1
|
Eşkin Tanrıverdi MD, Kaya Sezginer E, Erol Koç EM, Moraloğlu Tekin Ö. Evaluation of serum and peritoneal fluid mannose-binding lectin associated serine protease-3, adipsin, properdin, and complement factor-H levels in endometriosis patients. Int J Gynaecol Obstet 2025. [PMID: 39907303 DOI: 10.1002/ijgo.16195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 01/12/2025] [Accepted: 01/20/2025] [Indexed: 02/06/2025]
Abstract
OBJECTIVE Endometriosis is a chronic disease which has been reported to be associated with distorted immune mechanisms. The alternative pathway is a complement system which plays a role in immune defense. The present study aimed to evaluate whether the level of alternative complement molecules differ in women with endometriosis compared to heathy individuals. METHODS A total of 58 women participated in this prospective research. Women with a diagnosis of endometriosis confirmed by laparoscopy (n = 32) were compared to healthy women (n = 26) in terms of serum adipsin, properdin, mannose-binding lectin-associated serine protease-3 and complement factor-H (CFH) levels. The peritoneal fluid samples which were taken during the endometriosis surgery were also analyzed in terms of the complement levels. The clinical and demographic data including the serum CA-125 level and pelvic pain were also analyzed. SPSS version 23.0 was used in statistical analysis. RESULTS The serum levels of adipsin and CFH were found to be significantly increased in women with endometriosis (P = 0.027 and P = 0.040, respectively). Serum adipsin level was found to significantly correlate with serum CA-125 level (r = 0.320, P = 0.015), serum CFH level (r = 0.705, P < 0.001), and degree of the pelvic pain complaint (r = 0.326, P = 0.013). A strong, positive correlation was also observed between peritoneal fluid levels of adipsin, and CFH (r = 0.593; P < 0.001). CONCLUSION To the best of our knowledge, the present study is the first to evaluate the alternative complement system in women with endometriosis. The current findings may be noteworthy to elucidate the possible role of the key molecules of the alternative pathway in endometriosis pathogenesis.
Collapse
Affiliation(s)
| | - Ecem Kaya Sezginer
- Department of Biochemistry, Ankara University Faculty of Pharmacy, Ankara, Turkey
| | - Esin Merve Erol Koç
- Department of Gynecology and Obstetrics, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Özlem Moraloğlu Tekin
- Department of Gynecology and Obstetrics, Ankara Bilkent City Hospital, Ankara, Turkey
| |
Collapse
|
2
|
Sato S, Miwa T, Gullipalli D, Golla M, Mohammadyari E, Zhou L, Palmer M, Song WC. Improved therapeutic efficacy of a bifunctional anti-C5 mAb-FH SCR1-5 fusion protein over anti-C5 mAb in an accelerated mouse model of C3 glomerulopathy. Immunohorizons 2025; 9:vlae006. [PMID: 39865974 PMCID: PMC11841979 DOI: 10.1093/immhor/vlae006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 10/23/2024] [Indexed: 01/28/2025] Open
Abstract
C3 glomerulopathy (C3G), a rare kidney disease caused by dysregulation of alternative pathway complement activation, is characterized by glomerular C3 deposition, proteinuria, crescentic glomerulonephritis, and renal failure. The anti-C5 monoclonal antibody (mAb) drug eculizumab has shown therapeutic effects in some but not all patients with C3G, and no approved therapy is currently available. Here, we developed and used a triple transgenic mouse model of fast progressing lethal C3G (FHm/mP-/-hFDKI/KI) to compare the therapeutic efficacy of a bifunctional anti-C5 mAb fused to a functional factor H (FH) fragment (short consensus repeat 1-5 [SCR1-5]) and the anti-C5 mAb itself. The new C3G mouse model is derived by humanizing factor D (hFDKI/KI) in a previously described FHm/mP-/- mouse that developed lethal C3G. We tested the effectiveness of these 2 complement inhibitors in triple transgenic mice with established C3G and glomerular disease. No FHm/mP-/-hFDKI/KI mice treated with vehicle survived the 30-d study period. All FHm/mP-/-hFDKI/KI mice treated with the C5 mAb-FH SCR1-5 fusion protein and 50% of mice treated with the anti-C5 mAb survived the 30-d treatment period. Moreover, mice treated with the C5 mAb-FH SCR1-5 fusion protein, but not those treated with the anti-C5 mAb, showed restored plasma alternative pathway complement control. The C5 mAb-FH SCR1-5 fusion protein reversed glomerular disease to a greater degree than the anti-C5 mAb. These data suggest that simultaneously inhibiting the terminal and proximal complement pathways, by anti-C5 mAb and FH SCR1-5, respectively, can reverse established C3G and is more efficacious than inhibiting the terminal pathway alone. A similar approach may be effective in treating human C3G.
Collapse
MESH Headings
- Animals
- Disease Models, Animal
- Mice
- Complement C5/immunology
- Complement C5/antagonists & inhibitors
- Mice, Transgenic
- Complement C3/metabolism
- Complement C3/immunology
- Humans
- Recombinant Fusion Proteins/therapeutic use
- Recombinant Fusion Proteins/pharmacology
- Recombinant Fusion Proteins/genetics
- Antibodies, Monoclonal
- Complement Factor H/genetics
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/pharmacology
- Glomerulonephritis/drug therapy
- Glomerulonephritis/immunology
- Glomerulonephritis, Membranoproliferative/drug therapy
- Glomerulonephritis, Membranoproliferative/immunology
- Complement Pathway, Alternative/drug effects
Collapse
Affiliation(s)
- Sayaka Sato
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Takashi Miwa
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Damodar Gullipalli
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Madhu Golla
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Eshagh Mohammadyari
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Lin Zhou
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Matthew Palmer
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Wen-Chao Song
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
3
|
Homan EA, Gilani A, Rubio-Navarro A, Johnson MA, Schaepkens OM, Cortada E, Pereira de Lima R, Stoll L, Lo JC. Complement 3a receptor 1 on macrophages and Kupffer cells is not required for the pathogenesis of metabolic dysfunction-associated steatotic liver disease. eLife 2025; 13:RP100708. [PMID: 39773465 PMCID: PMC11709426 DOI: 10.7554/elife.100708] [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] [Indexed: 01/30/2025] Open
Abstract
Together with obesity and type 2 diabetes, metabolic dysfunction-associated steatotic liver disease (MASLD) is a growing global epidemic. Activation of the complement system and infiltration of macrophages has been linked to progression of metabolic liver disease. The role of complement receptors in macrophage activation and recruitment in MASLD remains poorly understood. In human and mouse, C3AR1 in the liver is expressed primarily in Kupffer cells, but is downregulated in humans with MASLD compared to obese controls. To test the role of complement 3a receptor (C3aR1) on macrophages and liver resident macrophages in MASLD, we generated mice deficient in C3aR1 on all macrophages (C3aR1-MφKO) or specifically in liver Kupffer cells (C3aR1-KpKO) and subjected them to a model of metabolic steatotic liver disease. We show that macrophages account for the vast majority of C3ar1 expression in the liver. Overall, C3aR1-MφKO and C3aR1-KpKO mice have similar body weight gain without significant alterations in glucose homeostasis, hepatic steatosis and fibrosis, compared to controls on a MASLD-inducing diet. This study demonstrates that C3aR1 deletion in macrophages or Kupffer cells, the predominant liver cell type expressing C3ar1, has no significant effect on liver steatosis, inflammation or fibrosis in a dietary MASLD model.
Collapse
Affiliation(s)
- Edwin A Homan
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| | - Ankit Gilani
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| | - Alfonso Rubio-Navarro
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| | - Maya A Johnson
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| | - Odin M Schaepkens
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| | - Eric Cortada
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| | - Renan Pereira de Lima
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| | - Lisa Stoll
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| | - James C Lo
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| |
Collapse
|
4
|
Zhang X, Jiang M, Zhang X, Zuo Y, Zhang H, Zhang T, Yang L, Lin J, Zhang Y, Dai X, Ge W, Sun C, Yang F, Zhang J, Liu Y, Wang Y, Qiang H, Yang X, Sun D. Adipsin improves diabetic hindlimb ischemia through SERPINE1 dependent angiogenesis. Cardiovasc Diabetol 2024; 23:429. [PMID: 39623437 PMCID: PMC11613494 DOI: 10.1186/s12933-024-02526-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 11/25/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Adipsin (complement factor D, CFD), as the first described adipokine, is well-known for its regulatory effects in diabetic cardiovascular complications. However, its role in diabetic hind-limb ischemia was not clarified. This study aimed to evaluate the possible therapeutic effect of Adipsin in hind-limb ischemia in type 2 diabetic mice and to elucidate the molecular mechanisms involved. METHODS A high-fat diet and streptozotocin (HFD/STZ)-induced diabetic mouse model, and a transgenic mouse model with adipose tissue-specific overexpression of Adipsin (Adipsin-Tg) were employed. Hindlimb ischemia was established by femoral artery ligation, and blood flow recovery was monitored using Laser Doppler perfusion imaging. Molecular mechanisms underlying Adipsin-potentiated angiogenesis were examined using RNA sequencing and co-immunoprecipitation/mass spectrometry (Co-IP/MS) analyses. RESULTS Adipsin expression was upregulated in non-diabetic mice following HLI, while suppressed in diabetic mice, indicating its potential role in ischemic recovery which is impaired in diabetes. Adipsin-Tg mice exhibited significantly improved blood flow recovery, increased capillary density, and enhanced muscle regeneration in comparison with non-transgenic (NTg) diabetic mice. Adipsin facilitated proliferation, migration, and tube formation of human umbilical vein endothelial cells (HUVECs) under hyperglycemic and hypoxic conditions. Additionally, it enhanced phosphorylation of AKT, ERK, and eNOS pathways both in vivo and in vitro. RNA sequencing and co-immunoprecipitation/mass spectrometry (Co-IP/MS) analyses identified that Adipsin promoted angiogenesis by interacting with SERBP1, which disrupted the binding of SERBP1 to SERPINE1 mRNA, resulting in reduced SERPINE1 expression and the subsequent activation of the VEGFR2 signaling cascade. CONCLUSIONS Adipsin promotes angiogenesis and facilitates blood perfusion recovery in diabetic mice with HLI by downregulating SERPINE1 through interaction with SERBP1. These findings elucidate a novel therapeutic potential for Adipsin in the management of PAD in diabetic patients, highlighting its role in enhancing angiogenesis and tissue repair.
Collapse
MESH Headings
- Animals
- Ischemia/physiopathology
- Ischemia/metabolism
- Ischemia/genetics
- Hindlimb
- Neovascularization, Physiologic
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/complications
- Humans
- Male
- Mice, Inbred C57BL
- Regional Blood Flow
- Human Umbilical Vein Endothelial Cells/metabolism
- Complement Factor D/metabolism
- Complement Factor D/genetics
- Muscle, Skeletal/blood supply
- Muscle, Skeletal/metabolism
- Signal Transduction
- Mice, Transgenic
- Diabetic Angiopathies/metabolism
- Diabetic Angiopathies/physiopathology
- Diabetic Angiopathies/genetics
- Diabetic Angiopathies/etiology
- Cell Proliferation
- Recovery of Function
- Mice
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/genetics
- Cell Movement
- Angiogenesis Inducing Agents/pharmacology
- Cells, Cultured
- Microvascular Density
- Angiogenesis
Collapse
Affiliation(s)
- Xiaohua Zhang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Mengyuan Jiang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Xuebin Zhang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Yixuan Zuo
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Huanle Zhang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Tingting Zhang
- Xijing 986 Hospital Department, Air Force Medical University, Xi'an, China
| | - Liyu Yang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Jie Lin
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Yan Zhang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Xinchun Dai
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Wen Ge
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Chuang Sun
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Fang Yang
- Basic Medical Teaching Experiment Center, Basic Medical College, Air Force Medical University, Xi'an, China
| | - Jiye Zhang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Yue Liu
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Yangyang Wang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Huanhuan Qiang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Xiaojie Yang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Dongdong Sun
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China.
| |
Collapse
|
5
|
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
|
6
|
Oakes A, Liu Y, Dubielecka PM. Complement or insult: the emerging link between complement cascade deficiencies and pathology of myeloid malignancies. J Leukoc Biol 2024; 116:966-984. [PMID: 38836653 PMCID: PMC11531810 DOI: 10.1093/jleuko/qiae130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/01/2024] [Accepted: 05/03/2024] [Indexed: 06/06/2024] Open
Abstract
The complement cascade is an ancient and highly conserved arm of the immune system. The accumulating evidence highlights elevated activity of the complement cascade in cancer microenvironment and emphasizes its effects on the immune, cancer, and cancer stroma cells, pointing to a role in inflammation-mediated etiology of neoplasms. The role the cascade plays in development, progression, and relapse of solid tumors is increasingly recognized, however its role in hematological malignancies, especially those of myeloid origin, has not been thoroughly assessed and remains obscure. As the role of inflammation and autoimmunity in development of myeloid malignancies is becoming recognized, in this review we focus on summarizing the links that have been identified so far for complement cascade involvement in the pathobiology of myeloid malignancies. Complement deficiencies are primary immunodeficiencies that cause an array of clinical outcomes including an increased risk of a range of infectious as well as local or systemic inflammatory and thrombotic conditions. Here, we discuss the impact that deficiencies in complement cascade initiators, mid- and terminal-components and inhibitors have on the biology of myeloid neoplasms. The emergent conclusions indicate that the links between complement cascade, inflammatory signaling, and the homeostasis of hematopoietic system exist, and efforts should continue to detail the mechanistic involvement of complement cascade in the development and progression of myeloid cancers.
Collapse
Affiliation(s)
- Alissa Oakes
- Department of Medicine, Alpert Medical School, Brown University, 69 Brown St, Providence, RI 02906, USA
- Division of Hematology/Oncology, Rhode Island Hospital, 69 Brown St, Providence, RI 02906, USA
- Therapeutic Sciences Graduate program, Brown University, 69 Brown St, Providence, RI 02906, USA
| | - Yuchen Liu
- Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, 22. S. Greene St., Baltimore, MD 21201-1595, USA
| | - Patrycja M Dubielecka
- Department of Medicine, Alpert Medical School, Brown University, 69 Brown St, Providence, RI 02906, USA
- Division of Hematology/Oncology, Rhode Island Hospital, 69 Brown St, Providence, RI 02906, USA
- Therapeutic Sciences Graduate program, Brown University, 69 Brown St, Providence, RI 02906, USA
- Legorreta Cancer Center, Brown University, One Hoppin St., Coro West, Suite 5.01, Providence, RI 02903, USA
| |
Collapse
|
7
|
Homan EA, Gilani A, Rubio-Navarro A, Johnson MA, Schaepkens OM, Cortada E, de Lima RP, Stoll L, Lo JC. Complement 3a Receptor 1 on Macrophages and Kupffer cells is not required for the Pathogenesis of Metabolic Dysfunction-Associated Steatotic Liver Disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.26.24309550. [PMID: 38978661 PMCID: PMC11230319 DOI: 10.1101/2024.06.26.24309550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Together with obesity and type 2 diabetes, metabolic dysfunction-associated steatotic liver disease (MASLD) is a growing global epidemic. Activation of the complement system and infiltration of macrophages has been linked to progression of metabolic liver disease. The role of complement receptors in macrophage activation and recruitment in MASLD remains poorly understood. In human and mouse, C3AR1 in the liver is expressed primarily in Kupffer cells, but is downregulated in humans with MASLD compared to obese controls. To test the role of complement 3a receptor (C3aR1) on macrophages and liver resident macrophages in MASLD, we generated mice deficient in C3aR1 on all macrophages (C3aR1-MφKO) or specifically in liver Kupffer cells (C3aR1-KpKO) and subjected them to a model of metabolic steatotic liver disease. We show that macrophages account for the vast majority of C3ar1 expression in the liver. Overall, C3aR1-MφKO and C3aR1-KpKO mice have similar body weight gain without significant alterations in glucose homeostasis, hepatic steatosis and fibrosis, compared to controls on a MASLD-inducing diet. This study demonstrates that C3aR1 deletion in macrophages or Kupffer cells, the predominant liver cell type expressing C3aR1, has no significant effect on liver steatosis, inflammation or fibrosis in a dietary MASLD model.
Collapse
Affiliation(s)
- Edwin A. Homan
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, New York, 10021
| | - Ankit Gilani
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, New York, 10021
| | - Alfonso Rubio-Navarro
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, New York, 10021
| | - Maya A. Johnson
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, New York, 10021
| | - Odin M. Schaepkens
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, New York, 10021
| | - Eric Cortada
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, New York, 10021
| | - Renan Pereira de Lima
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, New York, 10021
| | - Lisa Stoll
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, New York, 10021
| | - James C. Lo
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, New York, 10021
| |
Collapse
|
8
|
Mohammadyari E, Miwa T, Golla M, Song WC. Therapeutic targeting of factor D and MASP3 in complement-mediated diseases: Lessons learned from mouse studies. Eur J Immunol 2024; 54:e2350845. [PMID: 39540581 DOI: 10.1002/eji.202350845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 11/16/2024]
Abstract
The alternative pathway (AP) plays a major role in many complement-mediated human diseases. Factor D (FD), a rate-limiting enzyme in AP complement activation, is an attractive therapeutic target. Unlike other complement proteins, FD is synthesized primarily in adipose tissue, and its levels in human blood are relatively low. However, because of FD's high turnover rate, therapeutic targeting with monoclonal antibodies and chemical inhibitors has been challenging. The recent discovery that FD activity is regulated by mannose-binding lectin-associated serine protease 3 (MASP3), through conversion of a zymogen to mature FD, has sparked interest in MASP3 inhibition as a new way to block FD function and AP complement activity. Here, we review studies of mouse models of FD and MASP3 inhibition. We additionally discuss the lessons learned from these studies and their implications for therapeutic targeting of human FD and MASP3.
Collapse
Affiliation(s)
- Eshagh Mohammadyari
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Takashi Miwa
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Madhu Golla
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Wen-Chao Song
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
9
|
Wei Y, Lv Z, Xiao T, Du Z. The role of MASP1 in the complement system and expression characteristics in response to GCRV infection in grass carp. FISH & SHELLFISH IMMUNOLOGY 2024; 151:109712. [PMID: 38901682 DOI: 10.1016/j.fsi.2024.109712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/17/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024]
Abstract
The grass carp (Ctenopharyngodon idella) constitutes a significant economic resource within the aquaculture sector of our nation, yet it has been chronically afflicted by the Grass Carp Reovirus (GCRV) disease. The complement system, a vital component of fish's innate immunity, plays a crucial role in combating viral infections. This research investigates the potential role of MASP1, a key molecule in the lectin pathway of the complement system, in the GCRV infection in grass carp. An analysis of the molecular characteristics of MASP1 in grass carp revealed that its identity and similarity percentages range from 35.10 to 91.00 % and 35.30-91.00 %, respectively, in comparison to other species. Phylogenetically, MASP1 in C. idella aligns closely with species such as Danio rerio, Cyprinus carpio, and Carassius carassius, exhibiting chromosomal collinearity with the zebrafish. Subsequent tissue analysis in both healthy and GCRV-infected grass carp indicated that MASP1's basal expression was predominantly in the liver. Post-GCRV infection, MASP1 expression in various tissues exhibited temporal variations: peaking in the liver on day 5, spleen on day 7, and kidney on day 14. Furthermore, employing Complement Component 3 (C3) as a benchmark for complement system activation, it was observed that MASP1 could activate and cleave C3 to C3b. MASP1 also demonstrated an inhibitory effect on GCRV replication (compared with the control group, VP2 and VP7 decreased by 6.82-fold and 4.37-fold) and enhanced the expression of antiviral genes, namely IRF3, IRF7 and IFN1 (compared with the control group, increased 2.25-fold, 45.38-fold and 22.37-fold, respectively). In vivo protein injection experiments substantiated MASP1's influence on the relative mRNA expression levels of C3 in various tissues and its protein expression in serum. This study also verified that C3 could modulate the expression of antiviral genes such as IFN1 and IRF3.
Collapse
Affiliation(s)
- Yuling Wei
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China; Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha, Hunan, 410128, China
| | - Zhao Lv
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha, Hunan, 410128, China
| | - Tiaoyi Xiao
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha, Hunan, 410128, China.
| | - Zongjun Du
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China.
| |
Collapse
|
10
|
Singh S, Hussain S, Yadav SS, Tiwari NP, Usman K, Sawlani KK, Khattri S. Association of Serum Adipsin Level with Insulin Resistance and Inflammatory Markers in Newly Diagnosed Type two Diabetes Mellitus Patients. Indian J Clin Biochem 2024; 39:415-420. [PMID: 39005860 PMCID: PMC11239611 DOI: 10.1007/s12291-023-01126-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 02/21/2023] [Indexed: 04/01/2023]
Abstract
Adipsin is an anti-inflammatory adipokines and its altered level was seen in obesity and type II DM. Our study investigated the clinical significance of serum adipsin levels as a risk marker for type 2 diabetes and its relationships with insulin resistance and various adipo-cytokines. The study included 110 treatment-naïve T2DM cases and 100 controls of similar age and gender from northern India. Clinical, biochemical, and anthropometric characteristics were all profiled. Serum adipo-cytokines were measured using ELISA methods. Adipsin was significantly inversely correlated with body mass index (BMI), waist circumference, fasting plasma glucose, glycated haemoglobin (HbA1C), total cholesterol (TC), triglyceride (TG), homeostasis model assessment-estimated insulin resistance (HOMA-IR), tumour necrosis factor- α (TNF-α) and interleulin-6 (IL-6) and positively correlated with high-density lipoprotein cholesterol (HDL-C) and homeostasis model assessment of β-cell function (HOMA-B) (P < 0.05). T2DM occurrence decreased with increasing concentration of adipsin with an odds ratio (OR) of 0.68 (95% CI = 0.58-0.79), P < 0.001. The area under curve (95% CI) for adipsin was 0.70 (0.63 to 0.76) with P < 0.001. The best cutoff value for adipsin to predict T2DM was < 5.50 µg/ml with 47.27% sensitivity and 82.00% specificity. FPG and WC were both independent predictors of serum adipsin levels. Our findings showed that high adipsin levels reduced the likelihood of T2DM and emerged as a potential risk marker in the prediction of T2DM. Graphical Abstract
Collapse
Affiliation(s)
- Somya Singh
- Department of Pharmacology and Therapeutics, King George's Medical University, Lucknow, 226003 India
| | - Sartaj Hussain
- Department of Pharmacology, All India Institute of Medical Science, Vijaypur, Jammu, 184120 India
| | - Suraj Singh Yadav
- Department of Pharmacology and Therapeutics, King George's Medical University, Lucknow, 226003 India
| | - Nitesh Pati Tiwari
- Department of Pharmacology Government Medical College, Badaun, 243601 India
| | - Kauser Usman
- Department of Medicine, King George's Medical University, Lucknow, 226003 India
| | - Kamal Kumar Sawlani
- Department of Medicine, King George's Medical University, Lucknow, 226003 India
| | - Sanjay Khattri
- Department of Pharmacology and Therapeutics, King George's Medical University, Lucknow, 226003 India
| |
Collapse
|
11
|
Zhang X, Tian L, Majumdar A, Scheller EL. Function and Regulation of Bone Marrow Adipose Tissue in Health and Disease: State of the Field and Clinical Considerations. Compr Physiol 2024; 14:5521-5579. [PMID: 39109972 PMCID: PMC11725182 DOI: 10.1002/cphy.c230016] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2025]
Abstract
Bone marrow adipose tissue (BMAT) is a metabolically and clinically relevant fat depot that exists within bone. Two subtypes of BMAT, regulated and constitutive, reside in hematopoietic-rich red marrow and fatty yellow marrow, respectively, and exhibit distinct characteristics compared to peripheral fat such as white and brown adipose tissues. Bone marrow adipocytes (BMAds) are evolutionally preserved in most vertebrates, start development after birth and expand throughout life, and originate from unique progenitor populations that control bone formation and hematopoiesis. Mature BMAds also interact closely with other cellular components of the bone marrow niche, serving as a nearby energy reservoir to support the skeletal system, a signaling hub that contributes to both local and systemic homeostasis, and a final fuel reserve for survival during starvation. Though BMAT and bone are often inversely correlated, more BMAT does not always mean less bone, and the prevention of BMAT expansion as a strategy to prevent bone loss remains questionable. BMAT adipogenesis and lipid metabolism are regulated by the nervous systems and a variety of circulating hormones. This contributes to the plasticity of BMAT, including BMAT expansion in common physiological or pathological conditions, and BMAT catabolism under certain extreme circumstances, which are often associated with malnutrition and/or systemic inflammation. Altogether, this article provides a comprehensive overview of the local and systemic functions of BMAT and discusses the regulation and plasticity of this unique adipose tissue depot in health and disease. © 2024 American Physiological Society. Compr Physiol 14:5521-5579, 2024.
Collapse
Affiliation(s)
- Xiao Zhang
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA
| | - Linda Tian
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA
| | - Anurag Majumdar
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, St. Louis, Missouri, USA
| | - Erica L. Scheller
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA
- Department of Cell Biology and Physiology, Washington University, St. Louis, Missouri, USA
| |
Collapse
|
12
|
Bitounis D, Jacquinet E, Rogers MA, Amiji MM. Strategies to reduce the risks of mRNA drug and vaccine toxicity. Nat Rev Drug Discov 2024; 23:281-300. [PMID: 38263456 DOI: 10.1038/s41573-023-00859-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2023] [Indexed: 01/25/2024]
Abstract
mRNA formulated with lipid nanoparticles is a transformative technology that has enabled the rapid development and administration of billions of coronavirus disease 2019 (COVID-19) vaccine doses worldwide. However, avoiding unacceptable toxicity with mRNA drugs and vaccines presents challenges. Lipid nanoparticle structural components, production methods, route of administration and proteins produced from complexed mRNAs all present toxicity concerns. Here, we discuss these concerns, specifically how cell tropism and tissue distribution of mRNA and lipid nanoparticles can lead to toxicity, and their possible reactogenicity. We focus on adverse events from mRNA applications for protein replacement and gene editing therapies as well as vaccines, tracing common biochemical and cellular pathways. The potential and limitations of existing models and tools used to screen for on-target efficacy and de-risk off-target toxicity, including in vivo and next-generation in vitro models, are also discussed.
Collapse
Affiliation(s)
- Dimitrios Bitounis
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
- Moderna, Inc., Cambridge, MA, USA
| | | | | | - Mansoor M Amiji
- Departments of Pharmaceutical Sciences and Chemical Engineering, Northeastern University, Boston, MA, USA.
| |
Collapse
|
13
|
Dare A, Chen SY. Adipsin in the pathogenesis of cardiovascular diseases. Vascul Pharmacol 2024; 154:107270. [PMID: 38114042 PMCID: PMC10939892 DOI: 10.1016/j.vph.2023.107270] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 12/21/2023]
Abstract
Adipsin is an adipokine predominantly synthesized in adipose tissues and released into circulation. It is also known as complement factor-D (CFD), acting as the rate-limiting factor in the alternative complement pathway and exerting essential functions on the activation of complement system. The deficiency of CFD in humans is a very rare condition. However, complement overactivation has been implicated in the etiology of numerous disorders, including cardiovascular disease (CVD). Increased circulating level of adipsin has been reported to promote vascular derangements, systemic inflammation, and endothelial dysfunction. Prospective and case-control studies showed that this adipokine is directly associated with all-cause death and rehospitalization in patients with coronary artery disease. Adipsin has also been implicated in pulmonary arterial hypertension, abdominal aortic aneurysm, pre-eclampsia, and type-2 diabetes which is a major risk factor for CVD. Importantly, serum adipsin has been recognized as a unique prognostic marker for assessing cardiovascular diseases. At present, there is paucity of experimental evidence about the precise role of adipsin in the etiology of CVD. However, this mini review provides some insight on the contribution of adipsin in the pathogenesis of CVD and highlights its role on endothelial, smooth muscle and immune cells that mediate cardiovascular functions.
Collapse
Affiliation(s)
- Ayobami Dare
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, USA
| | - Shi-You Chen
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, USA; The Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA.
| |
Collapse
|
14
|
Xiao MT, Ellsworth CR, Qin X. Emerging role of complement in COVID-19 and other respiratory virus diseases. Cell Mol Life Sci 2024; 81:94. [PMID: 38368584 PMCID: PMC10874912 DOI: 10.1007/s00018-024-05157-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 01/03/2024] [Accepted: 02/03/2024] [Indexed: 02/19/2024]
Abstract
The complement system, a key component of innate immunity, provides the first line of defense against bacterial infection; however, the COVID-19 pandemic has revealed that it may also engender severe complications in the context of viral respiratory disease. Here, we review the mechanisms of complement activation and regulation and explore their roles in both protecting against infection and exacerbating disease. We discuss emerging evidence related to complement-targeted therapeutics in COVID-19 and compare the role of the complement in other respiratory viral diseases like influenza and respiratory syncytial virus. We review recent mechanistic studies and animal models that can be used for further investigation. Novel knockout studies are proposed to better understand the nuances of the activation of the complement system in respiratory viral diseases.
Collapse
Affiliation(s)
- Mark T Xiao
- Division of Comparative Pathology, Tulane National Primate Research Center, Health Sciences Campus, 18703 Three Rivers Road, Covington, LA, 70433, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Calder R Ellsworth
- Division of Comparative Pathology, Tulane National Primate Research Center, Health Sciences Campus, 18703 Three Rivers Road, Covington, LA, 70433, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Xuebin Qin
- Division of Comparative Pathology, Tulane National Primate Research Center, Health Sciences Campus, 18703 Three Rivers Road, Covington, LA, 70433, USA.
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, 70112, USA.
| |
Collapse
|
15
|
Wei Y, Lv Z, Liu Q, Yu J, Xiao Y, Du Z, Xiao T. Structural comparison and expression function analysis of BF/C2 in Ctenopharyngodon idella and Squaliobarbus curriculus. FISH & SHELLFISH IMMUNOLOGY 2023; 142:109154. [PMID: 37821003 DOI: 10.1016/j.fsi.2023.109154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 09/17/2023] [Accepted: 10/08/2023] [Indexed: 10/13/2023]
Abstract
Ctenopharyngodon idella and Squaliobarbus curriculus, members of the Cyprinidae family and Yaroideae subfamily, have shown different levels of resistance to grass carp reo virus (GCRV), with S. curriculus exhibiting higher resilience. In the pursuit to explore the distinctions in the structural and expression traits of BF/C2 (A,B) between the two species, we conducted an analysis involving the cloning and examination of various coding sequences (CDS). We successfully cloned the CDS of ci-BF/C2A and ci-BF/C2B from C. idella, which spanned 2259 bp and 2514 bp respectively, encoding 752 and 837 amino acids. Similarly, the CDS of sc-BF/C2A and sc-BF/C2B from S. curriculus were cloned, featuring lengths of 1353 bp and 2517 bp and encoding 450 and 838 amino acids, respectively. A chromosome collinearity assessment revealed that ci-BF/C2A demonstrated collinearity with sc-BF/C2A, a finding not replicated with ci-BF/C2B and sc-BF/C2B. Delving into gene structure, we discerned that ci-BF/C2A harbored a greater number of Tryp_SPc domains compared to sc-BF/C2A. Following this, we engineered and purified six prokaryotic recombinant proteins: CI-BF/C2A, CI-BF/C2A1 (a variant resulting from the deletion of the Tryp_SPc domain of CI-BF/C2A), CI-BF/C2A2 (representing the Tryp_SPc domain of CI-BF/C2A), CI-BF/C2B, SC-BF/C2A, and SC-BF/C2B. Through serum co-incubation tests with these recombinant proteins, we established the activation of the complement marker C3 in each case. Utilizing fluorescence quantitative expression analysis, we observed ubiquitous expression of ci-BF/C2A and ci-BF/C2B across all grass carp tissues, predominantly in the liver. This pattern mirrored in S. curriculus, where sc-BF/C2A was highly expressed in the gills, and sc-BF/C2B manifested notably in the liver. Kidney cell infection experiments on both species revealed enhanced resistance to GCRV post-incubation with the recombinant proteins. Notably, cells treated with SC-BF/C2 (A, B) exhibited pronounced resilience compared to those treated with CI-BF/C2 (A, B, A1, A2). However, cells incubated with CI-BF/C2A1 and CI-BF/C2A2 showed strengthen resistance relative to cells treated with CI-BF/C2A and CI-BF/C2B. In GCRV infection trials on grass carp, ci-BF/C2A and ci-BF/C2B expressions reached a zenith on the seventh day post-infection, highlighting a distinctive functional mode in immune defense against GCRV infection orchestrated by BF/C2. The empirical data underscores the pivotal role of the Tryp_SPc domain in immune responses to GCRV infection, pinpointing its influence on ci-BF/C2A expression. Conclusively, this investigation provides a foundational understanding of the unique immune function characteristics of BF/C2 in grass carp, paving the way for further scholarly exploration in this realm.
Collapse
Affiliation(s)
- Yuling Wei
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China; Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha, Hunan 410128, China
| | - Zhao Lv
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha, Hunan 410128, China
| | - Qiaolin Liu
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha, Hunan 410128, China
| | - Jianbo Yu
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha, Hunan 410128, China
| | - Yu Xiao
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha, Hunan 410128, China
| | - Zongjun Du
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Tiaoyi Xiao
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha, Hunan 410128, China.
| |
Collapse
|
16
|
Kim J, Oh CM, Kim H. The Interplay of Adipokines and Pancreatic Beta Cells in Metabolic Regulation and Diabetes. Biomedicines 2023; 11:2589. [PMID: 37761031 PMCID: PMC10526203 DOI: 10.3390/biomedicines11092589] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/19/2023] [Accepted: 09/20/2023] [Indexed: 09/29/2023] Open
Abstract
The interplay between adipokines and pancreatic beta cells, often referred to as the adipo-insular axis, plays a crucial role in regulating metabolic homeostasis. Adipokines are signaling molecules secreted by adipocytes that have profound effects on several physiological processes. Adipokines such as adiponectin, leptin, resistin, and visfatin influence the function of pancreatic beta cells. The reciprocal communication between adipocytes and beta cells is remarkable. Insulin secreted by beta cells affects adipose tissue metabolism, influencing lipid storage and lipolysis. Conversely, adipokines released from adipocytes can influence beta cell function and survival. Chronic obesity and insulin resistance can lead to the release of excess fatty acids and inflammatory molecules from the adipose tissue, contributing to beta cell dysfunction and apoptosis, which are key factors in developing type 2 diabetes. Understanding the complex interplay of the adipo-insular axis provides insights into the mechanisms underlying metabolic regulation and pathogenesis of metabolic disorders. By elucidating the molecular mediators involved in this interaction, new therapeutic targets and strategies may emerge to reduce the risk and progression of diseases, such as type 2 diabetes and its associated complications. This review summarizes the interactions between adipokines and pancreatic beta cells, and their roles in the pathogenesis of diabetes and metabolic diseases.
Collapse
Affiliation(s)
- Joon Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea;
| | - Chang-Myung Oh
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea;
| | - Hyeongseok Kim
- Department of Biochemistry, College of Medicine, Chungnam National University, Daejeon 35105, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35105, Republic of Korea
| |
Collapse
|
17
|
de Boer ECW, Thielen AJF, Langereis JD, Kamp A, Brouwer MC, Oskam N, Jongsma ML, Baral AJ, Spaapen RM, Zeerleder S, Vidarsson G, Rispens T, Wouters D, Pouw RB, Jongerius I. The contribution of the alternative pathway in complement activation on cell surfaces depends on the strength of classical pathway initiation. Clin Transl Immunology 2023; 12:e1436. [PMID: 36721662 PMCID: PMC9881211 DOI: 10.1002/cti2.1436] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 10/31/2022] [Accepted: 12/23/2022] [Indexed: 01/28/2023] Open
Abstract
Objectives The complement system is an important component of innate immunity. The alternative pathway (AP) amplification loop is considered an essential feed forward mechanism for complement activation. However, the role of the AP in classical pathway (CP) activation has only been studied in ELISA settings. Here, we investigated its contribution on physiologically relevant surfaces of human cells and bacterial pathogens and in antibody-mediated complement activation, including in autoimmune haemolytic anaemia (AIHA) setting with autoantibodies against red blood cells (RBCs). Methods We evaluated the contribution of the AP to complement responses initiated through the CP on human RBCs by serum of AIHA patients and recombinant antibodies. Moreover, we studied complement activation on Neisseria meningitidis and Escherichia coli. The effect of the AP was examined using either AP-depleted sera or antibodies against factor B and factor D. Results We show that the amplification loop is redundant when efficient CP activation takes place. This is independent of the presence of membrane-bound complement regulators. The role of the AP may become significant when insufficient CP complement activation occurs, but this depends on antibody levels and (sub)class. Our data indicate that therapeutic intervention in the amplification loop will most likely not be effective to treat antibody-mediated diseases. Conclusion The AP can be bypassed through efficient CP activation. The AP amplification loop has a role in complement activation during conditions of modest activation via the CP, when it can allow for efficient complement-mediated killing.
Collapse
Affiliation(s)
- Esther CW de Boer
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands,Department of Pediatric Immunology, Rheumatology, and Infectious Diseases, Emma Children's HospitalAmsterdam University Medical CentreAmsterdamThe Netherlands
| | - Astrid JF Thielen
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands
| | - Jeroen D Langereis
- Laboratory of Medical Immunology, Radboud Institute for Molecular Life SciencesRadboudumcNijmegenThe Netherlands,Radboud Center for Infectious Diseases, RadboudumcNijmegenThe Netherlands
| | - Angela Kamp
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands
| | - Mieke C Brouwer
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands
| | - Nienke Oskam
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands
| | - Marlieke L Jongsma
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands
| | - April J Baral
- Translational and Clinical Research InstituteNewcastle upon TyneUK
| | - Robbert M Spaapen
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands
| | - Sacha Zeerleder
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands,Department of Hematology, Luzerner KantonsspitalLuzern and University of BernBernSwitzerland,Department for BioMedical ResearchUniversity of BernBernSwitzerland
| | - Gestur Vidarsson
- Department of Experimental Immunohematology, Sanquin Research, and Landsteiner LaboratoryAmsterdam University Medical CenterAmsterdamThe Netherlands
| | - Theo Rispens
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands
| | - Diana Wouters
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands,Centre for Infectious Disease ControlNational Institute for Public Health and the Environment (RIVM)BilthovenThe Netherlands
| | - Richard B Pouw
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands,Sanquin Health SolutionsAmsterdamThe Netherlands
| | - Ilse Jongerius
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands,Department of Pediatric Immunology, Rheumatology, and Infectious Diseases, Emma Children's HospitalAmsterdam University Medical CentreAmsterdamThe Netherlands
| |
Collapse
|
18
|
Abstract
Complement factor D (FD) is a serine protease that plays an essential role in the activation of the alternative pathway (AP) by cleaving complement factor B (FB) and generating the C3 convertases C3(H2 O)Bb and C3bBb. FD is produced mainly from adipose tissue and circulates in an activated form. On the contrary, the other serine proteases of the complement system are mainly synthesized in the liver. The activation mechanism of FD has long been unknown. Recently, a serendipitous discovery in the mechanism of FD activation has been provided by a generation of Masp1 gene knockout mice lacking both the serine protease MASP-1 and its alternative splicing variant MASP-3, designated MASP-1/3-deficient mice. Sera from the MASP-1/3-deficient mice had little-to-no lectin pathway (LP) and AP activity with circulating zymogen or proenzyme FD (pro-FD). Sera from patients with 3MC syndrome carrying mutations in the MASP1 gene also had circulating pro-FD, suggesting that MASP-1 and/or MASP-3 are involved in activation of FD. Here, we summarize the current knowledge of the mechanism of FD activation that was finally elucidated using the sera of mice monospecifically deficient for MASP-1 or MASP-3. Sera of the MASP-1-deficient mice lacked LP activity, but those of the MASP-3-deficient mice lacked AP activity with pro-FD. This review illustrates the pivotal role of MASP-3 in the physiological activation of the AP via activation of FD.
Collapse
Affiliation(s)
- Hideharu Sekine
- Department of Immunology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Takeshi Machida
- Department of Immunology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Teizo Fujita
- Fukushima Prefectural General Hygiene Institute, Fukushima, Japan
| |
Collapse
|
19
|
Schubart A, Flohr S, Junt T, Eder J. Low-molecular weight inhibitors of the alternative complement pathway. Immunol Rev 2023; 313:339-357. [PMID: 36217774 PMCID: PMC10092480 DOI: 10.1111/imr.13143] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Dysregulation of the alternative complement pathway predisposes individuals to a number of diseases. It can either be evoked by genetic alterations in or by stabilizing antibodies to important pathway components and typically leads to severe diseases such as paroxysmal nocturnal hemoglobinuria, atypical hemolytic uremic syndrome, C3 glomerulopathy, and age-related macular degeneration. In addition, the alternative pathway may also be involved in many other diseases where its amplifying function for all complement pathways might play a role. To identify specific alternative pathway inhibitors that qualify as therapeutics for these diseases, drug discovery efforts have focused on the two central proteases of the pathway, factor B and factor D. Although drug discovery has been challenging for a number of reasons, potent and selective low-molecular weight (LMW) oral inhibitors have now been discovered for both proteases and several molecules are in clinical development for multiple complement-mediated diseases. While the clinical development of these inhibitors initially focuses on diseases with systemic and/or peripheral tissue complement activation, the availability of LMW inhibitors may also open up the prospect of inhibiting complement in the central nervous system where its activation may also play an important role in several neurodegenerative diseases.
Collapse
Affiliation(s)
- Anna Schubart
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Stefanie Flohr
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Tobias Junt
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Jörg Eder
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| |
Collapse
|
20
|
Gibson BG, Cox TE, Marchbank KJ. Contribution of animal models to the mechanistic understanding of Alternative Pathway and Amplification Loop (AP/AL)-driven Complement-mediated Diseases. Immunol Rev 2023; 313:194-216. [PMID: 36203396 PMCID: PMC10092198 DOI: 10.1111/imr.13141] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
This review aimed to capture the key findings that animal models have provided around the role of the alternative pathway and amplification loop (AP/AL) in disease. Animal models, particularly mouse models, have been incredibly useful to define the role of complement and the alternative pathway in health and disease; for instance, the use of cobra venom factor and depletion of C3 provided the initial insight that complement was essential to generate an appropriate adaptive immune response. The development of knockout mice have further underlined the importance of the AP/AL in disease, with the FH knockout mouse paving the way for the first anti-complement drugs. The impact from the development of FB, properdin, and C3 knockout mice closely follows this in terms of mechanistic understanding in disease. Indeed, our current understanding that complement plays a role in most conditions at one level or another is rooted in many of these in vivo studies. That C3, in particular, has roles beyond the obvious in innate and adaptive immunity, normal physiology, and cellular functions, with or without other recognized AP components, we would argue, only extends the reach of this arm of the complement system. Humanized mouse models also continue to play their part. Here, we argue that the animal models developed over the last few decades have truly helped define the role of the AP/AL in disease.
Collapse
Affiliation(s)
- Beth G. Gibson
- Complement Therapeutics Research Group and Newcastle University Translational and Clinical Research InstituteFaculty of Medical ScienceNewcastle‐upon‐TyneUK
- National Renal Complement Therapeutics CentreaHUS ServiceNewcastle upon TyneUK
| | - Thomas E. Cox
- Complement Therapeutics Research Group and Newcastle University Translational and Clinical Research InstituteFaculty of Medical ScienceNewcastle‐upon‐TyneUK
- National Renal Complement Therapeutics CentreaHUS ServiceNewcastle upon TyneUK
| | - Kevin J. Marchbank
- Complement Therapeutics Research Group and Newcastle University Translational and Clinical Research InstituteFaculty of Medical ScienceNewcastle‐upon‐TyneUK
- National Renal Complement Therapeutics CentreaHUS ServiceNewcastle upon TyneUK
| |
Collapse
|
21
|
Sood S, Feehan J, Itsiopoulos C, Wilson K, Plebanski M, Scott D, Hebert JR, Shivappa N, Mousa A, George ES, de Courten B. Higher Adherence to a Mediterranean Diet Is Associated with Improved Insulin Sensitivity and Selected Markers of Inflammation in Individuals Who Are Overweight and Obese without Diabetes. Nutrients 2022; 14:nu14204437. [PMID: 36297122 PMCID: PMC9608711 DOI: 10.3390/nu14204437] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/17/2022] [Accepted: 10/20/2022] [Indexed: 11/07/2022] Open
Abstract
Insulin resistance (IR) and chronic low-grade inflammation are risk factors for chronic diseases including type 2 diabetes (T2D) and cardiovascular disease. This study aimed to investigate two dietary indices: Mediterranean Diet Score (MDS) and Dietary Inflammatory Index (DII®), and their associations with direct measures of glucose metabolism and adiposity, and biochemical measures including lipids, cytokines and adipokines in overweight/obese adults. This cross-sectional study included 65 participants (males = 63%; age 31.3 ± 8.5 years). Dietary intake via 3-day food diaries was used to measure adherence to MDS (0-45 points); higher scores indicating adherence. Energy-adjusted DII (E-DII) scores were calculated with higher scores indicating a pro-inflammatory diet. IR was assessed using hyperinsulinemic euglycemic clamps, insulin secretion by intravenous glucose tolerance test, adiposity by dual-energy X-ray absorptiometry, and circulating cytokine and adipokine concentrations by multiplex assays. Higher MDS was associated with greater insulin sensitivity (β = 0.179; 95%CI: 0.39, 0.318) after adjusting for age, sex and % body fat, and lower NF-κB, higher adiponectin and adipsin in unadjusted and adjusted models. Higher E-DII score was associated with increased total cholesterol (β = 0.364; 95%CI: 0.066, 0.390) and LDL-cholesterol (β = 0.305; 95%CI: 0.019, 0.287) but not with adiposity, glucose metabolism, cytokines or adipokines. Greater MDS appears to be associated with decreased IR and inflammatory markers in overweight/obese adults.
Collapse
Affiliation(s)
- Surbhi Sood
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Faculty of Health, Deakin University, Geelong, VIC 3216, Australia
| | - Jack Feehan
- Institute for Health and Sport, Victoria University, Footscray, VIC 3011, Australia
| | - Catherine Itsiopoulos
- Department of Medicine, Nursing and Health Sciences, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia
| | - Kirsty Wilson
- Department of Medicine, Nursing and Health Sciences, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia
| | - Magdalena Plebanski
- Department of Medicine, Nursing and Health Sciences, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia
| | - David Scott
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Faculty of Health, Deakin University, Geelong, VIC 3216, Australia
- Department of Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC 3168, Australia
| | - James R. Hebert
- Cancer Prevention and Control Program and Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
- Department of Nutrition, Connecting Health Innovations LLC, Columbia, SC 29208, USA
| | - Nitin Shivappa
- Cancer Prevention and Control Program and Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
- Department of Nutrition, Connecting Health Innovations LLC, Columbia, SC 29208, USA
| | - Aya Mousa
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Monash University, Clayton, VIC 3168, Australia
| | - Elena S. George
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Faculty of Health, Deakin University, Geelong, VIC 3216, Australia
| | - Barbora de Courten
- Department of Medicine, Nursing and Health Sciences, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia
- Department of Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC 3168, Australia
- Correspondence: ; Tel.: +61-3-9925-6474
| |
Collapse
|
22
|
The complement C3-complement factor D-C3a receptor signalling axis regulates cardiac remodelling in right ventricular failure. Nat Commun 2022; 13:5409. [PMID: 36109509 PMCID: PMC9478115 DOI: 10.1038/s41467-022-33152-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 08/25/2022] [Indexed: 11/24/2022] Open
Abstract
Failure of the right ventricle plays a critical role in any type of heart failure. However, the mechanism remains unclear, and there is no specific therapy. Here, we show that the right ventricle predominantly expresses alternative complement pathway-related genes, including Cfd and C3aR1. Complement 3 (C3)-knockout attenuates right ventricular dysfunction and fibrosis in a mouse model of right ventricular failure. C3a is produced from C3 by the C3 convertase complex, which includes the essential component complement factor D (Cfd). Cfd-knockout mice also show attenuation of right ventricular failure. Moreover, the plasma concentration of CFD correlates with the severity of right ventricular failure in patients with chronic right ventricular failure. A C3a receptor (C3aR) antagonist dramatically improves right ventricular dysfunction in mice. In summary, we demonstrate the crucial role of the C3-Cfd-C3aR axis in right ventricular failure and highlight potential therapeutic targets for right ventricular failure. Right ventricular (RV) failure is clinically crucial, but there is no specific therapy. Here, the authors show that the complement alternative pathway is activated in RV failure and that blockade of the pathway ameliorates RV failure in mice.
Collapse
|
23
|
Man W, Song X, Xiong Z, Gu J, Lin J, Gu X, Yu D, Li C, Jiang M, Zhang X, Yang Z, Cao Y, Zhang Y, Shu X, Wu D, Wang H, Ji G, Sun D. Exosomes derived from pericardial adipose tissues attenuate cardiac remodeling following myocardial infarction by Adipsin-regulated iron homeostasis. Front Cardiovasc Med 2022; 9:1003282. [PMID: 36172581 PMCID: PMC9510661 DOI: 10.3389/fcvm.2022.1003282] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 08/09/2022] [Indexed: 11/13/2022] Open
Abstract
As a vital adipokine, Adipsin is closely associated with cardiovascular risks. Nevertheless, its role in the onset and development of cardiovascular diseases remains elusive. This study was designed to examine the effect of Adipsin on survival, cardiac dysfunction and adverse remodeling in the face of myocardial infarction (MI) injury. In vitro experiments were conducted to evaluate the effects of Adipsin on cardiomyocyte function in the face of hypoxic challenge and the mechanisms involved. Our results showed that Adipsin dramatically altered expression of proteins associated with iron metabolism and ferroptosis. In vivo results demonstrated that Adipsin upregulated levels of Ferritin Heavy Chain (FTH) while downregulating that of Transferrin Receptor (TFRC) in peri-infarct regions 1 month following MI. Adipsin also relieved post-MI-associated lipid oxidative stress as evidenced by decreased expression of COX2 and increased GPX4 level. Co-immunoprecipitation and immunofluorescence imaging prove a direct interaction between Adipsin and IRP2. As expected, cardioprotection provided by Adipsin depends on the key molecule of IRP2. These findings revealed that Adipsin could be efficiently delivered to the heart by exosomes derived from pericardial adipose tissues. In addition, Adipsin interacted with IRP2 to protect cardiomyocytes against ferroptosis and maintain iron homeostasis. Therefore, Adipsin-overexpressed exosomes derived from pericardial adipose tissues may be a promising therapeutic strategy to prevent adverse cardiac remodeling following ischemic heart injury.
Collapse
Affiliation(s)
- Wanrong Man
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Xinglong Song
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Zhenyu Xiong
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Jing Gu
- School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Jie Lin
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Xiaoming Gu
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi’an, China
| | - Duan Yu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Congye Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Mengyuan Jiang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Xuebin Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Zhi Yang
- Department of Radiation Oncology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yang Cao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yan Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Xiaofei Shu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Dexi Wu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Haichang Wang
- Heart Hospital, Xi’an International Medical Center, Xi’an, China
- Haichang Wang,
| | - Gang Ji
- Department of Gastrointestinal Surgery, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, China
- Gang Ji,
| | - Dongdong Sun
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- *Correspondence: Dongdong Sun,
| |
Collapse
|
24
|
Duan Y, Zhang X, Zhang X, Lin J, Shu X, Man W, Jiang M, Zhang Y, Wu D, Zhao Z, Sun D. Inhibition of macrophage-derived foam cells by Adipsin attenuates progression of atherosclerosis. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166533. [PMID: 36064133 DOI: 10.1016/j.bbadis.2022.166533] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 08/03/2022] [Accepted: 08/23/2022] [Indexed: 10/14/2022]
Abstract
Phagocytosis of oxidized low-density lipoprotein (OxLDL) by macrophages yields "foam cells" and serves as a hallmark of atherosclerotic lesion. Adipsin is a critical component of the complement activation pathway. Recent evidence has indicated an obligatory role for Adipsin in pathological models including ischemia-reperfusion and sepsis. Adipsin levels are significantly decreased in patients with asymptomatic carotid atherosclerosis, implying the role for Adipsin as a potential marker of asymptomatic carotid atherosclerosis. This study was designed to evaluate the role for Adipsin in atherosclerosis and the mechanisms involved using both in vivo and in vitro experiments. ApoE-/-/AdipsinTg mice were constructed and were fed a high-fat diet for 12 weeks. Compared with ApoE-/- mice, area of the sclerotic plaques was reduced, along with lower macrophage deposition within the plaque in ApoE-/-/AdipsinTg mice. RAW264.7 cells and bone marrow-derived macrophages (BMDMs) were stimulated with oxLDL (50 μg/ml). Adenovirus vectors containing the Adipsin gene were transfected into macrophages. Lipid accumulation was observed by Oil red O staining. Western blot and reverse transcription-polymerase chain reaction data revealed that Adipsin overexpression inhibited oxLDL-induced lipid uptake and foam cell formation and upregulation of CD36 and PPARγ in Ad-Adipsin-transfected macrophages. In addition, the PPARγ-specific agonist GW1929 reversed Adipsin overexpression-evoked inhibitory effect on lipid uptake. These results demonstrate unequivocally that Adipsin inhibits lipid uptake in a PPARγ/CD36-dependent manner and prevents the formation of foam cells, implying that Adipsin may be a potential therapeutic target against atherosclerosis.
Collapse
Affiliation(s)
- Yu Duan
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xuebin Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiao Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jie Lin
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiaofei Shu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Wanrong Man
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Mengyuan Jiang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yan Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Dexi Wu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhijing Zhao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Dongdong Sun
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
25
|
Kusakari K, Machida T, Ishida Y, Omori T, Suzuki T, Sekimata M, Wada I, Fujita T, Sekine H. The complex formation of MASP-3 with pattern recognition molecules of the lectin complement pathway retains MASP-3 in the circulation. Front Immunol 2022; 13:907023. [PMID: 36052069 PMCID: PMC9425028 DOI: 10.3389/fimmu.2022.907023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 07/27/2022] [Indexed: 12/02/2022] Open
Abstract
The complement system plays an important role in host defense and is activated via three different activation pathways. We have previously reported that mannose-binding lectin-associated serine protease (MASP)-3, unlike its splicing variant MASP-1, circulates in an active form and is essential for the activation of the alternative pathway (AP) via the activation of complement factor D (FD). On the other hand, like MASP-1 and MASP-2 of the lectin pathway (LP), MASP-3 forms a complex with the pattern recognition molecules (PRMs) of the LP (LP-PRMs). Both MASP-1 and MASP-2 can be activated efficiently when the LP-PRMs complexed with them bind to their ligands. On the other hand, it remains unclear how MASP-3 is activated, or whether complex formation of MASP-3 with LP-PRMs is involved in activation of MASP-3 or its efficiency in the circulation. To address these issues, we generated wild-type (WT) and four mutant recombinant mouse MASP-3 proteins fused with PA (human podoplanin dodecapeptide)-tag (rmMASP-3-PAs), the latter of which have single amino acid substitution for alanine in the CUB1 or CUB2 domain responsible for binding to LP-PRMs. The mutant rmMASP-3-PAs showed significantly reduced in-vivo complex formation with LP-PRMs when compared with WT rmMASP-3-PA. In the in-vivo kinetic analysis of MASP-3 activation, both WT and mutant rmMASP-3-PAs were cleaved into the active forms as early as 30 minutes in the circulation of mice, and no significant difference in the efficiency of MASP-3 cleavage was observed throughout an observation period of 48 hours after intravenous administration. All sera collected 3 hours after administration of each rmMASP-3-PA showed full restoration of the active FD and AP activity in MASP-3-deficient mouse sera at the same levels as WT mouse sera. Unexpectedly, all mutant rmMASP-3-PAs showed faster clearance from the circulation than the WT rmMASP-3-PA. To our knowledge, the current study is the first to show in-vivo kinetics of MASP-3 demonstrating rapid activation and clearance in the circulation. In conclusion, our results demonstrated that the complex formation of MASP-3 with LP-PRMs is not required for in-vivo activation of MASP-3 or its efficiency, but may contribute to the long-term retention of MASP-3 in the circulation.
Collapse
Affiliation(s)
- Kohei Kusakari
- Department of Immunology, Fukushima Medical University, Fukushima, Japan
| | - Takeshi Machida
- Department of Immunology, Fukushima Medical University, Fukushima, Japan
- *Correspondence: Takeshi Machida,
| | - Yumi Ishida
- Department of Immunology, Fukushima Medical University, Fukushima, Japan
| | - Tomoko Omori
- Department of Immunology, Fukushima Medical University, Fukushima, Japan
| | - Toshiyuki Suzuki
- Radioisotope Research Center, Fukushima Medical University, Fukushima, Japan
| | - Masayuki Sekimata
- Radioisotope Research Center, Fukushima Medical University, Fukushima, Japan
| | - Ikuo Wada
- Department of Cell Science, Institute of Biomedical Sciences, Fukushima Medical University, Fukushima, Japan
| | - Teizo Fujita
- Fukushima Prefectural General Hygiene Institute, Fukushima, Japan
| | - Hideharu Sekine
- Department of Immunology, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
26
|
Gil E, Noursadeghi M, Brown JS. Streptococcus pneumoniae interactions with the complement system. Front Cell Infect Microbiol 2022; 12:929483. [PMID: 35967850 PMCID: PMC9366601 DOI: 10.3389/fcimb.2022.929483] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 07/08/2022] [Indexed: 11/21/2022] Open
Abstract
Host innate and adaptive immunity to infection with Streptococcus pneumoniae is critically dependent on the complement system, demonstrated by the high incidence of invasive S. pneumoniae infection in people with inherited deficiency of complement components. The complement system is activated by S. pneumoniae through multiple mechanisms. The classical complement pathway is activated by recognition of S. pneumoniae by C-reactive protein, serum amyloid P, C1q, SIGN-R1, or natural or acquired antibody. Some S. pneumoniae strains are also recognised by ficolins to activate the mannose binding lectin (MBL) activation pathway. Complement activation is then amplified by the alternative complement pathway, which can also be activated by S. pneumoniae directly. Complement activation results in covalent linkage of the opsonic complement factors C3b and iC3b to the S. pneumoniae surface which promote phagocytic clearance, along with complement-mediated immune adherence to erythrocytes, thereby protecting against septicaemia. The role of complement for mucosal immunity to S. pneumoniae is less clear. Given the major role of complement in controlling infection with S. pneumoniae, it is perhaps unsurprising that S. pneumoniae has evolved multiple mechanisms of complement evasion, including the capsule, multiple surface proteins, and the toxin pneumolysin. There is considerable variation between S. pneumoniae capsular serotypes and genotypes with regards to sensitivity to complement which correlates with ability to cause invasive infections. However, at present we only have a limited understanding of the main mechanisms causing variations in complement sensitivity between S. pneumoniae strains and to non-pathogenic streptococci.
Collapse
Affiliation(s)
- Eliza Gil
- Division of Infection and Immunity, University College London, London, United Kingdom
- *Correspondence: Eliza Gil,
| | - Mahdad Noursadeghi
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Jeremy S. Brown
- Division of Medicine, University College London, London, United Kingdom
| |
Collapse
|
27
|
Salukhov VV, Lopatin YR, Minakov AA. Adipsin – summing up large-scale results: A review. CONSILIUM MEDICUM 2022. [DOI: 10.26442/20751753.2022.5.201280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Adipsin is one of the first discovered adipokines hormones produced by adipose tissue. Adipsin performs the function of a regulator of carbohydrate and lipid metabolism and participates in the adaptation of metabolism to the real needs of the body, being a powerful stimulant of anabolic processes. A characteristic feature of adipsin is that it is also a complement factor D, which is necessary for the normal functioning of an alternative pathway of activation of the complement system. Due to this, adipsin is represented in the body as a link between the energy block of the endocrine system and the humoral block of the immune system. Adipsin is known as a regulator of the function of pancreatic beta cells, a stimulator of lipogenesis, a modulator of inflammation processes. Recently, there have been works indicating the effect of adipsin on the microbiota, as well as its role in non-alcoholic fatty liver disease. To date, there are a large number of publications describing the biochemical structure, functions of adipsin, mechanisms of regulation of its synthesis, as well as changes in the level of adipsin in various pathological conditions. Attempts are also described to pharmacologically influence adipsin in order to modulate its functions or use it as a biomarker for the diagnosis of diseases. However, there is currently no structured review that summarizes and systematizes all available information about this adipokine. This is exactly the task we set ourselves in this study. The paper contains the results of all available studies on adipsin. In some cases, they are contradictory in nature, which indicates the need for further research in detecting connections between the body's systems.
Collapse
|
28
|
Gavriilaki E, Papakonstantinou A, Agrios KA. Novel Insights into Factor D Inhibition. Int J Mol Sci 2022; 23:7216. [PMID: 35806224 PMCID: PMC9267021 DOI: 10.3390/ijms23137216] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/23/2022] [Accepted: 06/23/2022] [Indexed: 01/15/2023] Open
Abstract
Complement-mediated diseases or complementopathies, such as Paroxysmal nocturnal hemoglobinuria (PNH), cold agglutinin disease (CAD), and transplant-associated thrombotic microangiopathy (TA-TMA), demand advanced complement diagnostics and therapeutics be adopted in a vast field of medical specialties, such as hematology, transplantation, rheumatology, and nephrology. The miracle of complement inhibitors as "orphan drugs" has dramatically improved morbidity and mortality in patients with otherwise life-threatening complementopathies. Efficacy has been significantly improved by upstream inhibition in patients with PNH. Different molecules may exert diverse characteristics in vitro and in vivo. Further studies remain to show safety and efficacy of upstream inhibition in other complementopathies. In addition, cost and availability issues are major drawbacks of current treatments. Therefore, further developments are warranted to address the unmet clinical needs in the field of complementopathies. This state-of-the-art narrative review aims to delineate novel insights into factor D inhibition as a promising target for complementopathies.
Collapse
Affiliation(s)
- Eleni Gavriilaki
- Hematology Department, G Papanicolaou Hospital, 57010 Thessaloniki, Greece
| | - Anna Papakonstantinou
- Department of Urology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Konstantinos A. Agrios
- Department of Chemistry, Villanova University, 800 Lancaster Ave., Villanova, PA 19085, USA
| |
Collapse
|
29
|
Gül FC, Kobat SG, Çelik F, Aydin S, Akkoç RF. Plasma and aqueous levels of alarin and adipsin ın patients with and without diabetic retinopathy. BMC Ophthalmol 2022; 22:176. [PMID: 35436912 PMCID: PMC9016965 DOI: 10.1186/s12886-022-02403-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 04/11/2022] [Indexed: 12/28/2022] Open
Abstract
Backround Diabetic retinopathy is a disease seen with microvascular complications as a result of hyperglycemia and insulin resistance. Alarin and Adipsin are molecules with a role in energy and glucose metabolism. The aim of this study was to determine plasma and aqueous levels of Alarin and Adipsin in patients with and without diabetic retinopathy to evaluate their potential roles in diabetic retinopathy. Methods The study included one eye from each of 20 cataract patients without diabetes (C), 20 cataract patients with diabetes and without diabetic retinopathy (DM + C), and 20 cataract patients with diabetes and diabetic retinopathy (DR + C). Plasma and aqueous humour samples were taken from all patients during the cataract operation. Alarin and Adipsin levels were examined with the enzyme-linked immunosorbent assay (ELISA) method. Results Both plasma and aqueous Alarin levels were significantly higher in the patients with diabetic retinopathy than in the control group (p < 0.001, p = 0.006). Adipsin levels were found to be significantly higher in plasma in the control group than in the DR + C group and significantly higher in aqueous in the DR + C group than in the control group (p < 0.001, p < 0.001). Conclusion These findings suggest that Alarin and Adipsin may play important role in diabetic retinopathy.
Collapse
Affiliation(s)
- Fatih Cem Gül
- Medical Doctor Ophthalmology Clinic, Universal Eye Center, Elazig, Turkey.
| | - Sabiha Güngör Kobat
- Medical Doctor Department of Ophthalmology Clinic, Firat University, Elazig, Turkey
| | - Fatih Çelik
- Medical Doctor Ophthalmology Clinic, Elazig City Hospital, Elazig, Turkey
| | - Süleyman Aydin
- Prof Dr Department of Biochemistry, Firat University, Elazig, Turkey
| | | |
Collapse
|
30
|
Milek M, Moulla Y, Kern M, Stroh C, Dietrich A, Schön MR, Gärtner D, Lohmann T, Dressler M, Kovacs P, Stumvoll M, Blüher M, Guiu-Jurado E. Adipsin Serum Concentrations and Adipose Tissue Expression in People with Obesity and Type 2 Diabetes. Int J Mol Sci 2022; 23:ijms23042222. [PMID: 35216336 PMCID: PMC8878597 DOI: 10.3390/ijms23042222] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/02/2022] [Accepted: 02/07/2022] [Indexed: 02/01/2023] Open
Abstract
(1) Adipsin is an adipokine that may link increased fat mass and adipose tissue dysfunction to obesity-related cardiometabolic diseases. Here, we investigated whether adipsin serum concentrations and adipose tissue (AT) adipsin mRNA expression are related to parameters of AT function, obesity and type 2 diabetes (T2D). (2) Methods: A cohort of 637 individuals with a wide range of age and body weight (Age: 18–85 years; BMI: 19–70 kg/m2) with (n = 237) or without (n = 400) T2D was analyzed for serum adipsin concentrations by ELISA and visceral (VAT) and subcutaneous (SAT) adipsin mRNA expression by RT-PCR. (3) Results: Adipsin serum concentrations were significantly higher in patients with T2D compared to normoglycemic individuals. We found significant positive univariate relationships of adipsin serum concentrations with age (r = 0.282, p < 0.001), body weight (r = 0.264, p < 0.001), fasting plasma glucose (r = 0.136, p = 0.006) and leptin serum concentrations (r = 0.362, p < 0.001). Neither VAT nor SAT adipsin mRNA expression correlated with adipsin serum concentrations after adjusting for age, sex and BMI. Independent of T2D status, we found significantly higher adipsin expression in SAT compared to VAT (4) Conclusions: Our data suggest that adipsin serum concentrations are strongly related to obesity and age. However, neither circulating adipsin nor adipsin AT expression reflects parameters of impaired glucose or lipid metabolism in patients with obesity with or without T2D.
Collapse
Affiliation(s)
- Margarete Milek
- Medical Department III—Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany; (M.M.); (P.K.); (M.S.)
| | - Yusef Moulla
- Clinic for Visceral, Transplantation and Thorax and Vascular Surgery, University Hospital Leipzig, 04103 Leipzig, Germany; (Y.M.); (A.D.)
| | - Matthias Kern
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany;
| | - Christine Stroh
- Department of General, Abdominal and Pediatric Surgery, Municipal Hospital, 07548 Gera, Germany;
| | - Arne Dietrich
- Clinic for Visceral, Transplantation and Thorax and Vascular Surgery, University Hospital Leipzig, 04103 Leipzig, Germany; (Y.M.); (A.D.)
| | - Michael R Schön
- Städtisches Klinikum Karlsruhe, Clinic of Visceral Surgery, 76133 Karlsruhe, Germany; (M.R.S.); (D.G.)
| | - Daniel Gärtner
- Städtisches Klinikum Karlsruhe, Clinic of Visceral Surgery, 76133 Karlsruhe, Germany; (M.R.S.); (D.G.)
| | - Tobias Lohmann
- Municipal Clinic Dresden-Neustadt, 01129 Dresden, Germany; (T.L.); (M.D.)
| | - Miriam Dressler
- Municipal Clinic Dresden-Neustadt, 01129 Dresden, Germany; (T.L.); (M.D.)
| | - Peter Kovacs
- Medical Department III—Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany; (M.M.); (P.K.); (M.S.)
| | - Michael Stumvoll
- Medical Department III—Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany; (M.M.); (P.K.); (M.S.)
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany;
| | - Matthias Blüher
- Medical Department III—Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany; (M.M.); (P.K.); (M.S.)
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany;
- Correspondence: (M.B.); (E.G.-J.); Tel.: +49-341-972-2901 (M.B.); +49-341-971-5895 (E.G.-J.)
| | - Esther Guiu-Jurado
- Medical Department III—Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany; (M.M.); (P.K.); (M.S.)
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany;
- Deutsches Zentrum für Diabetesforschung e.V., 85764 Oberschleißheim, Germany
- Correspondence: (M.B.); (E.G.-J.); Tel.: +49-341-972-2901 (M.B.); +49-341-971-5895 (E.G.-J.)
| |
Collapse
|
31
|
Al-Muraikhy S, Ramanjaneya M, Dömling AS, Bettahi I, Donati F, Botre F, Abou-Samra AB, Sellami M, Elrayess MA. High Endurance Elite Athletes Show Age-dependent Lower Levels of Circulating Complements Compared to Low/Moderate Endurance Elite Athletes. Front Mol Biosci 2021; 8:715035. [PMID: 34631796 PMCID: PMC8494969 DOI: 10.3389/fmolb.2021.715035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 09/06/2021] [Indexed: 12/19/2022] Open
Abstract
Introduction: Aerobic exercise activates the complement system in the peripheral blood. However, the effect of age and high intensity endurance training on the levels of circulating complements and sassociated inflammatory cytokines, oxidative stress markers and cellular aging remains unknown. Methods: In this study, serum samples from 79 elite athletes who belong to high (n = 48) and low/moderate (n = 31) endurance sports and two age groups (below 30 years old, n = 53, and above 30 years old, n = 26) were profiled for 14 complements. Linear models were used to assess differences in complements levels between sport and age groups. Spearmann’s correlation was used to assess the relationship among detected complements and proinflammatory cytokines, oxidative stress markers and telomere lengths. Results: High endurance elite athletes exhibited significantly lower levels of circulating C2, C3b/iC3b and adipsin complements than their age-matched low/moderate endurance counterparts. Levels of C2, adipsin and C3b/iC3b were positively correlated with most detected complements, the pro-inflammatory cytokines TNF-alpha and IL-22 and the anti-oxidant enzyme catalase. However, they were negatively correlated with telomere length only in younger elite athletes regardless of their sport groups. Furthermore, high endurance elite athletes showed significantly lower concentrations of C3b/iC3b, C4b, C5, C5a, C1q, C3, C4, factor H and properdin in younger athletes compared to their older counterparts. Conclusion: Our novel data suggest that high endurance elite athletes exhibit age-independent lower levels of circulating C2, C3b/iC3b and adipsin, associated with lower inflammatory, oxidative stress and cellular aging, as well as lower levels of 10 other complements in younger athletes compared to older counterparts. Assessing the effect of various levels of endurance sports on complements-based immune response provides a better understanding of exercise physiology and pathophysiology of elite athletes.
Collapse
Affiliation(s)
- Shamma Al-Muraikhy
- Biomedical Research Center, Qatar University, Doha, Qatar.,Department of Drug Design, University of Groningen, Groningen, Netherlands
| | - Manjunath Ramanjaneya
- Qatar Metabolic Institute, Hamad Medical Corporation, Doha, Qatar.,Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | | | - Ilham Bettahi
- Qatar Metabolic Institute, Hamad Medical Corporation, Doha, Qatar.,Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Francesco Donati
- Laboratorio Antidoping, Federazione Medico Sportiva Italiana, Rome, Italy
| | - Francesco Botre
- Laboratorio Antidoping, Federazione Medico Sportiva Italiana, Rome, Italy
| | | | - Maha Sellami
- Physical Education Department, College of Education, Qatar University, Doha, Qatar
| | | |
Collapse
|
32
|
Meeks KAC, Bentley AR, Gouveia MH, Chen G, Zhou J, Lei L, Adeyemo AA, Doumatey AP, Rotimi CN. Genome-wide analyses of multiple obesity-related cytokines and hormones informs biology of cardiometabolic traits. Genome Med 2021; 13:156. [PMID: 34620218 PMCID: PMC8499470 DOI: 10.1186/s13073-021-00971-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 09/16/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND A complex set of perturbations occur in cytokines and hormones in the etiopathogenesis of obesity and related cardiometabolic conditions such as type 2 diabetes (T2D). Evidence for the genetic regulation of these cytokines and hormones is limited, particularly in African-ancestry populations. In order to improve our understanding of the biology of cardiometabolic traits, we investigated the genetic architecture of a large panel of obesity- related cytokines and hormones among Africans with replication analyses in African Americans. METHODS We performed genome-wide association studies (GWAS) in 4432 continental Africans, enrolled from Ghana, Kenya, and Nigeria as part of the Africa America Diabetes Mellitus (AADM) study, for 13 obesity-related cytokines and hormones, including adipsin, glucose-dependent insulinotropic peptide (GIP), glucagon-like peptide-1 (GLP-1), interleukin-1 receptor antagonist (IL1-RA), interleukin-6 (IL-6), interleukin-10 (IL-10), leptin, plasminogen activator inhibitor-1 (PAI-1), resistin, visfatin, insulin, glucagon, and ghrelin. Exact and local replication analyses were conducted in African Americans (n = 7990). The effects of sex, body mass index (BMI), and T2D on results were investigated through stratified analyses. RESULTS GWAS identified 39 significant (P value < 5 × 10-8) loci across all 13 traits. Notably, 14 loci were African-ancestry specific. In this first GWAS for adipsin and ghrelin, we detected 13 and 4 genome-wide significant loci respectively. Stratified analyses by sex, BMI, and T2D showed a strong effect of these variables on detected loci. Eight novel loci were successfully replicated: adipsin (3), GIP (1), GLP-1 (1), and insulin (3). Annotation of these loci revealed promising links between these adipocytokines and cardiometabolic outcomes as illustrated by rs201751833 for adipsin and blood pressure and locus rs759790 for insulin level and T2D in lean individuals. CONCLUSIONS Our study identified genetic variants underlying variation in multiple adipocytokines, including the first loci for adipsin and ghrelin. We identified population differences in variants associated with adipocytokines and highlight the importance of stratification for discovery of loci. The high number of African-specific loci detected emphasizes the need for GWAS in African-ancestry populations, as these loci could not have been detected in other populations. Overall, our work contributes to the understanding of the biology linking adipocytokines to cardiometabolic traits.
Collapse
Affiliation(s)
- Karlijn A C Meeks
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive Bldg 12A rm 4047, Bethesda, MD, 20814, USA
| | - Amy R Bentley
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive Bldg 12A rm 4047, Bethesda, MD, 20814, USA
| | - Mateus H Gouveia
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive Bldg 12A rm 4047, Bethesda, MD, 20814, USA
| | - Guanjie Chen
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive Bldg 12A rm 4047, Bethesda, MD, 20814, USA
| | - Jie Zhou
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive Bldg 12A rm 4047, Bethesda, MD, 20814, USA
| | - Lin Lei
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive Bldg 12A rm 4047, Bethesda, MD, 20814, USA
| | - Adebowale A Adeyemo
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive Bldg 12A rm 4047, Bethesda, MD, 20814, USA
| | - Ayo P Doumatey
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive Bldg 12A rm 4047, Bethesda, MD, 20814, USA.
| | - Charles N Rotimi
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive Bldg 12A rm 4047, Bethesda, MD, 20814, USA.
| |
Collapse
|
33
|
Nan X, Teng Y, Tian J, Hu Z, Fang Q. A comprehensive assessment of the biocompatibility of Magnetospirillum gryphiswaldense MSR-1 bacterial magnetosomes in vitro and in vivo. Toxicology 2021; 462:152949. [PMID: 34534559 DOI: 10.1016/j.tox.2021.152949] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/01/2021] [Accepted: 09/08/2021] [Indexed: 10/20/2022]
Abstract
Bacterial magnetosomes (BMs) are iron oxide nanoparticles synthesized naturally by magnetotactic bacteria, made up of nano-sized inorganic crystals enclosed by a lipid bilayer membrane. Due to several superior characteristics, such as the narrow size distribution, uniform morphology, high purity and crystallinity, single magnetic domain as well as easy surface modification, increasing biomedical and biotechnological applications of BMs have been developed. The attracted wide attentions raise the urge for the evaluation of safety and toxicity. In this work, we performed a rather comprehensive and systematic assessment of in vitro and in vivo toxicity of BMs from MSR-1, including the cytotoxicity, mice bodyweights, blood test, organ coefficients, inflammation, and hemocompatibility study. We found that BMs have good biocompatibility except for influences on the immune response as demonstrated by enhanced activation of the complement system and inhibition of lymphocyte proliferation when used with an excessive concentration. BMs induced the production of reactive oxygen species (ROS) in macrophages at a dose-dependent manner but did not cause cell membrane damage and cell cycle arrest until the concentration is approximately 40 times the clinical dosage. We anticipate our work will guide modifications of BMs and expand their future applications.
Collapse
Affiliation(s)
- Xiaohui Nan
- Laboratory of Theoretical and Computational Nanoscience, CAS Key Laboratory of Nanophotonic Materials and Devices, CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, Beijing Key Laboratory of Ambient Particles Health Effects and Prevention Techniques, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, 100190, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yan Teng
- Wedocare Inc, Beijing, 100190, China
| | - Jiesheng Tian
- State Key Laboratories for Agrobiotechnology and College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Zhiyuan Hu
- Laboratory of Theoretical and Computational Nanoscience, CAS Key Laboratory of Nanophotonic Materials and Devices, CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, Beijing Key Laboratory of Ambient Particles Health Effects and Prevention Techniques, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, 100190, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing, 100190, China.
| | - Qiaojun Fang
- Laboratory of Theoretical and Computational Nanoscience, CAS Key Laboratory of Nanophotonic Materials and Devices, CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, Beijing Key Laboratory of Ambient Particles Health Effects and Prevention Techniques, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, 100190, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing, 100190, China.
| |
Collapse
|
34
|
Advancing therapeutic complement inhibition in hematologic diseases: PNH and beyond. Blood 2021; 139:3571-3582. [PMID: 34482398 DOI: 10.1182/blood.2021012860] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/11/2021] [Indexed: 11/20/2022] Open
Abstract
Complement is an elaborate system of the innate immunity. Genetic variants and autoantibodies leading to excessive complement activation are implicated in a variety of human diseases. Among them, the hematologic disease paroxysmal nocturnal hemoglobinuria (PNH) remains the prototype model of complement activation and inhibition. Eculizumab, the first-in-class complement inhibitor, was approved for PNH in 2007. Addressing some of the unmet needs, a long-acting C5 inhibitor, ravulizumab, and a C3 inhibitor, pegcetacoplan have been also now approved with PNH. Novel agents, such as factor B and factor D inhibitors, are under study with very promising results. In this era of several approved targeted complement therapeutics, selection of the proper drug needs to be based on a personalized approach. Beyond PNH, complement inhibition has also shown efficacy and safety in cold agglutinin disease (CAD), primarily with the C1s inhibitor of the classical complement pathway, sutimlimab, but also with pegcetacoplan. Furthermore, C5 inhibition with eculizumab and ravulizumab, as well as inhibition of the lectin pathway with narsoplimab, are investigated in transplant-associated thrombotic microangiopathy (TA-TMA). With this revolution of next-generation complement therapeutics, additional hematologic entities, such as delayed hemolytic transfusion reaction (DHTR) or immune thrombocytopenia (ITP), might also benefit from complement inhibitors. Therefore, this review aims to describe state-of-the-art knowledge of targeting complement in hematologic diseases focusing on: a) complement biology for the clinician, b) complement activation and therapeutic inhibition in prototypical complement-mediated hematologic diseases, c) hematologic entities under investigation for complement inhibition, and d) other complement-related disorders of potential interest to hematologists.
Collapse
|
35
|
Sommerfeld O, Dahlke K, Sossdorf M, Claus RA, Scherag A, Bauer M, Bloos F. Complement factor D is linked to platelet activation in human and rodent sepsis. Intensive Care Med Exp 2021; 9:41. [PMID: 34396466 PMCID: PMC8364893 DOI: 10.1186/s40635-021-00405-8] [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: 03/10/2021] [Accepted: 07/20/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The complement factor D (CFD) exerts a regulatory role during infection. However, its physiological function in coagulopathy and its impact on the course of an infection remains unclear. MATERIALS Wild-type and CFD-deficient mice (n = 91) were subjected to cecal ligation and puncture to induce sepsis. At several time points, markers of coagulation and the host-immune response were determined. Furthermore, in patients (n = 79) with sepsis or SIRS, CFD levels were related to clinical characteristics, use of antiplatelet drugs and outcome. RESULTS Septic CFD-deficient mice displayed higher TAT complexes (p = 0.02), impaired maximal clot firmness, but no relevant platelet drop and reduced GPIIb/IIIa surface expression on platelets (p = 0.03) compared to septic wild-type mice. In humans, higher CFD levels (non-survivors, 5.0 µg/ml to survivors, 3.6 µg/ml; p = 0.015) were associated with organ failure (SOFA score: r = 0.33; p = 0.003) and mortality (75% percentile, 61.1% to 25% percentile, 26.3%). CFD level was lower in patients with antiplatelet drugs (4.5-5.3 µg/ml) than in patients without. CONCLUSION In mice, CFD is linked to pronounced platelet activation, depicted by higher GPIIb/IIIa surface expression in wild-type mice. This might be of clinical importance since high CFD plasma concentrations were also associated with increased mortality in sepsis patients.
Collapse
Affiliation(s)
- O Sommerfeld
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany. .,Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany.
| | - K Dahlke
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany.,Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | - M Sossdorf
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany.,Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | - R A Claus
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany.,Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | - A Scherag
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany.,Institute of Medical Statistics, Computer and Data Sciences, Jena University Hospital, Jena, Germany
| | - M Bauer
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany.,Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | - F Bloos
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany. .,Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany.
| |
Collapse
|
36
|
Ma L, Sahu SK, Cano M, Kuppuswamy V, Bajwa J, McPhatter J, Pine A, Meizlish ML, Goshua G, Chang CH, Zhang H, Price C, Bahel P, Rinder H, Lei T, Day A, Reynolds D, Wu X, Schriefer R, Rauseo AM, Goss CW, O’Halloran JA, Presti RM, Kim AH, Gelman AE, Dela Cruz CS, Lee AI, Mudd PA, Chun HJ, Atkinson JP, Kulkarni HS. Increased complement activation is a distinctive feature of severe SARS-CoV-2 infection. Sci Immunol 2021; 6:eabh2259. [PMID: 34446527 PMCID: PMC8158979 DOI: 10.1126/sciimmunol.abh2259] [Citation(s) in RCA: 150] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 05/11/2021] [Indexed: 12/13/2022]
Abstract
Complement activation has been implicated in the pathogenesis of severe SARS-CoV-2 infection. However, it remains to be determined whether increased complement activation is a broad indicator of critical illness (and thus, no different in COVID-19). It is also unclear which pathways are contributing to complement activation in COVID-19, and if complement activation is associated with certain features of severe SARS-CoV-2 infection, such as endothelial injury and hypercoagulability. To address these questions, we investigated complement activation in the plasma from patients with COVID-19 prospectively enrolled at two tertiary care centers: Washington University School of Medicine (n=134) and Yale School of Medicine (n=49). We compared our patients to two non-COVID cohorts: (a) patients hospitalized with influenza (n=54), and (b) patients admitted to the intensive care unit (ICU) with acute respiratory failure requiring invasive mechanical ventilation (IMV, n=22). We demonstrate that circulating markers of complement activation are elevated in patients with COVID-19 compared to those with influenza and to patients with non-COVID-19 respiratory failure. Further, the results facilitate distinguishing those who are at higher risk of worse outcomes such as requiring ICU admission, or IMV. Moreover, the results indicate enhanced activation of the alternative complement pathway is most prevalent in patients with severe COVID-19 and is associated with markers of endothelial injury (i.e., angiopoietin-2) as well as hypercoagulability (i.e., thrombomodulin and von Willebrand factor). Our findings identify complement activation to be a distinctive feature of COVID-19, and provide specific targets that may be utilized for risk prognostication, drug discovery and personalized clinical trials.
Collapse
Affiliation(s)
- Lina Ma
- Division of Pulmonary and Critical Care Medicine, John T. Milliken Department of Medicine, Washington University School of Medicine; St. Louis, USA
| | - Sanjaya K. Sahu
- Division of Pulmonary and Critical Care Medicine, John T. Milliken Department of Medicine, Washington University School of Medicine; St. Louis, USA
| | - Marlene Cano
- Division of Pulmonary and Critical Care Medicine, John T. Milliken Department of Medicine, Washington University School of Medicine; St. Louis, USA
| | - Vasanthan Kuppuswamy
- Division of Hospital Medicine, John T. Milliken Department of Medicine, Washington University School of Medicine; St. Louis, USA
| | - Jamal Bajwa
- Division of Pulmonary and Critical Care Medicine, John T. Milliken Department of Medicine, Washington University School of Medicine; St. Louis, USA
- Marian University; Indianapolis, USA
| | - Ja’Nia McPhatter
- Division of Pulmonary and Critical Care Medicine, John T. Milliken Department of Medicine, Washington University School of Medicine; St. Louis, USA
- University of Pittsburgh; Pittsburgh, USA
| | - Alexander Pine
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine; New Haven, USA
| | | | - George Goshua
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine; New Haven, USA
| | - C-Hong Chang
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine; New Haven, USA
| | - Hanming Zhang
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine; New Haven, USA
| | - Christina Price
- Section of Immunology, Department of Internal Medicine, Yale School of Medicine; New Haven, USA
| | | | | | - Tingting Lei
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, USA
| | - Aaron Day
- Department of Emergency Medicine, Washington University School of Medicine; St. Louis, USA
| | - Daniel Reynolds
- Division of Pulmonary and Critical Care Medicine, John T. Milliken Department of Medicine, Washington University School of Medicine; St. Louis, USA
| | - Xiaobo Wu
- Division of Rheumatology, John T. Milliken Department of Medicine, Washington University School of Medicine; St. Louis, USA
| | - Rebecca Schriefer
- Division of Rheumatology, John T. Milliken Department of Medicine, Washington University School of Medicine; St. Louis, USA
| | - Adriana M. Rauseo
- Division of Infectious Diseases, John T. Milliken Department of Medicine, Washington University School of Medicine; St. Louis, USA
| | - Charles W. Goss
- Division of Biostatistics, Washington University School of Medicine; St. Louis, USA
| | - Jane A. O’Halloran
- Division of Infectious Diseases, John T. Milliken Department of Medicine, Washington University School of Medicine; St. Louis, USA
| | - Rachel M. Presti
- Division of Infectious Diseases, John T. Milliken Department of Medicine, Washington University School of Medicine; St. Louis, USA
| | - Alfred H. Kim
- Division of Rheumatology, John T. Milliken Department of Medicine, Washington University School of Medicine; St. Louis, USA
| | - Andrew E. Gelman
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, USA
- Division of Biostatistics, Washington University School of Medicine; St. Louis, USA
| | - Charles S. Dela Cruz
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine; New Haven, USA
| | - Alfred I. Lee
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine; New Haven, USA
| | - Philip A. Mudd
- Department of Emergency Medicine, Washington University School of Medicine; St. Louis, USA
| | - Hyung J. Chun
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine; New Haven, USA
| | - John P. Atkinson
- Division of Rheumatology, John T. Milliken Department of Medicine, Washington University School of Medicine; St. Louis, USA
| | - Hrishikesh S. Kulkarni
- Division of Pulmonary and Critical Care Medicine, John T. Milliken Department of Medicine, Washington University School of Medicine; St. Louis, USA
| |
Collapse
|
37
|
Suvarna K, Biswas D, Pai MGJ, Acharjee A, Bankar R, Palanivel V, Salkar A, Verma A, Mukherjee A, Choudhury M, Ghantasala S, Ghosh S, Singh A, Banerjee A, Badaya A, Bihani S, Loya G, Mantri K, Burli A, Roy J, Srivastava A, Agrawal S, Shrivastav O, Shastri J, Srivastava S. Proteomics and Machine Learning Approaches Reveal a Set of Prognostic Markers for COVID-19 Severity With Drug Repurposing Potential. Front Physiol 2021; 12:652799. [PMID: 33995121 PMCID: PMC8120435 DOI: 10.3389/fphys.2021.652799] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 03/12/2021] [Indexed: 12/13/2022] Open
Abstract
The pestilential pathogen SARS-CoV-2 has led to a seemingly ceaseless pandemic of COVID-19. The healthcare sector is under a tremendous burden, thus necessitating the prognosis of COVID-19 severity. This in-depth study of plasma proteome alteration provides insights into the host physiological response towards the infection and also reveals the potential prognostic markers of the disease. Using label-free quantitative proteomics, we performed deep plasma proteome analysis in a cohort of 71 patients (20 COVID-19 negative, 18 COVID-19 non-severe, and 33 severe) to understand the disease dynamics. Of the 1200 proteins detected in the patient plasma, 38 proteins were identified to be differentially expressed between non-severe and severe groups. The altered plasma proteome revealed significant dysregulation in the pathways related to peptidase activity, regulated exocytosis, blood coagulation, complement activation, leukocyte activation involved in immune response, and response to glucocorticoid biological processes in severe cases of SARS-CoV-2 infection. Furthermore, we employed supervised machine learning (ML) approaches using a linear support vector machine model to identify the classifiers of patients with non-severe and severe COVID-19. The model used a selected panel of 20 proteins and classified the samples based on the severity with a classification accuracy of 0.84. Putative biomarkers such as angiotensinogen and SERPING1 and ML-derived classifiers including the apolipoprotein B, SERPINA3, and fibrinogen gamma chain were validated by targeted mass spectrometry-based multiple reaction monitoring (MRM) assays. We also employed an in silico screening approach against the identified target proteins for the therapeutic management of COVID-19. We shortlisted two FDA-approved drugs, namely, selinexor and ponatinib, which showed the potential of being repurposed for COVID-19 therapeutics. Overall, this is the first most comprehensive plasma proteome investigation of COVID-19 patients from the Indian population, and provides a set of potential biomarkers for the disease severity progression and targets for therapeutic interventions.
Collapse
Affiliation(s)
- Kruthi Suvarna
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Deeptarup Biswas
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Medha Gayathri J. Pai
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Arup Acharjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Renuka Bankar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Viswanthram Palanivel
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Akanksha Salkar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Ayushi Verma
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Amrita Mukherjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Manisha Choudhury
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Saicharan Ghantasala
- Centre for Research in Nanotechnology and Sciences, Indian Institute of Technology Bombay, Mumbai, India
| | - Susmita Ghosh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Avinash Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Arghya Banerjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Apoorva Badaya
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai, India
| | - Surbhi Bihani
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Gaurish Loya
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Krishi Mantri
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Ananya Burli
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Jyotirmoy Roy
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Alisha Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
- Department of Genetics, University of Delhi, New Delhi, India
| | - Sachee Agrawal
- Kasturba Hospital for Infectious Diseases, Mumbai, India
| | - Om Shrivastav
- Kasturba Hospital for Infectious Diseases, Mumbai, India
| | | | - Sanjeeva Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| |
Collapse
|
38
|
Sun R, Qiao Y, Yan G, Wang D, Zuo W, Ji Z, Zhang X, Yao Y, Ma G, Tang C. Association between serum adipsin and plaque vulnerability determined by optical coherence tomography in patients with coronary artery disease. J Thorac Dis 2021; 13:2414-2425. [PMID: 34012589 PMCID: PMC8107545 DOI: 10.21037/jtd-21-259] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Background Early identification of vulnerable plaques is important for patients with coronary artery disease (CAD) to reduce acute coronary events and improve their prognosis. We sought to examine the relationship between adipsin, an adipokine secreted from adipocytes, and plaque vulnerability in CAD patients. Methods A total of 103 plaques from 99 consecutive patients who underwent coronary angiography were assessed by optical coherence tomography. The serum level of adipsin was measured using enzyme-linked immunosorbent assay (ELISA). The accuracy of adipsin for detecting thin-cap fibroatheroma (TCFA) was determined by the area under the receiver operating characteristic curve (AUC). Results Of the 99 patients, 49 were classified into the low adipsin group and 50 into the high adipsin group according to the median level of serum adipsin (2.43 µg/mL). The plaques from the high adipsin group exhibited a greater lipid index (2,700.0 vs. 1,975.9° × mm, P=0.015) and an increased proportion of TCFAs (41.2% vs. 21.2%, P=0.028) compared with the low adipsin group. Serum adipsin was found to be negatively correlated with fibrous cap thickness (ρ=−0.322, P=0.002), while it was positively correlated with average lipid arc (ρ=0.253, P=0.015), maximum lipid arc (ρ=0.211, P=0.044), lipid core length (ρ=0.241, P=0.021), lipid index (ρ=0.335, P=0.001), and vulnerability score (ρ=0.254, P=0.014). Furthermore, adipsin had a significant association with TCFAs (OR: 1.290, 95% CI: 1.048–1.589, P=0.016) in the multivariate analysis, while having a moderate diagnostic accuracy for TCFAs (AUC: 0.710, 95% CI: 0.602–0.817, P<0.001). Conclusions Our findings suggest that serum adipsin is significantly and positively correlated with the incidence of TCFAs. The application of adipsin as a biomarker may offer improvement in the diagnosis of vulnerable plaques and clinical benefits for CAD patients.
Collapse
Affiliation(s)
- Renhua Sun
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China.,Department of Cardiology, Yancheng No.1 People's Hospital, Yancheng, China
| | - Yong Qiao
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Gaoliang Yan
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Dong Wang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Wenjie Zuo
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Zhenjun Ji
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Xiaoguo Zhang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Yuyu Yao
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Genshan Ma
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Chengchun Tang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
39
|
Weiss SAI, Rehm SRT, Perera NC, Biniossek ML, Schilling O, Jenne DE. Origin and Expansion of the Serine Protease Repertoire in the Myelomonocyte Lineage. Int J Mol Sci 2021; 22:ijms22041658. [PMID: 33562184 PMCID: PMC7914634 DOI: 10.3390/ijms22041658] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/26/2021] [Accepted: 02/04/2021] [Indexed: 02/07/2023] Open
Abstract
The deepest evolutionary branches of the trypsin/chymotrypsin family of serine proteases are represented by the digestive enzymes of the gastrointestinal tract and the multi-domain proteases of the blood coagulation and complement system. Similar to the very old digestive system, highly diverse cleavage specificities emerged in various cell lineages of the immune defense system during vertebrate evolution. The four neutrophil serine proteases (NSPs) expressed in the myelomonocyte lineage, neutrophil elastase, proteinase 3, cathepsin G, and neutrophil serine protease 4, collectively display a broad repertoire of (S1) specificities. The origin of NSPs can be traced back to a circulating liver-derived trypsin-like protease, the complement factor D ancestor, whose activity is tightly controlled by substrate-induced activation and TNFα-induced locally upregulated protein secretion. However, the present-day descendants are produced and converted to mature enzymes in precursor cells of the bone marrow and are safely sequestered in granules of circulating neutrophils. The potential site and duration of action of these cell-associated serine proteases are tightly controlled by the recruitment and activation of neutrophils, by stimulus-dependent regulated secretion of the granules, and by various soluble inhibitors in plasma, interstitial fluids, and in the inflammatory exudate. An extraordinary dynamic range and acceleration of immediate defense responses have been achieved by exploiting the high structural plasticity of the trypsin fold.
Collapse
Affiliation(s)
- Stefanie A. I. Weiss
- Comprehensive Pneumology Center (CPC-M), Institute of Lung Biology and Disease (iLBD) Helmholtz Zentrum München and University Hospital of the Ludwig-Maximilians University (LMU), 81377 Munich, Germany; (S.A.I.W.); (S.R.T.R.)
| | - Salome R. T. Rehm
- Comprehensive Pneumology Center (CPC-M), Institute of Lung Biology and Disease (iLBD) Helmholtz Zentrum München and University Hospital of the Ludwig-Maximilians University (LMU), 81377 Munich, Germany; (S.A.I.W.); (S.R.T.R.)
| | | | - Martin L. Biniossek
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany;
| | - Oliver Schilling
- Institute of Surgical Pathology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany;
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Dieter E. Jenne
- Comprehensive Pneumology Center (CPC-M), Institute of Lung Biology and Disease (iLBD) Helmholtz Zentrum München and University Hospital of the Ludwig-Maximilians University (LMU), 81377 Munich, Germany; (S.A.I.W.); (S.R.T.R.)
- Max Planck Institute of Neurobiology, 82152 Planegg-Martinsried, Germany
- Correspondence:
| |
Collapse
|
40
|
Aaron N, Kraakman MJ, Zhou Q, Liu Q, Costa S, Yang J, Liu L, Yu L, Wang L, He Y, Fan L, Hirakawa H, Ding L, Lo J, Wang W, Zhao B, Guo E, Sun L, Rosen CJ, Qiang L. Adipsin promotes bone marrow adiposity by priming mesenchymal stem cells. eLife 2021; 10:69209. [PMID: 34155972 PMCID: PMC8219379 DOI: 10.7554/elife.69209] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 06/07/2021] [Indexed: 01/12/2023] Open
Abstract
Background Marrow adipose tissue (MAT) has been shown to be vital for regulating metabolism and maintaining skeletal homeostasis in the bone marrow (BM) niche. As a reflection of BM remodeling, MAT is highly responsive to nutrient fluctuations, hormonal changes, and metabolic disturbances such as obesity and diabetes mellitus. Expansion of MAT has also been strongly associated with bone loss in mice and humans. However, the regulation of BM plasticity remains poorly understood, as does the mechanism that links changes in marrow adiposity with bone remodeling. Methods We studied deletion of Adipsin, and its downstream effector, C3, in C57BL/6 mice as well as the bone-protected PPARγ constitutive deacetylation 2KR mice to assess BM plasticity. The mice were challenged with thiazolidinedione treatment, calorie restriction, or aging to induce bone loss and MAT expansion. Analysis of bone mineral density and marrow adiposity was performed using a μCT scanner and by RNA analysis to assess adipocyte and osteoblast markers. For in vitro studies, primary bone marrow stromal cells were isolated and subjected to osteoblastogenic or adipogenic differentiation or chemical treatment followed by morphological and molecular analyses. Clinical data was obtained from samples of a previous clinical trial of fasting and high-calorie diet in healthy human volunteers. Results We show that Adipsin is the most upregulated adipokine during MAT expansion in mice and humans in a PPARγ acetylation-dependent manner. Genetic ablation of Adipsin in mice specifically inhibited MAT expansion but not peripheral adipose depots, and improved bone mass during calorie restriction, thiazolidinedione treatment, and aging. These effects were mediated through its downstream effector, complement component C3, to prime common progenitor cells toward adipogenesis rather than osteoblastogenesis through inhibiting Wnt/β-catenin signaling. Conclusions Adipsin promotes new adipocyte formation and affects skeletal remodeling in the BM niche. Our study reveals a novel mechanism whereby the BM sustains its own plasticity through paracrine and endocrine actions of a unique adipokine. Funding This work was supported by the National Institutes of Health T32DK007328 (NA), F31DK124926 (NA), R01DK121140 (JCL), R01AR068970 (BZ), R01AR071463 (BZ), R01DK112943 (LQ), R24DK092759 (CJR), and P01HL087123 (LQ).
Collapse
Affiliation(s)
- Nicole Aaron
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Pharmacology, Columbia UniversityNew YorkUnited States
| | - Michael J Kraakman
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Medicine, Columbia UniversityNew YorkUnited States
| | - Qiuzhong Zhou
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical SchoolSingaporeSingapore
| | - Qiongming Liu
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Pathology and Cellular Biology, Columbia UniversityNew YorkUnited States
| | - Samantha Costa
- Center for Molecular Medicine, Maine Medical Center Research InstituteScarboroughUnited States,School of Medicine, Tufts UniversityBostonUnited States,Graduate School of Biomedical Science and Engineering, University of MaineOronoUnited States
| | - Jing Yang
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Pathology and Cellular Biology, Columbia UniversityNew YorkUnited States
| | - Longhua Liu
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Pathology and Cellular Biology, Columbia UniversityNew YorkUnited States
| | - Lexiang Yu
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Pathology and Cellular Biology, Columbia UniversityNew YorkUnited States
| | - Liheng Wang
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Medicine, Columbia UniversityNew YorkUnited States
| | - Ying He
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Pathology and Cellular Biology, Columbia UniversityNew YorkUnited States
| | - Lihong Fan
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Pathology and Cellular Biology, Columbia UniversityNew YorkUnited States
| | - Hiroyuki Hirakawa
- Department of Microbiology and Immunology, Columbia UniversityNew YorkUnited States,Department of Rehabilitation and Regenerative Medicine, Vagelos College of Physicians and SurgeonsNew YorkUnited States
| | - Lei Ding
- Department of Microbiology and Immunology, Columbia UniversityNew YorkUnited States,Department of Rehabilitation and Regenerative Medicine, Vagelos College of Physicians and SurgeonsNew YorkUnited States
| | - James Lo
- Weill Center for Metabolic Health, Cardiovascular Research Institute, and Division of Cardiology, Weill Cornell Medical CollegeNew YorkUnited States
| | - Weidong Wang
- Department of Medicine, Division of Endocrinology, Harold Hamm Diabetes Center, The University of Oklahoma Health Science CenterOklahoma CityUnited States
| | - Baohong Zhao
- Arthritis and Tissue Degeneration Program and The David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, Department of Medicine, Weill Cornell Medical College; Graduate Program in Cell & Developmental Biology, Weill Cornell Graduate School of Medical SciencesNew YorkUnited States
| | - Edward Guo
- Department of Biomedical Engineering, Columbia UniversityNew YorkUnited States
| | - Lei Sun
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical SchoolSingaporeSingapore
| | - Cliff J Rosen
- Center for Molecular Medicine, Maine Medical Center Research InstituteScarboroughUnited States
| | - Li Qiang
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Pathology and Cellular Biology, Columbia UniversityNew YorkUnited States
| |
Collapse
|
41
|
Alshubrami S, Al-Regaiey K, Alfadda AA, Iqbal M. Impact of Gastric Sleeve Surgery on Plasma Retinol Binding Protein 4 and Adipsin Levels in Healthy Male Population. Pak J Med Sci 2020; 36:1495-1499. [PMID: 33235563 PMCID: PMC7674898 DOI: 10.12669/pjms.36.7.2329] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Objectives Bariatric surgery provides most substantial and sustainable weight loss measures in individuals with obesity. Caloric restriction is not only intervention, changes in hormonal secretions are also leading contributory mechanisms to reduce body weight and improve the glycaemic control. The aim of this study was to evaluate the impact of gastric sleeve surgery on plasma retinol binding protein 4 (RBP4) and adipsin levels among Saudi male obese population. Methods This prospective study was conducted in the Departments of Physiology and Surgery, College of Medicine, King Saud University. Thirty-three obese (BMI>38.3) male patients age ranged from 25 to 50 years were recruited. RBP4 and adipsin levels were analyzed before and 6-12 months after gastric sleeve surgery by ELISA along with plasma glucose, insulin, HOMA-IR and lipid profile. Results Circulating RBP4 levels were not significantly changed by bariatric surgery (4382.85±40.35 ng before, and 4393.28±33.13 ng after surgery, p=0.842), neither did adipsin (2949.68±46.86 pg before, and 2917.90±41.90 pg after surgery, p=0.535). Segregation of study participants into two age groups, 25-35 and 35-50 years of age, revealed that before surgery older age group (35-50) had higher RBP4 levels compared to younger group (25-35) (p=0.016). However, after surgery RBP4 levels were decreased in older group but not to a significant level (p=0.174). In younger age group after surgery, there was a near significant increase in RBP4 levels (p=0.052). There were no significant changes in RBP4 levels in both age groups after surgery (p=0.461). For adipsin, there were no significant differences before and after surgery in both age groups. Insulin, BMI and HOMA-IR index were decreased after surgery, however there was no correlation with RBP4 and adipsin levels. Conclusions The present study findings do not suggest a role for RBP4 and adipsin in the improvement of insulin sensitivity in Saudi male obese population after gastric sleeve surgery. However, a decrease in RBP4 levels in older individuals after surgery needs further investigations to understand its effect on weight and glycemic control.
Collapse
Affiliation(s)
- Suad Alshubrami
- Suad Alshubrami Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia. Current Address: Director of Academic and Training Affairs Continuous Professional, King Salman Specialist Hospital, Hail, Saudi Arabia
| | - Khalid Al-Regaiey
- Khalid Al-Regaiey Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Assim A Alfadda
- Assim A. Alfadda Obesity Research Center, Department of Medicine, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | | |
Collapse
|
42
|
HFD-induced hepatic lipid accumulation and inflammation are decreased in Factor D deficient mouse. Sci Rep 2020; 10:17593. [PMID: 33067533 PMCID: PMC7568538 DOI: 10.1038/s41598-020-74617-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 09/30/2020] [Indexed: 12/12/2022] Open
Abstract
Excessive intake of fat causes accumulation of fat in liver, leading to non-alcoholic fatty liver disease (NAFLD). High-fat diet (HFD) upregulates the expression of Factor D, a complement pathway component, in the liver of mice. However, the functions of Factor D in liver are not well known. Therefore, the current study investigated the relationship between Factor D and hepatic lipid accumulation using CRISPR/Cas9-mediated Factor D knockout (FD-KO) mice. Factor D deficiency downregulated expression of genes related to fatty acid uptake and de novo lipogenesis in the liver. Furthermore, Factor D deficiency reduced the expression of inflammatory factors (Tnf and Ccl2) and fibrosis markers and decreased accumulation of F4/80-positive macrophages. These data suggest that the Factor D deficiency improved hepatic lipid accumulation and hepatic inflammation in HFD-fed mice.
Collapse
|
43
|
Li Y, Zou W, Brestoff JR, Rohatgi N, Wu X, Atkinson JP, Harris CA, Teitelbaum SL. Fat-Produced Adipsin Regulates Inflammatory Arthritis. Cell Rep 2020; 27:2809-2816.e3. [PMID: 31167128 DOI: 10.1016/j.celrep.2019.05.032] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/21/2019] [Accepted: 05/09/2019] [Indexed: 12/22/2022] Open
Abstract
We explored the relationship of obesity and inflammatory arthritis (IA) by selectively expressing diphtheria toxin in adipose tissue yielding "fat-free" (FF) mice completely lacking white and brown fat. FF mice exhibit systemic neutrophilia and elevated serum acute phase proteins suggesting a predisposition to severe IA. Surprisingly, FF mice are resistant to K/BxN serum-induced IA and attendant bone destruction. Despite robust systemic basal neutrophilia, neutrophil infiltration into joints of FF mice does not occur when challenged with K/BxN serum. Absence of adiponectin, leptin, or both has no effect on joint disease, but deletion of the adipokine adipsin (complement factor D) completely prevents serum-induced IA. Confirming that fat-expressed adipsin modulates the disorder, transplantation of wild-type (WT) adipose tissue into FF mice restores susceptibility to IA, whereas recipients of adipsin-deficient fat remain resistant. Thus, adipose tissue regulates development of IA through a pathway in which adipocytes modify neutrophil responses in distant tissues by producing adipsin.
Collapse
Affiliation(s)
- Yongjia Li
- Division of Anatomic and Molecular Pathology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Wei Zou
- Division of Anatomic and Molecular Pathology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jonathan R Brestoff
- Division of Laboratory and Genomic Medicine, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Nidhi Rohatgi
- Division of Anatomic and Molecular Pathology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Xiaobo Wu
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - John P Atkinson
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Charles A Harris
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Steven L Teitelbaum
- Division of Anatomic and Molecular Pathology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Division of Bone and Mineral Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Shriners Hospitals for Children, St. Louis, MO 63110, USA.
| |
Collapse
|
44
|
Adipokines as key players in β cell function and failure. Clin Sci (Lond) 2020; 133:2317-2327. [PMID: 31769478 DOI: 10.1042/cs20190523] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 11/11/2019] [Accepted: 11/13/2019] [Indexed: 12/13/2022]
Abstract
The growing prevalence of obesity and its related metabolic diseases, mainly Type 2 diabetes (T2D), has increased the interest in adipose tissue (AT) and its role as a principal metabolic orchestrator. Two decades of research have now shown that ATs act as an endocrine organ, secreting soluble factors termed adipocytokines or adipokines. These adipokines play crucial roles in whole-body metabolism with different mechanisms of action largely dependent on the tissue or cell type they are acting on. The pancreatic β cell, a key regulator of glucose metabolism due to its ability to produce and secrete insulin, has been identified as a target for several adipokines. This review will focus on how adipokines affect pancreatic β cell function and their impact on pancreatic β cell survival in disease contexts such as diabetes. Initially, the "classic" adipokines will be discussed, followed by novel secreted adipocyte-specific factors that show therapeutic promise in regulating the adipose-pancreatic β cell axis.
Collapse
|
45
|
Zhang Y, Keenan A, Dai DF, May KS, Anderson EE, Lindorfer MA, Henrich JB, Pitcher GR, Taylor RP, Smith RJ. C3(H2O) prevents rescue of complement-mediated C3 glomerulopathy in Cfh-/- Cfd-/- mice. JCI Insight 2020; 5:135758. [PMID: 32376801 DOI: 10.1172/jci.insight.135758] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 04/01/2020] [Indexed: 11/17/2022] Open
Abstract
Therapeutic complement inhibition is a major focus for novel drug development. Of upstream targets, factor D (FD) is appealing because it circulates in plasma at low concentrations and has a single function: to cleave factor B to generate C3 convertase of the alternative pathway (AP). Mice with a targeted deletion of factor H (FH; Cfh-/- mice) develop C3 glomerulopathy (C3G) due to uncontrolled AP activity. To assess the impact of FD inhibition, we studied Cfh-/- Cfd-/- mice. We show that C3G in Cfh-/- mice is not rescued by removing FD. We used serum from Cfh-/- Cfd-/- mice to demonstrate that residual AP function occurs even when both FD and FH are missing and that hemolytic activity is present due to the action of C3(H2O). We propose that uncontrolled tick-over leads to slow activation of the AP in Cfh-/- Cfd-/- mice and that a minimal threshold of FH is necessary if tissue deposition of C3 is to be prevented. The FD/FH ratio dictates serum C3 level and renal C3b deposition. In C3G patients with chronic renal disease, the FD/FH ratio correlates inversely with C3 and C5 serum levels, suggesting that continuous AP control may be difficult to achieve by targeting FD.
Collapse
Affiliation(s)
- Yuzhou Zhang
- Molecular Otolaryngology and Renal Research Laboratories, and
| | - Adam Keenan
- Molecular Otolaryngology and Renal Research Laboratories, and
| | - Dao-Fu Dai
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Kristofer S May
- Molecular Otolaryngology and Renal Research Laboratories, and
| | | | - Margaret A Lindorfer
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - John B Henrich
- Molecular Otolaryngology and Renal Research Laboratories, and
| | | | - Ronald P Taylor
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | | |
Collapse
|
46
|
Immunosuppression in Cows following Intramammary Infusion of Mycoplasma bovis. Infect Immun 2020; 88:IAI.00521-19. [PMID: 31843962 DOI: 10.1128/iai.00521-19] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 12/06/2019] [Indexed: 01/14/2023] Open
Abstract
Mycoplasma bovis is a destructive pathogen that causes large economic losses in rearing cattle for beef and dairy worldwide. M. bovis causes suppression of and evades the host immune response; however, the mechanisms of host immune function involved in M. bovis mastitis have not been elucidated. The purpose of this study was to elucidate the characteristics of the bovine immune response to mycoplasmal mastitis. We evaluated the responsiveness of the bovine mammary gland following infusion of M. bovis Somatic cell counts and bacterial counts in milk from the infected quarter were increased. However, the proliferation of peripheral blood mononuclear cells (blood MNCs) and mononuclear cells isolated from M. bovis-stimulated mammary lymph nodes (lymph node MNCs) did not differ from that in the unstimulated cells. Transcriptome analysis revealed that the mRNA levels of innate immune system-related genes in blood MNCs, complement factor D (CFD), ficolin 1 (FCN1), and tumor necrosis factor superfamily member 13 (TNFSF13) decreased following intramammary infusion of M. bovis The mRNA levels of immune exhaustion-related genes, programmed cell death 1 (PD-1), programmed cell death-ligand 1 (PD-L1), lymphocyte activation gene 3 (LAG3), and cytotoxic T-lymphocyte-associated protein 4 (CTLA4) of milk mononuclear cells (milk MNCs) in the infected quarter were increased compared with those before infusion. Increase in immune exhaustion-related gene expression and decrease in innate immune response-related genes of MNCs in quarters from cows were newly characterized by M. bovis-induced mastitis. These results suggested that M. bovis-induced mastitis affected the immune function of bovine MNCs, which is associated with prolonged duration of infection with M. bovis.
Collapse
|
47
|
Paré F, Tardif G, Fahmi H, Ouhaddi Y, Pelletier JP, Martel-Pelletier J. In vivo protective effect of adipsin-deficiency on spontaneous knee osteoarthritis in aging mice. Aging (Albany NY) 2020; 12:2880-2896. [PMID: 32012117 PMCID: PMC7041762 DOI: 10.18632/aging.102784] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 01/18/2020] [Indexed: 12/12/2022]
Abstract
The adipokine adipsin is an emerging mediator of human osteoarthritis (OA) progression. Here, we investigated its in vivo role in the development of spontaneous OA in aging mice. We compared articular knee joint morphology, histology in knee cartilage, synovial membrane, subchondral bone, meniscus, and anterior cruciate ligament (ACL); and chondrogenesis in the ACL from adipsin-deficient (Df-/-) and wild-type (Df+/+) 20-week- and 20-month-old mice. Serum levels of a panel of adipokines, inflammatory factors, and metalloproteases known to be implicated in OA were investigated. Data first revealed that the early manifestation of OA appeared in the ACL of 20-week-old mice, progressing to severe alterations in the 20 month-old wild-type mice. Further results demonstrated that adipsin-deficiency protected the articular tissues from spontaneous OA progression and triggered significantly higher serum levels of the adipokines adiponectin and FGF-21 while lowering levels of the inflammatory factor interleukin 6 (IL-6) in both young and old mice. This work further underlines the clinical relevance of adipsin as a novel therapeutic approach of human OA. Moreover, this study shows the potential beneficial effect of the adipokine FGF-21 against OA, and provides support for this factor to be a new biomarker and/or target of primary OA therapeutic avenues.
Collapse
Affiliation(s)
- Frédéric Paré
- Osteoarthritis Research Unit, University of Montreal Hospital Research Centre (CRCHUM), Montréal, Québec, Canada
| | - Ginette Tardif
- Osteoarthritis Research Unit, University of Montreal Hospital Research Centre (CRCHUM), Montréal, Québec, Canada
| | - Hassan Fahmi
- Osteoarthritis Research Unit, University of Montreal Hospital Research Centre (CRCHUM), Montréal, Québec, Canada
| | - Yassine Ouhaddi
- Osteoarthritis Research Unit, University of Montreal Hospital Research Centre (CRCHUM), Montréal, Québec, Canada
| | - Jean-Pierre Pelletier
- Osteoarthritis Research Unit, University of Montreal Hospital Research Centre (CRCHUM), Montréal, Québec, Canada
| | - Johanne Martel-Pelletier
- Osteoarthritis Research Unit, University of Montreal Hospital Research Centre (CRCHUM), Montréal, Québec, Canada
| |
Collapse
|
48
|
Funcke JB, Scherer PE. Beyond adiponectin and leptin: adipose tissue-derived mediators of inter-organ communication. J Lipid Res 2019; 60:1648-1684. [PMID: 31209153 PMCID: PMC6795086 DOI: 10.1194/jlr.r094060] [Citation(s) in RCA: 211] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 06/17/2019] [Indexed: 01/10/2023] Open
Abstract
The breakthrough discoveries of leptin and adiponectin more than two decades ago led to a widespread recognition of adipose tissue as an endocrine organ. Many more adipose tissue-secreted signaling mediators (adipokines) have been identified since then, and much has been learned about how adipose tissue communicates with other organs of the body to maintain systemic homeostasis. Beyond proteins, additional factors, such as lipids, metabolites, noncoding RNAs, and extracellular vesicles (EVs), released by adipose tissue participate in this process. Here, we review the diverse signaling mediators and mechanisms adipose tissue utilizes to relay information to other organs. We discuss recently identified adipokines (proteins, lipids, and metabolites) and briefly outline the contributions of noncoding RNAs and EVs to the ever-increasing complexities of adipose tissue inter-organ communication. We conclude by reflecting on central aspects of adipokine biology, namely, the contribution of distinct adipose tissue depots and cell types to adipokine secretion, the phenomenon of adipokine resistance, and the capacity of adipose tissue to act both as a source and sink of signaling mediators.
Collapse
Affiliation(s)
- Jan-Bernd Funcke
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX
| | - Philipp E Scherer
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
49
|
Peng M, Li Z, Niu D, Liu X, Dong Z, Li J. Complement factor B/C2 in molluscs regulates agglutination and illuminates evolution of the Bf/C2 family. FASEB J 2019; 33:13323-13333. [PMID: 31550175 DOI: 10.1096/fj.201901142rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Complement factor B/C2 family (Bf/C2F) proteins are core complement system components in vertebrates that are absent in invertebrates and have been lost by numerous species, raising evolutionary questions. At least 3 duplication events have occurred from Cnidaria (ancestor) to mammals. Type II Bf/C2 genes appeared during separation of Proterostomia and Deuterostomes. The second event occurred during separation of vertebrates and invertebrates, yielding type II-2 Bf/C2. The third event occurred when jawed and jawless fish were separated, eventually producing Bf and C2 genes. Herein, we report the second mollusc Sinonovacula constricta Bf/C2-type gene (ScBf). ScBf is similar to Ruditapes decussatus Bf-like because both lack the first complement control protein module at the N terminus present in mammalian Bf/C2 proteins. Uniquely, the Ser protease (SP) module at the C terminus of ScBf is ∼50 aa longer than in other complement factor B/C2-type (Bf/C2T) proteins, and is Glu-rich. Bf/C2T proteins in molluscs lack the catalytic Ser in the SP module. Surprisingly, ScBf regulates rabbit erythrocyte agglutination, during which it is localized on the erythrocyte surface. Thus, ScBf may mediate the agglutination cascade and may be an upstream regulator of this process. Our findings provide new insight into the origin of the Bf/C2F.-Peng, M., Li, Z., Niu, D., Liu, X., Dong, Z., Li, J. Complement factor B/C2 in molluscs regulates agglutination and illuminates evolution of the Bf/C2 family.
Collapse
Affiliation(s)
- Maoxiao Peng
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Shanghai Ocean University, Shanghai, China.,College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Zhi Li
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Shanghai Ocean University, Shanghai, China.,College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Donghong Niu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Shanghai Ocean University, Shanghai, China.,College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China.,National Demonstration Centre for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China.,Co-Innovation Centre of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang, China; and
| | - Xiaojun Liu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Shanghai Ocean University, Shanghai, China.,College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Zhiguo Dong
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Shanghai Ocean University, Shanghai, China.,College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China.,Co-Innovation Centre of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang, China; and
| | - Jiale Li
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Shanghai Ocean University, Shanghai, China.,College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China.,Co-Innovation Centre of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang, China; and
| |
Collapse
|
50
|
Chiarelli N, Ritelli M, Zoppi N, Colombi M. Cellular and Molecular Mechanisms in the Pathogenesis of Classical, Vascular, and Hypermobile Ehlers‒Danlos Syndromes. Genes (Basel) 2019; 10:E609. [PMID: 31409039 PMCID: PMC6723307 DOI: 10.3390/genes10080609] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/30/2019] [Accepted: 08/09/2019] [Indexed: 12/12/2022] Open
Abstract
The Ehlers‒Danlos syndromes (EDS) constitute a heterogenous group of connective tissue disorders characterized by joint hypermobility, skin abnormalities, and vascular fragility. The latest nosology recognizes 13 types caused by pathogenic variants in genes encoding collagens and other molecules involved in collagen processing and extracellular matrix (ECM) biology. Classical (cEDS), vascular (vEDS), and hypermobile (hEDS) EDS are the most frequent types. cEDS and vEDS are caused respectively by defects in collagen V and collagen III, whereas the molecular basis of hEDS is unknown. For these disorders, the molecular pathology remains poorly studied. Herein, we review, expand, and compare our previous transcriptome and protein studies on dermal fibroblasts from cEDS, vEDS, and hEDS patients, offering insights and perspectives in their molecular mechanisms. These cells, though sharing a pathological ECM remodeling, show differences in the underlying pathomechanisms. In cEDS and vEDS fibroblasts, key processes such as collagen biosynthesis/processing, protein folding quality control, endoplasmic reticulum homeostasis, autophagy, and wound healing are perturbed. In hEDS cells, gene expression changes related to cell-matrix interactions, inflammatory/pain responses, and acquisition of an in vitro pro-inflammatory myofibroblast-like phenotype may contribute to the complex pathogenesis of the disorder. Finally, emerging findings from miRNA profiling of hEDS fibroblasts are discussed to add some novel biological aspects about hEDS etiopathogenesis.
Collapse
Affiliation(s)
- Nicola Chiarelli
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, 25121 Brescia, Italy
| | - Marco Ritelli
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, 25121 Brescia, Italy
| | - Nicoletta Zoppi
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, 25121 Brescia, Italy
| | - Marina Colombi
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, 25121 Brescia, Italy.
| |
Collapse
|