1
|
Sunamoto M, Morohoshi K, Sato B, Mihashi R, Inui M, Yamada M, Miyado K, Kawano N. Complement Factor B Deficiency Is Dispensable for Female Fertility but Affects Microbiome Diversity and Complement Activity. Int J Mol Sci 2025; 26:1393. [PMID: 39941161 PMCID: PMC11818189 DOI: 10.3390/ijms26031393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/27/2025] [Accepted: 02/04/2025] [Indexed: 02/16/2025] Open
Abstract
Complement factor B (CFB) is a crucial component for the activation of the alternative pathway due to the formation of the C3 convertase with C3b, which further produces C3b to enhance the overall complement activity. Although Cfb is expressed not only in the immune tissues, but also in the reproductive tract, the physiological role of the alternative complement pathway in reproduction remains unclear. In this study, we addressed this issue by producing Cfb-knockout (KO) mice and analyzing their phenotypes. Sperm function, number of ovulated oocytes, and litter size were normal in KO mice. In contrast, the diversity of microbiomes in the gut and vaginal tract significantly increased in KO mice. Some serine protease activity in the serum from KO mice was lower than that of wild-type mice. Since the serum from KO mice showed significantly lower activity of the alternative complement pathway, CFB was found to be essential for this pathway. Our results indicate that although the alternative pathway is dispensable for normal fertility and development, it maintains the gut and vaginal microbiomes by suppressing their diversity and activating the alternative complement pathway.
Collapse
Affiliation(s)
- Manato Sunamoto
- Laboratory of Regulatory Biology, Department of Life Sciences, School of Agriculture, Meiji University, 1-1-1 Higashimita, Kawasaki 214-8571, Japan; (M.S.); (K.M.); (B.S.)
| | - Kazunori Morohoshi
- Laboratory of Regulatory Biology, Department of Life Sciences, School of Agriculture, Meiji University, 1-1-1 Higashimita, Kawasaki 214-8571, Japan; (M.S.); (K.M.); (B.S.)
- Division of Biomedical Engineering, National Defense Medical College Research Institute, 3-2 Namiki, Tokorozawa 359-8513, Japan
| | - Ban Sato
- Laboratory of Regulatory Biology, Department of Life Sciences, School of Agriculture, Meiji University, 1-1-1 Higashimita, Kawasaki 214-8571, Japan; (M.S.); (K.M.); (B.S.)
| | - Ryo Mihashi
- Laboratory of Regulatory Biology, Department of Life Sciences, School of Agriculture, Meiji University, 1-1-1 Higashimita, Kawasaki 214-8571, Japan; (M.S.); (K.M.); (B.S.)
| | - Masafumi Inui
- Laboratory of Animal Regeneration Systemology, Department of Life Sciences, School of Agriculture, Meiji University, 1-1-1 Higashimita, Kawasaki 214-8571, Japan;
| | - Mitsutoshi Yamada
- Department of Obstetrics and Gynecology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan;
| | - Kenji Miyado
- Department of Reproductive Biology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya, Tokyo 157-8535, Japan;
| | - Natsuko Kawano
- Laboratory of Regulatory Biology, Department of Life Sciences, School of Agriculture, Meiji University, 1-1-1 Higashimita, Kawasaki 214-8571, Japan; (M.S.); (K.M.); (B.S.)
| |
Collapse
|
2
|
Wang Z, Zhang T, Wang X, Zhai J, He L, Ma S, Zuo Q, Zhang G, Guo Y. Association between circulatory complement activation and hypertensive renal damage: a case-control study. Ren Fail 2024; 46:2365396. [PMID: 38874150 PMCID: PMC11182054 DOI: 10.1080/0886022x.2024.2365396] [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: 02/05/2024] [Accepted: 06/03/2024] [Indexed: 06/15/2024] Open
Abstract
OBJECTIVE The aim of this study was to investigate the potential importance of complement system activation, with particular emphasis on the complement alternative pathway (AP), in the pathogenesis of hypertensive renal damage. METHODS Serum complement C3, complement Factor H (CFH) and AP activation were assessed in 66 participants with established essential hypertension with renal damage (RD). Fifty-nine patients with age- and sex-matched essential hypertension without renal damage (NRD) and 58 healthy participants (normal) were selected. RESULTS Our study revealed that C3 and AP50 continuously increased from normal to NRD to RD (p < 0.05, respectively), while CFH was significantly lower than that in NRD and healthy participants (p < 0.05, respectively). After multifactorial logistic regression analysis corrected for confounders, elevated serum C3 (p = 0.001) and decreased CFH (p < 0.001) were found to be independent risk factors for hypertension in healthy participants; elevated serum C3 (p = 0.034), elevated AP50 (p < 0.001), decreased CFH (p < 0.001), increased age (p = 0.011) and increased BMI (p = 0.013) were found to be independent risk factors for the progression of hypertension to hypertensive renal damage; elevated serum C3 (p = 0.017), elevated AP50 (p = 0.023), decreased CFH (p = 0.005) and increased age (p = 0.041) were found to be independent risk factors for the development of hypertensive renal damage in healthy participants. CONCLUSION Abnormal activation of complement, particularly complement AP, may be a risk factor for the development and progression of hypertensive renal damage.
Collapse
Affiliation(s)
- Zhongli Wang
- Department of Physical Examination Center, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Tingting Zhang
- Department of Geriatric Cardiology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Xinyu Wang
- College of Postgraduate, Hebei North University, Zhangjiakou, Hebei, China
| | - Jianlong Zhai
- Department of Cardiology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Lili He
- Department of Geriatric Cardiology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Sai Ma
- Department of Internal Medicine, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Qingjuan Zuo
- Department of Geriatric Cardiology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Guorui Zhang
- Department of Cardiology, The Third Hospital of Shijiazhuang City Affiliated to Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yifang Guo
- Department of Geriatric Cardiology, Hebei General Hospital, Shijiazhuang, Hebei, China
| |
Collapse
|
3
|
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
|
4
|
Hantikainen E, Weichenberger CX, Dordevic N, Verri Hernandes V, Foco L, Gögele M, Melotti R, Pattaro C, Ralser M, Amari F, Farztdinov V, Mülleder M, Pramstaller PP, Rainer J, Domingues FS. Metabolite and protein associations with general health in the population-based CHRIS study. Sci Rep 2024; 14:26635. [PMID: 39496618 PMCID: PMC11535378 DOI: 10.1038/s41598-024-75627-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/07/2024] [Indexed: 11/06/2024] Open
Abstract
Identifying biomarkers able to discriminate individuals on different health trajectories is crucial to understand the molecular basis of age-related morbidity. We investigated multi-omics signatures of general health and organ-specific morbidity, as well as their interconnectivity. We examined cross-sectional metabolome and proteome data from 3,142 adults of the Cooperative Health Research in South Tyrol (CHRIS) study, an Alpine population study designed to investigate how human biology, environment, and lifestyle factors contribute to people's health over time. We had 174 metabolites and 148 proteins quantified from fasting serum and plasma samples. We used the Cumulative Illness Rating Scale (CIRS) Comorbidity Index (CMI), which considers morbidity in 14 organ systems, to assess health status (any morbidity vs. healthy). Omics-signatures for health status were identified using random forest (RF) classifiers. Linear regression models were fitted to assess directionality of omics markers and health status associations, as well as to identify omics markers related to organ-specific morbidity. Next to age, we identified 21 metabolites and 10 proteins as relevant predictors of health status and results confirmed associations for serotonin and glutamate to be age-independent. Considering organ-specific morbidity, several metabolites and proteins were jointly related to endocrine, cardiovascular, and renal morbidity. To conclude, circulating serotonin was identified as a potential novel predictor for overall morbidity.
Collapse
Affiliation(s)
| | | | | | | | - Luisa Foco
- Institute for Biomedicine, Eurac Research, Bolzano, Italy
| | - Martin Gögele
- Institute for Biomedicine, Eurac Research, Bolzano, Italy
| | | | | | - Markus Ralser
- Department of Biochemistry, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Fatma Amari
- Department of Biochemistry, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Vadim Farztdinov
- Core Facility, High-Throughput Mass Spectrometry, Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Michael Mülleder
- Core Facility, High-Throughput Mass Spectrometry, Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | | | | | | |
Collapse
|
5
|
Wojciuk B, Frulenko I, Brodkiewicz A, Kita D, Baluta M, Jędrzejczyk F, Budkowska M, Turkiewicz K, Proia P, Ciechanowicz A, Kostrzewa-Nowak D, Nowak R. The Complement System as a Part of Immunometabolic Post-Exercise Response in Adipose and Muscle Tissue. Int J Mol Sci 2024; 25:11608. [PMID: 39519159 PMCID: PMC11545998 DOI: 10.3390/ijms252111608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 10/17/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
The precise molecular processes underlying the complement's activation, which follows exposure to physical stress still remain to be fully elucidated. However, some possible mechanisms could play a role in initiating changes in the complement's activity, which are observed post-exposure to physical stress stimuli. These are mainly based on metabolic shifts that occur in the microenvironment of muscle tissue while performing its function with increased intensity, as well as the adipose tissue's role in sterile inflammation and adipokine secretion. This review aims to discuss the current opinions on the possible link between the complement activation and diet, age, sex, and health disorders with a particular emphasis on endocrinopathies and, furthermore, the type of physical activity and overall physical fitness. It has been indicated that regular physical activity incorporated into therapeutic strategies potentially improves the management of particular diseases, such as, e.g., autoimmune conditions. Moreover, it represents a favorable influence on immunoaging processes. A better understanding of the complement system's interaction with physical activity will support established clinical therapies targeting complement components.
Collapse
Affiliation(s)
- Bartosz Wojciuk
- Department of Diagnostic Immunology, Chair of Microbiology, Immunology and Laboratory Medicine, Pomeranian Medical University in Szczecin, 72 Powstańców Wlkp. Al., 70-111 Szczecin, Poland;
| | - Ignacy Frulenko
- Pomeranian Medical University in Szczecin, 1 Rybacka St., 70-204 Szczecin, Poland;
- Department of Pathology, Pomeranian Medical University in Szczecin, 1 Unii Lubelskiej St., 71-242 Szczecin, Poland
| | - Andrzej Brodkiewicz
- Department of Pediatrics, Pediatric Nephrology, Dialysis and Acute Intoxications, Pomeranian Medical University, 4 Mączna St., 70-204 Szczecin, Poland; (A.B.); (D.K.); (M.B.); (F.J.)
| | - Dagmara Kita
- Department of Pediatrics, Pediatric Nephrology, Dialysis and Acute Intoxications, Pomeranian Medical University, 4 Mączna St., 70-204 Szczecin, Poland; (A.B.); (D.K.); (M.B.); (F.J.)
| | - Monica Baluta
- Department of Pediatrics, Pediatric Nephrology, Dialysis and Acute Intoxications, Pomeranian Medical University, 4 Mączna St., 70-204 Szczecin, Poland; (A.B.); (D.K.); (M.B.); (F.J.)
| | - Filip Jędrzejczyk
- Department of Pediatrics, Pediatric Nephrology, Dialysis and Acute Intoxications, Pomeranian Medical University, 4 Mączna St., 70-204 Szczecin, Poland; (A.B.); (D.K.); (M.B.); (F.J.)
| | - Marta Budkowska
- Department of Medical Analytics, Pomeranian Medical University of Szczecin, 72 Powstańców Wlkp. Al., 70-111 Szczecin, Poland;
| | - Karolina Turkiewicz
- Department of Laboratory Diagnostics, University Clinical Hospital No. 2, Pomeranian Medical University in Szczecin, 72 Powstańców Wlkp. Al., 70-111 Szczecin, Poland;
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University in Szczecin, 72 Powstańców Wlkp. Al., 70-111 Szczecin, Poland;
| | - Patrizia Proia
- Sport and Exercise Sciences Research Unit, Department of Psychology, Educational Science and Human Movement, University of Palermo, 90144 Palermo, Italy;
| | - Andrzej Ciechanowicz
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University in Szczecin, 72 Powstańców Wlkp. Al., 70-111 Szczecin, Poland;
| | - Dorota Kostrzewa-Nowak
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University in Szczecin, 72 Powstańców Wlkp. Al., 70-111 Szczecin, Poland;
- Department of Biopharmaceutics and Pharmacodynamics, Faculty of Pharmacy, Medical University of Gdańsk, 107 Hallera St., 80-416 Gdańsk, Poland
| | - Robert Nowak
- Department of Pathology, Pomeranian Medical University in Szczecin, 1 Unii Lubelskiej St., 71-242 Szczecin, Poland
- Institute of Physical Culture Sciences, University of Szczecin, 17C Narutowicza St., 70-240 Szczecin, Poland
| |
Collapse
|
6
|
He Q, Wei Y, Qian Y, Zhong M. Pathophysiological dynamics in the contact, coagulation, and complement systems during sepsis: Potential targets for nafamostat mesilate. JOURNAL OF INTENSIVE MEDICINE 2024; 4:453-467. [PMID: 39310056 PMCID: PMC11411436 DOI: 10.1016/j.jointm.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/17/2024] [Accepted: 02/07/2024] [Indexed: 09/25/2024]
Abstract
Sepsis is a life-threatening syndrome resulting from a dysregulated host response to infection. It is the primary cause of death in the intensive care unit, posing a substantial challenge to human health and medical resource allocation. The pathogenesis and pathophysiology of sepsis are complex. During its onset, pro-inflammatory and anti-inflammatory mechanisms engage in intricate interactions, possibly leading to hyperinflammation, immunosuppression, and long-term immune disease. Of all critical outcomes, hyperinflammation is the main cause of early death among patients with sepsis. Therefore, early suppression of hyperinflammation may improve the prognosis of these patients. Nafamostat mesilate is a serine protease inhibitor, which can inhibit the activation of the complement system, coagulation system, and contact system. In this review, we discuss the pathophysiological changes occurring in these systems during sepsis, and describe the possible targets of the serine protease inhibitor nafamostat mesilate in the treatment of this condition.
Collapse
Affiliation(s)
- Qiaolan He
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yilin Wei
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yiqi Qian
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ming Zhong
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Lung Inflammation and Injury, Shanghai, China
- Shanghai Institute of Infectious Disease and Biosecurity, School of Public Health, Fudan University, Shanghai, China
| |
Collapse
|
7
|
Scott MA, Valeris-Chacin R, Thompson AC, Woolums AR, Karisch BB. Comprehensive time-course gene expression evaluation of high-risk beef cattle to establish immunological characteristics associated with undifferentiated bovine respiratory disease. Front Immunol 2024; 15:1412766. [PMID: 39346910 PMCID: PMC11427276 DOI: 10.3389/fimmu.2024.1412766] [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: 04/05/2024] [Accepted: 08/20/2024] [Indexed: 10/01/2024] Open
Abstract
Bovine respiratory disease (BRD) remains the leading infectious disease in beef cattle production systems. Host gene expression upon facility arrival may indicate risk of BRD development and severity. However, a time-course approach would better define how BRD development influences immunological and inflammatory responses after disease occurrences. Here, we evaluated whole blood transcriptomes of high-risk beef cattle at three time points to elucidate BRD-associated host response. Sequenced jugular whole blood mRNA from 36 cattle (2015: n = 9; 2017: n = 27) across three time points (n = 100 samples; days [D]0, D28, and D63) were processed through ARS-UCD1.2 reference-guided assembly (HISAT2/Stringtie2). Samples were categorized into BRD-severity cohorts (Healthy, n = 14; Treated 1, n = 11; Treated 2+, n = 11) via frequency of antimicrobial clinical treatment. Assessment of gene expression patterns over time within each BRD cohort was modeled through an autoregressive hidden Markov model (EBSeq-HMM; posterior probability ≥ 0.5, FDR < 0.01). Mixed-effects negative binomial models (glmmSeq; FDR < 0.05) and edgeR (FDR < 0.10) identified differentially expressed genes between and across cohorts overtime. A total of 2,580, 2,216, and 2,381 genes were dynamically expressed across time in Healthy, Treated 1, and Treated 2+ cattle, respectively. Genes involved in the production of specialized resolving mediators (SPMs) decreased at D28 and then increased by D63 across all three cohorts. Accordingly, SPM production and alternative complement were differentially expressed between Healthy and Treated 2+ at D0, but not statistically different between the three groups by D63. Magnitude, but not directionality, of gene expression related to SPM production, alternative complement, and innate immune response signified Healthy and Treated 2+ cattle. Differences in gene expression at D63 across the three groups were related to oxygen binding and carrier activity, natural killer cell-mediated cytotoxicity, cathelicidin production, and neutrophil degranulation, possibly indicating prolonged airway pathology and inflammation weeks after clinical treatment for BRD. These findings indicate genomic mechanisms indicative of BRD development and severity over time.
Collapse
Affiliation(s)
- Matthew A Scott
- Veterinary Education, Research, and Outreach Program, Texas A&M University, Canyon, TX, United States
| | - Robert Valeris-Chacin
- Veterinary Education, Research, and Outreach Program, Texas A&M University, Canyon, TX, United States
| | - Alexis C Thompson
- Texas A&M Veterinary Medical Diagnostic Laboratory, Canyon, TX, United States
| | - Amelia R Woolums
- Department of Pathobiology and Population Medicine, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, United States
| | - Brandi B Karisch
- Department of Animal and Dairy Sciences, Mississippi State University, Mississippi State, MS, United States
| |
Collapse
|
8
|
Reeve J. De-stabilizing innate immunity in COVID-19: effects of its own positive feedback and erratic viraemia on the alternative pathway of complement. ROYAL SOCIETY OPEN SCIENCE 2024; 11:221597. [PMID: 38234438 PMCID: PMC10791537 DOI: 10.1098/rsos.221597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 12/08/2023] [Indexed: 01/19/2024]
Abstract
Complement provides powerful, fast responses in the human circulation to SARS-CoV-2 (COVID-19 virus) infection of the lower respiratory tract. COVID-19 effects were investigated in a revised human in silico Mass Action model of complement's alternative pathway (AP) responses. Bursts of newly circulating virions increased the fission of Complement protein C3 into C3a and C3b via stimulation of the lectin pathway or inhibited complement factor H. Viral reproduction sub-models incorporated smoothly exponential or step-wise exponential growth. Starting complement protein concentrations were drawn randomly from published normal male or female ranges and each infection model run for 10 days. C3 and factor B (FB) syntheses driven by Lectin Pathway stimulation led to declining plasma C3 and increasing FB concentrations. The C3-convertase concentration, a driver of viral elimination, could match viral growth over three orders of magnitude but near-complete exhaustion of circulating C3 was more prevalent with step-wise than with 'smooth' increases in viral stimulation. C3 exhaustion could be prolonged. Type 2 Diabetes and hypertension led to greatly increased peak C3-convertase concentrations, as did short-term variability of COVID-19 viraemia, pulmonary capillary clotting and secondary acidosis. Positive feedback in the AP greatly extends its response range at the expense of stability.
Collapse
Affiliation(s)
- Jonathan Reeve
- Senior Research Fellow, Nuffield Department of Orthopaedics, Rheumatological and Musculoskeletal Sciences, University of Oxford Botnar Research Centre, Windmill Road, Oxford OX3 7LD, UK
| |
Collapse
|
9
|
Chen JY, Zhang L, Yang M, Hughes ED, Freeman ZT, Saunders TL, Lin F. Development of a C3 Humanized Rat as a New Model for Evaluating Novel C3 Inhibitors. J Innate Immun 2023; 16:56-65. [PMID: 38035563 PMCID: PMC10786584 DOI: 10.1159/000534963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
INTRODUCTION C3 is central for all complement activation pathways, thus making it an attractive therapeutic target. Many C3-targeted agents are under extensive development with one already approved for clinical use. However, most, if not all, C3 inhibitors are human or nonhuman primate C3-specific, making evaluating their efficacies in vivo before a clinical trial extremely difficult and costly. METHODS We first studied the compatibility of human C3 in the rat complement system, then developed a C3 humanized rat using the CRISPR/Cas9 technology. We thoroughly characterized the resultant human C3 humanized rats and tested the treatment efficacy of an established primate-specific C3 inhibitor in a model of complement-mediated hemolysis in the C3 humanized rats. RESULTS We found that supplementing human C3 protein into the C3-deficient rat blood restored its complement activity, which was inhibited by rat factor H or compstatin, suggesting that human C3 is compatible to the rat complement system. The newly developed C3 humanized rats appeared healthy and expressed human but not rat C3 without detectable spontaneous C3 activation. More importantly, complement-mediated hemolysis in the C3 humanized rats was also inhibited by compstatin both in vitro and in vivo. CONCLUSION The successfully developed C3 humanized rats provided a much-desired rodent model to evaluate novel C3 inhibitors in vivo as potential drugs.
Collapse
Affiliation(s)
- Jin Y. Chen
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Lingjun Zhang
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Maojing Yang
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | - Zachary T. Freeman
- Transgenic Animal Model Core, University of Michigan, Ann Arbor, MI, USA
| | - Thomas L. Saunders
- Transgenic Animal Model Core, University of Michigan, Ann Arbor, MI, USA
| | - Feng Lin
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
10
|
Brown LN, Barth JL, Jafri S, Rumschlag JA, Jenkins TR, Atkinson C, Lang H. Complement factor B is essential for the proper function of the peripheral auditory system. Front Neurol 2023; 14:1214408. [PMID: 37560455 PMCID: PMC10408708 DOI: 10.3389/fneur.2023.1214408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 07/05/2023] [Indexed: 08/11/2023] Open
Abstract
Sensorineural hearing loss is associated with dysfunction of cochlear cells. Although immune cells play a critical role in maintaining the inner ear microenvironment, the precise immune-related molecular mechanisms underlying the pathophysiology of hearing loss remain unclear. The complement cascade contributes to the regulation of immune cell activity. Additionally, activation of the complement cascade can lead to the cellular opsonization of cells and pathogens, resulting in their engulfment and elimination by phagocytes. Complement factor B (fB) is an essential activator protein in the alternative complement pathway, and variations in the fB gene are associated with age-related macular degeneration. Here we show that mice of both sexes deficient in fB functional alleles (fB-/-) demonstrate progressive hearing impairment. Transcriptomic analysis of auditory nerves from adult mice detected 706 genes that were significantly differentially expressed between fB-/- and wild-type control animals, including genes related to the extracellular matrix and neural development processes. Additionally, a subset of differentially expressed genes was related to myelin function and neural crest development. Histological and immunohistochemical investigations revealed pathological alterations in auditory nerve myelin sheathes of fB-/- mice. Pathological alterations were also seen in the stria vascularis of the cochlear lateral wall in these mice. Our results implicate fB as an integral regulator of myelin maintenance and stria vascularis integrity, underscoring the importance of understanding the involvement of immune signaling pathways in sensorineural hearing loss.
Collapse
Affiliation(s)
- LaShardai N. Brown
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Jeremy L. Barth
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
| | - Shabih Jafri
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Jeffrey A. Rumschlag
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Tyreek R. Jenkins
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Carl Atkinson
- Division of Pulmonary Medicine, University of Florida, Gainesville, FL, United States
| | - Hainan Lang
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
11
|
Lopez-Tello J, Yong HEJ, Sandovici I, Dowsett GKC, Christoforou ER, Salazar-Petres E, Boyland R, Napso T, Yeo GSH, Lam BYH, Constancia M, Sferruzzi-Perri AN. Fetal manipulation of maternal metabolism is a critical function of the imprinted Igf2 gene. Cell Metab 2023; 35:1195-1208.e6. [PMID: 37437545 DOI: 10.1016/j.cmet.2023.06.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/12/2023] [Accepted: 06/09/2023] [Indexed: 07/14/2023]
Abstract
Maternal-offspring interactions in mammals involve both cooperation and conflict. The fetus has evolved ways to manipulate maternal physiology to enhance placental nutrient transfer, but the mechanisms involved remain unclear. The imprinted Igf2 gene is highly expressed in murine placental endocrine cells. Here, we show that Igf2 deletion in these cells impairs placental endocrine signaling to the mother, without affecting placental morphology. Igf2 controls placental hormone production, including prolactins, and is crucial to establish pregnancy-related insulin resistance and to partition nutrients to the fetus. Consequently, fetuses lacking placental endocrine Igf2 are growth restricted and hypoglycemic. Mechanistically, Igf2 controls protein synthesis and cellular energy homeostasis, actions dependent on the placental endocrine cell type. Igf2 loss also has additional long-lasting effects on offspring metabolism in adulthood. Our study provides compelling evidence for an intrinsic fetal manipulation system operating in placenta that modifies maternal metabolism and fetal resource allocation, with long-term consequences for offspring metabolic health.
Collapse
Affiliation(s)
- Jorge Lopez-Tello
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK.
| | - Hannah E J Yong
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A(∗)STAR), 30 Medical Drive, Singapore 117609, Singapore
| | - Ionel Sandovici
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge CB2 0SW, UK; Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science and, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Georgina K C Dowsett
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science and, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Efthimia R Christoforou
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Esteban Salazar-Petres
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Rebecca Boyland
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; Royal Devon and Exeter Hospital NHS Trust, Barrack Rd, Exeter EX2 5DW, UK
| | - Tina Napso
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Giles S H Yeo
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science and, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Brian Y H Lam
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science and, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Miguel Constancia
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge CB2 0SW, UK; Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science and, University of Cambridge, Cambridge CB2 0QQ, UK.
| | - Amanda N Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK.
| |
Collapse
|
12
|
Sarkar MS, Mia MM, Amin MA, Hossain MS, Islam MZ. Bioinformatics and network biology approach to identifying type 2 diabetes genes and pathways that influence the progression of breast cancer. Heliyon 2023; 9:e16151. [PMID: 37234659 PMCID: PMC10205526 DOI: 10.1016/j.heliyon.2023.e16151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 04/28/2023] [Accepted: 05/07/2023] [Indexed: 05/28/2023] Open
Abstract
Breast cancer is the second most prevalent malignancy affecting women. Postmenopausal women breast tumor is one of the top causes of death in women, accounting for 23% of cancer cases. Type 2 diabetes, a worldwide pandemic, has been connected to a heightened risk of several malignancies, although its association with breast cancer is still uncertain. In comparison to non-diabetic women, women with T2DM had a 23% elevated likelihood of developing breast cancer. It is difficult to determine causative or genetic susceptibility that connect T2DM and breast cancer. We created a large-scale network-based quantitative approach employing unbiased methods to discover abnormally amplified genes in both T2DM and breast cancer, to solve these issues. We performed transcriptome analysis to uncover identical genetic biomarkers and pathways to clarify the connection between T2DM and breast cancer patients. In this study, two RNA-seq datasets (GSE103001 and GSE86468) from the Gene Expression Omnibus (GEO) are used to identify mutually differentially expressed genes (DEGs) for breast cancer and T2DM, as well as common pathways and prospective medicines. Firstly, 45 shared genes (30 upregulated and 15 downregulated) between T2D and breast cancer were detected. We employed gene ontology and pathway enrichment to characterize prevalent DEGs' molecular processes and signal transduction pathways and observed that T2DM has certain connections to the progression of breast cancer. Using several computational and statistical approaches, we created a protein-protein interactions (PPI) network and revealed hub genes. These hub genes can be potential biomarkers, which may also lead to new therapeutic strategies for investigated diseases. We conducted TF-gene interactions, gene-microRNA interactions, protein-drug interactions, and gene-disease associations to find potential connections between T2DM and breast cancer pathologies. We assume that the potential drugs that emerged from this study could be useful therapeutic values. Researchers, doctors, biotechnologists, and many others may benefit from this research.
Collapse
Affiliation(s)
- Md Sumon Sarkar
- Department of Pharmacy, Islamic University, Kushtia-7003, Bangladesh
| | - Md Misor Mia
- Department of Pharmacy, Islamic University, Kushtia-7003, Bangladesh
| | - Md Al Amin
- Department of Computer Science & Engineering, Prime University, Dhaka-1216, Bangladesh
| | - Md Sojib Hossain
- Department of Mathematics, Govt. Bangla College, Dhaka-1216, Bangladesh
| | - Md Zahidul Islam
- Department of Information & Communication Technology, Islamic University, Kushtia-7003, Bangladesh
| |
Collapse
|
13
|
Smith-Jackson K, Harrison RA. Alternative pathway activation in pregnancy, a measured amount "complements" a successful pregnancy, too much results in adverse events. Immunol Rev 2023; 313:298-319. [PMID: 36377667 PMCID: PMC10100418 DOI: 10.1111/imr.13169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
During pregnancy, the maternal host must adapt in order to enable growth of the fetus. These changes affect all organ systems and are designed both to protect the fetus and to minimize risk to the mother. One of the most prominent adaptations involves the immune system. The semi-allogenic fetoplacental unit has non-self components and must be protected against attack from the host. This requires both attenuation of adaptive immunity and protection from innate immune defense mechanisms. One of the key innate immune players is complement, and it is important that the fetoplacental unit is not identified as non-self and subjected to complement attack. Adaptation of the complement response must, however, be managed in such a way that maternal protection against infection is not compromised. As the complement system also plays a significant facilitating role in many of the stages of a normal pregnancy, it is also important that any necessary adaptation to accommodate the semi-allogenic aspects of the fetoplacental unit does not compromise this. In this review, both the physiological role of the alternative pathway of complement in facilitating a normal pregnancy, and its detrimental participation in pregnancy-specific disorders, are discussed.
Collapse
Affiliation(s)
- Kate Smith-Jackson
- Complement Therapeutics Research Group, Translational and Clinical Research Institute, Faculty of Medical Science, Newcastle University, Newcastle-upon-Tyne, UK.,The National Renal Complement Therapeutics Centre (NRCTC), Newcastle-upon-Tyne Hospitals NHS Foundation Trust, Newcastle-upon-Tyne, UK
| | | |
Collapse
|
14
|
Luo P, Xu J, Xu K, Jing W, Liu L, Xu P. Exploring the genetic relationship between deep vein thrombosis and plasma protein: a new research idea. Expert Rev Hematol 2022; 15:867-873. [PMID: 35857435 DOI: 10.1080/17474086.2022.2104707] [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] [Indexed: 02/08/2023]
Abstract
BACKGROUND The aim of this article is to scan and analyze the genetic correlation between plasma proteome and deep venous thrombosis(DVT), and to explore the correlation between plasma protein and DVT. RESEARCH DESIGN AND METHODS GWAS data of DVT and plasma proteins were analyzed with linkage disequilibrium scores, and plasma proteins that were genetically associated with DVT were screened out. To ascertain the causal link between potential plasma proteins and DVT, a Mendelian randomized (MR) study was used. This study used STRING to examine the pathogenesis of DVT in connection with the gene encoding plasma protein. RESULTS Several suggestive plasma proteins were detected for DVT, such as Complement factor B (correlation coefficient =0.3883 P value=0.0177), Chromogranin-A (correlation coefficient =-0.4786, P value=0.0158). Through MR analysis, we found that there was a significant positive causal relationship between Chromogranin-A (exposure) and DVT (outcome) (β=-0.0117, SE=0.0013, P<0.0001). Our STRING analysis revealed that hsa04610 was associated with coagulation cascade in the KEGG pathway of Complement factor B(P<0.0001), which was based on GO and KEGG analysis of 8 selected plasma proteins. CONCLUSIONS A genetic link between plasma protein and DVT was thoroughly investigated. Our findings provide a fresh perspective on the genetics and pathogenesis of DVT.
Collapse
Affiliation(s)
- Pan Luo
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, Shanxi, 710054, China
| | - Jiawen Xu
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, 37# Guoxue Road, Chengdu, 610041, People's Republic of China
| | - Ke Xu
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, Shanxi, 710054, China
| | - Wensen Jing
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, Shanxi, 710054, China
| | - Lin Liu
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, Shanxi, 710054, China
| | - Peng Xu
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, Shanxi, 710054, China
| |
Collapse
|
15
|
Ekhzaimy AA, Masood A, Benabdelkamel H, Elhassan T, Musambil M, Alfadda AA. Plasma proteomics reveals an improved cardio-metabolic profile in patients with type 2 diabetes post-liraglutide treatment. Diab Vasc Dis Res 2022; 19:14791641221094322. [PMID: 35616478 PMCID: PMC9152203 DOI: 10.1177/14791641221094322] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Diabetes mellitus is a chronic multisystem disease with a high global prevalence, including in Saudi Arabia. The Glucagon-like Peptide (GLP-1) receptor agonist liraglutide is known to lower glucose levels, reduce weight and improve cardiovascular outcome. However, mechanisms underlying the benefits of liraglutide treatment in patients with type 2 diabetes mellitus (T2DM) remain unclear. METHODS In the present study, a 2D-DIGE MALDI-TOF mass spectrometric approach combined with bioinformatics and network pathway analysis explore the plasma proteomic profile. The study involved 20 patients with T2DM with mean age of 54.4 ± 9.5 years and Hemoglobin A1c (HbA1c) between 8% and 11% (inclusive). RESULTS A statistically significant change (p < .006) was observed in HbA1c with no significant changes in body weight, renal function, or markers of dyslipidemia post-treatment with liraglutide. 2 D-DIGE gel analysis identified significant changes (⩾1.5-fold change, Analysis of variance (ANOVA), p ⩽ 0.05) in 72 proteins, (62 down and 10 up) in liraglutide pre-treatment compared to the post-treatment state. Proteins identified in our study were found to regulate metabolic processes including acute phase response proteins, enzymes, apolipoproteins with involvement of the inflammatory signaling pathways, NF-κB, AKT, and p38 MAPK. CONCLUSION Liraglutide treatment decreased levels of acute phase response that to reduce the systemic chronic inflammatory state and oxidative stress, and eventually improve the cardio-metabolic profile in these patients.
Collapse
Affiliation(s)
- Aishah A Ekhzaimy
- Department of Medicine, College of Medicine and King Saud Medical City, King Saud University, Riyadh, Saudi Arabia
| | - Afshan Masood
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Hicham Benabdelkamel
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Tasnem Elhassan
- Department of Medicine, College of Medicine and King Saud Medical City, King Saud University, Riyadh, Saudi Arabia
| | - Mohthash Musambil
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Assim A Alfadda
- Department of Medicine, College of Medicine and King Saud Medical City, King Saud University, Riyadh, Saudi Arabia
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
- Strategic Center for Diabetes Research, College of Medicine, King Saud University, Riyadh, Saudi Arabia
- Assim A Alfadda, Department of Medicine, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia.
| |
Collapse
|
16
|
Wang C, Wang Z, Zhang W. The potential role of complement alternative pathway activation in hypertensive renal damage. Exp Biol Med (Maywood) 2022; 247:797-804. [PMID: 35473318 DOI: 10.1177/15353702221091986] [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] [Indexed: 11/17/2022] Open
Abstract
Hypertensive renal damage is a common secondary kidney disease caused by poor control of blood pressure. Recent evidence has revealed abnormal activation of the complement alternative pathway (AP) in hypertensive patients and animal models and that this phenomenon is related to hypertensive renal damage. Conditions in the setting of hypertension, including high renin concentration, reduced binding of factor H to the glomerular basement membrane, and abnormal local synthesis of complement proteins, potentially promote the AP activation in the kidney. The products of the AP activation promote the phenotypic transition of mesangial cells and tubular cells, attack endothelial cells and recruit immunocytes to worsen hypertensive renal damage. The effects of complement inhibition on hypertensive renal damage are contradictory. Although clinical data support the use of C5 monoclonal antibody in malignant hypertension, pharmacological inhibition in hypertensive animals provides little benefit to kidney function. Therefore, the role of the complement AP in the pathogenesis of hypertensive renal damage and the value of complement inhibition in hypertensive renal damage treatment must be further explored.
Collapse
Affiliation(s)
- Chongjian Wang
- Division of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhiyu Wang
- Division of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wen Zhang
- Division of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
17
|
Yin X, Takov K, Straube R, Voit-Bak K, Graessler J, Julius U, Tselmin S, Rodionov RN, Barbir M, Walls M, Theofilatos K, Mayr M, Bornstein SR. Precision Medicine Approach for Cardiometabolic Risk Factors in Therapeutic Apheresis. Horm Metab Res 2022; 54:238-249. [PMID: 35413745 DOI: 10.1055/a-1776-7943] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Lipoprotein apheresis (LA) is currently the most powerful intervention possible to reach a maximal reduction of lipids in patients with familial hypercholesterolemia and lipoprotein(a) hyperlipidemia. Although LA is an invasive method, it has few side effects and the best results in preventing further major cardiovascular events. It has been suggested that the highly significant reduction of cardiovascular complications in patients with severe lipid disorders achieved by LA is mediated not only by the potent reduction of lipid levels but also by the removal of other proinflammatory and proatherogenic factors. Here we performed a comprehensive proteomic analysis of patients on LA treatment using intra-individually a set of differently sized apheresis filters with the INUSpheresis system. This study revealed that proteomic analysis correlates well with routine clinical chemistry in these patients. The method is eminently suited to discover new biomarkers and risk factors for cardiovascular disease in these patients. Different filters achieve reduction and removal of proatherogenic proteins in different quantities. This includes not only apolipoproteins, C-reactive protein, fibrinogen, and plasminogen but also proteins like complement factor B (CFAB), protein AMBP, afamin, and the low affinity immunoglobulin gamma Fc region receptor III-A (FcγRIIIa) among others that have been described as atherosclerosis and metabolic vascular diseases promoting factors. We therefore conclude that future trials should be designed to develop an individualized therapy approach for patients on LA based on their metabolic and vascular risk profile. Furthermore, the power of such cascade filter treatment protocols may improve the prevention of cardiometabolic disease and its complications.
Collapse
Affiliation(s)
- X Yin
- Kings College London, London, UK
| | - K Takov
- Kings College London, London, UK
| | - R Straube
- Zentrum für Apherese- und Hämofiltration am INUS Tagesklinikum, Cham, Germany
| | - K Voit-Bak
- Zentrum für Apherese- und Hämofiltration am INUS Tagesklinikum, Cham, Germany
| | - J Graessler
- Department and Outpatient Department of Medicine III, University Hospital Carl Gustav Carus, Dresden, Germany
| | - U Julius
- Department and Outpatient Department of Medicine III, University Hospital Carl Gustav Carus, Dresden, Germany
| | - S Tselmin
- Department and Outpatient Department of Medicine III, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Roman N Rodionov
- Department and Outpatient Department of Medicine III, University Hospital Carl Gustav Carus, Dresden, Germany
| | - M Barbir
- Royal Brompton Hospital, London, UK
| | | | | | - M Mayr
- Kings College London, London, UK
- Technische Universität Dresden, Dresden, Germany
| | - S R Bornstein
- Kings College London, London, UK
- Department and Outpatient Department of Medicine III, University Hospital Carl Gustav Carus, Dresden, Germany
| |
Collapse
|
18
|
Pathak E, Mishra R. Deciphering the link between Diabetes mellitus and SARS-CoV-2 infection through differential targeting of microRNAs in the human pancreas. J Endocrinol Invest 2022; 45:537-550. [PMID: 34669152 PMCID: PMC8527307 DOI: 10.1007/s40618-021-01693-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 10/10/2021] [Indexed: 01/08/2023]
Abstract
PURPOSE Coronavirus Disease 2019 (COVID-19) severity and Diabetes mellitus affect each other bidirectionally. However, the cause of severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) infection on the incidence of diabetes is unclear. In the SARS-CoV-2-infected cells, host microRNAs (miRNAs) may target the native gene transcripts as well as the viral genomic and subgenomic RNAs. Here, we investigated the role of miRNAs in linking Diabetes to SARS-CoV-2 infection in the human pancreas. METHODS Differential gene expression and disease enrichment analyses were performed on an RNA-Seq dataset of human embryonic stem cell-derived (hESC) mock-infected and SARS-CoV-2-infected pancreatic organoids to obtain the dysregulated Diabetes-associated genes. The miRNA target prediction for the Diabetes-associated gene transcripts and the SARS-CoV-2 RNAs has been made to determine the common miRNAs targeting them. Minimum Free Energy (MFE) analysis was done to identify the miRNAs, preferably targeting SARS-CoV-2 RNAs over the Diabetes-associated gene transcripts. RESULTS The gene expression and disease enrichment analyses of the RNA-Seq data have revealed five biomarker genes, i.e., CP, SOCS3, AGT, PSMB8 and CFB that are associated with Diabetes and get significantly upregulated in the pancreas following SARS-CoV-2-infection. Four miRNAs, i.e., hsa-miR-298, hsa-miR-3925-5p, hsa-miR-4691-3p and hsa-miR-5196-5p, showed preferential targeting of the SARS-CoV-2 genome over the cell's Diabetes-associated messenger RNAs (mRNAs) in the human pancreas. CONCLUSION Our study proposes that the differential targeting of the Diabetes-associated host genes by the miRNAs may lead to diabetic complications or new-onset Diabetes that can worsen the condition of COVID-19 patients.
Collapse
Affiliation(s)
| | - R Mishra
- Bioinformatics, MMV, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
19
|
Shen Y, Li J, Tian H, Ji Y, Li Z, Lu J, Lu H, Liu B, Liu F. High level of complement factor Ba within first prenatal test of gestation increases the risk of subsequent gestational diabetes: a propensity score-matched study. Gynecol Endocrinol 2022; 38:158-163. [PMID: 34643127 DOI: 10.1080/09513590.2021.1989400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
OBJECTIVE This study was to assess the alteration of circulating complement factor Ba (CFBa) within 11 to 17 weeks of gestation and its association with subsequent gestational diabetes mellitus (GDM) and its delivery outcome. METHODS Biochemical parameters and blood samples were collected from 399 pregnant women within 11 to 17 weeks of gestation. At 24 to 28 weeks of gestation, all participants underwent 75-g oral glucose tolerance test and were assigned to GDM group (n = 80) and normal control group (n = 319). Perinatal data were collected after delivery. A propensity score-matched (PSM) analysis was performed to reduce the impact of confounding factors on glucose metabolism during pregnancy between the two groups. RESULTS Two groups of 74 well-matched patients who maintained balance in terms of baseline characteristics. The levels of CFBa in pregnant women who later developed GDM were significantly higher than those in healthy pregnant women [0.4(0.1-0.8) vs. 0.2(0.2-0.3), p = 0.024]. Logistic regression analysis results confirmed that the level of CFBa was an independent impact factor for the occurrence of GDM (OR = 1.57, 95% CI: 1.118-2.210, p = 0.009). Further grouping according to the median level of CFBa, it was found that the incidence of GDM in category two (>0.23 ng/ml, n = 74) was markedly higher than that in the first category (≤0.23 ng/ml, n = 74) (p = 0.021). CONCLUSIONS High level of the CFBa within 11 to 17 weeks of gestation increases the risk of subsequent GDM, and maybe a biomarker for predicting GDM.
Collapse
Affiliation(s)
- Ying Shen
- Department of Endocrinology & Metabolism, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, China
| | - Junxian Li
- Department of Endocrinology & Metabolism, Shanghai Jiao-Tong University School of Medicine Affiliated Shanghai General Hospital, Shanghai, China
| | - Hairong Tian
- Department of Endocrinology and Metabolism, Jin Shan Branch of Shanghai Sixth People's Hospital, Shanghai, China
| | - Ye Ji
- Department of Endocrinology and Metabolism, Jin Shan Branch of Shanghai Sixth People's Hospital, Shanghai, China
| | - Ziyun Li
- Department of Endocrinology and Metabolism, Jin Shan Branch of Shanghai Sixth People's Hospital, Shanghai, China
| | - Junxi Lu
- Department of Endocrinology & Metabolism, Shanghai Key Laboratory of Diabetes, Shanghai Clinical Medical Center of Diabetes, Shanghai Key Clinical Center of Metabolic Diseases, Shanghai Institute for Diabetes, Shanghai Jiao-Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Huijuan Lu
- Department of Endocrinology & Metabolism, Shanghai Key Laboratory of Diabetes, Shanghai Clinical Medical Center of Diabetes, Shanghai Key Clinical Center of Metabolic Diseases, Shanghai Institute for Diabetes, Shanghai Jiao-Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Bo Liu
- Department of Endocrinology and Metabolism, Jin Shan Branch of Shanghai Sixth People's Hospital, Shanghai, China
| | - Fang Liu
- Department of Endocrinology & Metabolism, Shanghai Jiao-Tong University School of Medicine Affiliated Shanghai General Hospital, Shanghai, China
- Department of Endocrinology & Metabolism, Shanghai Key Laboratory of Diabetes, Shanghai Clinical Medical Center of Diabetes, Shanghai Key Clinical Center of Metabolic Diseases, Shanghai Institute for Diabetes, Shanghai Jiao-Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
20
|
Lee MJ, Na K, Shin H, Kim CY, Cho JY, Kang CM, Kim SH, Kim H, Choi HJ, Lee CK, Bae S, Son S, Paik YK. Early Diagnostic Ability of Human Complement Factor B in Pancreatic Cancer Is Partly Linked to Its Potential Tumor-Promoting Role. J Proteome Res 2021; 20:5315-5328. [PMID: 34766501 DOI: 10.1021/acs.jproteome.1c00805] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Although plasma complement factor B (CFB, NX_P00751), both alone and in combination with CA19-9 (i.e., the ComB-CAN), previously exhibited a reliable diagnostic ability for pancreatic cancer (PC), its detectability of the early stages and the cancer detection mechanism remained elusive. We first evaluated the diagnostic accuracy of ComB-CAN using plasma samples from healthy donors (HDs), patients with chronic pancreatitis (CP), and patients with different PC stages (I/II vs III/IV). An analysis of the area under the curve (AUC) by PanelComposer using logistic regression revealed that ComB-CAN has a superior diagnostic ability for early-stage PC (97.1.% [95% confidence interval (CI): (97.1-97.2)]) compared with CFB (94.3% [95% CI: 94.2-94.4]) or CA19-9 alone (34.3% [95% CI: 34.1-34.4]). In the comparisons of all stages of patients with PC vs CP and HDs, the AUC values of ComB-CAN, CFB, and CA19-9 were 0.983 (95% CI: 0.983-0.983), 0.950 (95% CI: 0.950-0.951), and 0.873 (95% CI: 0.873-0.874), respectively. We then investigated the molecular mechanism underlying the detection of early-stage PC by using stable cell lines of CFB knockdown and CFB overexpression. A global transcriptomic analysis coupled to cell invasion assays of both CFB-modulated cell lines suggested that CFB plays a tumor-promoting role in PC, which likely initiates the PI3K-AKT cancer signaling pathway. Thus our study establishes ComB-CAN as a reliable early diagnostic marker for PC that can be clinically applied for early PC screening in the general public.
Collapse
Affiliation(s)
- Min Jung Lee
- Yonsei Proteome Research Center, Yonsei University, 50 Yonsei-ro, Seodaemoon-ku, Seoul 03722, South Korea
| | - Keun Na
- Yonsei Proteome Research Center, Yonsei University, 50 Yonsei-ro, Seodaemoon-ku, Seoul 03722, South Korea
| | - Heon Shin
- Yonsei Proteome Research Center, Yonsei University, 50 Yonsei-ro, Seodaemoon-ku, Seoul 03722, South Korea
| | - Chae-Yeon Kim
- Yonsei Proteome Research Center, Yonsei University, 50 Yonsei-ro, Seodaemoon-ku, Seoul 03722, South Korea
| | - Jin-Young Cho
- Yonsei Proteome Research Center, Yonsei University, 50 Yonsei-ro, Seodaemoon-ku, Seoul 03722, South Korea
| | | | | | | | | | | | - Sumi Bae
- JW Bioscience Corp., 2477, Nambusunhwan-ro, Seocho-gu, Seoul 06725, South Korea
| | - Sunghwa Son
- JW Holdings Corp., 2477, Nambusunhwan-ro, Seocho-gu, Seoul 06725, South Korea
| | - Young-Ki Paik
- Yonsei Proteome Research Center, Yonsei University, 50 Yonsei-ro, Seodaemoon-ku, Seoul 03722, South Korea
| |
Collapse
|
21
|
Hypoglycemia-induced changes in complement pathways in type 2 diabetes. ATHEROSCLEROSIS PLUS 2021; 46:35-45. [PMID: 36643727 PMCID: PMC9833243 DOI: 10.1016/j.athplu.2021.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/05/2021] [Accepted: 11/11/2021] [Indexed: 01/18/2023]
Abstract
Background and aims An association between hypoglycaemia and adverse cardiovascular events has been suggested from longitudinal and retrospective cohort studies. The complement pathway proteins in hypoglycemia are not well studied. Here, we hypothesized that these circulating proteins would be elevated in response to hypoglycemia in type 2 diabetes (T2D) through the inflammatory response. Methods A prospective, parallel study in T2D (n = 23) and controls (n = 23). Subjects underwent insulin-induced hypoglycemia with blood sampling at baseline, hypoglycemia and post-hypoglycemia; SOMAscan proteomic analysis of complement pathway-related proteins, cytokines and inflammatory proteins was undertaken. Results At baseline: Complement C2 (p < 0.05) and Factor B (p < 0.05) were elevated in T2D. At hypoglycemia: Complement C2 (p < 0.05) and Factor B (p < 0.01) remained elevated, whilst Factor I became elevated (p < 0.05) in T2D; Complement C4b became elevated in controls (p < 0.05). In the post-hypoglycemia follow up period, Complement C2, Factor B and Factor I remained elevated in T2D; in addition, Factor D, Factor H and mannose-binding protein C showed elevations in T2D, whilst properdin, complement C3b, Factor H-related protein 5, complement C1q and decay-accelerating factor (DAF) showed elevations in controls. Granger causality analysis showed that inflammatory proteins appeared to drive complement protein changes in T2D; conversely, in controls, complement proteins drove inflammatory protein changes. Conclusions Baseline elevations in C2 and Factor B indicate upregulation of the complement pathway in T2D. Changes in complement pathway-related protein levels in response to hypoglycemia suggest both intrinsic and alternative pathway activation at 2-h that appears driven by the underlying inflammation in T2D and could contribute to a cardiovascular event.ClinicalTrials.gov NCT03102801. Date of registration April 6, 2017, retrospectively registered. https://clinicaltrials.gov/ct2/show/NCT03102801?term=NCT03102801&draw=2&rank=1.
Collapse
|
22
|
Carruthers NJ, Strieder-Barboza C, Caruso JA, Flesher CG, Baker NA, Kerk SA, Ky A, Ehlers AP, Varban OA, Lyssiotis CA, Lumeng CN, Stemmer PM, O'Rourke RW. The human type 2 diabetes-specific visceral adipose tissue proteome and transcriptome in obesity. Sci Rep 2021; 11:17394. [PMID: 34462518 PMCID: PMC8405693 DOI: 10.1038/s41598-021-96995-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 08/11/2021] [Indexed: 01/21/2023] Open
Abstract
Dysfunctional visceral adipose tissue (VAT) in obesity is associated with type 2 diabetes (DM) but underlying mechanisms remain unclear. Our objective in this discovery analysis was to identify genes and proteins regulated by DM to elucidate aberrant cellular metabolic and signaling mediators. We performed label-free proteomics and RNA-sequencing analysis of VAT from female bariatric surgery subjects with DM and without DM (NDM). We quantified 1965 protein groups, 23 proteins, and 372 genes that were differently abundant in DM vs. NDM VAT. Proteins downregulated in DM were related to fatty acid synthesis and mitochondrial function (fatty acid synthase, FASN; dihydrolipoyl dehydrogenase, mitochondrial, E3 component, DLD; succinate dehydrogenase-α, SDHA) while proteins upregulated in DM were associated with innate immunity and transcriptional regulation (vitronectin, VTN; endothelial protein C receptor, EPCR; signal transducer and activator of transcription 5B, STAT5B). Transcriptome indicated defects in innate inflammation, lipid metabolism, and extracellular matrix (ECM) function, and components of complement classical and alternative cascades. The VAT proteome and transcriptome shared 13 biological processes impacted by DM, related to complement activation, cell proliferation and migration, ECM organization, lipid metabolism, and gluconeogenesis. Our data revealed a marked effect of DM in downregulating FASN. We also demonstrate enrichment of complement factor B (CFB), coagulation factor XIII A chain (F13A1), thrombospondin 1 (THBS1), and integrins at mRNA and protein levels, albeit with lower q-values and lack of Western blot or PCR confirmation. Our findings suggest putative mechanisms of VAT dysfunction in DM.
Collapse
Affiliation(s)
- Nicholas J Carruthers
- Proteomics Core Facility, Wayne State University, 42 W. Warren Ave, Detroit, MI, 48202, USA
| | - Clarissa Strieder-Barboza
- Department of Surgery, University of Michigan Medical School, 1301 Catherine St, Ann Arbor, MI, 48109, USA
| | - Joseph A Caruso
- Department of Chemistry, University of Michigan Medical School, 1301 Catherine St, Ann Arbor, MI, 48109, USA
| | - Carmen G Flesher
- Department of Surgery, University of Michigan Medical School, 1301 Catherine St, Ann Arbor, MI, 48109, USA
| | - Nicki A Baker
- Department of Surgery, University of Michigan Medical School, 1301 Catherine St, Ann Arbor, MI, 48109, USA
| | - Samuel A Kerk
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, 1301 Catherine St, Ann Arbor, MI, 48109, USA
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan Medical School, 1301 Catherine St, Ann Arbor, MI, 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, 1301 Catherine St, Ann Arbor, MI, 48109, USA
| | - Alexander Ky
- Department of Surgery, University of Michigan Medical School, 1301 Catherine St, Ann Arbor, MI, 48109, USA
| | - Anne P Ehlers
- Department of Surgery, University of Michigan Medical School, 1301 Catherine St, Ann Arbor, MI, 48109, USA
- Department of Surgery, Veterans Affairs Ann Arbor Healthcare System, 2215 Fuller Rd, Ann Arbor, MI, 48105, USA
| | - Oliver A Varban
- Department of Surgery, University of Michigan Medical School, 1301 Catherine St, Ann Arbor, MI, 48109, USA
| | - Costas A Lyssiotis
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, 1301 Catherine St, Ann Arbor, MI, 48109, USA
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan Medical School, 1301 Catherine St, Ann Arbor, MI, 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, 1301 Catherine St, Ann Arbor, MI, 48109, USA
| | - Carey N Lumeng
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, 1301 Catherine St, Ann Arbor, MI, 48109, USA
- Graduate Program in Immunology, University of Michigan Medical School, 1301 Catherine St, Ann Arbor, MI, 48109, USA
- Graduate Program in Cellular and Molecular Biology, University of Michigan Medical School, 1301 Catherine St, Ann Arbor, MI, 48109, USA
| | - Paul M Stemmer
- Proteomics Core Facility, Wayne State University, 42 W. Warren Ave, Detroit, MI, 48202, USA
| | - Robert W O'Rourke
- Department of Surgery, University of Michigan Medical School, 1301 Catherine St, Ann Arbor, MI, 48109, USA.
- Department of Surgery, Veterans Affairs Ann Arbor Healthcare System, 2215 Fuller Rd, Ann Arbor, MI, 48105, USA.
- Section of General Surgery, Department of Surgery, University of Michigan, 2210 Taubman Center-5343, 1500 E. Medical Center Drive, Ann Arbor, MI, 48109-5343, USA.
| |
Collapse
|
23
|
Plasma protein expression profiles, cardiovascular disease, and religious struggles among South Asians in the MASALA study. Sci Rep 2021; 11:961. [PMID: 33441605 PMCID: PMC7806901 DOI: 10.1038/s41598-020-79429-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 12/07/2020] [Indexed: 11/12/2022] Open
Abstract
Blood protein concentrations are clinically useful, predictive biomarkers of cardiovascular disease (CVD). Despite a higher burden of CVD among U.S. South Asians, no CVD-related proteomics study has been conducted in this sub-population. The aim of this study is to investigate the associations between plasma protein levels and CVD incidence, and to assess the potential influence of religiosity/spirituality (R/S) on significant protein-CVD associations, in South Asians from the MASALA Study. We used a nested case–control design of 50 participants with incident CVD and 50 sex- and age-matched controls. Plasma samples were analyzed by SOMAscan for expression of 1305 proteins. Multivariable logistic regression models and model selection using Akaike Information Criteria were performed on the proteins and clinical covariates, with further effect modification analyses conducted to assess the influence of R/S measures on significant associations between proteins and incident CVD events. We identified 36 proteins that were significantly expressed differentially among CVD cases compared to matched controls. These proteins are involved in immune cell recruitment, atherosclerosis, endothelial cell differentiation, and vascularization. A final multivariable model found three proteins (Contactin-5 [CNTN5], Low affinity immunoglobulin gamma Fc region receptor II-a [FCGR2A], and Complement factor B [CFB]) associated with incident CVD after adjustment for diabetes (AUC = 0.82). Religious struggles that exacerbate the adverse impact of stressful life events, significantly modified the effect of Contactin-5 and Complement factor B on risk of CVD. Our research is this first assessment of the relationship between protein concentrations and risk of CVD in a South Asian sample. Further research is needed to understand patterns of proteomic profiles across diverse ethnic communities, and the influence of resources for resiliency on proteomic signatures and ultimately, risk of CVD.
Collapse
|
24
|
Xu B, Wang L, Zhan H, Zhao L, Wang Y, Shen M, Xu K, Li L, Luo X, Zhou S, Tang A, Liu G, Song L, Li Y. Investigation of the Mechanism of Complement System in Diabetic Nephropathy via Bioinformatics Analysis. J Diabetes Res 2021; 2021:5546199. [PMID: 34124269 PMCID: PMC8169258 DOI: 10.1155/2021/5546199] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/02/2021] [Accepted: 05/05/2021] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVES Diabetic nephropathy (DN) is a major cause of end-stage renal disease (ESRD) throughout the world, and the identification of novel biomarkers via bioinformatics analysis could provide research foundation for future experimental verification and large-group cohort in DN models and patients. METHODS GSE30528, GSE47183, and GSE104948 were downloaded from Gene Expression Omnibus (GEO) database to find differentially expressed genes (DEGs). The difference of gene expression between normal renal tissues and DN renal tissues was firstly screened by GEO2R. Then, the protein-protein interactions (PPIs) of DEGs were performed by STRING database, the result was integrated and visualized via applying Cytoscape software, and the hub genes in this PPI network were selected by MCODE and topological analysis. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were carried out to determine the molecular mechanisms of DEGs involved in the progression of DN. Finally, the Nephroseq v5 online platform was used to explore the correlation between hub genes and clinical features of DN. RESULTS There were 64 DEGs, and 32 hub genes were identified, enriched pathways of hub genes involved in several functions and expression pathways, such as complement binding, extracellular matrix structural constituent, complement cascade related pathways, and ECM proteoglycans. The correlation analysis and subgroup analysis of 7 complement cascade-related hub genes and the clinical characteristics of DN showed that C1QA, C1QB, C3, CFB, ITGB2, VSIG4, and CLU may participate in the development of DN. CONCLUSIONS We confirmed that the complement cascade-related hub genes may be the novel biomarkers for DN early diagnosis and targeted treatment.
Collapse
Affiliation(s)
- Bojun Xu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072 Sichuan, China
| | - Lei Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Huakui Zhan
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072 Sichuan, China
| | - Liangbin Zhao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072 Sichuan, China
| | - Yuehan Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072 Sichuan, China
| | - Meng Shen
- Chengdu Seventh People's Hospital, Chengdu, 610213 Sichuan, China
| | - Keyang Xu
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong
| | - Li Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072 Sichuan, China
| | - Xu Luo
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning 530023, China
| | - Shasha Zhou
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072 Sichuan, China
| | - Anqi Tang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072 Sichuan, China
| | - Gang Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072 Sichuan, China
| | - Lu Song
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072 Sichuan, China
| | - Yan Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072 Sichuan, China
| |
Collapse
|
25
|
Rasal KD, Iquebal MA, Dixit S, Vasam M, Raza M, Sahoo L, Jaiswal S, Nandi S, Mahapatra KD, Rasal A, Udit UK, Meher PK, Murmu K, Angadi UB, Rai A, Kumar D, Sundaray JK. Revealing Alteration in the Hepatic Glucose Metabolism of Genetically Improved Carp, Jayanti Rohu Labeo rohita Fed a High Carbohydrate Diet Using Transcriptome Sequencing. Int J Mol Sci 2020; 21:E8180. [PMID: 33142948 PMCID: PMC7662834 DOI: 10.3390/ijms21218180] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 01/25/2023] Open
Abstract
Although feed cost is the greatest concern in aquaculture, the inclusion of carbohydrates in the fish diet, and their assimilation, are still not well understood in aquaculture species. We identified molecular events that occur due to the inclusion of high carbohydrate levels in the diets of genetically improved 'Jayanti rohu' Labeo rohita. To reveal transcriptional changes in the liver of rohu, a feeding experiment was conducted with three doses of gelatinized starch (20% (control), 40%, and 60%). Transcriptome sequencing revealed totals of 15,232 (4464 up- and 4343 down-regulated) and 15,360 (4478 up- and 4171 down-regulated) differentially expressed genes. Up-regulated transcripts associated with glucose metabolisms, such as hexokinase, PHK, glycogen synthase and PGK, were found in fish fed diets with high starch levels. Interestingly, a de novo lipogenesis mechanism was found to be enriched in the livers of treated fish due to up-regulated transcripts such as FAS, ACCα, and PPARγ. The insulin signaling pathways with enriched PPAR and mTOR were identified by Kyoto Encyclopedia of Genes and Genome (KEGG) as a result of high carbohydrates. This work revealed for the first time the atypical regulation transcripts associated with glucose metabolism and lipogenesis in the livers of Jayanti rohu due to the inclusion of high carbohydrate levels in the diet. This study also encourages the exploration of early nutritional programming for enhancing glucose efficiency in carp species, for sustainable and cost-effective aquaculture production.
Collapse
Affiliation(s)
- Kiran D. Rasal
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Mir Asif Iquebal
- Centre for Agricultural Bioinformatics (CABin), ICAR-Indian Agricultural Statistics Research Institute, Library Avenue, PUSA, New Delhi 110012, India; (M.A.I.); (M.R.); (S.J.); (U.A.); (A.R.); (D.K.)
| | - Sangita Dixit
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Manohar Vasam
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Mustafa Raza
- Centre for Agricultural Bioinformatics (CABin), ICAR-Indian Agricultural Statistics Research Institute, Library Avenue, PUSA, New Delhi 110012, India; (M.A.I.); (M.R.); (S.J.); (U.A.); (A.R.); (D.K.)
| | - Lakshman Sahoo
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Sarika Jaiswal
- Centre for Agricultural Bioinformatics (CABin), ICAR-Indian Agricultural Statistics Research Institute, Library Avenue, PUSA, New Delhi 110012, India; (M.A.I.); (M.R.); (S.J.); (U.A.); (A.R.); (D.K.)
| | - Samiran Nandi
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Kanta Das Mahapatra
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Avinash Rasal
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Uday Kumar Udit
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Prem Kumar Meher
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Khuntia Murmu
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - UB Angadi
- Centre for Agricultural Bioinformatics (CABin), ICAR-Indian Agricultural Statistics Research Institute, Library Avenue, PUSA, New Delhi 110012, India; (M.A.I.); (M.R.); (S.J.); (U.A.); (A.R.); (D.K.)
| | - Anil Rai
- Centre for Agricultural Bioinformatics (CABin), ICAR-Indian Agricultural Statistics Research Institute, Library Avenue, PUSA, New Delhi 110012, India; (M.A.I.); (M.R.); (S.J.); (U.A.); (A.R.); (D.K.)
| | - Dinesh Kumar
- Centre for Agricultural Bioinformatics (CABin), ICAR-Indian Agricultural Statistics Research Institute, Library Avenue, PUSA, New Delhi 110012, India; (M.A.I.); (M.R.); (S.J.); (U.A.); (A.R.); (D.K.)
| | - Jitendra Kumar Sundaray
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| |
Collapse
|
26
|
Szpirer C. Rat models of human diseases and related phenotypes: a systematic inventory of the causative genes. J Biomed Sci 2020; 27:84. [PMID: 32741357 PMCID: PMC7395987 DOI: 10.1186/s12929-020-00673-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/09/2020] [Indexed: 12/13/2022] Open
Abstract
The laboratory rat has been used for a long time as the model of choice in several biomedical disciplines. Numerous inbred strains have been isolated, displaying a wide range of phenotypes and providing many models of human traits and diseases. Rat genome mapping and genomics was considerably developed in the last decades. The availability of these resources has stimulated numerous studies aimed at discovering causal disease genes by positional identification. Numerous rat genes have now been identified that underlie monogenic or complex diseases and remarkably, these results have been translated to the human in a significant proportion of cases, leading to the identification of novel human disease susceptibility genes, helping in studying the mechanisms underlying the pathological abnormalities and also suggesting new therapeutic approaches. In addition, reverse genetic tools have been developed. Several genome-editing methods were introduced to generate targeted mutations in genes the function of which could be clarified in this manner [generally these are knockout mutations]. Furthermore, even when the human gene causing a disease had been identified without resorting to a rat model, mutated rat strains (in particular KO strains) were created to analyze the gene function and the disease pathogenesis. Today, over 350 rat genes have been identified as underlying diseases or playing a key role in critical biological processes that are altered in diseases, thereby providing a rich resource of disease models. This article is an update of the progress made in this research and provides the reader with an inventory of these disease genes, a significant number of which have similar effects in rat and humans.
Collapse
Affiliation(s)
- Claude Szpirer
- Université Libre de Bruxelles, B-6041, Gosselies, Belgium.
- , Waterloo, Belgium.
| |
Collapse
|
27
|
Yang Y, Ma L, Song M, Li X, He F, Wang C, Chen M, Zhou J, Mei C. The role of the complement factor B-arginase-polyamine molecular axis in uremia-induced cardiac remodeling in mice. Eur J Immunol 2019; 50:220-233. [PMID: 31777959 DOI: 10.1002/eji.201948227] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 08/27/2019] [Accepted: 11/25/2019] [Indexed: 01/19/2023]
Abstract
The role of complement system in heart diseases is controversial. Besides, the mechanisms by which complement components participate in cardiac remodeling (CR) and heart failure during uremia are unclear. In this study, 5/6 nephrectomy was performed to adult mice to establish the uremic model and CR deteriorated over the course of uremia. Although complement pathways were not further activated over the course of the disease, soluble complement factor B (CFB) was upregulated at post-nephrectomy day 90 (PNx90) compared with PNx30. Further, CFB notably deteriorated CR in uremic mice but this effect was reversed by depletion of macrophages with liposomal clodronate. In vivo and in vitro CFB upregulated arginase 1 (ARG1) expression, increased ARG1 enzymatic activity, and stimulated the syntheses of ornithine, leading to polyamine overproduction in macrophages. Putrescine, an important polyamine, promoted cardiac fibroblast proliferation and collagen production, resulting in progressive CR. In vivo the inhibition of ARG1 activity with Nω -hydroxyl-l-arginine remarkably improved the general survival rates, inhibited the infiltration of cardiac fibroblasts, and alleviated progression of CR in uremic mice. Taken together, the CFB-ARG1-putrescine axis is related to progression of CR and ARG1 hyperactivity in macrophages may provide a novel therapeutic target against the heart injury in uremia.
Collapse
Affiliation(s)
- Yang Yang
- Kidney Therapeutic Center of the Chinese People's Liberation Army, Beidaihe Rehabilitation and Recuperation Center of the Chinese People's Liberation Army, Qinhuangdao, China
| | - Lu Ma
- Kidney Therapeutic Center of the Chinese People's Liberation Army, Beidaihe Rehabilitation and Recuperation Center of the Chinese People's Liberation Army, Qinhuangdao, China
| | - Minghui Song
- Kidney Therapeutic Center of the Chinese People's Liberation Army, Beidaihe Rehabilitation and Recuperation Center of the Chinese People's Liberation Army, Qinhuangdao, China
| | - Xiaomeng Li
- Ultrasonic Department, Beidaihe Rehabilitation and Recuperation Center of the Chinese People's Liberation Army, Qinhuangdao, China
| | - Fagui He
- Kidney Therapeutic Center of the Chinese People's Liberation Army, Beidaihe Rehabilitation and Recuperation Center of the Chinese People's Liberation Army, Qinhuangdao, China
| | - Chao Wang
- Kidney Therapeutic Center of the Chinese People's Liberation Army, Beidaihe Rehabilitation and Recuperation Center of the Chinese People's Liberation Army, Qinhuangdao, China
| | - Meihan Chen
- Kidney Institution of the Chinese People's Liberation Army, Chang Zheng Hospital, the Second Military Medical University, Shanghai, China
| | - Jie Zhou
- Kidney Institution of the Chinese People's Liberation Army, Chang Zheng Hospital, the Second Military Medical University, Shanghai, China
| | - Changlin Mei
- Kidney Institution of the Chinese People's Liberation Army, Chang Zheng Hospital, the Second Military Medical University, Shanghai, China
| |
Collapse
|
28
|
Cruz Junho CV, Trentin-Sonoda M, Alvim JM, Gaisler-Silva F, Carneiro-Ramos MS. Ca2+/Calmodulin-dependent kinase II delta B is essential for cardiomyocyte hypertrophy and complement gene expression after LPS and HSP60 stimulation in vitro. ACTA ACUST UNITED AC 2019; 52:e8732. [PMID: 31314855 PMCID: PMC6644523 DOI: 10.1590/1414-431x20198732] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 05/21/2019] [Indexed: 12/23/2022]
Abstract
Inflammation plays an important role in the development of cardiovascular diseases (CVDs), suggesting that the immune system is a target of therapeutic interventions used for treating CVDs. This study evaluated mechanisms underlying inflammatory response and cardiomyocyte hypertrophy associated with bacterial lipopolysaccharide (LPS)- or heat shock protein 60 (HSP60)-induced Toll-like receptor (TLR) stimulation and the effect of a small interfering RNA (siRNA) against Ca2+/calmodulin-dependent kinase II delta B (CaMKIIδB) on these outcomes. Our results showed that treatment with HSP60 or LPS (TLR agonists) induced cardiomyocyte hypertrophy and complement system C3 and factor B gene expression. In vitro silencing of CaMKIIδB prevented complement gene transcription and cardiomyocyte hypertrophy associated with TLR 2/4 activation but did not prevent the increase in interleukin-6 and tumor necrosis factor-alfa gene expression in primary cultured cardiomyocytes. Moreover, CaMKIIδB silencing attenuated nuclear factor-kappa B expression. These findings supported the hypothesis that CaMKIIδB acts as a link between inflammation and cardiac hypertrophy. Furthermore, the present study is the first to show that extracellular HSP60 activated complement gene expression through CaMKIIδB. Our results indicated that a stress stimulus induced by LPS or HSP60 treatment promoted cardiomyocyte hypertrophy and initiated an inflammatory response through the complement system. However, CaMKIIδB silencing prevented the cardiomyocyte hypertrophy independent of inflammatory response induced by LPS or HSP60 treatment.
Collapse
Affiliation(s)
- C V Cruz Junho
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Santo André, SP, Brasil
| | - M Trentin-Sonoda
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Santo André, SP, Brasil.,Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - J M Alvim
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Santo André, SP, Brasil.,Laboratorio de Genética e Cardiologia Molecular, Instituto do Coração, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - F Gaisler-Silva
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Santo André, SP, Brasil
| | - M S Carneiro-Ramos
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Santo André, SP, Brasil
| |
Collapse
|
29
|
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
|
30
|
Lim HK, Sull JW, Park BS, Mun JY, Hong MH, Lee Y, Hwang MJ, Lee MN, Lee JY, Kim IS. Relationship of Metabolic Diseases with Physical Activity Depending on Age. KOREAN JOURNAL OF CLINICAL LABORATORY SCIENCE 2018. [DOI: 10.15324/kjcls.2018.50.2.144] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Hyo Kyung Lim
- Department of Biomedical Laboratory Science, School of Medicine, Eulji University, Daejeon, Korea
| | - Jae Woong Sull
- Department of Biomedical Laboratory Science, College of Health Science, Eulji University, Seongnam, Korea
- Department of Senior Healthcare, BK21 plus program, Graduate School, Eulji University, Seongnam, Korea
| | - Beom Seok Park
- Department of Biomedical Laboratory Science, College of Health Science, Eulji University, Seongnam, Korea
- Department of Senior Healthcare, BK21 plus program, Graduate School, Eulji University, Seongnam, Korea
| | - Ji Young Mun
- Department of Biomedical Laboratory Science, College of Health Science, Eulji University, Seongnam, Korea
- Department of Senior Healthcare, BK21 plus program, Graduate School, Eulji University, Seongnam, Korea
| | - Min Hwa Hong
- Department of Senior Healthcare, BK21 plus program, Graduate School, Eulji University, Daejeon, Korea
| | - Yoori Lee
- Department of Senior Healthcare, BK21 plus program, Graduate School, Eulji University, Daejeon, Korea
| | - Min Ji Hwang
- Department of Biomedical Laboratory Science, School of Medicine, Eulji University, Daejeon, Korea
| | - Mi Na Lee
- Department of Biomedical Laboratory Science, School of Medicine, Eulji University, Daejeon, Korea
| | - Ji Young Lee
- Department of Biomedical Laboratory Science, School of Medicine, Eulji University, Daejeon, Korea
| | - In Sik Kim
- Department of Biomedical Laboratory Science, School of Medicine, Eulji University, Daejeon, Korea
- Department of Senior Healthcare, BK21 plus program, Graduate School, Eulji University, Daejeon, Korea
| |
Collapse
|
31
|
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
|