1
|
Darılmaz SE, Tugrul Aksakal MZ, Ceylan G, Kucukgergin C, Yetim Sahin A, Bekpinar S. Increased serum anti-angiogenic factor levels in insulin-resistant obese children and adolescents with or without liver steatosis (NAFL). Scand J Clin Lab Invest 2025:1-6. [PMID: 40366768 DOI: 10.1080/00365513.2025.2502947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 05/16/2025]
Abstract
This study aimed to investigate the effect of obesity on angiogenesis and its relationship with liver steatosis in obese children and adolescents. The study ıncluded 81 obese ın chıldren and 30 healthy controls. Obese subjects were subdıvıded by ultrasound ınto three groups: no steatosıs, grade 1 lıver steatosıs (NAFL), and grade 2 NAFL. Obese individuals, regardless of the presence of NAFL, exhibited significant insulin resistance (p < .01) compared to their lean counterparts. All obese subjects showed elevated serum ALT, wıth a sıgnıfıcantly greater ın those wıth NAFL. Marked dyslipidemia by decreased high-density lipoprotein (HDL) levels and elevated triglycerides, was observed in obese individuals with NAFL. The serum levels of angiopoietin-1 (Ang-1) and vascular endothelial growth factor-165b (VEGF165b) were measured as anti-angiogenic markers, while vascular endothelial growth factor-A (VEGF-A), fibroblast growth factor-2 (FGF-2) and P-selectin were assessed as pro-angiogenic factors. Compared to normal-weight children (5558 ± 674 pg/mL), Ang-1 levels were significantly elevated in all obese subgroups (8861 ± 1026; 8105 ± 615; 7388 ± 924, respectıvely). However, no significant differences in Ang-1 levels were observed among the obese subgroups. Ang-1 and VEGF165b levels were significantly higher in insulin-resistant individuals (7575 ± 747 pg/mL and 293 ± 44.4 pg/mL, respectively) compared to insulin-sensitive subjects (6143 ± 557 pg/mL and 179 ± 31.4 pg/mL, respectively). These findings suggest that insulin resistance in obese children is associated with altered angiogenic signaling. However, no significant differences in the serum levels of angiogenic factors were observed between obese groups with and without NAFL.
Collapse
Affiliation(s)
- Samed Emre Darılmaz
- Department of Biochemistry, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Melike Zeynep Tugrul Aksakal
- Department of Pediatrics, Division of Adolescent Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Gozde Ceylan
- Department of Biochemistry, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Canan Kucukgergin
- Department of Biochemistry, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Aylin Yetim Sahin
- Department of Pediatrics, Division of Adolescent Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Seldag Bekpinar
- Department of Biochemistry, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
2
|
Sharma S, Pierce J, Neverson JC, Khan R, Lee CF, Uppuluri S, Parry C, Amelotte E, Butler CA, Sellke FW, Harrington EO, Choudhary G, Morrison AR, Mantsounga CS. Macrophage Proangiogenic VEGF-A Is Required for Inflammatory Arteriogenesis During Vascular Injury. Biomedicines 2025; 13:828. [PMID: 40299401 PMCID: PMC12024885 DOI: 10.3390/biomedicines13040828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 03/14/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Background: Peripheral artery disease is associated with significant morbidity and mortality. Mechanical revascularization strategies are a mainstay of treatment but are often limited by the anatomic complexity of atherosclerotic lesions. Therapeutic angiogenesis has fallen short of being impactful due to fundamental gaps in our understanding of postdevelopmental angiogenesis. Methods: Using a preclinical model of peripheral artery disease involving acute vascular injury by femoral artery ligation along with cellular and molecular studies of VEGF-A expression, we sought to further understand the early role of macrophages in inflammatory angiogenesis and arteriogenesis. Results: Macrophage depletion studies revealed that the optimal levels of tissue VEGF-A expression, endothelial cell recruitment, and blood flow recovery were dependent on early macrophage recruitment. Proangiogenic VEGF-A expression was highest in macrophages polarized towards an inflammatory phenotype. Myeloid VEGF-A-deletion, while having no impact on the potent inflammatory cytokine, IL-1β, led to reductions in ischemic tissue VEGF-A, endothelial cell recruitment, and blood flow recovery due to impaired angiogenesis and arteriogenesis. Transplant of inflammatory polarized macrophages rescued the myeloid VEGF-A-deletion phenotype, leading to full blood flow recovery. Conclusions: Macrophages are a necessary and sufficient source of tissue VEGF-A during inflammatory-driven angiogenesis and arteriogenesis in response to vascular injury. Although further study is needed, cell-based therapeutic angiogenesis strategies involving the polarization of macrophages toward an inflammatory state, in order to produce high levels of proangiogenic VEGF-A, may be quite effective for improving revascularization in the context of PAD.
Collapse
Affiliation(s)
- Sheila Sharma
- Vascular Research Laboratory, Providence VA Medical Center, Providence, RI 02908, USA
- Ocean State Research Institute, Inc., Providence, RI 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Julia Pierce
- Vascular Research Laboratory, Providence VA Medical Center, Providence, RI 02908, USA
- Ocean State Research Institute, Inc., Providence, RI 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Jade C. Neverson
- Vascular Research Laboratory, Providence VA Medical Center, Providence, RI 02908, USA
- Ocean State Research Institute, Inc., Providence, RI 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Rachel Khan
- Vascular Research Laboratory, Providence VA Medical Center, Providence, RI 02908, USA
- Ocean State Research Institute, Inc., Providence, RI 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Cadence F. Lee
- Vascular Research Laboratory, Providence VA Medical Center, Providence, RI 02908, USA
- Ocean State Research Institute, Inc., Providence, RI 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Saketh Uppuluri
- Vascular Research Laboratory, Providence VA Medical Center, Providence, RI 02908, USA
- Ocean State Research Institute, Inc., Providence, RI 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Crystal Parry
- Vascular Research Laboratory, Providence VA Medical Center, Providence, RI 02908, USA
- Ocean State Research Institute, Inc., Providence, RI 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Elizabeth Amelotte
- Vascular Research Laboratory, Providence VA Medical Center, Providence, RI 02908, USA
- Ocean State Research Institute, Inc., Providence, RI 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Celia A. Butler
- Vascular Research Laboratory, Providence VA Medical Center, Providence, RI 02908, USA
- Ocean State Research Institute, Inc., Providence, RI 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Frank W. Sellke
- Vascular Research Laboratory, Providence VA Medical Center, Providence, RI 02908, USA
- Ocean State Research Institute, Inc., Providence, RI 02908, USA
- Cardiovascular Research Center, Brown University Health, Rhode Island Hospital, Providence, RI 02903, USA
| | - Elizabeth O. Harrington
- Vascular Research Laboratory, Providence VA Medical Center, Providence, RI 02908, USA
- Ocean State Research Institute, Inc., Providence, RI 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Gaurav Choudhary
- Vascular Research Laboratory, Providence VA Medical Center, Providence, RI 02908, USA
- Ocean State Research Institute, Inc., Providence, RI 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, RI 02903, USA
- Cardiovascular Research Center, Brown University Health, Rhode Island Hospital, Providence, RI 02903, USA
| | - Alan R. Morrison
- Vascular Research Laboratory, Providence VA Medical Center, Providence, RI 02908, USA
- Ocean State Research Institute, Inc., Providence, RI 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Chris S. Mantsounga
- Vascular Research Laboratory, Providence VA Medical Center, Providence, RI 02908, USA
- Ocean State Research Institute, Inc., Providence, RI 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, RI 02903, USA
| |
Collapse
|
3
|
Ahmed B, Farb MG, Gokce N. Cardiometabolic implications of adipose tissue aging. Obes Rev 2024; 25:e13806. [PMID: 39076025 DOI: 10.1111/obr.13806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 03/14/2024] [Accepted: 07/05/2024] [Indexed: 07/31/2024]
Abstract
Adipose tissue is a large endocrine organ that serves numerous physiological functions. As we age, adipose tissue remodels and can develop functional changes that alters its phenotype, potentially contributing to metabolic and cardiovascular disorders. Aging adipose tissue is characterized by regional redistribution of fat, accumulation of senescent cells, fibrosis, and decline in adipocyte differentiation capacities, which collectively impact adipose tissue function and whole body health. A notable transformation involves increased accumulation of intra-abdominal visceral adipose tissue and ectopic fat around internal organs such as the heart, blood vessels, liver, and kidneys that alter their functions. Other changes associated with aging include alterations in adipokine secretion and changes in adipocyte size and numbers. Aging adipocytes play a role in mediating chronic inflammation, metabolic dysfunction, and insulin resistance. Visceral adipose tissue, which increases in volume with aging, is in particular associated with inflammation, angiogenic dysfunction, and microvascular abnormalities, and mediators released by visceral fat may have adverse consequences systemically in multiple target organs, including the cardiovascular system. Understanding mechanisms underlying adipose tissue aging and its impact on cardiovascular health are important for developing interventions and treatments to promote healthy aging and reduce cardiometabolic disease risk.
Collapse
Affiliation(s)
- Bulbul Ahmed
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Melissa G Farb
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Noyan Gokce
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
4
|
Dronkers J, van Veldhuisen DJ, van der Meer P, Meems LMG. Heart Failure and Obesity: Unraveling Molecular Mechanisms of Excess Adipose Tissue. J Am Coll Cardiol 2024; 84:1666-1677. [PMID: 39415402 DOI: 10.1016/j.jacc.2024.07.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 07/01/2024] [Accepted: 07/11/2024] [Indexed: 10/18/2024]
Abstract
Obesity is an ongoing pandemic and is associated with the development of heart failure (HF), and especially HF with preserved ejection fraction. The definition of obesity is currently based on anthropometric measurements but neglects the location and molecular properties of excess fat. Important depots associated with HF development are subcutaneous adipose tissue and visceral adipose tissue, both located in the abdominal region, and epicardial adipose tissue (EAT) surrounding the myocardium. However, mechanisms linking these different adipose tissue depots to HF development are incompletely understood. EAT in particular is of great interest because of its close proximity to the heart. In this review, we therefore focus on the characteristics of different adipose tissue depots and their response to obesity. In addition, we evaluate how different mechanisms associated with EAT expansion potentially contribute to HF and in particular HF with preserved ejection fraction development.
Collapse
Affiliation(s)
- Just Dronkers
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Dirk J van Veldhuisen
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Peter van der Meer
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Laura M G Meems
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands.
| |
Collapse
|
5
|
Jia G, Sowers JR, Whaley-Connell A. Obesity in Hypertension: The Role of the Expanding Waistline Over the Years and Insights Into the Future. Hypertension 2024; 81:687-690. [PMID: 38018438 PMCID: PMC10954419 DOI: 10.1161/hypertensionaha.123.21719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2023]
Affiliation(s)
- Guanghong Jia
- Department of Medicine-Endocrinology and Metabolism, University of Missouri School of Medicine, Columbia, MO, 65212, USA
- Research Service, Harry S Truman Memorial Veterans Hospital, Research Service, 800 Hospital Dr, Columbia, MO, 65201, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, 65212, USA
| | - James R. Sowers
- Department of Medicine-Endocrinology and Metabolism, University of Missouri School of Medicine, Columbia, MO, 65212, USA
- Research Service, Harry S Truman Memorial Veterans Hospital, Research Service, 800 Hospital Dr, Columbia, MO, 65201, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, 65212, USA
- Department of Medicine–Nephrology and Hypertension, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Adam Whaley-Connell
- Department of Medicine-Endocrinology and Metabolism, University of Missouri School of Medicine, Columbia, MO, 65212, USA
- Research Service, Harry S Truman Memorial Veterans Hospital, Research Service, 800 Hospital Dr, Columbia, MO, 65201, USA
- Department of Medicine–Nephrology and Hypertension, University of Missouri School of Medicine, Columbia, MO 65212, USA
| |
Collapse
|
6
|
Engin A. Adipose Tissue Hypoxia in Obesity: Clinical Reappraisal of Hypoxia Hypothesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:329-356. [PMID: 39287857 DOI: 10.1007/978-3-031-63657-8_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Obese subjects exhibit lower adipose tissue oxygen consumption in accordance with the lower adipose tissue blood flow. Thereby, compared to lean subjects, obese individuals have almost half lower capillary density and more than half lower vascular endothelial growth factor (VEGF). The VEGF expression together with hypoxia-inducible transcription factor-1 alpha (HIF-1α) activity also requires phosphatidylinositol 3-kinase (PI3K) and mammalian target of rapamycin (mTOR)-mediated signaling. Especially HIF-1α is an important signaling molecule for hypoxia to induce the inflammatory responses. Hypoxia contributes to several biological functions, such as angiogenesis, cell proliferation, apoptosis, inflammation, and insulin resistance (IR). Pathogenesis of obesity-related comorbidities is attributed to intermittent hypoxia (IH), which is mostly observed in visceral obesity. Proinflammatory phenotype of the adipose tissue is a crucial link between IH and the development of IR. Inhibition of adaptive unfolded protein response (UPR) in hypoxia increases β cell death. Moreover, deletion of HIF-1α worsens β cell function. Oxidative stress, as well as the release of proinflammatory cytokines/adipokines in obesity, is proportional to the severity of IH. Reactive oxygen species (ROS) generation at mitochondria is responsible for propagation of the hypoxic signal; however, mitochondrial ROS production is required for hypoxic HIF-1α protein stabilization. Alterations in oxygen availability of adipose tissue directly affect the macrophage polarization and are responsible for the dysregulated adipocytokines production in obesity. Hypoxia both inhibits adipocyte differentiation from preadipocytes and macrophage migration from the hypoxic adipose tissue. Upon reaching a hypertrophic threshold beyond the adipocyte fat loading capacity, excess extracellular matrix (ECM) components are deposited, causing fibrosis. HIF-1α initiates the whole pathological process of fibrosis and inflammation in the obese adipose tissue. In addition to stressed adipocytes, hypoxia contributes to immune cell migration and activation which further aggravates adipose tissue fibrosis. Therefore, targeting HIF-1α might be an efficient way to suppress hypoxia-induced pathological changes in the ECM. The fibrosis score of adipose tissue correlates negatively with the body mass index and metabolic parameters. Inducers of browning/beiging adipocytes and adipokines, as well as modulations of matrix remodeling enzyme inhibitors, and associated gene regulators, are potential pharmacological targets for treating obesity.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
7
|
Ahmed B, Farb MG, Karki S, D'Alessandro S, Edwards NM, Gokce N. Pericardial Adipose Tissue Thrombospondin-1 Associates With Antiangiogenesis in Ischemic Heart Disease. Am J Cardiol 2024; 210:201-207. [PMID: 37863116 PMCID: PMC10842123 DOI: 10.1016/j.amjcard.2023.09.104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 09/15/2023] [Accepted: 09/24/2023] [Indexed: 10/22/2023]
Abstract
Accumulation of ectopic pericardial adipose tissue has been associated with cardiovascular complications which, in part, may relate to adipose-derived factors that regulate vascular responses and angiogenesis. We sought to characterize adipose tissue microvascular angiogenic capacity in subjects who underwent elective cardiac surgeries including aortic, valvular, and coronary artery bypass grafting. Pericardial adipose tissue was collected intraoperatively and examined for angiogenic capacity. Capillary sprouting was significantly blunted (twofold, p <0.001) in subjects with coronary artery disease (CAD) (age 60 ± 9 years, body mass index [BMI] 32 ± 4 kg/m2, low-density lipoprotein cholesterol [LDL-C] 95 ± 46 mg/100 ml, n = 29) compared with age-, BMI-, and LDL-C matched subjects without angiographic obstructive CAD (age 59 ± 10 y, BMI 35 ± 9 kg/m2, LDL-C 101 ± 40 mg/100 ml, n = 12). For potential mechanistic insight, we performed mRNA expression analyses using quantitative real-time polymerase chain reaction and observed no significant differences in pericardial fat gene expression of proangiogenic mediators vascular endothelial growth factor-A (VEGF-A), fibroblast growth factor-2 (FGF-2), and angiopoietin-1 (angpt1), or anti-angiogenic factors soluble fms-like tyrosine kinase-1 (sFlt-1) and endostatin. In contrast, mRNA expression of anti-angiogenic thrombospondin-1 (TSP-1) was significantly upregulated (twofold, p = 0.008) in CAD compared with non-CAD subjects, which was confirmed by protein western-immunoblot analysis. TSP-1 gene knockdown using short hairpin RNA lentiviral delivery significantly improved angiogenic deficiency in CAD (p <0.05). In conclusion, pericardial fat in subjects with CAD may be associated with an antiangiogenic profile linked to functional defects in vascularization capacity. Local paracrine actions of TSP-1 in adipose depots surrounding the heart may play a role in mechanisms of ischemic heart disease.
Collapse
Affiliation(s)
- Bulbul Ahmed
- Evans Department of Medicine, Boston University School of Medicine, Boston, Massachusetts; Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
| | - Melissa G Farb
- Evans Department of Medicine, Boston University School of Medicine, Boston, Massachusetts; Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
| | - Shakun Karki
- Evans Department of Medicine, Boston University School of Medicine, Boston, Massachusetts; Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
| | - Sophia D'Alessandro
- Evans Department of Medicine, Boston University School of Medicine, Boston, Massachusetts; Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
| | - Niloo M Edwards
- Division of Cardiac Surgery, Boston Medical Center, Boston, Massachusetts
| | - Noyan Gokce
- Evans Department of Medicine, Boston University School of Medicine, Boston, Massachusetts; Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts.
| |
Collapse
|
8
|
AlZaim I, de Rooij LPMH, Sheikh BN, Börgeson E, Kalucka J. The evolving functions of the vasculature in regulating adipose tissue biology in health and obesity. Nat Rev Endocrinol 2023; 19:691-707. [PMID: 37749386 DOI: 10.1038/s41574-023-00893-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/17/2023] [Indexed: 09/27/2023]
Abstract
Adipose tissue is an endocrine organ and a crucial regulator of energy storage and systemic metabolic homeostasis. Additionally, adipose tissue is a pivotal regulator of cardiovascular health and disease, mediated in part by the endocrine and paracrine secretion of several bioactive products, such as adipokines. Adipose vasculature has an instrumental role in the modulation of adipose tissue expansion, homeostasis and metabolism. The role of the adipose vasculature has been extensively explored in the context of obesity, which is recognized as a global health problem. Obesity-induced accumulation of fat, in combination with vascular rarefaction, promotes adipocyte dysfunction and induces oxidative stress, hypoxia and inflammation. It is now recognized that obesity-associated endothelial dysfunction often precedes the development of cardiovascular diseases. Investigations have revealed heterogeneity within the vascular niche and dynamic reciprocity between vascular and adipose cells, which can become dysregulated in obesity. Here we provide a comprehensive overview of the evolving functions of the vasculature in regulating adipose tissue biology in health and obesity.
Collapse
Affiliation(s)
- Ibrahim AlZaim
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Laura P M H de Rooij
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Bilal N Sheikh
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich, Leipzig, Germany
- Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Emma Börgeson
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Joanna Kalucka
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark.
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark.
| |
Collapse
|
9
|
Do T, Van A, Ataei A, Sharma S, Mohandas R. Microvascular Dysfunction in Obesity-Hypertension. Curr Hypertens Rep 2023; 25:447-453. [PMID: 37837517 DOI: 10.1007/s11906-023-01272-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2023] [Indexed: 10/16/2023]
Abstract
PURPOSE OF REVIEW This review aims to explore the role of microvascular dysfunction in obesity-hypertension, discuss the effects obesity has on renal microvasculature, review the current methods for assessing microvascular dysfunction and available therapeutic options, and identify critical areas for further research. RECENT FINDINGS There is a strong association between obesity and hypertension. However, the pathophysiology of obesity-hypertension is not clear. Microvascular dysfunction has been linked to hypertension and obesity and could be an important mediator of obesity-related hypertension. Newer therapies for hypertension and obesity could have ameliorating effects on microvascular dysfunction, including GLP-1 agonists and SGLT-2 inhibitors. There is still much progress to be made in our understanding of the complex interplay between obesity, hypertension, and microvascular dysfunction. Continued efforts to understand microvascular dysfunction and its role in obesity-hypertension are crucial to develop precision therapy to target obesity-hypertension.
Collapse
Affiliation(s)
- Tammy Do
- Department of Medicine, LSU Health Sciences Center - New Orleans, New Orleans, LA, USA
| | - Ashley Van
- Department of Medicine, LSU Health Sciences Center - New Orleans, New Orleans, LA, USA
| | - Arash Ataei
- Department of Medicine, LSU Health Sciences Center - New Orleans, New Orleans, LA, USA
| | - Swati Sharma
- Section of Nephrology and Hypertension, LSU Health Sciences Center - New Orleans, 2021 Perdido Street, Ste 4325, New Orleans, LA, 70112, USA
| | - Rajesh Mohandas
- Section of Nephrology and Hypertension, LSU Health Sciences Center - New Orleans, 2021 Perdido Street, Ste 4325, New Orleans, LA, 70112, USA.
| |
Collapse
|
10
|
Gao H, Li Y, Jin Y, Zhang L, Xia X, Liu J, Wang H, Xie Y, Ding W. Electroacupuncture activates angiogenesis by regulating the PI3K/Pten/Thbs1 signaling pathway to promote the browning of adipose tissue in HFD-induced obese mice. Biomed Pharmacother 2023; 166:115386. [PMID: 37651803 DOI: 10.1016/j.biopha.2023.115386] [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: 06/25/2023] [Revised: 08/23/2023] [Accepted: 08/23/2023] [Indexed: 09/02/2023] Open
Abstract
This study investigated the effect of electroacupuncture (EA) on the browning of white adipose tissue (WAT) via angiogenesis and its potential mechanism in obese mice. Four-week-old male C56BL/6 mice were randomly divided into a high-fat diet (HFD) and a normal chow diet (ND) group. After 12 weeks, HFD mice were randomly divided into two groups to receive or not receive EA for 3 weeks. After EA treatment, body weight, adipocyte size, serum glucose (GLU), triacylglycerol (TG), cholesterol (CHO), leptin (Lep), monocyte chemoattractant protein-1 (MCP-1), WAT browning-related genes, angiogenesis-related genes, and the PI3K/Pten/Thbs1 signaling pathway were evaluated. The results indicated that EA significantly reduced body weight, adipocyte size, and serum concentrations of GLU, TG, CHO, Lep and MCP-1 and promoted WAT browning. Angiogenesis and the PI3K/Pten/Thbs1 signaling pathway were all activated by EA intervention. The expression levels were consistent with the results of RNA-seq and confirmed via qRTPCR and WB. Our study showed that EA may activate angiogenesis via the PI3K/Pten/Thbs1 signaling pathway in WAT, thereby promoting the browning and thermogenesis of adipose tissue.
Collapse
Affiliation(s)
- Hongyan Gao
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yanhui Li
- Chongqing Hospital of Traditional Chinese Medicine, Chongqing 400021, China
| | - Yue Jin
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Li Zhang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xiuwen Xia
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jinkun Liu
- Chongqing Hospital of Traditional Chinese Medicine, Chongqing 400021, China
| | - Huaifu Wang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ya Xie
- Sichuan Provincial People's Hospital Jinniu Hospital, Chengdu 610007, China
| | - Weijun Ding
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
11
|
Roy PK, Islam J, Lalhlenmawia H. Prospects of potential adipokines as therapeutic agents in obesity-linked atherogenic dyslipidemia and insulin resistance. Egypt Heart J 2023; 75:24. [PMID: 37014444 PMCID: PMC10073393 DOI: 10.1186/s43044-023-00352-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 03/28/2023] [Indexed: 04/05/2023] Open
Abstract
BACKGROUND In normal circumstances, AT secretes anti-inflammatory adipokines (AAKs) which regulates lipid metabolism, insulin sensitivity, vascular hemostasis, and angiogenesis. However, during obesity AT dysfunction occurs and leads to microvascular imbalance and secretes several pro-inflammatory adipokines (PAKs), thereby favoring atherogenic dyslipidemia and insulin resistance. Literature suggests decreased levels of circulating AAKs and increased levels of PAKs in obesity-linked disorders. Importantly, AAKs have been reported to play a vital role in obesity-linked metabolic disorders mainly insulin resistance, type-2 diabetes mellitus and coronary heart diseases. Interestingly, AAKs counteract the microvascular imbalance in AT and exert cardioprotection via several signaling pathways such as PI3-AKT/PKB pathway. Although literature reviews have presented a number of investigations detailing specific pathways involved in obesity-linked disorders, literature concerning AT dysfunction and AAKs remains sketchy. In view of the above, in the present contribution an effort has been made to provide an insight on the AT dysfunction and role of AAKs in modulating the obesity and obesity-linked atherogenesis and insulin resistance. MAIN BODY "Obesity-linked insulin resistance", "obesity-linked cardiometabolic disease", "anti-inflammatory adipokines", "pro-inflammatory adipokines", "adipose tissue dysfunction" and "obesity-linked microvascular dysfunction" are the keywords used for searching article. Google scholar, Google, Pubmed and Scopus were used as search engines for the articles. CONCLUSIONS This review offers an overview on the pathophysiology of obesity, management of obesity-linked disorders, and areas in need of attention such as novel therapeutic adipokines and their possible future perspectives as therapeutic agents.
Collapse
Affiliation(s)
- Probin Kr Roy
- Department of Pharmacy, Regional Institute of Paramedical and Nursing Sciences (RIPANS), Aizawl, Mizoram, 796017, India.
| | - Johirul Islam
- Coromandel International Limited, Hyderabad, Telangana, 500101, India
| | - Hauzel Lalhlenmawia
- Department of Pharmacy, Regional Institute of Paramedical and Nursing Sciences (RIPANS), Aizawl, Mizoram, 796017, India
| |
Collapse
|
12
|
Pan J, Yin J, Gan L, Xue J. Two-sided roles of adipose tissue: Rethinking the obesity paradox in various human diseases from a new perspective. Obes Rev 2023; 24:e13521. [PMID: 36349390 DOI: 10.1111/obr.13521] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 08/05/2022] [Accepted: 10/17/2022] [Indexed: 11/11/2022]
Abstract
Overweight and obesity, as a result of excess fat accumulation, have become a worldwide public health issue. Recent studies have shown that obesity is closely related to many human diseases, such as cancer, cardiovascular diseases, and type 2 diabetes mellitus, in which adipose tissue plays a dual role. In addition to thermal and mechanical insulation and a critical role in energy storage and heat production, adipose tissue is also a highly plastic endocrine and signaling organ that secretes multiple bioactive molecules for inter-organ crosstalk. The phenotypic and biological changes of adipose tissue under pathological conditions, especially in obesity, increase the challenge of deciphering the positive or negative effects of adipose tissue in disease. Despite numerous studies on obesity and adipose tissue, the ambiguous role of adipose tissue on specific organs or tissues in different diseases is not fully understood, and the definite mechanisms remain obscure. In this review, we first summarize the basic biological characteristics of adipose tissue in the physiological state and the abnormal remodeling of adipose tissue during obesity. We then discuss the complex and disparate effects of obesity on various human diseases, with a particular focus on the dual roles and underlying mechanisms of adipose tissue, a quintessential player in obesity, in this process. More importantly, rethinking the causes of the "obesity paradox" phenomenon in diseases from the perspective of adipose homeostasis and dysfunction provides a novel strategy for disease treatment by intervening in fat function.
Collapse
Affiliation(s)
- Jing Pan
- Thoracic Oncology Ward, Cancer Center and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Jianqiong Yin
- Thoracic Oncology Ward, Cancer Center and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Lu Gan
- Research Laboratory of Emergency Medicine, Department of Emergency Medicine, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Jianxin Xue
- Thoracic Oncology Ward, Cancer Center and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.,Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
13
|
Balakrishnan B, Gupta A, Basri R, Sharma VM, Slayton M, Gentner K, Becker CC, Karki S, Muturi H, Najjar SM, Loria AS, Gokce N, Puri V. Endothelial-Specific Expression of CIDEC Improves High-Fat Diet-Induced Vascular and Metabolic Dysfunction. Diabetes 2023; 72:19-32. [PMID: 36256836 PMCID: PMC9797323 DOI: 10.2337/db22-0294] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 10/10/2022] [Indexed: 01/19/2023]
Abstract
Cell death-inducing DNA fragmentation factor-α-like effector C (CIDEC), originally identified to be a lipid droplet-associated protein in adipocytes, positively associates with insulin sensitivity. Recently, we discovered that it is expressed abundantly in human endothelial cells and regulates vascular function. The current study was designed to characterize the physiological effects and molecular actions of endothelial CIDEC in the control of vascular phenotype and whole-body glucose homeostasis. To achieve this, we generated a humanized mouse model expressing endothelial-specific human CIDEC (E-CIDECtg). E-CIDECtg mice exhibited protection against high-fat diet-induced glucose intolerance, insulin resistance, and dyslipidemia. Moreover, these mice displayed improved insulin signaling and endothelial nitric oxide synthase activation, enhanced endothelium-dependent vascular relaxation, and improved vascularization of adipose tissue, skeletal muscle, and heart. Mechanistically, we identified a novel interplay of CIDEC-vascular endothelial growth factor A (VEGFA)-vascular endothelial growth factor receptor 2 (VEGFR2) that reduced VEGFA and VEGFR2 degradation, thereby increasing VEGFR2 activation. Overall, our results demonstrate a protective role of endothelial CIDEC against obesity-induced metabolic and vascular dysfunction, in part, by modulation of VEGF signaling. These data suggest that CIDEC may be investigated as a potential future therapeutic target for mitigating obesity-related cardiometabolic disease.
Collapse
Affiliation(s)
- Bijinu Balakrishnan
- Department of Biomedical Sciences and Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
| | - Abhishek Gupta
- Department of Biomedical Sciences and Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
| | - Rabia Basri
- Department of Biomedical Sciences and Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
| | - Vishva M. Sharma
- Department of Biomedical Sciences and Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
| | - Mark Slayton
- Department of Biomedical Sciences and Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
| | - Kailey Gentner
- Department of Biomedical Sciences and Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
| | - Chloe C. Becker
- Department of Biomedical Sciences and Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
| | - Shakun Karki
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA
| | - Harrison Muturi
- Department of Biomedical Sciences and Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
| | - Sonia M. Najjar
- Department of Biomedical Sciences and Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
| | - Analia S. Loria
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY
| | - Noyan Gokce
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA
| | - Vishwajeet Puri
- Department of Biomedical Sciences and Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
| |
Collapse
|
14
|
Liao ZZ, Ran L, Qi XY, Wang YD, Wang YY, Yang J, Liu JH, Xiao XH. Adipose endothelial cells mastering adipose tissues metabolic fate. Adipocyte 2022; 11:108-119. [PMID: 35067158 PMCID: PMC8786343 DOI: 10.1080/21623945.2022.2028372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/04/2022] [Accepted: 01/07/2022] [Indexed: 11/16/2022] Open
Abstract
Dynamic communication within adipose tissue depends on highly vascularized structural characteristics to maintain systemic metabolic homoeostasis. Recently, it has been noted that adipose endothelial cells (AdECs) act as essential bridges for biological information transmission between adipose-resident cells. Hence, paracrine regulators that mediate crosstalk between AdECs and adipose stromal cells were summarized. We also highlight the importance of AdECs to maintain adipocytes metabolic homoeostasis by regulating insulin sensitivity, lipid turnover and plasticity. The differential regulation of AdECs in adipose plasticity often depends on vascular density and metabolic states. Although choosing pro-angiogenic or anti-angiogenic therapies for obesity is still a matter of debate in clinical settings, the growing numbers of drugs have been confirmed to play an anti-obesity effect by affecting vascularization. Pharmacologic angiogenesis intervention has great potential as therapeutic strategies for obesity.
Collapse
Affiliation(s)
- Zhe-Zhen Liao
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Li Ran
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xiao-Yan Qi
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Ya-Di Wang
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yuan-Yuan Wang
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jing Yang
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jiang-Hua Liu
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xin-Hua Xiao
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
15
|
Abstract
While most tissues exhibit their greatest growth during development, adipose tissue is capable of additional massive expansion in adults. Adipose tissue expandability is advantageous when temporarily storing fuel for use during fasting, but becomes pathological upon continuous food intake, leading to obesity and its many comorbidities. The dense vasculature of adipose tissue provides necessary oxygen and nutrients, and supports delivery of fuel to and from adipocytes under fed or fasting conditions. Moreover, the vasculature of adipose tissue comprises a major niche for multipotent progenitor cells, which give rise to new adipocytes and are necessary for tissue repair. Given the multiple, pivotal roles of the adipose tissue vasculature, impairments in angiogenic capacity may underlie obesity-associated diseases such as diabetes and cardiometabolic disease. Exciting new studies on the single-cell and single-nuclei composition of adipose tissues in mouse and humans are providing new insights into mechanisms of adipose tissue angiogenesis. Moreover, new modes of intercellular communication involving micro vesicle and exosome transfer of proteins, nucleic acids and organelles are also being recognized to play key roles. This review focuses on new insights on the cellular and signaling mechanisms underlying adipose tissue angiogenesis, and on their impact on obesity and its pathophysiological consequences.
Collapse
|
16
|
Sakers A, De Siqueira MK, Seale P, Villanueva CJ. Adipose-tissue plasticity in health and disease. Cell 2022; 185:419-446. [PMID: 35120662 PMCID: PMC11152570 DOI: 10.1016/j.cell.2021.12.016] [Citation(s) in RCA: 439] [Impact Index Per Article: 146.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/08/2021] [Accepted: 12/13/2021] [Indexed: 12/11/2022]
Abstract
Adipose tissue, colloquially known as "fat," is an extraordinarily flexible and heterogeneous organ. While historically viewed as a passive site for energy storage, we now appreciate that adipose tissue regulates many aspects of whole-body physiology, including food intake, maintenance of energy levels, insulin sensitivity, body temperature, and immune responses. A crucial property of adipose tissue is its high degree of plasticity. Physiologic stimuli induce dramatic alterations in adipose-tissue metabolism, structure, and phenotype to meet the needs of the organism. Limitations to this plasticity cause diminished or aberrant responses to physiologic cues and drive the progression of cardiometabolic disease along with other pathological consequences of obesity.
Collapse
Affiliation(s)
- Alexander Sakers
- Institute for Diabetes, Obesity & Metabolism, Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Mirian Krystel De Siqueira
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, Los Angeles, CA 90095-7070 USA; Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095-7070 USA
| | - Patrick Seale
- Institute for Diabetes, Obesity & Metabolism, Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104 USA.
| | - Claudio J Villanueva
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, Los Angeles, CA 90095-7070 USA; Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095-7070 USA.
| |
Collapse
|
17
|
Macrophage IL-1β promotes arteriogenesis by autocrine STAT3- and NF-κB-mediated transcription of pro-angiogenic VEGF-A. Cell Rep 2022; 38:110309. [PMID: 35108537 PMCID: PMC8865931 DOI: 10.1016/j.celrep.2022.110309] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 09/20/2021] [Accepted: 01/07/2022] [Indexed: 11/23/2022] Open
Abstract
Peripheral artery disease (PAD) leads to considerable morbidity, yet strategies for therapeutic angiogenesis fall short of being impactful. Inflammatory macrophage subsets play an important role in orchestrating post-developmental angiogenesis, but the underlying mechanisms are unclear. Here, we find that macrophage VEGF-A expression is dependent upon the potent inflammatory cytokine, IL-1β. IL-1β promotes pro-angiogenic VEGF-A165a isoform transcription via activation and promoter binding of STAT3 and NF-κB, as demonstrated by gene-deletion, gain-of-function, inhibition, and chromatin immunoprecipitation assays. Conversely, IL-1β-deletion or inhibition of STAT3 or NF-κB increases anti-angiogenic VEGF-A165b isoform expression, indicating IL-1β signaling may also direct splice variant selection. In an experimental PAD model of acute limb ischemia, macrophage IL-1β expression is required for pro-angiogenic VEGF-A expression and for VEGF-A-induced blood flow recovery via angio- or arteriogenesis. Though further study is needed, macrophage IL-1β-dependent transcription of VEGF-A via STAT3 and NF-κB may have potential to therapeutically promote angiogenesis in the setting of PAD. Mantsounga et al. show inflammatory macrophage IL-1β expression to be required for pro-angiogenic VEGF-A expression and consequent post-developmental angio- or arteriogenesis in an experimental model of peripheral artery disease. Autocrine IL-1β signaling promotes transcription of pro-angiogenic VEGF-A165a isoform expression relative to anti-angiogenic isoform, VEGF-A165b, through activation of STAT3 and NF-κB.
Collapse
|
18
|
Oh KK, Adnan M, Cho DH. Elucidating Drug-Like Compounds and Potential Mechanisms of Corn Silk ( Stigma Maydis) against Obesity: A Network Pharmacology Study. Curr Issues Mol Biol 2021; 43:1906-1936. [PMID: 34889899 PMCID: PMC8929052 DOI: 10.3390/cimb43030133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/27/2021] [Accepted: 11/03/2021] [Indexed: 11/16/2022] Open
Abstract
Corn silk (Stigma Maydis) has been utilized as an important herb against obesity by Chinese, Korean, and Native Americans, but its phytochemicals and mechanisms(s) against obesity have not been deciphered completely. This study aimed to identify promising bioactive constituents and mechanism of action(s) of corn silk (CS) against obesity via network pharmacology. The compounds from CS were identified using Gas Chromatography Mass Spectrometry (GC-MS) and were confirmed ultimately by Lipinski's rule via SwissADME. The relationships of the compound-targets or obesity-related targets were confirmed by public bioinformatics. The signaling pathways related to obesity, protein-protein interaction (PPI), and signaling pathways-targets-bioactives (STB) were constructed, visualized, and analyzed by RPackage. Lastly, Molecular Docking Test (MDT) was performed to validate affinity between ligand(s) and protein(s) on key signaling pathway(s). We identified a total of 36 compounds from CS via GC-MS, all accepted by Lipinski's rule. The number of 36 compounds linked to 154 targets, 85 among 154 targets related directly to obesity-targets (3028 targets). Of the final 85 targets, we showed that the PPI network (79 edges, 357 edges), 12 signaling pathways on a bubble chart, and STB network (67 edges, 239 edges) are considered as therapeutic components. The MDT confirmed that two key activators (β-Amyrone, β-Stigmasterol) bound most stably to PPARA, PPARD, PPARG, FABP3, FABP4, and NR1H3 on the PPAR signaling pathway, also, three key inhibitors (Neotocopherol, Xanthosine, and β-Amyrone) bound most tightly to AKT1, IL6, FGF2, and PHLPP1 on the PI3K-Akt signaling pathway. Overall, we provided promising key signaling pathways, targets, and bioactives of CS against obesity, suggesting crucial pharmacological evidence for further clinical testing.
Collapse
Affiliation(s)
| | | | - Dong-Ha Cho
- Department of Bio-Health Convergence, College of Biomedical Science, Kangwon National University, Chuncheon 24341, Korea; (K.-K.O.); (M.A.)
| |
Collapse
|
19
|
Cornelius VA, Fulton JR, Margariti A. Alternative Splicing: A Key Mediator of Diabetic Vasculopathy. Genes (Basel) 2021; 12:1332. [PMID: 34573314 PMCID: PMC8469645 DOI: 10.3390/genes12091332] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/24/2021] [Accepted: 08/24/2021] [Indexed: 12/25/2022] Open
Abstract
Cardiovascular disease is the leading cause of death amongst diabetic individuals. Atherosclerosis is the prominent driver of diabetic vascular complications, which is triggered by the detrimental effects of hyperglycemia and oxidative stress on the vasculature. Research has extensively shown diabetes to result in the malfunction of the endothelium, the main component of blood vessels, causing severe vascular complications. The pathogenic mechanism in which diabetes induces vascular dysfunction, however, remains largely unclear. Alternative splicing of protein coding pre-mRNAs is an essential regulatory mechanism of gene expression and is accepted to be intertwined with cellular physiology. Recently, a role for alternative splicing has arisen within vascular health, with aberrant mis-splicing having a critical role in disease development, including in atherosclerosis. This review focuses on the current knowledge of alternative splicing and the roles of alternatively spliced isoforms within the vasculature, with a particular focus on disease states. Furthermore, we explore the recent elucidation of the alternatively spliced QKI gene within vascular cell physiology and the onset of diabetic vasculopathy. Potential therapeutic strategies to restore aberrant splicing are also discussed.
Collapse
Affiliation(s)
| | | | - Andriana Margariti
- The Wellcome-Wolfson Institute of Experimental Medicine, Belfast BT9 7BL, UK; (V.A.C.); (J.R.F.)
| |
Collapse
|
20
|
Aasbrenn M, Svendstrup M, Schnurr TM, Lindqvist Hansen D, Worm D, Balslev-Harder M, Grarup N, Burgdorf KS, Vestergaard H, Pedersen O, Ängquist L, Fenger M, Sørensen TIA, Madsbad S, Hansen T. Genetic markers of abdominal obesity and weight loss after gastric bypass surgery. PLoS One 2021; 16:e0252525. [PMID: 34048505 PMCID: PMC8162622 DOI: 10.1371/journal.pone.0252525] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 05/17/2021] [Indexed: 11/18/2022] Open
Abstract
Background Weight loss after bariatric surgery varies widely between individuals, partly due to genetic differences. In addition, genetic determinants of abdominal obesity have been shown to attenuate weight loss after dietary intervention with special attention paid to the rs1358980-T risk allele in the VEGFA locus. Here we aimed to test if updated genetic risk scores (GRSs) for adiposity measures and the rs1358980-T risk allele are linked with weight loss following gastric bypass surgery. Methods Five hundred seventy six patients with morbid obesity underwent Roux-en-Y gastric bypass. A GRS for BMI and a GRS for waist-hip-ratio adjusted for BMI (proxy for abdominal obesity), respectively, were constructed. All patients were genotyped for the rs1358980-T risk allele. Associations between the genetic determinants and weight loss after bariatric surgery were evaluated. Results The GRS for BMI was not associated with weight loss (β = -2.0 kg/100 risk alleles, 95% CI -7.5 to 3.3, p = 0.45). Even though the GRS for abdominal obesity was associated with an attenuated weight loss response adjusted for age, sex and center (β = -14.6 kg/100 risk alleles, 95% CI -25.4 to -3.8, p = 0.008), it was not significantly associated with weight loss after adjustment for baseline BMI (β = -7.9 kg/100 risk alleles, 95% CI -17.5 to 1.6, p = 0.11). Similarly, the rs1358980-T risk allele was not significantly associated with weight loss (β = -0.8 kg/risk allele, 95% CI -2.2 to 0.6, p = 0.25). Discussion GRSs for adiposity derived from large meta-analyses and the rs1358980-T risk allele in the VEGFA locus did not predict weight loss after gastric bypass surgery. The association between a GRS for abdominal obesity and the response to bariatric surgery may be dependent on the association between the GRS and baseline BMI.
Collapse
Affiliation(s)
- Martin Aasbrenn
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Geriatric Research Unit, Department of Geriatrics, Bispebjerg-Frederiksberg University Hospital, Copenhagen, Denmark
- * E-mail:
| | - Mathilde Svendstrup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Endocrinology, Bispebjerg-Frederiksberg University Hospital, Copenhagen, Denmark
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Theresia M. Schnurr
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Dorte Worm
- Department of Medicine, Amager Hospital, Copenhagen, Denmark
| | - Marie Balslev-Harder
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Henrik Vestergaard
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Bornholms Hospital, Rønne, Denmark
| | - Oluf Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lars Ängquist
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mogens Fenger
- Department of Clinical Biochemistry, Hvidovre University Hospital, Copenhagen, Denmark
| | - Thorkild I. A. Sørensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Sten Madsbad
- Department of Endocrinology, Hvidovre University Hospital, Copenhagen, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
21
|
Capillary Rarefaction in Obesity and Metabolic Diseases-Organ-Specificity and Possible Mechanisms. Cells 2020; 9:cells9122683. [PMID: 33327460 PMCID: PMC7764934 DOI: 10.3390/cells9122683] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/02/2020] [Accepted: 12/10/2020] [Indexed: 12/11/2022] Open
Abstract
Obesity and its comorbidities like diabetes, hypertension and other cardiovascular disorders are the leading causes of death and disability worldwide. Metabolic diseases cause vascular dysfunction and loss of capillaries termed capillary rarefaction. Interestingly, obesity seems to affect capillary beds in an organ-specific manner, causing morphological and functional changes in some tissues but not in others. Accordingly, treatment strategies targeting capillary rarefaction result in distinct outcomes depending on the organ. In recent years, organ-specific vasculature and endothelial heterogeneity have been in the spotlight in the field of vascular biology since specialized vascular systems have been shown to contribute to organ function by secreting varying autocrine and paracrine factors and by providing niches for stem cells. This review summarizes the recent literature covering studies on organ-specific capillary rarefaction observed in obesity and metabolic diseases and explores the underlying mechanisms, with multiple modes of action proposed. It also provides a glimpse of the reported therapeutic perspectives targeting capillary rarefaction. Further studies should address the reasons for such organ-specificity of capillary rarefaction, investigate strategies for its prevention and reversibility and examine potential signaling pathways that can be exploited to target it.
Collapse
|
22
|
Kikuchi R, Tsuboi N, Sada KE, Nakatochi M, Yokoe Y, Suzuki A, Maruyama S, Murohara T, Matsushita T, Amano K, Atsumi T, Takasaki Y, Ito S, Hasegawa H, Dobashi H, Ito T, Makino H, Matsuo S. Vascular endothelial growth factor (VEGF)-A and VEGF-A 165b are associated with time to remission of granulomatosis with polyangiitis in a nationwide Japanese prospective cohort study. Ann Clin Biochem 2020; 58:86-94. [PMID: 33081494 DOI: 10.1177/0004563220968371] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Effective prognostic markers are needed for antineutrophil cytoplasmic antibody-associated vasculitis (AAV). This study evaluated the clinical associations of serum vascular endothelial growth factor-A (sVEGF-A) and sVEGF-A165b (an antiangiogenic isoform of VEGF-A) concentrations with time to remission of AAV in a nationwide Japanese prospective follow-up cohort. METHODS We collected samples from patients with AAV who were enrolled in the nationwide Japanese cohort study (RemIT-JAV-RPGN). We measured sVEGF-A and sVEGF-A165b concentrations using enzyme-linked immunosorbent assays in 57 serum samples collected 6 months before and after initiation of AAV treatment. Patients were classified based on AAV disease subtypes: microscopic polyangiitis, granulomatosis with polyangiitis and eosinophilic granulomatosis with polyangiitis (EGPA). RESULTS Results revealed significant reductions in sVEGF-A and sVEGF-A165b concentrations in patients with microscopic polyangiitis and EGPA, respectively. However, despite the comparable concentrations of sVEGF-A and sVEGF-A165b during the 6 months of treatment in granulomatosis with polyangiitis patients, correlation analysis revealed that the differences in log2-transformed concentrations of sVEGF-A and sVEGF-A165b were inversely correlated with time to remission in granulomatosis with polyangiitis patients. CONCLUSION These results suggest that sVEGF-A and -A165b can serve as potential markers of time to remission in patients with granulomatosis with polyangiitis.
Collapse
Affiliation(s)
- Ryosuke Kikuchi
- Department of Medical Technique, Nagoya University Hospital, Nagoya, Japan
| | - Naotake Tsuboi
- Department of Nephrology, School of Medicine, Fujita Health University, Nagoya, Japan
| | - Ken-Ei Sada
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.,Department of Clinical Epidemiology, Kochi Medical School, Kochi University, Nankoku, Japan
| | - Masahiro Nakatochi
- Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuki Yokoe
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Atsuo Suzuki
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shoichi Maruyama
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tadashi Matsushita
- Department of Clinical Laboratory, Nagoya University Hospital, Nagoya, Japan.,Department of Transfusion Medicine, Nagoya University Hospital, Nagoya, Japan
| | | | - Koichi Amano
- Department of Rheumatology and Clinical Immunology, Saitama Medical Center, Saitama Medical University, Saitama, Japan
| | - Tatsuya Atsumi
- Rheumatology, Endocrinology and Nephrology, Faculty of Medicine, Hokkaido University, Hokkaido, Japan
| | - Yoshinari Takasaki
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| | - Satoshi Ito
- Department of Rheumatology, Niigata Rheumatic Center, Niigata, Japan
| | - Hitoshi Hasegawa
- Department of Hematology, Clinical Immunology, and Infectious Diseases, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Hiroaki Dobashi
- Division of Hematology, Rheumatology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Takafumi Ito
- Division of Nephrology, Shimane University, Faculty of Medicine, Shimane, Japan
| | | | | |
Collapse
|
23
|
Mamer SB, Wittenkeller A, Imoukhuede PI. VEGF-A splice variants bind VEGFRs with differential affinities. Sci Rep 2020; 10:14413. [PMID: 32879419 PMCID: PMC7468149 DOI: 10.1038/s41598-020-71484-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 08/04/2020] [Indexed: 12/29/2022] Open
Abstract
Vascular endothelial growth factor A (VEGF-A) and its binding to VEGFRs is an important angiogenesis regulator, especially the earliest-known isoform, VEGF-A165a. Yet several additional splice variants play prominent roles in regulating angiogenesis in health and in vascular disease, including VEGF-A121 and an anti-angiogenic variant, VEGF-A165b. Few studies have attempted to distinguish these forms from their angiogenic counterparts, experimentally. Previous studies of VEGF-A:VEGFR binding have measured binding kinetics for VEGFA165 and VEGF-A121, but binding kinetics of the other two pro- and all anti-angiogenic splice variants are not known. We measured the binding kinetics for VEGF-A165, -A165b, and -A121 with VEGFR1 and VEGF-R2 using surface plasmon resonance. We validated our methods by reproducing the known affinities between VEGF-A165a:VEGFR1 and VEGF-A165a:VEGFR2, 1.0 pM and 10 pM respectively, and validated the known affinity VEGF-A121:VEGFR2 as KD = 0.66 nM. We found that VEGF-A121 also binds VEGFR1 with an affinity KD = 3.7 nM. We further demonstrated that the anti-angiogenic variant, VEGF-A165b selectively prefers VEGFR2 binding at an affinity = 0.67 pM while binding VEGFR1 with a weaker affinity-KD = 1.4 nM. These results suggest that the - A165b anti-angiogenic variant would preferentially bind VEGFR2. These discoveries offer a new paradigm for understanding VEGF-A, while further stressing the need to take care in differentiating the splice variants in all future VEGF-A studies.
Collapse
Affiliation(s)
- Spencer B Mamer
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| | - Ashley Wittenkeller
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - P I Imoukhuede
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
24
|
Fang Y, Kaszuba T, Imoukhuede PI. Systems Biology Will Direct Vascular-Targeted Therapy for Obesity. Front Physiol 2020; 11:831. [PMID: 32760294 PMCID: PMC7373796 DOI: 10.3389/fphys.2020.00831] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 06/22/2020] [Indexed: 12/12/2022] Open
Abstract
Healthy adipose tissue expansion and metabolism during weight gain require coordinated angiogenesis and lymphangiogenesis. These vascular growth processes rely on the vascular endothelial growth factor (VEGF) family of ligands and receptors (VEGFRs). Several studies have shown that controlling vascular growth by regulating VEGF:VEGFR signaling can be beneficial for treating obesity; however, dysregulated angiogenesis and lymphangiogenesis are associated with several chronic tissue inflammation symptoms, including hypoxia, immune cell accumulation, and fibrosis, leading to obesity-related metabolic disorders. An ideal obesity treatment should minimize adipose tissue expansion and the advent of adverse metabolic consequences, which could be achieved by normalizing VEGF:VEGFR signaling. Toward this goal, a systematic investigation of the interdependency of vascular and metabolic systems in obesity and tools to predict personalized treatment ranges are necessary to improve patient outcomes through vascular-targeted therapies. Systems biology can identify the critical VEGF:VEGFR signaling mechanisms that can be targeted to regress adipose tissue expansion and can predict the metabolic consequences of different vascular-targeted approaches. Establishing a predictive, biologically faithful platform requires appropriate computational models and quantitative tissue-specific data. Here, we discuss the involvement of VEGF:VEGFR signaling in angiogenesis, lymphangiogenesis, adipogenesis, and macrophage specification – key mechanisms that regulate adipose tissue expansion and metabolism. We then provide useful computational approaches for simulating these mechanisms, and detail quantitative techniques for acquiring tissue-specific parameters. Systems biology, through computational models and quantitative data, will enable an accurate representation of obese adipose tissue that can be used to direct the development of vascular-targeted therapies for obesity and associated metabolic disorders.
Collapse
Affiliation(s)
- Yingye Fang
- Imoukhuede Systems Biology Laboratory, Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, United States
| | - Tomasz Kaszuba
- Imoukhuede Systems Biology Laboratory, Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, United States
| | - P I Imoukhuede
- Imoukhuede Systems Biology Laboratory, Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
25
|
Tang X, Miao Y, Luo Y, Sriram K, Qi Z, Lin FM, Gu Y, Lai CH, Hsu CY, Peterson KL, Van Keuren-Jensen K, Fueger PT, Yeo GW, Natarajan R, Zhong S, Chen ZB. Suppression of Endothelial AGO1 Promotes Adipose Tissue Browning and Improves Metabolic Dysfunction. Circulation 2020; 142:365-379. [PMID: 32393053 DOI: 10.1161/circulationaha.119.041231] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Metabolic disorders such as obesity and diabetes mellitus can cause dysfunction of endothelial cells (ECs) and vascular rarefaction in adipose tissues. However, the modulatory role of ECs in adipose tissue function is not fully understood. Other than vascular endothelial growth factor-vascular endothelial growth factor receptor-mediated angiogenic signaling, little is known about the EC-derived signals in adipose tissue regulation. We previously identified Argonaute 1 (AGO1; a key component of microRNA-induced silencing complex) as a crucial regulator in hypoxia-induced angiogenesis. In this study, we intend to determine the AGO1-mediated EC transcriptome, the functional importance of AGO1-regulated endothelial function in vivo, and the relevance to adipose tissue function and obesity. METHODS We generated and subjected mice with EC-AGO1 deletion (EC-AGO1-knockout [KO]) and their wild-type littermates to a fast food-mimicking, high-fat high-sucrose diet and profiled the metabolic phenotypes. We used crosslinking immunoprecipitation- and RNA-sequencing to identify the AGO1-mediated mechanisms underlying the observed metabolic phenotype of EC-AGO1-KO. We further leveraged cell cultures and mouse models to validate the functional importance of the identified molecular pathway, for which the translational relevance was explored using human endothelium isolated from healthy donors and donors with obesity/type 2 diabetes mellitus. RESULTS We identified an antiobesity phenotype of EC-AGO1-KO, evident by lower body weight and body fat, improved insulin sensitivity, and enhanced energy expenditure. At the organ level, we observed the most significant phenotype in the subcutaneous and brown adipose tissues of KO mice, with greater vascularity and enhanced browning and thermogenesis. Mechanistically, EC-AGO1 suppression results in inhibition of thrombospondin-1 (THBS1/TSP1), an antiangiogenic and proinflammatory cytokine that promotes insulin resistance. In EC-AGO1-KO mice, overexpression of TSP1 substantially attenuated the beneficial phenotype. In human endothelium isolated from donors with obesity or type 2 diabetes mellitus, AGO1 and THBS1 are expressed at higher levels than the healthy controls, supporting a pathological role of this pathway. CONCLUSIONS Our study suggests a novel mechanism by which ECs, through the AGO1-TSP1 pathway, control vascularization and function of adipose tissues, insulin sensitivity, and whole-body metabolic state.
Collapse
Affiliation(s)
- Xiaofang Tang
- Department of Diabetes Complications and Metabolism (X.T., Y.M., Y.L., K.S., F.L., C.H.L., R.N., Z.C.), City of Hope, Duarte, CA
| | - Yifei Miao
- Department of Diabetes Complications and Metabolism (X.T., Y.M., Y.L., K.S., F.L., C.H.L., R.N., Z.C.), City of Hope, Duarte, CA
| | - Yingjun Luo
- Department of Diabetes Complications and Metabolism (X.T., Y.M., Y.L., K.S., F.L., C.H.L., R.N., Z.C.), City of Hope, Duarte, CA
| | - Kiran Sriram
- Department of Diabetes Complications and Metabolism (X.T., Y.M., Y.L., K.S., F.L., C.H.L., R.N., Z.C.), City of Hope, Duarte, CA.,Irell and Manella Graduate School of Biological Sciences (K.S., P.T.F., R.N., Z.C.), City of Hope, Duarte, CA
| | - Zhijie Qi
- Department of Bioengineering (Z.Q., S.Z.), University of California at San Diego, La Jolla
| | - Feng-Mao Lin
- Department of Diabetes Complications and Metabolism (X.T., Y.M., Y.L., K.S., F.L., C.H.L., R.N., Z.C.), City of Hope, Duarte, CA
| | - Yusu Gu
- Department of Medicine (Y.G., K.L.P.), University of California at San Diego, La Jolla
| | - Chih-Hung Lai
- Department of Diabetes Complications and Metabolism (X.T., Y.M., Y.L., K.S., F.L., C.H.L., R.N., Z.C.), City of Hope, Duarte, CA
| | - Chien-Yi Hsu
- Department of Internal Medicine, Taipei Medical University Hospital, Taiwan (C.Y.H)
| | - Kirk L Peterson
- Department of Medicine (Y.G., K.L.P.), University of California at San Diego, La Jolla
| | | | - Patrick T Fueger
- Irell and Manella Graduate School of Biological Sciences (K.S., P.T.F., R.N., Z.C.), City of Hope, Duarte, CA.,Department of Molecular and Cellular Endocrinology (P.T.F.), City of Hope, Duarte, CA
| | - Gene W Yeo
- Department of Cellular and Molecular Medicine (G.W.Y.), University of California at San Diego, La Jolla
| | - Rama Natarajan
- Department of Diabetes Complications and Metabolism (X.T., Y.M., Y.L., K.S., F.L., C.H.L., R.N., Z.C.), City of Hope, Duarte, CA.,Irell and Manella Graduate School of Biological Sciences (K.S., P.T.F., R.N., Z.C.), City of Hope, Duarte, CA
| | - Sheng Zhong
- Department of Bioengineering (Z.Q., S.Z.), University of California at San Diego, La Jolla
| | - Zhen Bouman Chen
- Department of Diabetes Complications and Metabolism (X.T., Y.M., Y.L., K.S., F.L., C.H.L., R.N., Z.C.), City of Hope, Duarte, CA.,Irell and Manella Graduate School of Biological Sciences (K.S., P.T.F., R.N., Z.C.), City of Hope, Duarte, CA
| |
Collapse
|
26
|
Vaccarino V, Badimon L, Bremner JD, Cenko E, Cubedo J, Dorobantu M, Duncker DJ, Koller A, Manfrini O, Milicic D, Padro T, Pries AR, Quyyumi AA, Tousoulis D, Trifunovic D, Vasiljevic Z, de Wit C, Bugiardini R. Depression and coronary heart disease: 2018 position paper of the ESC working group on coronary pathophysiology and microcirculation. Eur Heart J 2020; 41:1687-1696. [PMID: 30698764 PMCID: PMC10941327 DOI: 10.1093/eurheartj/ehy913] [Citation(s) in RCA: 229] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 10/25/2018] [Accepted: 01/03/2019] [Indexed: 12/13/2022] Open
Affiliation(s)
- Viola Vaccarino
- Department of Epidemiology, Rollins School of Public Health,
Emory University, 1518 Clifton Road Northeast, Atlanta, GA, 30322, USA
- Department of Medicine (Cardiology), Emory University School of
Medicine, 1518 Clifton Road Northeast, Atlanta, GA, 30322, USA
| | - Lina Badimon
- Cardiovascular Program (ICCC), IR-Hospital de la Santa Creu i Sant Pau.
CiberCV-Institute Carlos III. Autonomous University of Barcelona, C/
Sant Antoni Maria Claret, 167, 08025, Barcelona, Spain
| | - J Douglas Bremner
- Department of Psychiatry and Behavioral Sciences, Emory University School
of Medicine, 12 Executive Park Drive Northeast, Atlanta, GA, 30329,
USA
- Department of Radiology, Emory University School of Medicine,
1364 Clifton Road Northeast, Atlanta, GA, 30322, USA
- Atlanta Veterans Administration Medical Center, 670 Clairmont
Road, Decatur, GA, 30033, USA
| | - Edina Cenko
- Department of Experimental, Diagnostic and Specialty Medicine, University
of Bologna, Via Giuseppe Massarenti 9, 40138, Bologna, Italy
| | - Judit Cubedo
- Cardiovascular Program (ICCC), IR-Hospital de la Santa Creu i Sant Pau.
CiberCV-Institute Carlos III. Autonomous University of Barcelona, C/
Sant Antoni Maria Claret, 167, 08025, Barcelona, Spain
| | - Maria Dorobantu
- Cardiology Department, University of Medicine and Pharmacy ‘Carol Davila’
of Bucharest, Emergency Clinical Hospital of Bucharest, Calea Floreasca
8, Sector 1, Bucuresti, 014461, Romania
| | - Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology,
Thoraxcenter, Cardiovascular Research Institute COEUR, Erasmus MC, University Medical
Center, Rotterdam, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The
Netherlands
| | - Akos Koller
- Institute of Natural Sciences, University of Physical
Education, Alkotas street, 44, 1123, Budapest, Hungary
- Department of Physiology, New York Medical College, Valhalla,
NY, 10595, USA
| | - Olivia Manfrini
- Department of Experimental, Diagnostic and Specialty Medicine, University
of Bologna, Via Giuseppe Massarenti 9, 40138, Bologna, Italy
| | - Davor Milicic
- Department for Cardiovascular Diseases, University Hospital Center Zagreb,
University of Zagreb, Kispaticeva 12, HR-10000, Zagreb, Croatia
| | - Teresa Padro
- Cardiovascular Program (ICCC), IR-Hospital de la Santa Creu i Sant Pau.
CiberCV-Institute Carlos III. Autonomous University of Barcelona, C/
Sant Antoni Maria Claret, 167, 08025, Barcelona, Spain
| | - Axel R Pries
- Department of Physiology, Charitè-University Medicine,
Thielallee 71, D-14195, Berlin, Germany
| | - Arshed A Quyyumi
- Department of Medicine (Cardiology), Emory University School of
Medicine, 1518 Clifton Road Northeast, Atlanta, GA, 30322, USA
| | - Dimitris Tousoulis
- First Department of Cardiology, Hippokration Hospital, University of Athens
Medical School, Vasilissis Sofias 114, TK 115 28, Athens, Greece
| | - Danijela Trifunovic
- Department of Cardiology, University Clinical Center of
Serbia, Pasterova 2, 11000, Belgrade, Serbia
- School of Medicine, University of Belgrade, Dr Subotica 8,
11000, Belgrade, Serbia
| | - Zorana Vasiljevic
- School of Medicine, University of Belgrade, Dr Subotica 8,
11000, Belgrade, Serbia
| | - Cor de Wit
- Institut für Physiologie, Universität zu Lübeck and Deutsches Zentrumfür
Herz-Kreislauf-Forschung (DZHK), Ratzeburger Allee 160, 23538, Lübeck,
Germany
| | - Raffaele Bugiardini
- Department of Experimental, Diagnostic and Specialty Medicine, University
of Bologna, Via Giuseppe Massarenti 9, 40138, Bologna, Italy
| |
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW Obesity is a pandemic, yet preventable healthcare problem. Insulin resistance, diabetes mellitus, dyslipidemia, and cardiovascular complications are core manifestation of obesity. While adipose tissue is a primary site of energy storage, it is also an endocrine organ, secreting a large number of adipokines and cytokines. Nonetheless in obesity, the secretion of cytokines and free fatty acids increases significantly and is associated with the degree of adiposity and insulin resistance. Fat-specific protein 27 (FSP27) has emerged as one of the major proteins that promote physiological storage of fat in adipose tissue. RECENT FINDINGS Review of number of recent findings suggests that FSP27 plays a crucial role in physiological storage of fat within the adipose tissue especially in humans. However, in disease conditions such as obesity, FSP27 may contribute to ectopic fat accumulation in non-adipose tissue. More studies are required to highlight the tissue-specific role of FSP27, especially in humans.
Collapse
Affiliation(s)
- Shakun Karki
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, 88 East Newton St, Boston, MA, 02118, USA.
| |
Collapse
|
28
|
Karki S, Farb MG, Sharma VM, Jash S, Zizza EJ, Hess DT, Carmine B, Carter CO, Pernar LI, Apovian CM, Puri V, Gokce N. Fat-Specific Protein 27 Regulation of Vascular Function in Human Obesity. J Am Heart Assoc 2019; 8:e011431. [PMID: 31433737 PMCID: PMC6585348 DOI: 10.1161/jaha.118.011431] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 04/15/2019] [Indexed: 12/11/2022]
Abstract
Background Pathophysiological mechanisms that connect obesity to cardiovascular disease are incompletely understood. FSP27 (Fat-specific protein 27) is a lipid droplet-associated protein that regulates lipolysis and insulin sensitivity in adipocytes. We unexpectedly discovered extensive FSP27 expression in human endothelial cells that is downregulated in association with visceral obesity. We sought to examine the functional role of FSP27 in the control of vascular phenotype. Methods and Results We biopsied paired subcutaneous and visceral fat depots from 61 obese individuals (body mass index 44±8 kg/m2, age 48±4 years) during planned bariatric surgery. We characterized depot-specific FSP27 expression in relation to adipose tissue microvascular insulin resistance, endothelial function and angiogenesis, and examined differential effects of FSP27 modification on vascular function. We observed markedly reduced vasodilator and angiogenic capacity of microvessels isolated from the visceral compared with subcutaneous adipose depots. Recombinant FSP27 and/or adenoviral FSP27 overexpression in human tissue increased endothelial nitric oxide synthase phosphorylation and nitric oxide production, and rescued vasomotor and angiogenic dysfunction (P<0.05), while siRNA-mediated FSP27 knockdown had opposite effects. Mechanistically, we observed that FSP27 interacts with vascular endothelial growth factor-A and exerts robust regulatory control over its expression. Lastly, in a subset of subjects followed longitudinally for 12±3 months after their bariatric surgery, 30% weight loss improved metabolic parameters and increased angiogenic capacity that correlated positively with increased FSP27 expression (r=0.79, P<0.05). Conclusions Our data strongly support a key role and functional significance of FSP27 as a critical endogenous modulator of human microvascular function that has not been previously described. FSP27 may serve as a previously unrecognized regulator of arteriolar vasomotor capacity and angiogenesis which are pivotal in the pathogenesis of cardiometabolic diseases linked to obesity.
Collapse
Affiliation(s)
- Shakun Karki
- Evans Department of Medicine and Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMA
| | - Melissa G. Farb
- Evans Department of Medicine and Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMA
| | - Vishva M. Sharma
- Department of Biomedical Sciences and Diabetes InstituteOhio UniversityAthensOH
| | - Sukanta Jash
- Department of Biomedical Sciences and Diabetes InstituteOhio UniversityAthensOH
| | - Elaina J. Zizza
- Evans Department of Medicine and Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMA
| | - Donald T. Hess
- Department of General SurgeryBoston University School of MedicineBostonMA
| | - Brian Carmine
- Department of General SurgeryBoston University School of MedicineBostonMA
| | - Cullen O. Carter
- Department of General SurgeryBoston University School of MedicineBostonMA
| | - Luise I. Pernar
- Department of General SurgeryBoston University School of MedicineBostonMA
| | - Caroline M. Apovian
- Evans Department of Medicine and Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMA
| | - Vishwajeet Puri
- Department of Biomedical Sciences and Diabetes InstituteOhio UniversityAthensOH
| | - Noyan Gokce
- Evans Department of Medicine and Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMA
| |
Collapse
|
29
|
Modulation of Receptor Tyrosine Kinase Activity through Alternative Splicing of Ligands and Receptors in the VEGF-A/VEGFR Axis. Cells 2019; 8:cells8040288. [PMID: 30925751 PMCID: PMC6523102 DOI: 10.3390/cells8040288] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 03/19/2019] [Accepted: 03/22/2019] [Indexed: 12/14/2022] Open
Abstract
Vascular endothelial growth factor A (VEGF-A) signaling is essential for physiological and pathological angiogenesis. Alternative splicing of the VEGF-A pre-mRNA gives rise to a pro-angiogenic family of isoforms with a differing number of amino acids (VEGF-Axxxa), as well as a family of isoforms with anti-angiogenic properties (VEGF-Axxxb). The biological functions of VEGF-A proteins are mediated by a family of cognate protein tyrosine kinase receptors, known as the VEGF receptors (VEGFRs). VEGF-A binds to both VEGFR-1, largely suggested to function as a decoy receptor, and VEGFR-2, the predominant signaling receptor. Both VEGFR-1 and VEGFR-2 can also be alternatively spliced to generate soluble isoforms (sVEGFR-1/sVEGFR-2). The disruption of the splicing of just one of these genes can result in changes to the entire VEGF-A/VEGFR signaling axis, such as the increase in VEGF-A165a relative to VEGF-A165b resulting in increased VEGFR-2 signaling and aberrant angiogenesis in cancer. Research into this signaling axis has recently focused on manipulating the splicing of these genes as a potential therapeutic avenue in disease. Therefore, further research into understanding the mechanisms by which the splicing of VEGF-A/VEGFR-1/VEGFR-2 is regulated will help in the development of drugs aimed at manipulating splicing or inhibiting specific splice isoforms in a therapeutic manner.
Collapse
|
30
|
Ngo DTM, Sverdlov AL, Karki S, Macartney-Coxson D, Stubbs RS, Farb MG, Carmine B, Hess DT, Colucci WS, Gokce N. Oxidative modifications of mitochondrial complex II are associated with insulin resistance of visceral fat in obesity. Am J Physiol Endocrinol Metab 2019; 316:E168-E177. [PMID: 30576243 PMCID: PMC6397365 DOI: 10.1152/ajpendo.00227.2018] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Obesity, particularly visceral adiposity, has been linked to mitochondrial dysfunction and increased oxidative stress, which have been suggested as mechanisms of insulin resistance. The mechanism(s) behind this remains incompletely understood. In this study, we hypothesized that mitochondrial complex II dysfunction plays a role in impaired insulin sensitivity in visceral adipose tissue of subjects with obesity. We obtained subcutaneous and visceral adipose tissue biopsies from 43 subjects with obesity (body mass index ≥ 30 kg/m2) during planned bariatric surgery. Compared with subcutaneous adipose tissue, visceral adipose tissue exhibited decreased complex II activity, which was restored with the reducing agent dithiothreitol (5 mM) ( P < 0.01). A biotin switch assay identified that cysteine oxidative posttranslational modifications (OPTM) in complex II subunit A (succinate dehydrogenase A) were increased in visceral vs. subcutaneous fat ( P < 0.05). Insulin treatment (100 nM) stimulated complex II activity in subcutaneous fat ( P < 0.05). In contrast, insulin treatment of visceral fat led to a decrease in complex II activity ( P < 0.01), which was restored with addition of the mitochondria-specific oxidant scavenger mito-TEMPO (10 µM). In a cohort of 10 subjects with severe obesity, surgical weight loss decreased OPTM and restored complex II activity, exclusively in the visceral depot. Mitochondrial complex II may be an unrecognized and novel mediator of insulin resistance associated with visceral adiposity. The activity of complex II is improved by weight loss, which may contribute to metabolic improvements associated with bariatric surgery.
Collapse
Affiliation(s)
- Doan T M Ngo
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine , Boston, Massachusetts
- School of Biomedical Sciences and Pharmacy, University of Newcastle , Newcastle, New South Wales , Australia
| | - Aaron L Sverdlov
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine , Boston, Massachusetts
- School of Medicine and Public Health, University of Newcastle , Newcastle, New South Wales , Australia
| | - Shakun Karki
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine , Boston, Massachusetts
| | - Donia Macartney-Coxson
- Biomarkers Group, Institute of Environmental Science and Research , Wellington , New Zealand
| | | | - Melissa G Farb
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine , Boston, Massachusetts
| | - Brian Carmine
- Department of General Surgery, Boston University School of Medicine , Boston, Massachusetts
| | - Donald T Hess
- Department of General Surgery, Boston University School of Medicine , Boston, Massachusetts
| | - Wilson S Colucci
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine , Boston, Massachusetts
| | - Noyan Gokce
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine , Boston, Massachusetts
| |
Collapse
|
31
|
Ganta VC, Choi M, Farber CR, Annex BH. Antiangiogenic VEGF 165b Regulates Macrophage Polarization via S100A8/S100A9 in Peripheral Artery Disease. Circulation 2019; 139:226-242. [PMID: 30586702 PMCID: PMC6322929 DOI: 10.1161/circulationaha.118.034165] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 08/21/2018] [Indexed: 11/16/2022]
Abstract
BACKGROUND Atherosclerotic occlusions decrease blood flow to the lower limbs, causing ischemia and tissue loss in patients with peripheral artery disease (PAD). No effective medical therapies are currently available to induce angiogenesis and promote perfusion recovery in patients with severe PAD. Clinical trials aimed at inducing vascular endothelial growth factor (VEGF)-A levels, a potent proangiogenic growth factor to induce angiogenesis, and perfusion recovery were not successful. Alternate splicing in the exon-8 of VEGF-A results in the formation of VEGFxxxa (VEGF165a) and VEGFxxxb (VEGF165b) isoforms with existing literature focusing on VEGF165b's role in inhibiting vascular endothelial growth factor receptor 2-dependent angiogenesis. However, we have recently shown that VEGF165b blocks VEGF-A-induced endothelial vascular endothelial growth factor receptor 1 (VEGFR1) activation in ischemic muscle to impair perfusion recovery. Because macrophage-secreted VEGF165b has been shown to decrease angiogenesis in peripheral artery disease, and macrophages were well known to play important roles in regulating ischemic muscle vascular remodeling, we examined the role of VEGF165b in regulating macrophage function in PAD. METHODS Femoral artery ligation and resection were used as an in vivo preclinical PAD model, and hypoxia serum starvation was used as an in vitro model for PAD. Experiments including laser-Doppler perfusion imaging, adoptive cell transfer to ischemic muscle, immunoblot analysis, ELISAs, immunostainings, flow cytometry, quantitative polymerase chain reaction analysis, and RNA sequencing were performed to determine a role of VEGF165b in regulating macrophage phenotype and function in PAD. RESULTS First, we found increased VEGF165b expression with increased M1-like macrophages in PAD versus non-PAD (controls) muscle biopsies. Next, using in vitro hypoxia serum starvation, in vivo pre clinical PAD models, and adoptive transfer of VEGF165b-expressing bone marrow-derived macrophages or VEGFR1+/- bone marrow-derived macrophages (M1-like phenotype), we demonstrate that VEGF165b inhibits VEGFR1 activation to induce an M1-like phenotype that impairs ischemic muscle neovascularization. Subsequently, we found S100A8/S100A9 as VEGFR1 downstream regulators of macrophage polarization by RNA-Seq analysis of hypoxia serum starvation-VEGFR1+/+ versus hypoxia serum starvation-VEGFR1+/- bone marrow-derived macrophages. CONCLUSIONS In our current study, we demonstrate that increased VEGF165b expression in macrophages induces an antiangiogenic M1-like phenotype that directly impairs angiogenesis. VEGFR1 inhibition by VEGF165b results in S100A8/S100A9-mediated calcium influx to induce an M1-like phenotype that impairs ischemic muscle revascularization and perfusion recovery.
Collapse
Affiliation(s)
- Vijay Chaitanya Ganta
- Robert M Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA
- Division Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, VA
| | - Min Choi
- Robert M Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA
| | - Charles R. Farber
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA
| | - Brian H. Annex
- Robert M Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA
- Division Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, VA
| |
Collapse
|
32
|
Hammarstedt A, Gogg S, Hedjazifar S, Nerstedt A, Smith U. Impaired Adipogenesis and Dysfunctional Adipose Tissue in Human Hypertrophic Obesity. Physiol Rev 2019; 98:1911-1941. [PMID: 30067159 DOI: 10.1152/physrev.00034.2017] [Citation(s) in RCA: 302] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The subcutaneous adipose tissue (SAT) is the largest and best storage site for excess lipids. However, it has a limited ability to expand by recruiting and/or differentiating available precursor cells. When inadequate, this leads to a hypertrophic expansion of the cells with increased inflammation, insulin resistance, and a dysfunctional prolipolytic tissue. Epi-/genetic factors regulate SAT adipogenesis and genetic predisposition for type 2 diabetes is associated with markers of an impaired SAT adipogenesis and development of hypertrophic obesity also in nonobese individuals. We here review mechanisms for the adipose precursor cells to enter adipogenesis, emphasizing the role of bone morphogenetic protein-4 (BMP-4) and its endogenous antagonist gremlin-1, which is increased in hypertrophic SAT in humans. Gremlin-1 is a secreted and a likely important mechanism for the impaired SAT adipogenesis in hypertrophic obesity. Transiently increasing BMP-4 enhances adipogenic commitment of the precursor cells while maintained BMP-4 signaling during differentiation induces a beige/brown oxidative phenotype in both human and murine adipose cells. Adipose tissue growth and development also requires increased angiogenesis, and BMP-4, as a proangiogenic molecule, may also be an important feedback regulator of this. Hypertrophic obesity is also associated with increased lipolysis. Reduced lipid storage and increased release of FFA by hypertrophic SAT are important mechanisms for the accumulation of ectopic fat in the liver and other places promoting insulin resistance. Taken together, the limited expansion and storage capacity of SAT is a major driver of the obesity-associated metabolic complications.
Collapse
Affiliation(s)
- Ann Hammarstedt
- Department of Molecular and Clinical Medicine, The Lundberg Laboratory for Diabetes Research, the Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - Silvia Gogg
- Department of Molecular and Clinical Medicine, The Lundberg Laboratory for Diabetes Research, the Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - Shahram Hedjazifar
- Department of Molecular and Clinical Medicine, The Lundberg Laboratory for Diabetes Research, the Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - Annika Nerstedt
- Department of Molecular and Clinical Medicine, The Lundberg Laboratory for Diabetes Research, the Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - Ulf Smith
- Department of Molecular and Clinical Medicine, The Lundberg Laboratory for Diabetes Research, the Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| |
Collapse
|
33
|
Kikuchi R, Stevens M, Harada K, Oltean S, Murohara T. Anti-angiogenic isoform of vascular endothelial growth factor-A in cardiovascular and renal disease. Adv Clin Chem 2019; 88:1-33. [PMID: 30612603 DOI: 10.1016/bs.acc.2018.10.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Accumulating evidence suggests that pathologic interactions between the heart and the kidney can contribute to the progressive dysfunction of both organs. Recently, there has been an increase in the prevalence of cardiovascular disease (CVD) and chronic kidney disease (CKD) due to increasing obesity rates. It has been reported that obesity causes various heart and renal disorders and appears to accelerate their progression. Vascular endothelial growth factor-A (VEGF-A) is a major regulator of angiogenesis and vessel permeability, and is associated with CVD and CKD. It is now recognized that alternative VEGF-A gene splicing generates VEGF-A isoforms that differ in their biological actions. Proximal splicing that includes an exon 8a sequence results in pro-angiogenic VEGF-A165a, whereas distal splicing inclusive of exon 8b yields the anti-angiogenic isoform of VEGF-A (VEGF-A165b). This review highlights several recent preclinical and clinical studies on the role of VEGF-A165b in CVD and CKD as a novel function of VEGF-A. This review also discusses potential therapeutic approaches of the use of VEGF-A in clinical settings as a potential circulating biomarker for CVD and CKD.
Collapse
Affiliation(s)
- Ryosuke Kikuchi
- Department of Medical Technique, Nagoya University Hospital, Nagoya, Japan.
| | - Megan Stevens
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, United Kingdom
| | - Kazuhiro Harada
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Sebastian Oltean
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, United Kingdom
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
34
|
Szulińska M, Łoniewski I, Skrypnik K, Sobieska M, Korybalska K, Suliburska J, Bogdański P. Multispecies Probiotic Supplementation Favorably Affects Vascular Function and Reduces Arterial Stiffness in Obese Postmenopausal Women-A 12-Week Placebo-Controlled and Randomized Clinical Study. Nutrients 2018; 10:E1672. [PMID: 30400570 PMCID: PMC6265939 DOI: 10.3390/nu10111672] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 10/19/2018] [Accepted: 10/29/2018] [Indexed: 12/22/2022] Open
Abstract
Obesity in the postmenopausal period is associated with an increased risk of cardiovascular diseases in women. One of the key drivers of cardiovascular risk is endothelial dysfunction; thus, this is also a crucial point for studies on new therapeutic methods of cardioprotective properties. The aim of the current study was to evaluate the effect of two doses of multispecies probiotic Ecologic® Barrier supplement on functional (primary endpoint) and biochemical parameters (secondary endpoint) of endothelial dysfunction in obese postmenopausal women in a 12-week randomized, placebo-controlled clinical trial. A total of 81 obese Caucasian women participated in the trial. The subjects were randomly assigned to three groups that received a placebo, a low dose (LD) (2.5 × 10⁸ colony forming units (CFU) per day), or a high dose (HD) (1 × 1010 CFU per day) of lyophilisate powder containing live multispecies probiotic bacteria. The probiotic supplement was administered each day for 12 weeks in two equal portions. A high dose probiotic supplementation for 12 weeks decreased systolic blood pressure, vascular endothelial growth factor, pulse wave analysis systolic pressure, pulse wave analysis pulse pressure, pulse wave analysis augmentation index, pulse wave velocity, interleukin-6, tumor necrosis factor alpha, and thrombomodulin. Low doses of probiotic supplementation decreased the systolic blood pressure and interleukin-6 levels. The mean changes in the estimated parameters, compared among the three groups, revealed significant differences in the vascular endothelial growth factor, the pulse wave analysis systolic pressure, the pulse wave analysis augmentation index, the pulse wave velocity, the tumor necrosis factor alpha, and thrombomodulin. The post hoc tests showed significant differences for all parameters between HD and the placebo group, and HD and LD (besides pulse wave analysis augmentation index). We show for the first time that supplementation with multispecies probiotic Ecologic® Barrier favorably modifies both functional and biochemical markers of vascular dysfunction in obese postmenopausal women.
Collapse
Affiliation(s)
- Monika Szulińska
- Department of Treatment of Obesity, Metabolic Disorders and Clinical Dietetics, University of Medical Sciences in Poznań, Szamarzewskiego Str. 84, 60-569 Poznań, Poland.
| | - Igor Łoniewski
- Department of Biochemistry and Human Nutrition, Pomeranian Medical University in Szczecin, Broniewskiego 24, 71-460 Szczecin, Poland.
| | - Katarzyna Skrypnik
- Institute of Human Nutrition and Dietetics, Poznan University of Life Sciences, Wojska Polskiego St. 31, 60-624 Poznań, Poland.
| | - Magdalena Sobieska
- Department of Rheumatology and Rehabilitation, Poznan University of Medical Sciences, 28. Czerwca 1956r 135/147, 61-55 Poznań, Poland.
| | - Katarzyna Korybalska
- Department of Pathophysiology, Poznan University of Medical Sciences, Rokietnicka 8, 60-806 Poznan, Poland.
| | - Joanna Suliburska
- Institute of Human Nutrition and Dietetics, Poznan University of Life Sciences, Wojska Polskiego St. 31, 60-624 Poznań, Poland.
| | - Paweł Bogdański
- Department of Treatment of Obesity, Metabolic Disorders and Clinical Dietetics, University of Medical Sciences in Poznań, Szamarzewskiego Str. 84, 60-569 Poznań, Poland.
| |
Collapse
|
35
|
Ved N, Da Vitoria Lobo ME, Bestall SM, L Vidueira C, Beazley-Long N, Ballmer-Hofer K, Hirashima M, Bates DO, Donaldson LF, Hulse RP. Diabetes-induced microvascular complications at the level of the spinal cord: a contributing factor in diabetic neuropathic pain. J Physiol 2018; 596:3675-3693. [PMID: 29774557 PMCID: PMC6092307 DOI: 10.1113/jp275067] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 05/08/2018] [Indexed: 02/06/2023] Open
Abstract
KEY POINTS Diabetes is thought to induce neuropathic pain through activation of dorsal horn sensory neurons in the spinal cord. Here we explore the impact of hyperglycaemia on the blood supply supporting the spinal cord and chronic pain development. In streptozotocin-induced diabetic rats, neuropathic pain is accompanied by a decline in microvascular integrity in the dorsal horn. Hyperglycaemia-induced degeneration of the endothelium in the dorsal horn was associated with a loss in vascular endothelial growth factor (VEGF)-A165 b expression. VEGF-A165 b treatment prevented diabetic neuropathic pain and degeneration of the endothelium in the spinal cord. Using an endothelial-specific VEGFR2 knockout transgenic mouse model, the loss of endothelial VEGFR2 signalling led to a decline in vascular integrity in the dorsal horn and the development of hyperalgesia in VEGFR2 knockout mice. This highlights that vascular degeneration in the spinal cord could be a previously unidentified factor in the development of diabetic neuropathic pain. ABSTRACT Abnormalities of neurovascular interactions within the CNS of diabetic patients is associated with the onset of many neurological disease states. However, to date, the link between the neurovascular network within the spinal cord and regulation of nociception has not been investigated despite neuropathic pain being common in diabetes. We hypothesised that hyperglycaemia-induced endothelial degeneration in the spinal cord, due to suppression of vascular endothelial growth factor (VEGF)-A/VEGFR2 signalling, induces diabetic neuropathic pain. Nociceptive pain behaviour was investigated in a chemically induced model of type 1 diabetes (streptozotocin induced, insulin supplemented; either vehicle or VEGF-A165 b treated) and an inducible endothelial knockdown of VEGFR2 (tamoxifen induced). Diabetic animals developed mechanical allodynia and heat hyperalgesia. This was associated with a reduction in the number of blood vessels and reduction in Evans blue extravasation in the lumbar spinal cord of diabetic animals versus age-matched controls. Endothelial markers occludin, CD31 and VE-cadherin were downregulated in the spinal cord of the diabetic group versus controls, and there was a concurrent reduction of VEGF-A165 b expression. In diabetic animals, VEGF-A165 b treatment (biweekly i.p., 20 ng g-1 ) restored normal Evans blue extravasation and prevented vascular degeneration, diabetes-induced central neuron activation and neuropathic pain. Inducible knockdown of VEGFR2 (tamoxifen treated Tie2CreERT2 -vegfr2flfl mice) led to a reduction in blood vessel network volume in the lumbar spinal cord and development of heat hyperalgesia. These findings indicate that hyperglycaemia leads to a reduction in the VEGF-A/VEGFR2 signalling cascade, resulting in endothelial dysfunction in the spinal cord, which could be an undiscovered contributing factor to diabetic neuropathic pain.
Collapse
Affiliation(s)
- N Ved
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine University of Nottingham, Nottingham, NG7 2UH, UK.,Institute of Ophthalmology, 11-43 Bath St, London, EC1V 9EL, UK.,Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - M E Da Vitoria Lobo
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine University of Nottingham, Nottingham, NG7 2UH, UK
| | - S M Bestall
- Arthritis Research UK Pain Centre and School of Life Sciences, The Medical School QMC, University of Nottingham, Nottingham, NG7 2UH, UK
| | - C L Vidueira
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine University of Nottingham, Nottingham, NG7 2UH, UK
| | - N Beazley-Long
- Arthritis Research UK Pain Centre and School of Life Sciences, The Medical School QMC, University of Nottingham, Nottingham, NG7 2UH, UK
| | | | - M Hirashima
- Division of Vascular Biology, Kobe University, Japan
| | - D O Bates
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine University of Nottingham, Nottingham, NG7 2UH, UK.,Centre of Membrane and Protein and Receptors (COMPARE), University of Birmingham, Birmingham and University of Nottingham, Nottingham, UK
| | - L F Donaldson
- Institute of Ophthalmology, 11-43 Bath St, London, EC1V 9EL, UK
| | - R P Hulse
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine University of Nottingham, Nottingham, NG7 2UH, UK.,School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, UK
| |
Collapse
|
36
|
Cheung CY, Anderson DF, Brace RA. Transport-associated pathway responses in ovine fetal membranes to changes in amniotic fluid dynamics. Physiol Rep 2018; 5:5/20/e13455. [PMID: 29051303 PMCID: PMC5661228 DOI: 10.14814/phy2.13455] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 08/25/2017] [Accepted: 08/29/2017] [Indexed: 12/12/2022] Open
Abstract
Current evidence suggests that amniotic fluid volume (AFV) is actively regulated by vesicular transport of amniotic fluid outward across the amnion and into the underlying fetal vasculature in the placenta. Our objective was to determine whether gene expression profiles of potential stimulators, inhibitors, and mediators of vesicular transport are altered in response to changes in intramembranous absorption (IMA) rate. Samples of ovine amnion and chorion were obtained from fetal sheep with normal, experimentally reduced or increased AFVs and IMA rates. Amnion and chorion levels of target mRNAs were determined by RT‐qPCR. In the amnion, caveolin‐1 and flotillin‐1 mRNA levels were unchanged during alterations in IMA rate. However, levels of both were significantly higher in amnion than in chorion. Tubulin‐α mRNA levels in the amnion but not in chorion were reduced when IMA rate decreased, and amnion levels correlated positively with IMA rate (P < 0.05). Dynamin‐2 mRNA levels were not altered by experimental conditions. Vascular endothelial growth factor (VEGF164 and VEGF164b) mRNA levels increased during both increases and decreases in IMA rate, whereas soluble Flt‐1 levels did not change. Neither HIF‐1α nor PBEF mRNA levels in the amnion were correlated with VEGF164 expression levels and were not related to IMA rate. Collectively, our findings suggest that changes in amnion microtubule expression may be important in the regulation of transcellular vesicular transport of amniotic fluid and thus modulate IMA rate. Further, our results are consistent with the concept that the amnion is the rate‐limiting layer for amniotic fluid transport.
Collapse
Affiliation(s)
- Cecilia Y Cheung
- Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, Oregon .,Center for Developmental Health, Oregon Health and Science University, Portland, Oregon
| | - Debra F Anderson
- Center for Developmental Health, Oregon Health and Science University, Portland, Oregon
| | - Robert A Brace
- Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, Oregon.,Center for Developmental Health, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
37
|
Hueso L, Ortega R, Selles F, Wu-Xiong NY, Ortega J, Civera M, Ascaso JF, Sanz MJ, Real JT, Piqueras L. Upregulation of angiostatic chemokines IP-10/CXCL10 and I-TAC/CXCL11 in human obesity and their implication for adipose tissue angiogenesis. Int J Obes (Lond) 2018; 42:1406-1417. [PMID: 29795466 DOI: 10.1038/s41366-018-0102-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 04/04/2018] [Accepted: 04/10/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND/AIMS Impaired angiogenesis is linked to adipose tissue (AT) dysfunction, inflammation, and insulin resistance in human obesity. Chemokine (C-X-C motif) receptor. (CXCR3) ligands are important regulators of angiogenesis in different disease contexts such as cancer; however, their role in human morbid obesity is unknown. We investigated the role of the CXCR3 axis in AT angiogenesis in morbidly obese patients. SUBJECTS/METHODS The study group comprised 50 morbidly obese patients (mean age 44 ± 1 years, body mass index 44 ± 1 kg/m2) who had undergone laparoscopic Roux-Y-gastric bypass surgery, and 25 age-matched non-obese control subjects. We measured the circulating levels of the CXCR3 ligands monokine induced by interferon-γ (MIG/CXCL9), interferon-γ inducible protein 10 (IP-10/CXCL10), and interferon-γ-inducible T-cell alpha chemoattractant (I-TAC/CXCL11) in all studied subjects. Additionally, the expression of CXCR3 ligands was analyzed in paired biopsies of subcutaneous and visceral AT obtained during the laparoscopic procedure in morbidly obese patients. Additionally, we explored the functional role of CXCR3 ligands on angiogenesis in AT from morbidly obese patients using an ex vivo assay. RESULTS Plasma levels of CXCL10 and CXCL11 were significantly higher in morbidly obese patients than in controls (p < 0.01). In ex vivo assays, angiogenic growth was markedly lower in visceral AT than in subcutaneous AT (p < 0.05), which was related to significant tissue upregulation of CXCL10, CXCL11 and CXCR3 (p < 0.05). CXCL10 or CXCL11 inhibited AT angiogenesis (p < 0.05), and blockade of CXCR3 function significantly increased capillary sprouting in visceral fat deposits (p < 0.05). Western blot analysis showed that the p38 mitogen-activated protein kinase signaling pathway was implicated in the angiostatic effects of CXCR3 in AT. CONCLUSIONS CXCL10 and CXCL11 may play. deleterious role in obesity as potential inhibitors of AT angiogenesis. Accordingly, pharmacological blockade of CXCR3 could represent. therapy to prevent AT dysfunction in obesity.
Collapse
Affiliation(s)
- Luisa Hueso
- Institute of Health Research-INCLIVA, Valencia, Spain
| | - Rebeca Ortega
- Institute of Health Research-INCLIVA, Valencia, Spain.,Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | | | - Ning Yun Wu-Xiong
- Institute of Health Research-INCLIVA, Valencia, Spain.,Endocrinology and Nutrition Service, University Clinic Hospital of Valencia, Valencia, Spain
| | - Joaquin Ortega
- Surgery Service, University Clinic Hospital of Valencia, Valencia, Spain.,Department of Surgery, University of Valencia, Valencia, Spain
| | - Miguel Civera
- Institute of Health Research-INCLIVA, Valencia, Spain.,Endocrinology and Nutrition Service, University Clinic Hospital of Valencia, Valencia, Spain
| | - Juan F Ascaso
- Institute of Health Research-INCLIVA, Valencia, Spain.,Endocrinology and Nutrition Service, University Clinic Hospital of Valencia, Valencia, Spain.,CIBERDEM: Diabetes and Associated Metabolic Diseases Networking Biomedical Research- ISCIII, Madrid, Spain
| | - Maria-Jesus Sanz
- Institute of Health Research-INCLIVA, Valencia, Spain.,Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - José T Real
- Institute of Health Research-INCLIVA, Valencia, Spain. .,Endocrinology and Nutrition Service, University Clinic Hospital of Valencia, Valencia, Spain. .,CIBERDEM: Diabetes and Associated Metabolic Diseases Networking Biomedical Research- ISCIII, Madrid, Spain.
| | - Laura Piqueras
- Institute of Health Research-INCLIVA, Valencia, Spain. .,Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain.
| |
Collapse
|
38
|
Clegg LE, Mac Gabhann F. A computational analysis of pro-angiogenic therapies for peripheral artery disease. Integr Biol (Camb) 2018; 10:18-33. [PMID: 29327758 PMCID: PMC7017937 DOI: 10.1039/c7ib00218a] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Inducing therapeutic angiogenesis to effectively form hierarchical, non-leaky networks of perfused vessels in tissue engineering applications and ischemic disease remains an unmet challenge, despite extensive research and multiple clinical trials. Here, we use a previously-developed, multi-scale, computational systems pharmacology model of human peripheral artery disease to screen a diverse array of promising pro-angiogenic strategies, including gene therapy, biomaterials, and antibodies. Our previously-validated model explicitly accounts for VEGF immobilization, Neuropilin-1 binding, and weak activation of VEGF receptor 2 (VEGFR2) by the "VEGFxxxb" isoforms. First, we examine biomaterial-based delivery of VEGF engineered for increased affinity to the extracellular matrix. We show that these constructs maintain VEGF close to physiological levels and extend the duration of VEGFR2 activation. We demonstrate the importance of sub-saturating VEGF dosing to prevent angioma formation. Second, we examine the potential of ligand- or receptor-based gene therapy to normalize VEGF receptor signaling. Third, we explore the potential for antibody-based pro-angiogenic therapy. Our model supports recent observations that improvement in perfusion following treatment with anti-VEGF165b in mice is mediated by VEGF-receptor 1, not VEGFR2. Surprisingly, the model predicts that the approved anti-VEGF cancer drug, bevacizumab, may actually improve signaling of both VEGFR1 and VEGFR2 via a novel 'antibody swapping' effect that we demonstrate here. Altogether, this model provides insight into the mechanisms of action of several classes of pro-angiogenic strategies within the context of the complex molecular and physiological processes occurring in vivo. We identify molecular signaling similarities between promising approaches and key differences between promising and ineffective strategies.
Collapse
Affiliation(s)
- Lindsay E Clegg
- Institute for Computational Medicine, Institute for NanoBioTechnology, and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
| | | |
Collapse
|
39
|
Shibata Y, Kikuchi R, Ishii H, Suzuki S, Harada K, Hirayama K, Suzuki A, Tatami Y, Kondo K, Murohara T. Balance between angiogenic and anti-angiogenic isoforms of VEGF-A is associated with the complexity and severity of coronary artery disease. Clin Chim Acta 2017; 478:114-119. [PMID: 29289620 DOI: 10.1016/j.cca.2017.12.042] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 12/26/2017] [Accepted: 12/27/2017] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Assessing the complexity of coronary artery disease (CAD) is clinically important. Vascular endothelial growth factor A (VEGF-A) is a powerful and the most important regulator of angiogenesis. It has been reported that the anti-angiogenic isoform of VEGF-A (VEGF-A165b) inhibits angiogenesis. The purpose of this study was to evaluate the relationship between the complexities of CAD using the Syntax score (SS) and the levels of circulating total VEGF-A or VEGF-A165b. METHODS A total of 268 patients who underwent percutaneous coronary intervention (PCI) were enrolled. Of these, 73 patients without acute coronary syndrome or previous revascularization were included in this study. These subjects were divided into two groups according to the SS. Circulating total VEGF-A and VEGF-A165b levels were measured using an enzyme-linked immunosorbent assay. RESULTS Circulating levels of total VEGF-A were significantly higher in the SS>22 (high SS) group than in the SS≤22 (low SS) group (p=0.018). Moreover, the ratio of VEGF-A165b to total VEGF-A was significantly lower for the high SS group (p=0.004). The levels of total VEGF-A independently predicted high SS after adjusting for other significant variables among patients who underwent PCI (odds ratio, 1.004; 95% CI, 1.001 to 1.006; p=0.01). CONCLUSIONS These data indicated that high SS was associated with circulating levels of total VEGF-A and the ratio of VEGF-A165b to total VEGF-A in patients with complex CAD.
Collapse
Affiliation(s)
- Yohei Shibata
- Department of Cardiology, Nagoya University Graduate School of Medicine, Japan
| | - Ryosuke Kikuchi
- Department of Medical Technique, Nagoya University Hospital, Japan.
| | - Hideki Ishii
- Department of Cardiology, Nagoya University Graduate School of Medicine, Japan
| | - Susumu Suzuki
- Department of Cardiology, Nagoya University Graduate School of Medicine, Japan
| | - Kazuhiro Harada
- Department of Cardiology, Nagoya University Graduate School of Medicine, Japan
| | - Kenshi Hirayama
- Department of Cardiology, Nagoya University Graduate School of Medicine, Japan
| | - Atsuo Suzuki
- Department of Medical Technique, Nagoya University Hospital, Japan
| | - Yosuke Tatami
- Department of Cardiology, Nagoya University Graduate School of Medicine, Japan
| | - Kazuhisa Kondo
- Department of Cardiology, Nagoya University Graduate School of Medicine, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Japan.
| |
Collapse
|
40
|
Afsar B, Afsar RE, Dagel T, Kaya E, Erus S, Ortiz A, Covic A, Kanbay M. Capillary rarefaction from the kidney point of view. Clin Kidney J 2017; 11:295-301. [PMID: 29988260 PMCID: PMC6007395 DOI: 10.1093/ckj/sfx133] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 10/04/2017] [Indexed: 12/18/2022] Open
Abstract
Capillary rarefaction is broadly defined as a reduction in vascular density. Capillary rarefaction in the kidneys is thought to promote hypoxia, impair hemodynamic responses and predispose to chronic kidney disease (CKD) progression and hypertension development. Various mechanisms have been suggested to play a role in the development of capillary rarefaction, including inflammation, an altered endothelial-tubular epithelial cell crosstalk, a relative deficiency in angiogenic growth factors, loss of pericytes, increased activity of Transforming growth factor -β1 and thrombospondin-1, vitamin D deficiency, a link to lymphatic neoangiogenesis and INK4a/ARF (Cylin-dependent kinase inhibitor 2a; CDKN2A). In this review, we summarize the tools available to monitor capillary rarefaction noninvasively in the clinic, the contribution of capillary rarefaction to CKD and hypertension, the known mechanisms of capillary rarefaction, and potential future strategies to attenuate capillary rarefaction and reduce its negative impact. Therapeutic strategies to be explored in more detail include optimization of antihypertensive therapy, vitamin D receptor activators, sirtuin 1 activators, Hypoxia inducible factor prolyl hydroxylase inhibitors and stem cell therapy.
Collapse
Affiliation(s)
- Baris Afsar
- Division of Nephrology, Department of Internal Medicine, Suleyman Demirel University School of Medicine, Isparta, Turkey
| | - Rengin E Afsar
- Division of Nephrology, Department of Internal Medicine, Suleyman Demirel University School of Medicine, Isparta, Turkey
| | - Tuncay Dagel
- Department of Nephrology, Koc University Hospital, Istanbul, Turkey
| | - Ege Kaya
- Koc University School of Medicine, Istanbul, Turkey
| | - Suat Erus
- Department of Thoracic Surgery, Koc University Hospital, Istanbul, Turkey
| | - Alberto Ortiz
- Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Fundación Renal Iñigo Alvarez de Toledo, Madrid, Spain
| | - Adrian Covic
- Department of Nephrology, Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania
| | - Mehmet Kanbay
- Division of Nephrology, Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| |
Collapse
|
41
|
Clegg LE, Ganta VC, Annex BH, Mac Gabhann F. Systems Pharmacology of VEGF165b in Peripheral Artery Disease. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2017; 6:833-844. [PMID: 29193887 PMCID: PMC5744173 DOI: 10.1002/psp4.12261] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 10/12/2017] [Accepted: 10/13/2017] [Indexed: 01/13/2023]
Abstract
We built a whole‐body computational model to study the role of the poorly understood vascular endothelial growth factor (VEGF)165b splice isoform in peripheral artery disease (PAD). This model was built and validated using published and new experimental data from cells, mice, and humans, and explicitly accounts for known properties of VEGF165b: lack of extracellular matrix (ECM)‐binding and weak phosphorylation of vascular endothelial growth factor receptor‐2 (VEGFR2) in vitro. The resulting model captures all known information about VEGF165b distribution and signaling in human PAD, and provides novel, nonintuitive insight into VEGF165b mechanism of action in vivo. Although VEGF165a and VEGF165b compete for VEGFR2 in vitro, simulations show that these isoforms do not compete for VEGFR2 at much lower physiological concentrations. Instead, reduced VEGF165a may drive impaired VEGFR2 signaling. The model predicts that VEGF165b does compete for binding to VEGFR1, supporting a VEGFR1‐mediated response to anti‐VEGF165b. The model predicts a key role for VEGF165b in PAD, but in a different way than previously hypothesized.
Collapse
Affiliation(s)
- Lindsay E Clegg
- Institute for Computational Medicine, Institute for NanoBioTechnology, and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Vijay C Ganta
- Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
| | - Brian H Annex
- Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA.,Department of Cardiovascular Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Feilim Mac Gabhann
- Institute for Computational Medicine, Institute for NanoBioTechnology, and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
42
|
Escobedo N, Oliver G. The Lymphatic Vasculature: Its Role in Adipose Metabolism and Obesity. Cell Metab 2017; 26:598-609. [PMID: 28844882 PMCID: PMC5629116 DOI: 10.1016/j.cmet.2017.07.020] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 06/22/2017] [Accepted: 07/27/2017] [Indexed: 02/07/2023]
Abstract
Obesity is a key risk factor for metabolic and cardiovascular diseases, and although we understand the mechanisms regulating weight and energy balance, the causes of some forms of obesity remain enigmatic. Despite the well-established connections between lymphatics and lipids, and the fact that intestinal lacteals play key roles in dietary fat absorption, the function of the lymphatic vasculature in adipose metabolism has only recently been recognized. It is well established that angiogenesis is tightly associated with the outgrowth of adipose tissue, as expanding adipose tissue requires increased nutrient supply from blood vessels. Results supporting a crosstalk between lymphatic vessels and adipose tissue, and linking lymphatic function with metabolic diseases, obesity, and adipose tissue, also started to accumulate in the last years. Here we review our current knowledge of the mechanisms by which defective lymphatics contribute to obesity and fat accumulation in mouse models, as well as our understanding of the lymphatic-adipose tissue relationship.
Collapse
Affiliation(s)
- Noelia Escobedo
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca, Chile
| | - Guillermo Oliver
- Center for Vascular and Developmental Biology, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
43
|
Dynamics and implications of circulating anti-angiogenic VEGF-A 165b isoform in patients with ST-elevation myocardial infarction. Sci Rep 2017; 7:9962. [PMID: 28855597 PMCID: PMC5577291 DOI: 10.1038/s41598-017-10505-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 08/09/2017] [Indexed: 12/22/2022] Open
Abstract
Angiogenesis is crucial to restore microvascular perfusion in the jeopardized myocardium in the weeks following reperfused ST-segment elevation myocardial infarction (STEMI). (VEGF)-A165b, an anti-angiogenic factor, has been identified as a regulator of vascularization; however, it has not been previously implicated in acute myocardial infarction. We sought to investigate the dynamics of circulating VEGF-A165b and its association with cardiac magnetic resonance-derived infarct size and left ventricular ejection fraction (LVEF). 50 STEMI patients and 23 controls were included. Compared with control individuals, serum VEGF-A165b was elevated in STEMI patients prior to primary percutaneous coronary intervention (PCI). Following PCI, serum VEGF-A165b increased further, reaching a maximum level at 24 h and decreased one month after reperfusion. VEGF-A165b levels at 24 h were associated with a large infarct size and inversely related to LVEF. VEGF-A165b expression was increased in myocardial infarct areas from patients with previous history of AMI. An ex vivo assay using serum from STEMI patients showed that neutralization of VEGF-A165b increased tubulogenesis. Overall, the study suggests that VEGF-A165b might play a deleterious role after AMI as an inhibitor of angiogenesis in the myocardium. Accordingly, neutralization of VEGF-A165b could represent a novel pro-angiogenic therapy for reperfusion of myocardium in STEMI.
Collapse
|
44
|
Karki S, Ngo DTM, Farb MG, Park SY, Saggese SM, Hamburg NM, Carmine B, Hess DT, Walsh K, Gokce N. WNT5A regulates adipose tissue angiogenesis via antiangiogenic VEGF-A 165b in obese humans. Am J Physiol Heart Circ Physiol 2017; 313:H200-H206. [PMID: 28411232 PMCID: PMC6148084 DOI: 10.1152/ajpheart.00776.2016] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 03/20/2017] [Accepted: 04/05/2017] [Indexed: 12/16/2022]
Abstract
Experimental studies have suggested that Wingless-related integration site 5A (WNT5A) is a proinflammatory secreted protein that is associated with metabolic dysfunction in obesity. Impaired angiogenesis in fat depots has been implicated in the development of adipose tissue capillary rarefaction, hypoxia, inflammation, and metabolic dysfunction. We have recently demonstrated that impaired adipose tissue angiogenesis is associated with overexpression of antiangiogenic factor VEGF-A165b in human fat and the systemic circulation. In the present study, we postulated that upregulation of WNT5A is associated with angiogenic dysfunction and examined its role in regulating VEGF-A165b expression in human obesity. We biopsied subcutaneous and visceral adipose tissue from 38 obese individuals (body mass index: 44 ± 7 kg/m2, age: 37 ± 11 yr) during planned bariatric surgery and characterized depot-specific protein expression of VEGF-A165b and WNT5A using Western blot analysis. In both subcutaneous and visceral fat, VEGF-A165b expression correlated strongly with WNT5A protein (r = 0.9, P < 0.001). In subcutaneous adipose tissue where angiogenic capacity is greater than in the visceral depot, exogenous human recombinant WNT5A increased VEGF-A165b expression in both whole adipose tissue and isolated vascular endothelial cell fractions (P < 0.01 and P < 0.05, respectively). This was associated with markedly blunted angiogenic capillary sprout formation in human fat pad explants. Moreover, recombinant WNT5A increased secretion of soluble fms-like tyrosine kinase-1, a negative regulator of angiogenesis, in the sprout media (P < 0.01). Both VEGF-A165b-neutralizing antibody and secreted frizzled-related protein 5, which acts as a decoy receptor for WNT5A, significantly improved capillary sprout formation and reduced soluble fms-like tyrosine kinase-1 production (P < 0.05). We demonstrated a significant regulatory nexus between WNT5A and antiangiogenic VEGF-A165b in the adipose tissue of obese subjects that was linked to angiogenic dysfunction. Elevated WNT5A expression in obesity may function as a negative regulator of angiogenesis.NEW & NOTEWORTHY Wingless-related integration site 5a (WNT5A) negatively regulates adipose tissue angiogenesis via VEGF-A165b in human obesity.
Collapse
Affiliation(s)
- Shakun Karki
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts; and
| | - Doan T M Ngo
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts; and
| | - Melissa G Farb
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts; and
| | - Song Young Park
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts; and
| | - Samantha M Saggese
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts; and
| | - Naomi M Hamburg
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts; and
| | - Brian Carmine
- Department of General Surgery, Boston University School of Medicine, Boston, Massachusetts
| | - Donald T Hess
- Department of General Surgery, Boston University School of Medicine, Boston, Massachusetts
| | - Kenneth Walsh
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts; and
| | - Noyan Gokce
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts; and
| |
Collapse
|
45
|
Breier G, Chavakis T, Hirsch E. Angiogenesis in metabolic-vascular disease. Thromb Haemost 2017; 117:1289-1295. [PMID: 28594427 DOI: 10.1160/th17-05-0325] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 05/29/2017] [Indexed: 12/15/2022]
Abstract
Angiogenesis, literally formation of new blood vessels, is the main process through which the vascular system expands during embryonic and postnatal development. Endothelial cells, which constitute the inner lining of all blood vessels, are typically in a quiescent state in the healthy adult organism. However, in vascular and metabolic diseases, the endothelium becomes unstable and dysfunctional. The resulting tissue hypoxia may thereby induce pathological angiogenesis, which is a hallmark of disease conditions like cancer or diabetic retinopathy. However, recent evidence suggests that angiogenesis is also a major player in the context of further metabolic diseases, especially in obesity. In particular, deregulated angiogenesis is linked with adipose tissue dysfunction and insulin resistance. On the other hand, signalling pathways, such as the PI3K pathway, may regulate metabolic activities in the endothelium. Endothelial cell metabolism emerges as an important regulator of angiogenesis. This review summarises the role of angiogenesis in metabolic-vascular disease, with specific focus on the role of angiogenesis in obesity-related metabolic dysfunction and on signaling pathways, especially PI3K, linking cell metabolism to endothelial function.
Collapse
Affiliation(s)
| | - Triantafyllos Chavakis
- Triantafyllos Chavakis, Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany, E-mail:
| | | |
Collapse
|
46
|
Fuster JJ, Ouchi N, Gokce N, Walsh K. Obesity-Induced Changes in Adipose Tissue Microenvironment and Their Impact on Cardiovascular Disease. Circ Res 2017; 118:1786-807. [PMID: 27230642 DOI: 10.1161/circresaha.115.306885] [Citation(s) in RCA: 452] [Impact Index Per Article: 56.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 02/16/2016] [Indexed: 02/07/2023]
Abstract
Obesity is causally linked with the development of cardiovascular disorders. Accumulating evidence indicates that cardiovascular disease is the collateral damage of obesity-driven adipose tissue dysfunction that promotes a chronic inflammatory state within the organism. Adipose tissues secrete bioactive substances, referred to as adipokines, which largely function as modulators of inflammation. The microenvironment of adipose tissue will affect the adipokine secretome, having actions on remote tissues. Obesity typically leads to the upregulation of proinflammatory adipokines and the downregulation of anti-inflammatory adipokines, thereby contributing to the pathogenesis of cardiovascular diseases. In this review, we focus on the microenvironment of adipose tissue and how it influences cardiovascular disorders, including atherosclerosis and ischemic heart diseases, through the systemic actions of adipokines.
Collapse
Affiliation(s)
- José J Fuster
- From the Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA (J.J.F., N.G., K.W.); and Department of Molecular Cardiology, Nagoya University School of Medicine, Nagoya, Japan (N.O.).
| | - Noriyuki Ouchi
- From the Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA (J.J.F., N.G., K.W.); and Department of Molecular Cardiology, Nagoya University School of Medicine, Nagoya, Japan (N.O.)
| | - Noyan Gokce
- From the Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA (J.J.F., N.G., K.W.); and Department of Molecular Cardiology, Nagoya University School of Medicine, Nagoya, Japan (N.O.)
| | - Kenneth Walsh
- From the Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA (J.J.F., N.G., K.W.); and Department of Molecular Cardiology, Nagoya University School of Medicine, Nagoya, Japan (N.O.).
| |
Collapse
|
47
|
RNA-Sequencing data supports the existence of novel VEGFA splicing events but not of VEGFA xxxb isoforms. Sci Rep 2017; 7:58. [PMID: 28246395 PMCID: PMC5427905 DOI: 10.1038/s41598-017-00100-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 02/06/2017] [Indexed: 01/08/2023] Open
Abstract
Vascular endothelial growth factor (VEGFA), a pivotal regulator of angiogenesis and valuable therapeutic target, is characterised by alternative splicing which generates three principal isoforms, VEGFA121, VEGFA165 and VEGFA189. A second set of anti-angiogenic isoforms termed VEGFAxxxb that utilise an alternative splice site in the final exon have been widely reported, with mRNA detection based principally upon RT-PCR assays. We sought confirmation of the existence of the VEGFAxxxb isoforms within the abundant RNA sequencing data available publicly. Whilst sequences derived specifically from each of the canonical VEGFA isoforms were present in many tissues, there were no sequences derived from VEGFAxxxb isoforms. Sequencing of approximately 50,000 RT-PCR products spanning the exon 7–8 junction in 10 tissues did not identify any VEGFAxxxb transcripts. The absence or extremely low expression of these transcripts in vivo indicates that VEGFAxxxb isoforms are unlikely to play a role in normal physiology. Our analyses also revealed multiple novel splicing events supported by more reads than previously reported for VEGFA145 and VEGFA148 isoforms, including three from novel first exons consistent with existing transcription start site data. These novel VEGFA isoforms may play significant roles in specific cell types.
Collapse
|
48
|
Imatoh T, Kamimura S, Miyazaki M. No causal impact of serum vascular endothelial growth factor level on temporal changes in body mass index in Japanese male workers: a five-year longitudinal study. Endocrine 2017; 55:831-838. [PMID: 27834039 DOI: 10.1007/s12020-016-1165-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 11/01/2016] [Indexed: 11/29/2022]
Abstract
PURPOSE It has been reported that adipocytes secrete vascular endothelial growth factor. Therefore, we conducted a 5-year longitudinal epidemiological study to further elucidate the association between vascular endothelial growth factor levels and temporal changes in body mass index. METHODS Our study subjects were Japanese male workers, who had regular health check-ups. Vascular endothelial growth factor levels were measured at baseline. To examine the association between vascular endothelial growth factor levels and overweight, we calculated the odds ratio using a multivariate logistic regression model. Moreover, linear mixed effect models were used to assess the association between vascular endothelial growth factor level and temporal changes in body mass index during the 5-year follow-up period. RESULTS Vascular endothelial growth factor levels were marginally higher in subjects with a body mass index greater than 25 kg/m2 compared with in those with a body mass index less than 25 kg/m2 (505.4 vs. 465.5 pg/mL, P = 0.1) and were weakly correlated with leptin levels (β: 0.05, P = 0.07). In multivariate logistic regression, subjects in the highest vascular endothelial growth factor quantile were significantly associated with an increased risk for overweight compared with those in the lowest quantile (odds ratio 1.65, 95 % confidential interval: 1.10-2.50). Moreover P for trend was significant (P for trend = 0.003). However, the linear mixed effect model revealed that vascular endothelial growth factor levels were not associated with changes in body mass index over a 5-year period (quantile 2, β: 0.06, P = 0.46; quantile 3, β: -0.06, P = 0.45; quantile 4, β: -0.10, P = 0.22; quantile 1 as reference). CONCLUSIONS Our results suggested that high vascular endothelial growth factor levels were significantly associated with overweight in Japanese males but high vascular endothelial growth factor levels did not necessarily cause obesity.
Collapse
Affiliation(s)
- Takuya Imatoh
- Department of Preventive Medicine and Public Health, Faculty of Medicine, Fukuoka University, Fukuoka, Japan.
| | - Seiichiro Kamimura
- Department of Internal Medicine, Division of Preventive Medicine, Tenjin Clinic, Fukuoka, Japan
| | - Motonobu Miyazaki
- Saitama City Institute of Health Science and Research, Saitama, Japan
| |
Collapse
|
49
|
Clegg LE, Mac Gabhann F. A computational analysis of in vivo VEGFR activation by multiple co-expressed ligands. PLoS Comput Biol 2017; 13:e1005445. [PMID: 28319199 PMCID: PMC5378411 DOI: 10.1371/journal.pcbi.1005445] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 04/03/2017] [Accepted: 03/08/2017] [Indexed: 12/16/2022] Open
Abstract
The splice isoforms of vascular endothelial growth A (VEGF) each have different affinities for the extracellular matrix (ECM) and the coreceptor NRP1, which leads to distinct vascular phenotypes in model systems expressing only a single VEGF isoform. ECM-immobilized VEGF can bind to and activate VEGF receptor 2 (VEGFR2) directly, with a different pattern of site-specific phosphorylation than diffusible VEGF. To date, the way in which ECM binding alters the distribution of isoforms of VEGF and of the related placental growth factor (PlGF) in the body and resulting angiogenic signaling is not well-understood. Here, we extend our previous validated cell-level computational model of VEGFR2 ligation, intracellular trafficking, and site-specific phosphorylation, which captured differences in signaling by soluble and immobilized VEGF, to a multi-scale whole-body framework. This computational systems pharmacology model captures the ability of the ECM to regulate isoform-specific growth factor distribution distinctly for VEGF and PlGF, and to buffer free VEGF and PlGF levels in tissue. We show that binding of immobilized growth factor to VEGF receptors, both on endothelial cells and soluble VEGFR1, is likely important to signaling in vivo. Additionally, our model predicts that VEGF isoform-specific properties lead to distinct profiles of VEGFR1 and VEGFR2 binding and VEGFR2 site-specific phosphorylation in vivo, mediated by Neuropilin-1. These predicted signaling changes mirror those observed in murine systems expressing single VEGF isoforms. Simulations predict that, contrary to the 'ligand-shifting hypothesis,' VEGF and PlGF do not compete for receptor binding at physiological concentrations, though PlGF is predicted to slightly increase VEGFR2 phosphorylation when over-expressed by 10-fold. These results are critical to design of appropriate therapeutic strategies to control VEGF availability and signaling in regenerative medicine applications.
Collapse
Affiliation(s)
- Lindsay E. Clegg
- Institute for Computational Medicine, Institute for NanoBioTechnology, and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Feilim Mac Gabhann
- Institute for Computational Medicine, Institute for NanoBioTechnology, and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| |
Collapse
|
50
|
Canavese M, Ngo DTM, Maddern GJ, Hardingham JE, Price TJ, Hauben E. Biology and therapeutic implications of VEGF-A splice isoforms and single-nucleotide polymorphisms in colorectal cancer. Int J Cancer 2017; 140:2183-2191. [PMID: 27943279 DOI: 10.1002/ijc.30567] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 11/16/2016] [Accepted: 11/30/2016] [Indexed: 12/30/2022]
Abstract
Tumor growth, dissemination and metastasis are dependent on angiogenesis. The predominant vascular endothelial growth factor (VEGF) isoform that plays a major role in angiogenesis is VEGF-A. Indeed, VEGF-A is implicated in promoting angiogenesis of numerous solid malignancies, including colorectal cancer (CRC). A large body of preclinical and clinical evidence indicates that the expression of specific VEGF-A isoforms represents a predominant pro-angiogenic factor, which is associated with formation of metastases and poor prognosis in CRC patients. Different isoforms of human VEGF-A have been identified, all of which arise from alternative splicing of the primary transcript of a single gene. Notably, it has been recently demonstrated that expression of type 3 isoform pattern is significantly correlated with venous involvement in CRC as well as in progression to metastatic colorectal cancer (mCRC), although it remains unclear what proportion of CRC tumors express these isoforms. This review highlights the importance of investigating the genetic and the epigenetic variations in VEGF-A pathways in CRC, the functions of different VEGF-A isoforms and their potential application as prognostic markers and/or therapeutic targets. Better understanding of the mechanisms controlling angiogenesis in liver metastases is necessary to address the limitations of current anti-angiogenic therapies.
Collapse
Affiliation(s)
- Miriam Canavese
- The Basil Hetzel Institute for Translational Health Research, Liver Metastasis Research Group, Discipline of Surgery, University of Adelaide, Adelaide, Australia
| | - Doan T M Ngo
- Cardiology Unit, the Queen Elizabeth Hospital and Basil Hetzel Institute, University of Adelaide, Adelaide, Australia
| | - Guy J Maddern
- Department of Surgery, University of Adelaide, The Queen Elizabeth Hospital, Adelaide, Australia
| | - Jennifer E Hardingham
- Department of Medical Oncology, The Queen Elizabeth Hospital, Adelaide, South Australia and School of Medicine, University of Adelaide, Adelaide
| | - Timothy J Price
- Department of Medical Oncology, The Queen Elizabeth Hospital, Adelaide, South Australia and School of Medicine, University of Adelaide, Adelaide
| | - Ehud Hauben
- The Basil Hetzel Institute for Translational Health Research, Liver Metastasis Research Group, Discipline of Surgery, University of Adelaide, Adelaide, Australia
| |
Collapse
|