1
|
Muraoka S, Brodie WD, Mattichak MN, Gurrea‐Rubio M, Ikari Y, Foster C, Amin MA, Khanna N, Amin H, Campbell PL, Vichaikul S, Model EN, Omara MM, Petrovski S, Kozicki K, Amarista C, Webber A, Ali M, Palisoc PJ, Hervoso J, Ruth JH, Tsoi LC, Varga J, Gudjonsson JE, Khanna D, Fox DA, Tsou P. Targeting CD13/Aminopeptidase N as a Novel Therapeutic Approach for Scleroderma Fibrosis. Arthritis Rheumatol 2025; 77:80-91. [PMID: 39175116 PMCID: PMC11684996 DOI: 10.1002/art.42973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 07/22/2024] [Accepted: 08/07/2024] [Indexed: 08/24/2024]
Abstract
OBJECTIVE Systemic sclerosis (SSc) is an autoimmune multisystem disease with poorly understood pathogenesis and ineffective treatment options. Soluble CD13 (sCD13), generated by the cleavage of cell surface CD13 via matrix metalloproteinase 14 (MMP14), signals through the bradykinin receptor B1 (B1R) to elicit pro-inflammatory, pro-arthritic, and pro-angiogenic responses. In this study, we explored the antifibrotic potential of targeting the sCD13-B1R axis in SSc. METHODS The expression of CD13, B1R, and MMP14 was examined in SSc skin and explanted dermal fibroblasts. The efficacy of B1R antagonists in the inhibition on fibrosis was determined in vitro and in vivo. RESULTS Expression of the genes for CD13, B1R, and MMP14 was elevated in skin biopsies from patients with diffuse cutaneous (dc) SSc. Notably, single-cell analysis of SSc skin biopsies revealed the highest BDKRB1 expression in COL8A1-positive myofibroblasts, a population exclusively seen in SSc. Transforming growth factor beta (TGFβ) induced the expression of BDKRB1 and production of sCD13 by dcSSc skin fibroblasts. Treatment of dcSSc fibroblasts with sCD13 promoted fibrotic gene expression, signaling, cell proliferation, migration, and gel contraction. The pro-fibrotic responses of sCD13 or TGFβ were prevented by a B1R antagonist. Mice lacking Cd13 or Bdkrb1 were resistant to bleomycin-induced skin fibrosis and inflammation. Pharmacological B1R inhibition had a comparable antifibrotic effect. CONCLUSION These results are the first to demonstrate a key role for sCD13 in SSc skin fibrosis and suggest that targeting the sCD13-B1R signaling axis is a promising novel therapeutic approach for SSc.
Collapse
Affiliation(s)
- Sei Muraoka
- University of Michigan, Ann Arbor, and Toho University School of MedicineTokyoJapan
| | | | | | | | - Yuzo Ikari
- University of Michigan, Ann Arbor and Showa University School of MedicineTokyoJapan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2
|
França PRDC, Paiva JPBD, Carvalho RRD, Figueiredo CP, Sirois P, Fernandes PD. R-954, a bradykinin B1 receptor antagonist, as a potential therapy in a preclinical endometriosis model. Peptides 2024; 181:171294. [PMID: 39265809 DOI: 10.1016/j.peptides.2024.171294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/14/2024]
Abstract
Endometriosis is a gynecological condition characterized by the growth of endometrium-like tissues outside of the uterine cavity. Currently available drugs are efficacious in treating endometriosis-related pain, however it's not a targeted treatment. The aim of this work is to evaluate the effects of R-954, a bradykinin B1 receptor antagonist, in a murine model of endometriosis. The model was induced in animals through autologous transplantation of part of the uterine horn. After 51 days, it was observed that implants developed into endometriotic lesions. The administration of R-954 or progesterone, for 15 consecutive days, prevented the progression of cyst development, reduced the size and weight of the cysts. Both treatments also reduced cellular infiltrate and production of inflammatory mediators (interleukin-1β, interleukin-6, tumor necrosis factor). However, only R-954 decreased angiogenic factors (VEGF and VEGF receptor). In addition, treatment with the antagonist did not interfere in the females' estrous cycle, as well as prevented gestational losses (reduction in the number of intermediate resorptions in pregnant females with endometriosis). Data suggested that R-954 has anti-inflammatory and anti-angiogenic effects; does not influence the estrous cycle; and prevents the number of gestational losses suggesting it as a good candidate for endometriosis treatment.
Collapse
Affiliation(s)
- Patricia Ribeiro de Carvalho França
- Universidade Federal do Rio de Janeiro, Instituto de Ciências Biomédicas, Programa de Pesquisa em Descoberta de Fármacos, Laboratório de Farmacologia da Dor e da Inflamação, Rio de Janeiro, Brasil
| | - João Pedro Barros de Paiva
- Universidade Federal do Rio de Janeiro, Instituto de Ciências Biomédicas, Programa de Pesquisa em Descoberta de Fármacos, Laboratório de Farmacologia da Dor e da Inflamação, Rio de Janeiro, Brasil
| | | | | | - Pierre Sirois
- Laval University, CHUL Research Center, Quebec, Canada
| | - Patricia Dias Fernandes
- Universidade Federal do Rio de Janeiro, Instituto de Ciências Biomédicas, Programa de Pesquisa em Descoberta de Fármacos, Laboratório de Farmacologia da Dor e da Inflamação, Rio de Janeiro, Brasil.
| |
Collapse
|
3
|
Kinin B1 Receptor Mediates Bidirectional Interaction between Neuroinflammation and Oxidative Stress. Antioxidants (Basel) 2023; 12:antiox12010150. [PMID: 36671012 PMCID: PMC9854481 DOI: 10.3390/antiox12010150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/03/2023] [Accepted: 01/05/2023] [Indexed: 01/10/2023] Open
Abstract
Hypertension is associated with increased expression of kinin B1 receptors (B1R) and increased levels of pro-inflammatory cytokines within the neurons. We previously reported that angiotensin II (Ang II) upregulates B1R expression and can induce neuroinflammation and oxidative stress in primary hypothalamic neurons. However, the order in which B1R activation, neuroinflammation, and oxidative stress occur has not yet been studied. Using primary hypothalamic neurons from neonatal mice, we show that tumor necrosis factor (TNF), lipopolysaccharides (LPS), and hydrogen peroxide (H2O2) can upregulate B1R expression and increase oxidative stress. Furthermore, our study shows that B1R blockade with R715, a specific B1R antagonist, can attenuate these effects. To further confirm our findings, we used a deoxycorticosterone acetate (DOCA)-salt model of hypertension to show that oxidative stress is upregulated in the hypothalamic paraventricular nucleus (PVN) of the brain. Together, these data provide novel evidence that relationship between oxidative stress, neuroinflammation, and B1R upregulation in the brain is bidirectional, and that B1R antagonism may have beneficial effects on neuroinflammation and oxidative stress in various disease pathologies.
Collapse
|
4
|
Rampa DR, Feng H, Allur-Subramaniyan S, Shim K, Pekcec A, Lee D, Doods H, Wu D. Kinin B1 receptor blockade attenuates hepatic fibrosis and portal hypertension in chronic liver diseases in mice. J Transl Med 2022; 20:590. [PMID: 36514072 PMCID: PMC9746183 DOI: 10.1186/s12967-022-03808-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 12/03/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND AIMS Kinin B1 receptors (B1Rs) are implicated in the pathogenesis of fibrosis. This study examined the anti-fibrotic effects of B1R blockade with BI 113823 in two established mouse models of hepatic fibrosis induced by intraperitoneal carbon tetrachloride (CCl4) injection or bile duct ligation (BDL). The mechanisms underlying the protection afforded by B1R inhibition were examined using human peripheral blood cells and LX2 human hepatic stellate cells (HSCs). METHODS Fibrotic liver diseases were induced in mice by intraperitoneal carbon tetrachloride (CCl4) injection for 6 weeks, and by bile duct ligation (BDL) for 3 weeks, respectively. Mice received daily treatment of vehicle or BI 113823 (B1R antagonist) from onset of the experiment until the end of the study. RESULTS B1Rs were strongly induced in fibrotic mouse liver. BI 113823 significantly attenuated liver fibrosis and portal hypertension (PH), and improved survival in both CCl4 and BDL mice. BI 113823 significantly reduced the expression of fibrotic proteins α-SMA, collagens 1, 3, 4, and profibrotic growth factors PDGF, TGFβ, CTGF, VEGF, proliferating cell nuclear antigen; and reduced hepatic Akt phosphorylation in CCl4- and BDL-induced liver fibrosis. BI 113823 also reduced expression of Cytokines IL-1, IL-6; chemokines MCP-1, MCP-3 and infiltration of inflammatory cells; and inhibited human monocyte and neutrophil activation, transmigration, TNF-α & MPO production in vitro. BI 113823 inhibited TGF-β and B1R agonist-stimulated human-HSC activation, contraction, proliferation, migration and fibrosis protein expression, and inhibited activation of PI3K/Akt signalling pathway. CONCLUSIONS B1Rs merits consideration as a novel therapeutic target for chronic liver fibrosis and PH.
Collapse
Affiliation(s)
- Dileep Reddy Rampa
- grid.411545.00000 0004 0470 4320Department of Bio-Nanotechnology and Bio-Convergence Engineering, Jeonbuk National University, Jeonju, South Korea
| | - Huiying Feng
- grid.411545.00000 0004 0470 4320Department of Bio-Nanotechnology and Bio-Convergence Engineering, Jeonbuk National University, Jeonju, South Korea
| | - Sivakumar Allur-Subramaniyan
- grid.411545.00000 0004 0470 4320Department of Animal Biotechnology & Agricultural Convergence Technology, Jeonbuk National University, Jeonju, South Korea
| | - Kwanseob Shim
- grid.411545.00000 0004 0470 4320Department of Animal Biotechnology & Agricultural Convergence Technology, Jeonbuk National University, Jeonju, South Korea
| | - Anton Pekcec
- grid.420061.10000 0001 2171 7500Research Beyond Borders, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Dongwon Lee
- grid.411545.00000 0004 0470 4320Department of Bio-Nanotechnology and Bio-Convergence Engineering, Jeonbuk National University, Jeonju, South Korea
| | - Henri Doods
- grid.420061.10000 0001 2171 7500Research Beyond Borders, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Dongmei Wu
- grid.411545.00000 0004 0470 4320Department of Bio-Nanotechnology and Bio-Convergence Engineering, Jeonbuk National University, Jeonju, South Korea ,grid.410396.90000 0004 0430 4458Department of Research, Mount Sinai Medical Center, Miami Beach, FL USA
| |
Collapse
|
5
|
Urwyler P, Moser S, Trendelenburg M, Sendi P, Osthoff M. Targeting thromboinflammation in COVID-19 - A narrative review of the potential of C1 inhibitor to prevent disease progression. Mol Immunol 2022; 150:99-113. [PMID: 36030710 PMCID: PMC9393183 DOI: 10.1016/j.molimm.2022.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 08/07/2022] [Accepted: 08/15/2022] [Indexed: 11/30/2022]
Abstract
Coronavirus disease 2019 (COVID-19) caused by SARS-CoV-2 is associated with a clinical spectrum ranging from asymptomatic carriers to critically ill patients with complications including thromboembolic events, myocardial injury, multisystemic inflammatory syndromes and death. Since the beginning of the pandemic several therapeutic options emerged, with a multitude of randomized trials, changing the medical landscape of COVID-19. The effect of various monoclonal antibodies, antiviral, anti-inflammatory and anticoagulation drugs have been studied, and to some extent, implemented into clinical practice. In addition, a multitude of trials improved the understanding of the disease and emerging evidence points towards a significant role of the complement system, kallikrein-kinin, and contact activation system as drivers of disease in severe COVID-19. Despite their involvement in COVID-19, treatments targeting these plasmatic cascades have neither been systematically studied nor introduced into clinical practice, and randomized studies with regards to these treatments are scarce. Given the multiple-action, multiple-target nature of C1 inhibitor (C1-INH), the natural inhibitor of these cascades, this drug may be an interesting candidate to prevent disease progression and combat thromboinflammation in COVID-19. This narrative review will discuss the current evidence with regards to the involvement of these plasmatic cascades as well as endothelial cells in COVID-19. Furthermore, we summarize the evidence of C1-INH in COVID-19 and potential benefits and pitfalls of C1-INH treatment in COVID-19.
Collapse
Affiliation(s)
- Pascal Urwyler
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Basel, Basel, Switzerland; Department of Clinical Research and Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Stephan Moser
- Department of Clinical Research and Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Marten Trendelenburg
- Department of Clinical Research and Department of Biomedicine, University of Basel, Basel, Switzerland; Division of Internal Medicine, University Hospital Basel, Basel, Switzerland
| | - Parham Sendi
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Michael Osthoff
- Department of Clinical Research and Department of Biomedicine, University of Basel, Basel, Switzerland; Division of Internal Medicine, University Hospital Basel, Basel, Switzerland.
| |
Collapse
|
6
|
Tsou PS, Lu C, Gurrea-Rubio M, Muraoka S, Campbell PL, Wu Q, Model EN, Lind ME, Vichaikul S, Mattichak MN, Brodie WD, Hervoso JL, Ory S, Amarista CI, Pervez R, Junginger L, Ali M, Hodish G, O’Mara MM, Ruth JH, Robida AM, Alt AJ, Zhang C, Urquhart AG, Lawton JN, Chung KC, Maerz T, Saunders TL, Groppi VE, Fox DA, Amin MA. Soluble CD13 induces inflammatory arthritis by activating the bradykinin receptor B1. J Clin Invest 2022; 132:151827. [PMID: 35439173 PMCID: PMC9151693 DOI: 10.1172/jci151827] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 04/08/2022] [Indexed: 11/17/2022] Open
Abstract
CD13, an ectoenzyme on myeloid and stromal cells, also circulates as a shed, soluble protein (sCD13) with powerful chemoattractant, angiogenic, and arthritogenic properties, which require engagement of a G protein-coupled receptor (GPCR). Here we identify the GPCR that mediates sCD13 arthritogenic actions as the bradykinin receptor B1 (B1R). Immunofluorescence and immunoblotting verified high expression of B1R in rheumatoid arthritis (RA) synovial tissue and fibroblast-like synoviocytes (FLSs), and demonstrated binding of sCD13 to B1R. Chemotaxis, and phosphorylation of Erk1/2, induced by sCD13, were inhibited by B1R antagonists. In ex vivo RA synovial tissue organ cultures, a B1R antagonist reduced secretion of inflammatory cytokines. Several mouse arthritis models, including serum transfer, antigen-induced, and local innate immune stimulation arthritis models, were attenuated in Cd13-/- and B1R-/- mice and were alleviated by B1R antagonism. These results establish a CD13/B1R axis in the pathogenesis of inflammatory arthritis and identify B1R as a compelling therapeutic target in RA and potentially other inflammatory diseases.
Collapse
Affiliation(s)
- Pei-Suen Tsou
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Chenyang Lu
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA.,Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Mikel Gurrea-Rubio
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Sei Muraoka
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Phillip L. Campbell
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Qi Wu
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Ellen N. Model
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Matthew E. Lind
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Sirapa Vichaikul
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Megan N. Mattichak
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - William D. Brodie
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jonatan L. Hervoso
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Sarah Ory
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Camila I. Amarista
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Rida Pervez
- Department of Orthopedic Surgery, University of Michigan Health System, Ann Arbor, Michigan, USA
| | - Lucas Junginger
- Department of Orthopedic Surgery, University of Michigan Health System, Ann Arbor, Michigan, USA
| | - Mustafa Ali
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Gal Hodish
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Morgan M. O’Mara
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jeffrey H. Ruth
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | | | | | | - Andrew G. Urquhart
- Department of Orthopedic Surgery, University of Michigan Health System, Ann Arbor, Michigan, USA
| | - Jeffrey N. Lawton
- Department of Orthopedic Surgery, University of Michigan Health System, Ann Arbor, Michigan, USA
| | - Kevin C. Chung
- Department of Orthopedic Surgery, University of Michigan Health System, Ann Arbor, Michigan, USA
| | - Tristan Maerz
- Department of Orthopedic Surgery, University of Michigan Health System, Ann Arbor, Michigan, USA
| | - Thomas L. Saunders
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA.,Biomedical Research Core Facilities, Transgenic Animal Model Core, and
| | - Vincent E. Groppi
- Center for Discovery of New Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - David A. Fox
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - M. Asif Amin
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
7
|
Zeng S, Yi R, Tan F, Sun P, Cheng Q, Zhao X. Lactobacillus plantarum HFY05 Attenuates Carrageenan-Induced Thrombosis in Mice by Regulating NF-κB Pathway-Associated Inflammatory Responses. Front Nutr 2022; 9:813899. [PMID: 35308280 PMCID: PMC8931398 DOI: 10.3389/fnut.2022.813899] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/31/2022] [Indexed: 01/02/2023] Open
Abstract
In this study, a carrageenan-induced thrombus model was established in mice to observe the ability of Lactobacillus plantarum KFY05 (LP-KFY05) to inhibit thrombosis through an NF-κB-associated pathway. Biochemical analysis, microscopical observations, quantitative polymerase chain reactions (qPCR) and western blot analysis were used to examine relevant serum and tissue indexes, and the composition of intestinal microorganisms was determined by examining the abundance of microorganisms in feces. The results showed that LP-KFY05 could markedly reduce the degree of black tail in thrombotic mice; increase the activated partial thromboplastin time (APTT); and decrease the thrombin time (TT), fibrinogen (FIB) level, and prothrombin time (PT). LP-KFY05 could also reduce tumor necrosis factor alpha (TNF-α), interleukin-6 (IL-6), and interleukin-1 beta (IL-1β) levels in sera and renal tissues of thrombotic mice. Hematoxylin and eosin staining showed that LP-KFY05 could alleviate renal tissue lesions and tail vein thrombosis. qPCR results showed that LP-KFY05 could down-regulate nuclear factor kappa-B (NF-κB) p65, IL-6, TNF-α, and interferon γ (IFN-γ) mRNA expression in renal tissues, as well as NF-κB p65, intercellular cell adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), and E-selectin mRNA expression in tail vein vascular tissues of thrombotic mice. Western blot analysis showed that LP-KFY05 also down-regulated NF-κB protein expression in renal and tail vein vascular tissues of thrombotic mice. Lastly, LP-KFY05 increased the abundances of Bacteroidetes, Lactobacillus, and Bifidobacterium, as well as decreased the abundance of Firmicutes. These results show that LP-KFY05 can reduce inflammation and inhibit thrombosis in thrombotic mice, and the effects of high concentrations of LP-KFY05 were most pronounced, which were similar to the effects of dipyridamole.
Collapse
Affiliation(s)
- Shi Zeng
- Department of Neurosurgery, People's Hospital of Chongqing Banan District, Chongqing, China
| | - Ruokun Yi
- Chongqing Collaborative Innovation Center for Functional Food, Chongqing Engineering Research Center of Functional Food, Chongqing Engineering Laboratory for Research and Development of Functional Food, Chongqing University of Education, Chongqing, China
| | - Fang Tan
- Department of Public Health, Our Lady of Fatima University, Valenzuela, Philippines
| | - Peng Sun
- Chongqing Collaborative Innovation Center for Functional Food, Chongqing Engineering Research Center of Functional Food, Chongqing Engineering Laboratory for Research and Development of Functional Food, Chongqing University of Education, Chongqing, China
| | - Qiang Cheng
- Department of Neurosurgery, People's Hospital of Chongqing Banan District, Chongqing, China
| | - Xin Zhao
- Chongqing Collaborative Innovation Center for Functional Food, Chongqing Engineering Research Center of Functional Food, Chongqing Engineering Laboratory for Research and Development of Functional Food, Chongqing University of Education, Chongqing, China
| |
Collapse
|
8
|
Molema G, Zijlstra JG, van Meurs M, Kamps JAAM. Renal microvascular endothelial cell responses in sepsis-induced acute kidney injury. Nat Rev Nephrol 2022; 18:95-112. [PMID: 34667283 DOI: 10.1038/s41581-021-00489-1] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2021] [Indexed: 12/29/2022]
Abstract
Microvascular endothelial cells in the kidney have been a neglected cell type in sepsis-induced acute kidney injury (sepsis-AKI) research; yet, they offer tremendous potential as pharmacological targets. As endothelial cells in distinct cortical microvascular segments are highly heterogeneous, this Review focuses on endothelial cells in their anatomical niche. In animal models of sepsis-AKI, reduced glomerular blood flow has been attributed to inhibition of endothelial nitric oxide synthase activation in arterioles and glomeruli, whereas decreased cortex peritubular capillary perfusion is associated with epithelial redox stress. Elevated systemic levels of vascular endothelial growth factor, reduced levels of circulating sphingosine 1-phosphate and loss of components of the glycocalyx from glomerular endothelial cells lead to increased microvascular permeability. Although coagulation disbalance occurs in all microvascular segments, the molecules involved differ between segments. Induction of the expression of adhesion molecules and leukocyte recruitment also occurs in a heterogeneous manner. Evidence of similar endothelial cell responses has been found in kidney and blood samples from patients with sepsis. Comprehensive studies are needed to investigate the relationships between segment-specific changes in the microvasculature and kidney function loss in sepsis-AKI. The application of omics technologies to kidney tissues from animals and patients will be key in identifying these relationships and in developing novel therapeutics for sepsis.
Collapse
Affiliation(s)
- Grietje Molema
- Dept. Pathology and Medical Biology, Medical Biology section, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
| | - Jan G Zijlstra
- Dept. Critical Care, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Matijs van Meurs
- Dept. Pathology and Medical Biology, Medical Biology section, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.,Dept. Critical Care, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Jan A A M Kamps
- Dept. Pathology and Medical Biology, Medical Biology section, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
9
|
Crosstalk between the renin-angiotensin, complement and kallikrein-kinin systems in inflammation. Nat Rev Immunol 2021; 22:411-428. [PMID: 34759348 PMCID: PMC8579187 DOI: 10.1038/s41577-021-00634-8] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2021] [Indexed: 12/28/2022]
Abstract
During severe inflammatory and infectious diseases, various mediators modulate the equilibrium of vascular tone, inflammation, coagulation and thrombosis. This Review describes the interactive roles of the renin–angiotensin system, the complement system, and the closely linked kallikrein–kinin and contact systems in cell biological functions such as vascular tone and leakage, inflammation, chemotaxis, thrombosis and cell proliferation. Specific attention is given to the role of these systems in systemic inflammation in the vasculature and tissues during hereditary angioedema, cardiovascular and renal glomerular disease, vasculitides and COVID-19. Moreover, we discuss the therapeutic implications of these complex interactions, given that modulation of one system may affect the other systems, with beneficial or deleterious consequences. The renin–angiotensin, complement and kallikrein–kinin systems comprise a multitude of mediators that modulate physiological responses during inflammatory and infectious diseases. This Review investigates the complex interactions between these systems and how these are dysregulated in various conditions, including cardiovascular diseases and COVID-19, as well as their therapeutic implications.
Collapse
|
10
|
Othman R, Cagnone G, Joyal JS, Vaucher E, Couture R. Kinins and Their Receptors as Potential Therapeutic Targets in Retinal Pathologies. Cells 2021; 10:1913. [PMID: 34440682 PMCID: PMC8391508 DOI: 10.3390/cells10081913] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/23/2021] [Accepted: 07/24/2021] [Indexed: 12/29/2022] Open
Abstract
The kallikrein-kinin system (KKS) contributes to retinal inflammation and neovascularization, notably in diabetic retinopathy (DR) and neovascular age-related macular degeneration (AMD). Bradykinin type 1 (B1R) and type 2 (B2R) receptors are G-protein-coupled receptors that sense and mediate the effects of kinins. While B2R is constitutively expressed and regulates a plethora of physiological processes, B1R is almost undetectable under physiological conditions and contributes to pathological inflammation. Several KKS components (kininogens, tissue and plasma kallikreins, and kinin receptors) are overexpressed in human and animal models of retinal diseases, and their inhibition, particularly B1R, reduces inflammation and pathological neovascularization. In this review, we provide an overview of the KKS with emphasis on kinin receptors in the healthy retina and their detrimental roles in DR and AMD. We highlight the crosstalk between the KKS and the renin-angiotensin system (RAS), which is known to be detrimental in ocular pathologies. Targeting the KKS, particularly the B1R, is a promising therapy in retinal diseases, and B1R may represent an effector of the detrimental effects of RAS (Ang II-AT1R).
Collapse
Affiliation(s)
- Rahmeh Othman
- School of Optometry, Université de Montréal, Montreal, QC H3T 1P1, Canada
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Gael Cagnone
- Department of Pediatry, Faculty of Medicine, CHU St Justine, Université de Montréal, Montreal, QC H3T 1J4, Canada; (G.C.); (J.-S.J.)
| | - Jean-Sébastien Joyal
- Department of Pediatry, Faculty of Medicine, CHU St Justine, Université de Montréal, Montreal, QC H3T 1J4, Canada; (G.C.); (J.-S.J.)
| | - Elvire Vaucher
- School of Optometry, Université de Montréal, Montreal, QC H3T 1P1, Canada
| | - Réjean Couture
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| |
Collapse
|
11
|
ACE2 in the renin-angiotensin system. Clin Sci (Lond) 2020; 134:3063-3078. [PMID: 33264412 DOI: 10.1042/cs20200478] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/12/2020] [Accepted: 11/19/2020] [Indexed: 01/01/2023]
Abstract
In 2020 we are celebrating the 20th anniversary of the angiotensin-converting enzyme 2 (ACE2) discovery. This event was a landmark that shaped the way that we see the renin-angiotensin system (RAS) today. ACE2 is an important molecular hub that connects the RAS classical arm, formed mainly by the octapeptide angiotensin II (Ang II) and its receptor AT1, with the RAS alternative or protective arm, formed mainly by the heptapeptides Ang-(1-7) and alamandine, and their receptors, Mas and MrgD, respectively. In this work we reviewed classical and modern literature to describe how ACE2 is a critical component of the protective arm, particularly in the context of the cardiac function, coagulation homeostasis and immune system. We also review recent literature to present a critical view of the role of ACE2 and RAS in the SARS-CoV-2 pandemic.
Collapse
|
12
|
Dagnino APA, Campos MM, Silva RBM. Kinins and Their Receptors in Infectious Diseases. Pharmaceuticals (Basel) 2020; 13:ph13090215. [PMID: 32867272 PMCID: PMC7558425 DOI: 10.3390/ph13090215] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/23/2020] [Accepted: 08/25/2020] [Indexed: 01/08/2023] Open
Abstract
Kinins and their receptors have been implicated in a series of pathological alterations, representing attractive pharmacological targets for several diseases. The present review article aims to discuss the role of the kinin system in infectious diseases. Literature data provides compelling evidence about the participation of kinins in infections caused by diverse agents, including viral, bacterial, fungal, protozoan, and helminth-related ills. It is tempting to propose that modulation of kinin actions and production might be an adjuvant strategy for management of infection-related complications.
Collapse
|
13
|
Kinin-B1 Receptor Stimulation Promotes Invasion and is Involved in Cell-Cell Interaction of Co-Cultured Glioblastoma and Mesenchymal Stem Cells. Sci Rep 2018; 8:1299. [PMID: 29358738 PMCID: PMC5777993 DOI: 10.1038/s41598-018-19359-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 12/29/2017] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma multiforme (GBM) represents the most lethal brain tumour, and these tumours have very limited treatment options. Mesenchymal stem cells (MSC) are considered as candidates for advanced cell therapies, due to their tropism towards GBM, possibly affecting their malignancy, thus also representing a potential therapeutic vector. Therefore, we aimed to compare the effects of bone-marrow-derived versus adipose-tissue-derived MSC (BM-/AT-MSC) on heterogeneous populations of tumour cells. This cells' interplay was addressed by the in-vitro two-dimensional (monolayer) and three-dimensional (spheroid) co-culture models, using U87 and U373 GBM cell lines, expressing genotypically different mesenchymal transcriptome profiles. U87 cell low mesenchymal profile expressed high levels of kinin receptor 1 (B1R) and their invasion was greatly enhanced by the B1R agonist des-Arg9-bradykinin upon BM-MSC co-culturing in 3D co-cultures. This correlated to significantly higher cell-cell interactions in U87/BM-MSC mixed spheroids. This was not observed with the U373 cells and not in AT-MSC co-cultures. Altogether, these data support the on-going exploration of B1R as target for adjuvant approach in GBM therapy. Secondly, the results emphasize the need for further careful exploration of the selectivity regarding the origin of MSC as potential candidates for cell therapies, particular in cancer, where they may adversely affect heterogeneous tumour cell populations.
Collapse
|
14
|
Kim JY, Choi SH, Cho YD, Yoon YH, Park JH, Lim CS. The impact of hypoxic conditions on apoptosis and Toll-like receptor 4 expression in polymorphonuclear neutrophils. EUR J INFLAMM 2018. [DOI: 10.1177/1721727x18756759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Many patients are admitted to the emergency department due to trauma. Trauma patients suffer from hypoxia due to massive hemorrhage, respiratory failure, and hypovolemic shock. Further damage is caused by reduced immune function and over-expression of inflammatory response. We conducted an experiment to determine the effects of hyperoxia and hypoxia on apoptosis and expression of Toll-like receptor 4 (TLR4) in polymorphonuclear neutrophils (PMNs). Initially, the PMNs were placed in normoxic and hypoxic conditions, and these PMNs were divided into two groups as stimulated or not stimulated with lipopolysaccharide (LPS). Levels of apoptosis and TLR4 expression were measured under normoxic, hypoxic, and hyperoxic conditions. Apoptosis decreased in the hypoxic group than in the normoxic group. With LPS stimulation, apoptosis was decreased in all three treatment groups and even more reduced in the hypoxic group. TLR4 expression increased in all three treatment groups with LPS stimulation, increased further in the hypoxic group, and to a lesser degree in the hyperoxic group. Unlike the cells exposed to hypoxic conditions, the cells exposed to the hyperoxic condition reacted similarly to the cells in the control (normoxic) group. Therefore, the inflammatory reactions can be stronger in the hypoxic group than in the other two groups.
Collapse
Affiliation(s)
- Jung-Youn Kim
- Department of Emergency Medicine, College of Medicine, Korea University, Seoul, Korea
| | - Sung-Hyuk Choi
- Department of Emergency Medicine, College of Medicine, Korea University, Seoul, Korea
- Department of Emergency Medicine, Korea University Guro Hospital, Seoul, Korea
| | - Young-Duck Cho
- Department of Emergency Medicine, College of Medicine, Korea University, Seoul, Korea
| | - Young-Hoon Yoon
- Department of Emergency Medicine, College of Medicine, Korea University, Seoul, Korea
| | - Jong-Hak Park
- Department of Emergency Medicine, College of Medicine, Korea University, Seoul, Korea
| | - Chae-Seung Lim
- Department of Laboratory Medicine, College of Medicine, Korea University, Seoul, Korea
| |
Collapse
|
15
|
Abstract
INTRODUCTION Kinins are peptide mediators exerting their pro-inflammatory actions by the selective stimulation of two distinct G-protein coupled receptors, termed BKB1R and BKB2R. While BKB2R is constitutively expressed in a multitude of tissues, BKB1R is hardly expressed at baseline but highly inducible by inflammatory mediators. In particular, BKB1R was shown to be involved in the pathogenesis of numerous inflammatory diseases. Areas covered: This review intends to evaluate the therapeutic potential of substances interacting with the BKB1R. To this purpose we summarize the published literature on animal studies with antagonists and knockout mice for this receptor. Expert Opinion: In most cases the pharmacological inhibition of BKB1R or its genetic deletion was beneficial for the outcome of the disease in animal models. Therefore, several companies have developed BKB1R antagonists and tested them in phase I and II clinical trials. However, none of the developed BKB1R antagonists was further developed for clinical use. We discuss possible reasons for this failure of translation of preclinical findings on BKB1R antagonists into the clinic.
Collapse
Affiliation(s)
- Fatimunnisa Qadri
- a Max-Delbrück Center for Molecular Medicine (MDC) , Berlin , Germany
| | - Michael Bader
- a Max-Delbrück Center for Molecular Medicine (MDC) , Berlin , Germany.,b Berlin Institute of Health (BIH) , Berlin , Germany.,c Charité University Medicine Berlin , Germany.,d German Center for Cardiovascular Research (DZHK) site Berlin , Berlin , Germany.,e Institute for Biology , University of Lübeck , Lübeck , Germany
| |
Collapse
|
16
|
Haddad Y, Couture R. Localization and Interaction between Kinin B1 Receptor and NADPH Oxidase in the Vascular System of Diabetic Rats. Front Physiol 2017; 8:861. [PMID: 29163205 PMCID: PMC5671568 DOI: 10.3389/fphys.2017.00861] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 10/16/2017] [Indexed: 01/17/2023] Open
Abstract
Kinin B1 receptor (B1R) enhanced superoxide anion (O2•-) production in the vasculature of diabetic rats. This study investigates the induction and distribution of B1R in diabetic blood vessels and addresses the hypothesis that B1R is co-localized with NADPH oxidase (NOX1 and NOX2) and produces its activation via protein kinase C (PKC). Diabetes was induced in rats with streptozotocin (STZ 65 mg.kg−1, i.p.). Two weeks later, the production of O2•- was measured in aorta rings in response to the B1R agonist (Sar[D-Phe8]-des-Arg9-BK, 20 μM) by the method of lucigenin-enhanced chemiluminescence. Various inhibitors were added (10 μM) to block PKCtotal (Ro-31-8220), PKCβ1/2 (LY333531), or NADPH oxidase (Diphenyleneiodonium). The cellular localization of B1R was studied in the aorta, popliteal artery, and renal glomerulus/arteries by immunofluorescence and confocal microscopy with markers of endothelial cells (anti-RECA-1), macrophages (anti-CD11), vascular smooth muscle cells (anti-SMA), and NADPH oxidase (anti-NOX1 and NOX2). Although B1R was largely distributed in resistant vessels, it was sparsely expressed in the aorta's endothelium. The greater basal production of O2•- in STZ-diabetic aorta was significantly enhanced by the B1R agonist (15–45 min). The peak response to the agonist (30 min) was inhibited by all inhibitors. Immunofluorescent staining for B1R, NOX1, and NOX2 was significantly increased in endothelial cells, vascular smooth muscle cells, and macrophages of STZ-diabetic aorta on which they were found co-localized. Data showed that B1R enhanced O2•- by activating vascular NADPH oxidase through PKCβ1/2. This was substantiated by the cellular co-localization of B1R with NOX1 and NOX2 and opens the possibility that B1R-enhanced oxidative stress is derived from vascular and infiltrating immune cells in diabetes.
Collapse
Affiliation(s)
- Youssef Haddad
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Réjean Couture
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
17
|
Soares DDM, Santos DR, Rummel C, Ott D, Melo MCC, Roth J, Calixto JB, Souza GEP. The relevance of kalikrein-kinin system via activation of B 2 receptor in LPS-induced fever in rats. Neuropharmacology 2017; 126:84-96. [PMID: 28826826 DOI: 10.1016/j.neuropharm.2017.08.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 08/05/2017] [Accepted: 08/11/2017] [Indexed: 12/21/2022]
Abstract
PURPOSE This study evaluated the involvement of endogenous kallikrein-kinin system and the bradykinin (BK) B1 and B2 receptors on LPS- induced fever and the POA cells involved in this response. MATERIAL AND METHODS Male Wistar rats received either i.v. (1 mg/kg), i.c.v. (20 nmol) or i.h. (2 nmol) injections of icatibant (B2 receptor antagonist) 30 or 60 min, respectively, before the stimuli. DALBK (B1 receptor antagonist) was given either 15min before BK (i.c.v.) or 30 min before LPS (i.v.). Captopril (5 mg/kg, sc.,) was given 1 h prior LPS or BK. Concentrations of BK and total kininogenon CSF, plasma and tissue kallikrein were evaluated. Rectal temperatures (rT) were assessed by telethermometry. Ca++ signaling in POA cells was performed in rat pup brain tissue microcultures. RESULTS Icatibant reduced LPS fever while, captopril exacerbated that response, an effect abolished by icatibant. Icatibant (i.h.) reduced fever to BK (i.h.) but not that induced by LPS (i.v.). BK increased intracellular calcium concentration in neurons and astrocytes. LPS increased levels of bradykinin, tissue kallikrein and total kininogen. BK (i.c.v.) increased rT and decreased tail skin temperature. Captopril potentiated BK-induced fever an effect abolished by icatibant. DALBK reduced the fever induced by BK. BK (i.c.v.) increased the CSF PGE2concentration. Effect abolished by indomethacin (i.p.). CONCLUSIONS LPS activates endogenous kalikrein-kinin system leading to production of BK, which by acting on B2-receptors of POA cells causes prostaglandin synthesis that in turn produces fever. Thus, a kinin B2-receptor antagonist that enters into the brain could constitute a new and interesting strategy to treat fever.
Collapse
Affiliation(s)
- Denis de Melo Soares
- Department of Medicament, Faculty of Pharmacy of Federal University of Bahia, Laboratory of Pharmacology, Ribeirão Preto, SP, Brazil.
| | - Danielle R Santos
- Pharmacology, Department of Physic and Chemistry, Faculty of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Christoph Rummel
- Veterinary Physiology, Faculty of Veterinary Medicine, Justus-Liebig-University of Giessen, Germany
| | - Daniela Ott
- Veterinary Physiology, Faculty of Veterinary Medicine, Justus-Liebig-University of Giessen, Germany
| | - Míriam C C Melo
- Pharmacology, Department of Physic and Chemistry, Faculty of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Joachim Roth
- Veterinary Physiology, Faculty of Veterinary Medicine, Justus-Liebig-University of Giessen, Germany
| | - João B Calixto
- Center of Innovation and Preclinical Research, Florianópolis, SC, Brazil
| | - Glória E P Souza
- Pharmacology, Department of Physic and Chemistry, Faculty of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
18
|
El Akoum S, Haddad Y, Couture R. Impact of pioglitazone and bradykinin type 1 receptor antagonist on type 2 diabetes in high-fat diet-fed C57BL/6J mice. Obes Sci Pract 2017; 3:352-362. [PMID: 29071111 PMCID: PMC5598024 DOI: 10.1002/osp4.117] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 05/01/2017] [Accepted: 05/19/2017] [Indexed: 01/24/2023] Open
Abstract
Aim Type 2 diabetes (T2D) is a major complication of obesity and a leading cause of morbidity and mortality. Antagonizing bradykinin type 1 receptor (B1R) improved body and tissue fat mass and reversed vascular and adipose tissue inflammation in a rat model of insulin resistance. This study aimed at evaluating further the role of B1R in a mouse model of T2D by comparing the antidiabetic and anti‐inflammatory effects of the B1R antagonist SSR240612 (SSR) in adipose tissue with those of pioglitazone (TZD), an activator of peroxisome proliferator‐activated receptor gamma. Methods C57BL/6J mice were fed with high‐fat diet (HFD) or standard diet (control) for 20 weeks. Yet, during the last 4 weeks, HFD‐fed mice were administered SSR and TZD (10 mg kg−1 d−1 each) as monotherapy or combined therapy subcutaneously. The impact of treatments was measured on metabolic hormones levels (ELISA), adipose tissue inflammatory status and the expression of candidate genes involved in T2D (quantitative real‐time polymerase chain reaction and western blot). Results SSR240612 and TZD treatments improved hyperglycaemia, hyperinsulinaemia, insulin resistance, adipose tissue inflammation (expression of B1R, chemokine ligand 2, F4/80 and tumour necrosis factor) and modulated adipogenesis (peroxisome proliferator‐activated receptor gamma, adipocytes' protein 2 and CD40 expressions) in HFD‐fed mice. Yet, SSR was more effective than TZD to reduce visceral fat mass and resistin. TZD/SSR combined treatment had an additive effect to improve insulin sensitivity and glucose intolerance. Conclusion Bradykinin type 1 receptor antagonism could represent a promising therapeutic tool in combination with TZD for the treatment of T2D, obesity and insulin resistance.
Collapse
Affiliation(s)
- S El Akoum
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine Université de Montréal Montréal Canada.,CHUM Research Center Montréal Canada
| | - Y Haddad
- Département de Pharmacologie et Physiologie, Faculté de Médecine Université de Montréal Montréal Canada
| | - R Couture
- Département de Pharmacologie et Physiologie, Faculté de Médecine Université de Montréal Montréal Canada
| |
Collapse
|
19
|
Analytical Management of Patients Undergoing Oral Anticoagulant Therapy Could Have a Strong Impact on Clinical Outcomes: A Follow-up Study. Cardiol Ther 2017; 6:261-271. [PMID: 28526929 PMCID: PMC5688967 DOI: 10.1007/s40119-017-0090-x] [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: 03/08/2017] [Indexed: 11/30/2022] Open
Abstract
Introduction Oral anticoagulant therapy, such as vitamin K antagonists (VKAs), is prominent for the prevention of cerebral ischemic stroke or systemic embolism and all-cause mortality in patients with atrial fibrillation, venous thromboembolism, and mechanical or biological valve. VKA treatment requires monitoring of the international normalized ratio (INR) in order to maintain it in a therapeutic range, avoiding side effects, the main and most significant of which is bleeding. The aim of the present study was to evaluate the event rates of several clinical composite outcomes, such as bleeding, thromboembolic events, and all-cause death. Methods We compared three organizational models distinguished by a total (from 1 January to 31 December 2015 in which PT/INR analysis with the relative internal and external quality controls was performed by the surveillance center) or partial (from 15 January to 15 July 2016 and from 15 August to 15 November 2016, in which the surveillance center had the ability to view only the PT/INR results or all patients analyses, including blood count, creatinine, liver enzymes, etc., respectively) analytical patient management. The present longitudinal follow-up study included 1225 patients, recruited from 1 January 2015 to 15 November 2016 at a surveillance center for the prevention of cerebral ischemic stroke and systemic embolism in Chieti (Italy). Results The results show a significant rise of the incidence rate ratio in patients undergoing VKA treatment during the period 15 January to 15 July 2016 compared to the previous one regarding total bleeding, especially for minor bleeding and digestive bleeding; thromboembolic events; and all-death cause. Conclusions These findings show that analytical and clinical data and information should be under the direct supervision and responsibility of the surveillance center. In fact, this approach seems to highlight the best results in terms of safety and therapeutic effectiveness.
Collapse
|
20
|
Dutra RC. Kinin receptors: Key regulators of autoimmunity. Autoimmun Rev 2017; 16:192-207. [DOI: 10.1016/j.autrev.2016.12.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 10/25/2016] [Indexed: 01/06/2023]
|
21
|
Haddad Y, Couture R. Interplay between the kinin B1 receptor and inducible nitric oxide synthase in insulin resistance. Br J Pharmacol 2016; 173:1988-2000. [PMID: 27059924 DOI: 10.1111/bph.13491] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 02/23/2016] [Accepted: 03/26/2016] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND AND PURPOSE Kinins are vasoactive and pro-inflammatory peptides whose biological effects are mediated by two GPCRs, named B1 and B2 receptors. While the B2 receptor plays a protective role in the cardiovascular system via the activation of endothelial NOS, the B1 receptor is associated with vascular inflammation, insulin resistance and diabetic complications. Because the B1 receptor is a potent activator of the inducible form of NOS (iNOS), this study has addressed the role of iNOS in the deleterious effects of B1 receptors in insulin resistance. EXPERIMENTAL APPROACH Male Sprague-Dawley rats (50-75 g) had free access to a drinking solution containing 10% d-glucose or tap water (control) for 9 weeks. During the last week, a selective iNOS inhibitor (1400W, 1 mg·kg(-1) twice daily) or its vehicle was administered s.c. KEY RESULTS Prolonged glucose treatment caused insulin resistance and several hallmarks of type 2 diabetes. Whereas the treatment with 1400W had no impact on the elevated systolic blood pressure and leptin levels in glucose-fed rats, it significantly reversed or attenuated hyperglycaemia, hyperinsulinaemia, insulin resistance (HOMA index), body weight gain, peroxynitrite formation (nitrotyrosine expression) and the up-regulation of biomarkers of inflammation (B1 receptor, carboxypeptidase M, iNOS and IL-1β) in renal cortex and aorta and to some extent in the liver. CONCLUSIONS AND IMPLICATIONS Pharmacological blockade of iNOS prevents the formation of peroxynitrite, which amplifies the pro-inflammatory effects of B1 receptors through a positive feedback mechanism. Hence, targeting iNOS can prevent the deleterious effects of B1 receptors in insulin resistance and peripheral inflammation.
Collapse
Affiliation(s)
- Youssef Haddad
- Department of Molecular and Integrative Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Réjean Couture
- Department of Molecular and Integrative Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|