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
|
Kip P, Sluiter TJ, MacArthur MR, Tao M, Jung J, Mitchell SJ, Kooijman S, Kruit N, Gorham J, Seidman JG, Quax PHA, Aikawa M, Ozaki CK, Mitchell JR, de Vries MR. Short-term Pre-operative Methionine Restriction Induces Browning of Perivascular Adipose Tissue and Improves Vein Graft Remodeling in Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.02.565269. [PMID: 37961405 PMCID: PMC10635070 DOI: 10.1101/2023.11.02.565269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Short-term preoperative methionine restriction (MetR) shows promise as a translatable strategy to modulate the body's response to surgical injury. Its application, however, to improve post-interventional vascular remodeling remains underexplored. Here, we find that MetR protects from arterial intimal hyperplasia in a focal stenosis model and adverse vascular remodeling after vein graft surgery. RNA sequencing reveals that MetR enhances the brown adipose tissue phenotype in arterial perivascular adipose tissue (PVAT) and induces it in venous PVAT. Specifically, PPAR-α was highly upregulated in PVAT-adipocytes. Furthermore, MetR dampens the post-operative pro-inflammatory response to surgery in PVAT-macrophages in vivo and in vitro . This study shows for the first time that the detrimental effects of dysfunctional PVAT on vascular remodeling can be reversed by MetR, and identifies pathways involved in browning of PVAT. Furthermore, we demonstrate the potential of short-term pre-operative MetR as a simple intervention to ameliorate vascular remodeling after vascular surgery.
Collapse
|
3
|
Shirasu T, Yodsanit N, Li J, Huang Y, Xie X, Tang R, Wang Q, Zhang M, Urabe G, Webb A, Wang Y, Wang X, Xie R, Wang B, Kent KC, Gong S, Guo LW. Neointima abating and endothelium preserving - An adventitia-localized nanoformulation to inhibit the epigenetic writer DOT1L. Biomaterials 2023; 301:122245. [PMID: 37467597 PMCID: PMC10530408 DOI: 10.1016/j.biomaterials.2023.122245] [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: 12/20/2022] [Revised: 06/05/2023] [Accepted: 07/12/2023] [Indexed: 07/21/2023]
Abstract
Open vascular reconstructions such as bypass are common treatments for cardiovascular disease. Unfortunately, neointimal hyperplasia (IH) follows, leading to treatment failure for which there is no approved therapy. Here we combined the strengths of tailoring nanoplatforms for open vascular reconstructions and targeting new epigenetic mechanisms. We produced adhesive nanoparticles (ahNP) that could be pen-brushed and immobilized on the adventitia to sustainably release pinometostat, an inhibitor drug selective to the epigenetic writer DOT1L that catalyzes histone-3 lysine-79 dimethylation (H3K79me2). This treatment not only reduced IH by 76.8% in injured arteries mimicking open reconstructions in obese Zucker rats with human-like diseases but also avoided the shortcoming of endothelial impairment in IH management. In mechanistic studies, chromatin immunoprecipitation (ChIP) sequencing revealed co-enrichment of the histone mark H3K27ac(acetyl) and its reader BRD4 at the gene of aurora kinase B (AURKB), where H3K79me2 was also enriched as indicated by ChIP-qPCR. Accordingly, DOT1L co-immunoprecipitated with H3K27ac. Furthermore, the known IH driver BRD4 governed the expression of DOT1L which controlled AURKB's protein level, revealing a BRD4- > DOT1L- > AURKB axis. Consistently, AURKB-selective inhibition reduced IH. Thus, this study presents a prototype nanoformulation suited for open vascular reconstructions, and the new insights into chromatin modulators may aid future translational advances.
Collapse
Affiliation(s)
- Takuro Shirasu
- Division of Surgical Sciences, Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Nisakorn Yodsanit
- Department of Biomedical Engineering and Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA; Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Jing Li
- Division of Surgical Sciences, Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Yitao Huang
- Division of Surgical Sciences, Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA; The Biomedical Sciences Graduate Program (BIMS), School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Xiujie Xie
- Division of Surgical Sciences, Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Runze Tang
- Division of Surgical Sciences, Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Qingwei Wang
- Division of Surgical Sciences, Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Mengxue Zhang
- Division of Surgical Sciences, Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Go Urabe
- Division of Surgical Sciences, Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Amy Webb
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Yuyuan Wang
- Department of Biomedical Engineering and Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA; Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Xiuxiu Wang
- Department of Biomedical Engineering and Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA; Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Ruosen Xie
- Department of Biomedical Engineering and Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA; Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Bowen Wang
- Division of Surgical Sciences, Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - K Craig Kent
- Division of Surgical Sciences, Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA.
| | - Shaoqin Gong
- Department of Biomedical Engineering and Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA; Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53715, USA.
| | - Lian-Wang Guo
- Division of Surgical Sciences, Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA; Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, 22908, USA; Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, 22908, USA.
| |
Collapse
|
4
|
Zhao S, Deslarzes-Dubuis C, Urfer S, Lambelet M, Déglise S, Allagnat F. Cystathionine Gamma Lyase Is Regulated by Flow and Controls Smooth Muscle Migration in Human Saphenous Vein. Antioxidants (Basel) 2023; 12:1731. [PMID: 37760034 PMCID: PMC10525225 DOI: 10.3390/antiox12091731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/28/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
The saphenous vein is the conduit of choice for bypass grafting. Unfortunately, the hemodynamic stress associated with the arterial environment of the bypass vein graft leads to the development of intimal hyperplasia (IH), an excessive cellular growth and collagen deposition that results in restenosis and secondary graft occlusion. Hydrogen sulfide (H2S) is a ubiquitous redox-modifying gasotransmitter that inhibits IH. H2S is produced via the reverse trans-sulfuration pathway by three enzymes: cystathionine γ-lyase (CSE), cystathionine β-synthase (CBS) and 3-mercaptopyruvate sulfurtransferase (3-MST). However, the expression and regulation of these enzymes in the human vasculature remains unclear. Here, we investigated the expression of CSE, CBS and 3-MST in segments of native human saphenous vein and large arteries. Furthermore, we evaluated the regulation of these enzymes in vein segments cultured under static, venous (7 mmHg pressure) or arterial (100 mmHg pressure) pressure. CSE was expressed in the media, neointima and intima of the vessels and was negatively regulated by arterial shear stress. Adenoviral-mediated CSE overexpression or RNA interference-mediated CSE knock-down revealed that CSE inhibited primary human VSMC migration but not proliferation. We propose that high shear stress in arteriovenous bypass grafts inhibits CSE expression in both the media and endothelium, which may contribute to increased VSMC migration in the context of IH.
Collapse
|
5
|
Bechelli C, Macabrey D, Deglise S, Allagnat F. Clinical Potential of Hydrogen Sulfide in Peripheral Arterial Disease. Int J Mol Sci 2023; 24:9955. [PMID: 37373103 DOI: 10.3390/ijms24129955] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
Peripheral artery disease (PAD) affects more than 230 million people worldwide. PAD patients suffer from reduced quality of life and are at increased risk of vascular complications and all-cause mortality. Despite its prevalence, impact on quality of life and poor long-term clinical outcomes, PAD remains underdiagnosed and undertreated compared to myocardial infarction and stroke. PAD is due to a combination of macrovascular atherosclerosis and calcification, combined with microvascular rarefaction, leading to chronic peripheral ischemia. Novel therapies are needed to address the increasing incidence of PAD and its difficult long-term pharmacological and surgical management. The cysteine-derived gasotransmitter hydrogen sulfide (H2S) has interesting vasorelaxant, cytoprotective, antioxidant and anti-inflammatory properties. In this review, we describe the current understanding of PAD pathophysiology and the remarkable benefits of H2S against atherosclerosis, inflammation, vascular calcification, and other vasculo-protective effects.
Collapse
Affiliation(s)
- Clémence Bechelli
- Department of Vascular Surgery, Lausanne University Hospital, 1005 Lausanne, Switzerland
| | - Diane Macabrey
- Department of Vascular Surgery, Lausanne University Hospital, 1005 Lausanne, Switzerland
| | - Sebastien Deglise
- Department of Vascular Surgery, Lausanne University Hospital, 1005 Lausanne, Switzerland
| | - Florent Allagnat
- Department of Vascular Surgery, Lausanne University Hospital, 1005 Lausanne, Switzerland
| |
Collapse
|
6
|
Kiesworo K, MacArthur MR, Kip P, Agius T, Macabrey D, Lambelet M, Hamard L, Ozaki CK, Mitchell JR, Déglise S, Mitchell SJ, Allagnat F, Longchamp A. Cystathionine-γ-lyase overexpression modulates oxidized nicotinamide adenine dinucleotide biosynthesis and enhances neovascularization. JVS Vasc Sci 2023; 4:100095. [PMID: 36852171 PMCID: PMC9958478 DOI: 10.1016/j.jvssci.2022.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 11/10/2022] [Indexed: 01/15/2023] Open
Abstract
Objective Hydrogen sulfide is a proangiogenic gas produced primarily by the transsulfuration enzyme cystathionine-γ-lyase (CGL). CGL-dependent hydrogen sulfide production is required for neovascularization in models of peripheral arterial disease. However, the benefits of increasing endogenous CGL and its mechanism of action have not yet been elucidated. Methods Male whole body CGL-overexpressing transgenic (CGLTg) mice and wild-type (WT) littermates (C57BL/6J) were subjected to the hindlimb ischemia model (age, 10-12 weeks). Functional recovery was assessed via the treadmill exercise endurance test. Leg perfusion was measured by laser Doppler imaging and vascular endothelial-cadherin immunostaining. To examine the angiogenic potential, aortic ring sprouting assay and postnatal mouse retinal vasculature development studies were performed. Finally, comparative metabolomics analysis, oxidized/reduced nicotinamide adenine dinucleotide (NAD+/NADH) analysis, and quantitative real-time polymerase chain reaction were performed on CGLWT and CGLTg gastrocnemius muscle. Results The restoration of blood flow occurred more rapidly in CGLTg mice. Compared with the CGLWT mice, the median ± standard deviation running distance and time were increased for the CGLTg mice after femoral artery ligation (159 ± 53 m vs 291 ± 74 m [P < .005] and 17 ± 4 minutes vs 27 ± 5 minutes [P < .05], respectively). Consistently, in the CGLTg ischemic gastrocnemius muscle, the capillary density was increased fourfold (0.05 ± 0.02 vs 0.20 ± 0.12; P < .005). Ex vivo, the endothelial cell (EC) sprouting length was increased in aorta isolated from CGLTg mice, especially when cultured in VEGFA (vascular endothelial growth factor A)-only media (63 ± 2 pixels vs 146 ± 52 pixels; P < .05). Metabolomics analysis demonstrated a higher level of niacinamide, a precursor of NAD+/NADH in the muscle of CGLTg mice (61.4 × 106 ± 5.9 × 106 vs 72.4 ± 7.7 × 106 area under the curve; P < .05). Similarly, the NAD+ salvage pathway gene expression was increased in CGLTg gastrocnemius muscle. Finally, CGL overexpression or supplementation with the NAD+ precursor nicotinamide mononucleotide improved EC migration in vitro (wound closure: control, 35% ± 9%; CGL, 55% ± 11%; nicotinamide mononucleotide, 42% ± 13%; P < .05). Conclusions Our results have demonstrated that CGL overexpression improves the neovascularization of skeletal muscle on hindlimb ischemia. These effects correlated with changes in the NAD pathway, which improved EC migration.
Collapse
Affiliation(s)
- Kevin Kiesworo
- Department of Vascular Surgery, Lausanne University Hospital, Lausanne, Switzerland
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | | | - Peter Kip
- Department of Surgery and Heart and Vascular Center, Brigham & Women's Hospital and Harvard Medical School, Boston, MA
| | - Thomas Agius
- Department of Vascular Surgery, Lausanne University Hospital, Lausanne, Switzerland
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Diane Macabrey
- 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
| | - Lauriane Hamard
- Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - C.-Keith Ozaki
- Department of Surgery and Heart and Vascular Center, Brigham & Women's Hospital and Harvard Medical School, Boston, MA
| | - James R. Mitchell
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Sébastien Déglise
- Department of Vascular Surgery, Lausanne University Hospital, Lausanne, Switzerland
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Sarah J. Mitchell
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Florent Allagnat
- 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
| |
Collapse
|
7
|
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: 24] [Impact Index Per Article: 24.0] [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
|
8
|
Anderson EM, Rozowsky JM, Fazzone BJ, Schmidt EA, Stevens BR, O’Malley KA, Scali ST, Berceli SA. Temporal Dynamics of the Intestinal Microbiome Following Short-Term Dietary Restriction. Nutrients 2022; 14:2785. [PMID: 35889742 PMCID: PMC9318361 DOI: 10.3390/nu14142785] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 06/28/2022] [Accepted: 07/02/2022] [Indexed: 12/04/2022] Open
Abstract
Short-term dietary restriction has been proposed as an intriguing pre-operative conditioning strategy designed to attenuate the surgical stress response and improve outcomes. However, it is unclear how this nutritional intervention influences the microbiome, which is known to modulate the systemic condition. Healthy individuals were recruited to participate in a four-day, 70% protein-restricted, 30% calorie-restricted diet, and stool samples were collected at baseline, after the restricted diet, and after resuming normal food intake. Taxonomy and functional pathway analysis was performed via shotgun metagenomic sequencing, prevalence filtering, and differential abundance analysis. High prevalence species were altered by the dietary intervention but quickly returned to baseline after restarting a regular diet. Composition and functional changes after the restricted diet included the decreased relative abundance of commensal bacteria and a catabolic phenotype. Notable species changes included Faecalibacterium prausnitzii and Roseburia intestinalis, which are major butyrate producers within the colon and are characteristically decreased in many disease states. The macronutrient components of the diet might have influenced these changes. We conclude that short-term dietary restriction modulates the ecology of the gut microbiome, with this modulation being characterized by a relative dysbiosis.
Collapse
Affiliation(s)
- Erik M. Anderson
- Department of Surgery, University of Florida College of Medicine, 1600 SW Archer Rd., Gainesville, FL 32610, USA; (E.M.A.); (J.M.R.); (B.J.F.); (E.A.S.); (K.A.O.); (S.T.S.)
- Department of Surgery, Malcolm Randall Veteran Affairs Medical Center, 1601 SW Archer Rd., Gainesville, FL 32610, USA
| | - Jared M. Rozowsky
- Department of Surgery, University of Florida College of Medicine, 1600 SW Archer Rd., Gainesville, FL 32610, USA; (E.M.A.); (J.M.R.); (B.J.F.); (E.A.S.); (K.A.O.); (S.T.S.)
- Department of Surgery, Malcolm Randall Veteran Affairs Medical Center, 1601 SW Archer Rd., Gainesville, FL 32610, USA
| | - Brian J. Fazzone
- Department of Surgery, University of Florida College of Medicine, 1600 SW Archer Rd., Gainesville, FL 32610, USA; (E.M.A.); (J.M.R.); (B.J.F.); (E.A.S.); (K.A.O.); (S.T.S.)
- Department of Surgery, Malcolm Randall Veteran Affairs Medical Center, 1601 SW Archer Rd., Gainesville, FL 32610, USA
| | - Emilie A. Schmidt
- Department of Surgery, University of Florida College of Medicine, 1600 SW Archer Rd., Gainesville, FL 32610, USA; (E.M.A.); (J.M.R.); (B.J.F.); (E.A.S.); (K.A.O.); (S.T.S.)
- Department of Surgery, Malcolm Randall Veteran Affairs Medical Center, 1601 SW Archer Rd., Gainesville, FL 32610, USA
| | - Bruce R. Stevens
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, 1600 SW Archer Rd., Gainesville, FL 32610, USA;
| | - Kerri A. O’Malley
- Department of Surgery, University of Florida College of Medicine, 1600 SW Archer Rd., Gainesville, FL 32610, USA; (E.M.A.); (J.M.R.); (B.J.F.); (E.A.S.); (K.A.O.); (S.T.S.)
- Department of Surgery, Malcolm Randall Veteran Affairs Medical Center, 1601 SW Archer Rd., Gainesville, FL 32610, USA
| | - Salvatore T. Scali
- Department of Surgery, University of Florida College of Medicine, 1600 SW Archer Rd., Gainesville, FL 32610, USA; (E.M.A.); (J.M.R.); (B.J.F.); (E.A.S.); (K.A.O.); (S.T.S.)
- Department of Surgery, Malcolm Randall Veteran Affairs Medical Center, 1601 SW Archer Rd., Gainesville, FL 32610, USA
| | - Scott A. Berceli
- Department of Surgery, University of Florida College of Medicine, 1600 SW Archer Rd., Gainesville, FL 32610, USA; (E.M.A.); (J.M.R.); (B.J.F.); (E.A.S.); (K.A.O.); (S.T.S.)
- Department of Surgery, Malcolm Randall Veteran Affairs Medical Center, 1601 SW Archer Rd., Gainesville, FL 32610, USA
| |
Collapse
|
9
|
Macabrey D, Longchamp A, Déglise S, Allagnat F. Clinical Use of Hydrogen Sulfide to Protect Against Intimal Hyperplasia. Front Cardiovasc Med 2022; 9:876639. [PMID: 35479275 PMCID: PMC9035533 DOI: 10.3389/fcvm.2022.876639] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 03/18/2022] [Indexed: 12/27/2022] Open
Abstract
Arterial occlusive disease is the narrowing of the arteries via atherosclerotic plaque buildup. The major risk factors for arterial occlusive disease are age, high levels of cholesterol and triglycerides, diabetes, high blood pressure, and smoking. Arterial occlusive disease is the leading cause of death in Western countries. Patients who suffer from arterial occlusive disease develop peripheral arterial disease (PAD) when the narrowing affects limbs, stroke when the narrowing affects carotid arteries, and heart disease when the narrowing affects coronary arteries. When lifestyle interventions (exercise, diet…) fail, the only solution remains surgical endovascular and open revascularization. Unfortunately, these surgeries still suffer from high failure rates due to re-occlusive vascular wall adaptations, which is largely due to intimal hyperplasia (IH). IH 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. Re-occlusive IH lesions result in costly and complex recurrent end-organ ischemia, and often lead to loss of limb, brain function, or life. Despite decades of IH research, limited therapies are currently available. Hydrogen sulfide (H2S) is an endogenous gasotransmitter derived from cysteine metabolism. Although environmental exposure to exogenous high H2S is toxic, endogenous H2S has important vasorelaxant, cytoprotective and anti-inflammatory properties. Its vasculo-protective properties have attracted a remarkable amount of attention, especially its ability to inhibit IH. This review summarizes IH pathophysiology and treatment, and provides an overview of the potential clinical role of H2S to prevent IH and restenosis.
Collapse
Affiliation(s)
- Diane Macabrey
- 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
| | - Sébastien Déglise
- 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
| |
Collapse
|
10
|
Hine C, Treviño-Villarreal JH, Mejia P, Longchamp A, Brace LE, Harputlugil E, Mitchell SJ, Yang J, Guan Y, Maciejewski JP, Jha BK, Mitchell JR. Sulfur Amino Acid Supplementation Abrogates Protective Effects of Caloric Restriction for Enhancing Bone Marrow Regrowth Following Ionizing Radiation. Nutrients 2022; 14:nu14071529. [PMID: 35406143 PMCID: PMC9002760 DOI: 10.3390/nu14071529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/30/2022] [Accepted: 04/02/2022] [Indexed: 02/01/2023] Open
Abstract
Radiation therapy damages and depletes total bone marrow (BM) cellularity, compromising safety and limiting effective dosing. Aging also strains total BM and BM hematopoietic stem and progenitor cell (HSPC) renewal and function, resulting in multi-system defects. Interventions that preserve BM and BM HSPC homeostasis thus have potential clinical significance. Here, we report that 50% calorie restriction (CR) for 7-days or fasting for 3-days prior to irradiation improved mouse BM regrowth in the days and weeks post irradiation. Specifically, one week of 50% CR ameliorated loss of total BM cellularity post irradiation compared to ad libitum-fed controls. CR-mediated BM protection was abrogated by dietary sulfur amino acid (i.e., cysteine, methionine) supplementation or pharmacological inhibition of sulfur amino acid metabolizing and hydrogen sulfide (H2S) producing enzymes. Up to 2-fold increased proliferative capacity of ex vivo-irradiated BM isolated from food restricted mice relative to control mice indicates cell autonomy of the protective effect. Pretreatment with H2S in vitro was sufficient to preserve proliferative capacity by over 50% compared to non-treated cells in ex vivo-irradiated BM and BM HSPCs. The exogenous addition of H2S inhibited Ten eleven translocation 2 (TET2) activity in vitro, thus providing a potential mechanism of action. Short-term CR or fasting therefore offers BM radioprotection and promotes regrowth in part via altered sulfur amino acid metabolism and H2S generation, with translational implications for radiation treatment and aging.
Collapse
Affiliation(s)
- Christopher Hine
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA;
- Department of Molecular Metabolism (Formally Genetics and Complex Diseases), Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (J.H.T.-V.) (P.M.); (A.L.); (L.E.B.); (E.H.); (S.J.M.); (J.R.M.)
- Correspondence:
| | - J. Humberto Treviño-Villarreal
- Department of Molecular Metabolism (Formally Genetics and Complex Diseases), Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (J.H.T.-V.) (P.M.); (A.L.); (L.E.B.); (E.H.); (S.J.M.); (J.R.M.)
- Service of Endocrinology, Department of Internal Medicine, University Hospital and School of Medicine, Universidad Autonoma de Nuevo Leon, Monterrey N.L. 64460, Mexico
| | - Pedro Mejia
- Department of Molecular Metabolism (Formally Genetics and Complex Diseases), Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (J.H.T.-V.) (P.M.); (A.L.); (L.E.B.); (E.H.); (S.J.M.); (J.R.M.)
| | - Alban Longchamp
- Department of Molecular Metabolism (Formally Genetics and Complex Diseases), Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (J.H.T.-V.) (P.M.); (A.L.); (L.E.B.); (E.H.); (S.J.M.); (J.R.M.)
- Department of Vascular Surgery, Centre Hospitalier Universitaire Vaudois, University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Lear E. Brace
- Department of Molecular Metabolism (Formally Genetics and Complex Diseases), Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (J.H.T.-V.) (P.M.); (A.L.); (L.E.B.); (E.H.); (S.J.M.); (J.R.M.)
| | - Eylul Harputlugil
- Department of Molecular Metabolism (Formally Genetics and Complex Diseases), Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (J.H.T.-V.) (P.M.); (A.L.); (L.E.B.); (E.H.); (S.J.M.); (J.R.M.)
| | - Sarah J. Mitchell
- Department of Molecular Metabolism (Formally Genetics and Complex Diseases), Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (J.H.T.-V.) (P.M.); (A.L.); (L.E.B.); (E.H.); (S.J.M.); (J.R.M.)
- Department of Health Sciences and Technology, ETH Zurich, 8005 Zurich, Switzerland
| | - Jie Yang
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA;
| | - Yihong Guan
- Department of Translational Hematology & Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (Y.G.); (J.P.M.); (B.K.J.)
| | - Jaroslaw P. Maciejewski
- Department of Translational Hematology & Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (Y.G.); (J.P.M.); (B.K.J.)
| | - Babal K. Jha
- Department of Translational Hematology & Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (Y.G.); (J.P.M.); (B.K.J.)
| | - James R. Mitchell
- Department of Molecular Metabolism (Formally Genetics and Complex Diseases), Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (J.H.T.-V.) (P.M.); (A.L.); (L.E.B.); (E.H.); (S.J.M.); (J.R.M.)
- Department of Health Sciences and Technology, ETH Zurich, 8005 Zurich, Switzerland
| |
Collapse
|
11
|
Kalafut KC, Mitchell SJ, MacArthur MR, Mitchell JR. Short-Term Ketogenic Diet Induces a Molecular Response That Is Distinct From Dietary Protein Restriction. Front Nutr 2022; 9:839341. [PMID: 35433789 PMCID: PMC9005751 DOI: 10.3389/fnut.2022.839341] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 02/17/2022] [Indexed: 12/17/2022] Open
Abstract
There is increasing interest in utilizing short-term dietary interventions in the contexts of cancer, surgical stress and metabolic disease. These short-term diets may be more feasible than extended interventions and may be designed to complement existing therapies. In particular, the high-fat, low-carbohydrate ketogenic diet (KD), traditionally used to treat epilepsy, has gained popularity as a potential strategy for weight loss and improved metabolic health. In mice, long-term KD improves insulin sensitivity and may extend lifespan and healthspan. Dietary protein restriction (PR) causes increased energy expenditure, weight loss and improved glucose homeostasis. Since KD is inherently a low-protein diet (10% of calories from protein vs. >18% in control diet), here we evaluated the potential for mechanistic overlap between PR and KD via activation of a PR response. Mice were fed control, protein-free (PF), or one of four ketogenic diets with varying protein content for 8 days. PF and KD both decreased body weight, fat mass, and liver weights, and reduced fasting glucose and insulin levels, compared to mice fed the control diet. However, PF-fed animals had significantly improved insulin tolerance compared to KD. Furthermore, contrary to the PF-fed mice, mice fed ketogenic diets containing more than 5% of energy from protein did not increase hepatic Fgf21 or brown adipose Ucp1 expression. Interestingly, mice fed KD lacking protein demonstrated greater elevations in hepatic Fgf21 than mice fed a low-fat PF diet. To further elucidate potential mechanistic differences between PF and KD and the interplay between dietary protein and carbohydrate restriction, we conducted RNA-seq analysis on livers from mice fed each of the six diets and identified distinct gene sets which respond to dietary protein content, dietary fat content, and ketogenesis. We conclude that KD with 10% of energy from protein does not induce a protein restriction response, and that the overlapping metabolic benefits of KD and PF diets may occur via distinct underlying mechanisms.
Collapse
Affiliation(s)
- Krystle C. Kalafut
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, United States
| | - Sarah J. Mitchell
- Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zürich, Zurich, Switzerland
| | - Michael R. MacArthur
- Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zürich, Zurich, Switzerland
| | - James R. Mitchell
- Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zürich, Zurich, Switzerland
| |
Collapse
|
12
|
Jiang X, MacArthur MR, Treviño-Villarreal JH, Kip P, Ozaki CK, Mitchell SJ, Mitchell JR. Intracellular H 2S production is an autophagy-dependent adaptive response to DNA damage. Cell Chem Biol 2021; 28:1669-1678.e5. [PMID: 34166610 PMCID: PMC8665944 DOI: 10.1016/j.chembiol.2021.05.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 03/08/2021] [Accepted: 05/26/2021] [Indexed: 12/21/2022]
Abstract
Hydrogen sulfide (H2S) is a gasotransmitter with broad physiological activities, including protecting cells against stress, but little is known about the regulation of cellular H2S homeostasis. We have performed a high-content small-molecule screen and identified genotoxic agents, including cancer chemotherapy drugs, as activators of intracellular H2S levels. DNA damage-induced H2S in vitro and in vivo. Mechanistically, DNA damage elevated autophagy and upregulated H2S-generating enzyme CGL; chemical or genetic disruption of autophagy or CGL impaired H2S induction. Importantly, exogenous H2S partially rescued autophagy-deficient cells from genotoxic stress. Furthermore, stressors that are not primarily genotoxic (growth factor depletion and mitochondrial uncoupler FCCP) increased intracellular H2S in an autophagy-dependent manner. Our findings highlight the role of autophagy in H2S production and suggest that H2S generation may be a common adaptive response to DNA damage and other stressors.
Collapse
Affiliation(s)
- Xiaofeng Jiang
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA.
| | - Michael R MacArthur
- Department of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland
| | | | - Peter Kip
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Department of Surgery and the Heart and Vascular Center, Brigham & Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Einthoven Laboratory for Experimental Vascular Medicine and Department of Surgery, Leiden University Medical Center, 2333 CC Leiden, the Netherlands
| | - C Keith Ozaki
- Department of Surgery and the Heart and Vascular Center, Brigham & Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Sarah J Mitchell
- Department of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland.
| | - James R Mitchell
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Department of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland
| |
Collapse
|
13
|
Kip P, Sluiter TJ, Moore JK, Hart A, Ruske J, O’Leary J, Jung J, Tao M, MacArthur MR, Heindel P, de Jong A, de Vries MR, Burak MF, Mitchell SJ, Mitchell JR, Ozaki CK. Short-Term Pre-Operative Protein Caloric Restriction in Elective Vascular Surgery Patients: A Randomized Clinical Trial. Nutrients 2021; 13:nu13114024. [PMID: 34836280 PMCID: PMC8621550 DOI: 10.3390/nu13114024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/03/2021] [Accepted: 11/04/2021] [Indexed: 01/12/2023] Open
Abstract
(1) Background: Vascular surgery operations are hampered by high failure rates and frequent occurrence of peri-operative cardiovascular complications. In pre-clinical studies, pre-operative restriction of proteins and/or calories (PCR) has been shown to limit ischemia-reperfusion damage, slow intimal hyperplasia, and improve metabolic fitness. However, whether these dietary regimens are feasible and safe in the vascular surgery patient population remains unknown. (2) Methods: We performed a randomized controlled trial in patients scheduled for any elective open vascular procedure. Participants were randomized in a 3:2 ratio to either four days of outpatient pre-operative PCR (30% calorie, 70% protein restriction) or their regular ad-libitum diet. Blood was drawn at baseline, pre-operative, and post-operative day 1 timepoints. A leukocyte subset flow cytometry panel was performed at these timepoints. Subcutaneous/perivascular adipose tissue was sampled and analyzed. Follow-up was one year post-op. (3) Results: 19 patients were enrolled, of whom 11 completed the study. No diet-related reasons for non-completion were reported, and there was no intervention group crossover. The PCR diet induced weight loss and BMI decrease without malnutrition. Insulin sensitivity was improved after four days of PCR (p = 0.05). Between diet groups, there were similar rates of re-intervention, wound infection, and cardiovascular complications. Leukocyte populations were maintained after four days of PCR. (4) Conclusions: Pre-operative PCR is safe and feasible in elective vascular surgery patients.
Collapse
Affiliation(s)
- Peter Kip
- Department of Surgery and the Heart and Vascular Center, Brigham & Women’s Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA; (P.K.); (T.J.S.); (A.H.); (J.R.); (J.O.); (M.T.); (P.H.); (M.F.B.)
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (J.J.); (M.R.M.); (S.J.M.); (J.R.M.)
- Einthoven Laboratory for Experimental Vascular Medicine, Department of Surgery, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands; (A.d.J.); (M.R.d.V.)
| | - Thijs J. Sluiter
- Department of Surgery and the Heart and Vascular Center, Brigham & Women’s Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA; (P.K.); (T.J.S.); (A.H.); (J.R.); (J.O.); (M.T.); (P.H.); (M.F.B.)
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (J.J.); (M.R.M.); (S.J.M.); (J.R.M.)
- Einthoven Laboratory for Experimental Vascular Medicine, Department of Surgery, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands; (A.d.J.); (M.R.d.V.)
| | - Jodene K. Moore
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA;
| | - Abby Hart
- Department of Surgery and the Heart and Vascular Center, Brigham & Women’s Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA; (P.K.); (T.J.S.); (A.H.); (J.R.); (J.O.); (M.T.); (P.H.); (M.F.B.)
| | - Jack Ruske
- Department of Surgery and the Heart and Vascular Center, Brigham & Women’s Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA; (P.K.); (T.J.S.); (A.H.); (J.R.); (J.O.); (M.T.); (P.H.); (M.F.B.)
| | - James O’Leary
- Department of Surgery and the Heart and Vascular Center, Brigham & Women’s Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA; (P.K.); (T.J.S.); (A.H.); (J.R.); (J.O.); (M.T.); (P.H.); (M.F.B.)
| | - Jonathan Jung
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (J.J.); (M.R.M.); (S.J.M.); (J.R.M.)
- School of Medicine, University of Glasgow, Glasgow G12 8QF, UK
| | - Ming Tao
- Department of Surgery and the Heart and Vascular Center, Brigham & Women’s Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA; (P.K.); (T.J.S.); (A.H.); (J.R.); (J.O.); (M.T.); (P.H.); (M.F.B.)
| | - Michael R. MacArthur
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (J.J.); (M.R.M.); (S.J.M.); (J.R.M.)
- Department of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland
| | - Patrick Heindel
- Department of Surgery and the Heart and Vascular Center, Brigham & Women’s Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA; (P.K.); (T.J.S.); (A.H.); (J.R.); (J.O.); (M.T.); (P.H.); (M.F.B.)
| | - Alwin de Jong
- Einthoven Laboratory for Experimental Vascular Medicine, Department of Surgery, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands; (A.d.J.); (M.R.d.V.)
| | - Margreet R. de Vries
- Einthoven Laboratory for Experimental Vascular Medicine, Department of Surgery, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands; (A.d.J.); (M.R.d.V.)
| | - M. Furkan Burak
- Department of Surgery and the Heart and Vascular Center, Brigham & Women’s Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA; (P.K.); (T.J.S.); (A.H.); (J.R.); (J.O.); (M.T.); (P.H.); (M.F.B.)
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (J.J.); (M.R.M.); (S.J.M.); (J.R.M.)
| | - Sarah J. Mitchell
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (J.J.); (M.R.M.); (S.J.M.); (J.R.M.)
- Department of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland
| | - James R. Mitchell
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (J.J.); (M.R.M.); (S.J.M.); (J.R.M.)
- Department of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland
| | - C. Keith Ozaki
- Department of Surgery and the Heart and Vascular Center, Brigham & Women’s Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA; (P.K.); (T.J.S.); (A.H.); (J.R.); (J.O.); (M.T.); (P.H.); (M.F.B.)
- Correspondence:
| |
Collapse
|
14
|
Synthesis of Reactive Sulfur Species in Cultured Vascular Endothelial Cells after Exposure to TGF-β 1: Induction of Cystathionine γ-Lyase and Cystathionine β-Synthase Expression Mediated by the ALK5-Smad2/3/4 and ALK5-Smad2/3-ATF4 Pathways. Int J Mol Sci 2021; 22:ijms222111762. [PMID: 34769192 PMCID: PMC8583730 DOI: 10.3390/ijms222111762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 11/17/2022] Open
Abstract
Transforming growth factor-β1 (TGF-β1) occurs at high levels at damage sites of vascular endothelial cell layers and regulates the functions of vascular endothelial cells. Reactive sulfur species (RSS), such as cysteine persulfide, glutathione persulfide, and hydrogen persulfide, are cytoprotective factors against electrophiles such as reactive oxygen species and heavy metals. Previously, we reported that sodium trisulfide, a sulfane sulfur donor, promotes vascular endothelial cell proliferation. The objective of the present study was to clarify the regulation and significance of RSS synthesis in vascular endothelial cells after exposure to TGF-β1. Bovine aortic endothelial cells in a culture system were treated with TGF-β1 to assess the expression of intracellular RSS, the effect of RSS on cell proliferation in the presence of TGF-β1, induction of RSS-producing enzymes by TGF-β1, and intracellular signal pathways that mediate this induction. The results suggest that TGF-β1 increased intracellular RSS levels to modulate its inhibitory effect on proliferation. The increased production of RSS, probably high-molecular-mass RSS, was due to the induction of cystathionine γ-lyase and cystathionine β-synthase, which are RSS-producing enzymes, and the induction was mediated by the ALK5-Smad2/3/4 and ALK5-Smad2/3-ATF4 pathways in vascular endothelial cells. TGF-β1 regulates vascular endothelial cell functions such as proliferation and fibrinolytic activity; intracellular high-molecular-mass RSS, which are increased by TGF-β1, may modulate the regulation activity in vascular endothelial cells.
Collapse
|
15
|
Hydrogen sulfide in ageing, longevity and disease. Biochem J 2021; 478:3485-3504. [PMID: 34613340 PMCID: PMC8589328 DOI: 10.1042/bcj20210517] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/16/2021] [Accepted: 08/18/2021] [Indexed: 12/21/2022]
Abstract
Hydrogen sulfide (H2S) modulates many biological processes, including ageing. Initially considered a hazardous toxic gas, it is now recognised that H2S is produced endogenously across taxa and is a key mediator of processes that promote longevity and improve late-life health. In this review, we consider the key developments in our understanding of this gaseous signalling molecule in the context of health and disease, discuss potential mechanisms through which H2S can influence processes central to ageing and highlight the emergence of novel H2S-based therapeutics. We also consider the major challenges that may potentially hinder the development of such therapies.
Collapse
|
16
|
The Gasotransmitter Hydrogen Sulfide (H 2S) Prevents Pathologic Calcification (PC) in Cartilage. Antioxidants (Basel) 2021; 10:antiox10091433. [PMID: 34573065 PMCID: PMC8471338 DOI: 10.3390/antiox10091433] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 08/31/2021] [Accepted: 09/02/2021] [Indexed: 01/09/2023] Open
Abstract
Pathologic calcification (PC) is a painful and disabling condition whereby calcium-containing crystals deposit in tissues that do not physiologically calcify: cartilage, tendons, muscle, vessels and skin. In cartilage, compression and inflammation triggered by PC leads to cartilage degradation typical of osteoarthritis (OA). The PC process is poorly understood and treatments able to target the underlying mechanisms of the disease are lacking. Here we show a crucial role of the gasotransmitter hydrogen sulfide (H2S) and, in particular, of the H2S-producing enzyme cystathionine γ-lyase (CSE), in regulating PC in cartilage. Cse deficiency (Cse KO mice) exacerbated calcification in both surgically-induced (menisectomy) and spontaneous (aging) murine models of cartilage PC, and augmented PC was closely associated with cartilage degradation (OA). On the contrary, Cse overexpression (Cse tg mice) protected from these features. In vitro, Cse KO chondrocytes showed increased calcification, potentially via enhanced alkaline phosphatase (Alpl) expression and activity and increased IL-6 production. The opposite results were obtained in Cse tg chondrocytes. In cartilage samples from patients with OA, CSE expression inversely correlated with the degree of tissue calcification and disease severity. Increased cartilage degradation in murine and human tissues lacking or expressing low CSE levels may be accounted for by dysregulated catabolism. We found higher levels of matrix-degrading metalloproteases Mmp-3 and -13 in Cse KO chondrocytes, whereas the opposite results were obtained in Cse tg cells. Finally, by high-throughput screening, we identified a novel small molecule CSE positive allosteric modulator (PAM), and demonstrated that it was able to increase cellular H2S production, and decrease murine and human chondrocyte calcification and IL-6 secretion. Together, these data implicate impaired CSE-dependent H2S production by chondrocytes in the etiology of cartilage PC and worsening of secondary outcomes (OA). In this context, enhancing CSE expression and/or activity in chondrocytes could represent a potential strategy to inhibit PC.
Collapse
|
17
|
Kip P, Tao M, Trocha KM, MacArthur MR, Peters HAB, Mitchell SJ, Mann CG, Sluiter TJ, Jung J, Patterson S, Quax PHA, de Vries MR, Mitchell JR, Keith Ozaki C. Periprocedural Hydrogen Sulfide Therapy Improves Vascular Remodeling and Attenuates Vein Graft Disease. J Am Heart Assoc 2020; 9:e016391. [PMID: 33146045 PMCID: PMC7763704 DOI: 10.1161/jaha.120.016391] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 08/31/2020] [Indexed: 11/25/2022]
Abstract
Background Failure rates after revascularization surgery remain high, both in vein grafts (VG) and arterial interventions. One promising approach to improve outcomes is endogenous upregulation of the gaseous transmitter-molecule hydrogen sulfide, via short-term dietary restriction. However, strict patient compliance stands as a potential translational barrier in the vascular surgery patient population. Here we present a new therapeutic approach, via a locally applicable gel containing the hydrogen sulfide releasing prodrug (GYY), to both mitigate graft failure and improve arterial remodeling. Methods and Results All experiments were performed on C57BL/6 (male, 12 weeks old) mice. VG surgery was performed by grafting a donor-mouse cava vein into the right common carotid artery of a recipient via an end-to-end anastomosis. In separate experiments arterial intimal hyperplasia was assayed via a right common carotid artery focal stenosis model. All mice were harvested at postoperative day 28 and artery/graft was processed for histology. Efficacy of hydrogen sulfide was first tested via GYY supplementation of drinking water either 1 week before VG surgery (pre-GYY) or starting immediately postoperatively (post-GYY). Pre-GYY mice had a 36.5% decrease in intimal/media+adventitia area ratio compared with controls. GYY in a 40% Pluronic gel (or vehicle) locally applied to the graft/artery had decreased intimal/media area ratios (right common carotid artery) and improved vessel diameters. GYY-geltreated VG had larger diameters at both postoperative days 14 and 28, and a 56.7% reduction in intimal/media+adventitia area ratios. Intimal vascular smooth muscle cell migration was decreased 30.6% after GYY gel treatment, which was reproduced in vitro. Conclusions Local gel-based treatment with the hydrogen sulfide-donor GYY stands as a translatable therapy to improve VG durability and arterial remodeling after injury.
Collapse
Affiliation(s)
- Peter Kip
- Department of Surgery and the Heart and Vascular CenterBrigham & Women’s HospitalHarvard Medical SchoolBostonMA
- Department of Molecular MetabolismHarvard T.H. Chan School of Public HealthBostonMA
- Einthoven Laboratory for Experimental Vascular Medicine and Department of SurgeryLeiden University Medical CenterLeidenThe Netherlands
| | - Ming Tao
- Department of Surgery and the Heart and Vascular CenterBrigham & Women’s HospitalHarvard Medical SchoolBostonMA
| | - Kaspar M. Trocha
- Department of Surgery and the Heart and Vascular CenterBrigham & Women’s HospitalHarvard Medical SchoolBostonMA
- Department of Molecular MetabolismHarvard T.H. Chan School of Public HealthBostonMA
| | - Michael R. MacArthur
- Department of Molecular MetabolismHarvard T.H. Chan School of Public HealthBostonMA
| | - Hendrika A. B. Peters
- Einthoven Laboratory for Experimental Vascular Medicine and Department of SurgeryLeiden University Medical CenterLeidenThe Netherlands
| | - Sarah J. Mitchell
- Department of Molecular MetabolismHarvard T.H. Chan School of Public HealthBostonMA
| | - Charlotte G. Mann
- Department of Molecular MetabolismHarvard T.H. Chan School of Public HealthBostonMA
| | - Thijs J. Sluiter
- Department of Surgery and the Heart and Vascular CenterBrigham & Women’s HospitalHarvard Medical SchoolBostonMA
- Department of Molecular MetabolismHarvard T.H. Chan School of Public HealthBostonMA
- Einthoven Laboratory for Experimental Vascular Medicine and Department of SurgeryLeiden University Medical CenterLeidenThe Netherlands
| | - Jonathan Jung
- Department of Molecular MetabolismHarvard T.H. Chan School of Public HealthBostonMA
- School of MedicineUniversity of GlasgowGlasgowUK
| | - Suzannah Patterson
- Department of Surgery and the Heart and Vascular CenterBrigham & Women’s HospitalHarvard Medical SchoolBostonMA
| | - Paul H. A. Quax
- Einthoven Laboratory for Experimental Vascular Medicine and Department of SurgeryLeiden University Medical CenterLeidenThe Netherlands
| | - Margreet R. de Vries
- Einthoven Laboratory for Experimental Vascular Medicine and Department of SurgeryLeiden University Medical CenterLeidenThe Netherlands
| | - James R. Mitchell
- Department of Molecular MetabolismHarvard T.H. Chan School of Public HealthBostonMA
| | - C. Keith Ozaki
- Department of Surgery and the Heart and Vascular CenterBrigham & Women’s HospitalHarvard Medical SchoolBostonMA
| |
Collapse
|
18
|
Transcriptional Induction of Cystathionine γ-Lyase, a Reactive Sulfur-Producing Enzyme, by Copper Diethyldithiocarbamate in Cultured Vascular Endothelial Cells. Int J Mol Sci 2020; 21:ijms21176053. [PMID: 32842680 PMCID: PMC7503448 DOI: 10.3390/ijms21176053] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/18/2020] [Accepted: 08/21/2020] [Indexed: 12/13/2022] Open
Abstract
As toxic substances can enter the circulating blood and cross endothelial monolayers to reach parenchymal cells in organs, vascular endothelial cells are an important target compartment for such substances. Reactive sulfur species protect cells against oxidative stress and toxic substances, including heavy metals. Reactive sulfur species are produced by enzymes, such as cystathionine γ-lyase (CSE), cystathionine β-synthase, 3-mercaptopyruvate sulfurtransferase, and cysteinyl-tRNA synthetase. However, little is known about the regulatory mechanisms underlying the expression of these enzymes in vascular endothelial cells. Bio-organometallics is a research field that analyzes biological systems using organic-inorganic hybrid molecules (organometallic compounds and metal coordinating compounds) as molecular probes. In the present study, we analyzed intracellular signaling pathways that mediate the expression of reactive sulfur species-producing enzymes in cultured bovine aortic endothelial cells, using copper diethyldithiocarbamate (Cu10). Cu10 selectively upregulated CSE gene expression in vascular endothelial cells independent of cell density. This transcriptional induction of endothelial CSE required both the diethyldithiocarbamate scaffold and the coordinated copper ion. Additionally, the present study revealed that ERK1/2, p38 MAPK, and hypoxia-inducible factor (HIF)-1α/HIF-1β pathways mediate transcriptional induction of endothelial CSE by Cu10. The transcription factors NF-κB, Sp1, and ATF4 were suggested to act in constitutive CSE expression, although the possibility that they are involved in the CSE induction by Cu10 cannot be excluded. The present study used a copper complex as a molecular probe to reveal that the transcription of CSE is regulated by multiple pathways in vascular endothelial cells, including ERK1/2, p38 MAPK, and HIF-1α/HIF-1β. Bio-organometallics appears to be an effective strategy for analyzing the functions of intracellular signaling pathways in vascular endothelial cells.
Collapse
|
19
|
Baganha F, de Jong A, Jukema JW, Quax PHA, de Vries MR. The Role of Immunomodulation in Vein Graft Remodeling and Failure. J Cardiovasc Transl Res 2020; 14:100-109. [PMID: 32542547 PMCID: PMC7892738 DOI: 10.1007/s12265-020-10001-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 04/01/2020] [Indexed: 12/18/2022]
Abstract
Obstructive arterial disease is a major cause of morbidity and mortality in the developed world. Venous bypass graft surgery is one of the most frequently used revascularization strategies despite its considerable short and long time failure rate. Due to vessel wall remodeling, inflammation, intimal hyperplasia, and accelerated atherosclerosis, vein grafts may (ultimately) fail to revascularize tissues downstream to occlusive atherosclerotic lesions. In the past decades, little has changed in the prevention of vein graft failure (VGF) although new insights in the role of innate and adaptive immunity in VGF have emerged. In this review, we discuss the pathophysiological mechanisms underlying the development of VGF, emphasizing the role of immune response and associated factors related to VG remodeling and failure. Moreover, we discuss potential therapeutic options that can improve patency based on data from both preclinical studies and the latest clinical trials. This review contributes to the insights in the role of immunomodulation in vein graft failure in humans. We describe the effects of immune cells and related factors in early (thrombosis), intermediate (inward remodeling and intimal hyperplasia), and late (intimal hyperplasia and accelerated atherosclerosis) failure based on both preclinical (mouse) models and clinical data.
Collapse
Affiliation(s)
- Fabiana Baganha
- Department of Vascular Surgery, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands.,Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, Aberdeen University, Aberdeen, UK
| | - Alwin de Jong
- Department of Vascular Surgery, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Paul H A Quax
- Department of Vascular Surgery, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Margreet R de Vries
- Department of Vascular Surgery, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands. .,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands.
| |
Collapse
|
20
|
Kip P, Trocha KM, Tao M, O'leary JJ, Ruske J, Giulietti JM, Trevino-Villareal JH, MacArthur MR, Bolze A, Burak MF, Patterson S, Ho KJ, Carmody RN, Guzman RJ, Mitchell JR, Ozaki CK. Insights From a Short-Term Protein-Calorie Restriction Exploratory Trial in Elective Carotid Endarterectomy Patients. Vasc Endovascular Surg 2019; 53:470-476. [PMID: 31216949 DOI: 10.1177/1538574419856453] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND Open vascular surgery interventions are not infrequently hampered by complication rates and durability. Preclinical surgical models show promising beneficial effects in modulating the host response to surgical injury via short-term dietary preconditioning. Here, we explore short-term protein-calorie restriction preconditioning in patients undergoing elective carotid endarterectomy to understand patient participation dynamics and practicalities of robust research approaches around nutritional/surgical interventions. METHODS We designed a pilot prospective, multicenter, randomized controlled study in patients undergoing carotid endarterectomy. After a 3:2 randomization to a 3-day preoperative protein-calorie restriction regimen (30% calorie/70% protein restriction) or ad libitum group, blood, clinical parameters, and stool samples were collected at baseline, pre-op, and post-op days 1 and 30. Subcutaneous and perivascular adipose tissues were harvested periprocedurally. Samples were analyzed for standard chemistries and cell counts, adipokines. Bacterial DNA isolation and 16S rRNA sequencing were performed on stool samples and the relative abundance of bacterial species was measured. RESULTS Fifty-one patients were screened, 9 patients consented to the study, 5 were randomized, and 4 completed the trial. The main reason for non-consent was a 3-day in-hospital stay. All 4 participants were randomized to the protein-calorie restriction group, underwent successful endarterectomy, reported no compliance difficulties, nor were there adverse events. Stool analysis trended toward increased abundance of the sulfide-producing bacterial species Bilophila wadsworthia after dietary intervention (P = .08). CONCLUSIONS Although carotid endarterectomy patients held low enthusiasm for a 3-day preoperative inpatient stay, there were no adverse effects in this small cohort. Multidisciplinary longitudinal research processes were successfully executed throughout the nutritional/surgical intervention. Future translational endeavors into dietary preconditioning of vascular surgery patients should focus on outpatient approaches.
Collapse
Affiliation(s)
- Peter Kip
- 1 Division of Vascular and Endovascular Surgery, Brigham & Women's Hospital, Boston, MA, USA.,2 Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,3 Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Kaspar M Trocha
- 1 Division of Vascular and Endovascular Surgery, Brigham & Women's Hospital, Boston, MA, USA.,2 Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Ming Tao
- 1 Division of Vascular and Endovascular Surgery, Brigham & Women's Hospital, Boston, MA, USA
| | - James J O'leary
- 1 Division of Vascular and Endovascular Surgery, Brigham & Women's Hospital, Boston, MA, USA
| | - Jack Ruske
- 1 Division of Vascular and Endovascular Surgery, Brigham & Women's Hospital, Boston, MA, USA
| | - Jennifer M Giulietti
- 1 Division of Vascular and Endovascular Surgery, Brigham & Women's Hospital, Boston, MA, USA
| | - Jose H Trevino-Villareal
- 2 Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Michael R MacArthur
- 2 Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Andrew Bolze
- 1 Division of Vascular and Endovascular Surgery, Brigham & Women's Hospital, Boston, MA, USA
| | - M Furkan Burak
- 1 Division of Vascular and Endovascular Surgery, Brigham & Women's Hospital, Boston, MA, USA.,2 Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Suzannah Patterson
- 1 Division of Vascular and Endovascular Surgery, Brigham & Women's Hospital, Boston, MA, USA
| | - Karen J Ho
- 4 Department of Vascular Surgery, Northwestern Medicine Feinberg School of Medicine, Chicago, IL, USA
| | - Rachel N Carmody
- 5 Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - Raul J Guzman
- 6 Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - James R Mitchell
- 2 Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - C Keith Ozaki
- 1 Division of Vascular and Endovascular Surgery, Brigham & Women's Hospital, Boston, MA, USA
| |
Collapse
|