1
|
Agius T, Emsley R, Lyon A, MacArthur MR, Kiesworo K, Faivre A, Stavart L, Lambelet M, Legouis D, de Seigneux S, Golshayan D, Lazeyras F, Yeh H, Markmann JF, Uygun K, Ocampo A, Mitchell SJ, Allagnat F, Déglise S, Longchamp A. Short-term hypercaloric carbohydrate loading increases surgical stress resilience by inducing FGF21. Nat Commun 2024; 15:1073. [PMID: 38316771 PMCID: PMC10844297 DOI: 10.1038/s41467-024-44866-3] [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: 08/18/2023] [Accepted: 01/08/2024] [Indexed: 02/07/2024] Open
Abstract
Dietary restriction promotes resistance to surgical stress in multiple organisms. Counterintuitively, current medical protocols recommend short-term carbohydrate-rich drinks (carbohydrate loading) prior to surgery, part of a multimodal perioperative care pathway designed to enhance surgical recovery. Despite widespread clinical use, preclinical and mechanistic studies on carbohydrate loading in surgical contexts are lacking. Here we demonstrate in ad libitum-fed mice that liquid carbohydrate loading for one week drives reductions in solid food intake, while nearly doubling total caloric intake. Similarly, in humans, simple carbohydrate intake is inversely correlated with dietary protein intake. Carbohydrate loading-induced protein dilution increases expression of hepatic fibroblast growth factor 21 (FGF21) independent of caloric intake, resulting in protection in two models of surgical stress: renal and hepatic ischemia-reperfusion injury. The protection is consistent across male, female, and aged mice. In vivo, amino acid add-back or genetic FGF21 deletion blocks carbohydrate loading-mediated protection from ischemia-reperfusion injury. Finally, carbohydrate loading induction of FGF21 is associated with the induction of the canonical integrated stress response (ATF3/4, NF-kB), and oxidative metabolism (PPARγ). Together, these data support carbohydrate loading drinks prior to surgery and reveal an essential role of protein dilution via FGF21.
Collapse
Affiliation(s)
- Thomas Agius
- Department of Vascular Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Raffaella Emsley
- Department of Vascular Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Arnaud Lyon
- Department of Vascular Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Michael R MacArthur
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Kevin Kiesworo
- Department of Vascular Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Anna Faivre
- Laboratory of Nephrology, Department of Internal Medicine Specialties and Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
- Service of Nephrology, Department of Internal Medicine Specialties, University Hospital of Geneva, Geneva, Switzerland
| | - Louis Stavart
- Transplantation Center, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Martine Lambelet
- Department of Vascular Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - David Legouis
- Laboratory of Nephrology, Department of Internal Medicine Specialties and Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
- Division of Intensive Care, Department of Acute Medicine, University Hospital of Geneva, Geneva, Switzerland
| | - Sophie de Seigneux
- Laboratory of Nephrology, Department of Internal Medicine Specialties and Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
- Service of Nephrology, Department of Internal Medicine Specialties, University Hospital of Geneva, Geneva, Switzerland
| | - Déla Golshayan
- Transplantation Center, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Francois Lazeyras
- Department of Radiology and Medical Informatics, University of Geneva, Geneva, Switzerland
- Center for Biomedical Imaging (CIBM), Geneva, Switzerland
| | - Heidi Yeh
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - James F Markmann
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Korkut Uygun
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Alejandro Ocampo
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Sarah J Mitchell
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Florent Allagnat
- Department of Vascular Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Sébastien Déglise
- Department of Vascular Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Alban Longchamp
- Department of Vascular Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland.
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Chai Y, Shangguan L, Yu H, Sun Y, Huang X, Zhu Y, Wang H, Liu Y. Near Infrared Light-Activatable Platelet-Mimicking NIR-II NO Nano-Prodrug for Precise Atherosclerosis Theranostics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304994. [PMID: 38037484 PMCID: PMC10797437 DOI: 10.1002/advs.202304994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/30/2023] [Indexed: 12/02/2023]
Abstract
Atherosclerosis is a chronic inflammatory disease that affects arteries and is the main cause of cardiovascular disease. Atherosclerotic plaque formation is usually asymptomatic and does not manifest until the occurrence of clinical events. Therefore, early diagnosis and treatment of atherosclerotic plaques is particularly important. Here, a series of NIR-II fluorescent dyes (RBT-NH) are developed for three photoresponsive NO prodrugs (RBT-NO), which can be controllably triggered by 808 nm laser to release NO and turn on the NIR-II emission in the clinical medicine "therapeutic window". Notably, RBT3-NO is selected for its exhibited high NO releasing efficiency and superior fluorescence signal enhancement. Subsequently, a platelet-mimicking nano-prodrug system (RBT3-NO-PEG@PM) is constructed by DSPE-mPEG5k and platelet membrane (PM) for effectively targeted diagnosis and therapy of atherosclerosis in mice. The results indicate that this platelet-mimicking NO nano-prodrug system can reduce the accumulation of lipids at the sites of atherosclerotic plaques, improve the inflammatory response at the lesion sites, and promote endothelial cell migration, thereby slowing the progression of plaques.
Collapse
Affiliation(s)
- Yun Chai
- State Key Laboratory of Natural Medicines, School of EngineeringChina Pharmaceutical UniversityNanjing211198China
| | - Lina Shangguan
- State Key Laboratory of Natural Medicines, School of EngineeringChina Pharmaceutical UniversityNanjing211198China
| | - Hui Yu
- State Key Laboratory of Natural Medicines, School of EngineeringChina Pharmaceutical UniversityNanjing211198China
| | - Ye Sun
- State Key Laboratory of Natural Medicines, School of EngineeringChina Pharmaceutical UniversityNanjing211198China
| | - Xiaoyan Huang
- State Key Laboratory of Natural Medicines, School of EngineeringChina Pharmaceutical UniversityNanjing211198China
| | - Yanyan Zhu
- State Key Laboratory of Natural Medicines, School of EngineeringChina Pharmaceutical UniversityNanjing211198China
| | - Hai‐Yan Wang
- School of Mechanical EngineeringSoutheast UniversityNanjing211189China
| | - Yi Liu
- State Key Laboratory of Natural Medicines, School of EngineeringChina Pharmaceutical UniversityNanjing211198China
| |
Collapse
|
3
|
Déglise S, Bechelli C, Allagnat F. Vascular smooth muscle cells in intimal hyperplasia, an update. Front Physiol 2023; 13:1081881. [PMID: 36685215 PMCID: PMC9845604 DOI: 10.3389/fphys.2022.1081881] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/12/2022] [Indexed: 01/05/2023] Open
Abstract
Arterial occlusive disease is the leading cause of death in Western countries. Core contemporary therapies for this disease include angioplasties, stents, endarterectomies and bypass surgery. However, these treatments suffer from high failure rates due to re-occlusive vascular wall adaptations and restenosis. Restenosis following vascular surgery is largely due to intimal hyperplasia. Intimal hyperplasia develops in response to vessel injury, leading to inflammation, vascular smooth muscle cells dedifferentiation, migration, proliferation and secretion of extra-cellular matrix into the vessel's innermost layer or intima. In this review, we describe the current state of knowledge on the origin and mechanisms underlying the dysregulated proliferation of vascular smooth muscle cells in intimal hyperplasia, and we present the new avenues of research targeting VSMC phenotype and proliferation.
Collapse
Affiliation(s)
| | | | - Florent Allagnat
- Department of Vascular Surgery, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
4
|
Pharmacological prevention of intimal hyperplasia: A state-of-the-art review. Pharmacol Ther 2022; 235:108157. [PMID: 35183591 DOI: 10.1016/j.pharmthera.2022.108157] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 02/04/2022] [Accepted: 02/14/2022] [Indexed: 02/06/2023]
Abstract
Intimal hyperplasia (IH) occurs in a considerable number of cases of blood vessel reconstruction by stenting or balloon angioplasty, venous bypass grafting, and arteriovenous dialysis accesses. It is triggered by endothelial injury during the vascular intervention and leads to vessel restenosis with life-threatening consequences for patients. To date, the drugs used for IH prevention in clinics-paclitaxel and rapalog drugs-have been focusing primarily on the vascular smooth muscle cell (VSMC) proliferation pathway of IH development. Limitations, such as endothelial toxicity and inappropriate drug administration timing, have spurred the search for new and efficient pharmacological approaches to control IH. In this state-of-the-art review, we present the pathways of IH development, focusing on the key events and actors involved in IH. Subsequently, we discuss different drugs and drug combinations interfering with these pathways based on their effect on peripheral circulation IH models in animal studies, or on clinical reports. The reports were obtained through an extensive search of peer-reviewed publications in Pubmed, Embase, and Google Scholar, with search equations composed based on five concepts around IH and their various combinations. To improve vascular intervention outcomes, rethinking of conventional therapeutic approaches to IH prevention is needed. Exploring local application of drugs and drug combinations acting on different pathophysiological pathways of IH development has the potential to provide effective and safe restenosis prevention.
Collapse
|
5
|
The Interplay of Endothelial P2Y Receptors in Cardiovascular Health: From Vascular Physiology to Pathology. Int J Mol Sci 2022; 23:ijms23115883. [PMID: 35682562 PMCID: PMC9180512 DOI: 10.3390/ijms23115883] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/19/2022] [Accepted: 05/23/2022] [Indexed: 12/19/2022] Open
Abstract
The endothelium plays a key role in blood vessel health. At the interface of the blood, it releases several mediators that regulate local processes that protect against the development of cardiovascular disease. In this interplay, there is increasing evidence for a role of extracellular nucleotides and endothelial purinergic P2Y receptors (P2Y-R) in vascular protection. Recent advances have revealed that endothelial P2Y1-R and P2Y2-R mediate nitric oxide-dependent vasorelaxation as well as endothelial cell proliferation and migration, which are processes involved in the regeneration of damaged endothelium. However, endothelial P2Y2-R, and possibly P2Y1-R, have also been reported to promote vascular inflammation and atheroma development in mouse models, with endothelial P2Y2-R also being described as promoting vascular remodeling and neointimal hyperplasia. Interestingly, at the interface with lipid metabolism, P2Y12-R has been found to trigger HDL transcytosis through endothelial cells, a process known to be protective against lipid deposition in the vascular wall. Better characterization of the role of purinergic P2Y-R and downstream signaling pathways in determination of the endothelial cell phenotype in healthy and pathological environments has clinical potential for the prevention and treatment of cardiovascular diseases.
Collapse
|
6
|
Macabrey D, Longchamp A, MacArthur MR, Lambelet M, Urfer S, Deglise S, Allagnat F. Sodium thiosulfate acts as a hydrogen sulfide mimetic to prevent intimal hyperplasia via inhibition of tubulin polymerisation. EBioMedicine 2022; 78:103954. [PMID: 35334307 PMCID: PMC8941337 DOI: 10.1016/j.ebiom.2022.103954] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 03/08/2022] [Accepted: 03/08/2022] [Indexed: 11/06/2022] Open
Abstract
Background Intimal hyperplasia (IH) remains a major limitation in the long-term success of any type of revascularisation. IH is due to vascular smooth muscle cell (VSMC) dedifferentiation, proliferation and migration. The gasotransmitter Hydrogen Sulfide (H2S), mainly produced in blood vessels by the enzyme cystathionine- γ-lyase (CSE), inhibits IH in pre-clinical models. However, there is currently no H2S donor available to treat patients. Here we used sodium thiosulfate (STS), a clinically-approved source of sulfur, to limit IH. Methods Low density lipoprotein receptor deleted (LDLR−/−), WT or Cse-deleted (Cse−/−) male mice randomly treated with 4 g/L STS in the water bottle were submitted to focal carotid artery stenosis to induce IH. Human vein segments were maintained in culture for 7 days to induce IH. Further in vitro studies were conducted in primary human vascular smooth muscle cells (VSMCs). Findings STS inhibited IH in WT mice, as well as in LDLR−/− and Cse−/− mice, and in human vein segments. STS inhibited cell proliferation in the carotid artery wall and in human vein segments. STS increased polysulfides in vivo and protein persulfidation in vitro, which correlated with microtubule depolymerisation, cell cycle arrest and reduced VSMC migration and proliferation. Interpretation STS, a drug used for the treatment of cyanide poisoning and calciphylaxis, protects against IH in a mouse model of arterial restenosis and in human vein segments. STS acts as an H2S donor to limit VSMC migration and proliferation via microtubule depolymerisation. Funding This work was supported by the Swiss National Science Foundation (grant FN-310030_176158 to FA and SD and PZ00P3-185927 to AL); the Novartis Foundation to FA; and the Union des Sociétés Suisses des Maladies Vasculaires to SD, and the Fondation pour la recherche en chirurgie vasculaire et thoracique.
Collapse
|
7
|
Hildebrand S, Ibrahim M, Schlitzer A, Maegdefessel L, Röll W, Pfeifer A. PDGF regulates guanylate cyclase expression and cGMP signaling in vascular smooth muscle. Commun Biol 2022; 5:197. [PMID: 35241778 PMCID: PMC8894477 DOI: 10.1038/s42003-022-03140-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 02/08/2022] [Indexed: 11/17/2022] Open
Abstract
The nitric oxide-cGMP (NO-cGMP) pathway is of outstanding importance for vascular homeostasis and has multiple beneficial effects in vascular disease. Neointimal hyperplasia after vascular injury is caused by increased proliferation and migration of vascular smooth muscle cells (VSMCs). However, the role of NO-cGMP signaling in human VSMCs in this process is still not fully understood. Here, we investigate the interaction between platelet derived growth factor (PDGF)-signaling, one of the major contributors to neointimal hyperplasia, and the cGMP pathway in vascular smooth muscle, focusing on NO-sensitive soluble guanylyl cyclase (sGC). We show that PDGF reduces sGC expression by activating PI3K and Rac1, which in turn alters Notch ligand signaling. These data are corroborated by gene expression analysis in human atheromas, as well as immunohistological analysis of diseased and injured arteries. Collectively, our data identify the crosstalk between PDGF and NO/sGC signaling pathway in human VSMCs as a potential target to tackle neointimal hyperplasia. PDGF reduces expression of nitric oxide-sensitive soluble guanylyl cyclase (NO-sGC) through PI3K-P-Rex1-Rac1 signaling in vascular smooth muscle cells. These insights provide possible avenues to prevent dysregulation of NO/cGMP signaling in vascular disease.
Collapse
Affiliation(s)
- Staffan Hildebrand
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany.
| | - Mohamed Ibrahim
- Quantitative Systems Biology, LIMES-Institute (Life and Medical Sciences Bonn), University of Bonn, Bonn, Germany
| | - Andreas Schlitzer
- Quantitative Systems Biology, LIMES-Institute (Life and Medical Sciences Bonn), University of Bonn, Bonn, Germany
| | - Lars Maegdefessel
- Experimental Vascular Surgery and Medicine, Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar - Technical University Munich, Munich, Germany.,Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Wilhelm Röll
- Department of Cardiac Surgery, University of Bonn, Bonn, Germany
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany.
| |
Collapse
|
8
|
Radial artery lumen diameter and intima thickness in patients with abdominal aortic aneurysm. JVS Vasc Sci 2022; 3:274-284. [PMID: 36052216 PMCID: PMC9424594 DOI: 10.1016/j.jvssci.2022.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/22/2022] [Indexed: 11/21/2022] Open
|
9
|
Macabrey D, Deslarzes-Dubuis C, Longchamp A, Lambelet M, Ozaki CK, Corpataux JM, Allagnat F, Déglise S. Hydrogen Sulphide Release via the Angiotensin Converting Enzyme Inhibitor Zofenopril Prevents Intimal Hyperplasia in Human Vein Segments and in a Mouse Model of Carotid Artery Stenosis. Eur J Vasc Endovasc Surg 2021; 63:336-346. [PMID: 34916111 DOI: 10.1016/j.ejvs.2021.09.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 09/01/2021] [Accepted: 09/17/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Hypertension is a major risk factor for intimal hyperplasia (IH) and re-stenosis following vascular and endovascular interventions. Preclinical studies suggest that hydrogen sulphide (H2S), an endogenous gasotransmitter, limits re-stenosis. While there is no clinically available pure H2S releasing compound, the sulfhydryl containing angiotensin converting enzyme inhibitor zofenopril is a source of H2S. Here, it was hypothesised that zofenopril, due to H2S release, would be superior to other non-sulfhydryl containing angiotensin converting enzyme inhibitors (ACEi) in reducing intimal hyperplasia. METHODS Spontaneously hypertensive male Cx40 deleted mice (Cx40-/-) or wild type (WT) littermates were randomly treated with enalapril 20 mg or zofenopril 30 mg. Discarded human vein segments and primary human smooth muscle cells (SMCs) were treated with the active compound enalaprilat or zofenoprilat. IH was evaluated in mice 28 days after focal carotid artery stenosis surgery and in human vein segments cultured for seven days ex vivo. Human primary smooth muscle cell (SMC) proliferation and migration were studied in vitro. RESULTS Compared with control animals (intima/media thickness 2.3 ± 0.33 μm), enalapril reduced IH in Cx40-/- hypertensive mice by 30% (1.7 ± 0.35 μm; p = .037), while zofenopril abrogated IH (0.4 ± 0.16 μm; p < .002 vs. control and p > .99 vs. sham operated Cx40-/- mice). In WT normotensive mice, enalapril had no effect (0.9665 ± 0.2 μm in control vs. 1.140 ± 0.27 μm; p > .99), while zofenopril also abrogated IH (0.1623 ± 0.07 μm; p < .008 vs. control and p > .99 vs. sham operated WT mice). Zofenoprilat, but not enalaprilat, also prevented IH in human vein segments ex vivo. The effect of zofenopril on carotid and SMCs correlated with reduced SMC proliferation and migration. Zofenoprilat inhibited the mitogen activated protein kinase and mammalian target of rapamycin pathways in SMCs and human vein segments. CONCLUSION Zofenopril provides extra beneficial effects compared with non-sulfhydryl ACEi in reducing SMC proliferation and re-stenosis, even in normotensive animals. These findings may hold broad clinical implications for patients suffering from vascular occlusive diseases and hypertension.
Collapse
Affiliation(s)
- Diane Macabrey
- Department of Vascular Surgery, Lausanne University Hospital, Lausanne, Switzerland; Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Céline Deslarzes-Dubuis
- Department of Vascular Surgery, Lausanne University Hospital, Lausanne, Switzerland; Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Alban Longchamp
- Department of Vascular Surgery, Lausanne University Hospital, Lausanne, Switzerland; Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Martine Lambelet
- Department of Vascular Surgery, Lausanne University Hospital, Lausanne, Switzerland; Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Charles K Ozaki
- Department of Surgery and the Heart and Vascular Centre, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jean-Marc Corpataux
- Department of Vascular Surgery, Lausanne University Hospital, Lausanne, Switzerland; Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Florent Allagnat
- Department of Vascular Surgery, Lausanne University Hospital, Lausanne, Switzerland; Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland.
| | - Sébastien Déglise
- Department of Vascular Surgery, Lausanne University Hospital, Lausanne, Switzerland; Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
10
|
Longchamp A, Kaur K, Macabrey D, Dubuis C, Corpataux JM, Déglise S, Matson JB, Allagnat F. Hydrogen sulfide-releasing peptide hydrogel limits the development of intimal hyperplasia in human vein segments. Acta Biomater 2019; 97:374-384. [PMID: 31352106 PMCID: PMC6801028 DOI: 10.1016/j.actbio.2019.07.042] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 07/19/2019] [Accepted: 07/24/2019] [Indexed: 12/12/2022]
Abstract
Currently available interventions for vascular occlusive diseases suffer from high failure rates due to re-occlusive vascular wall adaptations, a process called intimal hyperplasia (IH). Naturally occurring hydrogen sulfide (H2S) works as a vasculoprotective gasotransmitter in vivo. However, given its reactive and hazardous nature, H2S is difficult to administer systemically. Here, we developed a hydrogel capable of localized slow release of precise amounts of H2S and tested its benefits on IH. The H2S-releasing hydrogel was prepared from a short peptide attached to an S-aroylthiooxime H2S donor. Upon dissolution in aqueous buffer, the peptide self-assembled into nanofibers, which formed a gel in the presence of calcium. This new hydrogel delivered H2S over the course of several hours, in contrast with fast-releasing NaHS. The H2S-releasing peptide/gel inhibited proliferation and migration of primary human vascular smooth muscle cells (VSMCs), while promoting proliferation and migration of human umbilical endothelial cells (ECs). Both NaHS and the H2S-releasing gel limited IH in human great saphenous vein segments obtained from vascular patients undergoing bypass surgery, with the H2S-releasing gel showing efficacy at a 5x lower dose than NaHS. These results suggest local perivascular H2S release as a new strategy to limit VSMC proliferation and IH while promoting EC proliferation, hence re-endothelialization. STATEMENT OF SIGNIFICANCE: Arterial occlusive disease is the leading cause of death in Western countries, yet current therapies suffer from high failure rates due to intimal hyperplasia (IH), a thickening of the vascular wall leading to secondary vessel occlusion. Hydrogen sulfide (H2S) is a gasotransmitter with vasculoprotective properties. Here we designed and synthesized a peptide-based H2S-releasing hydrogel and found that local application of the gel reduced IH in human vein segments obtained from patients undergoing bypass surgery. This work provides the first evidence of H2S efficacy against IH in human tissue, and the results show that the gel is more effective than NaHS, a common instantaneous H2S donor.
Collapse
Affiliation(s)
- Alban Longchamp
- Department of Vascular Surgery, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Kuljeet Kaur
- Virginia Tech, Department of Chemistry, Macromolecules Innovation Institute and Virginia Tech Center for Drug Discovery, Blacksburg, VA, USA
| | - Diane Macabrey
- Department of Vascular Surgery, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Celine Dubuis
- Department of Vascular Surgery, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Jean-Marc Corpataux
- Department of Vascular Surgery, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Sébastien Déglise
- Department of Vascular Surgery, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - John B Matson
- Virginia Tech, Department of Chemistry, Macromolecules Innovation Institute and Virginia Tech Center for Drug Discovery, Blacksburg, VA, USA.
| | - Florent Allagnat
- Department of Vascular Surgery, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland.
| |
Collapse
|
11
|
Mylonaki I, Allain E, Strano F, Allémann E, Corpataux JM, Meda P, Jordan O, Delie F, Rougemont AL, Haefliger JA, Saucy F. Evaluating intimal hyperplasia under clinical conditions. Interact Cardiovasc Thorac Surg 2019; 27:427-436. [PMID: 29617792 DOI: 10.1093/icvts/ivy101] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 03/04/2018] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES Open arterial revascularization using venous segments is frequently associated with the development of intimal hyperplasia (IH), leading to severe restenosis and graft failure. The lack of treatment to prevent this pathology is a major problem. Therefore, we generated a new porcine model, which closely mimics the clinical development of human IH, to test the therapeutic potential of candidate drugs. METHODS A patch of jugular vein was sutured to the right common carotid artery of pigs, to expose the vein to haemodynamic conditions of the arterial bed. Four weeks after surgery, the operated vessels which received no further treatment (the control group) were compared with (i) contralateral, non-operated vessels (the healthy group); (ii) vessels of pigs that received a perivascular application of a drug-free microparticle gel (the placebo group) and (iii) vessels of pigs that perioperatively received the same gel loaded with 10-mg atorvastatin (the atorvastatin group). RESULTS When compared with non-operated vessels, all operated segments displayed a sizable IH which was thicker in the venous patch than in the host artery. These alterations were associated with a thickening of the intima layer of both vessels in the absence of inflammation. The intima/media ratio has been significantly increased by 2000-fold in the vein patches. Perivascular application of atorvastatin did not prevent IH formation. However, the drug increased the adventitial neovascularization in the operated vessels. CONCLUSIONS The novel porcine model allows for monitoring IH formation under haemodynamic conditions which mimic clinical situations. It should facilitate the screening of innovative treatments to prevent restenosis.
Collapse
Affiliation(s)
- Ioanna Mylonaki
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland
| | - Elisabeth Allain
- Department of Vascular Surgery, Lausanne University Hospital, Lausanne, Switzerland
| | - Francesco Strano
- Department of Vascular Surgery, Lausanne University Hospital, Lausanne, Switzerland
| | - Eric Allémann
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland
| | - Jean-Marc Corpataux
- Department of Vascular Surgery, Lausanne University Hospital, Lausanne, Switzerland
| | - Paolo Meda
- Department of Cell Physiology and Metabolism, University of Geneva, Medical Center, Geneva, Switzerland
| | - Olivier Jordan
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland
| | - Florence Delie
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland
| | - Anne-Laure Rougemont
- Division of Clinical Pathology, Geneva University Hospitals, Geneva, Switzerland
| | | | - François Saucy
- Department of Vascular Surgery, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
12
|
Cui C, Wang X, Shang XM, Li L, Ma Y, Zhao GY, Song YX, Geng XB, Zhao BQ, Tian MR, Wang HL. lncRNA 430945 promotes the proliferation and migration of vascular smooth muscle cells via the ROR2/RhoA signaling pathway in atherosclerosis. Mol Med Rep 2019; 19:4663-4672. [PMID: 30957191 PMCID: PMC6522828 DOI: 10.3892/mmr.2019.10137] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 03/27/2019] [Indexed: 11/09/2022] Open
Abstract
The proliferation and migration of vascular smooth muscle cells (VSMCs) are major cellular events in hypertension‑induced vascular remodeling, which is closely involved in the progression of atherosclerosis (AS). Although long non‑coding RNAs (lncRNAs) are gaining recognition as novel regulators of VSMCs, their functioning and role in AS remain to be elucidated. In the present study, the role of lncRNA ENST00000430945 (lncRNA 430945) in AS was investigated. VSMCs transfected with a small interfering RNA (siRNA; si‑430945) and a negative control (si‑NC) were used. Cell Counting Kit‑8, wound‑healing and Transwell migration arrays were performed to determine whether lncRNA 430945 influenced VSMC proliferation and migration. Furthermore, the study examined whether a correlation exists between lncRNA 430945 and the receptor tyrosine kinase‑like orphan receptor 2 (ROR2) signaling pathway. It was found that the expression of lncRNA 430945 was high in human AS tissues, which in turn promoted angiotensin II (AngII)‑induced VSMC proliferation. Reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) and western blot analyses showed that lncRNA 430945 mediated the AngII‑induced upregulation of ROR2. In addition, the microarray and RT‑qPCR results showed that the expression of lncRNA 430945 was increased considerably in AS tissues. The downregulation of lncRNA 430945 significantly suppressed AngII‑induced VSMC proliferation and migration. In addition, ROR2 levels in VSMCs transfected with si‑430945 were markedly lower than those cells transfected with si‑NC. Additionally, western blotting showed that lncRNA 430945 activated the signaling pathways associated with ROR2 and Ras homolog gene family member A (RhoA). The upregulation of lncRNA 430945 in AS promoted the proliferation and migration of VSMCs via activation of the ROR2/RhoA signaling pathway. Therefore, targeting ROR2 or RhoA may be a promising technique in developing therapeutic strategies for treating AS.
Collapse
Affiliation(s)
- Chuan Cui
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
- Department of Cardiology, Tangshan Workers Hospital, Tangshan, Hebei 063000, P.R. China
| | - Xing Wang
- Department of Internal Medicine, Qian'an Hospital of Traditional Chinese Medicine, Qian'an, Hebei 064400, P.R. China
| | - Xiao-Ming Shang
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
- Department of Cardiology, Tangshan Workers Hospital, Tangshan, Hebei 063000, P.R. China
| | - Li Li
- Department of Cardiology, Tangshan Workers Hospital, Tangshan, Hebei 063000, P.R. China
| | - Yi Ma
- Department of Cardiology, Tangshan Workers Hospital, Tangshan, Hebei 063000, P.R. China
| | - Guo-Yu Zhao
- Department of Cardiology, Tangshan Workers Hospital, Tangshan, Hebei 063000, P.R. China
| | - Yu-Xin Song
- Department of Cardiology, Tangshan Workers Hospital, Tangshan, Hebei 063000, P.R. China
| | - Xue-Bin Geng
- Department of Cardiology, Tangshan Workers Hospital, Tangshan, Hebei 063000, P.R. China
| | - Bi-Qiong Zhao
- Department of Cardiology, Tangshan Workers Hospital, Tangshan, Hebei 063000, P.R. China
| | - Mei-Rong Tian
- Department of Cardiology, Tangshan Workers Hospital, Tangshan, Hebei 063000, P.R. China
| | - Hong-Ling Wang
- Department of Cardiology, Tangshan Workers Hospital, Tangshan, Hebei 063000, P.R. China
| |
Collapse
|
13
|
Le Gal L, Pellegrin M, Santoro T, Mazzolai L, Kurtz A, Meda P, Wagner C, Haefliger J. Connexin37-Dependent Mechanisms Selectively Contribute to Modulate Angiotensin II -Mediated Hypertension. J Am Heart Assoc 2019; 8:e010823. [PMID: 30943815 PMCID: PMC6507190 DOI: 10.1161/jaha.118.010823] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 01/30/2019] [Indexed: 12/23/2022]
Abstract
Background Gap junction channels made of Connexin37 (Cx37) are expressed by aortic endothelial and smooth muscle cells of hypertensive mice, as well as by the renin-secreting cells of kidneys. Methods and Results To decipher whether Cx37 has any role in hypertension, angiotensin II (Ang II ) was infused in normotensive wild-type and Cx37-deficient mice (Cx37-/-). After 2 to 4 weeks, the resulting increase in blood pressure was lower in Cx37-/- than in wild-type mice, suggesting an alteration in the Ang II response. To investigate this possibility, mice were submitted to a 2-kidney, 1-clip procedure, a renin-dependent model of hypertension. Two weeks after this clipping, Cx37-/- mice were less hypertensive than wild-type mice and, 2 weeks later, their blood pressure had returned to control values, in spite of abnormally high plasma renin levels. In contrast, Cx37-/- and wild-type mice that received N-nitro-l-arginine-methyl-ester, a renin-independent model of hypertension, featured a similar and sustained increase in blood pressure. The data indicate that loss of Cx37 selectively altered the Ang II -dependent pathways. Consistent with this conclusion, aortas of Cx37-/- mice featured an increased basal expression of the Ang II type 2 receptors ( AT 2R), and increased transcripts levels of downstream signaling proteins, such as Cnksr1 and Ptpn6 ( SHP -1). Accordingly, the response of Cx37-/- mice aortas to an ex vivo Ang II exposure was altered, since phosphorylation levels of several proteins of the Ang II pathway ( MLC 2, ERK , and AKT ) remained unchanged. Conclusions These findings provide evidence that Cx37 selectively influences Ang II signaling, mostly via a modulation of the expression of the Ang II type 2 receptor.
Collapse
Affiliation(s)
- Loïc Le Gal
- Department of MedicineUniversity of LausanneSwitzerland
| | - Maxime Pellegrin
- Division of AngiologyHeart and Vessel DepartmentCentre Hospitalier Universitaire VaudoisUniversity of LausanneSwitzerland
| | - Tania Santoro
- Department of MedicineUniversity of LausanneSwitzerland
| | - Lucia Mazzolai
- Division of AngiologyHeart and Vessel DepartmentCentre Hospitalier Universitaire VaudoisUniversity of LausanneSwitzerland
| | - Armin Kurtz
- Department of PhysiologyUniversity of RegensburgGermany
| | - Paolo Meda
- Department of Cell Physiology and MetabolismSchool of MedicineCMUUniversity of GenevaSwitzerland
| | | | | |
Collapse
|
14
|
Yilmaz E, Kaya‐Sezginer E, Yilmaz‐Oral D, Cengiz T, Bayatli N, Gur S. Effects of hydrogen sulphide donor, sodium hydrosulphide treatment on the erectile dysfunction in L‐NAME‐induced hypertensive rats. Andrologia 2019; 51:e13240. [DOI: 10.1111/and.13240] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/15/2018] [Accepted: 01/05/2019] [Indexed: 12/27/2022] Open
Affiliation(s)
- Enis Yilmaz
- Department of Pharmacology, Faculty of Pharmacy Ankara University Ankara Turkey
| | - Ecem Kaya‐Sezginer
- Department of Biochemistry, Faculty of Pharmacy Ankara University Ankara Turkey
| | - Didem Yilmaz‐Oral
- Department of Pharmacology, Faculty of Pharmacy Ankara University Ankara Turkey
- Department of Pharmacology, Faculty of Pharmacy Cukurova University Adana Turkey
| | - Tugba Cengiz
- Department of Pharmacology, Faculty of Pharmacy Ankara University Ankara Turkey
| | - Nur Bayatli
- Department of Pharmacology, Faculty of Pharmacy Ankara University Ankara Turkey
| | - Serap Gur
- Department of Pharmacology, Faculty of Pharmacy Ankara University Ankara Turkey
- Department of Urology Tulane University Health Sciences Center New Orleans Los Angeles
| |
Collapse
|
15
|
Mendes Garrido Abregú F, Gobetto MN, Juriol LV, Caniffi C, Elesgaray R, Tomat AL, Arranz C. Developmental programming of vascular dysfunction by prenatal and postnatal zinc deficiency in male and female rats. J Nutr Biochem 2018. [PMID: 29525532 DOI: 10.1016/j.jnutbio.2018.01.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Micronutrient malnutrition during intrauterine and postnatal growth may program cardiovascular diseases in adulthood. We examined whether moderate zinc restriction in male and female rats throughout fetal life, lactation and/or postweaning growth induces alterations that can predispose to the onset of vascular dysfunction in adulthood. Female Wistar rats were fed low- or control zinc diets from pregnancy to offspring weaning. After weaning, offspring were fed either a low- or a control zinc diet until 81 days. We evaluated systolic blood pressure (SBP), thoracic aorta morphology, nitric oxide (NO) system and vascular reactivity in 6- and/or 81-day-old offspring. At day 6, zinc-deficient male and female offspring showed a decrease in aortic NO synthase (NOS) activity accompanied by an increase in oxidative stress. Zinc-deficient 81-day-old male rats exhibited an increase in collagen deposition in tunica media, as well as lower activity of endothelial NOS (eNOS) that could not be reversed with an adequate zinc diet during postweaning life. Zinc deficiency programmed a reduction in eNOS protein expression and higher SBP only in males. Adult zinc-deficient rats of both sexes showed reduced vasodilator response dependent on eNOS activity and impaired aortic vasoconstrictor response to angiotensin-II associated with alterations in intracellular calcium mobilization. Female rats were less sensitive to the effects of zinc deficiency and exhibited higher eNOS activity and/or expression than males, without alterations in SBP or aortic histology. This work strengthens the importance of a balanced intake of micronutrients during perinatal growth to ensure adequate vascular function in adult life.
Collapse
Affiliation(s)
- Facundo Mendes Garrido Abregú
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Fisiología, Buenos Aires, Argentina; CONICET, Universidad de Buenos Aires Instituto de la Química y Metabolismo del Fármaco (IQUIMEFA), Buenos Aires, Argentina
| | - María Natalia Gobetto
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Fisiología, Buenos Aires, Argentina; CONICET, Universidad de Buenos Aires Instituto de la Química y Metabolismo del Fármaco (IQUIMEFA), Buenos Aires, Argentina
| | - Lorena Vanesa Juriol
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Fisiología, Buenos Aires, Argentina; CONICET, Universidad de Buenos Aires Instituto de la Química y Metabolismo del Fármaco (IQUIMEFA), Buenos Aires, Argentina
| | - Carolina Caniffi
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Fisiología, Buenos Aires, Argentina; CONICET, Universidad de Buenos Aires Instituto de la Química y Metabolismo del Fármaco (IQUIMEFA), Buenos Aires, Argentina
| | - Rosana Elesgaray
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Fisiología, Buenos Aires, Argentina; CONICET, Universidad de Buenos Aires Instituto de la Química y Metabolismo del Fármaco (IQUIMEFA), Buenos Aires, Argentina
| | - Analía Lorena Tomat
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Fisiología, Buenos Aires, Argentina; CONICET, Universidad de Buenos Aires Instituto de la Química y Metabolismo del Fármaco (IQUIMEFA), Buenos Aires, Argentina.
| | - Cristina Arranz
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Fisiología, Buenos Aires, Argentina; CONICET, Universidad de Buenos Aires Instituto de la Química y Metabolismo del Fármaco (IQUIMEFA), Buenos Aires, Argentina
| |
Collapse
|
16
|
Lambelet M, Terra LF, Fukaya M, Meyerovich K, Labriola L, Cardozo AK, Allagnat F. Dysfunctional autophagy following exposure to pro-inflammatory cytokines contributes to pancreatic β-cell apoptosis. Cell Death Dis 2018; 9:96. [PMID: 29367588 PMCID: PMC5833699 DOI: 10.1038/s41419-017-0121-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 10/31/2017] [Accepted: 11/02/2017] [Indexed: 12/14/2022]
Abstract
Type 1 diabetes (T1D) results from β-cell destruction due to concerted action of both innate and adaptive immune responses. Pro-inflammatory cytokines, such as interleukin-1β and interferon-γ, secreted by the immune cells invading islets of Langerhans, contribute to pancreatic β-cell death in T1D. Cytokine-induced endoplasmic reticulum (ER) stress plays a central role in β-cell demise. ER stress can modulate autophagic response; however, no study addressed the regulation of autophagy during the pathophysiology of T1D. In this study, we document that cytokines activate the AMPK-ULK-1 pathway while inhibiting mTORC1, which stimulates autophagy activity in an ER stress-dependent manner. On the other hand, time-course analysis of LC3-II accumulation in autophagosomes revealed that cytokines block the autophagy flux in an ER stress independent manner, leading to the formation of large dysfunctional autophagosomes and worsening of ER stress. Cytokines rapidly impair lysosome function, leading to lysosome membrane permeabilization, Cathepsin B leakage and lysosomal cell death. Blocking cathepsin activity partially protects against cytokine-induced or torin1-induced apoptosis, whereas blocking autophagy aggravates cytokine-induced CHOP overexpression and β-cell apoptosis. In conclusion, cytokines stimulate the early steps of autophagy while blocking the autophagic flux, which aggravate ER stress and trigger lysosomal cell death. Restoration of autophagy/lysosomal function may represent a novel strategy to improve β-cell resistance in the context of T1D.
Collapse
Affiliation(s)
- Martine Lambelet
- Department of Vascular Surgery, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Leticia F Terra
- Departamento de Bioquimica, Instituto de Quimica, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Makiko Fukaya
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Kira Meyerovich
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Leticia Labriola
- Departamento de Bioquimica, Instituto de Quimica, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Alessandra K Cardozo
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Florent Allagnat
- Department of Vascular Surgery, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland.
| |
Collapse
|
17
|
Yuhong L, Zhengzhong B, Feng T, Quanyu Y, Ge RL. L-arginine Attenuates Hypobaric Hypoxia-Induced Increase in Ornithine Decarboxylase 1. Wilderness Environ Med 2017; 28:285-290. [PMID: 28735657 DOI: 10.1016/j.wem.2017.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 05/15/2017] [Accepted: 05/27/2017] [Indexed: 11/18/2022]
Abstract
BACKGROUND Chronic hypoxia-induced pulmonary hypertension and vascular remodeling have been shown to be associated with ornithine decarboxylase 1 (ODC1). However, few animal studies have investigated the role of ODC1 in acute hypoxia. OBJECTIVES We investigated ODC1 gene expression, morphologic and functional changes, and the effect of L-arginine as an attenuator in lung tissues of rats exposed to acute hypobaric hypoxia at a simulated altitude of 6000 m. METHODS Sprague-Dawley rats exposed to simulated hypobaric hypoxia (6000 m) for 24, 48, or 72 hours were treated with L-arginine (L-arginine group, 20 mg/100 g intraperitoneal; n=15) or untreated (non-L-arginine group, n=15). Control rats (n=5) were maintained at 2260 m in a normal environment for the same amount of time but were treated without L-arginine. The mean pulmonary artery pressure was measured by PowerLab system. The morphologic and immunohistochemical changes in lung tissue were observed under a microscope. The mRNA and protein levels of ODC1 were measured by real-time polymerase chain reaction and Western-blot, respectively. RESULTS Hypobaric hypoxia induced pulmonary interstitial hyperemia and capillary expansion in the lungs of rats exposed to acute hypoxia at 6000 m. The mean pulmonary artery pressure and the mRNA and protein levels of ODC1 were significantly increased, which could be attenuated by treatment with L-arginine. CONCLUSIONS L-arginine attenuates acute hypobaric hypoxia-induced increase in mean pulmonary artery pressure and ODC1 gene expression in lung tissues of rats. ODC1 gene contributes to the development of hypoxic pulmonary hypertension.
Collapse
Affiliation(s)
- Li Yuhong
- Research Center for High Altitude Medicine; Key Laboratories Development Program of Qinghai Province; and Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Qinghai University, Xining, China; Department of Respiratory Medicine, the Affiliated Hospital of Qinghai University, Xining, China
| | - Bai Zhengzhong
- Research Center for High Altitude Medicine; Key Laboratories Development Program of Qinghai Province; and Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Qinghai University, Xining, China
| | - Tang Feng
- Research Center for High Altitude Medicine; Key Laboratories Development Program of Qinghai Province; and Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Qinghai University, Xining, China
| | - Yang Quanyu
- Research Center for High Altitude Medicine; Key Laboratories Development Program of Qinghai Province; and Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Qinghai University, Xining, China
| | - Ri-Li Ge
- Research Center for High Altitude Medicine; Key Laboratories Development Program of Qinghai Province; and Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Qinghai University, Xining, China.
| |
Collapse
|
18
|
Allagnat F, Dubuis C, Lambelet M, Le Gal L, Alonso F, Corpataux JM, Déglise S, Haefliger JA. Connexin37 reduces smooth muscle cell proliferation and intimal hyperplasia in a mouse model of carotid artery ligation. Cardiovasc Res 2017; 113:805-816. [PMID: 28449099 DOI: 10.1093/cvr/cvx079] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 04/20/2017] [Indexed: 12/11/2022] Open
Abstract
AIMS Intimal hyperplasia (IH) is an abnormal response to vessel injury characterized by the dedifferentiation, migration, and proliferation of quiescent vascular smooth muscle cells (VSMC) to form a neointima layer. Vascular connexins (Cx) are involved in the pathophysiology of various vascular diseases, and Cx43, the main Cx expressed in VSMC, has been shown to promote VSMC proliferation and IH. The aim of this study was to investigate the participation of another Cx, namely Cx37, in the formation of the neointima layer. METHODS AND RESULTS Wild-type (WT) and Cx37-deficient (Cx37-/-) C57BL/6J mice were subjected to carotid artery ligation (CAL), a model of vessel injury and IH. The neointima developed linearly in WT until 28 days post surgery. In contrast, the neointima layer was almost absent 14 days after surgery in Cx37-/- mice, and twice as more developed after 28 days compared to WT mice. This large neointima formation correlated with a two-fold increase in cell proliferation in the media and neointima regions between 14 and 28 days in Cx37-/- mice compared to WT mice. The CAL triggered Cx43 overexpression in the media and neointima layers of ligated carotids in WT mice, and selectively up-regulated Cx37 expression in the media layer, but not in the neointima layer. The de novo expression of Cx37 in human primary VSMC reduced cell proliferation and P-Akt levels, in association with lower Cx43 levels, whereas Cx43 overexpression increased P-Akt levels. CONCLUSION The presence of Cx37 in the media layer of injured arteries restrains VSMC proliferation and limits the development of IH, presumably by interfering with the pro-proliferative effect of Cx43 and the Akt pathway.
Collapse
MESH Headings
- Aged
- Animals
- Carotid Arteries/metabolism
- Carotid Arteries/pathology
- Carotid Arteries/surgery
- Carotid Artery Injuries/genetics
- Carotid Artery Injuries/metabolism
- Carotid Artery Injuries/pathology
- Carotid Stenosis/genetics
- Carotid Stenosis/metabolism
- Carotid Stenosis/pathology
- Cell Proliferation
- Cells, Cultured
- Connexin 43/metabolism
- Connexins/deficiency
- Connexins/genetics
- Connexins/metabolism
- Disease Models, Animal
- Female
- Humans
- Hyperplasia
- Ligation
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neointima
- Phosphorylation
- Proto-Oncogene Proteins c-akt/metabolism
- Signal Transduction
- Time Factors
- Gap Junction alpha-4 Protein
Collapse
Affiliation(s)
- Florent Allagnat
- Department of Vascular Surgery, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Céline Dubuis
- Department of Vascular Surgery, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Martine Lambelet
- Department of Vascular Surgery, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Loïc Le Gal
- Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV), Laboratory of Experimental Medicine, c/o Department of Physiology, Bugnon 7a, 1005 Lausanne, Switzerland
| | - Florian Alonso
- Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV), Laboratory of Experimental Medicine, c/o Department of Physiology, Bugnon 7a, 1005 Lausanne, Switzerland
| | - Jean-Marc Corpataux
- Department of Vascular Surgery, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Sébastien Déglise
- Department of Vascular Surgery, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Jacques-Antoine Haefliger
- Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV), Laboratory of Experimental Medicine, c/o Department of Physiology, Bugnon 7a, 1005 Lausanne, Switzerland
| |
Collapse
|