1
|
Rodríguez-Espinosa D, Cuadrado-Payán E, Morantes L, Gomez M, Maduell F, Broseta JJ. Lipid and immunophenotypic profiles in hemodialysis patients with citrate vs. acetate dialysates. Front Cardiovasc Med 2025; 12:1497353. [PMID: 40276257 PMCID: PMC12018436 DOI: 10.3389/fcvm.2025.1497353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 03/18/2025] [Indexed: 04/26/2025] Open
Abstract
Background Chronic kidney disease (CKD) is a significant cardiovascular (CV) risk factor, with dialysis-dependent CKD (DD-CKD) patients facing high mortality rates. Hypercholesterolemia is another crucial CV risk factor, typically managed with lipid-lowering therapy, though its efficacy in DD-CKD remains uncertain. Evidence shows mixed results regarding the benefits of statins in these patients. Citrate-based dialysates are known to reduce inflammatory biomarkers compared to acetate-based ones, potentially impacting lipid profiles and immune responses. This study aimed to determine the effects of citrate vs. acetate dialysate on lipid profiles and immunophenotypes in DD-CKD patients. Methods This unicentric, cross-over, prospective study included 21 hemodialysis patients (10 males, 11 females, average age 62.25 years). Each patient underwent 24 dialysis sessions (12 with each dialysate) and acted as their own control. Lipid profiles, immunological parameters, and nutritional and inflammatory markers were measured before the last session with each dialysate. Results After twelve dialysis sessions with citrate dialysate (CD), compared to acetate dialysate (AD), there was a statistically significant decline in TG and remnant cholesterol, with a decrease in HDL and an increase in LDL. Regarding immunology, C3 complement levels were higher, while CD3+ CD8+ and CD16+ 56+ lymphocytes were lower. Finally, total lymphocytes were lower with AD than with CD. We found no difference in predialysis nutritional nor inflammatory parameters except for ESR, which was higher when subjects used CD than AD. Conclusion There are significant differences in lipid and immunophenotypic profiles with CD in comparison to AD. Interestingly, there could be an advantageous profile given the reduced amount of remnant cholesterol and TG. However, further studies are needed to understand if the observed changes lead to beneficial hard clinical outcomes in DD-CKD patients.
Collapse
Affiliation(s)
| | | | | | | | | | - José Jesús Broseta
- Department of Nephrology and Renal Transplantation, Hospital Clínic of Barcelona, Barcelona, Spain
| |
Collapse
|
2
|
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
|
3
|
Okumuş EB, Böke ÖB, Turhan SŞ, Doğan A. From development to future prospects: The adipose tissue & adipose tissue organoids. Life Sci 2024; 351:122758. [PMID: 38823504 DOI: 10.1016/j.lfs.2024.122758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/22/2024] [Accepted: 05/27/2024] [Indexed: 06/03/2024]
Abstract
Living organisms store their energy in different forms of fats including lipid droplets, triacylglycerols, and steryl esters. In mammals and some non-mammal species, the energy is stored in adipose tissue which is the innervated specialized connective tissue that incorporates a variety of cell types such as macrophages, fibroblasts, pericytes, endothelial cells, adipocytes, blood cells, and several kinds of immune cells. Adipose tissue is so complex that the scope of its function is not only limited to energy storage, it also encompasses to thermogenesis, mechanical support, and immune defense. Since defects and complications in adipose tissue are heavily related to certain chronic diseases such as obesity, cardiovascular diseases, type 2 diabetes, insulin resistance, and cholesterol metabolism defects, it is important to further study adipose tissue to enlighten further mechanisms behind those diseases to develop possible therapeutic approaches. Adipose organoids are accepted as very promising tools for studying fat tissue development and its underlying molecular mechanisms, due to their high recapitulation of the adipose tissue in vitro. These organoids can be either derived using stromal vascular fractions or pluripotent stem cells. Due to their great vascularization capacity and previously reported incontrovertible regulatory role in insulin sensitivity and blood glucose levels, adipose organoids hold great potential to become an excellent candidate for the source of stem cell therapy. In this review, adipose tissue types and their corresponding developmental stages and functions, the importance of adipose organoids, and the potential they hold will be discussed in detail.
Collapse
Affiliation(s)
- Ezgi Bulut Okumuş
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, İstanbul, Turkey
| | - Özüm Begüm Böke
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, İstanbul, Turkey
| | - Selinay Şenkal Turhan
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, İstanbul, Turkey
| | - Ayşegül Doğan
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, İstanbul, Turkey.
| |
Collapse
|
4
|
Deng T, Liang M, Du L, Li K, Li J, Qian L, Xue Q, Qiu S, Xu L, Zhang L, Gao X, Li J, Lan X, Gao H. Transcriptome Analysis of Compensatory Growth and Meat Quality Alteration after Varied Restricted Feeding Conditions in Beef Cattle. Int J Mol Sci 2024; 25:2704. [PMID: 38473950 DOI: 10.3390/ijms25052704] [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: 01/24/2024] [Revised: 02/17/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Compensatory growth (CG) is a physiological response that accelerates growth following a period of nutrient limitation, with the potential to improve growth efficiency and meat quality in cattle. However, the underlying molecular mechanisms remain poorly understood. In this study, 60 Huaxi cattle were divided into one ad libitum feeding (ALF) group and two restricted feeding groups (75% restricted, RF75; 50% restricted, RF50) undergoing a short-term restriction period followed by evaluation of CG. Detailed comparisons of growth performance during the experimental period, as well as carcass and meat quality traits, were conducted, complemented by a comprehensive transcriptome analysis of the longissimus dorsi muscle using differential expression analysis, gene set enrichment analysis (GSEA), gene set variation analysis (GSVA), and weighted correlation network analysis (WGCNA). The results showed that irrespective of the restriction degree, the restricted animals exhibited CG, achieving final body weights comparable to the ALF group. Compensating animals showed differences in meat quality traits, such as pH, cooking loss, and fat content, compared to the ALF group. Transcriptomic analysis revealed 57 genes and 31 pathways differentially regulated during CG, covering immune response, acid-lipid metabolism, and protein synthesis. Notably, complement-coagulation-fibrinolytic system synergy was identified as potentially responsible for meat quality optimization in RF75. This study provides novel and valuable genetic insights into the regulatory mechanisms of CG in beef cattle.
Collapse
Affiliation(s)
- Tianyu Deng
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Mang Liang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Lili Du
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Keanning Li
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Jinnan Li
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Li Qian
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Qingqing Xue
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Shiyuan Qiu
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Lingyang Xu
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Lupei Zhang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xue Gao
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Junya Li
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xianyong Lan
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Huijiang Gao
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| |
Collapse
|
5
|
Nguyen VD, Hughes TR, Zhou Y. From complement to complosome in non-alcoholic fatty liver disease: When location matters. Liver Int 2024; 44:316-329. [PMID: 38010880 DOI: 10.1111/liv.15796] [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: 09/10/2023] [Revised: 10/21/2023] [Accepted: 11/09/2023] [Indexed: 11/29/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a growing public health threat and becoming the leading cause of liver transplantation. Nevertheless, no approved specific treatment is currently available for NAFLD. The pathogenesis of NAFLD is multifaceted and not yet fully understood. Accumulating evidence suggests a significant role of the complement system in the development and progression of NAFLD. Here, we provide an overview of the complement system, incorporating the novel concept of complosome, and summarise the up-to-date evidence elucidating the association between complement dysregulation and the pathogenesis of NAFLD. In this process, the extracellular complement system is activated through various pathways, thereby directly contributing to, or working together with other immune cells in the disease development and progression. We also introduce the complosome and assess the evidence that implicates its potential influence in NAFLD through its direct impact on hepatocytes or non-parenchymal liver cells. Additionally, we expound upon how complement system and the complosome may exert their effects in relation with hepatic zonation in NAFLD. Furthermore, we discuss the potential therapeutic implications of targeting the complement system, extracellularly and intracellularly, for NAFLD treatment. Finally, we present future perspectives towards a better understanding of the complement system's contribution to NAFLD.
Collapse
Affiliation(s)
- Van-Dien Nguyen
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| | - Timothy R Hughes
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| | - You Zhou
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| |
Collapse
|
6
|
Nakamura M, Imaoka M, Sakai K, Kubo T, Imai R, Hida M, Tazaki F, Orui J, Inoue T, Takeda M. Complement component C3 is associated with body composition parameters and sarcopenia in community-dwelling older adults: a cross-sectional study in Japan. BMC Geriatr 2024; 24:102. [PMID: 38279167 PMCID: PMC10821262 DOI: 10.1186/s12877-024-04720-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 01/18/2024] [Indexed: 01/28/2024] Open
Abstract
BACKGROUND Chronic inflammation is a factor in the pathogenesis of sarcopenia, which is characterized by low muscle mass and reduced strength. Complement C3 is important in the management of the immune network system. This study seeks to determine the relationship between serum C3 levels and body composition and sarcopenia-related status in community-dwelling older adults. METHODS Study participants were 269 older adults living in rural Japan. A bioelectrical impedance analysis device was used to measure body composition parameters including body mass index (BMI), body fat percentage, waist-hip-ratio, and appendicular skeletal muscle mass index (SMI). Muscle function was measured by handgrip strength and 6-m walking speed. The correlation coefficients for C3 level and measurements were calculated using Pearson correlation analysis. Participants were categorized into normal, pre-sarcopenia, dynapenia, or sarcopenia groups. Sarcopenia was defined according to 2019 Asian Working Group for Sarcopenia definition, dynapenia was defined as low muscle function without low muscle mass, and pre-sarcopenia was defined as the presence of low muscle mass only. The C3 threshold score for sarcopenia status was evaluated by receiver operating characteristic curve (ROC) analysis. RESULTS Significant positive correlations were found between C3 and BMI, body fat percentage, and waist-hip ratio in both sexes, and further positive correlations with SMI were found in women. The relationship with body fat percentage was particularly strong. Body composition measurements (BMI, body fat percentage, and waist- hip ratio) and C3 levels were lowest in the sarcopenia group compared with the others. ROC analysis showed that the significant threshold of C3 for discriminating between the normal and sarcopenia groups was 105 mg/dL. Multiple logistic regression analysis showed that participants with C3 < 105 mg/dL had an odds ratio of 3.27 (95% confidence interval, 1.49-7.18) for sarcopenia adjusted by sex, age and body fat percentage. CONCLUSION C3 levels are suggested to be related to body composition and pathophysiological functions of sarcopenia. C3 is expected to become a useful biomarker for sarcopenia, for predicting the onset of the disease and for predicting the effectiveness of interventions.
Collapse
Affiliation(s)
- Misa Nakamura
- Department of Rehabilitation, Osaka Kawasaki Rehabilitation University, 158 Mizuma, Kaizuka, Osaka, 597-0104, Japan.
| | - Masakazu Imaoka
- Department of Rehabilitation, Osaka Kawasaki Rehabilitation University, 158 Mizuma, Kaizuka, Osaka, 597-0104, Japan
| | - Keiko Sakai
- Department of Rehabilitation, Osaka Kawasaki Rehabilitation University, 158 Mizuma, Kaizuka, Osaka, 597-0104, Japan
| | - Takanari Kubo
- Department of Rehabilitation, Osaka Kawasaki Rehabilitation University, 158 Mizuma, Kaizuka, Osaka, 597-0104, Japan
| | - Ryota Imai
- Department of Rehabilitation, Osaka Kawasaki Rehabilitation University, 158 Mizuma, Kaizuka, Osaka, 597-0104, Japan
| | - Mitsumasa Hida
- Department of Rehabilitation, Osaka Kawasaki Rehabilitation University, 158 Mizuma, Kaizuka, Osaka, 597-0104, Japan
| | - Fumie Tazaki
- Department of Rehabilitation, Osaka Kawasaki Rehabilitation University, 158 Mizuma, Kaizuka, Osaka, 597-0104, Japan
| | - Junya Orui
- Department of Rehabilitation, Osaka Kawasaki Rehabilitation University, 158 Mizuma, Kaizuka, Osaka, 597-0104, Japan
| | - Takao Inoue
- Department of Rehabilitation, Osaka Kawasaki Rehabilitation University, 158 Mizuma, Kaizuka, Osaka, 597-0104, Japan
| | - Masatoshi Takeda
- Department of Rehabilitation, Osaka Kawasaki Rehabilitation University, 158 Mizuma, Kaizuka, Osaka, 597-0104, Japan
| |
Collapse
|
7
|
García-González M, Gómez-Bernal F, Quevedo-Abeledo JC, Fernández-Cladera Y, González-Rivero AF, López-Mejías R, Díaz-González F, González-Gay MÁ, Ferraz-Amaro I. HDL Cholesterol Efflux and the Complement System Are Linked in Systemic Lupus Erythematosus. J Clin Med 2023; 12:5405. [PMID: 37629447 PMCID: PMC10455830 DOI: 10.3390/jcm12165405] [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: 07/22/2023] [Revised: 08/07/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
Cholesterol efflux capacity (CEC), the ability of high-density lipoprotein (HDL) cholesterol to accept cholesterol from macrophages, has been linked to cardiovascular events. Systemic lupus erythematosus (SLE) is characterized by the consumption of complement (C) proteins and has been associated with an increased risk of cardiovascular disease. CEC is reduced in SLE patients compared to controls. In the present work, our objective was to analyze whether the disruption of C influences CEC in patients with SLE. New-generation functional assays of the three pathways of the C system were performed in 207 patients with SLE. Additionally, serum levels of inactive (C1q, C2, C3, C4, and factor D) and activated (C3a) molecules, and regulators (C1-inhibitor and factor H) of C system were measured. CEC, using an in vitro assay, and lipoprotein serum concentrations were assessed. Multivariable linear regression analysis was performed to assess the relationship between C system and CEC. After full multivariable analysis, the alternative C cascade functional test showed a significant and negative relationship with CEC. This was also the case for C2 and C3, in which the associations were found to be positive and statistically significant, after adjustment for covariates. In conclusion, C system and CEC are interconnected in patients with SLE.
Collapse
Affiliation(s)
- María García-González
- Division of Rheumatology, Hospital Universitario de Canarias, 38320 Tenerife, Spain; (M.G.-G.); (F.D.-G.)
| | - Fuensanta Gómez-Bernal
- Division of Central Laboratory, Hospital Universitario de Canarias, 38320 Tenerife, Spain; (F.G.-B.); (Y.F.-C.); (A.F.G.-R.)
| | | | - Yolanda Fernández-Cladera
- Division of Central Laboratory, Hospital Universitario de Canarias, 38320 Tenerife, Spain; (F.G.-B.); (Y.F.-C.); (A.F.G.-R.)
| | - Agustín F. González-Rivero
- Division of Central Laboratory, Hospital Universitario de Canarias, 38320 Tenerife, Spain; (F.G.-B.); (Y.F.-C.); (A.F.G.-R.)
| | - Raquel López-Mejías
- Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Instituto de Investigación sanitaria Marqués de Valdecilla (IDIVAL), 39011 Santander, Spain;
| | - Federico Díaz-González
- Division of Rheumatology, Hospital Universitario de Canarias, 38320 Tenerife, Spain; (M.G.-G.); (F.D.-G.)
- Department of Internal Medicine, University of La Laguna (ULL), 38200 Tenerife, Spain
| | - Miguel Á. González-Gay
- Division of Rheumatology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain
- Department of Medicine and Psychiatry, University of Cantabria, 39005 Santander, Spain
| | - Iván Ferraz-Amaro
- Division of Rheumatology, Hospital Universitario de Canarias, 38320 Tenerife, Spain; (M.G.-G.); (F.D.-G.)
- Department of Internal Medicine, University of La Laguna (ULL), 38200 Tenerife, Spain
| |
Collapse
|
8
|
Sun ZJ, Chang DY, Chen M, Zhao MH. Deficiency of CFB attenuates renal tubulointerstitial damage by inhibiting ceramide synthesis in diabetic kidney disease. JCI Insight 2022; 7:156748. [PMID: 36546481 PMCID: PMC9869976 DOI: 10.1172/jci.insight.156748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 11/09/2022] [Indexed: 12/24/2022] Open
Abstract
Accumulating evidence suggests the pathogenic role of immunity and metabolism in diabetic kidney disease (DKD). Herein, we aimed to investigate the effect of complement factor B (CFB) on lipid metabolism in the development of DKD. We found that in patients with diabetic nephropathy, the staining of Bb, CFB, C3a, C5a, and C5b-9 was markedly elevated in renal tubulointerstitium. Cfb-knockout diabetic mice had substantially milder tubulointerstitial injury and less ceramide biosynthesis. The in vitro study demonstrated that cytokine secretion, endoplasmic reticulum stress, oxidative stress, and cell apoptosis were ameliorated in HK-2 cells transfected with siRNA of CFB under high-glucose conditions. Exogenous ceramide supplementation attenuated the protective effect of CFB knockdown in HK-2 cells, while inhibiting ceramide synthases (CERS) with fumonisin B1 in CFB-overexpressing cells rescued the cell injury. CFB knockdown could downregulate the expression of NF-κB p65, which initiates the transcription of CERS3. Furthermore, C3 knockdown abolished CFB-mediated cytokine secretion, NF-κB signaling activation, and subsequently ceramide biosynthesis. Thus, CFB deficiency inhibited activation of the complement alternative pathway and attenuated kidney damage in DKD, especially tubulointerstitial injury, by inhibiting the NF-κB signaling pathway, further blocking the transcription of CERS, which regulates the biosynthesis of ceramide. CFB may be a promising therapeutic target of DKD.
Collapse
Affiliation(s)
- Zi-jun Sun
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China.,Peking University Institute of Nephrology, Beijing, China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China.,Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Dong-yuan Chang
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China.,Peking University Institute of Nephrology, Beijing, China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China.,Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Min Chen
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China.,Peking University Institute of Nephrology, Beijing, China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China.,Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Ming-hui Zhao
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China.,Peking University Institute of Nephrology, Beijing, China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China.,Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Beijing, China
| |
Collapse
|
9
|
Feng L, Zhao Y, Wang WL. Association between complement C3 and the prevalence of metabolic-associated fatty liver disease in a Chinese population: a cross-sectional study. BMJ Open 2021; 11:e051218. [PMID: 34711595 PMCID: PMC8557272 DOI: 10.1136/bmjopen-2021-051218] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVES Recently studies demonstrated that adipose tissue can produce and release complement C3 and serum complement C3 levels were associated with diabetes mellitus, metabolic syndrome and non-alcoholic fatty liver disease (NAFLD). Thus, we plan to investigate the association of complement C3 levels and the presence of metabolic-associated fatty liver disease (MAFLD). DESIGN Observational study with a cross-sectional sample. SETTING This study surveyed 4729 participants in Zhejiang province, China. PARTICIPANTS 55 participants were excluded for acute infection and 1001 participants were excluded for lack of ultrasonography diagnoses and complete or partial absence of laboratory tests. The final sample size was 3673 participants. OUTCOME MEASURES Spearman correlation analysis was used to examine the correlations between complement C3 levels and variables. Binary logistic regression was carried out to evaluate the association between complement C3 levels and the presence of MAFLD after adjustment for demographic and biochemical variables. Mediation effects were used to explore whether insulin resistance (IR), hyperlipidaemia and obesity mediated the association between complement C3 and MAFLD. RESULTS Participants with MAFLD had higher complement C3 levels and complement C3 levels were closely associated with body mass index, waist circumference, alanine aminotransferase, aspartate aminotransferase, γ-glutamyl transpeptidase and homoeostasis model assessment (HOMA)-IR. The presence of MAFLD increased with the increase of complement C3 levels and the presence of MAFLD were highest in the HOMA-IR ≥2.5 participants. We found the OR and Cl of standardised C3 for MAFLD was 1.333 (1.185-1.500), each 1 SD increase in C3 would increase the presence of MAFLD by 33.3%, and obesity partly mediated the effect of complement C3 on the presence of MAFLD. CONCLUSIONS The present results suggest that complement C3 can be used as a risk factor for the presence of MAFLD after adjustment for confounding variables and obesity may partly mediate the effect of complement C3 on the presence of MAFLD.
Collapse
Affiliation(s)
- Limin Feng
- Department of Laboratory Medicine, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Ying Zhao
- Department of Laboratory Medicine, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Wei-Lin Wang
- Department of Hepatobiliary and Pancreatic Surgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, China
| |
Collapse
|
10
|
DeKorver NW, Chaudoin TR, Zhao G, Wang D, Arikkath J, Bonasera SJ. Complement Component C3 Loss leads to Locomotor Deficits and Altered Cerebellar Internal Granule Cell In Vitro Synaptic Protein Expression in C57BL/6 Mice. Mol Neurobiol 2021; 58:5857-5875. [PMID: 34415487 DOI: 10.1007/s12035-021-02480-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 06/24/2021] [Indexed: 01/14/2023]
Abstract
Complement component 3 (C3) expression is increased in the cerebellum of aging mice that demonstrate locomotor impairments and increased excitatory synapse density. However, C3 regulation of locomotion, as well as C3 roles in excitatory synapse function, remains poorly understood. Here, we demonstrate that constitutive loss of C3 function in mice evokes a locomotor phenotype characterized by decreased speed, increased active state locomotor probability, and gait ataxia. C3 loss does not alter metabolism or body mass composition. No evidence of significant muscle weakness or degenerative arthritis was found in C3 knockout mice to explain decreased gait speeds. In an enriched primary cerebellar granule cell culture model, loss of C3 protein results in increased excitatory synaptic density and increased response to KCl depolarization. Our analysis of excitatory synaptic density in the cerebellar internal granule cell and molecular layers did not demonstrate increased synaptic density in vivo, suggesting the presence of compensatory mechanisms regulating synaptic development. Functional deficits in C3 knockout mice are therefore more likely to result from altered synaptic function and/or connectivity than gross synaptic deficits. Our data demonstrate a novel role for complement proteins in cerebellar regulation of locomotor output and control.
Collapse
Affiliation(s)
- Nicholas W DeKorver
- Department of Internal Medicine, Division of Geriatrics, University of Nebraska Medical Center, 3028 Durham Research Center II, Omaha, NE, 68198-5039, USA
| | - Tammy R Chaudoin
- Department of Internal Medicine, Division of Geriatrics, University of Nebraska Medical Center, 3028 Durham Research Center II, Omaha, NE, 68198-5039, USA
| | - Gang Zhao
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, 986125 Nebraska Medical Center, PDD 3020, Omaha, NE, 68198-6125, USA
| | - Dong Wang
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, 986125 Nebraska Medical Center, PDD 3020, Omaha, NE, 68198-6125, USA
| | - Jyothi Arikkath
- Monroe-Meyer Institute, University of Nebraska Medical Center, 3031 Durham Research Center II, Omaha, NE, 68198-5960, USA
| | - Stephen J Bonasera
- Department of Internal Medicine, Division of Geriatrics, University of Nebraska Medical Center, 3028 Durham Research Center II, Omaha, NE, 68198-5039, USA.
| |
Collapse
|
11
|
Martin WP, Conroy C, Naicker SD, Cormican S, Griffin TP, Islam MN, McCole EM, McConnell I, Lamont J, FitzGerald P, Ferguson JP, Richardson C, Logue SE, Griffin MD. Multiplex Serum Biomarker Assays Improve Prediction of Renal and Mortality Outcomes in Chronic Kidney Disease. KIDNEY360 2021; 2:1225-1239. [PMID: 34849485 PMCID: PMC7612046 DOI: 10.34067/kid.0007552020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND We investigated the predictive value of 11 serum biomarkers for renal and mortality end points in people with CKD. METHODS Adults with CKD (n=139) were enrolled from outpatient clinics between February 2014 and November 2016. Biomarker quantification was performed using two multiplex arrays on a clinical-grade analyzer. Relationships between biomarkers and renal and mortality end points were investigated by random forests and Cox proportional hazards regression. RESULTS The cohort was 56% male. The mean age was 63 years and median (IQR) CKD-EPI eGFR was 33 (24-51) ml/min per BSA. A total of 56 (40%) people developed a composite end point defined as ≥40% decline in eGFR, doubling of serum creatinine, RRT, or death over median (IQR) follow-up of 5.4 (4.7-5.7) years. Prediction of the composite end point was better with random forests trained on serum biomarkers compared with clinical variables (area under the curve of 0.81 versus 0.78). The predictive performance of biomarkers was further enhanced when considered alongside clinical variables (area under the curve of 0.83 versus 0.81 for biomarkers alone). Patients (n=27, 19%) with high soluble TNF receptor-1 (≥3 ng/ml) and neutrophil gelatinase-associated lipocalin (≥156 ng/ml), coupled with low complement 3a des-arginine (<2368 ng/ml), almost universally (96%) developed the composite renal and mortality end point. C-reactive protein (adjusted hazard ratio, 1.4; 95% CI, 1.1 to 1.8), neutrophil gelatinase-associated lipocalin (adjusted hazard ratio, 2.8; 95% CI, 1.3 to 6.1) and complement 3a desarginine (adjusted hazard ratio, 0.6; 95% CI, 0.4 to 0.96) independently predicted time to the composite end point. CONCLUSIONS Outpatients with the triad of high soluble TNF receptor-1 and neutrophil gelatinase-associated lipocalin coupled with low complement 3a des-arginine had high adverse event rates over 5-year follow-up. Incorporation of serum biomarkers alongside clinical variables improved prediction of CKD progression and mortality. Our findings require confirmation in larger, more diverse patient cohorts.
Collapse
Affiliation(s)
- William P. Martin
- Diabetes Complications Research Centre, School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Chloe Conroy
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Serika D. Naicker
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, School of Medicine, National University of Ireland Galway, Galway, Ireland
| | - Sarah Cormican
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, School of Medicine, National University of Ireland Galway, Galway, Ireland,Nephrology Services, Galway University Hospitals, Saolta University Health Care Group, Galway, Ireland
| | - Tomás P. Griffin
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, School of Medicine, National University of Ireland Galway, Galway, Ireland,Centre for Diabetes, Endocrinology and Metabolism, Galway University Hospitals, Saolta University Health Care Group, Galway, Ireland
| | - Md Nahidul Islam
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, School of Medicine, National University of Ireland Galway, Galway, Ireland
| | | | - Ivan McConnell
- Randox Laboratories Limited, Crumlin, Antrim, Northern Ireland
| | - John Lamont
- Randox Laboratories Limited, Crumlin, Antrim, Northern Ireland
| | | | - John P. Ferguson
- Health Research Board Clinical Research Facility, National University of Ireland Galway, Galway, Ireland
| | | | - Susan E. Logue
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, School of Medicine, National University of Ireland Galway, Galway, Ireland
| | - Matthew D. Griffin
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, School of Medicine, National University of Ireland Galway, Galway, Ireland,Nephrology Services, Galway University Hospitals, Saolta University Health Care Group, Galway, Ireland
| |
Collapse
|
12
|
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
|
13
|
West EE, Kunz N, Kemper C. Complement and human T cell metabolism: Location, location, location. Immunol Rev 2020; 295:68-81. [PMID: 32166778 PMCID: PMC7261501 DOI: 10.1111/imr.12852] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 02/19/2020] [Accepted: 02/25/2020] [Indexed: 12/26/2022]
Abstract
The complement system represents one of the evolutionary oldest arms of our immune system and is commonly recognized as a liver-derived and serum-active system critical for providing protection against invading pathogens. Recent unexpected findings, however, have defined novel and rather "uncommon" locations and activities of complement. Specifically, the discovery of an intracellularly active complement system-the complosome-and its key role in the regulation of cell metabolic pathways that underly normal human T cell responses have taught us that there is still much to be discovered about this system. Here, we summarize the current knowledge about the emerging functions of the complosome in T cell metabolism. We further place complosome activities among the non-canonical roles of other intracellular innate danger sensing systems and argue that a "location-centric" view of complement evolution could logically justify its close connection with the regulation of basic cell physiology.
Collapse
Affiliation(s)
- Erin E. West
- Complement and Inflammation Research Section, National Heart, Lung and Blood Institute, Bethesda, MD, USA
| | - Natalia Kunz
- Complement and Inflammation Research Section, National Heart, Lung and Blood Institute, Bethesda, MD, USA
| | - Claudia Kemper
- Complement and Inflammation Research Section, National Heart, Lung and Blood Institute, Bethesda, MD, USA
- Faculty of Life Sciences and Medicine, King’s College London, London, UK
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| |
Collapse
|
14
|
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
|
15
|
Saleh J, Al-Maqbali M, Abdel-Hadi D. Role of Complement and Complement-Related Adipokines in Regulation of Energy Metabolism and Fat Storage. Compr Physiol 2019; 9:1411-1429. [PMID: 31688967 DOI: 10.1002/cphy.c170037] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Adipose tissue releases many cytokines and inflammatory factors described as adipokines. In obesity, adipokines released from expanding adipose tissue are implicated in disease progression and metabolic dysfunction. However, mechanisms controlling the progression of adiposity and metabolic complications are not fully understood. It has been suggested that expanding fat mass and sustained release of inflammatory adipokines in adipose tissue lead to hypoxia, oxidative stress, apoptosis, and cellular damage. These changes trigger an immune response involving infiltration of adipose tissue with immune cells, complement activation and generation of factors involved in opsonization and clearance of damaged cells. Abundant evidence now indicates that adipose tissue is an active secretory source of complement and complement-related adipokines that, in addition to their inflammatory role, contribute to the regulation of metabolic function. This article highlights advances in knowledge regarding the role of these adipokines in energy regulation of adipose tissue through modulating lipogenic and lipolytic pathways. Several adipokines will be discussed including adipsin, Factor H, properdin, C3a, Acylation-Stimulating Protein, C1q/TNF-related proteins, and response gene to complement-32 (RGC-32). Interactions between these factors will be described considering their immune-metabolic roles in the adipose tissue microenvironment and their potential contribution to progression of adiposity and metabolic dysfunction. The differential expression and the role of complement factors in gender-related fat partitioning will also be addressed. Identifying lipogenic adipokines and their specific autocrine/paracrine roles may provide means for adipose-tissue-targeted therapeutic interventions that may disrupt the vicious circle of adiposity and disease progression. © 2019 American Physiological Society. Compr Physiol 9:1411-1429, 2019.
Collapse
Affiliation(s)
- Jumana Saleh
- Biochemistry Department, College of Medicine & Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Muna Al-Maqbali
- Biochemistry Department, College of Medicine, Sultan Qaboos University, Muscat, Oman
| | | |
Collapse
|
16
|
Corvillo F, Akinci B. An overview of lipodystrophy and the role of the complement system. Mol Immunol 2019; 112:223-232. [PMID: 31177059 DOI: 10.1016/j.molimm.2019.05.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 05/28/2019] [Accepted: 05/29/2019] [Indexed: 12/19/2022]
Abstract
The complement system is a major component of innate immunity playing essential roles in the destruction of pathogens, the clearance of apoptotic cells and immune complexes, the enhancement of phagocytosis, inflammation, and the modulation of adaptive immune responses. During the last decades, numerous studies have shown that the complement system has key functions in the biology of certain tissues. For example, complement contributes to normal brain and embryonic development and to the homeostasis of lipid metabolism. However, the complement system is subjected to the effective balance between activation-inactivation to maintain complement homeostasis and to prevent self-injury to cells or tissues. When this control is disrupted, serious pathologies eventually develop, such as C3 glomerulopathy, autoimmune conditions and infections. Another heterogeneous group of ultra-rare diseases in which complement abnormalities have been described are the lipodystrophy syndromes. These diseases are characterized by the loss of adipose tissue throughout the entire body or partially. Complement over-activation has been reported in most of the patients with acquired partial lipodystrophy (also called Barraquer-Simons Syndrome) and in some cases of the generalized variety of the disease (Lawrence Syndrome). Even so, the mechanism through which the complement system induces adipose tissue abnormalities remains unclear. This review focuses on describing the link between the complement system and certain forms of lipodystrophy. In addition, we present an overview regarding the clinical presentation, differential diagnosis, classification, and management of patients with lipodystrophy associated with complement abnormalities.
Collapse
Affiliation(s)
- F Corvillo
- Complement Research Group, La Paz University Hospital Research Institute (IdiPAZ), La Paz University Hospital, Madrid, Spain; Center for Biomedical Network Research on Rare Diseases (CIBERER U754), Madrid, Spain.
| | - B Akinci
- Division of Endocrinology, Department of Internal Medicine, Dokuz Eylul University, Izmir, Turkey; Brehm Center for Diabetes Research, Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan, 1000 Wall Street, Room 5313, Ann Arbor, MI, 48105, USA
| |
Collapse
|
17
|
Valverde-Franco G, Tardif G, Mineau F, Paré F, Lussier B, Fahmi H, Pelletier JP, Martel-Pelletier J. High in vivo levels of adipsin lead to increased knee tissue degradation in osteoarthritis: data from humans and animal models. Rheumatology (Oxford) 2018; 57:1851-1860. [PMID: 29982662 DOI: 10.1093/rheumatology/key181] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Indexed: 01/02/2023] Open
Abstract
Objective This study explored the role of the adipokine adipsin in OA. Methods Control and OA articular tissues, cells and serum were obtained from human individuals. Serum adipsin levels of human OA individuals were compared with cartilage volume loss as assessed by MRI at 48 months. Human adipsin expression was determined by PCR, its production in tissues by immunohistochemistry, and in SF and serum by a specific assay. OA was surgically induced in wild-type (Df+/+) and adipsin-deficient (Df-/-) mice, and synovial membrane and cartilage processed for histology and immunohistochemistry. Results Adipsin levels were significantly increased in human OA serum, SF, synovial membrane and cartilage compared with controls, but the expression was similar in chondrocytes, synoviocytes and osteoblasts. Multivariate analysis demonstrated that human serum adipsin levels were significantly associated (P = 0.045) with cartilage volume loss in the lateral compartment of the knee. Destabilization of the medial meniscus-Df-/- mice showed a preservation of the OA synovial membrane and cartilage lesions (P ⩽ 0.026), the latter corroborated by the decreased production of cartilage degradation products and proteases (P ⩽ 0.047). The adipsin effect is likely due to a deficient alternative complement pathway (P ⩽ 0.036). Conclusion In human OA, higher serum adipsin levels were associated with greater cartilage volume loss in the lateral compartment, and adipsin deficiency led to a preservation of knee structure. Importantly, we documented an association between adipsin and OA synovial membrane and cartilage degeneration through the activation of the complement pathway. This study highlights the clinical relevance of adipsin as a valuable biomarker and potential therapeutic target for OA.
Collapse
Affiliation(s)
- Gladys Valverde-Franco
- Osteoarthritis Research Unit, University of Montreal Hospital Research Centre (CRCHUM), Montreal, Canada
| | - Ginette Tardif
- Osteoarthritis Research Unit, University of Montreal Hospital Research Centre (CRCHUM), Montreal, Canada
| | - François Mineau
- Osteoarthritis Research Unit, University of Montreal Hospital Research Centre (CRCHUM), Montreal, Canada
| | - Frédéric Paré
- Osteoarthritis Research Unit, University of Montreal Hospital Research Centre (CRCHUM), Montreal, Canada
| | - Bertrand Lussier
- Osteoarthritis Research Unit, University of Montreal Hospital Research Centre (CRCHUM), Montreal, Canada.,Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, Quebec, Canada
| | - Hassan Fahmi
- Osteoarthritis Research Unit, University of Montreal Hospital Research Centre (CRCHUM), Montreal, Canada
| | - Jean-Pierre Pelletier
- Osteoarthritis Research Unit, University of Montreal Hospital Research Centre (CRCHUM), Montreal, Canada
| | - Johanne Martel-Pelletier
- Osteoarthritis Research Unit, University of Montreal Hospital Research Centre (CRCHUM), Montreal, Canada
| |
Collapse
|
18
|
Gonzalez LL, Garrie K, Turner MD. Type 2 diabetes - An autoinflammatory disease driven by metabolic stress. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3805-3823. [PMID: 30251697 DOI: 10.1016/j.bbadis.2018.08.034] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 08/27/2018] [Indexed: 02/06/2023]
Abstract
Type 2 diabetes has traditionally been viewed as a metabolic disorder characterised by chronic high glucose levels, insulin resistance, and declining insulin secretion from the pancreas. Modern lifestyle, with abundant nutrient supply and reduced physical activity, has resulted in dramatic increases in the rates of obesity-associated disease conditions, including diabetes. The associated excess of nutrients induces a state of systemic low-grade chronic inflammation that results from production and secretion of inflammatory mediators from the expanded pool of activated adipocytes. Here, we review the mechanisms by which obesity induces adipose tissue dysregulation, detailing the roles of adipose tissue secreted factors and their action upon other cells and tissues central to glucose homeostasis and type 2 diabetes. Furthermore, given the emerging importance of adipokines, cytokines and chemokines in disease progression, we suggest that type 2 diabetes should now be viewed as an autoinflammatory disease, albeit one that is driven by metabolic dysregulation.
Collapse
Affiliation(s)
- Laura L Gonzalez
- Interdisciplinary Biomedical Research Centre, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham NG11 8NS, United Kingdom
| | - Karin Garrie
- Interdisciplinary Biomedical Research Centre, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham NG11 8NS, United Kingdom
| | - Mark D Turner
- Interdisciplinary Biomedical Research Centre, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham NG11 8NS, United Kingdom.
| |
Collapse
|
19
|
Núñez K, Thevenot P, Alfadhli A, Cohen A. Complement Activation in Liver Transplantation: Role of Donor Macrosteatosis and Implications in Delayed Graft Function. Int J Mol Sci 2018; 19:1750. [PMID: 29899265 PMCID: PMC6032339 DOI: 10.3390/ijms19061750] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 05/28/2018] [Accepted: 06/08/2018] [Indexed: 12/16/2022] Open
Abstract
The complement system anchors the innate inflammatory response by triggering both cell-mediated and antibody-mediated immune responses against pathogens. The complement system also plays a critical role in sterile tissue injury by responding to damage-associated molecular patterns. The degree and duration of complement activation may be a critical variable controlling the balance between regenerative and destructive inflammation following sterile injury. Recent studies in kidney transplantation suggest that aberrant complement activation may play a significant role in delayed graft function following transplantation, confirming results obtained from rodent models of renal ischemia/reperfusion (I/R) injury. Deactivating the complement cascade through targeting anaphylatoxins (C3a/C5a) might be an effective clinical strategy to dampen reperfusion injury and reduce delayed graft function in liver transplantation. Targeting the complement cascade may be critical in donor livers with mild to moderate steatosis, where elevated lipid burden amplifies stress responses and increases hepatocyte turnover. Steatosis-driven complement activation in the donor liver may also have implications in rejection and thrombolytic complications following transplantation. This review focuses on the roles of complement activation in liver I/R injury, strategies to target complement activation in liver I/R, and potential opportunities to translate these strategies to transplanting donor livers with mild to moderate steatosis.
Collapse
Affiliation(s)
- Kelley Núñez
- Institute of Translational Research, Ochsner Health System, New Orleans, LA 70121, USA.
| | - Paul Thevenot
- Institute of Translational Research, Ochsner Health System, New Orleans, LA 70121, USA.
| | - Abeer Alfadhli
- Institute of Translational Research, Ochsner Health System, New Orleans, LA 70121, USA.
| | - Ari Cohen
- Institute of Translational Research, Ochsner Health System, New Orleans, LA 70121, USA.
| |
Collapse
|
20
|
Kolev M, Markiewski MM. Targeting complement-mediated immunoregulation for cancer immunotherapy. Semin Immunol 2018; 37:85-97. [PMID: 29454575 PMCID: PMC5984681 DOI: 10.1016/j.smim.2018.02.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 01/31/2018] [Accepted: 02/02/2018] [Indexed: 12/21/2022]
Abstract
Complement was initially discovered as an assembly of plasma proteins "complementing" the cytolytic activity of antibodies. However, our current knowledge places this complex system of several plasma proteins, receptors, and regulators in the center of innate immunity as a bridge between the initial innate responses and adaptive immune reactions. Consequently, complement appears to be pivotal for elimination of pathogens, not only as an early response defense, but by directing the subsequent adaptive immune response. The discovery of functional intracellular complement and its roles in cellular metabolism opened novel avenues for research and potential therapeutic implications. The recent studies demonstrating immunoregulatory functions of complement in the tumor microenvironment and the premetastatic niche shifted the paradigm on our understanding of functions of the complement system in regulating immunity. Several complement proteins, through their interaction with cells in the tumor microenvironment and in metastasis-targeted organs, contribute to modulating tumor growth, antitumor immunity, angiogenesis, and therefore, the overall progression of malignancy and, perhaps, responsiveness of cancer to different therapies. Here, we focus on recent progress in our understanding of immunostimulatory vs. immunoregulatory functions of complement and potential applications of these findings to the design of novel therapies for cancer patients.
Collapse
Affiliation(s)
- Martin Kolev
- Complement and Inflammation Research Section, DIR, NHLBI, NIH, Bethesda, MD, 20892, United States.
| | - Maciej M Markiewski
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, United States.
| |
Collapse
|
21
|
Hertle E, Arts ICW, Kallen CJHVD, Feskens EJM, Schalkwijk CG, Stehouwer CDA, Greevenbroek MMJV. The alternative complement pathway is longitudinally associated with adverse cardiovascular outcomes. Thromb Haemost 2017; 115:446-57. [DOI: 10.1160/th15-05-0439] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 09/08/2015] [Indexed: 01/01/2023]
Abstract
SummaryThe alternative pathway of complement activation is highly reactive and can be activated spontaneously in the vasculature. Activation may contribute to vascular damage and development of cardiovascular disease (CVD). We aimed to investigate functional components of the alternative pathway in cardiovascular risk. We studied 573 individuals who were followed-up for seven years. At baseline, we measured the enhancer properdin; the rate-limiting protease factor D (FD); and a marker of systemic activation, Bb. Using generalised estimating equations, we investigated their longitudinal associations with cardiovascular events (CVE, N=89), CVD (N=159), low-grade inflammation (LGI), endothelial dysfunction (ED) and carotid intima-media thickness (cIMT). Furthermore, we investigated associations with incident CVE (N=39) and CVD (N=73) in 342 participants free of CVD at baseline. CVE included myocardial infarction, stroke, cardiac angioplasty and/or cardiac bypass. CVD additionally included ischaemia on an electrocardiogram and/or ankle-brachial index < 0.9. In adjusted analyses, properdin was positively associated with CVE (per 1SD, longitudinal OR=1.36 [1.07; 1.74], OR for incident CVE=1.53 [1.06; 2.20]), but not with CVD. Properdin was also positively associated with ED (β=0.13 [95 %CI 0.06; 0.20]), but not with LGI or cIMT. FD and Bb were positively associated with LGI (per 1SD, FD: β=0.21 [0.12; 0.29], Bb: β=0.14 [0.07; 0.21]), and ED (FD: β=0.20 [0.11; 0.29], Bb: β=0.10 [0.03; 0.18]), but not with cIMT, CVE or CVD. Taken together, this suggests that the alternative complement pathway contributes to processes of vascular damage, and that in particular a high potential to enhance alternative pathway activation may promote unfavourable cardiovascular outcomes in humans.Supplementary Material to this article is available online at www.thrombosis-online.com.
Collapse
|
22
|
Longitudinal associations of the alternative and terminal pathways of complement activation with adiposity: The CODAM study. Obes Res Clin Pract 2017; 12:286-292. [PMID: 29174517 DOI: 10.1016/j.orcp.2017.11.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 10/25/2017] [Accepted: 11/03/2017] [Indexed: 12/19/2022]
Abstract
OBJECTIVE To investigate longitudinal associations of components of the alternative (C3, C3a, Bb, factor D [FD], factor H [FH], and properdin) and the terminal complement pathway (C5a, sC5b-9) with adiposity. METHODS A prospective human cohort study (n=574 at baseline, n=489 after 7 years follow-up) was analyzed. Generalized estimating equations were used to evaluate the longitudinal associations between complement components (standardized values) and adiposity (main outcome BMI [kg/m2]). Multiple linear regression models were used to investigate the associations between change in complement levels and change in BMI. Analyses were adjusted for age, sex, medication and lifestyle. RESULTS Over the 7-year period, baseline C3 was positively associated with BMI (β=1.72 [95% confidence interval (CI): 1.35; 2.09]). Positive associations were also observed for C3a (β=0.64 [0.31; 0.97]), FD (β=1.00 [0.59; 1.42]), FH (β=1.17 [0.82; 1.53]), and properdin (β=0.60 [0.28; 0.92]), but not for Bb, C5a or sC5b-9. Moreover, changes in C3 (β=0.52 [0.34; 0.71]) and FH (β=0.51 [0.32; 0.70]) were significantly associated with changes in BMI. CONCLUSIONS The complement system, particularly activation of the alternative pathway, may be involved in development of adiposity. Whether individual aspects of alternative pathway activation have a causal role in human obesity, remains to be investigated.
Collapse
|
23
|
Moreno-Navarrete JM, Fernández-Real JM. The complement system is dysfunctional in metabolic disease: Evidences in plasma and adipose tissue from obese and insulin resistant subjects. Semin Cell Dev Biol 2017; 85:164-172. [PMID: 29107169 DOI: 10.1016/j.semcdb.2017.10.025] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 10/20/2017] [Accepted: 10/24/2017] [Indexed: 02/03/2023]
Abstract
The relationship among chronic low-grade inflammation, insulin resistance and other obesity-associated metabolic disturbances is increasingly recognized. The possible mechanisms that trigger these immunologic alterations remain to be fully understood. The complement system is a crucial element of immune defense system, being important in the activation of innate and adaptative immune response, promoting the clearance of apoptotic and damaged endogenous cells and participating in processes of tissue development, degeneration, and regeneration. Circulating components of the complement system appear to be dysregulated in obesity-associated metabolic disturbances. The activation of the complement system is also evident in adipose tissue from obese subjects, in association with subclinical inflammation and alterations in glucose metabolism. The possible contribution of some components of the complement system in the development of insulin resistance and obesity-associated metabolic disturbances, and the possible role of complement system in adipose tissue physiology is reviewed here. The modulation of the complement system could constitute a potential target in the pathophysiology and therapy of obesity and associated metabolic disease.
Collapse
Affiliation(s)
- José María Moreno-Navarrete
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), CIBEROBN (CB06/03/010) and Instituto de Salud Carlos III (ISCIII), Girona, Spain.
| | - José Manuel Fernández-Real
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), CIBEROBN (CB06/03/010) and Instituto de Salud Carlos III (ISCIII), Girona, Spain.
| |
Collapse
|
24
|
Coan PM, Barrier M, Alfazema N, Carter RN, Marion de Procé S, Dopico XC, Garcia Diaz A, Thomson A, Jackson-Jones LH, Moyon B, Webster Z, Ross D, Moss J, Arends MJ, Morton NM, Aitman TJ. Complement Factor B Is a Determinant of Both Metabolic and Cardiovascular Features of Metabolic Syndrome. Hypertension 2017; 70:624-633. [PMID: 28739975 PMCID: PMC5548512 DOI: 10.1161/hypertensionaha.117.09242] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 02/22/2017] [Accepted: 06/02/2017] [Indexed: 12/28/2022]
Abstract
CFB (complement factor B) is elevated in adipose tissue and serum from patients with type 2 diabetes mellitus and cardiovascular disease, but the causal relationship to disease pathogenesis is unclear. Cfb is also elevated in adipose tissue and serum of the spontaneously hypertensive rat, a well-characterized model of metabolic syndrome. To establish the role of CFB in metabolic syndrome, we knocked out the Cfb gene in the spontaneously hypertensive rat. Cfb-/- rats showed improved glucose tolerance and insulin sensitivity, redistribution of visceral to subcutaneous fat, increased adipocyte mitochondrial respiration, and marked changes in gene expression. Cfb-/- rats also had lower blood pressure, increased ejection fraction and fractional shortening, and reduced left ventricular mass. These changes in metabolism and gene expression, in adipose tissue and left ventricle, suggest new adipose tissue-intrinsic and blood pressure-independent mechanisms for insulin resistance and cardiac hypertrophy in the spontaneously hypertensive rat. In silico analysis of the human CFB locus revealed 2 cis-regulated expression quantitative trait loci for CFB expression significantly associated with visceral fat, circulating triglycerides and hypertension in genome-wide association studies. Together, these data demonstrate a key role for CFB in the development of spontaneously hypertensive rat metabolic syndrome phenotypes and of related traits in humans and indicate the potential for CFB as a novel target for treatment of cardiometabolic disease.
Collapse
Affiliation(s)
- Philip M Coan
- From the Centre for Genomic and Experimental Medicine, MRC Institute for Genetics and Molecular Medicine, Edinburgh, United Kingdom (P.M.C., M.B., N.A., S.M.P., X.C.D., D.R., J.M., T.J.A.); British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute (P.M.C., M.B., N.A., R.N.C., A.T., L.H.J.-J., N.M.M., T.J.A.) and Royal (Dick) School of Veterinary Studies (X.C.D.), University of Edinburgh, United Kingdom; Department of Medicine (A.G.D., T.J.A) and Embryonic Stem Cell and Transgenics Facility, MRC Clinical Sciences Centre (B.M., Z.W.), Imperial College London, United Kingdom; and Division of Pathology, Centre for Comparative Pathology, Cancer Research UK Edinburgh Centre, United Kingdom (M.J.A.).
| | - Marjorie Barrier
- From the Centre for Genomic and Experimental Medicine, MRC Institute for Genetics and Molecular Medicine, Edinburgh, United Kingdom (P.M.C., M.B., N.A., S.M.P., X.C.D., D.R., J.M., T.J.A.); British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute (P.M.C., M.B., N.A., R.N.C., A.T., L.H.J.-J., N.M.M., T.J.A.) and Royal (Dick) School of Veterinary Studies (X.C.D.), University of Edinburgh, United Kingdom; Department of Medicine (A.G.D., T.J.A) and Embryonic Stem Cell and Transgenics Facility, MRC Clinical Sciences Centre (B.M., Z.W.), Imperial College London, United Kingdom; and Division of Pathology, Centre for Comparative Pathology, Cancer Research UK Edinburgh Centre, United Kingdom (M.J.A.)
| | - Neza Alfazema
- From the Centre for Genomic and Experimental Medicine, MRC Institute for Genetics and Molecular Medicine, Edinburgh, United Kingdom (P.M.C., M.B., N.A., S.M.P., X.C.D., D.R., J.M., T.J.A.); British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute (P.M.C., M.B., N.A., R.N.C., A.T., L.H.J.-J., N.M.M., T.J.A.) and Royal (Dick) School of Veterinary Studies (X.C.D.), University of Edinburgh, United Kingdom; Department of Medicine (A.G.D., T.J.A) and Embryonic Stem Cell and Transgenics Facility, MRC Clinical Sciences Centre (B.M., Z.W.), Imperial College London, United Kingdom; and Division of Pathology, Centre for Comparative Pathology, Cancer Research UK Edinburgh Centre, United Kingdom (M.J.A.)
| | - Roderick N Carter
- From the Centre for Genomic and Experimental Medicine, MRC Institute for Genetics and Molecular Medicine, Edinburgh, United Kingdom (P.M.C., M.B., N.A., S.M.P., X.C.D., D.R., J.M., T.J.A.); British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute (P.M.C., M.B., N.A., R.N.C., A.T., L.H.J.-J., N.M.M., T.J.A.) and Royal (Dick) School of Veterinary Studies (X.C.D.), University of Edinburgh, United Kingdom; Department of Medicine (A.G.D., T.J.A) and Embryonic Stem Cell and Transgenics Facility, MRC Clinical Sciences Centre (B.M., Z.W.), Imperial College London, United Kingdom; and Division of Pathology, Centre for Comparative Pathology, Cancer Research UK Edinburgh Centre, United Kingdom (M.J.A.)
| | - Sophie Marion de Procé
- From the Centre for Genomic and Experimental Medicine, MRC Institute for Genetics and Molecular Medicine, Edinburgh, United Kingdom (P.M.C., M.B., N.A., S.M.P., X.C.D., D.R., J.M., T.J.A.); British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute (P.M.C., M.B., N.A., R.N.C., A.T., L.H.J.-J., N.M.M., T.J.A.) and Royal (Dick) School of Veterinary Studies (X.C.D.), University of Edinburgh, United Kingdom; Department of Medicine (A.G.D., T.J.A) and Embryonic Stem Cell and Transgenics Facility, MRC Clinical Sciences Centre (B.M., Z.W.), Imperial College London, United Kingdom; and Division of Pathology, Centre for Comparative Pathology, Cancer Research UK Edinburgh Centre, United Kingdom (M.J.A.)
| | - Xaquin C Dopico
- From the Centre for Genomic and Experimental Medicine, MRC Institute for Genetics and Molecular Medicine, Edinburgh, United Kingdom (P.M.C., M.B., N.A., S.M.P., X.C.D., D.R., J.M., T.J.A.); British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute (P.M.C., M.B., N.A., R.N.C., A.T., L.H.J.-J., N.M.M., T.J.A.) and Royal (Dick) School of Veterinary Studies (X.C.D.), University of Edinburgh, United Kingdom; Department of Medicine (A.G.D., T.J.A) and Embryonic Stem Cell and Transgenics Facility, MRC Clinical Sciences Centre (B.M., Z.W.), Imperial College London, United Kingdom; and Division of Pathology, Centre for Comparative Pathology, Cancer Research UK Edinburgh Centre, United Kingdom (M.J.A.)
| | - Ana Garcia Diaz
- From the Centre for Genomic and Experimental Medicine, MRC Institute for Genetics and Molecular Medicine, Edinburgh, United Kingdom (P.M.C., M.B., N.A., S.M.P., X.C.D., D.R., J.M., T.J.A.); British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute (P.M.C., M.B., N.A., R.N.C., A.T., L.H.J.-J., N.M.M., T.J.A.) and Royal (Dick) School of Veterinary Studies (X.C.D.), University of Edinburgh, United Kingdom; Department of Medicine (A.G.D., T.J.A) and Embryonic Stem Cell and Transgenics Facility, MRC Clinical Sciences Centre (B.M., Z.W.), Imperial College London, United Kingdom; and Division of Pathology, Centre for Comparative Pathology, Cancer Research UK Edinburgh Centre, United Kingdom (M.J.A.)
| | - Adrian Thomson
- From the Centre for Genomic and Experimental Medicine, MRC Institute for Genetics and Molecular Medicine, Edinburgh, United Kingdom (P.M.C., M.B., N.A., S.M.P., X.C.D., D.R., J.M., T.J.A.); British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute (P.M.C., M.B., N.A., R.N.C., A.T., L.H.J.-J., N.M.M., T.J.A.) and Royal (Dick) School of Veterinary Studies (X.C.D.), University of Edinburgh, United Kingdom; Department of Medicine (A.G.D., T.J.A) and Embryonic Stem Cell and Transgenics Facility, MRC Clinical Sciences Centre (B.M., Z.W.), Imperial College London, United Kingdom; and Division of Pathology, Centre for Comparative Pathology, Cancer Research UK Edinburgh Centre, United Kingdom (M.J.A.)
| | - Lucy H Jackson-Jones
- From the Centre for Genomic and Experimental Medicine, MRC Institute for Genetics and Molecular Medicine, Edinburgh, United Kingdom (P.M.C., M.B., N.A., S.M.P., X.C.D., D.R., J.M., T.J.A.); British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute (P.M.C., M.B., N.A., R.N.C., A.T., L.H.J.-J., N.M.M., T.J.A.) and Royal (Dick) School of Veterinary Studies (X.C.D.), University of Edinburgh, United Kingdom; Department of Medicine (A.G.D., T.J.A) and Embryonic Stem Cell and Transgenics Facility, MRC Clinical Sciences Centre (B.M., Z.W.), Imperial College London, United Kingdom; and Division of Pathology, Centre for Comparative Pathology, Cancer Research UK Edinburgh Centre, United Kingdom (M.J.A.)
| | - Ben Moyon
- From the Centre for Genomic and Experimental Medicine, MRC Institute for Genetics and Molecular Medicine, Edinburgh, United Kingdom (P.M.C., M.B., N.A., S.M.P., X.C.D., D.R., J.M., T.J.A.); British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute (P.M.C., M.B., N.A., R.N.C., A.T., L.H.J.-J., N.M.M., T.J.A.) and Royal (Dick) School of Veterinary Studies (X.C.D.), University of Edinburgh, United Kingdom; Department of Medicine (A.G.D., T.J.A) and Embryonic Stem Cell and Transgenics Facility, MRC Clinical Sciences Centre (B.M., Z.W.), Imperial College London, United Kingdom; and Division of Pathology, Centre for Comparative Pathology, Cancer Research UK Edinburgh Centre, United Kingdom (M.J.A.)
| | - Zoe Webster
- From the Centre for Genomic and Experimental Medicine, MRC Institute for Genetics and Molecular Medicine, Edinburgh, United Kingdom (P.M.C., M.B., N.A., S.M.P., X.C.D., D.R., J.M., T.J.A.); British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute (P.M.C., M.B., N.A., R.N.C., A.T., L.H.J.-J., N.M.M., T.J.A.) and Royal (Dick) School of Veterinary Studies (X.C.D.), University of Edinburgh, United Kingdom; Department of Medicine (A.G.D., T.J.A) and Embryonic Stem Cell and Transgenics Facility, MRC Clinical Sciences Centre (B.M., Z.W.), Imperial College London, United Kingdom; and Division of Pathology, Centre for Comparative Pathology, Cancer Research UK Edinburgh Centre, United Kingdom (M.J.A.)
| | - David Ross
- From the Centre for Genomic and Experimental Medicine, MRC Institute for Genetics and Molecular Medicine, Edinburgh, United Kingdom (P.M.C., M.B., N.A., S.M.P., X.C.D., D.R., J.M., T.J.A.); British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute (P.M.C., M.B., N.A., R.N.C., A.T., L.H.J.-J., N.M.M., T.J.A.) and Royal (Dick) School of Veterinary Studies (X.C.D.), University of Edinburgh, United Kingdom; Department of Medicine (A.G.D., T.J.A) and Embryonic Stem Cell and Transgenics Facility, MRC Clinical Sciences Centre (B.M., Z.W.), Imperial College London, United Kingdom; and Division of Pathology, Centre for Comparative Pathology, Cancer Research UK Edinburgh Centre, United Kingdom (M.J.A.)
| | - Julie Moss
- From the Centre for Genomic and Experimental Medicine, MRC Institute for Genetics and Molecular Medicine, Edinburgh, United Kingdom (P.M.C., M.B., N.A., S.M.P., X.C.D., D.R., J.M., T.J.A.); British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute (P.M.C., M.B., N.A., R.N.C., A.T., L.H.J.-J., N.M.M., T.J.A.) and Royal (Dick) School of Veterinary Studies (X.C.D.), University of Edinburgh, United Kingdom; Department of Medicine (A.G.D., T.J.A) and Embryonic Stem Cell and Transgenics Facility, MRC Clinical Sciences Centre (B.M., Z.W.), Imperial College London, United Kingdom; and Division of Pathology, Centre for Comparative Pathology, Cancer Research UK Edinburgh Centre, United Kingdom (M.J.A.)
| | - Mark J Arends
- From the Centre for Genomic and Experimental Medicine, MRC Institute for Genetics and Molecular Medicine, Edinburgh, United Kingdom (P.M.C., M.B., N.A., S.M.P., X.C.D., D.R., J.M., T.J.A.); British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute (P.M.C., M.B., N.A., R.N.C., A.T., L.H.J.-J., N.M.M., T.J.A.) and Royal (Dick) School of Veterinary Studies (X.C.D.), University of Edinburgh, United Kingdom; Department of Medicine (A.G.D., T.J.A) and Embryonic Stem Cell and Transgenics Facility, MRC Clinical Sciences Centre (B.M., Z.W.), Imperial College London, United Kingdom; and Division of Pathology, Centre for Comparative Pathology, Cancer Research UK Edinburgh Centre, United Kingdom (M.J.A.)
| | - Nicholas M Morton
- From the Centre for Genomic and Experimental Medicine, MRC Institute for Genetics and Molecular Medicine, Edinburgh, United Kingdom (P.M.C., M.B., N.A., S.M.P., X.C.D., D.R., J.M., T.J.A.); British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute (P.M.C., M.B., N.A., R.N.C., A.T., L.H.J.-J., N.M.M., T.J.A.) and Royal (Dick) School of Veterinary Studies (X.C.D.), University of Edinburgh, United Kingdom; Department of Medicine (A.G.D., T.J.A) and Embryonic Stem Cell and Transgenics Facility, MRC Clinical Sciences Centre (B.M., Z.W.), Imperial College London, United Kingdom; and Division of Pathology, Centre for Comparative Pathology, Cancer Research UK Edinburgh Centre, United Kingdom (M.J.A.)
| | - Timothy J Aitman
- From the Centre for Genomic and Experimental Medicine, MRC Institute for Genetics and Molecular Medicine, Edinburgh, United Kingdom (P.M.C., M.B., N.A., S.M.P., X.C.D., D.R., J.M., T.J.A.); British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute (P.M.C., M.B., N.A., R.N.C., A.T., L.H.J.-J., N.M.M., T.J.A.) and Royal (Dick) School of Veterinary Studies (X.C.D.), University of Edinburgh, United Kingdom; Department of Medicine (A.G.D., T.J.A) and Embryonic Stem Cell and Transgenics Facility, MRC Clinical Sciences Centre (B.M., Z.W.), Imperial College London, United Kingdom; and Division of Pathology, Centre for Comparative Pathology, Cancer Research UK Edinburgh Centre, United Kingdom (M.J.A.)
| |
Collapse
|
25
|
Kolev M, Kemper C. Keeping It All Going-Complement Meets Metabolism. Front Immunol 2017; 8:1. [PMID: 28149297 PMCID: PMC5241319 DOI: 10.3389/fimmu.2017.00001] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 01/03/2017] [Indexed: 01/22/2023] Open
Abstract
The complement system is an evolutionary old and crucial component of innate immunity, which is key to the detection and removal of invading pathogens. It was initially discovered as a liver-derived sentinel system circulating in serum, the lymph, and interstitial fluids that mediate the opsonization and lytic killing of bacteria, fungi, and viruses and the initiation of the general inflammatory responses. Although work performed specifically in the last five decades identified complement also as a critical instructor of adaptive immunity—indicating that complement’s function is likely broader than initially anticipated—the dominant opinion among researchers and clinicians was that the key complement functions were in principle defined. However, there is now a growing realization that complement activity goes well beyond “classic” immune functions and that this system is also required for normal (neuronal) development and activity and general cell and tissue integrity and homeostasis. Furthermore, the recent discovery that complement activation is not confined to the extracellular space but occurs within cells led to the surprising understanding that complement is involved in the regulation of basic processes of the cell, particularly those of metabolic nature—mostly via novel crosstalks between complement and intracellular sensor, and effector, pathways that had been overlooked because of their spatial separation. These paradigm shifts in the field led to a renaissance in complement research and provide new platforms to now better understand the molecular pathways underlying the wide-reaching effects of complement functions in immunity and beyond. In this review, we will cover the current knowledge about complement’s emerging relationship with the cellular metabolism machinery with a focus on the functional differences between serum-circulating versus intracellularly active complement during normal cell survival and induction of effector functions. We will also discuss how taking a closer look into the evolution of key complement components not only made the functional connection between complement and metabolism rather “predictable” but how it may also give clues for the discovery of additional roles for complement in basic cellular processes.
Collapse
Affiliation(s)
- Martin Kolev
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital , London , UK
| | - Claudia Kemper
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK; Laboratory of Molecular Immunology, The Immunology Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
26
|
Bavia L, Cogliati B, Dettoni JB, Ferreira Alves VA, Isaac L. The complement component C5 promotes liver steatosis and inflammation in murine non-alcoholic liver disease model. Immunol Lett 2016; 177:53-61. [PMID: 27477770 DOI: 10.1016/j.imlet.2016.07.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 07/27/2016] [Indexed: 02/05/2023]
Abstract
Non-Alcoholic Fatty Liver Disease (NALD) is considering a hepatic manifestation of metabolic syndrome. Although the pathogenesis of NALD is not completely understood, insulin resistance and inflammatory cytokines are implicated. Considering that component C5 is a central mediator of inflammation, we investigated the role of C5 in the establishment of NALD. Eight to ten-week old B6 C5(+) and A/J C5(-) male mice were fed a high fat diet containing glucose (HFDG) for 6 and 10 weeks. We observed that B6 C5(+) mice HFDG-fed for 10 weeks developed hepatomegaly, triglycerides (TG) accumulation, steatosis and enhanced liver TNF-α, IL-6, IL-12p70 and IL-17 levels when compared to A/J C5(-) mice. Next, B6 C5(+) mice were compared with congenic B6 C5(-) mice. Again, B6 C5(+) HFDG-fed mice developed more steatosis, liver centro-lobular inflammation and presented higher levels of liver IL-1β, IL-12p70, IL-17 and TFG-β than B6 C5(-) mice under the same conditions. B6 C5(+) mice HFDG-fed also presented lower concentrations of serum albumin, serum cholesterol, blood leukocytes and liver NO production when compared with B6 C5(-) mice. We concluded that murine C5 contributes effectively to liver steatosis and inflammation in NALD pathogenesis. In addition, C5 is also important to control serum cholesterol and albumin levels in the C57BL/6 genetic background.
Collapse
Affiliation(s)
- Lorena Bavia
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil.
| | - Bruno Cogliati
- Departamento de Patologia, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São Paulo, São Paulo, Brazil
| | | | | | - Lourdes Isaac
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
27
|
Wu J, Jiao ZY, Zhang Z, Tang ZH, Zhang HH, Lu HL, Cianflone K. Cross-talk between α7 nAChR-mediated cholinergic pathway and acylation stimulating protein signaling in 3T3-L1 adipocytes: role of NFκB and STAT3. Biochem Cell Biol 2015; 93:335-42. [PMID: 25985797 DOI: 10.1139/bcb-2015-0023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Inflammation is a key feature in adipose tissue, especially in association with obesity comorbidies. The novel adipokine acylation stimulating protein (ASP) is one factor implicated in the inflammatory response. The disruption of the α7 nicotine acetylcholine receptor (α7nAChR), an important component of the endogenous non-neural cholinergic defense system, may exacerbate sustained inflammatory phenotype. We examined cholinergic regulation of ASP-initiated inflammatory response in 3T3-L1 adipocytes. Our results show that preincubation of 3T3-L1 cells with α7nAChR agonist GTS-21 significantly reduces ASP-mediated chemokine MCP-1 secretion, which is regulated though nuclear factor κB (NFκB) and signal transducer and activator of transcription 3 (STAT3). Treatment of 3T3-L1 cells with GTS-21 significantly reduced NFκB activation by DNA binding and STAT3 activation by disturbing post-translational modification.
Collapse
Affiliation(s)
- Jing Wu
- a Department of Pediatrics, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Zhou-yang Jiao
- b Department of Cardiovascular Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Zhe Zhang
- c Department of Histology & Embryology, College of Basic Medical Science, Zhengzhou University, Zhengzhou, 450001, China
| | - Zhi-hui Tang
- a Department of Pediatrics, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Hao-hao Zhang
- d Department of Endocrinology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Hui-ling Lu
- e Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Katherine Cianflone
- f Centre de Recherche Institut Universitaire de Cardiologie & Pneumologie de Québec, Université Laval, QC, G1V 4G5, Canada
| |
Collapse
|
28
|
Kwiterovich PO. Detection and Treatment of Children and Adolescents with Dyslipidemia. DYSLIPIDEMIAS 2015. [DOI: 10.1007/978-1-60761-424-1_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
29
|
Al Riyami B, El-Tahir M, Al Maskari S, Johnson EH, Saleh J. Acute effects of exogenous hormone administration on postprandial acylation stimulating protein levels in ovariectomized rats after a fat load. J Nutr Metab 2014; 2014:510916. [PMID: 25525514 PMCID: PMC4267214 DOI: 10.1155/2014/510916] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 11/14/2014] [Indexed: 11/20/2022] Open
Abstract
Background. ASP, a potent lipogenic factor, was linked to female fat metabolism in association studies. Aim. To investigate acute effects of sex hormone treatment on postprandial ASP levels in vivo. Methods. 24 female rats were randomly divided into 4 groups including controls. The rats were ovariectomized and injected with progesterone, estrogen, or testosterone. An hour later, olive oil was administered orally. Plasma ASP and triglycerides were measured at several postprandial time points. Area under the curve (TG-AUC) represented TG clearance. Results. Only the progesterone treated group had a significant postprandial ASP increase at two hours compared to basal levels (439.8 ± 62.4 versus 253.4 ± 59.03 μg/mL, P = 0.04). Interestingly, increased ASP levels coordinated negatively with corresponding TG levels and TG-AUC postprandially, mostly evident in the opposite effects in the progesterone and testosterone treated groups. ASP levels increased 3-fold in the progesterone versus testosterone treated groups, whereas TG-AUC was significantly lower. Conclusion. These findings suggest that progesterone enhances ASP production and TG clearance simultaneously, supporting the notion of a stimulatory role for progesterone on ASP-mediated TG clearance. This is the first functional study demonstrating a cause-effect relationship between hormone treatment and ASP levels in vivo and may contribute to understanding the mechanism of progesterone function as a female lipogenic hormone.
Collapse
Affiliation(s)
- Bashair Al Riyami
- Department of Biochemistry, College of Medicine, Sultan Qaboos University, Al-Khod, 123 Muscat, Oman
| | - Marah El-Tahir
- Department of Microbiology and Immunology, College of Medicine, Sultan Qaboos University, Al-Khod, 123 Muscat, Oman
| | - Sultan Al Maskari
- Department of Biochemistry, College of Medicine, Sultan Qaboos University, Al-Khod, 123 Muscat, Oman
| | - Eugene H. Johnson
- Department of Animal and Veterinary Sciences, College of Agricultural and Marine Sciences, Sultan Qaboos University, Al-Khod, 123 Muscat, Oman
| | - Jumana Saleh
- Department of Biochemistry, College of Medicine, Sultan Qaboos University, Al-Khod, 123 Muscat, Oman
| |
Collapse
|
30
|
The complement system in human cardiometabolic disease. Mol Immunol 2014; 61:135-48. [PMID: 25017306 DOI: 10.1016/j.molimm.2014.06.031] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 06/18/2014] [Accepted: 06/23/2014] [Indexed: 02/07/2023]
Abstract
The complement system has been implicated in obesity, fatty liver, diabetes and cardiovascular disease (CVD). Complement factors are produced in adipose tissue and appear to be involved in adipose tissue metabolism and local inflammation. Thereby complement links adipose tissue inflammation to systemic metabolic derangements, such as low-grade inflammation, insulin resistance and dyslipidaemia. Furthermore, complement has been implicated in pathophysiological mechanisms of diet- and alcohol induced liver damage, hyperglycaemia, endothelial dysfunction, atherosclerosis and fibrinolysis. In this review, we summarize current evidence on the role of the complement system in several processes of human cardiometabolic disease. C3 is the central component in complement activation, and has most widely been studied in humans. C3 concentrations are associated with insulin resistance, liver dysfunction, risk of the metabolic syndrome, type 2 diabetes and CVD. C3 can be activated by the classical, the lectin and the alternative pathway of complement activation; and downstream activation of C3 activates the terminal pathway. Complement may also be activated via extrinsic proteases of the coagulation, fibrinolysis and the kinin systems. Studies on the different complement activation pathways in human cardiometabolic disease are limited, but available evidence suggests that they may have distinct roles in processes underlying cardiometabolic disease. The lectin pathway appeared beneficial in some studies on type 2 diabetes and CVD, while factors of the classical and the alternative pathway were related to unfavourable cardiometabolic traits. The terminal complement pathway was also implicated in insulin resistance and liver disease, and appears to have a prominent role in acute and advanced CVD. The available human data suggest a complex and potentially causal role for the complement system in human cardiometabolic disease. Further, preferably longitudinal studies are needed to disentangle which aspects of the complement system and complement activation affect the different processes in human cardiometabolic disease.
Collapse
|
31
|
Takeshita A, Kusakabe KT, Hiyama M, Kuniyoshi N, Kondo T, Kano K, Kiso Y, Okada T. Dynamics and reproductive effects of complement factors in the spontaneous abortion model of CBA/J×DBA/2 mice. Immunobiology 2014; 219:385-91. [DOI: 10.1016/j.imbio.2014.01.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 12/04/2013] [Accepted: 01/03/2014] [Indexed: 11/16/2022]
|
32
|
Protective role for properdin in progression of experimental murine atherosclerosis. PLoS One 2014; 9:e92404. [PMID: 24667818 PMCID: PMC3965423 DOI: 10.1371/journal.pone.0092404] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Accepted: 02/21/2014] [Indexed: 12/29/2022] Open
Abstract
Genetic, dietary and immune factors contribute to the pathogenesis of atherosclerosis in humans and mice. Complement activation is an integral part of the innate immune defence but also shapes cellular responses and influences directly triglyceride synthesis. Deficiency of Factor B of the alternative pathway (AP) of complement is beneficial in LDLR(-/-) mice fed a high fat diet. The serum glycoprotein properdin is a key positive regulator of the AP but has not been studied in experimental atherosclerosis. Atherosclerosis was assessed after feeding low fat (LFD) or high fat (HFD) Western type diets to newly generated LDLR(-/-) Properdin(KO) (LDLR(-/-)P(KO)) and LDLR-/-PWT mice. Lipids, lymphocytes and monocytes were similar among genotypes, genders and diets. Complement C3, but not C3adesarg, levels were enhanced in LDLR(-/-)P(KO) mice regardless of diet type or gender. Non-esterified fatty acids (NEFA) were decreased in male LDLR(-/-)P(KO) fed a HFD compared with controls. All mice showed significant atherosclerotic burden in aortae and at aortic roots but male LDLR(-/-) mice fed a LFD were affected to the greatest extent by the absence of properdin. The protective effect of properdin expression was overwhelmed in both genders of LDLR(-/-)mice when fed a HFD. We conclude that properdin plays an unexpectedly beneficial role in the development and progression of early atherosclerotic lesions.
Collapse
|
33
|
Poursharifi P, Lapointe M, Fisette A, Lu H, Roy C, Munkonda MN, Fairlie DP, Cianflone K. C5aR and C5L2 act in concert to balance immunometabolism in adipose tissue. Mol Cell Endocrinol 2014; 382:325-333. [PMID: 24397921 DOI: 10.1016/j.mce.2013.10.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 10/15/2013] [Indexed: 12/24/2022]
Abstract
Recent studies suggested that the immunometabolic receptors; C5aR and C5L2, constitutively self-associate into homo-/heterodimers and that acylation stimulating protein (ASP/C3adesArg) or C5a treatment of adipocytes increased their colocalization. The present study evaluates the C5aR contribution in adipocytes to the metabolic and immune responses elicited by ligand stimulation. The effects of C5a, ASP, and insulin on cytokine production, triglyceride synthesis (TGS), and key signaling pathways were evaluated in isolated primary adipocytes and cultured 3T3-L1 differentiated adipocytes. In addition, mRNA expression of IRS1 and PGC1α was compared in adipose tissue samples from WT vs. C5aRKO mice. Both C5a and ASP directly increased MCP-1 (238±4%; P<0.001, and 377±2% vs. basal 100%; P<0.001, respectively) and KC (413±11%; P<0.001, and 529±16%; P<0.001 vs. basal 100%, respectively) secretion, TGS (131±1%; P<0.001, and 152±6%; P<0.001, vs. basal 100% respectively), and Akt/NFκB phosphorylation pathways in adipocytes. However, in C5aRKO adipocytes, C5a effects were disrupted, while stimulatory effects of ASP were mostly maintained. Addition of C5a completely blocked ASP signaling and activity in both C5aRKO and WT adipocytes as well as 3T3-L1 adipocytes. Furthermore, C5aRKO adipocytes revealed impaired insulin stimulation of cytokine production, with partial impairment of signaling and TGS stimulation, consistent with decreased IRS1 and PGC1α mRNA expression in adipose tissue. These observations indicate the importance of C5aR in adipose tissue metabolism and immunity, which may be regulated through heterodimerization with C5L2.
Collapse
Affiliation(s)
- Pegah Poursharifi
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Québec, QC, Canada; Department of Medicine, Laval University, Québec, QC, Canada
| | - Marc Lapointe
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Québec, QC, Canada
| | - Alexandre Fisette
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Québec, QC, Canada; Department of Medicine, Laval University, Québec, QC, Canada
| | - Huiling Lu
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Québec, QC, Canada
| | - Christian Roy
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Québec, QC, Canada; Department of Medicine, Laval University, Québec, QC, Canada
| | - Mercedes Nancy Munkonda
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Québec, QC, Canada; Department of Medicine, Laval University, Québec, QC, Canada
| | - David P Fairlie
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Katherine Cianflone
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Québec, QC, Canada; Department of Medicine, Laval University, Québec, QC, Canada.
| |
Collapse
|
34
|
Kemper C, Köhl J. Novel roles for complement receptors in T cell regulation and beyond. Mol Immunol 2013; 56:181-90. [PMID: 23796748 DOI: 10.1016/j.molimm.2013.05.223] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 05/20/2013] [Indexed: 12/16/2022]
Abstract
Complement receptors are expressed on cells of the innate and the adaptive immune system. They play important roles in pathogen and danger sensing as they translate the information gathered by complement fluid phase sensors into cellular responses. Further, they control complement activation on viable and apoptotic host cells, clearance of immune complexes and mediate opsonophagocytosis. More recently, evidence has accumulated that complement receptors form a complex network with other innate receptors systems such as the Toll-like receptors, the Notch signaling system, IgG Fc receptors and C-type lectin receptors contributing to the benefit and burden of innate and adaptive immune responses in autoimmune and allergic diseases as well as in cancer and transplantation. Here, we will discuss recent developments and emerging concepts of complement receptor activation and regulation with a particular focus on the differentiation, maintenance and contraction of effector and regulatory T cells.
Collapse
Affiliation(s)
- Claudia Kemper
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK.
| | | |
Collapse
|
35
|
The role of the complement system in metabolic organs and metabolic diseases. Semin Immunol 2013; 25:47-53. [PMID: 23684628 DOI: 10.1016/j.smim.2013.04.003] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 04/13/2013] [Indexed: 12/20/2022]
Abstract
Emerging evidence points to a close crosstalk between metabolic organs and innate immunity in the course of metabolic disorders. In particular, cellular and humoral factors of innate immunity are thought to contribute to metabolic dysregulation of the adipose tissue or the liver, as well as to dysfunction of the pancreas; all these conditions are linked to the development of insulin resistance and diabetes mellitus. A central component of innate immunity is the complement system. Interestingly, the classical view of complement as a major system of host defense that copes with infections is changing to that of a multi-functional player in tissue homeostasis, degeneration, and regeneration. In the present review, we will discuss the link between complement and metabolic organs, focusing on the pancreas, adipose tissue, and liver and the diverse effects of complement system on metabolic disorders.
Collapse
|
36
|
Roy C, Gupta A, Fisette A, Lapointe M, Poursharifi P, Richard D, Lu H, Lu B, Gerard N, Gerard C, Cianflone K. C5a receptor deficiency alters energy utilization and fat storage. PLoS One 2013; 8:e62531. [PMID: 23667486 PMCID: PMC3646841 DOI: 10.1371/journal.pone.0062531] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 03/22/2013] [Indexed: 01/26/2023] Open
Abstract
OBJECTIVE To investigate the impact of whole body C5a receptor (C5aR) deficiency on energy metabolism and fat storage. DESIGN Male wildtype (WT) and C5aR knockout (C5aRKO) mice were fed a low fat (CHOW) or a high fat high sucrose diet-induced obesity (DIO) diet for 14 weeks. Body weight and food intake were measured weekly. Indirect calorimetry, dietary fatload clearance, insulin and glucose tolerance tests were also evaluated. Liver, muscle and adipose tissue mRNA gene expression were measured by RT-PCR. RESULTS At week one and 12, C5aRKO mice on DIO had increased oxygen consumption. After 12 weeks, although food intake was comparable, C5aRKO mice had lower body weight (-7% CHOW, -12% DIO) as well as smaller gonadal (-38% CHOW, -36% DIO) and inguinal (-29% CHOW, -30% DIO) fat pads than their WT counterparts. Conversely, in WT mice, C5aR was upregulated in DIO vs CHOW diets in gonadal adipose tissue, muscle and liver, while C5L2 mRNA expression was lower in C5aRKO on both diet. Furthermore, blood analysis showed lower plasma triglyceride and non-esterified fatty acid levels in both C5aRKO groups, with faster postprandial triglyceride clearance after a fatload. Additionally, C5aRKO mice showed lower CD36 expression in gonadal and muscle on both diets, while DGAT1 expression was higher in gonadal (CHOW) and liver (CHOW and DIO) and PPARγ was increased in muscle and liver. CONCLUSION These observations point towards a role (either direct or indirect) for C5aR in energy expenditure and fat storage, suggesting a dual role for C5aR in metabolism as well as in immunity.
Collapse
Affiliation(s)
- Christian Roy
- Centre de Recherche Institut Universitaire de Cardiologie & Pneumologie de Québec, Université Laval, Québec, Québec, Canada
| | - Abhishek Gupta
- Centre de Recherche Institut Universitaire de Cardiologie & Pneumologie de Québec, Université Laval, Québec, Québec, Canada
| | - Alexandre Fisette
- Centre de Recherche Institut Universitaire de Cardiologie & Pneumologie de Québec, Université Laval, Québec, Québec, Canada
| | - Marc Lapointe
- Centre de Recherche Institut Universitaire de Cardiologie & Pneumologie de Québec, Université Laval, Québec, Québec, Canada
| | - Pegah Poursharifi
- Centre de Recherche Institut Universitaire de Cardiologie & Pneumologie de Québec, Université Laval, Québec, Québec, Canada
| | - Denis Richard
- Centre de Recherche Institut Universitaire de Cardiologie & Pneumologie de Québec, Université Laval, Québec, Québec, Canada
| | - HuiLing Lu
- Centre de Recherche Institut Universitaire de Cardiologie & Pneumologie de Québec, Université Laval, Québec, Québec, Canada
| | - Bao Lu
- Ina Sue Perlmutter Lab, Children’s Hospital, Harvard Medical School, Boston, Massachusetts United States of America
| | - Norma Gerard
- Ina Sue Perlmutter Lab, Children’s Hospital, Harvard Medical School, Boston, Massachusetts United States of America
| | - Craig Gerard
- Ina Sue Perlmutter Lab, Children’s Hospital, Harvard Medical School, Boston, Massachusetts United States of America
| | - Katherine Cianflone
- Centre de Recherche Institut Universitaire de Cardiologie & Pneumologie de Québec, Université Laval, Québec, Québec, Canada
| |
Collapse
|
37
|
Coelho M, Oliveira T, Fernandes R. Biochemistry of adipose tissue: an endocrine organ. Arch Med Sci 2013; 9:191-200. [PMID: 23671428 PMCID: PMC3648822 DOI: 10.5114/aoms.2013.33181] [Citation(s) in RCA: 754] [Impact Index Per Article: 62.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Revised: 07/02/2012] [Accepted: 07/04/2012] [Indexed: 02/06/2023] Open
Abstract
Adipose tissue is no longer considered to be an inert tissue that stores fat. This tissue is capable of expanding to accommodate increased lipids through hypertrophy of existing adipocytes and by initiating differentiation of pre-adipocytes. Adipose tissue metabolism exerts an impact on whole-body metabolism. As an endocrine organ, adipose tissue is responsible for the synthesis and secretion of several hormones. These are active in a range of processes, such as control of nutritional intake (leptin, angiotensin), control of sensitivity to insulin and inflammatory process mediators (tumor necrosis factor α (TNF-α), interleukin-6 (IL-6), resistin, visfatin, adiponectin, among others) and pathways (plasminogen activator inhibitor 1 (PAI-1) and acylation stimulating protein (ASP) for example). This paper reviews some of the biochemical and metabolic aspects of adipose tissue and its relationship to inflammatory disease and insulin resistance.
Collapse
Affiliation(s)
- Marisa Coelho
- Ciências Químicas e das Biomoléculas e Unidade de Mecanismos Moleculares da Doença do Centro de Investigação em Saúde e Ambiente, Escola Superior de Tecnologia da Saúde do Porto, Instituto Politécnico do Porto, Portugal ; Centro de Farmacologia e Biopatologia Química (U38-FCT), Faculdade de Medicina da Universidade do Porto, Portugal
| | | | | |
Collapse
|
38
|
Tom FQ, Gauvreau D, Lapointe M, Lu H, Poursharifi P, Luo XP, Cianflone K. Differential chemoattractant response in adipocytes and macrophages to the action of acylation stimulating protein. Eur J Cell Biol 2012; 92:61-9. [PMID: 23245988 DOI: 10.1016/j.ejcb.2012.10.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Revised: 10/12/2012] [Accepted: 10/28/2012] [Indexed: 01/02/2023] Open
Abstract
Obesity is characterized by chronic low-grade inflammation with increased adipose tissue pro-inflammatory cytokine production. Acylation stimulating protein (ASP) stimulates triglyceride synthesis and glucose transport via its receptor C5L2. Circulating ASP is increased in obesity, insulin resistance and metabolic syndrome. The present study examines the effects of normal (50 nM), high physiological (200 nM) and pathological (600 nM) levels of ASP on inflammatory changes in 3T3-L1 adipocytes and J774 macrophages and the underlying mechanisms involved. Treatment with ASP for 24h increased monocyte chemoattractant protein-1 (MCP1, 800%, P<0.001) and keratinocyte-derived chemokine (KC, >150%, P<0.01) secretion in adipocytes in a dose-dependent manner, with no effect on IL-6 or adiponectin. In macrophages, ASP had no effect on these cytokines. C5a, a ligand for C5L2 and C5aR receptors, differed from ASP. Macrophage-adipocyte coculture increased MCP-1 and adiponectin secretion, and ASP further enhanced secretion (P<0.001 and P<0.05, respectively) at doses of 50 nM and 200 nM. ASP increased Ser(468) and Ser(536) phosphorylation of p65 NFκB in a time- and concentration-dependent manner (P<0.05) as well as phosphorylation of Akt Ser(473) (p=0.02). ASP and insulin stimulations of Ser(536) p65 NFκB phosphorylation were comparable (both p<0.05) but not additive. Both inhibition of PI3kinase (with wortmannin) and NFκB (with BAY11-7085) prevented ASP stimulation of MCP-1 and KC secretion in adipocytes. These findings suggest that ASP, especially at high physiologic doses, may stimulate specific inflammatory cytokines in adipocytes through PI3kinase- and NFκB-dependant pathways, thus further promoting macrophage infiltration and local inflammation in obese adipose tissue.
Collapse
Affiliation(s)
- Fun-Qun Tom
- Centre de Recherche Institut Universitaire de Cardiologie & Pneumologie de Québec, Université Laval, Québec, Canada
| | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
The innate immune system is a prewired set of cellular and humoral components that has developed to sense perturbations in normal physiology and trigger responses to restore the system back to baseline. It is now understood that many of these components can also sense the physiologic changes that occur with obesity and be activated. While the exact reasons for this chronic immune response to obesity are unclear, there is strong evidence to suggest that innate inflammatory systems link obesity and disease. Based on this, anti-inflammatory therapies for diseases like type 2 diabetes and metabolic syndrome may form the core of future treatment plans. This review will highlight the components involved in the innate immune response and discuss the evidence that they contribute to the pathogenesis of obesity-associated diseases.
Collapse
Affiliation(s)
- Carey N Lumeng
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI, United States.
| |
Collapse
|
40
|
Munkonda MN, Lapointe M, Miegueu P, Roy C, Gauvreau D, Richard D, Cianflone K. Recombinant acylation stimulating protein administration to C3-/- mice increases insulin resistance via adipocyte inflammatory mechanisms. PLoS One 2012; 7:e46883. [PMID: 23056509 PMCID: PMC3466186 DOI: 10.1371/journal.pone.0046883] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 09/07/2012] [Indexed: 12/28/2022] Open
Abstract
Background Complement 3 (C3), a key component of the innate immune system, is involved in early inflammatory responses. Acylation stimulating protein (ASP; aka C3adesArg), a C3 cleavage product, is produced in adipose tissue and stimulates lipid storage. We hypothesized that, depending on the diet, chronic ASP administration in C3−/− mice would affect lipid metabolism and insulin sensitivity via an adaptive adipose tissue inflammatory response. Methodology/Principal Findings C3−/− mice on normal low fat diet (ND) or high fat diet (HFD) were chronically administered recombinant ASP (rASP) for 25 days via an osmotic mini-pump. While there was no effect on food intake, there was a decrease in activity, with a relative increase in adipose tissue weight on ND, and a shift in adipocyte size distribution. While rASP administration to C3−/− mice on a ND increased insulin sensitivity, on a HFD, rASP administration had the opposite effect. Specifically, rASP administration in C3−/− HFD mice resulted in decreased gene expression of IRS1, GLUT4, SREBF1 and NFκB in muscle, and decreased C5L2 but increased JNK, CD36, CD11c, CCR2 and NFκB gene expression in adipose tissue as well as increased secretion of proinflammatory cytokines (Rantes, KC, MCP-1, IL-6 and G-CSF). In adipose tissue, although IRS1 and GLUT4 mRNA were unchanged, insulin response was reduced. Conclusion The effects of chronic rASP administration are tissue and diet specific, rASP administration enhances the HFD induced inflammatory response leading to an insulin-resistant state. These results suggest that, in humans, the increased plasma ASP associated with obesity and cardiovascular disease could be an additional factor directly contributing to development of metabolic syndrome, insulin resistance and diabetes.
Collapse
Affiliation(s)
- Mercedes Nancy Munkonda
- Centre de Recherche Institut Universitaire de Cardiologie & Pneumologie de Québec, Université Laval, Québec, Canada
| | - Marc Lapointe
- Centre de Recherche Institut Universitaire de Cardiologie & Pneumologie de Québec, Université Laval, Québec, Canada
| | - Pierre Miegueu
- Centre de Recherche Institut Universitaire de Cardiologie & Pneumologie de Québec, Université Laval, Québec, Canada
| | - Christian Roy
- Centre de Recherche Institut Universitaire de Cardiologie & Pneumologie de Québec, Université Laval, Québec, Canada
| | - Danny Gauvreau
- Centre de Recherche Institut Universitaire de Cardiologie & Pneumologie de Québec, Université Laval, Québec, Canada
| | - Denis Richard
- Centre de Recherche Institut Universitaire de Cardiologie & Pneumologie de Québec, Université Laval, Québec, Canada
| | - Katherine Cianflone
- Centre de Recherche Institut Universitaire de Cardiologie & Pneumologie de Québec, Université Laval, Québec, Canada
- * E-mail:
| |
Collapse
|
41
|
Gauvreau D, Roy C, Tom FQ, Lu H, Miegueu P, Richard D, Song WC, Stover C, Cianflone K. A new effector of lipid metabolism: Complement factor properdin. Mol Immunol 2012; 51:73-81. [DOI: 10.1016/j.molimm.2012.02.110] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Revised: 02/01/2012] [Accepted: 02/06/2012] [Indexed: 10/28/2022]
|
42
|
Ortega FJ, Moreno-Navarrete JM, Sabater M, Ricart W, Frühbeck G, Fernández-Real JM. Circulating glucagon is associated with inflammatory mediators in metabolically compromised subjects. Eur J Endocrinol 2011; 165:639-45. [PMID: 21798957 DOI: 10.1530/eje-11-0384] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Acute phase mediators promote metabolic changes by modifying circulating hormones. However, there is virtually no data about the link between glucagon and inflammatory parameters in obesity-related chronic low-grade inflammation. STUDY DESIGN We performed both cross-sectional and longitudinal (diet-induced weight loss) studies. METHODS Circulating glucagon concentrations (ELISA), parameters of glucose and lipid metabolism, interleukin 6 (IL6), and complement factor B (CFB) were analyzed in 316 subjects (250 men and 66 women). The effects of weight loss were investigated in an independent cohort of 20 subjects. RESULTS Circulating glucagon significantly correlated with glucose (r=0.407, P<0.0001), HbAlc (r=0.426, P<0.0001), fasting triglycerides (r=0.356, P=0.001), and parameters of innate immune response system such as IL6 (r=0.342, P=0.050) and CFB (r=0.404, P=0.002) in obese subjects with altered glucose tolerance, but not in individuals with normal glucose tolerance (NGT). In obese and NGT subjects, glucagon was associated with fasting triglycerides (r=0.475, P=0.003) and CFB (r=0.624, P=0.001). In obese subjects, glucagon (P=0.019) and CFB (P=0.002) independently contributed to 26% of fasting triglyceride variance (P<0.0001) after controlling for the effects of age and fasting serum glucose concentration in multiple lineal regression models. Moreover, concomitant with fat mass, fasting triglycerides, and CFB, weight loss led to significantly decreased circulating glucagon (-23.1%, P=0.004). CONCLUSIONS According to the current results, acute phase reactants such as IL6 and CFB are associated with fasting glucagon in metabolically compromised subjects. This suggests that glucagon may be behind the association between inflammatory and metabolic parameters in obesity-associated chronic low-grade inflammation.
Collapse
Affiliation(s)
- Francisco J Ortega
- Service of Diabetes, Endocrinology and Nutrition (UDEN), Institut d'Investigació Biomédica de Girona (IdIBGi), Hospital of Girona Dr Josep Trueta, Girona, Spain
| | | | | | | | | | | |
Collapse
|
43
|
Oberbach A, Blüher M, Wirth H, Till H, Kovacs P, Kullnick Y, Schlichting N, Tomm JM, Rolle-Kampczyk U, Murugaiyan J, Binder H, Dietrich A, von Bergen M. Combined proteomic and metabolomic profiling of serum reveals association of the complement system with obesity and identifies novel markers of body fat mass changes. J Proteome Res 2011; 10:4769-88. [PMID: 21823675 DOI: 10.1021/pr2005555] [Citation(s) in RCA: 167] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Obesity is associated with multiple adverse health effects and a high risk of developing metabolic and cardiovascular diseases. Therefore, there is a great need to identify circulating parameters that link changes in body fat mass with obesity. This study combines proteomic and metabolomic approaches to identify circulating molecules that discriminate healthy lean from healthy obese individuals in an exploratory study design. To correct for variations in physical activity, study participants performed a one hour exercise bout to exhaustion. Subsequently, circulating factors differing between lean and obese individuals, independent of physical activity, were identified. The DIGE approach yielded 126 differentially abundant spots representing 39 unique proteins. Differential abundance of proteins was confirmed by ELISA for antithrombin-III, clusterin, complement C3 and complement C3b, pigment epithelium-derived factor (PEDF), retinol binding protein 4 (RBP4), serum amyloid P (SAP), and vitamin-D binding protein (VDBP). Targeted serum metabolomics of 163 metabolites identified 12 metabolites significantly related to obesity. Among those, glycine (GLY), glutamine (GLN), and glycero-phosphatidylcholine 42:0 (PCaa 42:0) serum concentrations were higher, whereas PCaa 32:0, PCaa 32:1, and PCaa 40:5 were decreased in obese compared to lean individuals. The integrated bioinformatic evaluation of proteome and metabolome data yielded an improved group separation score of 2.65 in contrast to 2.02 and 2.16 for the single-type use of proteomic or metabolomics data, respectively. The identified circulating parameters were further investigated in an extended set of 30 volunteers and in the context of two intervention studies. Those included 14 obese patients who had undergone sleeve gastrectomy and 12 patients on a hypocaloric diet. For determining the long-term adaptation process the samples were taken six months after the treatment. In multivariate regression analyses, SAP, CLU, RBP4, PEDF, GLN, and C18:2 showed the strongest correlation to changes in body fat mass. The combined serum proteomic and metabolomic profiling reveals a link between the complement system and obesity and identifies both novel (C3b, CLU, VDBP, and all metabolites) and confirms previously discovered markers (PEDF, RBP4, C3, ATIII, and SAP) of body fat mass changes.
Collapse
Affiliation(s)
- Andreas Oberbach
- IFB Adiposity Diseases, Leipzig University Medical Centre, Leipzig, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Pathway knockout and redundancy in metabolic networks. J Theor Biol 2011; 270:63-9. [DOI: 10.1016/j.jtbi.2010.11.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2010] [Revised: 11/07/2010] [Accepted: 11/08/2010] [Indexed: 11/23/2022]
|
45
|
Malik TH, Cortini A, Carassiti D, Boyle JJ, Haskard DO, Botto M. The alternative pathway is critical for pathogenic complement activation in endotoxin- and diet-induced atherosclerosis in low-density lipoprotein receptor-deficient mice. Circulation 2010; 122:1948-56. [PMID: 20974996 PMCID: PMC2978131 DOI: 10.1161/circulationaha.110.981365] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2010] [Accepted: 08/30/2010] [Indexed: 11/16/2022]
Abstract
BACKGROUND The early components of the classical and lectin complement pathways have been shown to protect low-density lipoprotein receptor-deficient mice (Ldlr(-/-)) from early atherogenesis. However, the role of the alternative pathway remained unknown, and that was investigated in this study. METHODS AND RESULTS Mice lacking factor B (Bf(-/-)), the initiator of the alternative pathway, were crossed with Ldlr(-/-) mice and studied under different proatherogenic conditions. There was no statistically significant difference in lipid profiles or atherosclerotic lesion development between Bf(-/-)/Ldlr(-/-) and Ldlr(-/-) mice fed a low-fat diet. However, in these groups, administration of bacterial lipopolysaccharide led to a significant increase in atherosclerosis only in Ldlr(-/-) and not in Bf(-/-)/Ldlr(-/-) mice, indicating that the alternative pathway is necessary for endotoxin-mediated atherogenesis. Bf(-/-)/Ldlr(-/-) mice also had significantly decreased cross-sectional aortic root lesion fraction area and reduced lesion complexity compared with Ldlr(-/-) animals after a 12-week period of high-fat diet, although this was also accompanied by reduced levels of serum cholesterol. Under both experimental conditions, the atherosclerotic changes in the Bf(-/-)/Ldlr(-/-) mice were accompanied by a marked reduction in complement activation in the circulation and in atherosclerotic plaques, with no statistically significant differences in immunoglobulin G deposition or in the serum antibody response to oxidized low-density lipoprotein. CONCLUSIONS These data demonstrate that amplification of complement activation by the alternative pathway in response to lipopolysaccharide or high-fat diet plays a proatherogenic role.
Collapse
Affiliation(s)
- Talat H. Malik
- Rheumatology Section, Division of Immunology and Inflammation, Department of Medicine, Imperial College, London
| | - Andrea Cortini
- Rheumatology Section, Division of Immunology and Inflammation, Department of Medicine, Imperial College, London
| | - Daniele Carassiti
- Rheumatology Section, Division of Immunology and Inflammation, Department of Medicine, Imperial College, London
| | - Joseph J Boyle
- Vascular Sciences Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College, London
| | - Dorian O. Haskard
- Vascular Sciences Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College, London
| | - Marina Botto
- Rheumatology Section, Division of Immunology and Inflammation, Department of Medicine, Imperial College, London
| |
Collapse
|
46
|
Gao Y, Gauvreau D, Cianflone K. Hormone and pharmaceutical regulation of ASP production in 3T3-L1 adipocytes. J Cell Biochem 2010; 109:896-905. [PMID: 20069551 DOI: 10.1002/jcb.22464] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Several studies have demonstrated increases in acylation stimulating protein (ASP), and precursor protein C3 in obesity, diabetes and dyslipidemia, however the nature of the regulation is unknown. To evaluate chronic hormonal and pharmaceutical mediated changes in ASP and potential mechanisms, 3T3-L1 adipocytes were treated with physiological concentrations of relevant hormones and drugs currently used in treatment of metabolic diseases for 48 h. Medium ASP production and C3 secretion were evaluated in relation to changes in adipocyte lipid metabolism (cellular triglyceride (TG) mass, non-esterified fatty acid (NEFA) release and real-time FA uptake). Chylomicrons increased ASP production (up to 411 +/- 133% P < 0.05), while leptin, triiodothyronine, and beta-blockers atenolol and propranolol had no effect. Dexamethasone, lovastatin, rosiglitazone and rimonabant decreased ASP production (-53 to -85%, P < 0.05), associated with a decrease in the precursor protein C3 (-37% to -65%, P < 0.01). By contrast, epinephrine, progesterone, testosterone, angiotensin II and metformin also decreased ASP (-54% to -100%, P < 0.05), but without change in precursor protein C3, suggesting a direct effect on convertase activity, possibly mediated by interference (except metformin) due to marked increases in NEFA (5.6-31-fold, increased P < 0.05). Both lovastatin and metformin induced decreases in ASP were also associated with decreased TG mass (maximal -60%, P < 0.05) and real-time FA uptake (maximum -75%, P < 0.05), suggesting a change in adipocyte differentiation status. These in vitro results are consistent with in vivo ASP profiles in subjects, and suggest that ASP may be regulated through precursor C3 availability, convertase activity and differentiation status.
Collapse
Affiliation(s)
- Ying Gao
- Centre de Recherche Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Laval University, Québec, Canada
| | | | | |
Collapse
|
47
|
Fisette A, Cianflone K. The ASP and C5L2 pathway: another bridge between inflammation and metabolic homeostasis. ACTA ACUST UNITED AC 2010. [DOI: 10.2217/clp.10.21] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
48
|
Paglialunga S, Fisette A, Munkonda M, Gao Y, Richard D, Cianflone K. The effects of acylation stimulating protein supplementation VS antibody neutralization on energy expenditure in wildtype mice. BMC PHYSIOLOGY 2010; 10:4. [PMID: 20416070 PMCID: PMC2875207 DOI: 10.1186/1472-6793-10-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2009] [Accepted: 04/23/2010] [Indexed: 11/10/2022]
Abstract
BACKGROUND Acylation stimulating protein (ASP) is an adipogenic hormone that stimulates triglyceride (TG) synthesis and glucose transport in adipocytes. Previous studies have shown that ASP-deficient C3 knockout mice are hyperphagic yet lean, as they display increased oxygen consumption and fatty acid oxidation compared to wildtype mice. In the present study, antibodies against ASP (Anti-ASP) and human recombinant ASP (rASP) were tested in vitro and in vivo. Continuous administration for 4 weeks via osmotic mini-pump of Anti-ASP or rASP was evaluated in wildtype mice on a high-fat diet (HFD) to examine their effects on body weight, food intake and energy expenditure. RESULTS In mature murine adipocytes, rASP significantly stimulated fatty acid uptake (+243% vs PBS, P < 0.05) while Anti-ASP neutralized the rASP response. Mice treated with Anti-ASP showed elevated energy expenditure (P < 0.0001), increased skeletal muscle glucose oxidation (+141%, P < 0.001), reduced liver glycogen (-34%, P < 0.05) and glucose-6-phosphate content (-64%, P = 0.08) compared to control mice. There was no change in body weight, food intake, fasting insulin, adiponectin, CRP or TG levels compared to controls. Interestingly, HFD mice treated with rASP showed the opposite phenotype with reduced energy expenditure (P < 0.0001) and increased body weight (P < 0.05), cumulative food intake (P < 0.0001) and liver glycogen content (+59%, P < 0.05). Again, there was no change in circulating insulin, adiponectin, CRP or TG levels, however, plasma free fatty acids were reduced (-48%, P < 0.05). CONCLUSION In vitro, Anti-ASP effectively neutralized ASP stimulated fatty acid uptake. In vivo, Anti-ASP treatment increased whole body energy utilization while rASP increased energy storage. Therefore, ASP is a potent anabolic hormone that may also be a mediator of energy expenditure.
Collapse
Affiliation(s)
- Sabina Paglialunga
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, QC, G1V 4G5, Canada
| | | | | | | | | | | |
Collapse
|
49
|
Takahashi M, Ishida Y, Iwaki D, Kanno K, Suzuki T, Endo Y, Homma Y, Fujita T. Essential role of mannose-binding lectin-associated serine protease-1 in activation of the complement factor D. J Exp Med 2010; 207:29-37. [PMID: 20038603 PMCID: PMC2812541 DOI: 10.1084/jem.20090633] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Accepted: 11/23/2009] [Indexed: 11/29/2022] Open
Abstract
The complement system is an essential component of innate immunity, participating in the pathogenesis of inflammatory diseases and in host defense. In the lectin complement pathway, mannose-binding lectin (MBL) and ficolins act as recognition molecules, and MBL-associated serine protease (MASP) is a key enzyme; MASP-2 is responsible for the lectin pathway activation. The function of other serine proteases (MASP-1 and MASP-3) is still obscure. In this study, we generated a MASP-1- and MASP-3-deficient mouse model (Masp1/3-/-) and found that no activation of the alternative pathway was observed in Masp1/3-/- serum. Mass spectrometric analysis revealed that circulating complement factor D (Df) in Masp1/3-/- mice is a zymogen (pro-Df) with the activation peptide QPRGR at its N terminus. These results suggested that Masp1/3-/- mice failed to convert pro-Df to its active form, whereas it was generally accepted that the activation peptide of pro-Df is removed during its secretion and factor D constitutively exists in an active form in the circulation. Furthermore, recombinant MASP-1 converted pro-Df to the active form in vitro, although the activation mechanism of pro-Df by MASP-1 is still unclear. Thus, it is clear that MASP-1 is an essential protease of both the lectin and alternative complement pathways.
Collapse
Affiliation(s)
- Minoru Takahashi
- Department of Immunology, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Moreno-Navarrete JM, Martínez-Barricarte R, Catalán V, Sabater M, Gómez-Ambrosi J, Ortega FJ, Ricart W, Blüher M, Frühbeck G, Rodríguez de Cordoba S, Fernández-Real JM. Complement factor H is expressed in adipose tissue in association with insulin resistance. Diabetes 2010; 59:200-9. [PMID: 19833879 PMCID: PMC2797922 DOI: 10.2337/db09-0700] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Activation of the alternative pathway of the complement system, in which factor H (fH; complement fH [CFH]) is a key regulatory component, has been suggested as a link between obesity and metabolic disorders. Our objective was to study the associations between circulating and adipose tissue gene expressions of CFH and complement factor B (fB; CFB) with obesity and insulin resistance. RESEARCH DESIGN AND METHODS Circulating fH and fB were determined by enzyme-linked immunosorbent assay in 398 subjects. CFH and CFB gene expressions were evaluated in 76 adipose tissue samples, in isolated adipocytes, and in stromovascular cells (SVC) (n = 13). The effects of weight loss and rosiglitazone were investigated in independent cohorts. RESULTS Both circulating fH and fB were associated positively with BMI, waist circumference, triglycerides, and inflammatory parameters and negatively with insulin sensitivity and HDL cholesterol. For the first time, CFH gene expression was detected in human adipose tissue (significantly increased in subcutaneous compared with omental fat). CFH gene expression in omental fat was significantly associated with insulin resistance. In contrast, CFB gene expression was significantly increased in omental fat but also in association with fasting glucose and triglycerides. The SVC fraction was responsible for these differences, although isolated adipocytes also expressed fB and fH at low levels. Both weight loss and rosiglitazone led to significantly decreased circulating fB and fH levels. CONCLUSIONS Increased circulating fH and fB concentrations in subjects with altered glucose tolerance could reflect increased SVC-induced activation of the alternative pathway of complement in omental adipose tissue linked to insulin resistance and metabolic disturbances.
Collapse
Affiliation(s)
- José María Moreno-Navarrete
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomédica de Girona, and CIBER Fisiopatologia Obesidad y Nutricion, Instituto de Salud Carlos III, Girona, Spain
| | | | - Victoria Catalán
- Department of Endocrinology and Metabolic Research Laboratory, Clínica Universitaria, University of Navarra, Pamplona, Spain, and CIBER Fisiopatologia Obesidad y Nutricion, Instituto de Salud Carlos III, Pamplona, Spain
| | - Mònica Sabater
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomédica de Girona, and CIBER Fisiopatologia Obesidad y Nutricion, Instituto de Salud Carlos III, Girona, Spain
| | - Javier Gómez-Ambrosi
- Department of Endocrinology and Metabolic Research Laboratory, Clínica Universitaria, University of Navarra, Pamplona, Spain, and CIBER Fisiopatologia Obesidad y Nutricion, Instituto de Salud Carlos III, Pamplona, Spain
| | - Francisco José Ortega
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomédica de Girona, and CIBER Fisiopatologia Obesidad y Nutricion, Instituto de Salud Carlos III, Girona, Spain
| | - Wifredo Ricart
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomédica de Girona, and CIBER Fisiopatologia Obesidad y Nutricion, Instituto de Salud Carlos III, Girona, Spain
| | - Mathias Blüher
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Gema Frühbeck
- Department of Endocrinology and Metabolic Research Laboratory, Clínica Universitaria, University of Navarra, Pamplona, Spain, and CIBER Fisiopatologia Obesidad y Nutricion, Instituto de Salud Carlos III, Pamplona, Spain
| | | | - José Manuel Fernández-Real
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomédica de Girona, and CIBER Fisiopatologia Obesidad y Nutricion, Instituto de Salud Carlos III, Girona, Spain
- Corresponding author: José Manuel Fernández-Real,
| |
Collapse
|