1
|
Baryła M, Skrzycki M, Danielewicz R, Kosieradzki M, Struga M. Protein biomarkers in assessing kidney quality before transplantation‑current status and future perspectives (Review). Int J Mol Med 2024; 54:107. [PMID: 39370783 PMCID: PMC11448562 DOI: 10.3892/ijmm.2024.5431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 07/31/2024] [Indexed: 10/08/2024] Open
Abstract
To meet the demand for kidney transplants (KTx), organs are frequently retrieved not only from standard criteria donors (SCD; a donor who is aged <50 years and suffered brain death from any number of causes, such as traumatic injuries or a stroke) but also from expanded criteria donors (any donor aged >60 years or donors aged >50 years with two of the following: A history of high blood pressure, a creatinine serum level ≥1.5 mg/dl or death resulting from a stroke). This comes at the cost of a higher risk of primary non‑function (the permanent hyperkalemia, hyperuremia and fluid overload that result in the need for continuous dialysis after KTx), delayed graft function (the need for dialysis session at least once during the first week after KTx), earlier graft loss and urinary complications (vesico‑ureteral reflux, obstruction of the vesico‑ureteral anastomosis, urine leakage). At present, there are no commercially available diagnostic tools for assessing kidney quality prior to KTx. Currently available predictive models based on clinical data, such as the Kidney Donor Profile Index, are insufficient. One promising option is the application of perfusion solutions for protein biomarkers of kidney quality and predictors of short‑ and long‑term outcomes. However, to date, protein markers that can be detected with ELISA, western blotting and cytotoxic assays have not been identified to be a beneficial predictors of kidney quality. These include lactate dehydrogenases, glutathione S‑transferases, fatty acid binding proteins, extracellular histones, IL‑18, neutrophil gelatinase‑associated lipocalin, MMPs and kidney injury molecule‑1. However, novel methods, including liquid chromatography‑mass spectrometry (LC‑MS) and microarrays, allow the analysis of all renal proteins suspended/dissolved in the acellular preservation solution used for kidney storage before KTx (including hypothermic machine perfusion as one of kidney storage methods) e.g. Belzer University of Wisconsin. Recent proteomic studies utilizing LC‑MS have identified complement pathway elements (C3, C1QB, C4BPA, C1S, C1R and C1RL), desmoplakin, blood coagulation pathway elements and immunoglobulin heavy variable 2‑26 to be novel predictors of kidney quality before transplantation. This was because they were found to correlate with estimated glomerular filtration rate at 3 and 12 months after kidney transplantation. However, further proteomic studies focusing on distinct markers obtained from hypothermic and normothermic machine perfusion are needed to confirm their predictive value and to improve kidney storage methods. Therefore, the present literature review from PubMed, Scopus, Embase and Web of Science was performed with the aims of summarizing the current knowledge on the most frequently studied single protein biomarkers. In addition, novel analytical methods and insights into organ injury during preservation were documented, where future directions in assessing organ quality before kidney transplantation were also discussed.
Collapse
Affiliation(s)
- Maksymilian Baryła
- Chair and Department of Biochemistry, Medical University of Warsaw, 02-097 Warsaw, Poland
- Department of General and Transplant Surgery, Infant Jesus Hospital, Medical University of Warsaw, 02-006 Warsaw, Poland
| | - Michał Skrzycki
- Chair and Department of Biochemistry, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Roman Danielewicz
- Department of General and Transplant Surgery, Infant Jesus Hospital, Medical University of Warsaw, 02-006 Warsaw, Poland
| | - Maciej Kosieradzki
- Department of General and Transplant Surgery, Infant Jesus Hospital, Medical University of Warsaw, 02-006 Warsaw, Poland
| | - Marta Struga
- Department of General and Transplant Surgery, Infant Jesus Hospital, Medical University of Warsaw, 02-006 Warsaw, Poland
| |
Collapse
|
2
|
Zhang Y, Li X, Li S, Zhou Y, Zhang T, Sun L. Immunotherapy for Pulmonary Arterial Hypertension: From the Pathogenesis to Clinical Management. Int J Mol Sci 2024; 25:8427. [PMID: 39125996 PMCID: PMC11313500 DOI: 10.3390/ijms25158427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 07/27/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
Pulmonary hypertension (PH) is a progressive cardiovascular disease, which may lead to severe cardiopulmonary dysfunction. As one of the main PH disease groups, pulmonary artery hypertension (PAH) is characterized by pulmonary vascular remodeling and right ventricular dysfunction. Increased pulmonary artery resistance consequently causes right heart failure, which is the major reason for morbidity and mortality in this disease. Although various treatment strategies have been available, the poor clinical prognosis of patients with PAH reminds us that further studies of the pathological mechanism of PAH are still needed. Inflammation has been elucidated as relevant to the initiation and progression of PAH, and plays a crucial and functional role in vascular remodeling. Many immune cells and cytokines have been demonstrated to be involved in the pulmonary vascular lesions in PAH patients, with the activation of downstream signaling pathways related to inflammation. Consistently, this influence has been found to correlate with the progression and clinical outcome of PAH, indicating that immunity and inflammation may have significant potential in PAH therapy. Therefore, we reviewed the pathogenesis of inflammation and immunity in PAH development, focusing on the potential targets and clinical application of anti-inflammatory and immunosuppressive therapy.
Collapse
Affiliation(s)
| | | | | | | | - Tiantai Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China; (Y.Z.); (X.L.); (S.L.); (Y.Z.)
| | - Lan Sun
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China; (Y.Z.); (X.L.); (S.L.); (Y.Z.)
| |
Collapse
|
3
|
Nadasy GL, Patai BB, Molnar AA, Hetthessy JR, Tokes AM, Varady Z, Dornyei G. Vicious Circle With Venous Hypertension, Irregular Flow, Pathological Venous Wall Remodeling, and Valve Destruction in Chronic Venous Disease: A Review. Angiology 2024:33197241256680. [PMID: 38839285 DOI: 10.1177/00033197241256680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
Substantial advances occurred in phlebological practice in the last two decades. With the use of modern diagnostic equipment, the patients' venous hemodynamics can be examined in detail in everyday practice. Application of venous segments for arterial bypasses motivated studies on the effect of hemodynamic load on the venous wall. New animal models have been developed to study hemodynamic effects on the venous system. In vivo and in vitro studies revealed cellular phase transitions of venous endothelial, smooth muscle, and fibroblastic cells and changes in connective tissue composition, under hemodynamic load and at different locations of the chronically diseased venous system. This review is an attempt to integrate our knowledge from epidemiology, paleoanthropology and anthropology, clinical and experimental hemodynamic studies, histology, cell physiology, cell pathology, and molecular biology on the complex pathomechanism of this frequent disease. Our conclusion is that the disease is initiated by limited genetic adaptation of mankind not to bipedalism but to bipedalism in the unmoving standing or sitting position. In the course of the disease several pathologic vicious circles emerge, sustained venous hypertension inducing cellular phase transitions, chronic wall inflammation, apoptosis of cells, pathologic dilation, and valvular damage which, in turn, further aggravate the venous hypertension.
Collapse
Affiliation(s)
- Gyorgy L Nadasy
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | | | - Andrea A Molnar
- Department of Cardiology, Semmelweis University, Budapest, Hungary
| | | | - Anna-Maria Tokes
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | | | - Gabriella Dornyei
- Department of Morphology and Physiology, Health Science Faculty, Semmelweis University, Budapest, Hungary
| |
Collapse
|
4
|
Zhang F, Tao H, Gluck JM, Wang L, Daneshmand MA, King MW. A textile-reinforced composite vascular graft that modulates macrophage polarization and enhances endothelial cell migration, adhesion and proliferation in vitro. SOFT MATTER 2023; 19:1624-1641. [PMID: 36752696 DOI: 10.1039/d2sm01190e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
At the present time, there is no successful off-the-shelf small-caliber vascular graft (<6 mm) for the repair or bypass of the coronary or carotid arteries. In this study, we engineer a textile-reinforced hydrogel vascular graft. The textile fibers are circularly knitted into a flexible yet robust conduit to serve as the backbone of the composite vascular graft and provide the primary mechanical support. It is embedded in the hydrogel matrix which seals the open structure of the knitted reinforcement and mediates cellular response toward a faster reendothelialization. The mechanical properties of the composite vascular graft, including bursting strength, suture retention strength and radial compliance, significantly surpass the requirement for the vascular graft application and can be adjusted by altering the structure of the textile reinforcement. The addition of hydrogel matrix, on the other hand, improves the survival, adhesion and proliferation of endothelial cells in vitro. The composite vascular graft also enhances macrophage activation and upregulates M1 and M2 related gene expression, which further improves the endothelial cell migration that might favor the reendothelialization of the vascular graft. Taken together, the textile-reinforced hydrogel shows it potential to be a promising scaffold material to fabricate a tissue engineered vascular graft.
Collapse
Affiliation(s)
- Fan Zhang
- Wilson College of Textiles, North Carolina State University, Raleigh, NC 27606, USA.
| | - Hui Tao
- College of Textiles, Donghua University, Shanghai, 201620, China
| | - Jessica M Gluck
- Wilson College of Textiles, North Carolina State University, Raleigh, NC 27606, USA.
| | - Lu Wang
- College of Textiles, Donghua University, Shanghai, 201620, China
| | - Mani A Daneshmand
- Department of Surgery, School of Medicine, Emory University, Atlanta, Georgia 30322, USA
| | - Martin W King
- Wilson College of Textiles, North Carolina State University, Raleigh, NC 27606, USA.
- College of Textiles, Donghua University, Shanghai, 201620, China
| |
Collapse
|
5
|
Wang B, Wang X, Kenneth A, Drena A, Pacheco A, Kalvin L, Ibrahim ES, Rossi PJ, Thatcher K, Lincoln J. Developing small-diameter vascular grafts with human amniotic membrane: long-term evaluation of transplantation outcomes in a small animal model. Biofabrication 2023; 15. [PMID: 36626826 DOI: 10.1088/1758-5090/acb1da] [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: 10/05/2022] [Accepted: 01/10/2023] [Indexed: 01/11/2023]
Abstract
While current clinical utilization of large vascular grafts for vascular transplantation is encouraging, tissue engineering of small grafts still faces numerous challenges. This study aims to investigate the feasibility of constructing a small vascular graft from decellularized amniotic membranes (DAMs). DAMs were rolled around a catheter and each of the resulting grafts was crosslinked with (a) 0.1% glutaraldehyde; (b) 1-ethyl-3-(3-dimethylaminopropyl) crbodiimidehydro-chloride (20 mM)-N-hydroxy-succinimide (10 mM); (c) 0.5% genipin; and (d) no-crosslinking, respectively. Our results demonstrated the feasibility of using a rolling technique followed by lyophilization to transform DAM into a vessel-like structure. The genipin-crosslinked DAM graft showed an improved integrated structure, prolonged stability, proper mechanical property, and superior biocompatibility. After transplantation in rat abdominal aorta, the genipin-crosslinked DAM graft remained patent up to 16 months, with both endothelial and smooth muscle cell regeneration, which suggests that the genipin-crosslinked DAM graft has great potential to beimplementedas a small tissue engineered graft for futurevasculartransplantation.
Collapse
Affiliation(s)
- Bo Wang
- Joint Department of Biomedical Engineering, Marquette University and the Medical College of Wisconsin, Milwaukee, WI 53226, United States of America
| | - Xiaolong Wang
- Joint Department of Biomedical Engineering, Marquette University and the Medical College of Wisconsin, Milwaukee, WI 53226, United States of America
| | - Allen Kenneth
- Biomedical Resource Center, Medical College of Wisconsin, Milwaukee, WI 53226, United States of America
| | - Alexander Drena
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States of America
| | - Arsenio Pacheco
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States of America
| | - Lindsey Kalvin
- Department of Medicine, Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, United States of America
| | - Ei-Sayed Ibrahim
- Department of Radiology, Medical College of Wisconsin, Milwaukee, WI 53226, United States of America
| | - Peter J Rossi
- Heart and Vascular Center, Froedtert Hospital, Milwaukee, WI 53226, United States of America
| | - Kaitlyn Thatcher
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, United States of America
| | - Joy Lincoln
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, United States of America
| |
Collapse
|
6
|
Hu S, Wang L, Xu Y, Li F, Wang T. Disulfiram attenuates hypoxia-induced pulmonary hypertension by inhibiting GSDMD cleavage and pyroptosis in HPASMCs. Respir Res 2022; 23:353. [PMID: 36527086 PMCID: PMC9756478 DOI: 10.1186/s12931-022-02279-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Pulmonary hypertension (PH) is characterized by progressive pulmonary arterial remodelling, associated with different severities of inflammation and altered immune processes. Disulfiram eliminates the formation of N-gasdermin D (GSDMD) plasma membrane pores to prevent pyroptosis. Pyroptosis is a form of lytic cell death characterized by inflammasome activation and proinflammatory cytokine release that acts in the development of PH. We sought to investigate whether disulfiram could alleviate hypoxia-induced PH by inhibiting pyroptosis. METHODS To investigate whether disulfiram alleviates the progression of pulmonary hypertension, rodents were exposed to chronic hypoxia (10% oxygen, 4 weeks) to induce PH. The severity of PH was assessed by measuring right ventricular systolic pressure, mean pulmonary artery pressure, and the degree of right ventricular hypertrophy. Western blotting was used to measure proteins associated with the pyroptosis pathway, and ELISA was performed to measure the secretion of IL-18 and IL-1β, both of which are the primary methods for assessing pyroptosis. RESULTS IL-18 and IL-1β concentrations were higher in patients with PH than in normal controls. Disulfiram suppressed the progression of PH in mice and rats through the alleviation of pulmonary arterial remodelling. Pyroptosis-related proteins and the inflammasome were activated in rodent models of PH. Disulfiram inhibited the processing of GSDMD into N-GSDMD and attenuated the secretion of IL-1β and IL18. In vivo experiments showed that disulfiram also inhibited lytic death in HPASMCs. CONCLUSIONS Disulfiram treatment reduces PH progression through suppressing vascular remodelling by inhibiting GSDMD cleavage and pyroptosis. It might become a novel therapeutic option for the treatment of PH.
Collapse
Affiliation(s)
- Shunlian Hu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
- The Center for Biomedical Research, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Lu Wang
- Department of Respiratory and Critical Care Medicine, Miyun Teaching Hospital of Capital Medical University, Beijing, People's Republic of China
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, Miyun District, Beijing, People's Republic of China
- Department of Respiratory and Critical Care Medicine, Beijing Miyun Hospital, Beijing, People's Republic of China
| | - Yahan Xu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
- The Center for Biomedical Research, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Fajiu Li
- The Sixth Hospital of Wuhan City, Affiliated Hospital of Jianghan University, Beijing, People's Republic of China
| | - Tao Wang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
- The Center for Biomedical Research, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.
| |
Collapse
|
7
|
Zhang F, King MW. Immunomodulation Strategies for the Successful Regeneration of a Tissue-Engineered Vascular Graft. Adv Healthc Mater 2022; 11:e2200045. [PMID: 35286778 PMCID: PMC11468936 DOI: 10.1002/adhm.202200045] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/18/2022] [Indexed: 01/02/2023]
Abstract
Cardiovascular disease leads to the highest morbidity worldwide. There is an urgent need to solve the lack of a viable arterial graft for patients requiring coronary artery bypass surgery. The current gold standard is to use the patient's own blood vessel, such as a saphenous vein graft. However, some patients do not have appropriate vessels to use because of systemic disease or secondary surgery. On the other hand, there is no commercially available synthetic vascular graft available on the market for small diameter (<6 mm) blood vessels like coronary, carotid, and peripheral popliteal arteries. Tissue-engineered vascular grafts (TEVGs) are studied in recent decades as a promising alternative to synthetic arterial prostheses. Yet only a few studies have proceeded to a clinical trial. Recent studies have uncovered that the host immune response can be directed toward increasing the success of a TEVG by shedding light on ways to modulate the macrophage response and improve the tissue regeneration outcome. In this review, the basic concepts of vascular tissue engineering and immunoengineering are considered. The state-of-art of TEVGs is summarized and the role of macrophages in TEVG regeneration is analyzed. Current immunomodulatory strategies based on biomaterials are also discussed.
Collapse
Affiliation(s)
- Fan Zhang
- Wilson College of TextilesNorth Carolina State UniversityRaleighNC27606USA
| | - Martin W. King
- Wilson College of TextilesNorth Carolina State UniversityRaleighNC27606USA
| |
Collapse
|
8
|
Evans BR, Yerly A, van der Vorst EPC, Baumgartner I, Bernhard SM, Schindewolf M, Döring Y. Inflammatory Mediators in Atherosclerotic Vascular Remodeling. Front Cardiovasc Med 2022; 9:868934. [PMID: 35600479 PMCID: PMC9114307 DOI: 10.3389/fcvm.2022.868934] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 04/11/2022] [Indexed: 12/23/2022] Open
Abstract
Atherosclerotic vascular disease remains the most common cause of ischemia, myocardial infarction, and stroke. Vascular function is determined by structural and functional properties of the arterial vessel wall, which consists of three layers, namely the adventitia, media, and intima. Key cells in shaping the vascular wall architecture and warranting proper vessel function are vascular smooth muscle cells in the arterial media and endothelial cells lining the intima. Pathological alterations of this vessel wall architecture called vascular remodeling can lead to insufficient vascular function and subsequent ischemia and organ damage. One major pathomechanism driving this detrimental vascular remodeling is atherosclerosis, which is initiated by endothelial dysfunction allowing the accumulation of intimal lipids and leukocytes. Inflammatory mediators such as cytokines, chemokines, and modified lipids further drive vascular remodeling ultimately leading to thrombus formation and/or vessel occlusion which can cause major cardiovascular events. Although it is clear that vascular wall remodeling is an elementary mechanism of atherosclerotic vascular disease, the diverse underlying pathomechanisms and its consequences are still insufficiently understood.
Collapse
Affiliation(s)
- Bryce R. Evans
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Anaïs Yerly
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Emiel P. C. van der Vorst
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
- Institute for Molecular Cardiovascular Research (IMCAR) and Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, Netherlands
| | - Iris Baumgartner
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Sarah Maike Bernhard
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Marc Schindewolf
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Yvonne Döring
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
- *Correspondence: Yvonne Döring
| |
Collapse
|
9
|
Christou H, Khalil RA. Mechanisms of pulmonary vascular dysfunction in pulmonary hypertension and implications for novel therapies. Am J Physiol Heart Circ Physiol 2022; 322:H702-H724. [PMID: 35213243 PMCID: PMC8977136 DOI: 10.1152/ajpheart.00021.2022] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/22/2022] [Accepted: 02/22/2022] [Indexed: 12/21/2022]
Abstract
Pulmonary hypertension (PH) is a serious disease characterized by various degrees of pulmonary vasoconstriction and progressive fibroproliferative remodeling and inflammation of the pulmonary arterioles that lead to increased pulmonary vascular resistance, right ventricular hypertrophy, and failure. Pulmonary vascular tone is regulated by a balance between vasoconstrictor and vasodilator mediators, and a shift in this balance to vasoconstriction is an important component of PH pathology, Therefore, the mainstay of current pharmacological therapies centers on pulmonary vasodilation methodologies that either enhance vasodilator mechanisms such as the NO-cGMP and prostacyclin-cAMP pathways and/or inhibit vasoconstrictor mechanisms such as the endothelin-1, cytosolic Ca2+, and Rho-kinase pathways. However, in addition to the increased vascular tone, many patients have a "fixed" component in their disease that involves altered biology of various cells in the pulmonary vascular wall, excessive pulmonary artery remodeling, and perivascular fibrosis and inflammation. Pulmonary arterial smooth muscle cell (PASMC) phenotypic switch from a contractile to a synthetic and proliferative phenotype is an important factor in pulmonary artery remodeling. Although current vasodilator therapies also have some antiproliferative effects on PASMCs, they are not universally successful in halting PH progression and increasing survival. Mild acidification and other novel approaches that aim to reverse the resident pulmonary vascular pathology and structural remodeling and restore a contractile PASMC phenotype could ameliorate vascular remodeling and enhance the responsiveness of PH to vasodilator therapies.
Collapse
Affiliation(s)
- Helen Christou
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Raouf A Khalil
- Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
10
|
Rong W, Liu C, Li X, Wan N, Wei L, Zhu W, Bai P, Li M, Ou Y, Li F, Wang L, Wu X, Liu J, Xing M, Zhao X, Liu H, Zhang H, Lyu A. Caspase-8 Promotes Pulmonary Hypertension by Activating Macrophage-Associated Inflammation and IL-1β (Interleukin 1β) Production. Arterioscler Thromb Vasc Biol 2022; 42:613-631. [PMID: 35387479 DOI: 10.1161/atvbaha.121.317168] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Macrophages are involved in the pathogenesis of pulmonary arterial hypertension (PAH). Caspase-8, an apical component of cell death pathways, is significantly upregulated in macrophages of PAH animal models. However, its role in PAH remains unclear. Caspase-8 plays a critical role in regulating inflammatory responses via inflammasome activation, cell death, and cytokine induction. This study investigated the mechanism of regulation of IL-1β (interleukin 1β) activation in macrophages by caspase-8. METHODS A hypoxia + SU5416-induced PAH mouse model and monocrotaline-induced rat model of PAH were constructed and the role of caspase-8 was analyzed. RESULTS Caspase-8 and cleaved-caspase-8 were significantly upregulated in the lung tissues of SU5416 and hypoxia-treated PAH mice and monocrotaline-treated rats. Pharmacological inhibition of caspase-8 alleviated PAH compared with wild-type mice, observed as a significant reduction in right ventricular systolic pressure, ratio of right ventricular wall to left ventricular wall plus ventricular septum, pulmonary vascular media thickness, and pulmonary vascular muscularization; caspase-8 ablated mice also showed significant remission. Mechanistically, increased proliferation of pulmonary arterial smooth muscle cellss is closely associated with activation of the NLRP3 (NOD [nucleotide oligomerization domain]-, LRR [leucine-rich repeat]-, and PYD [pyrin domain]-containing protein 3) inflammasome and the IL-1β signaling pathway. Although caspase-8 did not affect extracellular matrix synthesis, it promoted inflammatory cell infiltration and pulmonary arterial smooth muscle cell proliferation via NLRP3/IL-1β activation during the development stage of PAH. CONCLUSIONS Taken together, our study suggests that macrophage-derived IL-1β via caspase-8-dependent canonical inflammasome is required for macrophages to play a pathogenic role in pulmonary perivascular inflammation.
Collapse
Affiliation(s)
- Wuwei Rong
- Department of Vascular & Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, China (W.R., C.L., N.W., L.W., W.Z., A.L.)
| | - Chenchen Liu
- Department of Vascular & Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, China (W.R., C.L., N.W., L.W., W.Z., A.L.)
| | - Xiaoming Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China (X.L., M.L., Y.O., F.L., L.W., X.W., J.L., M.X., X.Z., H.L., H.Z.)
| | - Naifu Wan
- Department of Vascular & Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, China (W.R., C.L., N.W., L.W., W.Z., A.L.)
| | - Lijiang Wei
- Department of Vascular & Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, China (W.R., C.L., N.W., L.W., W.Z., A.L.)
| | - Wentong Zhu
- Department of Vascular & Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, China (W.R., C.L., N.W., L.W., W.Z., A.L.)
| | - Peiyuan Bai
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China (P.B.)
| | - Ming Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China (X.L., M.L., Y.O., F.L., L.W., X.W., J.L., M.X., X.Z., H.L., H.Z.)
| | - Yangjing Ou
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China (X.L., M.L., Y.O., F.L., L.W., X.W., J.L., M.X., X.Z., H.L., H.Z.)
| | - Fang Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China (X.L., M.L., Y.O., F.L., L.W., X.W., J.L., M.X., X.Z., H.L., H.Z.)
| | - Lingxia Wang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China (X.L., M.L., Y.O., F.L., L.W., X.W., J.L., M.X., X.Z., H.L., H.Z.)
| | - Xuanhui Wu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China (X.L., M.L., Y.O., F.L., L.W., X.W., J.L., M.X., X.Z., H.L., H.Z.)
| | - Jianling Liu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China (X.L., M.L., Y.O., F.L., L.W., X.W., J.L., M.X., X.Z., H.L., H.Z.)
| | - Mingyan Xing
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China (X.L., M.L., Y.O., F.L., L.W., X.W., J.L., M.X., X.Z., H.L., H.Z.)
| | - Xiaoming Zhao
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China (X.L., M.L., Y.O., F.L., L.W., X.W., J.L., M.X., X.Z., H.L., H.Z.)
| | - Han Liu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China (X.L., M.L., Y.O., F.L., L.W., X.W., J.L., M.X., X.Z., H.L., H.Z.)
| | - Haibing Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China (X.L., M.L., Y.O., F.L., L.W., X.W., J.L., M.X., X.Z., H.L., H.Z.)
| | - Ankang Lyu
- Department of Vascular & Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, China (W.R., C.L., N.W., L.W., W.Z., A.L.)
| |
Collapse
|
11
|
Somarathna M, Hwang PT, Millican RC, Alexander GC, Isayeva-Waldrop T, Sherwood JA, Brott BC, Falzon I, Northrup H, Shiu YT, Stubben CJ, Totenhagen J, Jun HW, Lee T. Nitric oxide releasing nanomatrix gel treatment inhibits venous intimal hyperplasia and improves vascular remodeling in a rodent arteriovenous fistula. Biomaterials 2022; 280:121254. [PMID: 34836683 PMCID: PMC8724452 DOI: 10.1016/j.biomaterials.2021.121254] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 01/03/2023]
Abstract
Vascular access is the lifeline for hemodialysis patients and the single most important component of the hemodialysis procedure. Arteriovenous fistula (AVF) is the preferred vascular access for hemodialysis patients, but nearly 60% of AVFs created fail to successfully mature due to early intimal hyperplasia development and poor outward remodeling. There are currently no therapies available to prevent AVF maturation failure. First, we showed the important regulatory role of nitric oxide (NO) on AVF development by demonstrating that intimal hyperplasia development was reduced in an overexpressed endothelial nitric oxide synthase (NOS3) mouse AVF model. This supported the rationale for the potential application of NO to the AVF. Thus, we developed a self-assembled NO releasing nanomatrix gel and applied it perivascularly at the arteriovenous anastomosis immediately following rat AVF creation to investigate its therapeutic effect on AVF development. We demonstrated that the NO releasing nanomatrix gel inhibited intimal hyperplasia formation (more than 70% reduction), as well as improved vascular outward remodeling (increased vein diameter) and hemodynamic adaptation (lower wall shear stress approaching the preoperative level and less vorticity). Therefore, direct application of the NO releasing nanomatrix gel to the AVF anastomosis immediately following AVF creation may enhance AVF development, thereby providing long-term and durable vascular access for hemodialysis.
Collapse
Affiliation(s)
- Maheshika Somarathna
- Department of Medicine and Division of Nephrology, University of Alabama at Birmingham, AL, 35294, USA
| | - Patrick Tj Hwang
- Department of Biomedical Engineering, University of Alabama at Birmingham, AL, 35294, USA; Endomimetics, LLC, Birmingham, AL, 35242, USA
| | | | - Grant C Alexander
- Department of Biomedical Engineering, University of Alabama at Birmingham, AL, 35294, USA; Endomimetics, LLC, Birmingham, AL, 35242, USA
| | - Tatyana Isayeva-Waldrop
- Department of Medicine and Division of Nephrology, University of Alabama at Birmingham, AL, 35294, USA
| | | | - Brigitta C Brott
- Endomimetics, LLC, Birmingham, AL, 35242, USA; Department of Medicine and Division of Cardiovascular Disease, University of Alabama at Birmingham, AL, 35233, USA
| | - Isabelle Falzon
- Division of Nephrology and Hypertension, Department of Internal Medicine, University of Utah, Salt Lake City, UT, 84132, USA
| | - Hannah Northrup
- Division of Nephrology and Hypertension, Department of Internal Medicine, University of Utah, Salt Lake City, UT, 84132, USA
| | - Yan-Ting Shiu
- Division of Nephrology and Hypertension, Department of Internal Medicine, University of Utah, Salt Lake City, UT, 84132, USA; Veterans Affairs Medical Center, Salt Lake City, UT, 84148, USA
| | - Chris J Stubben
- Bioinformatics Shared Resource, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, 84112, USA
| | - John Totenhagen
- Department of Radiology, University of Alabama at Birmingham, AL, 35294, USA
| | - Ho-Wook Jun
- Department of Biomedical Engineering, University of Alabama at Birmingham, AL, 35294, USA; Endomimetics, LLC, Birmingham, AL, 35242, USA
| | - Timmy Lee
- Department of Medicine and Division of Nephrology, University of Alabama at Birmingham, AL, 35294, USA; Veterans Affairs Medical Center, Birmingham, AL, 35233, USA
| |
Collapse
|
12
|
Gu P, Hui X, Zheng Q, Gao Y, Jin L, Jiang W, Zhou C, Liu T, Huang Y, Liu Q, Nie T, Wang Y, Wang Y, Zhao J, Xu A. Mitochondrial uncoupling protein 1 antagonizes atherosclerosis by blocking NLRP3 inflammasome-dependent interleukin-1β production. SCIENCE ADVANCES 2021; 7:eabl4024. [PMID: 34878840 PMCID: PMC8654294 DOI: 10.1126/sciadv.abl4024] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/15/2021] [Indexed: 12/14/2022]
Abstract
Mitochondrial uncoupling protein 1 (UCP1) is the hallmark of brown adipocytes responsible for cold- and diet-induced thermogenesis. Here, we report a previously unidentified role of UCP1 in maintaining vascular health through its anti-inflammatory actions possibly in perivascular adipose tissue. UCP1 deficiency exacerbates dietary obesity-induced endothelial dysfunction, vascular inflammation, and atherogenesis in mice, which was not rectified by reconstitution of UCP1 in interscapular brown adipose tissue. Mechanistically, lack of UCP1 augments mitochondrial membrane potential and mitochondrial superoxide, leading to hyperactivation of the NLRP3-inflammasome and caspase-1–mediated maturation of interleukin-1β (IL-1β). UCP1 deficiency–evoked deterioration of vascular dysfunction and atherogenesis is reversed by IL-1β neutralization or a chemical mitochondrial uncoupler. Furthermore, UCP1 knockin pigs (which lack endogenous UCP1) are refractory to vascular inflammation and coronary atherosclerosis. Thus, UCP1 acts as a gatekeeper to prevent NLRP3 inflammasome activation and IL-1β production in the vasculature, thereby conferring a protective effect against cardiovascular diseases.
Collapse
Affiliation(s)
- Ping Gu
- State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong, China
- Department of Medicine, University of Hong Kong, Hong Kong, China
- Department of Endocrinology, Jinling Hospital, Nanjing University, School of Medicine, Nanjing, China
| | - Xiaoyan Hui
- State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong, China
- Department of Medicine, University of Hong Kong, Hong Kong, China
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, China
- Corresponding author. (A.X.); (X.H.); (J.Z.)
| | - Qiantao Zheng
- State Key Laboratory of Stem Cell and Reproductive Biology, Chinese Academy of Sciences, Chaoyang District, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Yuan Gao
- State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong, China
- Department of Medicine, University of Hong Kong, Hong Kong, China
| | - Leigang Jin
- State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong, China
- Department of Medicine, University of Hong Kong, Hong Kong, China
| | - Weimin Jiang
- Department of Cardiology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Changsheng Zhou
- Department of Medical Imaging, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Tianxia Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Chinese Academy of Sciences, Chaoyang District, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Yu Huang
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Qing Liu
- State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong, China
- Department of Medicine, University of Hong Kong, Hong Kong, China
| | - Tao Nie
- Clinical Department of Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Yanfang Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong, China
- Department of Pharmacy and Pharmacology, University of Hong Kong, Hong Kong, China
| | - Jianguo Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Chinese Academy of Sciences, Chaoyang District, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
- Corresponding author. (A.X.); (X.H.); (J.Z.)
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong, China
- Department of Medicine, University of Hong Kong, Hong Kong, China
- Department of Pharmacy and Pharmacology, University of Hong Kong, Hong Kong, China
- Corresponding author. (A.X.); (X.H.); (J.Z.)
| |
Collapse
|
13
|
Jala VR, Bodduluri SR, Ghosh S, Chheda Z, Singh R, Smith ME, Chilton PM, Fleming CJ, Mathis SP, Sharma RK, Knight R, Yan J, Haribabu B. Absence of CCR2 reduces spontaneous intestinal tumorigenesis in the Apc Min /+ mouse model. Int J Cancer 2021; 148:2594-2607. [PMID: 33497467 DOI: 10.1002/ijc.33477] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/15/2020] [Accepted: 01/08/2021] [Indexed: 12/19/2022]
Abstract
The biological activities of chemokine (C-C motif) ligand 2 (CCL2) are mediated via C-C chemokine receptor-2 (CCR2). Increased CCL2 level is associated with metastasis of many cancers. In our study, we investigated the role of the CCL2/CCR2 axis in the development of spontaneous intestinal tumorigenesis using the ApcMin/+ mouse model. Ablation of CCR2 in ApcMin/+ mice significantly increased the overall survival and reduced intestinal tumor burden. Immune cell analysis showed that CCR2-/- ApcMin/+ mice exhibited significant reduction in the myeloid cell population and increased interferon γ (IFN-γ) producing T cells both in spleen and mesenteric lymph nodes compared to ApcMin/+ mice. The CCR2-/- ApcMin/+ tumors showed significantly reduced levels of interleukin (IL)-17 and IL-23 and increased IFN-γ and Granzyme B compared to ApcMin/+ tumors. Transfer of CCR2+/+ ApcMin/+ CD4+ T cells into Rag2-/- mice led to development of colitis phenotype with increased CD4+ T cells hyper proliferation and IL-17 production. In contrast, adoptive transfer of CCR2-/- ApcMin/+ CD4+ T cells into Rag2-/- mice failed to enhance colonic inflammation or IL-17 production. These results a suggest novel additional role for CCR2, where it regulates migration of IL-17 producing cells mediating tumor-promoting inflammation in addition to its role in migration of tumor associated macrophages.
Collapse
Affiliation(s)
- Venkatakrishna Rao Jala
- Department of Microbiology and Immunology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| | - Sobha Rani Bodduluri
- Department of Microbiology and Immunology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| | - Sweta Ghosh
- Department of Microbiology and Immunology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| | - Zinal Chheda
- Department of Microbiology and Immunology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| | - Rajbir Singh
- Department of Microbiology and Immunology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| | - Michelle E Smith
- Department of Microbiology and Immunology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| | - Paula M Chilton
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA
| | - Christopher J Fleming
- Department of Microbiology and Immunology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| | - Steven Paul Mathis
- Department of Microbiology and Immunology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| | - Rajesh Kumar Sharma
- James Graham Brown Cancer Center, Department of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Rob Knight
- Center for Microbiome Innovation, University of California San Diego, La Jolla, California, USA
| | - Jun Yan
- Department of Microbiology and Immunology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| | - Bodduluri Haribabu
- Department of Microbiology and Immunology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
14
|
Masaki N, Adachi O, Katahira S, Saiki Y, Horii A, Kawamoto S, Saiki Y. Progression of vascular remodeling in pulmonary vein obstruction. J Thorac Cardiovasc Surg 2020; 160:777-790.e5. [PMID: 32222412 DOI: 10.1016/j.jtcvs.2020.01.098] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 12/24/2019] [Accepted: 01/20/2020] [Indexed: 01/24/2023]
Abstract
OBJECTIVES Pulmonary vein obstruction (PVO) frequently occurs after repair of total anomalous pulmonary vein connection with progression of intimal hyperplasia from the anastomotic site toward upstream pulmonary veins (PVs). However, the understanding of mechanism in PVO progression is constrained by lack of data derived from a physiological model of the disease, and no prophylaxis has been established. We developed a new PVO animal model, investigated the mechanisms of PVO progression, and examined a new prophylactic strategy. METHODS We developed a chronic PVO model using infant domestic pigs by cutting and resuturing the left lower PV followed by weekly hemodynamic parameter measurement and angiographic assessment of the anastomosed PV. Subsequently, we tested a novel therapeutic strategy with external application of rapamycin-eluting film to the anastomotic site. RESULTS We found the pig PVO model mimicked human PVO hemodynamically and histopathologically. This model exhibited increased expression levels of Ki-67 and phospho-mammalian target of rapamycin in smooth muscle-like cells at the anastomotic neointima. In addition, contractile to synthetic phenotypic transition; that is, dedifferentiation of smooth muscle cells and mammalian target of rapamycin pathway activation in the neointima of upstream PVs were observed. Rapamycin-eluting films externally applied around the anastomotic site inhibited the activation of mammalian target of rapamycin in the smooth muscle-like cells of neointima, and delayed PV anastomotic stenosis. CONCLUSIONS We demonstrate the evidence on dedifferentiation of smooth muscle-like cells and mammalian target of rapamycin pathway activation in the pathogenesis of PVO progression. Delivery of rapamycin to the anastomotic site from the external side delayed PV anastomotic stenosis, implicating a new therapeutic strategy to prevent PVO progression.
Collapse
Affiliation(s)
- Naoki Masaki
- Division of Cardiovascular Surgery, Tohoku University Graduate School of Medicine, Aoba-ku, Sendai, Japan
| | - Osamu Adachi
- Division of Cardiovascular Surgery, Tohoku University Graduate School of Medicine, Aoba-ku, Sendai, Japan
| | - Shintaro Katahira
- Division of Cardiovascular Surgery, Tohoku University Graduate School of Medicine, Aoba-ku, Sendai, Japan
| | - Yuriko Saiki
- Department of Molecular Pathology, Tohoku University Graduate School of Medicine, Aoba-ku, Sendai, Japan
| | - Akira Horii
- Department of Molecular Pathology, Tohoku University Graduate School of Medicine, Aoba-ku, Sendai, Japan
| | - Shunsuke Kawamoto
- Division of Cardiovascular Surgery, Tohoku University Graduate School of Medicine, Aoba-ku, Sendai, Japan
| | - Yoshikatsu Saiki
- Division of Cardiovascular Surgery, Tohoku University Graduate School of Medicine, Aoba-ku, Sendai, Japan.
| |
Collapse
|
15
|
Structure and Function of Porcine Arteries Are Preserved for up to 6 Days Using the HypoRP Cold-storage Solution. Transplantation 2020; 104:e125-e134. [PMID: 32000259 DOI: 10.1097/tp.0000000000003141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Maintaining functional vessels during preservation of vascularized composite allografts (VCAs) remains a major challenge. The University of Wisconsin (UW) solution has demonstrated significant short-term benefits (4-6 h). Here we determined whether the new hypothermic resuscitation and preservation solution HypoRP improves both structure, survival, and function of pig arteries during storage for up to 6 days. METHODS Using porcine swine mesenteric arteries, the effects of up to 6-day incubation in a saline (PBS), UW, or HypoRP solution on the structure, cell viability, metabolism, and function were determined. RESULTS After incubation at 4°C, for up to 6 days, the structures of the arteries were significantly disrupted, especially the tunica media, following incubation in PBS, in contrast with incubation in the HypoRP solution and to a lesser extent, in UW solution. Those disruptions were associated with increased active caspase 3 indicative of apoptosis. Additionally, while incubation in PBS led to a significant decrease in the metabolic activity, UW and HypoRP solutions allowed a stable to increased metabolic activity following 6 days of cold storage. Functional responsiveness to phenylephrine (PE) and sodium nitroprusside (SNP) decreased over time for artery rings stored in PBS and UW solution but not for those stored in HypoRP solution. Moreover, artery rings cold-stored in HypoRP solution were more sensitive to ATP. CONCLUSIONS The HypoRP solution improved long-term cold storage of porcine arteries by limiting structural alterations, including the collagen matrix, reducing apoptosis, and maintaining artery contraction-relaxation functions for up to 6 days.
Collapse
|
16
|
Ding X, Chen J, Wu C, Wang G, Zhou C, Chen S, Wang K, Zhang A, Ye P, Wu J, Chen S, Zhang H, Xu K, Wang S, Xia J. Nucleotide-Binding Oligomerization Domain-Like Receptor Protein 3 Deficiency in Vascular Smooth Muscle Cells Prevents Arteriovenous Fistula Failure Despite Chronic Kidney Disease. J Am Heart Assoc 2020; 8:e011211. [PMID: 30587058 PMCID: PMC6405733 DOI: 10.1161/jaha.118.011211] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background The arteriovenous fistula (AVF) is the preferred hemodialysis access for patients with chronic kidney disease. Chronic kidney disease can increase neointima formation, which greatly contributes to AVF failure by an unknown mechanism. Our study aimed to determine the role of nucleotide‐binding oligomerization domain‐like receptor protein 3 (NLRP3) in neointima formation induced by experimental AVFs in the presence of chronic kidney disease. Methods and Results From our findings, NLRP3 was upregulated in the intimal lesions of AVFs in both uremic mice and patients. Smooth muscle–specific knockout NLRP3 mice exhibited markedly decreased neointima formation in the outflow vein of AVFs. Compared with primary vascular smooth muscle cells isolated from control mice, those isolated from smooth muscle–specific knockout NLRP3 mice showed compromised proliferation, migration, phenotypic switching, and a weakened ability to activate mononuclear macrophages. To identify how NLRP3 functions, several small‐molecule inhibitors were used. The results showed that NLRP3 regulates smooth muscle cell proliferation and migration through Smad2/3 phosphorylation rather than through caspase‐1/interleukin‐1 signaling. Unexpectedly, the selective NLRP3‐inflammasome inhibitor MCC950 also repressed Smad2/3 phosphorylation and relieved chronic kidney disease–promoted AVF failure independent of macrophages. Conclusions Our findings suggest that NLRP3 in vascular smooth muscle cells may play a crucial role in uremia‐associated AVF failure and may be a promising therapeutic target for the treatment of AVF failure.
Collapse
Affiliation(s)
- Xiangchao Ding
- 1 Department of Cardiovascular Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Jiuling Chen
- 1 Department of Cardiovascular Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Chuangyan Wu
- 1 Department of Cardiovascular Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,2 Department of Thoracic Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Guohua Wang
- 1 Department of Cardiovascular Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Cheng Zhou
- 1 Department of Cardiovascular Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Shanshan Chen
- 3 Key Laboratory for Molecular Diagnosis of Hubei Province Central Hospital of Wuhan Tongji Medical College Huazhong University of Science and Technology Wuhan China.,4 Central Laboratory Central Hospital of Wuhan Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Ke Wang
- 6 Department of Respiratory and Critical Care Medicine Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Anchen Zhang
- 5 Department of Cardiovascular Medicine Central Hospital of Wuhan Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Ping Ye
- 5 Department of Cardiovascular Medicine Central Hospital of Wuhan Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Jie Wu
- 1 Department of Cardiovascular Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Shanshan Chen
- 1 Department of Cardiovascular Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Hao Zhang
- 1 Department of Cardiovascular Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Kaiying Xu
- 2 Department of Thoracic Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Sihua Wang
- 2 Department of Thoracic Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Jiahong Xia
- 1 Department of Cardiovascular Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| |
Collapse
|
17
|
Hudalla H, Michael Z, Christodoulou N, Willis GR, Fernandez-Gonzalez A, Filatava EJ, Dieffenbach P, Fredenburgh LE, Stearman RS, Geraci MW, Kourembanas S, Christou H. Carbonic Anhydrase Inhibition Ameliorates Inflammation and Experimental Pulmonary Hypertension. Am J Respir Cell Mol Biol 2019; 61:512-524. [PMID: 30951642 PMCID: PMC6775956 DOI: 10.1165/rcmb.2018-0232oc] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 04/02/2019] [Indexed: 01/07/2023] Open
Abstract
Inflammation and vascular smooth muscle cell (VSMC) phenotypic switching are causally linked to pulmonary arterial hypertension (PAH) pathogenesis. Carbonic anhydrase inhibition induces mild metabolic acidosis and exerts protective effects in hypoxic pulmonary hypertension. Carbonic anhydrases and metabolic acidosis are further known to modulate immune cell activation. To evaluate if carbonic anhydrase inhibition modulates macrophage activation, inflammation, and VSMC phenotypic switching in severe experimental pulmonary hypertension, pulmonary hypertension was assessed in Sugen 5416/hypoxia (SU/Hx) rats after treatment with acetazolamide or ammonium chloride (NH4Cl). We evaluated pulmonary and systemic inflammation and characterized the effect of carbonic anhydrase inhibition and metabolic acidosis in alveolar macrophages and bone marrow-derived macrophages (BMDMs). We further evaluated the treatment effects on VSMC phenotypic switching in pulmonary arteries and pulmonary artery smooth muscle cells (PASMCs) and corroborated some of our findings in lungs and pulmonary arteries of patients with PAH. Both patients with idiopathic PAH and SU/Hx rats had increased expression of lung inflammatory markers and signs of PASMC dedifferentiation in pulmonary arteries. Acetazolamide and NH4Cl ameliorated SU/Hx-induced pulmonary hypertension and blunted pulmonary and systemic inflammation. Expression of carbonic anhydrase isoform 2 was increased in alveolar macrophages from SU/Hx animals, classically (M1) and alternatively (M2) activated BMDMs, and lungs of patients with PAH. Carbonic anhydrase inhibition and acidosis had distinct effects on M1 and M2 markers in BMDMs. Inflammatory cytokines drove PASMC dedifferentiation, and this was inhibited by acetazolamide and acidosis. The protective antiinflammatory effect of acetazolamide in pulmonary hypertension is mediated by a dual mechanism of macrophage carbonic anhydrase inhibition and systemic metabolic acidosis.
Collapse
MESH Headings
- Acetazolamide/therapeutic use
- Acidosis/chemically induced
- Acidosis/complications
- Acidosis/immunology
- Ammonium Chloride/therapeutic use
- Animals
- Carbonic Anhydrase Inhibitors/therapeutic use
- Carbonic Anhydrases/physiology
- Cell Differentiation/drug effects
- Contractile Proteins/biosynthesis
- Contractile Proteins/genetics
- Drug Evaluation, Preclinical
- Humans
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/enzymology
- Hypertension, Pulmonary/etiology
- Hypertension, Pulmonary/pathology
- Hypoxia/complications
- Inflammation
- Macrophages/drug effects
- Macrophages/enzymology
- Macrophages, Alveolar/drug effects
- Macrophages, Alveolar/enzymology
- Male
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Protein Isoforms/antagonists & inhibitors
- Pulmonary Artery/pathology
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Rats
- Rats, Sprague-Dawley
Collapse
Affiliation(s)
- Hannes Hudalla
- Department of Pediatric Newborn Medicine and
- Department of Neonatology, Heidelberg University Children’s Hospital, Heidelberg, Germany
- Harvard Medical School, Boston, Massachusetts
| | - Zoe Michael
- Department of Pediatric Newborn Medicine and
- Harvard Medical School, Boston, Massachusetts
| | | | - Gareth R. Willis
- Harvard Medical School, Boston, Massachusetts
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, Massachusetts; and
| | - Angeles Fernandez-Gonzalez
- Harvard Medical School, Boston, Massachusetts
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, Massachusetts; and
| | | | - Paul Dieffenbach
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Laura E. Fredenburgh
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Robert S. Stearman
- Division of Pulmonary, Critical Care Medicine, Sleep, and Occupational Medicine, Department of Medicine, School of Medicine, Indiana University, Indianapolis, Indiana
| | - Mark W. Geraci
- Division of Pulmonary, Critical Care Medicine, Sleep, and Occupational Medicine, Department of Medicine, School of Medicine, Indiana University, Indianapolis, Indiana
| | - Stella Kourembanas
- Department of Pediatric Newborn Medicine and
- Harvard Medical School, Boston, Massachusetts
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, Massachusetts; and
| | - Helen Christou
- Department of Pediatric Newborn Medicine and
- Harvard Medical School, Boston, Massachusetts
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, Massachusetts; and
| |
Collapse
|
18
|
Rehfuss JP, DeSart KM, Rozowsky JM, O'Malley KA, Moldawer LL, Baker HV, Wang Y, Wu R, Nelson PR, Berceli SA. Hyperacute Monocyte Gene Response Patterns Are Associated With Lower Extremity Vein Bypass Graft Failure. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2019. [PMID: 29530886 DOI: 10.1161/circgen.117.001970] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND Despite being the definitive treatment for lower extremity peripheral arterial disease, vein bypass grafts fail in half of all cases. Early repair mechanisms after implantation, governed largely by the immune environment, contribute significantly to long-term outcomes. The current study investigates the early response patterns of circulating monocytes as a determinant of graft outcome. METHODS In 48 patients undergoing infrainguinal vein bypass grafting, the transcriptomes of circulating monocytes were analyzed preoperatively and at 1, 7, and 28 days post-operation. RESULTS Dynamic clustering algorithms identified 50 independent gene response patterns. Three clusters (64 genes) were differentially expressed, with a hyperacute response pattern defining those patients with failed versus patent grafts 12 months post-operation. A second independent data set, comprised of 96 patients subjected to major trauma, confirmed the value of these 64 genes in predicting an uncomplicated versus complicated recovery. Causal network analysis identified 8 upstream elements that regulate these mediator genes, and Bayesian analysis with a priori knowledge of the biological interactions was integrated to create a functional network describing the relationships among the regulatory elements and downstream mediator genes. Linear models predicted the removal of either STAT3 (signal transducer and activator of transcription 3) or MYD88 (myeloid differentiation primary response 88) to shift mediator gene expression levels toward those seen in successful grafts. CONCLUSIONS A novel combination of dynamic gene clustering, linear models, and Bayesian network analysis has identified a core set of regulatory genes whose manipulations could migrate vein grafts toward a more favorable remodeling phenotype.
Collapse
Affiliation(s)
- Jonathan P Rehfuss
- From the Malcom Randall Veterans Affairs Medical Center, Gainesville, FL (J.P.R., K.M.D., J.M.R., K.A.O., S.A.B.); Department of Surgery (J.P.R., K.M.D., J.M.R., K.A.O., L.L.M., S.A.B.) and Department of Molecular Genetics and Microbiology (H.V.B.), University of Florida, Gainesville; Department of Biostatistics, Rutgers University, New Brunswick, NJ (Y.W.); Center for Statistical Genetics, Pennsylvania State University, Hershey (R.W.); and Department of Surgery, University of South Florida, Tampa (P.R.N.)
| | - Kenneth M DeSart
- From the Malcom Randall Veterans Affairs Medical Center, Gainesville, FL (J.P.R., K.M.D., J.M.R., K.A.O., S.A.B.); Department of Surgery (J.P.R., K.M.D., J.M.R., K.A.O., L.L.M., S.A.B.) and Department of Molecular Genetics and Microbiology (H.V.B.), University of Florida, Gainesville; Department of Biostatistics, Rutgers University, New Brunswick, NJ (Y.W.); Center for Statistical Genetics, Pennsylvania State University, Hershey (R.W.); and Department of Surgery, University of South Florida, Tampa (P.R.N.)
| | - Jared M Rozowsky
- From the Malcom Randall Veterans Affairs Medical Center, Gainesville, FL (J.P.R., K.M.D., J.M.R., K.A.O., S.A.B.); Department of Surgery (J.P.R., K.M.D., J.M.R., K.A.O., L.L.M., S.A.B.) and Department of Molecular Genetics and Microbiology (H.V.B.), University of Florida, Gainesville; Department of Biostatistics, Rutgers University, New Brunswick, NJ (Y.W.); Center for Statistical Genetics, Pennsylvania State University, Hershey (R.W.); and Department of Surgery, University of South Florida, Tampa (P.R.N.)
| | - Kerri A O'Malley
- From the Malcom Randall Veterans Affairs Medical Center, Gainesville, FL (J.P.R., K.M.D., J.M.R., K.A.O., S.A.B.); Department of Surgery (J.P.R., K.M.D., J.M.R., K.A.O., L.L.M., S.A.B.) and Department of Molecular Genetics and Microbiology (H.V.B.), University of Florida, Gainesville; Department of Biostatistics, Rutgers University, New Brunswick, NJ (Y.W.); Center for Statistical Genetics, Pennsylvania State University, Hershey (R.W.); and Department of Surgery, University of South Florida, Tampa (P.R.N.)
| | - Lyle L Moldawer
- From the Malcom Randall Veterans Affairs Medical Center, Gainesville, FL (J.P.R., K.M.D., J.M.R., K.A.O., S.A.B.); Department of Surgery (J.P.R., K.M.D., J.M.R., K.A.O., L.L.M., S.A.B.) and Department of Molecular Genetics and Microbiology (H.V.B.), University of Florida, Gainesville; Department of Biostatistics, Rutgers University, New Brunswick, NJ (Y.W.); Center for Statistical Genetics, Pennsylvania State University, Hershey (R.W.); and Department of Surgery, University of South Florida, Tampa (P.R.N.)
| | - Henry V Baker
- From the Malcom Randall Veterans Affairs Medical Center, Gainesville, FL (J.P.R., K.M.D., J.M.R., K.A.O., S.A.B.); Department of Surgery (J.P.R., K.M.D., J.M.R., K.A.O., L.L.M., S.A.B.) and Department of Molecular Genetics and Microbiology (H.V.B.), University of Florida, Gainesville; Department of Biostatistics, Rutgers University, New Brunswick, NJ (Y.W.); Center for Statistical Genetics, Pennsylvania State University, Hershey (R.W.); and Department of Surgery, University of South Florida, Tampa (P.R.N.)
| | - Yaqun Wang
- From the Malcom Randall Veterans Affairs Medical Center, Gainesville, FL (J.P.R., K.M.D., J.M.R., K.A.O., S.A.B.); Department of Surgery (J.P.R., K.M.D., J.M.R., K.A.O., L.L.M., S.A.B.) and Department of Molecular Genetics and Microbiology (H.V.B.), University of Florida, Gainesville; Department of Biostatistics, Rutgers University, New Brunswick, NJ (Y.W.); Center for Statistical Genetics, Pennsylvania State University, Hershey (R.W.); and Department of Surgery, University of South Florida, Tampa (P.R.N.)
| | - Rongling Wu
- From the Malcom Randall Veterans Affairs Medical Center, Gainesville, FL (J.P.R., K.M.D., J.M.R., K.A.O., S.A.B.); Department of Surgery (J.P.R., K.M.D., J.M.R., K.A.O., L.L.M., S.A.B.) and Department of Molecular Genetics and Microbiology (H.V.B.), University of Florida, Gainesville; Department of Biostatistics, Rutgers University, New Brunswick, NJ (Y.W.); Center for Statistical Genetics, Pennsylvania State University, Hershey (R.W.); and Department of Surgery, University of South Florida, Tampa (P.R.N.)
| | - Peter R Nelson
- From the Malcom Randall Veterans Affairs Medical Center, Gainesville, FL (J.P.R., K.M.D., J.M.R., K.A.O., S.A.B.); Department of Surgery (J.P.R., K.M.D., J.M.R., K.A.O., L.L.M., S.A.B.) and Department of Molecular Genetics and Microbiology (H.V.B.), University of Florida, Gainesville; Department of Biostatistics, Rutgers University, New Brunswick, NJ (Y.W.); Center for Statistical Genetics, Pennsylvania State University, Hershey (R.W.); and Department of Surgery, University of South Florida, Tampa (P.R.N.)
| | - Scott A Berceli
- From the Malcom Randall Veterans Affairs Medical Center, Gainesville, FL (J.P.R., K.M.D., J.M.R., K.A.O., S.A.B.); Department of Surgery (J.P.R., K.M.D., J.M.R., K.A.O., L.L.M., S.A.B.) and Department of Molecular Genetics and Microbiology (H.V.B.), University of Florida, Gainesville; Department of Biostatistics, Rutgers University, New Brunswick, NJ (Y.W.); Center for Statistical Genetics, Pennsylvania State University, Hershey (R.W.); and Department of Surgery, University of South Florida, Tampa (P.R.N.).
| |
Collapse
|
19
|
Exercise Training-Induced Changes in MicroRNAs: Beneficial Regulatory Effects in Hypertension, Type 2 Diabetes, and Obesity. Int J Mol Sci 2018; 19:ijms19113608. [PMID: 30445764 PMCID: PMC6275070 DOI: 10.3390/ijms19113608] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 10/19/2018] [Indexed: 12/21/2022] Open
Abstract
MicroRNAs are small non-coding RNAs that regulate gene expression post-transcriptionally. They are involved in the regulation of physiological processes, such as adaptation to physical exercise, and also in disease settings, such as systemic arterial hypertension (SAH), type 2 diabetes mellitus (T2D), and obesity. In SAH, microRNAs play a significant role in the regulation of key signaling pathways that lead to the hyperactivation of the renin-angiotensin-aldosterone system, endothelial dysfunction, inflammation, proliferation, and phenotypic change in smooth muscle cells, and the hyperactivation of the sympathetic nervous system. MicroRNAs are also involved in the regulation of insulin signaling and blood glucose levels in T2D, and participate in lipid metabolism, adipogenesis, and adipocyte differentiation in obesity, with specific microRNA signatures involved in the pathogenesis of each disease. Many studies report the benefits promoted by exercise training in cardiovascular diseases by reducing blood pressure, glucose levels, and improving insulin signaling and lipid metabolism. The molecular mechanisms involved, however, remain poorly understood, especially regarding the participation of microRNAs in these processes. This review aimed to highlight microRNAs already known to be associated with SAH, T2D, and obesity, as well as their possible regulation by exercise training.
Collapse
|
20
|
Macarie RD, Vadana M, Ciortan L, Tucureanu MM, Ciobanu A, Vinereanu D, Manduteanu I, Simionescu M, Butoi E. The expression of MMP-1 and MMP-9 is up-regulated by smooth muscle cells after their cross-talk with macrophages in high glucose conditions. J Cell Mol Med 2018; 22:4366-4376. [PMID: 29992758 PMCID: PMC6111860 DOI: 10.1111/jcmm.13728] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 05/18/2018] [Indexed: 01/29/2023] Open
Abstract
Patients with diabetes mellitus have an increased risk of myocardial infarction and coronary artery disease-related death, exhibiting highly vulnerable plaques. Many studies have highlighted the major role of macrophages (MAC) and smooth muscle cells (SMC) and the essential part of metalloproteases (MMPs) in atherosclerotic plaque vulnerability. We hypothesize that in diabetes, the interplay between MAC and SMC in high glucose conditions may modify the expression of MMPs involved in plaque vulnerability. The SMC-MAC cross-talk was achieved using trans-well chambers, where human SMC were grown at the bottom and human MAC in the upper chamber in normal (NG) or high (HG) glucose concentration. After cross-talk, the conditioned media and cells were isolated and investigated for the expression of MMPs, MCP-1 and signalling molecules. We found that upon cross-talk with MAC in HG, SMC exhibit: (i) augmented expression of MMP-1 and MMP-9; (ii) significant increase in the enzymatic activity of MMP-9; (iii) higher levels of soluble MCP-1 chemokine which is functionally active and involved in MMPs up-regulation; (iv) activated PKCα signalling pathway which, together with NF-kB are responsible for MMP-1 and MMP-9 up-regulation, and (v) impaired function of collagen assembly. Taken together, our data indicate that MCP-1 released by cell cross-talk in diabetic conditions binds to CCR2 and triggers MMP-1 and MMP-9 over-expression and activity, features that could explain the high vulnerability of atherosclerotic plaque found at diabetic patients.
Collapse
Affiliation(s)
- Razvan Daniel Macarie
- Institute of Cellular Biology and Pathology “Nicolae Simionescu”, Biopathology and Therapy of InflammationBucharestRomania
| | - Mihaela Vadana
- Institute of Cellular Biology and Pathology “Nicolae Simionescu”, Biopathology and Therapy of InflammationBucharestRomania
| | - Letitia Ciortan
- Institute of Cellular Biology and Pathology “Nicolae Simionescu”, Biopathology and Therapy of InflammationBucharestRomania
| | - Monica M. Tucureanu
- Institute of Cellular Biology and Pathology “Nicolae Simionescu”, Biopathology and Therapy of InflammationBucharestRomania
| | - Andrea Ciobanu
- Cardiology DepartmentCarol Davila University of Medicine and Pharmacy, University and Emergency Hospital BucharestBucharestRomania
| | - Dragos Vinereanu
- Cardiology DepartmentCarol Davila University of Medicine and Pharmacy, University and Emergency Hospital BucharestBucharestRomania
| | - Ileana Manduteanu
- Institute of Cellular Biology and Pathology “Nicolae Simionescu”, Biopathology and Therapy of InflammationBucharestRomania
| | - Maya Simionescu
- Institute of Cellular Biology and Pathology “Nicolae Simionescu”, Biopathology and Therapy of InflammationBucharestRomania
| | - Elena Butoi
- Institute of Cellular Biology and Pathology “Nicolae Simionescu”, Biopathology and Therapy of InflammationBucharestRomania
| |
Collapse
|
21
|
Roy C, Tabiasco J, Caillon A, Delneste Y, Merot J, Favre J, Guihot AL, Martin L, Nascimento DC, Ryffel B, Robson SC, Sévigny J, Henrion D, Kauffenstein G. Loss of vascular expression of nucleoside triphosphate diphosphohydrolase-1/CD39 in hypertension. Purinergic Signal 2017; 14:73-82. [PMID: 29236227 DOI: 10.1007/s11302-017-9597-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 11/29/2017] [Indexed: 12/21/2022] Open
Abstract
Ectonucleoside triphosphate diphosphohydrolase-1, the major vascular/immune ectonucleotidase, exerts anti-thrombotic and immunomodulatory actions by hydrolyzing extracellular nucleotides (danger signals). Hypertension is characterized by vascular wall remodeling, endothelial dysfunction, and immune infiltration. Here our aim was to investigate the impact of arterial hypertension on CD39 expression and activity in mice. Arterial expression of CD39 was determined by reverse transcription quantitative real-time PCR in experimental models of hypertension, including angiotensin II (AngII)-treated mice (1 mg/kg/day, 21 days), deoxycorticosterone acetate-salt mice (1% salt and uninephrectomy, 21 days), and spontaneously hypertensive rats. A decrease in CD39 expression occurred in the resistance and conductance arteries of hypertensive animals with no effect on lymphoid organs. In AngII-treated mice, a decrease in CD39 protein levels (Western blot) was corroborated by reduced arterial nucleotidase activity, as evaluated by fluorescent (etheno)-ADP hydrolysis. Moreover, serum-soluble ADPase activity, supported by CD39, was significantly decreased in AngII-treated mice. Experiments were conducted in vitro on vascular cells to determine the elements underlying this downregulation. We found that CD39 transcription was reduced by proinflammatory cytokines interleukin (IL)-1β and tumor necrosis factor alpha on vascular smooth muscle cells and by IL-6 and anti-inflammatory and profibrotic cytokine transforming growth factor beta 1 on endothelial cells. In addition, CD39 expression was downregulated by mechanical stretch on vascular cells. Arterial expression and activity of CD39 were decreased in hypertension as a result of both a proinflammatory environment and mechanical strain exerted on vascular cells. Reduced ectonucleotidase activity may alter the vascular condition, thus enhancing arterial damage, remodeling, or thrombotic events.
Collapse
Affiliation(s)
- Charlotte Roy
- MITOVASC Institute - UMR CNRS 6015 INSERM U1083 University of Angers, Angers, France
| | - Julie Tabiasco
- CNRS UMR 6299, INSERM 892, CRCNA, University of Angers, Angers, France
| | - Antoine Caillon
- MITOVASC Institute - UMR CNRS 6015 INSERM U1083 University of Angers, Angers, France
| | - Yves Delneste
- CNRS UMR 6299, INSERM 892, CRCNA, University of Angers, Angers, France.,University Hospital of Angers, Angers, France
| | - Jean Merot
- L'institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France
| | - Julie Favre
- MITOVASC Institute - UMR CNRS 6015 INSERM U1083 University of Angers, Angers, France
| | - Anne Laure Guihot
- MITOVASC Institute - UMR CNRS 6015 INSERM U1083 University of Angers, Angers, France
| | - Ludovic Martin
- MITOVASC Institute - UMR CNRS 6015 INSERM U1083 University of Angers, Angers, France.,University Hospital of Angers, Angers, France
| | - Daniele C Nascimento
- CNRS, UMR 7355, Orleans, France.,CNRS UMR 7355, INEM, University of Orleans, Orleans, France
| | - Bernhard Ryffel
- CNRS, UMR 7355, Orleans, France.,CNRS UMR 7355, INEM, University of Orleans, Orleans, France
| | - Simon C Robson
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard University, Boston, MA, USA
| | - Jean Sévigny
- Département de microbiologie-infectiologie et d'immunologie, Faculté de Médecine, Université Laval, Québec City, QC, G1V 0A6, Canada.,Centre de recherche du CHU de Québec - Université Laval, Québec City, QC, G1V 4G2, Canada
| | - Daniel Henrion
- MITOVASC Institute - UMR CNRS 6015 INSERM U1083 University of Angers, Angers, France.,University Hospital of Angers, Angers, France
| | - Gilles Kauffenstein
- MITOVASC Institute - UMR CNRS 6015 INSERM U1083 University of Angers, Angers, France. .,University Hospital of Angers, Angers, France.
| |
Collapse
|
22
|
Simons KH, Peters HAB, Jukema JW, de Vries MR, Quax PHA. A protective role of IRF3 and IRF7 signalling downstream TLRs in the development of vein graft disease via type I interferons. J Intern Med 2017; 282:522-536. [PMID: 28857295 DOI: 10.1111/joim.12679] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Toll like receptors (TLR) play an important role in vein graft disease (VGD). Interferon regulatory factors (IRF) 3 and 7 are the transcriptional regulators of type I interferons (IFN) and type I IFN responsive genes and are downstream factors of TLRs. Relatively little is known with regard to the interplay of IRFs and TLRs in VGD development. The aim of this study was to investigate the role of IRF3 and IRF7 signaling downstream TLRs and the effect of IRF3 and IRF7 in VGD. METHODS AND RESULTS In vitro activation of TLR3 induced IRF3 and IRF7 dependent IFNβ expression in bone marrow macrophages and vascular smooth muscle cells. Activation of TLR4 showed to regulate pro-inflammatory cytokines via IRF3. Vein graft surgery was performed in Irf3-/- , Irf7-/- and control mice. After 14 days Irf3-/- vein grafts had an increased vessel wall thickness compared to both control (P = 0.01) and Irf7-/- (P = 0.02) vein grafts. After 28 days, vessel wall thickness increased in Irf3-/- (P = 0.0003) and Irf7-/- (P = 0.04) compared to control vein grafts and also increased in Irf7-/- compared to Irf3-/- vein grafts (P = 0.02). Immunohistochemical analysis showed a significant higher influx of macrophages after 14 days in Irf3-/- vein grafts and after 28 days in Irf7-/- vein grafts compared to control vein grafts. CONCLUSIONS The present study is the first to describe a protective role of both IRF3 and IRF7 in VGD. IRFs regulate VGD downstream TLRs since Irf3-/- and Irf7-/- vein grafts show increased vessel wall thickening after respectively 14 and 28 days after surgery.
Collapse
Affiliation(s)
- K H Simons
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands.,Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - H A B Peters
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands.,Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - J W Jukema
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands.,Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - M R de Vries
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands.,Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - P H A Quax
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands.,Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
23
|
Jiang WJ, Ren WH, Liu XJ, Liu Y, Wu FJ, Sun LZ, Lan F, Du J, Zhang HJ. Disruption of mechanical stress in extracellular matrix is related to Stanford type A aortic dissection through down-regulation of Yes-associated protein. Aging (Albany NY) 2017; 8:1923-1939. [PMID: 27608489 PMCID: PMC5076445 DOI: 10.18632/aging.101033] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Accepted: 09/06/2016] [Indexed: 01/19/2023]
Abstract
In this study, we assessed whether the down-regulation of Yes-associated protein (YAP) is involved in the pathogenesis of extracellular matrix (ECM) mechanical stress-induced Stanford type A aortic dissection (STAAD). Human aortic samples were obtained from heart transplantation donors as normal controls and from STAAD patients undergoing surgical replacement of the ascending aorta. Decreased maximum aortic wall velocity, ECM disorders, increased VSMC apoptosis, and YAP down-regulation were identified in STAAD samples. In a mouse model of STAAD, YAP was down-regulated over time during the development of ECM damage, and increased VSMC apoptosis was also observed. YAP knockdown induced VSMC apoptosis under static conditions in vitro, and the change in mechanical stress induced YAP down-regulation and VSMC apoptosis. This study provides evidence that YAP down-regulation caused by the disruption of mechanical stress is associated with the development of STAAD via the induction of apoptosis in aortic VSMCs. As STAAD is among the most elusive and life-threatening vascular diseases, better understanding of the molecular pathogenesis of STAAD is critical to improve clinical outcome.
Collapse
Affiliation(s)
- Wen-Jian Jiang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 10029, China.,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 10029, China.,Beijing Laboratory for Cardiovascular Precision Medicine, Beijing 10029, China.,The Key Laboratory of Remodeling-related Cardiovascular Disease, Ministry of Education, Beijing 10029, China.,Beijing Aortic Disease Center, Cardiovascular Surgery Center, Beijing 10029, China.,Beijing Engineering Research Center for Vascular Prostheses, Beijing, China, 10029
| | - Wei-Hong Ren
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 10029, China.,Beijing Laboratory for Cardiovascular Precision Medicine, Beijing 10029, China
| | - Xu-Jie Liu
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 10029, China.,Beijing Laboratory for Cardiovascular Precision Medicine, Beijing 10029, China
| | - Yan Liu
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 10029, China.,Beijing Laboratory for Cardiovascular Precision Medicine, Beijing 10029, China
| | - Fu-Jian Wu
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 10029, China.,Beijing Laboratory for Cardiovascular Precision Medicine, Beijing 10029, China
| | - Li-Zhong Sun
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 10029, China.,Beijing Laboratory for Cardiovascular Precision Medicine, Beijing 10029, China.,The Key Laboratory of Remodeling-related Cardiovascular Disease, Ministry of Education, Beijing 10029, China.,Beijing Aortic Disease Center, Cardiovascular Surgery Center, Beijing 10029, China.,Beijing Engineering Research Center for Vascular Prostheses, Beijing, China, 10029
| | - Feng Lan
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 10029, China.,Beijing Laboratory for Cardiovascular Precision Medicine, Beijing 10029, China
| | - Jie Du
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 10029, China.,Beijing Laboratory for Cardiovascular Precision Medicine, Beijing 10029, China
| | - Hong-Jia Zhang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 10029, China.,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 10029, China.,Beijing Laboratory for Cardiovascular Precision Medicine, Beijing 10029, China.,The Key Laboratory of Remodeling-related Cardiovascular Disease, Ministry of Education, Beijing 10029, China.,Beijing Aortic Disease Center, Cardiovascular Surgery Center, Beijing 10029, China.,Beijing Engineering Research Center for Vascular Prostheses, Beijing, China, 10029
| |
Collapse
|
24
|
Pan J, Li K, Huang W, Zhang X. MiR-137 inhibited cell proliferation and migration of vascular smooth muscle cells via targeting IGFBP-5 and modulating the mTOR/STAT3 signaling. PLoS One 2017; 12:e0186245. [PMID: 29016699 PMCID: PMC5634643 DOI: 10.1371/journal.pone.0186245] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Accepted: 09/27/2017] [Indexed: 12/28/2022] Open
Abstract
Abnormal proliferation of vascular smooth muscle cells (VSMCs) contributes to the development of cardiovascular diseases. Studies have shown the great impact of microRNAs (miRNAs) on the cell proliferation of VSMCs. This study examined the effects of miR-137 on the cell proliferation and migration of VSMCs and also explored the underlying molecular mechanisms. The mRNA and protein expression levels were determined by qRT-PCR and western blot assays, respectively. The CCK-8 assay, wound healing assay and transwell migration assay were performed to measure cell proliferation and migration of VSMCs. The miR-137-targeted 3’untranslated region of insulin-like growth factor-binding protein-5 (IGFBP-5) was confirmed by luciferase reporter assay. Platelet-derived growth factor-bb (PDGF-bb) treatment enhanced cell proliferation and suppressed the expression of miR-137 in VSMCs. The gain-of-function and loss-of-function assays showed that overexpression of miR-137 suppressed the cell proliferation and migration, and also inhibited the expression of matrix genes of VSMCs; down-regulation of miR-137 had the opposite effects on VSMCs. Bioinformatics analysis and luciferase report assay results showed that IGFBP-5 was a direct target of miR-137, and miR-137 overexpression suppressed the IGFBP-5 expression and down-regulation of miR-137 increased the IGFBP-5 expression in VSMCs. PDGF-bb treatment also increased the IGFBP-5 mRNA expression. In addition, enforced expression of IGFBP-5 reversed the inhibitory effects of miR-137 on cell proliferation and migration of VSMCs. More importantly, overexpression of miR-137 also suppressed the activity of mTOR/STAT3 signaling in VSMCs. Taken together, the results suggest that miR-137 may suppress cell proliferation and migration of VSMCs via targeting IGFBP-5 and modulating mTOR/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Jin Pan
- Clinical Medical College, Xi'an Medical University, Xi'an City, Shaanxi Province, China
- * E-mail:
| | - Kai Li
- Department of Cardiology, the First Affiliated Hospital of Xi'an Medical University, Xi'an City, Shaanxi Province, China
| | - Wei Huang
- Clinical Medical College, Xi'an Medical University, Xi'an City, Shaanxi Province, China
| | - Xiaoqing Zhang
- Clinical Medical College, Xi'an Medical University, Xi'an City, Shaanxi Province, China
| |
Collapse
|
25
|
Garay-Lugo N, Domínguez-Lopez A, Miliar García A, Aguilar Barrera E, Gómez López M, Gómez Alcalá A, Martínez Godinez MDLA, Lara-Padilla E. n-3 Fatty acids modulate the mRNA expression of the Nlrp3 inflammasome and Mtor in the liver of rats fed with high-fat or high-fat/fructose diets. Immunopharmacol Immunotoxicol 2017; 38:353-63. [PMID: 27367537 DOI: 10.1080/08923973.2016.1208221] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
CONTEXT There is evidence that n-3 polyunsaturated fatty acids (n-3-PUFAs) can inhibit mTORC1, which should potentiate autophagy and eliminate NLRP3 inflammasome activity. OBJECTIVE Evaluate the effect of a high-fat or high-fat/fructose diet with and without n-3-PUFAs on hepatic gene expression. MATERIALS AND METHODS We examined the mRNA expression by RT-PCR of Mtor, Nlrp3, and other 22 genes associated with inflammation in rats livers after a 9-week diet. The dietary regimens were low-fat (control, CD), high-fat (HF), high-fat/fructose (HF-Fr), and also each of these supplemented with n-3-PUFAs (CD-n-3-PUFAs, HF-n-3-PUFAs, and HF-Fr-n-3-PUFAs). These data were processed by GeneMania and STRING databases. RESULTS Compared to the control, the HF group showed a significant increase (between p < 0.05 and p < 0.0001) in 20 of these genes (Il1b, Il18, Rxra, Nlrp3, Casp1, Il33, Tnf, Acaca, Mtor, Eif2s1, Eif2ak4, Nfkb1, Srebf1, Hif1a, Ppara, Ppard, Pparg, Mlxipl, Fasn y Scd1), and a decrease in Sirt1 (p < 0.05). With the HF-Fr diet, a significant increase (between p < 0.05 and p < 0.005) was also found in the expression of 16 evaluated genes (Srebf1, Mlxipl, Rxra, Abca1, Il33, Nfkb1, Hif1a, Pparg, Casp1, Il1b, Il-18, Tnf, Ppard, Acaca, Fasn, Scd1), along with a decrease in the transcription of Mtor and Elovl6 (p < 0.05). Contrarily, many of the genes whose expression increased with the HF and HF-Fr diets did not significantly increase with the HF-n-3-PUFAs or HF-Fr-n-3-PUFAs diet. DISCUSSION AND CONCLUSION We found the interrelation of the genes for the mTORC1 complex, the NLRP3 inflammasome, and other metabolically important proteins, and that these genes respond to n-3-PUFAs.
Collapse
Affiliation(s)
- Natalia Garay-Lugo
- a Laboratorio de Biología Molecular , Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis and Salvador Díaz Mirón , México , D.F , México
| | - Aarón Domínguez-Lopez
- a Laboratorio de Biología Molecular , Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis and Salvador Díaz Mirón , México , D.F , México
| | - Angel Miliar García
- a Laboratorio de Biología Molecular , Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis and Salvador Díaz Mirón , México , D.F , México
| | - Eliud Aguilar Barrera
- a Laboratorio de Biología Molecular , Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis and Salvador Díaz Mirón , México , D.F , México
| | - Modesto Gómez López
- a Laboratorio de Biología Molecular , Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis and Salvador Díaz Mirón , México , D.F , México
| | - Alejandro Gómez Alcalá
- a Laboratorio de Biología Molecular , Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis and Salvador Díaz Mirón , México , D.F , México
| | - Maria de Los Angeles Martínez Godinez
- a Laboratorio de Biología Molecular , Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis and Salvador Díaz Mirón , México , D.F , México
| | - Eleazar Lara-Padilla
- b Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis and Díaz Mirón , México , D.F , México
| |
Collapse
|
26
|
Prakash YS. Emerging concepts in smooth muscle contributions to airway structure and function: implications for health and disease. Am J Physiol Lung Cell Mol Physiol 2016; 311:L1113-L1140. [PMID: 27742732 DOI: 10.1152/ajplung.00370.2016] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 10/06/2016] [Indexed: 12/15/2022] Open
Abstract
Airway structure and function are key aspects of normal lung development, growth, and aging, as well as of lung responses to the environment and the pathophysiology of important diseases such as asthma, chronic obstructive pulmonary disease, and fibrosis. In this regard, the contributions of airway smooth muscle (ASM) are both functional, in the context of airway contractility and relaxation, as well as synthetic, involving production and modulation of extracellular components, modulation of the local immune environment, cellular contribution to airway structure, and, finally, interactions with other airway cell types such as epithelium, fibroblasts, and nerves. These ASM contributions are now found to be critical in airway hyperresponsiveness and remodeling that occur in lung diseases. This review emphasizes established and recent discoveries that underline the central role of ASM and sets the stage for future research toward understanding how ASM plays a central role by being both upstream and downstream in the many interactive processes that determine airway structure and function in health and disease.
Collapse
Affiliation(s)
- Y S Prakash
- Departments of Anesthesiology, and Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
27
|
Parpaleix A, Amsellem V, Houssaini A, Abid S, Breau M, Marcos E, Sawaki D, Delcroix M, Quarck R, Maillard A, Couillin I, Ryffel B, Adnot S. Role of interleukin-1 receptor 1/MyD88 signalling in the development and progression of pulmonary hypertension. Eur Respir J 2016; 48:470-83. [DOI: 10.1183/13993003.01448-2015] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 05/03/2016] [Indexed: 11/05/2022]
Abstract
Pulmonary artery smooth muscle cell (PA-SMC) proliferation and inflammation are key components of pulmonary arterial hypertension (PAH). Interleukin (IL)-1β binds to IL-1 receptor (R)1, thereby recruiting the molecular adaptor myeloid differentiation primary response protein 88 (MyD88) (involved in IL-1R1 and Toll-like receptor signal transduction) and inducing IL-1, IL-6 and tumour necrosis factor-α synthesis through nuclear factor-κB activation.We investigated the IL-1R1/MyD88 pathway in the pathogenesis of pulmonary hypertension.Marked IL-1R1 and MyD88 expression with predominant PA-SMC immunostaining was found in lungs from patients with idiopathic PAH, mice with hypoxia-induced pulmonary hypertension and SM22-5-HTT+mice. Elevations in lung IL-1β, IL-1R1, MyD88 and IL-6 preceded pulmonary hypertension in hypoxic mice. IL-1R1−/−, MyD88−/−and control mice given the IL-1R1 antagonist anakinra were protected similarly against hypoxic pulmonary hypertension and perivascular macrophage recruitment. Anakinra reversed pulmonary hypertension partially in SM22-5-HTT+mice and markedly in monocrotaline-treated rats. IL-1β-mediated stimulation of mouse PA-SMC growth was abolished by anakinra and absent in IL-1R1−/−and MyD88−/−mice. Gene deletion confined to the myeloid lineage (M.lys-Cre MyD88fl/flmice) decreased pulmonary hypertension severityversuscontrols, suggesting IL-1β-mediated effects on PA-SMCs and macrophages. The growth-promoting effect of media conditioned by M1 or M2 macrophages from M.lys-Cre MyD88fl/flmice was attenuated.Pulmonary vessel remodelling and inflammation during pulmonary hypertension require IL-1R1/MyD88 signalling. Targeting the IL-1β/IL-1R1 pathway may hold promise for treating human PAH.
Collapse
|
28
|
Wang B, Wei G, Liu B, Zhou X, Xiao H, Dong N, Li F. The Role of High Mobility Group Box 1 Protein in Interleukin-18-Induced Myofibroblastic Transition of Valvular Interstitial Cells. Cardiology 2016; 135:168-178. [PMID: 27395056 DOI: 10.1159/000447483] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 06/07/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND Increased levels of interleukin-18 (IL-18) and high mobility group box 1 protein (HMGB1) have been reported in patients with calcific aortic valve disease (CAVD). However, the role of IL-18 and HMGB1 in the modulation of the valvular interstitial cell (VIC) phenotype remains unclear. We hypothesized that HMGB1 mediates IL-18-induced myofibroblastic transition of VICs. METHODS The expression of IL-18, HMGB1 and α-smooth muscle actin (α-SMA) in human aortic valves was evaluated by immunohistochemical staining, real-time polymerase chain reaction and immunoblotting. Plasma concentrations of IL-18 and HMGB1 were measured using the ELISA kit. Cultured human aortic VICs were used as an in vitro model. RESULTS Immunohistochemistry and immunoblotting revealed increased levels of IL-18, HMGB1 and α-SMA in calcific valves. Circulating IL-18 and HMGB1 levels were also higher in CAVD patients. In vitro, IL-18 induced upregulation of HMGB1 and α-SMA in VICs. Moreover, IL-18 induced secretion of HMGB1 to the extracellular space and activation of nuclear factor kappa-B (NF-κB). Blockade of NF-κB abrogated the upregulation and release of HMGB1 induced by IL-18. Whereas HMGB1 inhibition attenuated the IL-18-induced expression of α-SMA, HMGB1 enhanced the effect of IL-18. CONCLUSIONS We demonstrated for the first time that both tissue and plasma levels of IL-18 and HMGB1 were increased in patients with CAVD. Mechanically, HMGB1 mediated IL-18-induced VIC myofibroblastic transition.
Collapse
Affiliation(s)
- Bo Wang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | | | | | | | | | | | | |
Collapse
|
29
|
Cross-talk between macrophages and smooth muscle cells impairs collagen and metalloprotease synthesis and promotes angiogenesis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1568-78. [DOI: 10.1016/j.bbamcr.2016.04.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 03/30/2016] [Accepted: 04/05/2016] [Indexed: 02/08/2023]
|
30
|
de Vries MR, Simons KH, Jukema JW, Braun J, Quax PHA. Vein graft failure: from pathophysiology to clinical outcomes. Nat Rev Cardiol 2016; 13:451-70. [PMID: 27194091 DOI: 10.1038/nrcardio.2016.76] [Citation(s) in RCA: 197] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Occlusive arterial disease is a leading cause of morbidity and mortality worldwide. Aside from balloon angioplasty, bypass graft surgery is the most commonly performed revascularization technique for occlusive arterial disease. Coronary artery bypass graft surgery is performed in patients with left main coronary artery disease and three-vessel coronary disease, whereas peripheral artery bypass graft surgery is used to treat patients with late-stage peripheral artery occlusive disease. The great saphenous veins are commonly used conduits for surgical revascularization; however, they are associated with a high failure rate. Therefore, preservation of vein graft patency is essential for long-term surgical success. With the exception of 'no-touch' techniques and lipid-lowering and antiplatelet (aspirin) therapy, no intervention has hitherto unequivocally proven to be clinically effective in preventing vein graft failure. In this Review, we describe both preclinical and clinical studies evaluating the pathophysiology underlying vein graft failure, and the latest therapeutic options to improve patency for both coronary and peripheral grafts.
Collapse
Affiliation(s)
- Margreet R de Vries
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Karin H Simons
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - J Wouter Jukema
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands.,Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Jerry Braun
- Department of Cardiothoracic Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Paul H A Quax
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| |
Collapse
|
31
|
A Novel Protective Function of 5-Methoxytryptophan in Vascular Injury. Sci Rep 2016; 6:25374. [PMID: 27146795 PMCID: PMC4857180 DOI: 10.1038/srep25374] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 04/18/2016] [Indexed: 12/22/2022] Open
Abstract
5-Methoxytryptophan (5-MTP), a 5-methoxyindole metabolite of tryptophan metabolism, was recently shown to suppress inflammatory mediator-induced cancer cell proliferation and migration. However, the role of 5-MTP in vascular disease is unknown. In this study, we investigated whether 5-MTP protects against vascular remodeling following arterial injury. Measurements of serum 5-MTP levels in healthy subjects and patients with coronary artery disease (CAD) showed that serum 5-MTP concentrations were inversely correlated with CAD. To test the role of 5-MTP in occlusive vascular disease, we subjected mice to a carotid artery ligation model of neointima formation and treated mice with vehicle or 5-MTP. Compared with vehicle-treated mice, 5-MTP significantly reduced intimal thickening by 40% 4 weeks after ligation. BrdU incorporation assays revealed that 5-MTP significantly reduced VSMC proliferation both in vivo and in vitro. Furthermore, 5-MTP reduced endothelial loss and detachment, ICAM-1 and VCAM-1 expressions, and inflammatory cell infiltration in the ligated arterial wall, suggesting attenuation of endothelial dysfunction. Signaling pathway analysis indicated that 5-MTP mediated its effects predominantly via suppressing p38 MAPK signaling in endothelial and VSMCs. Our data demonstrate a novel vascular protective function of 5-MTP against arterial injury-induced intimal hyperplasia. 5-MTP might be a therapeutic target for preventing and/or treating vascular remodeling.
Collapse
|
32
|
Kan X, Wu Y, Ma Y, Zhang C, Li P, Wu L, Zhang S, Li Y, Du J. Deficiency of IL-12p35 improves cardiac repair after myocardial infarction by promoting angiogenesis. Cardiovasc Res 2016; 109:249-259. [DOI: 10.1093/cvr/cvv255] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|
33
|
The ADP antagonist MRS2179 regulates the phenotype of smooth muscle cells to limit intimal hyperplasia. Cardiovasc Drugs Ther 2015; 29:23-9. [PMID: 25528944 DOI: 10.1007/s10557-014-6561-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE ADP plays an important part in platelet aggregation by activating P2Y1 and P2Y12 receptors. The ADP antagonist MRS2179 has been used in thrombosis-related treatments but its effects on vein graft (VG) remodeling is undefined. We examined the effect of MRS2179 on VG intimal hyperplasia and explored the mechanism of action. METHODS A mouse model of VG transplantation was established. Mice underwent surgery and received MRS2179 by intraperitoneal injection every other day for 3 weeks. VG remodeling was assessed 4-weeks later. Vascular smooth muscle cells (VSMCs) were isolated and treated with MRS2179. The effect of MRS2179 on the proliferation, migration and inflammatory-cytokine expression of VSMCs was also evaluated. RESULTS MRS2179 significantly inhibited VSMC proliferation compared with the control group. Significant inhibitory effects of MRS2179 on VSMC migration was observed in two-dimensional and three-dimensional models. The extent of intimal hyperplasia was significantly less in MRS2179 treated mice than in controls. Reduced migration of macrophage was found in MRS2179 treated mice. Expression of the inflammatory cytokines IL-1β and TNF-α was decreased significantly in the MRS2179 treated group. In addition, decreased phosphorylation was found on Akt, Erk1/2 and p38. CONCLUSIONS These data demonstrate that MRS2179 inhibits neointima formation in VGs by regulating the proliferation, and migration of VSMCs, macrophage migration, inflammatory-cytokine secretion and related signaling pathway. Our study provides novel insights regarding purinergic signaling in SMCs in vivo. The P2Y1 receptor may serve as a therapeutic target in neointima formation.
Collapse
|
34
|
Liu Y, Wang Y, Shi H, Jia L, Cheng J, Cui W, Li H, Li P, Du J. CARD9 mediates necrotic smooth muscle cell-induced inflammation in macrophages contributing to neointima formation of vein grafts. Cardiovasc Res 2015; 108:148-58. [PMID: 26243429 DOI: 10.1093/cvr/cvv211] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 07/27/2015] [Indexed: 12/11/2022] Open
Abstract
AIMS Inflammation plays an important role in the neointima formation of grafted veins. However, the initiation of inflammation in grafted veins is still unclear. Here, we investigated the role and underlying mechanism of an innate immunity signalling protein, caspase-associated recruitment domain 9 (CARD9) in vein grafts in mice. METHODS AND RESULTS In early murine vein grafts, we observed robust death of smooth muscle cells (SMCs), which was accompanied by infiltration of macrophages and expression of pro-inflammatory cytokines. Meanwhile, SMC necrosis was associated with the expression of pro-inflammatory cytokines in macrophages in vitro. To explore the mediators of necrotic SMC-induced inflammation in grafted veins from mice, we examined the expression of CARD family proteins and found CARD9 highly expressed in infiltrated macrophages of grafted veins. CARD9-knockout (KO) inhibited necrotic SMC-induced pro-inflammatory cytokine expression and NF-κB activation. Furthermore, CARD9-KO suppressed necrotic SMC-induced expression of VEGF in macrophages. Finally, CARD9-KO decreased neointima formation of grafted veins in mice. CONCLUSION The innate immune protein CARD9 in macrophages may mediate necrotic SMC-induced inflammation by activating NF-κB and contributed to neointima formation in the vein grafts.
Collapse
Affiliation(s)
- Yan Liu
- Beijing Anzhen Hospital, Capital Medical University, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Collaborative Innovative Research Center for Cardiovascular Diseases, Beijing 100029, China
| | - Ying Wang
- Beijing Anzhen Hospital, Capital Medical University, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Collaborative Innovative Research Center for Cardiovascular Diseases, Beijing 100029, China
| | - Hongtao Shi
- Beijing Anzhen Hospital, Capital Medical University, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Collaborative Innovative Research Center for Cardiovascular Diseases, Beijing 100029, China
| | - Lixin Jia
- Beijing Anzhen Hospital, Capital Medical University, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Collaborative Innovative Research Center for Cardiovascular Diseases, Beijing 100029, China
| | - Jizhong Cheng
- Beijing Anzhen Hospital, Capital Medical University, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Collaborative Innovative Research Center for Cardiovascular Diseases, Beijing 100029, China
| | - Wei Cui
- Beijing Anzhen Hospital, Capital Medical University, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Collaborative Innovative Research Center for Cardiovascular Diseases, Beijing 100029, China
| | - Huihua Li
- Beijing Anzhen Hospital, Capital Medical University, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Collaborative Innovative Research Center for Cardiovascular Diseases, Beijing 100029, China
| | - Ping Li
- Beijing Anzhen Hospital, Capital Medical University, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Collaborative Innovative Research Center for Cardiovascular Diseases, Beijing 100029, China
| | - Jie Du
- Beijing Anzhen Hospital, Capital Medical University, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Collaborative Innovative Research Center for Cardiovascular Diseases, Beijing 100029, China
| |
Collapse
|
35
|
Affiliation(s)
- Valerie Z Wall
- From the Departments of Pathology (V.Z.W., K.E.B.) and Medicine, Division of Metabolism, Endocrinology and Nutrition (K.E.B.), Diabetes and Obesity Center of Excellence, University of Washington School of Medicine, Seattle
| | - Karin E Bornfeldt
- From the Departments of Pathology (V.Z.W., K.E.B.) and Medicine, Division of Metabolism, Endocrinology and Nutrition (K.E.B.), Diabetes and Obesity Center of Excellence, University of Washington School of Medicine, Seattle.
| |
Collapse
|