marleen M J van Greevenbroek
- Department of Internal Medicine, Maastricht University Medical Centre and CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
37
|
't Hart LM, Vogelzangs N, Mook-Kanamori DO, Brahimaj A, Nano J, van der Heijden AAWA, Willems van Dijk K, Slieker RC, Steyerberg EW, Ikram MA, Beekman M, Boomsma DI, van Duijn CM, Slagboom PE, Stehouwer CDA, Schalkwijk CG, Arts ICW, Dekker JM, Dehghan A, Muka T, van der Kallen CJH, Nijpels G, van Greevenbroek MMJ. Blood Metabolomic Measures Associate With Present and Future Glycemic Control in Type 2 Diabetes. J Clin Endocrinol Metab 2018; 103:4569-4579. [PMID: 30113659 DOI: 10.1210/jc.2018-01165] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 07/30/2018] [Indexed: 02/06/2023]
Abstract
OBJECTIVE We studied whether blood metabolomic measures in people with type 2 diabetes (T2D) are associated with insufficient glycemic control and whether this association is influenced differentially by various diabetes drugs. We then tested whether the same metabolomic profiles were associated with the initiation of insulin therapy. METHODS A total of 162 metabolomic measures were analyzed using a nuclear magnetic resonance-based method in people with T2D from four cohort studies (n = 2641) and one replication cohort (n = 395). Linear and logistic regression analyses with adjustment for potential confounders, followed by meta-analyses, were performed to analyze associations with hemoglobin A1c levels, six glucose-lowering drug categories, and insulin initiation during a 7-year follow-up period (n = 698). RESULTS After Bonferroni correction, 26 measures were associated with insufficient glycemic control (HbA1c >53 mmol/mol). The strongest association was with glutamine (OR, 0.66; 95% CI, 0.61 to 0.73; P = 7.6 × 10-19). In addition, compared with treatment-naive patients, 31 metabolomic measures were associated with glucose-lowering drug use (representing various metabolite categories; P ≤ 3.1 × 10-4 for all). In drug-stratified analyses, associations with insufficient glycemic control were only mildly affected by different glucose-lowering drugs. Five of the 26 metabolomic measures (apolipoprotein A1 and medium high-density lipoprotein subclasses) were also associated with insulin initiation during follow-up in both discovery and replication. The strongest association was observed for medium high-density lipoprotein cholesteryl ester (OR, 0.54; 95% CI, 0.42 to 0.71; P = 4.5 × 10-6). CONCLUSION Blood metabolomic measures were associated with present and future glycemic control and might thus provide relevant cues to identify those at increased risk of treatment failure.
Collapse
Affiliation(s)
- Leen M 't Hart
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden ZA, Netherlands
- Section of Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden ZA, Netherlands
- Department of Epidemiology and Biostatistics, Amsterdam Public Health Research Institute, VU University Medical Center, Amsterdam HV, Netherlands
| | - Nicole Vogelzangs
- Cardiovascular Research Institute Maastricht and Maastricht Centre for Systems Biology, Maastricht University, Maastricht LK, Netherlands
| | - Dennis O Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden ZA, Netherlands
- Department of Public Health and Primary Care, Leiden University Medical Center, Leiden ZA, Netherlands
| | - Adela Brahimaj
- Department of Epidemiology, Erasmus Medical Center, Rotterdam GD, Netherlands
| | - Jana Nano
- Department of Epidemiology, Erasmus Medical Center, Rotterdam GD, Netherlands
- Institute of Epidemiology, German Research Center for Environment Health, Helmholtz Zentrum Munich, Munich, Germany
- German Center for Diabetes Research (Deutsches Zentrum für Diabetesforschung), Munich, Germany
| | - Amber A W A van der Heijden
- Department of General Practice and Elderly Care Medicine, Amsterdam Public Health Research Institute, VU University Medical Center, Amsterdam, Netherlands
| | - Ko Willems van Dijk
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden ZA, Netherlands
- Department of Human Genetics, Leiden University Medical Center, Leiden ZA, Netherlands
- Division of Endocrinology, Department of Internal Medicine, Leiden University Medical Center, Leiden ZA, Netherlands
| | - Roderick C Slieker
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden ZA, Netherlands
- Department of Epidemiology and Biostatistics, Amsterdam Public Health Research Institute, VU University Medical Center, Amsterdam HV, Netherlands
| | - Ewout W Steyerberg
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden ZA, Netherlands
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus Medical Center, Rotterdam GD, Netherlands
| | - Marian Beekman
- Section of Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden ZA, Netherlands
| | - Dorret I Boomsma
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam HV, Netherlands
| | | | - P Eline Slagboom
- Section of Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden ZA, Netherlands
| | - Coen D A Stehouwer
- Cardiovascular Research Institute Maastricht, School for Cardiovascular Diseases, Maastricht University, Maastricht LK, Netherlands
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht LK, Netherlands
| | - Casper G Schalkwijk
- Cardiovascular Research Institute Maastricht, School for Cardiovascular Diseases, Maastricht University, Maastricht LK, Netherlands
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht LK, Netherlands
| | - Ilja C W Arts
- Cardiovascular Research Institute Maastricht and Maastricht Centre for Systems Biology, Maastricht University, Maastricht LK, Netherlands
| | - Jacqueline M Dekker
- Department of Epidemiology and Biostatistics, Amsterdam Public Health Research Institute, VU University Medical Center, Amsterdam HV, Netherlands
| | - Abbas Dehghan
- Department of Epidemiology, Erasmus Medical Center, Rotterdam GD, Netherlands
- Department of Biostatistics and Epidemiology, MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London, United Kingdom
| | - Taulant Muka
- Department of Epidemiology, Erasmus Medical Center, Rotterdam GD, Netherlands
| | - Carla J H van der Kallen
- Cardiovascular Research Institute Maastricht, School for Cardiovascular Diseases, Maastricht University, Maastricht LK, Netherlands
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht LK, Netherlands
| | - Giel Nijpels
- Department of General Practice and Elderly Care Medicine, Amsterdam Public Health Research Institute, VU University Medical Center, Amsterdam, Netherlands
| | - Marleen M J van Greevenbroek
- Cardiovascular Research Institute Maastricht, School for Cardiovascular Diseases, Maastricht University, Maastricht LK, Netherlands
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht LK, Netherlands
| |
Collapse
|
38
|
Muniyappa R, Tella SH, Sortur S, Mszar R, Grewal S, Abel BS, Auh S, Chang DC, Krakoff J, Skarulis MC. Predictive Accuracy of Surrogate Indices for Hepatic and Skeletal Muscle Insulin Sensitivity. J Endocr Soc 2018; 3:108-118. [PMID: 30675598 DOI: 10.1210/js.2018-00206] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 09/20/2018] [Indexed: 12/25/2022] Open
Abstract
Context Surrogate indices of muscle and hepatic insulin sensitivity derived from an oral glucose tolerance test (OGTT) are frequently used in clinical studies. However, the predictive accuracy of these indices has not been validated. Design In this cross-sectional study, hyperinsulinemic-euglycemic glucose clamp with tritiated glucose infusion and a 75-g OGTT were performed in individuals (n = 659, aged 18 to 49 years, body mass index of 16 to 64 kg/m2) with varying degrees of glucose tolerance. A calibration model was used to assess the ability of OGTT-derived, tissue-specific surrogate indices [hepatic insulin resistance index (HIRI) and muscle insulin sensitivity index (MISI)] to predict insulin sensitivity/resistance indices derived from the reference glucose clamp [Hepatic-IRbasal, a product of fasting plasma insulin and hepatic glucose production (HGP), Hepatic-IRclamp, reciprocal of the percent suppression of HGP during the insulin clamp corrected for plasma insulin concentration, and Muscle-ISclamp, a measure of peripheral glucose disposal]. Predictive accuracy was assessed by root mean squared error of prediction and leave-one-out, cross-validation-type square root of the mean squared error of prediction. Results HIRI and MISI were correlated with their respective clamp-derived indices. HIRI was negatively related to Muscle-ISclamp (r = -0.62, P < 0.0001) and MISI correlated with Hepatic-IR derived from the clamp (Hepatic-IRbasal: r = -0.48, P < 0.0001 and Hepatic-IRclamp: r = -0.41, P < 0.0001). However, the accuracy of HIRI and MISI to predict Hepatic-IR (basal or during clamp) was not significantly different. Likewise, the ability of HIRI and MISI to predict Muscle-ISclamp was also similar. Conclusion Our findings indicate that the surrogate indices derived from an OGTT are accurate in predicting insulin sensitivity but are not tissue specific.
Collapse
Affiliation(s)
- Ranganath Muniyappa
- Diabetes, Endocrinology, and Obesity Branch, Clinical Endocrinology Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Sri Harsha Tella
- Diabetes, Endocrinology, and Obesity Branch, Clinical Endocrinology Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Shrayus Sortur
- Diabetes, Endocrinology, and Obesity Branch, Clinical Endocrinology Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Reed Mszar
- Diabetes, Endocrinology, and Obesity Branch, Clinical Endocrinology Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Shivraj Grewal
- Diabetes, Endocrinology, and Obesity Branch, Clinical Endocrinology Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Brent S Abel
- Diabetes, Endocrinology, and Obesity Branch, Clinical Endocrinology Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Sungyoung Auh
- Diabetes, Endocrinology, and Obesity Branch, Clinical Endocrinology Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Douglas C Chang
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona
| | - Jonathan Krakoff
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona
| | - Monica C Skarulis
- Diabetes, Endocrinology, and Obesity Branch, Clinical Endocrinology Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
39
|
Comparative Proteomic Analysis of Two Differently Extracted Coptis chinensis in the Treatment of Type 2 Diabetic Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:3248521. [PMID: 30302116 PMCID: PMC6158947 DOI: 10.1155/2018/3248521] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 08/08/2018] [Accepted: 08/16/2018] [Indexed: 12/29/2022]
Abstract
Coptis chinensis (CC) is widely used to treat diabetes in traditional Chinese medicine due to its significant hypoglycemic and hypolipidemic effects. It was reported that CC powders are more effective than CC decoctions. In this study, a rat model of type 2 diabetes was established and treated with supercritical-extracted CC and gastric juice extracted CC, respectively. Body weight, fasting plasma insulin, insulin resistance index, and lipid profiles were measured along with oral glucose tolerance tests (OGTTs). In addition, the levels of plasma proteins were compared between type 2 diabetic rats and CC-treated rats using an iTRAQ-based quantitative proteomic analysis. The results showed that the plasma levels of triglyceride (TC), total cholesterol (TG), and low-density lipoprotein (LDL) in rats of both CC-treated groups were significantly decreased. In addition, the proteomic analysis identified 929 proteins, while 15 proteins were selected from these 929 proteins based on their expression levels and bioinformatic results. Among these 15 proteins, 9 proteins (IGF-1, Igfbp4, Igfbp-6, Igfals, C2, C4, Cfi, Prdx-2, and Prdx-3) were upregulated in the two CC-treated groups, while 6 proteins (Pla2g7, Pcyox1, ApoC-1, ApoC-3, ApoB-100, and ApoE) were downregulated. The functions of these proteins are associated with glucose metabolism, insulin action, immunity, inflammation, lipid metabolism, oxidation, and antioxidation. The two differently extracted CC did not show significant differences in terms of their treatment efficacy. This research expanded our understanding on the therapeutic effects and mechanisms of CC in the treatment of type 2 diabetes.
Collapse
|
40
|
Castellano-Castillo D, Moreno-Indias I, Fernandez-Garcia JC, Clemente-Postigo M, Castro-Cabezas M, Tinahones FJ, Queipo-Ortuño MI, Cardona F. Complement Factor C3 Methylation and mRNA Expression Is Associated to BMI and Insulin Resistance in Obesity. Genes (Basel) 2018; 9:E410. [PMID: 30104553 PMCID: PMC6116013 DOI: 10.3390/genes9080410] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 07/20/2018] [Accepted: 08/08/2018] [Indexed: 12/24/2022] Open
Abstract
Epigenetic marks, and especially DNA methylation, are becoming an important factor in obesity, which could help to explain its etiology and associated comorbidities. Adipose tissue, now considered as an important endocrine organ, produces complement system factors. Complement component 3 (C3) turns out to be an important protein in metabolic disorders, via either inflammation or the C3 subproduct acylation stimulating protein (ASP) which directly stimulates lipid storage. In this study, we analyze C3 DNA methylation in adipose tissue from subjects with a different grade of obesity. Adipose tissue samples were collected from subjects with a different degree of obesity determined by their body mass index (BMI) as: Overweight subjects (BMI ≥ 25 and <30), obese class 1/2 subjects (BMI ≥ 30 and <40) and obese class 3 subjects (BMI ≥ 40). C3 DNA methylation was measured for 7 CpGs by pyrosequencition using the Pyromark technology (Qiagen, Madrid Spain). C3 messenger RNA (mRNA) levels were analyzed by pre-designed Taqman assays (Applied biosystems, Foster City, CA, USA) and ASP/C3a was measured using a ELISA kit. The data were analyzed using the statistic package SPSS. C3 DNA methylation levels were lower in the morbid obese group. Accordingly, C3 methylation correlated negatively with BMI and leptin. However, C3 mRNA levels were more associated with insulin resistance, and positive correlations with insulin, glucose and homeostasis model assessment-estimated insulin resistance (HOMA-IR) existed. ASP correlated negatively with high density lipoprotein (HDL) cholesterol. C3 methylation levels were associated to adiposity variables, such as BMI and leptin, while the C3 mRNA levels were associated to glucose metabolism.
Collapse
Affiliation(s)
- Daniel Castellano-Castillo
- Unidad de Gestión Clínica de Endocrinología y Nutrición del Hospital Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Malaga, 29010 Malaga, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERobn), 28029 Madrid, Spain.
| | - Isabel Moreno-Indias
- Unidad de Gestión Clínica de Endocrinología y Nutrición del Hospital Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Malaga, 29010 Malaga, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERobn), 28029 Madrid, Spain.
| | - Jose Carlos Fernandez-Garcia
- Unidad de Gestión Clínica de Endocrinología y Nutrición del Hospital Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Malaga, 29010 Malaga, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERobn), 28029 Madrid, Spain.
| | - Mercedes Clemente-Postigo
- Unidad de Gestión Clínica de Endocrinología y Nutrición del Hospital Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Malaga, 29010 Malaga, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERobn), 28029 Madrid, Spain.
| | - Manuel Castro-Cabezas
- Department of Internal Medicine Center for Endocrinology, Diabetes and Vascular Medicine St. Franciscus Gasthuis Hospital, 3045 PM Rotterdam, The Netherlands.
| | - Francisco José Tinahones
- Unidad de Gestión Clínica de Endocrinología y Nutrición del Hospital Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Malaga, 29010 Malaga, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERobn), 28029 Madrid, Spain.
| | - María Isabel Queipo-Ortuño
- Unidad de Gestión Clínica de Endocrinología y Nutrición del Hospital Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Malaga, 29010 Malaga, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERobn), 28029 Madrid, Spain.
| | - Fernando Cardona
- Unidad de Gestión Clínica de Endocrinología y Nutrición del Hospital Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Malaga, 29010 Malaga, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERobn), 28029 Madrid, Spain.
| |
Collapse
|
41
|
Ratter JM, Tack CJ, Netea MG, Stienstra R. Environmental Signals Influencing Myeloid Cell Metabolism and Function in Diabetes. Trends Endocrinol Metab 2018; 29:468-480. [PMID: 29789206 DOI: 10.1016/j.tem.2018.04.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 04/18/2018] [Accepted: 04/19/2018] [Indexed: 12/13/2022]
Abstract
The environment induces metabolic reprogramming of immune cells via specific signaling pathways. Recent studies have revealed that changes in cell metabolism affect key immune cell functions including cytokine production and migration. In diabetes, these functions are either insufficiently or excessively activated, translating into diabetes-associated complications, including increased susceptibility to infection and accelerated cardiovascular disease. Diabetes alters the abundance of environmental signals, including glucose, insulin, and lipids. Subsequently, changes in environmental signals drive metabolic reprogramming, impair immune cell function, and ultimately contribute to diabetes-associated complications. We review here recent studies on changes in innate immune cell metabolism, especially in myeloid cells, that are driven by environmental signals relevant to diabetes, and discuss therapeutic perspectives of targeting metabolism of immune cells in diabetes.
Collapse
Affiliation(s)
- Jacqueline M Ratter
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands; Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Cees J Tack
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands; Department for Genomics and Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Rinke Stienstra
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands; Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands.
| |
Collapse
|
42
|
Bao X, Borné Y, Johnson L, Muhammad IF, Persson M, Niu K, Engström G. Comparing the inflammatory profiles for incidence of diabetes mellitus and cardiovascular diseases: a prospective study exploring the 'common soil' hypothesis. Cardiovasc Diabetol 2018; 17:87. [PMID: 29895294 PMCID: PMC5996509 DOI: 10.1186/s12933-018-0733-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 06/06/2018] [Indexed: 12/15/2022] Open
Abstract
Background Chronic low-grade inflammation and associated insulin resistance and metabolic abnormalities have been proposed as ‘common soil’ for diabetes mellitus (DM) and cardiovascular disease (CVD). This paper aimed to investigate the inflammatory profiles of DM and CVD and to distinguish their shared and specific markers. Methods Based on the Malmö Diet and Cancer cohort, total and differential leukocyte counts were measured in 25,969 participants without previous DM or CVD and were studied in relation to incident DM (mean follow-up 17.4 ± 5.58 years) and incident CVD (i.e., coronary events, including fatal and nonfatal myocardial infarction, or stroke); mean follow-up 17.7 ± 5.46 years, using multivariable Cox regression models. Furthermore, plasma concentrations of another seven inflammatory markers were examined in relation to incident DM and incident CVD in a sub-cohort of 4658 participants. The associations of each inflammatory marker with incident DM versus incident CVD were compared using the Lunn–McNeil competing risks approach. In sensitivity analyses, those who developed both DM and CVD during follow-up were excluded. Results After adjustment for conventional risk factors, total and differential leukocyte counts, orosomucoid, and C-reactive protein were associated with an increased risk of both DM and CVD. Neutrophil to lymphocyte ratio, ceruloplasmin, alpha1-antitrypsin and soluble urokinase plasminogen activator receptor predicted increased risk of CVD but not DM, while haptoglobin and complement C3 showed the opposite pattern. In competing risks analyses, lymphocyte count and complement C3 had stronger associations with risk of DM than with risk of CVD (p for equal associations = 0.020 and 0.006). The reverse was true for neutrophil to lymphocyte ratio (p for equal associations = 0.025). Results were consistent in sensitivity analyses. Conclusions The results indicated substantial similarities in the inflammatory profiles associated with DM and CVD. However, there are also significant differences. These findings may help discriminate between individuals at elevated risk of DM and those at elevated risk of CVD, which is a prerequisite for targeted therapies. Electronic supplementary material The online version of this article (10.1186/s12933-018-0733-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xue Bao
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China. .,Department of Clinical Sciences, Lund University, CRC 60:13, Jan Waldenströms gata 35, 20502, Malmö, Sweden.
| | - Yan Borné
- Department of Clinical Sciences, Lund University, CRC 60:13, Jan Waldenströms gata 35, 20502, Malmö, Sweden
| | - Linda Johnson
- Department of Clinical Sciences, Lund University, CRC 60:13, Jan Waldenströms gata 35, 20502, Malmö, Sweden
| | - Iram Faqir Muhammad
- Department of Clinical Sciences, Lund University, CRC 60:13, Jan Waldenströms gata 35, 20502, Malmö, Sweden
| | - Margaretha Persson
- Department of Clinical Sciences, Lund University, CRC 60:13, Jan Waldenströms gata 35, 20502, Malmö, Sweden
| | - Kaijun Niu
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Gunnar Engström
- Department of Clinical Sciences, Lund University, CRC 60:13, Jan Waldenströms gata 35, 20502, Malmö, Sweden
| |
Collapse
|
43
|
ten Klooster JP, Sotiriou A, Boeren S, Vaessen S, Vervoort J, Pieters R. Type 2 diabetes-related proteins derived from an in vitro model of inflamed fat tissue. Arch Biochem Biophys 2018. [PMID: 29526533 DOI: 10.1016/j.abb.2018.03.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
|
44
|
Hertle E, Arts ICW, van der Kallen CJH, Feskens EJM, Schalkwijk CG, Stehouwer CDA, van Greevenbroek MMJ. Classical Pathway of Complement Activation: Longitudinal Associations of C1q and C1-INH With Cardiovascular Outcomes: The CODAM Study (Cohort on Diabetes and Atherosclerosis Maastricht)-Brief Report. Arterioscler Thromb Vasc Biol 2018; 38:1242-1244. [PMID: 29567681 DOI: 10.1161/atvbaha.118.310806] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 02/27/2018] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The classical complement pathway has been assigned both protective and pathological effects in cardiovascular disease (CVD), but human data are lacking. We determined the associations of the pattern recognition factor C1q and the regulator C1-INH (C1-inhibitor) with incident CVD, carotid intima-media thickness, endothelial dysfunction, and low-grade inflammation. APPROACH AND RESULTS Baseline concentrations of C1q and C1-INH were measured in the CODAM study (Cohort on Diabetes and Atherosclerosis Maastricht; n=574; 61% men; age, 60±7 years). The 7-year incidence of CVD in participants free of CVD at baseline was evaluated using logistic regression analyses (n=342; 73 cases). The lowest incidence of CVD was observed in the middle tertile of C1q (Tlow compared with Tmiddle: odds ratio, 2.38 [95% confidence interval, 1.14-4.95]; Thigh compared with Tmiddle: odds ratio, 1.96 [95% confidence interval, 0.94-4.07]). C1-INH was not associated with CVD. During the 7-year follow-up period, C1q and C1-INH were not, or inconsistently, associated with carotid intima-media thickness or with biomarker scores reflecting endothelial dysfunction and low-grade inflammation. CONCLUSIONS Our results suggest a nonlinear association between C1q and incident CVD. This supports the concept that early steps in classical pathway activation may have both protective and pathological effects on human CVD.
Collapse
Affiliation(s)
- Elisabeth Hertle
- From the CARIM School for Cardiovascular Diseases (E.H., I.C.W.A., C.J.H.v.d.K., C.G.S., C.D.A.S., M.M.J.v.G.).,Department of Internal Medicine (E.H., C.J.H.v.d.K., C.G.S., C.D.A.S., M.M.J.v.G.)
| | - Ilja C W Arts
- From the CARIM School for Cardiovascular Diseases (E.H., I.C.W.A., C.J.H.v.d.K., C.G.S., C.D.A.S., M.M.J.v.G.).,Department of Epidemiology (I.C.W.A.).,Maastricht Centre for Systems Biology (I.C.W.A.), Maastricht University, The Netherlands
| | - Carla J H van der Kallen
- From the CARIM School for Cardiovascular Diseases (E.H., I.C.W.A., C.J.H.v.d.K., C.G.S., C.D.A.S., M.M.J.v.G.).,Department of Internal Medicine (E.H., C.J.H.v.d.K., C.G.S., C.D.A.S., M.M.J.v.G.)
| | - Edith J M Feskens
- Division of Human Nutrition, Section of Nutrition and Epidemiology, Wageningen University, The Netherlands (E.J.M.F.)
| | - Casper G Schalkwijk
- From the CARIM School for Cardiovascular Diseases (E.H., I.C.W.A., C.J.H.v.d.K., C.G.S., C.D.A.S., M.M.J.v.G.).,Department of Internal Medicine (E.H., C.J.H.v.d.K., C.G.S., C.D.A.S., M.M.J.v.G.)
| | - Coen D A Stehouwer
- From the CARIM School for Cardiovascular Diseases (E.H., I.C.W.A., C.J.H.v.d.K., C.G.S., C.D.A.S., M.M.J.v.G.).,Department of Internal Medicine (E.H., C.J.H.v.d.K., C.G.S., C.D.A.S., M.M.J.v.G.)
| | - Marleen M J van Greevenbroek
- From the CARIM School for Cardiovascular Diseases (E.H., I.C.W.A., C.J.H.v.d.K., C.G.S., C.D.A.S., M.M.J.v.G.) .,Department of Internal Medicine (E.H., C.J.H.v.d.K., C.G.S., C.D.A.S., M.M.J.v.G.)
| |
Collapse
|
45
|
Atanes P, Ruz-Maldonado I, Pingitore A, Hawkes R, Liu B, Zhao M, Huang GC, Persaud SJ, Amisten S. C3aR and C5aR1 act as key regulators of human and mouse β-cell function. Cell Mol Life Sci 2018; 75:715-726. [PMID: 28921001 PMCID: PMC5769825 DOI: 10.1007/s00018-017-2655-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 08/22/2017] [Accepted: 09/06/2017] [Indexed: 12/23/2022]
Abstract
AIMS Complement components 3 and 5 (C3 and C5) play essential roles in the complement system, generating C3a and C5a peptides that are best known as chemotactic and inflammatory factors. In this study we characterised islet expression of C3 and C5 complement components, and the impact of C3aR and C5aR1 activation on islet function and viability. MATERIALS AND METHODS Human and mouse islet mRNAs encoding key elements of the complement system were quantified by qPCR and distribution of C3 and C5 proteins was determined by immunohistochemistry. Activation of C3aR and C5aR1 was determined using DiscoverX beta-arrestin assays. Insulin secretion from human and mouse islets was measured by radioimmunoassay, and intracellular calcium ([Ca2+]i), ATP generation and apoptosis were assessed by standard techniques. RESULTS C3 and C5 proteins and C3aR and C5aR1 were expressed by human and mouse islets, and C3 and C5 were mainly localised to β- and α-cells. Conditioned media from islets exposed for 1 h to 5.5 and 20 mM glucose stimulated C3aR and C5aR1-driven beta-arrestin recruitment. Activation of C3aR and C5aR1 potentiated glucose-induced insulin secretion from human and mouse islets, increased [Ca2+]i and ATP generation, and protected islets against apoptosis induced by a pro-apoptotic cytokine cocktail or palmitate. CONCLUSIONS Our observations demonstrate a functional link between activation of components of the innate immune system and improved β-cell function, suggesting that low-level chronic inflammation may improve glucose homeostasis through direct effects on β-cells.
Collapse
Affiliation(s)
- Patricio Atanes
- Diabetes Research Group, Division of Diabetes and Nutritional Sciences, Faculty of Life Sciences and Medicine, Hodgkin Building, King's College London, Guy's Campus, London, SE1 1UL, UK.
| | - Inmaculada Ruz-Maldonado
- Diabetes Research Group, Division of Diabetes and Nutritional Sciences, Faculty of Life Sciences and Medicine, Hodgkin Building, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Attilio Pingitore
- Diabetes Research Group, Division of Diabetes and Nutritional Sciences, Faculty of Life Sciences and Medicine, Hodgkin Building, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Ross Hawkes
- Diabetes Research Group, Division of Diabetes and Nutritional Sciences, Faculty of Life Sciences and Medicine, Hodgkin Building, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Bo Liu
- Diabetes Research Group, Division of Diabetes and Nutritional Sciences, Faculty of Life Sciences and Medicine, Hodgkin Building, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Min Zhao
- Diabetes Research Group, Division of Diabetes and Nutritional Sciences, Faculty of Life Sciences and Medicine, Hodgkin Building, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Guo Cai Huang
- Diabetes Research Group, Division of Diabetes and Nutritional Sciences, Faculty of Life Sciences and Medicine, Hodgkin Building, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Shanta J Persaud
- Diabetes Research Group, Division of Diabetes and Nutritional Sciences, Faculty of Life Sciences and Medicine, Hodgkin Building, King's College London, Guy's Campus, London, SE1 1UL, UK.
| | - Stefan Amisten
- Diabetes Research Group, Division of Diabetes and Nutritional Sciences, Faculty of Life Sciences and Medicine, Hodgkin Building, King's College London, Guy's Campus, London, SE1 1UL, UK
| |
Collapse
|
46
|
Mishra I, Jones C, Patel V, Deodhar S, Cistola DP. Early detection of metabolic dysregulation using water T 2 analysis of biobanked samples. Diabetes Metab Syndr Obes 2018; 11:807-818. [PMID: 30538517 PMCID: PMC6260129 DOI: 10.2147/dmso.s180655] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND The ability to use frozen biobanked samples from cohort studies and clinical trials is critically important for biomarker discovery and validation. Here we investigated whether plasma and serum water transverse relaxation times (T2) from frozen biobanked samples could be used as biomarkers for metabolic syndrome (MetS) and its underlying conditions, specifically insulin resistance, dyslipidemia, and subclinical inflammation. METHODS Plasma and serum aliquots from 44 asymptomatic, non-diabetic human subjects were biobanked at -80°C for 7-9 months. Water T2 measurements were recorded at 37°C on 50 µL of unmodified plasma or serum using benchtop nuclear magnetic resonance relaxometry. The T2 values for freshly drawn and once-frozen-thawed ("frozen") samples were compared using Huber M-values (M), Lin concordance correlation coefficients (ρc), and Bland-Altman plots. Water T2 values from frozen plasma and serum samples were compared with >130 metabolic biomarkers and analyzed using multi-variable linear/logistic regression and ROC curves. RESULTS Frozen plasma water T2 values were highly correlated with fresh (M=0.94, 95% CI 0.89, 0.97) but showed a lower level of agreement (ρc=0.74, 95% CI 0.62, 0.82) because of an average offset of -5.6% (-7.1% for serum). Despite the offset, frozen plasma water T2 was strongly correlated with markers of hyperinsulinemia, dyslipidemia, and inflammation and detected these conditions with 89% sensitivity and 91% specificity (100%/63% for serum). Using optimized cut points, frozen plasma and serum water T2 detected hyperinsulinemia, dyslipidemia, and inflammation in 23 of 44 subjects, including nine with an early stage of metabolic dysregulation that did not meet the clinical thresholds for prediabetes or MetS. CONCLUSION Plasma and serum water T2 values from once-frozen-thawed biobanked samples detect metabolic dysregulation with high sensitivity and specificity. However, the cut points for frozen biobanked samples must be calibrated independent of those for freshly drawn plasma and serum.
Collapse
Affiliation(s)
- Ina Mishra
- Nanoparticle Diagnostics Laboratory, Institute for Cardiovascular and Metabolic Diseases, Department of Physiology & Anatomy, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA,
- Center of Emphasis in Diabetes & Metabolism, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, 79905, USA,
| | - Clinton Jones
- Nanoparticle Diagnostics Laboratory, Institute for Cardiovascular and Metabolic Diseases, Department of Physiology & Anatomy, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA,
- Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Vipulkumar Patel
- Nanoparticle Diagnostics Laboratory, Institute for Cardiovascular and Metabolic Diseases, Department of Physiology & Anatomy, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA,
- Center of Emphasis in Diabetes & Metabolism, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, 79905, USA,
| | - Sneha Deodhar
- Nanoparticle Diagnostics Laboratory, Institute for Cardiovascular and Metabolic Diseases, Department of Physiology & Anatomy, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA,
| | - David P Cistola
- Nanoparticle Diagnostics Laboratory, Institute for Cardiovascular and Metabolic Diseases, Department of Physiology & Anatomy, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA,
- Center of Emphasis in Diabetes & Metabolism, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, 79905, USA,
| |
Collapse
|
47
|
Karkhaneh M, Qorbani M, Mohajeri-Tehrani MR, Hoseini S. Association of serum complement C3 with metabolic syndrome components in normal weight obese women. J Diabetes Metab Disord 2017; 16:49. [PMID: 29299442 PMCID: PMC5745599 DOI: 10.1186/s40200-017-0330-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 11/28/2017] [Indexed: 01/21/2023]
Abstract
BACKGROUND Increased serum complement C3 has been related to body fat mass, metabolic syndrome and chronic diseases. The purpose of this study was to evaluate the levels of C3 in the subjects of normal weight obese (hereafter NWO) as well as their possible relationships with metabolic syndrome and inflammation. METHODS In this case-control study, 40 obese women with normal weight (body mass index (BMI) = 18.5-24.9 kg/m2) and body fat percentage above 30% (fat mass (FM) > 30%) and 30 non-obese women (BMI = 18.5-24.9 kg/m2) and fat percentage less than 25% (FM < 25%) were selected as the study sample. Body composition was analyzed using Bio Impedance analyzer. Blood samples were then collected and analyzed for fasting serum concentration of lipid components of metabolic syndrome, insulin, serum complement C3 and High sensitivity C reactive protein (hsCRP). RESULTS Mean waist and hip circumferences in NWO was higher than non-NWO (74.78 ± 4.81 versus 70.76 ± 2.91 and 99.12 ± 4.32 versus 93.16 ± 2/91, respectively, P-value < 0.001). However, the mean waist-to-hip ratio did not differ significantly (p = 0.448). The mean fasting serum concentration of complement C3, hsCRP and insulin was higher in NWO compared to that in non-NWO (P-value < 0.05). Moreover, insulin sensitivity in NWO was lower than that in non-NWO (0.357 versus 0.374, p-value = 0.043). Moreover, a significant correlation was found between body fat percentage and fasting serum complement C3 and insulin concentration (r = 0.417 and r = 0.254, p-value < 0.005, respectively). CONCLUSION Obese women with normal body mass index but high body fat percentage have higher serum C3 and are at a higher risk for metabolic dysregulation and metabolic syndrome than the healthy non-obese subjects.
Collapse
Affiliation(s)
- Maryam Karkhaneh
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Mostafa Qorbani
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohamad Reza Mohajeri-Tehrani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeed Hoseini
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Hojatdost street, Naderi street, Keshavarz Blv., Tehran, Iran
| |
Collapse
|
48
|
Vlaicu SI, Tatomir A, Boodhoo D, Vesa S, Mircea PA, Rus H. The role of complement system in adipose tissue-related inflammation. Immunol Res 2017; 64:653-64. [PMID: 26754764 DOI: 10.1007/s12026-015-8783-5] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
As the common factor linking adipose tissue to the metabolic context of obesity, insulin resistance and atherosclerosis are associated with a low-grade chronic inflammatory status, to which the complement system is an important contributor. Adipose tissue synthesizes complement proteins and is a target of complement activation. C3a-desArg/acylation-stimulating protein stimulates lipogenesis and affects lipid metabolism. The C3a receptor and C5aR are involved in the development of adipocytes' insulin resistance through macrophage infiltration and the activation of adipose tissue. The terminal complement pathway has been found to be instrumental in promoting hyperglycemia-associated tissue damage, which is characteristic of the major vascular complications of diabetes mellitus and diabetic ketoacidosis. As a mediator of the effects of the terminal complement complex C5b-9, RGC-32 has an impact on energy expenditure as well as lipid and glucose metabolic homeostasis. All of this evidence, taken together, indicates an important role for complement activation in metabolic diseases.
Collapse
Affiliation(s)
- Sonia I Vlaicu
- Department of Neurology, University of Maryland, School of Medicine, 655 W Baltimore St, BRB 12-033, Baltimore, MD, 21201, USA.,Department of Internal Medicine, "Iuliu Haţieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Alexandru Tatomir
- Department of Neurology, University of Maryland, School of Medicine, 655 W Baltimore St, BRB 12-033, Baltimore, MD, 21201, USA.,Research Service, Veterans Administration Maryland Health Care System, Baltimore, MD, USA
| | - Dallas Boodhoo
- Department of Neurology, University of Maryland, School of Medicine, 655 W Baltimore St, BRB 12-033, Baltimore, MD, 21201, USA
| | - Stefan Vesa
- Department of Pharmacology, Toxicology and Clinical Pharmacology, "Iuliu Haţieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Petru A Mircea
- Department of Internal Medicine, "Iuliu Haţieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Horea Rus
- Department of Neurology, University of Maryland, School of Medicine, 655 W Baltimore St, BRB 12-033, Baltimore, MD, 21201, USA. .,Research Service, Veterans Administration Maryland Health Care System, Baltimore, MD, USA. .,Veterans Administration Multiple Sclerosis Center of Excellence, Baltimore, MD, USA.
| |
Collapse
|
49
|
Gómez-Abril SÁ, Morillas-Ariño C, Ponce-Marco JL, Torres-Sánchez T, Delgado-Gomis F, Hernández-Mijares A, Rocha M. Short- and Long-Term Effects of Weight Loss on the Complement Component C3 After Laparoscopic Gastric Bypass in Obese Patients. Obes Surg 2017; 26:2756-2763. [PMID: 27143095 DOI: 10.1007/s11695-016-2195-z] [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] [Indexed: 12/22/2022]
Abstract
BACKGROUND The C3 complement component (C3c) is increasingly recognized as a cardiometabolic risk factor, but how it is affected after weight loss through gastric bypass is a question yet to be answered. METHODS A total of 66 obese patients underwent laparoscopic gastric bypass. Anthropometric parameters, total cholesterol (TC), triglycerides, high-density lipoprotein cholesterol (HDLc), low-density lipoprotein cholesterol (LDLc), glucose, insulin, HOMA-IR, liver enzymes, high-sensitivity C-reactive protein (hsCRP), and C3c levels were evaluated at baseline and at 1 and 5 years post-surgery. RESULTS All anthropometric and biochemical parameters improved significantly after surgery, although a deterioration was detected with respect to the percentage of excess of weight loss, insulin, TC, LDLc, and lactate dehydrogenase 5 years post-surgery. Despite this, a remission rate of 84 % was observed in the presence of metabolic syndrome after 5 years follow-up. hsCRP and C3c were reduced significantly after surgery and maintained throughout the experimental period. In addition, C3c was correlated with BMI and insulin at all time points. The multivariate regression model, in which C3c was a dependent variable, revealed that aspartate aminotransferase and BMI were independent variables at baseline, alkaline phosphatase and insulin were independent at 1 year post-surgery, and insulin, BMI, and TC were independent at 5 years post-surgery. CONCLUSIONS C3c may be a marker of the chronic inflammatory process underlying insulin resistance. Its association with BMI and liver enzymes supports a major role in metabolic activity, although future research is needed to clarify the nature of the molecular mechanisms involved and the physiological significance of these findings.
Collapse
Affiliation(s)
- Segundo Á Gómez-Abril
- Department of General and Digestive Surgery, University Hospital Doctor Peset-FISABIO, Valencia, Spain
| | - Carlos Morillas-Ariño
- Service of Endocrinology and Nutrition, University Hospital Doctor Peset-FISABIO, Av. Gaspar, Aguilar 90, 46017, Valencia, Spain
| | - Jose L Ponce-Marco
- Department of General and Digestive Surgery, University Hospital La Fe, Valencia, Spain
| | - Teresa Torres-Sánchez
- Department of General and Digestive Surgery, University Hospital Doctor Peset-FISABIO, Valencia, Spain
| | - Fernando Delgado-Gomis
- Department of General and Digestive Surgery, University Hospital Doctor Peset-FISABIO, Valencia, Spain
| | - Antonio Hernández-Mijares
- Service of Endocrinology and Nutrition, University Hospital Doctor Peset-FISABIO, Av. Gaspar, Aguilar 90, 46017, Valencia, Spain. .,Institute of Health Research INCLIVA, University of Valencia, Valencia, Spain. .,Department of Medicine, University of Valencia, Valencia, Spain.
| | - Milagros Rocha
- Service of Endocrinology and Nutrition, University Hospital Doctor Peset-FISABIO, Av. Gaspar, Aguilar 90, 46017, Valencia, Spain. .,Institute of Health Research INCLIVA, University of Valencia, Valencia, Spain. .,CIBER CB06/04/0071 Research Group, CIBER Hepatic and Digestive Diseases, University of Valencia, Av Blasco Ibáñez 15, 46010, Valencia, Spain.
| |
Collapse
|
50
|
Borné Y, Muhammad IF, Lorés-Motta L, Hedblad B, Nilsson PM, Melander O, de Jong EK, Blom AM, den Hollander AI, Engström G. Complement C3 Associates With Incidence of Diabetes, but No Evidence of a Causal Relationship. J Clin Endocrinol Metab 2017; 102:4477-4485. [PMID: 29029276 DOI: 10.1210/jc.2017-00948] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 09/26/2017] [Indexed: 01/03/2023]
Abstract
PURPOSE This study explored whether complement factor 3 (C3) in plasma is associated with incidence of diabetes in a population-based cohort. We also identified genetic variants related to C3 and explored whether C3 and diabetes share common genetic determinants. METHODS C3 was analyzed in plasma from 4368 nondiabetic subjects, 46 to 68 years old, from the Malmö Diet and Cancer Study. Incidence of diabetes was studied in relationship to C3 levels during 17.7± 4.4 years of follow-up. Genotypes associated with C3 were identified in a genome-wide association study. Diabetes Genetics Replication and Meta-Analysis and the European Genetic Database were used for in silico look-up. RESULTS In all, 538 (12.3%) subjects developed diabetes during 18 years of follow-up. High C3 was significantly associated with incidence of diabetes after risk factor adjustments (hazard ratio comparing 4th vs 1st quartile, 1.54 (95% confidence interval, 1.13 to 2.09; P = 0.005). C3 was associated with polymorphisms at the complement factor H locus (P < 10-8). However, no relationship with diabetes was observed for this locus. Another eight loci were associated with C3 with P < 10-5. One of them, the glucose kinase regulatory protein (GCKR) locus, has been previously associated with diabetes. The relationship between C3 levels and the GCKR locus was replicated in the European Genetic Database cohort. CONCLUSIONS Plasma concentration of C3 is a risk marker for incidence of diabetes. The results suggest that this association could, in part, be explained by pleiotropic effects related to the GCKR gene.
Collapse
Affiliation(s)
- Yan Borné
- Department of Clinical Sciences, Lund University, Sweden
| | | | - Laura Lorés-Motta
- Department of Ophthalmology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, The Netherlands
| | - Bo Hedblad
- Department of Clinical Sciences, Lund University, Sweden
| | | | - Olle Melander
- Department of Clinical Sciences, Lund University, Sweden
| | - Eiko K de Jong
- Department of Ophthalmology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, The Netherlands
| | - Anna M Blom
- Department of Translational Medicine, Lund University, Sweden
| | - Anneke I den Hollander
- Department of Ophthalmology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, The Netherlands
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, The Netherlands
| | | |
Collapse
|