1
|
Nadeau A, Ouellet M, Béland R, Mercier C, Robillard S, Lizotte F, Despatis MA, Bentzinger CF, Geraldes P. Endothelial SHP-1 regulates diabetes-induced abnormal collateral vessel formation and endothelial cell senescence. J Mol Cell Cardiol 2025; 202:50-63. [PMID: 40068773 DOI: 10.1016/j.yjmcc.2025.03.005] [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: 08/06/2024] [Revised: 03/01/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND Critical limb ischemia is a major cause of peripheral arterial disease and morbidity affecting patients with diabetes. Diabetes-induced premature senescence of endothelial cells (EC) has been proposed as a mechanism leading to impaired ischemia-driven angiogenesis. We showed that hyperglycemia induced expression of the protein tyrosine phosphatase SHP-1, which reduced angiogenic factor activity in ischemic muscle of diabetic mice. Here, we evaluate the impact of SHP-1 deletion on EC function and senescence. METHODS Ligation of the femoral artery was performed in nondiabetic (NDM) and 3 months diabetic (DM) mice with EC-specific deletion of SHP-1. Cell migration, proliferation and protein expression were evaluated in EC exposed to normal (NG) or high glucose (HG) concentrations. Gastrocnemius and tibial artery of patients with diabetes were collected and analyzed. RESULTS Blood flow reperfusion and limb function were reduced by 43 % and 82 %, respectively in DM mice as compared to NDM mice. EC-specific deletion of SHP-1 in DM mice restored blood flow reperfusion by 60 %, and limb function by 86 %, while capillary density was similar to NDM mice. Moreover, ablation of SHP-1 in EC prevented diabetes-induced expression of the senescence markers p53 and p21 and counteracted Nrf2 downregulation. In EC, elevated expression of beta-galactosidase, p21 and p53, and suppression of Nrf2 and VEGF actions were observed in EC exposed to HG levels and human muscle and artery of patients with diabetes, effects that were reversed by overexpression of dominant negative SHP-1. CONCLUSION SHP-1 in EC is a central effector of diabetes-induced senescence and induces aberrant collateral vessel formation and blood flow reperfusion. Reduced SHP-1 expression counteracts these pathologic features.
Collapse
Affiliation(s)
- Alexandre Nadeau
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Québec, Canada
| | - Marike Ouellet
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Québec, Canada
| | - Raphaël Béland
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Québec, Canada
| | - Clément Mercier
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Québec, Canada
| | - Stéphanie Robillard
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Québec, Canada
| | - Farah Lizotte
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Québec, Canada
| | | | - C Florian Bentzinger
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Québec, Canada; Department of Pharmacology and Physiology, Québec, Canada
| | - Pedro Geraldes
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Québec, Canada; Department of Medicine, Université de Sherbrooke, Québec, Canada.
| |
Collapse
|
2
|
Bhattacharjee S, Gao J, Lu YW, Eisa-Beygi S, Wu H, Li K, Birsner AE, Wong S, Song Y, Shyy JYJ, Cowan DB, Huang W, Wei W, Aikawa M, Shi J, Chen H. Endothelial FOXM1 and Dab2 promote diabetic wound healing. JCI Insight 2025; 10:e186504. [PMID: 39846251 PMCID: PMC11790024 DOI: 10.1172/jci.insight.186504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/26/2024] [Indexed: 01/24/2025] Open
Abstract
Diabetes mellitus can cause impaired and delayed wound healing, leading to lower extremity amputations; however, the mechanisms underlying the regulation of vascular endothelial growth factor-dependent (VEGF-dependent) angiogenesis remain unclear. In our study, the molecular underpinnings of endothelial dysfunction in diabetes are investigated, focusing on the roles of disabled-2 (Dab2) and Forkhead box M1 (FOXM1) in VEGF receptor 2 (VEGFR2) signaling and endothelial cell function. Bulk RNA-sequencing analysis identified significant downregulation of Dab2 in high-glucose-treated primary mouse skin endothelial cells. In diabetic mice with endothelial deficiency of Dab2, in vivo and in vitro angiogenesis and wound healing were reduced when compared with wild-type diabetic mice. Restoration of Dab2 expression by injected mRNA-containing, LyP-1-conjugated lipid nanoparticles rescued impaired angiogenesis and wound healing in diabetic mice. Furthermore, FOXM1 was downregulated in skin endothelial cells under high-glucose conditions as determined by RNA-sequencing analysis. FOXM1 was found to bind to the Dab2 promoter, regulating its expression and influencing VEGFR2 signaling. The FOXM1 inhibitor FDI-6 reduced Dab2 expression and phosphorylation of VEGFR2. Our study provides evidence of the crucial roles of Dab2 and FOXM1 in diabetic endothelial dysfunction and establishes targeted delivery as a promising treatment for diabetic vascular complications.
Collapse
Affiliation(s)
- Sudarshan Bhattacharjee
- Vascular Biology Program, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Surgery and
| | - Jianing Gao
- Vascular Biology Program, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Surgery and
| | - Yao Wei Lu
- Vascular Biology Program, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Surgery and
| | - Shahram Eisa-Beygi
- Vascular Biology Program, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Surgery and
| | - Hao Wu
- Vascular Biology Program, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Surgery and
| | - Kathryn Li
- Vascular Biology Program, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Amy E. Birsner
- Vascular Biology Program, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| | - Scott Wong
- Vascular Biology Program, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Surgery and
| | - Yudong Song
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - John Y-J. Shyy
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Douglas B. Cowan
- Vascular Biology Program, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Surgery and
| | - Wendong Huang
- Division of Molecular Diabetes Research, Department of Diabetes and Metabolic Diseases, City of Hope National Medical Center, Duarte, California, USA
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, and
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jinjun Shi
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Hong Chen
- Vascular Biology Program, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Surgery and
| |
Collapse
|
3
|
Zhang E, Shi Y, Han X, Zhu H, Song B, Yang C, Cao Z. An injectable and biodegradable zwitterionic gel for extending the longevity and performance of insulin infusion catheters. Nat Biomed Eng 2024; 8:1197-1213. [PMID: 37884794 DOI: 10.1038/s41551-023-01108-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 09/18/2023] [Indexed: 10/28/2023]
Abstract
Continuous subcutaneous insulin infusion (CSII) is an essential insulin replacement therapy in the management of diabetes. However, the longevity of clinical CSII is limited by skin complications, by impaired insulin absorption and by occlusions associated with the subcutaneous insertion of CSII catheters, which require replacement and rotation of the insertion site every few days. Here we show that a biodegradable zwitterionic gel covering the tip end of commercial off-the-shelf CSII catheters fully resolves early skin irritations, extends the longevity of catheters and improves the rate of insulin absorption (also with respect to conventional syringe-based subcutaneous injection) for longer than 6 months in diabetic mice, and by 11 days in diabetic minipigs (from 2 to 13 days, under standard CSII-wearing conditions of insulin pump therapy and in a continuous basal-plus-bolus-infusion setting). The implanted gel displayed anti-inflammatory and anti-foreign-body-reaction properties and promoted the local formation of new blood vessels. The gel is subcutaneously injected before the tip of catheter is inserted into it, and should be generally applicable to CSII catheters and other implantable devices.
Collapse
Affiliation(s)
- Ershuai Zhang
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA
| | - Yuanjie Shi
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA
| | - Xiangfei Han
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA
| | - Hui Zhu
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA
| | - Boyi Song
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA
| | - Chengbiao Yang
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA
| | - Zhiqiang Cao
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
4
|
Gaur A, Varatharajan S, Balan Y, Taranikanti M, John NA, Umesh M, Ganji V, Medala K. Brain-derived neurotrophic factor (BDNF) and other neurotrophic factors in type 2 diabetes mellitus and their association with neuropathy. Ir J Med Sci 2024; 193:2287-2292. [PMID: 38806878 DOI: 10.1007/s11845-024-03716-3] [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: 02/18/2024] [Accepted: 05/14/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND Diabetes mellitus (DM) is associated with increased risk of morbidity and premature mortality due to its various complications. In an Indian study, the prevalence of diabetic peripheral neuropathy (DPN) in type 2 diabetic subjects was shown to be 29.2%. There is increasing evidence that a deficiency of nerve growth factor (NGF) in diabetes, as well as the calcitonin gene-related peptide (CGRP), may also contribute to the development of DPN. The aim of the current study was to evaluate nerve growth factor levels with neuropathy in type 2 DM. MATERIALS AND METHODS Forty healthy controls and 40 patients with type 2 DM were recruited; they were asked to report to Dept. of Physiology for initial history taking, general examination and neuropathy examination. A total of 5 mL of blood was collected for neurotrophic factor estimation as well as glycemic profile estimation. RESULTS The brain-derived neurotrophic factor (BDNF) values were significantly lower in the DM group whereas the insulin levels were also quite high in DM. The hot thresholds for both the upper limb and lower limb were greater in the DM group suggesting the impending neuropathy. Similarly, the Michigan scores were also greater in the DM group. The neuropathy parameters especially the Michigan A and B and the hot thresholds were positively correlated with duration of DM and glucose profile. CONCLUSION The neurotrophic factors especially BDNF are drastically reduced in DM patients and are negatively associated with neuropathy, and hence, BDNF can be utilized as a therapeutic target to treat and prevent neuropathy.
Collapse
Affiliation(s)
- Archana Gaur
- Department of Physiology, All India Institute of Medical Sciences, Bibinagar, Hyderabad, Telangana, 508126, India.
| | - Sakthivadivel Varatharajan
- Department of General Medicine, All India Institute of Medical Sciences, Bibinagar, Telangana, 508126, India
| | - Yuvaraj Balan
- Department of Biochemistry, All India Institute of Medical Sciences, Madurai, India
| | - Madhuri Taranikanti
- Department of Physiology, All India Institute of Medical Sciences, Bibinagar, Hyderabad, Telangana, 508126, India
| | - Nitin Ashok John
- Department of Physiology, All India Institute of Medical Sciences, Bibinagar, Hyderabad, Telangana, 508126, India
| | - Madhusudhan Umesh
- Department of Physiology, All India Institute of Medical Sciences, Bibinagar, Hyderabad, Telangana, 508126, India
| | - Vidya Ganji
- Department of Physiology, All India Institute of Medical Sciences, Bibinagar, Hyderabad, Telangana, 508126, India
| | - Kalpana Medala
- Department of Physiology, All India Institute of Medical Sciences, Bibinagar, Hyderabad, Telangana, 508126, India
| |
Collapse
|
5
|
Bhattacharjee S, Gao J, Lu YW, Eisa-Beygi S, Wu H, Li K, Birsner AE, Wong S, Song Y, Shyy JYJ, Cowan DB, Wei W, Aikawa M, Shi J, Chen H. Interplay Between FoxM1 and Dab2 Promotes Endothelial Cell Responses in Diabetic Wound Healing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.07.579019. [PMID: 39253510 PMCID: PMC11383039 DOI: 10.1101/2024.02.07.579019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Diabetes mellitus can cause impaired and delayed wound healing, leading to lower extremity amputations; however, the mechanisms underlying the regulation of vascular endothelial growth factor (VEGF)-dependent angiogenesis remain uncertain and could reveal new therapeutic targets. In our study, the molecular underpinnings of endothelial dysfunction in diabetes were investigated, focusing on the roles of Disabled-2 (Dab2) and Forkhead Box M1 (FoxM1) in VEGF receptor 2 (VEGFR2) signaling and endothelial cell (EC) function. Bulk RNA-sequencing analysis identified significant downregulation of Dab2 in high concentrations glucose treated primary mouse skin ECs, simulating hyperglycemic conditions in diabetes mellitus. In diabetic mice with a genetic EC deficiency of Dab2 angiogenesis was reduced in vivo and in vitro when compared with wild-type mice. Restoration of Dab2 expression by injected mRNA-containing lipid nanoparticles rescued impaired angiogenesis and wound healing in diabetic mice. At the same time, FoxM1 was downregulated in skin ECs subjected to high glucose conditions as determined by RNA-sequencing analysis. FoxM1 was found to bind to the Dab2 promoter, regulating its expression and influencing VEGFR2 signaling. The FoxM1 inhibitor FDI-6 reduced Dab2 expression and phosphorylation of VEGFR2. These findings indicate that restoring Dab2 expression through targeted therapies can enhance angiogenesis and wound repair in diabetes. To explore this therapeutic potential, we tested LyP-1-conjugated lipid nanoparticles (LNPs) containing Dab2 or control mRNAs to target ECs and found the former significantly improved wound healing and angiogenesis in diabetic mice. This study provides evidence of the crucial roles of Dab2 and FoxM1 in diabetic endothelial dysfunction and establishes targeted delivery as a promising treatment for diabetic vascular complications.
Collapse
Affiliation(s)
- Sudarshan Bhattacharjee
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Jianing Gao
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Yao Wei Lu
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Shahram Eisa-Beygi
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Hao Wu
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Kathryn Li
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Amy E. Birsner
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA
| | - Scott Wong
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Yudong Song
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - John Y-J. Shyy
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Douglas B. Cowan
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jinjun Shi
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hong Chen
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
6
|
Kamal R, Awasthi A, Pundir M, Thakur S. Healing the diabetic wound: Unlocking the secrets of genes and pathways. Eur J Pharmacol 2024; 975:176645. [PMID: 38759707 DOI: 10.1016/j.ejphar.2024.176645] [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: 03/04/2024] [Revised: 05/03/2024] [Accepted: 05/13/2024] [Indexed: 05/19/2024]
Abstract
Diabetic wounds (DWs) are open sores that can occur anywhere on a diabetic patient's body. They are often complicated by infections, hypoxia, oxidative stress, hyperglycemia, and reduced growth factors and nucleic acids. The healing process involves four phases: homeostasis, inflammation, proliferation, and remodeling, regulated by various cellular and molecular events. Numerous genes and signaling pathways such as VEGF, TGF-β, NF-κB, PPAR-γ, MMPs, IGF, FGF, PDGF, EGF, NOX, TLR, JAK-STAT, PI3K-Akt, MAPK, ERK, JNK, p38, Wnt/β-catenin, Hedgehog, Notch, Hippo, FAK, Integrin, and Src pathways are involved in these events. These pathways and genes are often dysregulated in DWs leading to impaired healing. The present review sheds light on the pathogenesis, healing process, signaling pathways, and genes involved in DW. Further, various therapeutic strategies that target these pathways and genes via nanotechnology are also discussed. Additionally, clinical trials on DW related to gene therapy are also covered in the present review.
Collapse
Affiliation(s)
- Raj Kamal
- Department of Quality Assurance, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Ankit Awasthi
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, 142001, India.
| | - Mandeep Pundir
- School of Pharmaceutical Sciences, RIMT University, Punjab, 142001, India; Chitkara College of Pharmacy, Chitkara University, Punjab, 142001, India
| | - Shubham Thakur
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, 142001, India
| |
Collapse
|
7
|
Du H, Li S, Lu J, Tang L, Jiang X, He X, Liang J, Liao X, Cui T, Huang Y, Liu H. Single-cell RNA-seq and bulk-seq identify RAB17 as a potential regulator of angiogenesis by human dermal microvascular endothelial cells in diabetic foot ulcers. BURNS & TRAUMA 2023; 11:tkad020. [PMID: 37605780 PMCID: PMC10440157 DOI: 10.1093/burnst/tkad020] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 01/10/2023] [Accepted: 03/22/2023] [Indexed: 08/23/2023]
Abstract
Background Angiogenesis is crucial in diabetic wound healing and is often impaired in diabetic foot ulcers (DFUs). Human dermal microvascular endothelial cells (HDMECs) are vital components in dermal angiogenesis; however, their functional and transcriptomic characteristics in DFU patients are not well understood. This study aimed to comprehensively analyse HDMECs from DFU patients and healthy controls and find the potential regulator of angiogenesis in DFUs. Methods HDMECs were isolated from skin specimens of DFU patients and healthy controls via magnetic-activated cell sorting. The proliferation, migration and tube-formation abilities of the cells were then compared between the experimental groups. Both bulk RNA sequencing (bulk-seq) and single-cell RNA-seq (scRNA-seq) were used to identify RAB17 as a potential marker of angiogenesis, which was further confirmed via weighted gene co-expression network analysis (WGCNA) and least absolute shrink and selection operator (LASSO) regression. The role of RAB17 in angiogenesis was examined through in vitro and in vivo experiments. Results The isolated HDMECs displayed typical markers of endothelial cells. HDMECs isolated from DFU patients showed considerably impaired tube formation, rather than proliferation or migration, compared to those from healthy controls. Gene set enrichment analysis (GSEA), fGSEA, and gene set variation analysis (GSVA) of bulk-seq and scRNA-seq indicated that angiogenesis was downregulated in DFU-HDMECs. LASSO regression identified two genes, RAB17 and CD200, as characteristic of DFU-HDMECs; additionally, the expression of RAB17 was found to be significantly reduced in DFU-HDMECs compared to that in the HDMECs of healthy controls. Overexpression of RAB17 was found to enhance angiogenesis, the expression of hypoxia inducible factor-1α and vascular endothelial growth factor A, and diabetic wound healing, partially through the mitogen-activated protein kinase/extracellular signal-regulated kinase signalling pathway. Conclusions Our findings suggest that the impaired angiogenic capacity in DFUs may be related to the dysregulated expression of RAB17 in HDMECs. The identification of RAB17 as a potential molecular target provides a potential avenue for the treatment of impaired angiogenesis in DFUs.
Collapse
Affiliation(s)
- Hengyu Du
- Department of Plastic Surgery of the First Affiliated Hospital of Jinan University, Institute of New Technology of Plastic Surgery of Jinan University, Key Laboratory of Regenerative Medicine of Ministry of Education, Guangzhou 510630, P.R. China
| | - Shenghong Li
- Department of Plastic Surgery of the First Affiliated Hospital of Jinan University, Institute of New Technology of Plastic Surgery of Jinan University, Key Laboratory of Regenerative Medicine of Ministry of Education, Guangzhou 510630, P.R. China
| | - Jinqiang Lu
- Department of Plastic Surgery of the First Affiliated Hospital of Jinan University, Institute of New Technology of Plastic Surgery of Jinan University, Key Laboratory of Regenerative Medicine of Ministry of Education, Guangzhou 510630, P.R. China
| | - Lingzhi Tang
- Department of Plastic Surgery of the First Affiliated Hospital of Jinan University, Institute of New Technology of Plastic Surgery of Jinan University, Key Laboratory of Regenerative Medicine of Ministry of Education, Guangzhou 510630, P.R. China
| | - Xiao Jiang
- Department of Plastic Surgery of the First Affiliated Hospital of Jinan University, Institute of New Technology of Plastic Surgery of Jinan University, Key Laboratory of Regenerative Medicine of Ministry of Education, Guangzhou 510630, P.R. China
| | - Xi He
- Department of Plastic Surgery of the First Affiliated Hospital of Jinan University, Institute of New Technology of Plastic Surgery of Jinan University, Key Laboratory of Regenerative Medicine of Ministry of Education, Guangzhou 510630, P.R. China
| | - Jiaji Liang
- Department of Plastic Surgery of the First Affiliated Hospital of Jinan University, Institute of New Technology of Plastic Surgery of Jinan University, Key Laboratory of Regenerative Medicine of Ministry of Education, Guangzhou 510630, P.R. China
| | - Xuan Liao
- Department of Plastic Surgery of the First Affiliated Hospital of Jinan University, Institute of New Technology of Plastic Surgery of Jinan University, Key Laboratory of Regenerative Medicine of Ministry of Education, Guangzhou 510630, P.R. China
| | - Taixing Cui
- Dalton Cardiovascular Research Center, Department of Medical Pharmacology and Physiology, University of Missouri, School of Medicine, 134 Research Park Dr, Columbia, MO 65211, USA
| | - Yuesheng Huang
- Institute of Wound Repair and Regeneration Medicine, Southern University of Science and Technology School of Medicine, and Department of Wound Repair, Southern University of Science and Technology Hospital, Shenzhen, Guangdong, 518055, China
| | - Hongwei Liu
- Department of Plastic Surgery of the First Affiliated Hospital of Jinan University, Institute of New Technology of Plastic Surgery of Jinan University, Key Laboratory of Regenerative Medicine of Ministry of Education, Guangzhou 510630, P.R. China
| |
Collapse
|
8
|
Bai T, Li M, Liu Y, Qiao Z, Zhang X, Wang Y, Wang Z. The promotion action of AURKA on post-ischemic angiogenesis in diabetes-related limb ischemia. Mol Med 2023; 29:39. [PMID: 36977984 PMCID: PMC10053687 DOI: 10.1186/s10020-023-00635-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
Abstract
Background
Diabetes-related limb ischemia is a challenge for lower extremity amputation. Aurora Kinase A (AURKA) is an essential serine/threonine kinase for mitosis, while its role in limb ischemia remains unclear.
Method
Human microvascular endothelial cells (HMEC-1) were cultured in high glucose (HG, 25 mmol/L d-glucose) and no additional growth factors (ND) medium to mimic diabetes and low growth factors deprivation as in vitro model. Diabetic C57BL/6 mice were induced by streptozotocin (STZ) administration. After seven days, ischemia was surgically performed by left unilateral femoral artery ligation on diabetic mice. The vector of adenovirus was utilized to overexpress AURKA in vitro and in vivo.
Results
In our study, HG and ND-mediated downregulation of AURKA impaired the cell cycle progression, proliferation, migration, and tube formation ability of HMEC-1, which were rescued by overexpressed AURKA. Increased expression of vascular endothelial growth factor A (VEGFA) induced by overexpressed AURKA were likely regulatory molecules that coordinate these events. Mice with AURKA overexpression exhibited improved angiogenesis in response to VEGF in Matrigel plug assay, with increased capillary density and hemoglobin content. In diabetic limb ischemia mice, AURKA overexpression rescued blood perfusion and motor deficits, accompanied by the recovery of gastrocnemius muscles observed by H&E staining and positive Desmin staining. Moreover, AURKA overexpression rescued diabetes-related impairment of angiogenesis, arteriogenesis, and functional recovery in the ischemic limb. Signal pathway results revealed that VEGFR2/PI3K/AKT pathway might be involved in AURKA triggered angiogenesis procedure. In addition, AURKA overexpression impeded oxidative stress and subsequent following lipid peroxidation both in vitro and in vivo, indicating another protective mechanism of AURKA function in diabetic limb ischemia. The changes in lipid peroxidation biomarkers (lipid ROS, GPX4, SLC7A11, ALOX5, and ASLC4) in in vitro and in vivo were suggestive of the possible involvement of ferroptosis and interaction between AUKRA and ferroptosis in diabetic limb ischemia, which need further investigation.
Conclusions
These results implicated a potent role of AURKA in diabetes-related impairment of ischemia-mediated angiogenesis and implied a potential therapeutic target for ischemic diseases of diabetes.
Collapse
|
9
|
Singh MV, Dokun AO. Diabetes mellitus in peripheral artery disease: Beyond a risk factor. Front Cardiovasc Med 2023; 10:1148040. [PMID: 37139134 PMCID: PMC10149861 DOI: 10.3389/fcvm.2023.1148040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/22/2023] [Indexed: 05/05/2023] Open
Abstract
Peripheral artery disease (PAD) is one of the major cardiovascular diseases that afflicts a large population worldwide. PAD results from occlusion of the peripheral arteries of the lower extremities. Although diabetes is a major risk factor for developing PAD, coexistence of PAD and diabetes poses significantly greater risk of developing critical limb threatening ischemia (CLTI) with poor prognosis for limb amputation and high mortality. Despite the prevalence of PAD, there are no effective therapeutic interventions as the molecular mechanism of how diabetes worsens PAD is not understood. With increasing cases of diabetes worldwide, the risk of complications in PAD have greatly increased. PAD and diabetes affect a complex web of multiple cellular, biochemical and molecular pathways. Therefore, it is important to understand the molecular components that can be targeted for therapeutic purposes. In this review, we describe some major developments in enhancing the understanding of the interactions of PAD and diabetes. We also provide results from our laboratory in this context.
Collapse
Affiliation(s)
- Madhu V. Singh
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Ayotunde O. Dokun
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Fraternal Order of Eagles Diabetes Research Centre, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Correspondence: Ayotunde O. Dokun
| |
Collapse
|
10
|
Goswami AG, Basu S, Huda F, Pant J, Ghosh Kar A, Banerjee T, Shukla VK. An appraisal of vascular endothelial growth factor (VEGF): the dynamic molecule of wound healing and its current clinical applications. Growth Factors 2022; 40:73-88. [PMID: 35584274 DOI: 10.1080/08977194.2022.2074843] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Angiogenesis is a critical step of wound healing, and its failure leads to chronic wounds. The idea of restoring blood flow to the damaged tissues by promoting neo-angiogenesis is lucrative and has been researched extensively. Vascular endothelial growth factor (VEGF), a key dynamic molecule of angiogenesis has been investigated for its functions. In this review, we aim to appraise its biology, the comprehensive role of this dynamic molecule in the wound healing process, and how this knowledge has been translated in clinical application in various types of wounds. Although, most laboratory research on the use of VEGF is promising, its clinical applications have not met great expectations. We discuss various lacunae that might exist in making its clinical application unsuccessful for commercial use, and provide insight to the foundation for future research.
Collapse
Affiliation(s)
- Aakansha Giri Goswami
- Department of General surgery, All India Institute of Medical Sciences, Rishikesh, India
| | - Somprakas Basu
- Department of General surgery, All India Institute of Medical Sciences, Rishikesh, India
| | - Farhanul Huda
- Department of General surgery, All India Institute of Medical Sciences, Rishikesh, India
| | - Jayanti Pant
- Department of Physiology, All India Institute of Medical Sciences, Rishikesh, India
| | - Amrita Ghosh Kar
- Department of Pathology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Tuhina Banerjee
- Department of Microbiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Vijay Kumar Shukla
- Department of General Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
11
|
Semenza GL. Hypoxia-inducible factors: roles in cardiovascular disease progression, prevention, and treatment. Cardiovasc Res 2022; 119:371-380. [PMID: 35687650 DOI: 10.1093/cvr/cvac089] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Indexed: 12/17/2022] Open
Abstract
Hypoxia-inducible factors (HIF)-1 and HIF-2 are master regulators of oxygen homeostasis that regulate the expression of thousands of genes in order to match O2 supply and demand. A large body of experimental data links HIF activity to protection against multiple disorders affecting the cardiovascular system: ischemic cardiovascular disease (including coronary artery disease and peripheral artery disease), through collateral blood vessel formation and preconditioning phenomena; emphysema; lymphedema; and lung transplant rejection. In these disorders, strategies to increase the expression of one or both HIFs may be of therapeutic utility. Conversely, extensive data link HIFs to the pathogenesis of pulmonary arterial hypertension and drugs that inhibit one or both HIFs may be useful in treating this disease.
Collapse
Affiliation(s)
- Gregg L Semenza
- Armstrong Oxygen Biology Research Center, Vascular Program, Institute for Cell Engineering; and Departments of Genetic Medicine, Pediatrics, Medicine, Oncology, Radiation Oncology, and Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
12
|
Rustagi Y, Abouhashem AS, Verma P, Verma SS, Hernandez E, Liu S, Kumar M, Guda PR, Srivastava R, Mohanty SK, Kacar S, Mahajan S, Wanczyk KE, Khanna S, Murphy MP, Gordillo GM, Roy S, Wan J, Sen CK, Singh K. Endothelial Phospholipase Cγ2 Improves Outcomes of Diabetic Ischemic Limb Rescue Following VEGF Therapy. Diabetes 2022; 71:1149-1165. [PMID: 35192691 PMCID: PMC9044136 DOI: 10.2337/db21-0830] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 02/15/2022] [Indexed: 11/13/2022]
Abstract
Therapeutic vascular endothelial growth factor (VEGF) replenishment has met with limited success for the management of critical limb-threatening ischemia. To improve outcomes of VEGF therapy, we applied single-cell RNA sequencing (scRNA-seq) technology to study the endothelial cells of the human diabetic skin. Single-cell suspensions were generated from the human skin followed by cDNA preparation using the Chromium Next GEM Single-cell 3' Kit v3.1. Using appropriate quality control measures, 36,487 cells were chosen for downstream analysis. scRNA-seq studies identified that although VEGF signaling was not significantly altered in diabetic versus nondiabetic skin, phospholipase Cγ2 (PLCγ2) was downregulated. The significance of PLCγ2 in VEGF-mediated increase in endothelial cell metabolism and function was assessed in cultured human microvascular endothelial cells. In these cells, VEGF enhanced mitochondrial function, as indicated by elevation in oxygen consumption rate and extracellular acidification rate. The VEGF-dependent increase in cell metabolism was blunted in response to PLCγ2 inhibition. Follow-up rescue studies therefore focused on understanding the significance of VEGF therapy in presence or absence of endothelial PLCγ2 in type 1 (streptozotocin-injected) and type 2 (db/db) diabetic ischemic tissue. Nonviral topical tissue nanotransfection technology (TNT) delivery of CDH5 promoter-driven PLCγ2 open reading frame promoted the rescue of hindlimb ischemia in diabetic mice. Improvement of blood flow was also associated with higher abundance of VWF+/CD31+ and VWF+/SMA+ immunohistochemical staining. TNT-based gene delivery was not associated with tissue edema, a commonly noted complication associated with proangiogenic gene therapies. Taken together, our study demonstrates that TNT-mediated delivery of endothelial PLCγ2, as part of combination gene therapy, is effective in diabetic ischemic limb rescue.
Collapse
Affiliation(s)
- Yashika Rustagi
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
| | - Ahmed S. Abouhashem
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
- Sharkia Clinical Research Department, Ministry of Health and Population, Cairo, Egypt
| | - Priyanka Verma
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
| | - Sumit S. Verma
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
| | - Edward Hernandez
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
| | - Sheng Liu
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN
| | - Manishekhar Kumar
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
| | - Poornachander R. Guda
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
| | - Rajneesh Srivastava
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
| | - Sujit K. Mohanty
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
| | - Sedat Kacar
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
| | - Sanskruti Mahajan
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
| | - Kristen E. Wanczyk
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
| | - Savita Khanna
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
| | - Michael P. Murphy
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
| | - Gayle M. Gordillo
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
| | - Sashwati Roy
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
| | - Jun Wan
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN
| | - Chandan K. Sen
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
| | - Kanhaiya Singh
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
13
|
Zhao C, Heuslein JL, Zhang Y, Annex BH, Popel AS. Dynamic Multiscale Regulation of Perfusion Recovery in Experimental Peripheral Arterial Disease: A Mechanistic Computational Model. JACC Basic Transl Sci 2022; 7:28-50. [PMID: 35128207 PMCID: PMC8807862 DOI: 10.1016/j.jacbts.2021.10.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/13/2021] [Accepted: 10/13/2021] [Indexed: 01/29/2023]
Abstract
In peripheral arterial disease (PAD), the degree of endogenous capacity to modulate revascularization of limb muscle is central to the management of leg ischemia. To characterize the multiscale and multicellular nature of revascularization in PAD, we have developed the first computational systems biology model that mechanistically incorporates intracellular, cellular, and tissue-level features critical for the dynamic reconstitution of perfusion after occlusion-induced ischemia. The computational model was specifically formulated for a preclinical animal model of PAD (mouse hindlimb ischemia [HLI]), and it has gone through multilevel model calibration and validation against a comprehensive set of experimental data so that it accurately captures the complex cellular signaling, cell-cell communication, and function during post-HLI perfusion recovery. As an example, our model simulations generated a highly detailed description of the time-dependent spectrum-like macrophage phenotypes in HLI, and through model sensitivity analysis we identified key cellular processes with potential therapeutic significance in the pathophysiology of PAD. Furthermore, we computationally evaluated the in vivo effects of different targeted interventions on post-HLI tissue perfusion recovery in a model-based, data-driven, virtual mouse population and experimentally confirmed the therapeutic effect of a novel model-predicted intervention in real HLI mice. This novel multiscale model opens up a new avenue to use integrative systems biology modeling to facilitate translational research in PAD.
Collapse
Key Words
- ARG1, arginase-1
- EC, endothelial cell
- HLI, hindlimb ischemia
- HMGB1, high-mobility group box 1
- HUVEC, human umbilical vein endothelial call
- IFN, interferon
- IL, interleukin
- MLKL, mixed lineage kinase domain-like protein
- PAD, peripheral arterial disease
- RT-PCR, reverse transcriptase polymerase chain reaction
- TLR4, Toll-like receptor 4
- TNF, tumor necrosis factor
- VEGF, vascular endothelial growth factor
- VMP, virtual mouse population
- hindlimb ischemia
- macrophage polarization
- mathematical modeling
- necrosis/necroptosis
- perfusion recovery
- peripheral arterial disease
- systems biology
- virtual mouse population
Collapse
Affiliation(s)
- Chen Zhao
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Joshua L. Heuslein
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
| | - Yu Zhang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Brian H. Annex
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Aleksander S. Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
14
|
Niknam Z, Samadi M, Ghalibafsabbaghi A, Chodari L. IGF-I combined with exercise improve diabetes-induced vascular dysfunction in heart of male Wistar rats. J Cardiovasc Thorac Res 2021; 14:34-41. [PMID: 35620752 PMCID: PMC9106942 DOI: 10.34172/jcvtr.2021.54] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 11/24/2021] [Indexed: 11/30/2022] Open
Abstract
Introduction: This research investigates the impact of insulin-like growth factor-I (IGF -I)and exercise on mediators associated with angiogenesis (VEGF-A, TSP-1, and NF-кβ) and capillarization status of the diabetic rats’ hearts.
Methods: Splitting of forty Wistar male rats into five groups occurred as following: control,diabetes, diabetes+IGF-I, diabetes+exercise, and diabetes+exercise+IGF-I.Through intraperitoneal administration of 60 mg/kg streptozotocin, the condition of Type 1diabetes was escalated. After four weeks of treatment with IGF-I (2 mg/kg/day) or treadmill exercise (17 m/min, zero degrees slope, 30 min/day), in the heart, microvascular density and protein levels of VEGF-A, TSP-1, and NF-кβ were determined by H&E staining and ELISA,respectively.
Results: Within the diabetic group, observations present a significant decrease in VEGF-A and MVD levels, whereas an increase in the TSP-1 and NF-Κb levels. While these impacts were reversed by either IGF-I or exercise treatments, simultaneous treatment had synergistic effects. Moreover, among diabetic rats, undesirable histologic alterations of the heart were demonstrated, including myonecrosis, interstitial edema, hemorrhage, and mononuclear immune cell infiltration, whereas treatments improved these changes.
Conclusion: These data manifest that IGF-I and exercise can increase the cardiac angiogenesis of diabetic rats through increasing expression of VEGF-A, and decreasing TSP-1 and NF-кβproteins level, also can improve myocardial tissue damages.
Collapse
Affiliation(s)
- Zahra Niknam
- Proteomics Research Center, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Mahrokh Samadi
- Nephrology and Kidney Transplant Research Center, Clinical Research Institute, Urmia University of Medical Sciences , Urmia, Iran
| | | | - leila Chodari
- Neurophysiology Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
- Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
15
|
Nishikai-Yan Shen T, Kado M, Hagiwara H, Fujimura S, Mizuno H, Tanaka R. MMP9 secreted from mononuclear cell quality and quantity culture mediates STAT3 phosphorylation and fibroblast migration in wounds. Regen Ther 2021; 18:464-471. [PMID: 34805452 PMCID: PMC8581454 DOI: 10.1016/j.reth.2021.10.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 09/21/2021] [Accepted: 10/20/2021] [Indexed: 01/13/2023] Open
Abstract
Introduction Intractable ulcers may ultimately lead to amputation. To promote wound healing, researchers developed a serum-free ex vivo peripheral blood mononuclear cell quality and quantity culture (MNC-QQc) as a source for cell therapy. In mice, pigs, and even humans, cell therapy with MNC-QQc reportedly yields a high regenerative efficacy. However, the mechanism of wound healing by MNC-QQc cells remains largely unknown. Hence, using an in vitro wound healing model, this study aimed to investigate MNC-QQc cells and the migratory potential of dermal fibroblasts. Methods After separation from a 50 mL blood sample from healthy individuals, mononuclear cells were cultured for 7 days in a serum-free ex vivo expansion system with five different cytokines (MNC-QQc method). The effects of MNC-QQc cells on human dermal fibroblast migration were observed by scratch assay. An angiogenesis array screened the MNC-QQc cell supernatant for proteins related to wound healing. Finally, fibroblast migration was confirmed by observing the intracellular signal transduction pathways via Western blot. Results The migration of fibroblasts co-cultured with MNC-QQc cells increased by matrix metallopeptidase-9 (MMP9) secretion, as suggested by the angiogenesis array. Furthermore, the phosphorylation of signal transducer and activator of transcription 3 (STAT3) in fibroblast/MNC-QQc cell co-culture and fibroblast culture with added recombinant human MMP9 protein increased. When fibroblasts were cultured with either an MMP9 inhibitor or a STAT3 inhibitor, both fibroblast migration and STAT3 phosphorylation were significantly suppressed. Conclusions MNC-QQc cells promote wound healing by the secretion of MMP9, which induces fibroblast migration via the STAT3 signaling pathway.
Collapse
Key Words
- BM, Bone marrow
- BMMNC, Bone marrow mononuclear cells
- Cell culture
- Cell therapy
- DMEM, Dulbecco's modified Eagle's medium
- EPC, Endothelial progenitor cells
- FBS, Fetal bovine serum
- HRP, Horseradish peroxidase
- MMP, Matrix metallopeptidase
- MMP9
- MNC, Monocyte cell
- MNC-QQc
- PB, Peripheral blood
- PBMNC, Peripheral blood monocyte cells
- PBS, Phosphate-buffered saline
- QQc, Quality and quantity culture
- SE, Standard error
- VEGF, Vascular endothelial growth factor
- Wound healing
Collapse
Affiliation(s)
- Tsubame Nishikai-Yan Shen
- Division of Regenerative Therapy, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan.,Intractable Disease Research Center, Juntendo University School of Medicine, Tokyo, Japan
| | - Makiko Kado
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Hiroko Hagiwara
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan.,Center for Genomic and Regenerative Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | - Satoshi Fujimura
- Division of Regenerative Therapy, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan.,Intractable Disease Research Center, Juntendo University School of Medicine, Tokyo, Japan
| | - Hiroshi Mizuno
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan.,Intractable Disease Research Center, Juntendo University School of Medicine, Tokyo, Japan.,Center for Genomic and Regenerative Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | - Rica Tanaka
- Division of Regenerative Therapy, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan.,Intractable Disease Research Center, Juntendo University School of Medicine, Tokyo, Japan.,Center for Genomic and Regenerative Medicine, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
16
|
Wang H, Pei S, Fang S, Jin S, Deng S, Zhao Y, Feng Y. Irisin restores high glucose-induced cell injury in vascular endothelial cells by activating Notch pathway via Notch receptor 1. Biosci Biotechnol Biochem 2021; 85:2093-2102. [PMID: 34329390 DOI: 10.1093/bbb/zbab137] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 07/19/2021] [Indexed: 01/12/2023]
Abstract
Diabetic foot ulcers (DFU) are a vascular complication of diabetes mellitus (DM). It has been confirmed that irisin is closely related to DM. However, the effect of irisin on DFU is obscure and needs further study. After human umbilical vein endothelial cell lines (HUVECs) were treated with different concentrations' irisin, normal glucose, high glucose (HG), HG plus irisin-high (H) or sh-Notch1, cell biological behaviors, LDH, and VEGFA were detected by cell function experiments. Apoptosis- and Notch pathway-related protein levels were evaluated by Western blot. Irisin has no cytotoxicity, and irisin-H elevated cell viability and inhibited apoptosis and LDH level in HG-induced HUVECs. Meanwhile, irisin-H restored HG-repressed migration and angiogenesis in HUVECs. Irisin-H inhibited apoptosis-related protein levels and promoted VEGFA and Notch pathway-related protein levels in HG-treated HUVECs. Additionally, sh-Notch1 reversed the protective effect of irisin-H in HG-treated HUVECs. Irisin restores HG-induced cell injury and angiogenesis in HUVECs by activating Notch pathway via Notch1.
Collapse
Affiliation(s)
- Hanrui Wang
- Departement of Vascular Surgery, The First Affiliated Hospital of Jiamusi University, Jiamusi University, Jiamusi, Heilongjiang, China
| | - Siying Pei
- Department of Biochemistry, School of Basic Medical Science, Jiamusi University, Jiamusi, Heilongjiang, China
| | - Shuqing Fang
- Department of Nephrology, The Central Hospital of Jia Mu Si City, Jiamusi, Heilongjiang, China
| | - Song Jin
- Departement of Vascular Surgery, The First Affiliated Hospital of Jiamusi University, Jiamusi University, Jiamusi, Heilongjiang, China
| | - Shuhua Deng
- Nursing Department, The Central Hospital of Jia Mu Si City, Jiamusi, Heilongjiang, China
| | - Yanan Zhao
- Department of Internal Medicine, Hospital of Traditional Chinese Medicine of Qingan County, Suihua, Heilongjiang, China
| | - Yao Feng
- Department of TCM, The First Affiliated Hospital of Jiamusi University; Jiamusi University, Jiamusi, Heilongjiang, China
| |
Collapse
|
17
|
Solly EL, Psaltis PJ, Bursill CA, Tan JTM. The Role of miR-181c in Mechanisms of Diabetes-Impaired Angiogenesis: An Emerging Therapeutic Target for Diabetic Vascular Complications. Front Pharmacol 2021; 12:718679. [PMID: 34483928 PMCID: PMC8414254 DOI: 10.3389/fphar.2021.718679] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/06/2021] [Indexed: 12/13/2022] Open
Abstract
Diabetes mellitus is estimated to affect up to 700 million people by the year 2045, contributing to an immense health and economic burden. People living with diabetes have a higher risk of developing numerous debilitating vascular complications, leading to an increased need for medical care, a reduced quality of life and increased risk of early death. Current treatments are not satisfactory for many patients who suffer from impaired angiogenesis in response to ischaemia, increasing their risk of ischaemic cardiovascular conditions. These vascular pathologies are characterised by endothelial dysfunction and abnormal angiogenesis, amongst a host of impaired signaling pathways. Therapeutic stimulation of angiogenesis holds promise for the treatment of diabetic vascular complications that stem from impaired ischaemic responses. However, despite significant effort and research, there are no established therapies that directly stimulate angiogenesis to improve ischaemic complications such as ischaemic heart disease and peripheral artery disease, highlighting the immense unmet need. However, despite significant effort and research, there are no established therapies that directly stimulate angiogenesis in a clinical setting, highlighting the immense unmet need. MicroRNAs (miRNAs) are emerging as powerful targets for multifaceted diseases including diabetes and cardiovascular disease. This review highlights the potential role of microRNAs as therapeutic targets for rescuing diabetes-impaired angiogenesis, with a specific focus on miR-181c, which we have previously identified as an important angiogenic regulator. Here we summarise the pathways currently known to be regulated by miR-181c, which include the classical angiogenesis pathways that are dysregulated in diabetes, mitochondrial function and axonal guidance, and describe how these relate both directly and indirectly to angiogenesis. The pleiotropic actions of miR-181c across multiple key angiogenic signaling pathways and critical cellular processes highlight its therapeutic potential as a novel target for treating diabetic vascular complications.
Collapse
Affiliation(s)
- Emma L Solly
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.,Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Peter J Psaltis
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.,Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Christina A Bursill
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.,Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia.,ARC Centre of Excellence for Nanoscale BioPhotonics, The University of Adelaide, Adelaide, SA, Australia
| | - Joanne T M Tan
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.,Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
18
|
Mercier C, Brazeau T, Lamoureux J, Boisvert E, Robillard S, Breton V, Paré M, Guay A, Lizotte F, Despatis MA, Geraldes P. Diabetes Impaired Ischemia-Induced PDGF (Platelet-Derived Growth Factor) Signaling Actions and Vessel Formation Through the Activation of Scr Homology 2-Containing Phosphatase-1. Arterioscler Thromb Vasc Biol 2021; 41:2469-2482. [PMID: 34320834 DOI: 10.1161/atvbaha.121.316638] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Objective Critical limb ischemia is a major complication of diabetes characterized by insufficient collateral vessel development and proper growth factor signaling unresponsiveness. Although mainly deactivated by hypoxia, phosphatases are important players in the deregulation of proangiogenetic pathways. Previously, SHP-1 (Scr homology 2-containing phosphatase-1) was found to be associated with the downregulation of growth factor actions in the diabetic muscle. Thus, we aimed to gain further understanding of the impact of SHP-1 on smooth muscle cell (SMC) function under hypoxic and diabetic conditions. Approach and Results Despite being inactivated under hypoxic conditions, high glucose level exposure sustained SHP-1 phosphatase activity in SMC and increased its interaction with PDGFR (platelet-derived growth factor receptor)-β, thus reducing PDGF proangiogenic actions. Overexpression of an inactive form of SHP-1 fully restored PDGF-induced proliferation, migration, and signaling pathways in SMC exposed to high glucose and hypoxia. Nondiabetic and diabetic mice with deletion of SHP-1 specifically in SMC were generated. Ligation of the femoral artery was performed, and blood flow was measured for 4 weeks. Blood flow reperfusion, vascular density and maturation, and limb survival were all improved while vascular apoptosis was attenuated in diabetic SMC-specific SHP-1 null mice as compared to diabetic mice. Conclusions Diabetes and high glucose level exposure maintained SHP-1 activity preventing hypoxia-induced PDGF actions in SMC. Specific deletion of SHP-1 in SMC partially restored blood flow reperfusion in the diabetic ischemic limb. Therefore, local modulation of SHP-1 activity in SMC could represent a potential therapeutic avenue to improve the proangiogenic properties of SMC under ischemia and diabetes.
Collapse
MESH Headings
- Angiogenesis Inducing Agents/pharmacology
- Animals
- Blood Glucose/metabolism
- Case-Control Studies
- Cattle
- Cell Hypoxia
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Diabetes Mellitus, Experimental/enzymology
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/physiopathology
- Diabetic Angiopathies/enzymology
- Diabetic Angiopathies/genetics
- Diabetic Angiopathies/physiopathology
- Enzyme Activation
- Hindlimb/blood supply
- Humans
- Ischemia/enzymology
- Ischemia/physiopathology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Neovascularization, Physiologic/drug effects
- Platelet-Derived Growth Factor/pharmacology
- Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics
- Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism
- Signal Transduction
- Mice
Collapse
Affiliation(s)
- Clément Mercier
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke (C.M., T.B., J.L., E.B., S.R., V.B., M.P., A.G., F.L., P.G.), Université de Sherbrooke, Québec, Canada
| | - Tristan Brazeau
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke (C.M., T.B., J.L., E.B., S.R., V.B., M.P., A.G., F.L., P.G.), Université de Sherbrooke, Québec, Canada
| | - Jérémy Lamoureux
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke (C.M., T.B., J.L., E.B., S.R., V.B., M.P., A.G., F.L., P.G.), Université de Sherbrooke, Québec, Canada
| | - Elizabeth Boisvert
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke (C.M., T.B., J.L., E.B., S.R., V.B., M.P., A.G., F.L., P.G.), Université de Sherbrooke, Québec, Canada
| | - Stéphanie Robillard
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke (C.M., T.B., J.L., E.B., S.R., V.B., M.P., A.G., F.L., P.G.), Université de Sherbrooke, Québec, Canada
| | - Valérie Breton
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke (C.M., T.B., J.L., E.B., S.R., V.B., M.P., A.G., F.L., P.G.), Université de Sherbrooke, Québec, Canada
| | - Martin Paré
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke (C.M., T.B., J.L., E.B., S.R., V.B., M.P., A.G., F.L., P.G.), Université de Sherbrooke, Québec, Canada
| | - Andréanne Guay
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke (C.M., T.B., J.L., E.B., S.R., V.B., M.P., A.G., F.L., P.G.), Université de Sherbrooke, Québec, Canada
| | - Farah Lizotte
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke (C.M., T.B., J.L., E.B., S.R., V.B., M.P., A.G., F.L., P.G.), Université de Sherbrooke, Québec, Canada
| | | | - Pedro Geraldes
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke (C.M., T.B., J.L., E.B., S.R., V.B., M.P., A.G., F.L., P.G.), Université de Sherbrooke, Québec, Canada
- Division of Endocrinology, Department of Medicine (P.G.), Université de Sherbrooke, Québec, Canada
| |
Collapse
|
19
|
Kant V, Jangir BL, Sharma M, Kumar V, Joshi VG. Topical application of quercetin improves wound repair and regeneration in diabetic rats. Immunopharmacol Immunotoxicol 2021; 43:536-553. [PMID: 34278923 DOI: 10.1080/08923973.2021.1950758] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Purpose: There is an urgent need of effective drug/formulation to speed up the healing process in diabetic wounds. In our earlier studies, quercetin has accelerated the healing of nondiabetic wounds. So, we investigated the wound-healing potentials of quercetin in diabetic rats.Materials and methods: A square-shaped cutaneous wound (≈400 mm2) was created on the back of nondiabetic and diabetic rats. They were divided into three groups, viz. healthy control (nondiabetic), diabetic control and diabetic-treated group. Ointment base was topically applied for 21 days in healthy and diabetic control groups. Quercetin (0.3%) ointment was similarly applied in third group. Effects of quercetin on repair and regenerations of diabetic wounds in terms of wound closure, inflammation, angiogenesis, fibroblast proliferation, collagen synthesis, epithelialization, axonal regeneration etc was studied.Results: Quercetin accelerated the wound closure and increased the expressions of IL-10, VEGF and TGF-β1 in granulation/healing tissue of diabetic wound. However, quercetin decreased the expression of TNF-α, IL-1β, and MMP-9. Histopathological evaluation revealed amelioration of persistence of inflammatory cells by quercetin in diabetic wounds. There was good quality of granulation tissue, marked fibroblast proliferation, well organized collagen deposition, early regeneration of epithelial layer etc. in the quercetin treated diabetic wounds in comparison to diabetic control group. Results of immunohistochemistry showed more angiogenesis, faster phenotypic switching of fibroblast to myofibroblasts and increased GAP-43 positive nerve fibers in quercetin-treated diabetic wounds.Conclusion: Quercetin ointment at 0.3% w/w concentration modulates cytokines, growth factors and protease, thereby improved repair and regenerations of cutaneous diabetic wounds in rats.
Collapse
Affiliation(s)
- Vinay Kant
- Department of Veterinary Pharmacology and Toxicology, Lala Lajpat Rai University of Veterinary & Animal Sciences (LUVAS), Hisar, Haryana, India
| | - Babu Lal Jangir
- Department of Veterinary Pathology, Lala Lajpat Rai University of Veterinary & Animal Sciences (LUVAS), Hisar, Haryana, India
| | - Maneesh Sharma
- Department of Veterinary Clinical Complex, College of Veterinary Sciences, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, Haryana, India
| | - Vinod Kumar
- Department of Veterinary Pharmacology and Toxicology, Lala Lajpat Rai University of Veterinary & Animal Sciences (LUVAS), Hisar, Haryana, India
| | - Vinay G Joshi
- Department of Animal Biotechnology, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, Haryana, India
| |
Collapse
|
20
|
Certelli A, Valente P, Uccelli A, Grosso A, Di Maggio N, D'Amico R, Briquez PS, Hubbell JA, Wolff T, Gürke L, Mujagic E, Gianni-Barrera R, Banfi A. Robust Angiogenesis and Arteriogenesis in the Skin of Diabetic Mice by Transient Delivery of Engineered VEGF and PDGF-BB Proteins in Fibrin Hydrogels. Front Bioeng Biotechnol 2021; 9:688467. [PMID: 34277588 PMCID: PMC8281302 DOI: 10.3389/fbioe.2021.688467] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/02/2021] [Indexed: 12/20/2022] Open
Abstract
Non-healing ulcers are a serious complication of diabetes mellitus and a major unmet medical need. A major cause for the lack of healing is the impairment of spontaneous vascularization in the skin, despite mostly normal blood flow in deeper large vessels. Therefore, pro-angiogenic treatments are needed to increase therapeutic perfusion by recruiting new arterial connections (therapeutic arteriogenesis). Vascular endothelial growth factor (VEGF) is the master regulator of angiogenesis in physiology and disease, but exploitation of its therapeutic potential requires careful control of its dose distribution in tissue. Co-delivery of platelet derived growth factor-BB (PDGF-BB) has been shown to expand the therapeutic window of VEGF and also improve associated arteriogenesis. We used a highly controlled protein delivery system, based on a clinically applicable fibrin-based platform, to investigate the angiogenic and arteriogenic potential of engineered versions (TG-) of VEGF and PDGF-BB proteins in the skin of diabetic and obese db/db mice. Intradermal delivery of therapeutically relevant doses of TG-VEGF and TG-PDGF-BB induced robust growth of new microvascular networks with similar efficacy as in normal littermate control mice. Further, TG-PDGF-BB prevented the formation of aberrant vascular enlargements by high TG-VEGF levels. As fibrin was degraded after the first week, the induced angiogenesis mostly regressed by 4 weeks, but it promoted effective arteriogenesis in the dermal layer. Therefore, controlled co-delivery of TG-VEGF and TG-PDGF-BB recombinant proteins is effective to induce angiogenesis and arteriogenesis in diabetic mouse skin and should be further investigated to promote diabetic wound healing.
Collapse
Affiliation(s)
- Alessandro Certelli
- Cell and Gene Therapy, Department of Biomedicine, University Hospital of Basel, University of Basel, Basel, Switzerland
| | - Paolo Valente
- Cell and Gene Therapy, Department of Biomedicine, University Hospital of Basel, University of Basel, Basel, Switzerland.,Vascular Surgery, Department of Surgery, University Hospital of Basel, University of Basel, Basel, Switzerland
| | - Andrea Uccelli
- Cell and Gene Therapy, Department of Biomedicine, University Hospital of Basel, University of Basel, Basel, Switzerland
| | - Andrea Grosso
- Cell and Gene Therapy, Department of Biomedicine, University Hospital of Basel, University of Basel, Basel, Switzerland
| | - Nunzia Di Maggio
- Cell and Gene Therapy, Department of Biomedicine, University Hospital of Basel, University of Basel, Basel, Switzerland
| | - Rosalinda D'Amico
- Cell and Gene Therapy, Department of Biomedicine, University Hospital of Basel, University of Basel, Basel, Switzerland.,Vascular Surgery, Department of Surgery, University Hospital of Basel, University of Basel, Basel, Switzerland
| | - Priscilla S Briquez
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Jeffrey A Hubbell
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Thomas Wolff
- Vascular Surgery, Department of Surgery, University Hospital of Basel, University of Basel, Basel, Switzerland
| | - Lorenz Gürke
- Vascular Surgery, Department of Surgery, University Hospital of Basel, University of Basel, Basel, Switzerland
| | - Edin Mujagic
- Vascular Surgery, Department of Surgery, University Hospital of Basel, University of Basel, Basel, Switzerland
| | - Roberto Gianni-Barrera
- Cell and Gene Therapy, Department of Biomedicine, University Hospital of Basel, University of Basel, Basel, Switzerland
| | - Andrea Banfi
- Cell and Gene Therapy, Department of Biomedicine, University Hospital of Basel, University of Basel, Basel, Switzerland.,Vascular Surgery, Department of Surgery, University Hospital of Basel, University of Basel, Basel, Switzerland
| |
Collapse
|
21
|
Ganta VC, Annex BH. Peripheral vascular disease: preclinical models and emerging therapeutic targeting of the vascular endothelial growth factor ligand-receptor system. Expert Opin Ther Targets 2021; 25:381-391. [PMID: 34098826 PMCID: PMC8573823 DOI: 10.1080/14728222.2021.1940139] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 06/04/2021] [Indexed: 10/21/2022]
Abstract
Introduction: Vascular endothelial growth factor (VEGF)-A is a sought therapeutic target for PAD treatment because of its potent role in angiogenesis. However, no therapeutic benefit was achieved in VEGF-A clinical trials, suggesting that our understanding of VEGF-A biology and ischemic angiogenic processes needs development. Alternate splicing in VEGF-A produces pro- and anti-angiogenic VEGF-A isoforms; the only difference being a 6-amino acid switch in the C-terminus of the final 8th exon of the gene. This finding has changed our understanding of VEGF-A biology and may explain the lack of benefit in VEGF-A clinical trials. It presents new therapeutic opportunities for peripheral arterial disease (PAD) treatment.Areas covered: Literature search was conducted to include: 1) predicted mechanism by which the anti-angiogenic VEGF-A isoform would inhibit angiogenesis, 2) unexpected mechanism of action, and 3) how this mechanism revealed novel signaling pathways that may enhance future therapeutics in PAD.Expert opinion: Inhibiting a specific anti-angiogenic VEGF-A isoform in ischemic muscle promotes perfusion recovery in preclinical PAD. Additional efforts focused on the production of these isoforms, and the pathways altered by modulating different VEGF receptor-ligand interactions, and how this new data may allow bedside progress offers new approaches to PAD are discussed.I.
Collapse
Affiliation(s)
- Vijay Chaitanya Ganta
- Department of Medicine and Vascular Biology Center, Augusta University, Augusta, GA, USA
| | - Brian H Annex
- Department of Medicine and Vascular Biology Center, Augusta University, Augusta, GA, USA
| |
Collapse
|
22
|
Huang HL, Kuo CS, Chang TY, Chou RH, Chen IC, Yang FC, Chen NJ, Lin SJ, Wu CC, Huang PH. An oral absorbent, AST-120, restores vascular growth and blood flow in ischemic muscles in diabetic mice via modulation of macrophage transition. J Mol Cell Cardiol 2021; 155:99-110. [PMID: 33713645 DOI: 10.1016/j.yjmcc.2021.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 02/06/2021] [Accepted: 03/04/2021] [Indexed: 10/21/2022]
Abstract
Background Diabetes has a pronounced effect on the peripheral vasculature. The accumulation of advanced glycation end products (AGEs) is regarded as the crucial mechanism responsible for vascular damage in diabetes, but it is not easy to be avoided from food. In this study, we aimed to investigate the effects of an oral absorbent, AST-120, on the accumulation of AGEs and changes in blood flow recovery in diabetic mice. Methods The mice were divided into four groups, wild-type (WT) mice without treatment, WT mice treated with 5% AST-120 mixed into pulverized chow, streptozotocin-induced diabetes mellitus (DM) mice, and DM mice treated with 5% AST-120. Six weeks after hind-limb ischemia surgery, blood flow reperfusion, histology, plasma AGE, and cytokine were examined. Bone marrow cells were cultured and derived into macrophages to evaluate the effects of AGEs on macrophage polarization. Results Plasma AGEs were significantly increased in diabetic mice. AST-120 could bind to AGEs and reduced their plasma concentrations. Histological analysis revealed fewer collateral vessels with corresponding impairment of blood flow recovery in diabetic mice. In these mice, AGE-positive and AGE receptor-positive macrophages were numerous in ischemic limbs compared with non- diabetic mice. In diabetic mice, macrophages in ischemic tissues demonstrated greater M1 polarization than M2 polarization; this pattern was reversed in the AST-120 treatment group. The change in macrophage polarization was associated with the corresponding expression of pro-inflammatory cytokines in the ischemic tissues. In cell cultures, AGEs triggered the transformation of bone marrow-derived macrophages into the M1 phenotype. The alterations in the polarization of macrophages were reversed after treatment with AST-120. Conclusions Oral administration of AST-120 decreased the serum levels of AGEs in diabetic mice and improved neovascularization of ischemic limbs. This benefit may be due to, at least partially, the alterations in macrophage polarization and the associated changes in inflammatory cytokines.
Collapse
Affiliation(s)
- Hsin-Lei Huang
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; National Taipei University of Nursing and Health Sciences, Taiwan
| | - Chin-Sung Kuo
- Division of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ting-Yung Chang
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Cardiovascular Research Center, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ruey-Hsing Chou
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Cardiovascular Research Center, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - I-Chun Chen
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Fu-Chen Yang
- Institute of Microbiology and Immunology, School of Life Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Nien-Jung Chen
- Institute of Microbiology and Immunology, School of Life Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan; Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shing-Jong Lin
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan; Cardiovascular Research Center, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Taipei Medical University, Taipei, Taiwan; Division of Cardiology, Heart Center, Cheng-Hsin General Hospital, Taipei, Taiwan
| | - Chih-Cheng Wu
- National Tsing-Hua University, Institute of Biomedical Engineering, Hsinchu, Taiwan; Cardiovascular Center, National Taiwan University Hospital, Hsinchu Branch, Taipei, Taiwan; National Taiwan University Hospital, College of Medicine, Taipei, Taiwan; Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan.
| | - Po-Hsun Huang
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Cardiovascular Research Center, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
23
|
Sabzevari Rad R, Shirvani H, Mahmoodzadeh Hosseini H, Shamsoddini A, Samadi M. Micro RNA-126 promoting angiogenesis in diabetic heart by VEGF/Spred-1/Raf-1 pathway: effects of high-intensity interval training. J Diabetes Metab Disord 2021; 19:1089-1096. [PMID: 33520826 DOI: 10.1007/s40200-020-00610-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 08/06/2020] [Indexed: 12/20/2022]
Abstract
Purpose This study aims to investigate the effect of high-intensity interval training (HIIT) on gene expression of MicroRNA-126 (miR-126) and serum concentration of vascular endothelial growth factor/ sprouty related EVH1 domain containing 1/ rapidly accelerated fibrosarcoma 1 (VEGF/Spred-1/Raf-1) proteins effective in cardiac tissue angiogenesis of diabetic rats. Methods Forty male Wistar rats were randomly divided into four groups of healthy control (HC), diabetic control (DC), diabetic with HIIT training (DT), and healthy with HIIT training (HT). HIIT was performed 6 days per week for 6 weeks (with the overload). Diabetes was induced via the combination of intraperitoneal injection of streptozotocin and high-fat foods. Results Diabetes remarkably diminished the expressions of miR-126, VEGF and Raf-1 proteins, and augmented Spred-1 expression. Meanwhile, the implementation of HIIT gave rise to a significant enhancement in expression of miR-126 heart tissue (P < 0.01), and subsequently increased the expression of VEGF and Raf-1 proteins (P < 0.01), and declined Spred-1 expression (P < 0.01) in the training group compared to the control group. Conclusion The results of this study show that HIIT increases the expression of miR-126 by activating the angiogenesis pathway of the heart tissue. Increased angiogenesis through the miR-126 pathway is vital to compensate for heart destruction induced by diabetes. Thus, the use of standard interval exercise can be introduced as a novel therapeutic target for diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Reza Sabzevari Rad
- Exercise Physiology Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hossein Shirvani
- Exercise Physiology Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hamideh Mahmoodzadeh Hosseini
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Alireza Shamsoddini
- Exercise Physiology Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Samadi
- Exercise Physiology Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Abstract
This review takes an inclusive approach to microvascular dysfunction in diabetes mellitus and cardiometabolic disease. In virtually every organ, dynamic interactions between the microvasculature and resident tissue elements normally modulate vascular and tissue function in a homeostatic fashion. This regulation is disordered by diabetes mellitus, by hypertension, by obesity, and by dyslipidemia individually (or combined in cardiometabolic disease), with dysfunction serving as an early marker of change. In particular, we suggest that the familiar retinal, renal, and neural complications of diabetes mellitus are late-stage manifestations of microvascular injury that begins years earlier and is often abetted by other cardiometabolic disease elements (eg, hypertension, obesity, dyslipidemia). We focus on evidence that microvascular dysfunction precedes anatomic microvascular disease in these organs as well as in heart, muscle, and brain. We suggest that early on, diabetes mellitus and/or cardiometabolic disease can each cause reversible microvascular injury with accompanying dysfunction, which in time may or may not become irreversible and anatomically identifiable disease (eg, vascular basement membrane thickening, capillary rarefaction, pericyte loss, etc.). Consequences can include the familiar vision loss, renal insufficiency, and neuropathy, but also heart failure, sarcopenia, cognitive impairment, and escalating metabolic dysfunction. Our understanding of normal microvascular function and early dysfunction is rapidly evolving, aided by innovative genetic and imaging tools. This is leading, in tissues like the retina, to testing novel preventive interventions at early, reversible stages of microvascular injury. Great hope lies in the possibility that some of these interventions may develop into effective therapies.
Collapse
Affiliation(s)
- William B Horton
- Division of Endocrinology and Metabolism, Department of Medicine
| | - Eugene J Barrett
- Division of Endocrinology and Metabolism, Department of Medicine
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia
| |
Collapse
|
25
|
Morifuji T, Tanaka M, Nakanishi R, Hirabayashi T, Kondo H, Fujino H. Preventive effects of low-intensity endurance exercise for severe hyperglycemia-induced capillary regression in non-obese type 2 diabetes rat skeletal muscle. Physiol Rep 2021; 9:e14712. [PMID: 33463898 PMCID: PMC7814498 DOI: 10.14814/phy2.14712] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 12/10/2020] [Accepted: 12/12/2020] [Indexed: 12/12/2022] Open
Abstract
Although endurance exercise is effective for reducing diabetes‐related capillary regression, it is difficult to prescribe high‐intensity endurance exercise due to the potential worsening of complications in patients with severe hyperglycemia. Therefore, this study aimed to examine whether chronic low‐intensity exercise training may prevent severe hyperglycemia‐induced capillary regression of skeletal muscle in non‐obese type 2 diabetes. Non‐diabetic Sprague Dawley rats were assigned to a control (Con) group and an exercise (Ex) group. Likewise, spontaneously diabetic Torii rats were assigned to a diabetic sedentary (DM) group or a diabetic exercise (DMEx) group. Rats in the Ex and DMEx groups were placed on a motor‐driven treadmill running at low speed (15 m/min) for 60 min/day, 5 days/week, for 14 weeks. Serum glucose levels were significantly increased in the DM group, but not in the DMEx group. Although the capillary‐to‐fiber ratio in the plantaris muscle was significantly lower in the DM group compared to the control group, the ratio in the DMEx group was significantly higher compared to the DM group. Moreover, the succinate dehydrogenase activity and expression levels of vascular endothelial growth factor and peroxisome proliferator‐activated receptor γ coactivator‐1α (PGC‐1α) were reduced in the plantaris muscle of the DM group. However, those in the DMEx group were significantly higher than those in the DM group. These results indicate that low‐intensity chronic endurance exercise training has the potential to prevent the progression of capillary regression in the skeletal muscles of non‐obese type 2 diabetes patients with severe hyperglycemia.
Collapse
Affiliation(s)
- Takeshi Morifuji
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan.,Department of Physical Therapy, Josai International University, Tougane, Japan
| | - Minoru Tanaka
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan.,Department of Rehabilitation Science, Osaka Health Science University, Osaka, Japan
| | - Ryosuke Nakanishi
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan.,Department of Physical Therapy, Faculty of Rehabilitation, Kobe international University, Kobe, Japan
| | - Takumi Hirabayashi
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Hiroyo Kondo
- Department of Food Science and Nutrition, Nagoya Women's University, Nagoya, Japan
| | - Hidemi Fujino
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| |
Collapse
|
26
|
Mercier C, Rousseau M, Geraldes P. Growth Factor Deregulation and Emerging Role of Phosphatases in Diabetic Peripheral Artery Disease. Front Cardiovasc Med 2021; 7:619612. [PMID: 33490120 PMCID: PMC7817696 DOI: 10.3389/fcvm.2020.619612] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/10/2020] [Indexed: 01/25/2023] Open
Abstract
Peripheral artery disease is caused by atherosclerosis of lower extremity arteries leading to the loss of blood perfusion and subsequent critical ischemia. The presence of diabetes mellitus is an important risk factor that greatly increases the incidence, the progression and the severity of the disease. In addition to accelerated disease progression, diabetic patients are also more susceptible to develop serious impairment of their walking abilities through an increased risk of lower limb amputation. Hyperglycemia is known to alter the physiological development of collateral arteries in response to ischemia. Deregulation in the production of several critical pro-angiogenic factors has been reported in diabetes along with vascular cell unresponsiveness in initiating angiogenic processes. Among the multiple molecular mechanisms involved in the angiogenic response, protein tyrosine phosphatases are potent regulators by dephosphorylating pro-angiogenic tyrosine kinase receptors. However, evidence has indicated that diabetes-induced deregulation of phosphatases contributes to the progression of several micro and macrovascular complications. This review provides an overview of growth factor alterations in the context of diabetes and peripheral artery disease, as well as a description of the role of phosphatases in the regulation of angiogenic pathways followed by an analysis of the effects of hyperglycemia on the modulation of protein tyrosine phosphatase expression and activity. Knowledge of the role of phosphatases in diabetic peripheral artery disease will help the development of future therapeutics to locally regulate phosphatases and improve angiogenesis.
Collapse
Affiliation(s)
- Clément Mercier
- Department of Medicine, Division of Endocrinology, Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Marina Rousseau
- Department of Medicine, Division of Endocrinology, Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Pedro Geraldes
- Department of Medicine, Division of Endocrinology, Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
27
|
UZUNER B, KETENCİ S, SALBAŞ E. Diyabetik Nöropatiye Genel Yaklaşım. ACTA MEDICA ALANYA 2020. [DOI: 10.30565/medalanya.788960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
28
|
|
29
|
Hopfner U, Maan ZN, Hu MS, Aitzetmüller MM, Zaussinger M, Kirsch M, Machens HG, Duscher D. Deferoxamine enhances the regenerative potential of diabetic Adipose Derived Stem Cells. J Plast Reconstr Aesthet Surg 2020; 73:1738-1746. [PMID: 32418841 DOI: 10.1016/j.bjps.2020.02.045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 12/05/2019] [Accepted: 02/16/2020] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Diabetes mellitus remains a significant public health problem, consuming over $400 billion every year. While Diabetes itself can be controlled effectively, impaired wound healing still occurs frequently in diabetic patients. Adipose-derived mesenchymal stem cells (ASCs) provide an especially appealing source for diabetic wound cell therapy. With autologous approaches, the functionality of ASCs largely underlie patient-dependent factors. Diabetes is a significant diminishing factor of MSC functionality. Here, we explore a novel strategy to enhance diabetic ASC functionality through deferoxamine (DFO) preconditioning. MATERIAL AND METHODS Human diabetic ASCs have been preconditioned with 150 µM and 300 µM DFO in vitro and analyzed for regenerative cytokine expression. Murine diabetic ASCs have been preconditioned with 150 µM DFO examined for their in vitro and in vivo vasculogenic capacity in Matrigel assays. Additionally, a diabetic murine wound healing model has been performed to assess the regenerative capacity of preconditioned cells. RESULTS DFO preconditioning enhances the VEGF expression of human diabetic ASCs through hypoxia-inducible factor upregulation. The use of 150 µM of DFO was an optimal concentration to induce regenerative effects. The vasculogenic potential of preconditioned diabetic ASCs is significantly greater in vitro and in vivo. The enhanced regenerative functionality of DFO preconditioned ASCs was further confirmed in a model of diabetic murine wound healing. CONCLUSION These results demonstrate that DFO significantly induced the upregulation of hypoxia-inducible factor-1 alpha and VEGF in diabetic ASCs and showed efficacy in the treatment of diabetes-associated deficits of wound healing. The favorable status of DFO as a small molecule drug approved since decades for multiple indications makes this approach highly translatable.
Collapse
Affiliation(s)
- Ursula Hopfner
- Department for Plastic Surgery and Hand Surgery, Klinikum rechts der Isar, Technical University of Munich, Germany
| | - Zeshaan N Maan
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Michael S Hu
- Department for Plastic Surgery, University of Pittsburgh, Pennsylvania, USA
| | - Matthias M Aitzetmüller
- Department for Plastic Surgery and Hand Surgery, Klinikum rechts der Isar, Technical University of Munich, Germany
| | - Maximilian Zaussinger
- Department for Plastic Surgery and Hand Surgery, Klinikum rechts der Isar, Technical University of Munich, Germany
| | - Manuela Kirsch
- Department for Plastic Surgery and Hand Surgery, Klinikum rechts der Isar, Technical University of Munich, Germany
| | - Hans-Günther Machens
- Department for Plastic Surgery and Hand Surgery, Klinikum rechts der Isar, Technical University of Munich, Germany
| | - Dominik Duscher
- Department for Plastic Surgery and Hand Surgery, Klinikum rechts der Isar, Technical University of Munich, Germany; Division for Plastic and Reconstructive Surgery, Kepler University Hospital, Linz, Austria.
| |
Collapse
|
30
|
Robillard S, Mercier C, Breton V, Paquin-Veillette J, Guay A, Lizotte F, Geraldes P. Ablation of angiotensin type 2 receptor prevents endothelial nitric oxide synthase glutathionylation and nitration in ischaemic abductor muscle of diabetic mice. Diab Vasc Dis Res 2020; 17:1479164119883978. [PMID: 31726870 PMCID: PMC7510371 DOI: 10.1177/1479164119883978] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Peripheral artery disease is a severe complication of diabetes. We have reported that the deletion of angiotensin type 2 receptor in diabetic mice promoted vascular angiogenesis in the ischaemic muscle 4 weeks following ischaemia. However, the angiotensin type 2 receptor deletion beneficial effects occurred 2 weeks post surgery suggesting that angiotensin type 2 receptor may regulate other pro-angiogenic signalling pathways during the early phases of ischaemia. Nondiabetic and diabetic angiotensin type 2 receptor-deficient mice (Agtr2-/Y) underwent femoral artery ligation after 2 months of diabetes. Blood perfusion was measured every week up to 2 weeks post surgery. Expression of vascular endothelial growth factor, vascular endothelial growth factor receptor and endothelial nitric oxide synthase expression and activity were evaluated. Blood flow reperfusion in the ischaemic muscle of diabetic Agtr2+/Y mice was recovered at 35% as compared to a 68% recovery in diabetic Agtr2-/Y mice. The expression of vascular endothelial growth factor and its receptors was diminished in diabetic Agtr2+/Y mice, an observation not seen in diabetic Agtr2-/Y mice. Interestingly, Agtr2-/Y mice were protected from diabetes-induced glutathionylation, nitration and decreased endothelial nitric oxide synthase expression, which correlated with reduced endothelial cell death and enhanced vascular density in diabetic ischaemic muscle. In conclusion, our results suggest that the deletion of angiotensin type 2 receptor promotes blood flow reperfusion in diabetes by favouring endothelial cell survival and function.
Collapse
MESH Headings
- Animals
- Apoptosis
- Blood Flow Velocity
- Cattle
- Cells, Cultured
- Diabetes Mellitus/enzymology
- Diabetes Mellitus/genetics
- Disease Models, Animal
- Endothelial Cells/enzymology
- Endothelial Cells/pathology
- Glutathione/metabolism
- Hindlimb
- Ischemia/enzymology
- Ischemia/genetics
- Ischemia/physiopathology
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Skeletal/blood supply
- Neovascularization, Physiologic
- Nitrates/metabolism
- Nitric Oxide Synthase Type III/metabolism
- Peripheral Arterial Disease/enzymology
- Peripheral Arterial Disease/genetics
- Peripheral Arterial Disease/physiopathology
- Protein Processing, Post-Translational
- Receptor, Angiotensin, Type 2/deficiency
- Receptor, Angiotensin, Type 2/genetics
- Recovery of Function
- Regional Blood Flow
Collapse
Affiliation(s)
- Stéphanie Robillard
- Research Center of the Centre
Hospitalier Universitaire de Sherbrooke, Québec, Canada
| | - Clément Mercier
- Research Center of the Centre
Hospitalier Universitaire de Sherbrooke, Québec, Canada
| | - Valérie Breton
- Research Center of the Centre
Hospitalier Universitaire de Sherbrooke, Québec, Canada
| | | | - Andréanne Guay
- Research Center of the Centre
Hospitalier Universitaire de Sherbrooke, Québec, Canada
| | - Farah Lizotte
- Research Center of the Centre
Hospitalier Universitaire de Sherbrooke, Québec, Canada
| | - Pedro Geraldes
- Research Center of the Centre
Hospitalier Universitaire de Sherbrooke, Québec, Canada
- Division of Endocrinology, Department of
Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada
- Pedro Geraldes, Division of Endocrinology,
Department of Medicine, Université de Sherbrooke, 3001 12e Ave Nord, Sherbrooke,
QC J1H 5N4, Canada.
| |
Collapse
|
31
|
Tamari T, Elimelech R, Cohen G, Cohen T, Doppelt O, Eskander-Hashoul L, Zigdon-Giladi H. Endothelial Progenitor Cells inhibit jaw osteonecrosis in a rat model: A major adverse effect of bisphosphonate therapy. Sci Rep 2019; 9:18896. [PMID: 31827217 PMCID: PMC6906486 DOI: 10.1038/s41598-019-55383-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 11/22/2019] [Indexed: 01/27/2023] Open
Abstract
Medication-related osteonecrosis of the jaw (MRONJ) is a serious adverse effect of antiresorptive and antiangiogenic therapies. MRONJ is identified by chronic wounds in the oral mucosa associated with exposed necrotic bone. We hypothesized that zoledronic acid (ZOL) impairs keratinocyte and fibroblast function and reduces soft tissue vascularization; therefore, treating MRONJ with proangiogenic cells may benefit MRONJ patients. The effect of ZOL and dexamethasone (DEX) on gingival fibroblasts and keratinocytes was investigated. In-vitro, ZOL inhibited fibroblast and keratinocyte proliferation, delaying scratch healing. In-vivo, exposed bone was detected at tooth extraction sites, mainly in ZOL(+)/DEX(+) rats; and was associated with significantly decreased soft tissue vascularization, serum-VEGF, and tissue-VEGF. Local injection of early and late endothelial progenitor cells (EPCs) healed 13 of 14 MRONJ lesions compared with 2/7 lesions in the mesenchymal stem cells, and 2/6, in culture-medium group. The EPCs reduced necrotic bone area, increased serum and tissue VEGF levels. EPCs engraftment was minimal, suggesting their paracrine role in MRONJ healing. The EPC-conditioned medium improved scratch healing of keratinocytes and fibroblasts via VEGF pathway and elevated mRNA of VEGFA and collagen1A1. In conclusion, a novel MRONJ treatment with EPCs, increased vascularization and improved epithelial and fibroblast functions as well as cured the lesion.
Collapse
Affiliation(s)
- Tal Tamari
- Laboratory for Bone Repair, Rambam Health Care Campus, Haifa, Israel
| | - Rina Elimelech
- Department of Periodontology, School of Graduate Dentistry, Rambam Health Care Campus, Haifa, Israel.,Laboratory for Bone Repair, Rambam Health Care Campus, Haifa, Israel
| | - Gal Cohen
- Laboratory for Bone Repair, Rambam Health Care Campus, Haifa, Israel
| | - Talia Cohen
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Ofri Doppelt
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Lana Eskander-Hashoul
- Department of Periodontology, School of Graduate Dentistry, Rambam Health Care Campus, Haifa, Israel.,Laboratory for Bone Repair, Rambam Health Care Campus, Haifa, Israel
| | - Hadar Zigdon-Giladi
- Department of Periodontology, School of Graduate Dentistry, Rambam Health Care Campus, Haifa, Israel. .,Laboratory for Bone Repair, Rambam Health Care Campus, Haifa, Israel. .,The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
32
|
Alleboina S, Ayalew D, Peravali R, Chen L, Wong T, Dokun AO. Dual specificity phosphatase 5 regulates perfusion recovery in experimental peripheral artery disease. Vasc Med 2019; 24:395-404. [PMID: 31451089 DOI: 10.1177/1358863x19866254] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Peripheral artery disease (PAD) is caused by atherosclerotic occlusions of vessels outside the heart, particularly those of the lower extremities. Angiogenesis is one critical physiological response to vessel occlusion in PAD, but our understanding of the molecular mechanisms involved in angiogenesis is incomplete. Dual specificity phosphatase 5 (DUSP5) has been shown to play a key role in embryonic vascular development, but its role in post-ischemic angiogenesis is not known. We induced hind limb ischemia in mice and found robust upregulation of Dusp5 expression in ischemic hind limbs. Moreover, in vivo knockdown of Dusp5 resulted in impaired perfusion recovery in ischemic limbs and was associated with increased limb necrosis. In vitro studies showed upregulation of DUSP5 in human endothelial cells exposed to ischemia, and knockdown of DUSP5 in these ischemic endothelial cells resulted in impaired endothelial cell proliferation and angiogenesis, but did not alter apoptosis. Finally, we show that these effects of DUSP5 on post-ischemic angiogenesis are a result of DUSP5-dependent decrease in ERK1/2 phosphorylation and p21 protein expression. Thus, we have identified a role of DUSP5 in post-ischemic angiogenesis and implicated a DUSP5-ERK-p21 pathway that may serve as a therapeutic target for the modulation of post-ischemic angiogenesis in PAD.
Collapse
Affiliation(s)
- Satyanarayana Alleboina
- Division of Endocrinology, Diabetes and Metabolism, Health Sciences Center, University of Tennessee, Memphis, TN, USA
| | - Dawit Ayalew
- Division of Endocrinology, Diabetes and Metabolism, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Rahul Peravali
- Division of Endocrinology, Diabetes and Metabolism, Health Sciences Center, University of Tennessee, Memphis, TN, USA
| | - Lingdan Chen
- Division of Endocrinology, Diabetes and Metabolism, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Thomas Wong
- Division of Endocrinology, Diabetes and Metabolism, Carver School of Medicine, University of Iowa, Iowa City, IA, USA
| | - Ayotunde O Dokun
- Division of Endocrinology, Diabetes and Metabolism, Carver School of Medicine, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
33
|
Glucose impairs angiogenesis and promotes ventricular remodelling following myocardial infarction via upregulation of microRNA-17. Exp Cell Res 2019; 381:191-200. [DOI: 10.1016/j.yexcr.2019.04.039] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 04/27/2019] [Accepted: 04/30/2019] [Indexed: 01/07/2023]
|
34
|
Yuan J, Tan JTM, Rajamani K, Solly EL, King EJ, Lecce L, Simpson PJL, Lam YT, Jenkins AJ, Bursill CA, Keech AC, Ng MKC. Fenofibrate Rescues Diabetes-Related Impairment of Ischemia-Mediated Angiogenesis by PPARα-Independent Modulation of Thioredoxin-Interacting Protein. Diabetes 2019; 68:1040-1053. [PMID: 30765336 DOI: 10.2337/db17-0926] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 02/07/2019] [Indexed: 12/18/2022]
Abstract
Fenofibrate, a peroxisome proliferator-activated receptor α (PPARα) agonist, reduces lower limb amputations in patients with type 2 diabetes. The mechanism is, however, unknown. In this study, we demonstrate that fenofibrate markedly attenuates diabetes-related impairment of ischemia-mediated angiogenesis. In a murine model of hindlimb ischemia, daily oral fenofibrate treatment restored diabetes-impaired blood flow recovery, foot movement, hindlimb capillary density, vessel diameter, and vascular endothelial growth factor signaling to nondiabetic levels in both wild-type and PPARα-knockout mice, indicating that these fenofibrate effects are largely PPARα independent. In vitro, fenofibric acid (FFA) rescued high glucose-induced (25 mmol/L) impairment of endothelial cell migration, tubulogenesis, and survival in a PPARα-independent manner. Interestingly, fenofibrate in vivo and FFA in vitro reversed high glucose-induced expression of thioredoxin-interacting protein (TXNIP), an exquisitely glucose-inducible gene previously identified as a critical mediator of diabetes-related impairment in neovascularization. Conversely, adenoviral overexpression of TXNIP abrogated the restorative effects of FFA on high glucose-impaired endothelial cell function in vitro, indicating that the effects of FFA are mediated by TXNIP. We conclude that fenofibrate rescues diabetic impairment in ischemia-mediated angiogenesis, in large part, by PPARα-independent regulation of TXNIP. These findings may therefore explain the reduction in amputations seen in patients with diabetes treated with fenofibrate.
Collapse
Affiliation(s)
- Jun Yuan
- Heart Research Institute, Newtown, New South Wales, Australia
- Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Joanne T M Tan
- Heart Research Institute, Newtown, New South Wales, Australia
- Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- Faculty of Health and Medical Sciences, Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Kushwin Rajamani
- Heart Research Institute, Newtown, New South Wales, Australia
- Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
- NHMRC Clinical Trials Centre, Camperdown, New South Wales, Australia
| | - Emma L Solly
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Emily J King
- Heart Research Institute, Newtown, New South Wales, Australia
| | - Laura Lecce
- Heart Research Institute, Newtown, New South Wales, Australia
| | | | - Yuen Ting Lam
- Heart Research Institute, Newtown, New South Wales, Australia
- Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Alicia J Jenkins
- Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
- NHMRC Clinical Trials Centre, Camperdown, New South Wales, Australia
| | - Christina A Bursill
- Heart Research Institute, Newtown, New South Wales, Australia
- Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- Faculty of Health and Medical Sciences, Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Anthony C Keech
- Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
- NHMRC Clinical Trials Centre, Camperdown, New South Wales, Australia
- Cardiology Department, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Martin K C Ng
- Heart Research Institute, Newtown, New South Wales, Australia
- Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
- Cardiology Department, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| |
Collapse
|
35
|
Crosby CO, Zoldan J. Mimicking the physical cues of the ECM in angiogenic biomaterials. Regen Biomater 2019; 6:61-73. [PMID: 30967961 PMCID: PMC6447000 DOI: 10.1093/rb/rbz003] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 12/02/2018] [Accepted: 12/29/2018] [Indexed: 12/12/2022] Open
Abstract
A functional microvascular system is imperative to build and maintain healthy tissue. Impaired microvasculature results in ischemia, thereby limiting the tissue's intrinsic regeneration capacity. Therefore, the ability to regenerate microvascular networks is key to the development of effective cardiovascular therapies. To stimulate the formation of new microvasculature, researchers have focused on fabricating materials that mimic the angiogenic properties of the native extracellular matrix (ECM). Here, we will review biomaterials that seek to imitate the physical cues that are natively provided by the ECM to encourage the formation of microvasculature in engineered constructs and ischemic tissue in the body.
Collapse
Affiliation(s)
- Cody O Crosby
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Janet Zoldan
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
36
|
Rudnicki M, Abdifarkosh G, Rezvan O, Nwadozi E, Roudier E, Haas TL. Female Mice Have Higher Angiogenesis in Perigonadal Adipose Tissue Than Males in Response to High-Fat Diet. Front Physiol 2018; 9:1452. [PMID: 30405427 PMCID: PMC6206240 DOI: 10.3389/fphys.2018.01452] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 09/25/2018] [Indexed: 01/21/2023] Open
Abstract
Background: Impaired capillary growth (angiogenesis) in skeletal muscle and adipose tissue contributes to the development of metabolic disorders in obese males. This association remains unexplored in females, despite mounting evidence that endothelial cells have sex-specific transcriptional profiles. Therefore, herein we assessed whether males and females show distinct angiogenic capacities in response to diet-induced obesity. Methods: Age-matched male and female mice were fed normal chow or high-fat obesogenic diets for 16 weeks. At the end of diet period, systemic glucose disposal was assessed as well as insulin sensitivity of skeletal muscle and visceral adipose tissue. Capillary content and the expression of angiogenic regulators were also evaluated in these tissues. Results: When placed on a high-fat diet, female mice gained less weight than males and showed a metabolic phenotype similar to NC-fed mice, contrasting with the impaired whole-body glucose metabolism observed in high-fat-fed males. However, high-fat-feeding elevated serum lipid levels similarly in male and female mice. Although skeletal muscle of high-fat-fed female mice had higher insulin sensitivity than male counterparts, no sex difference was detected in muscle capillarization. Metabolic functions of perigonadal white adipose tissue (pgWAT) were retained in high-fat-fed females, as evidenced by smaller adipocytes with preserved insulin sensitivity, greater responsiveness to isoproterenol, higher expression of Adiponectin and a lower ratio of Leptin:Adiponectin mRNA. An enhanced browning phenotype was detected in HF-fed female adipocytes with upregulation of Ucp1 expression. PgWAT from high-fat-fed females also showed augmented capillary number and expression of endothelial cell markers, which was associated with elevated mRNA levels of pro-angiogenic mediators, including vascular endothelial growth factor A (Vegfa) and its receptor (Vegfr2), the Notch ligand Jagged-1 (Jag1) and Angiopoietin-2 (Angpt2). Conclusion: Taken together, our findings provide novel evidence that visceral adipose tissue of female mice display greater levels of pro-angiogenic factors and vascularity than males in response to high-fat diet. This phenotype is associated with preserved metabolic homeostasis at both tissue and systemic levels. Our study discloses that a thus-far-unappreciated sex-specific difference in the regulation of adipose angiogenesis may contribute to an individual's susceptibility to developing adipose dysfunction and obesity-related metabolic disturbances.
Collapse
Affiliation(s)
- Martina Rudnicki
- Angiogenesis Research Group, School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, ON, Canada
| | - Ghoncheh Abdifarkosh
- Angiogenesis Research Group, School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, ON, Canada
| | - Omid Rezvan
- Angiogenesis Research Group, School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, ON, Canada
| | - Emmanuel Nwadozi
- Angiogenesis Research Group, School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, ON, Canada
| | - Emilie Roudier
- Angiogenesis Research Group, School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, ON, Canada
| | - Tara L Haas
- Angiogenesis Research Group, School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, ON, Canada
| |
Collapse
|
37
|
Is there a Chance to Promote Arteriogenesis by DPP4 Inhibitors Even in Type 2 Diabetes? A Critical Review. Cells 2018; 7:cells7100181. [PMID: 30360455 PMCID: PMC6210696 DOI: 10.3390/cells7100181] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/08/2018] [Accepted: 10/18/2018] [Indexed: 12/18/2022] Open
Abstract
Cardiovascular diseases (CVD) are still the prevailing cause of death not only in industrialized countries, but even worldwide. Type 2 diabetes mellitus (type 2 DM) and hyperlipidemia, a metabolic disorder that is often associated with diabetes, are major risk factors for developing CVD. Recently, clinical trials proved the safety of gliptins in treating patients with type 2 DM. Gliptins are dipeptidyl-peptidase 4 (DPP4/CD26) inhibitors, which stabilize glucagon-like peptide-1 (GLP-1), thereby increasing the bioavailability of insulin. Moreover, blocking DPP4 results in increased levels of stromal cell derived factor 1 (SDF-1). SDF-1 has been shown in pre-clinical animal studies to improve heart function and survival after myocardial infarction, and to promote arteriogenesis, the growth of natural bypasses, compensating for the function of an occluded artery. Clinical trials, however, failed to demonstrate a superiority of gliptins compared to placebo treated type 2 DM patients in terms of cardiovascular (CV) outcomes. This review highlights the function of DPP4 inhibitors in type 2 DM, and in treating cardiovascular diseases, with special emphasis on arteriogenesis. It critically addresses the potency of currently available gliptins and gives rise to hope by pointing out the most relevant questions that need to be resolved.
Collapse
|
38
|
Hourigan ST, Solly EL, Nankivell VA, Ridiandries A, Weimann BM, Henriquez R, Tepper ER, Zhang JQJ, Tsatralis T, Clayton ZE, Vanags LZ, Robertson S, Nicholls SJ, Ng MKC, Bursill CA, Tan JTM. The regulation of miRNAs by reconstituted high-density lipoproteins in diabetes-impaired angiogenesis. Sci Rep 2018; 8:13596. [PMID: 30206364 PMCID: PMC6133943 DOI: 10.1038/s41598-018-32016-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 08/28/2018] [Indexed: 01/08/2023] Open
Abstract
Diabetic vascular complications are associated with impaired ischaemia-driven angiogenesis. We recently found that reconstituted high-density lipoproteins (rHDL) rescue diabetes-impaired angiogenesis. microRNAs (miRNAs) regulate angiogenesis and are transported within HDL to sites of injury/repair. The role of miRNAs in the rescue of diabetes-impaired angiogenesis by rHDL is unknown. Using a miRNA array, we found that rHDL inhibits hsa-miR-181c-5p expression in vitro and using a hsa-miR-181c-5p mimic and antimiR identify a novel anti-angiogenic role for miR-181c-5p. miRNA expression was tracked over time post-hindlimb ischaemic induction in diabetic mice. Early post-ischaemia when angiogenesis is important, rHDL suppressed hindlimb mmu-miR-181c-5p. mmu-miR-181c-5p was not detected in the plasma or within HDL, suggesting rHDL specifically targets mmu-miR-181c-5p at the ischaemic site. Three known angiogenic miRNAs (mmu-miR-223-3p, mmu-miR-27b-3p, mmu-miR-92a-3p) were elevated in the HDL fraction of diabetic rHDL-infused mice early post-ischaemia. This was accompanied by a decrease in plasma levels. Only mmu-miR-223-3p levels were elevated in the hindlimb 3 days post-ischaemia, indicating that rHDL regulates mmu-miR-223-3p in a time-dependent and site-specific manner. The early regulation of miRNAs, particularly miR-181c-5p, may underpin the rescue of diabetes-impaired angiogenesis by rHDL and has implications for the treatment of diabetes-related vascular complications.
Collapse
Affiliation(s)
- Samuel T Hourigan
- The Heart Research Institute, Sydney, Australia.,The University of Sydney, Sydney Medical School, Sydney, Australia
| | - Emma L Solly
- Heart Health Theme, South Australian Health & Medical Research Institute, Adelaide, Australia
| | - Victoria A Nankivell
- Heart Health Theme, South Australian Health & Medical Research Institute, Adelaide, Australia
| | - Anisyah Ridiandries
- The Heart Research Institute, Sydney, Australia.,The University of Sydney, Sydney Medical School, Sydney, Australia
| | - Benjamin M Weimann
- Heart Health Theme, South Australian Health & Medical Research Institute, Adelaide, Australia.,Adelaide Medical School, Faculty of Health & Medical Sciences, The University of Adelaide, Adelaide, Australia
| | | | - Edward R Tepper
- The Heart Research Institute, Sydney, Australia.,The University of Sydney, Sydney Medical School, Sydney, Australia
| | - Jennifer Q J Zhang
- The Heart Research Institute, Sydney, Australia.,The University of Sydney, Sydney Medical School, Sydney, Australia
| | | | - Zoe E Clayton
- The Heart Research Institute, Sydney, Australia.,The University of Sydney, Sydney Medical School, Sydney, Australia
| | - Laura Z Vanags
- The Heart Research Institute, Sydney, Australia.,The University of Sydney, Sydney Medical School, Sydney, Australia
| | - Stacy Robertson
- The Heart Research Institute, Sydney, Australia.,The University of Sydney, Sydney Medical School, Sydney, Australia
| | - Stephen J Nicholls
- Heart Health Theme, South Australian Health & Medical Research Institute, Adelaide, Australia.,Adelaide Medical School, Faculty of Health & Medical Sciences, The University of Adelaide, Adelaide, Australia
| | - Martin K C Ng
- The Heart Research Institute, Sydney, Australia.,The University of Sydney, Sydney Medical School, Sydney, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Christina A Bursill
- The Heart Research Institute, Sydney, Australia.,The University of Sydney, Sydney Medical School, Sydney, Australia.,Heart Health Theme, South Australian Health & Medical Research Institute, Adelaide, Australia.,Adelaide Medical School, Faculty of Health & Medical Sciences, The University of Adelaide, Adelaide, Australia
| | - Joanne T M Tan
- The Heart Research Institute, Sydney, Australia. .,The University of Sydney, Sydney Medical School, Sydney, Australia. .,Heart Health Theme, South Australian Health & Medical Research Institute, Adelaide, Australia. .,Adelaide Medical School, Faculty of Health & Medical Sciences, The University of Adelaide, Adelaide, Australia.
| |
Collapse
|
39
|
Hedhli J, Slania SLL, Płoska A, Czerwinski A, Konopka CJ, Wozniak M, Banach M, Dobrucki IT, Kalinowski L, Dobrucki LW. Evaluation of a dimeric-cRGD peptide for targeted PET-CT imaging of peripheral angiogenesis in diabetic mice. Sci Rep 2018; 8:5401. [PMID: 29599497 PMCID: PMC5876368 DOI: 10.1038/s41598-018-23372-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 03/06/2018] [Indexed: 11/09/2022] Open
Abstract
The α V β3 integrin plays an important role in many physiological functions and pathological disorders. α V β3 is minimally expressed in normal quiescent endothelial cells, but significantly upregulated during neovascularization. In this study, we evaluated a 64Cu-labeled dimeric cRGD tracer targeted at α V β3 integrin and report its applicability to assess peripheral angiogenesis in diabetes mellitus (DM). We established a murine model of type-1 DM characterized by elevated glucose, glycated serum protein (GSP), and glycated hemoglobin A1c (HbA1c). We demonstrated that our imaging probe is specific to α V β3 integrin under both normo- and hyperglycemic conditions. We found that the analysis of in vivo PET-CT images correlated well with gamma well counting (GWC). Both GWC and PET-CT imaging demonstrated increased uptake of 64Cu-NOTA-PEG4-cRGD2 in the ischemic hindlimb in contrast to non-ischemic control. GWC of the distal ischemic tissue from DM mice showed significantly lower probe accumulation than in non-DM mice. The immunofluorescence staining of the ischemic tissues showed a 3-fold reduction in CD31 and 4-fold reduction in the α V β3 expression in DM vs. non-DM animals. In conclusion, we successfully demonstrated that diabetes-associated reductions in peripheral angiogenesis can be non-invasively detected with PET-CT imaging using targeted dimeric-cRGD probe.
Collapse
Affiliation(s)
- Jamila Hedhli
- Beckman Institute for Advanced Science and Technology, Urbana, IL, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Stephanie L L Slania
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Agata Płoska
- Beckman Institute for Advanced Science and Technology, Urbana, IL, USA
- Department of Medical Laboratory Diagnostics and Central Bank of Frozen Tissues & Genetic Specimens, Medical University of Gdansk, Gdansk, Poland
| | | | - Christian J Konopka
- Beckman Institute for Advanced Science and Technology, Urbana, IL, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Marcin Wozniak
- Beckman Institute for Advanced Science and Technology, Urbana, IL, USA
- Department of Medical Laboratory Diagnostics and Central Bank of Frozen Tissues & Genetic Specimens, Medical University of Gdansk, Gdansk, Poland
| | - Maciej Banach
- Department of Hypertension, Medical University of Lodz, Lodz, Poland
| | - Iwona T Dobrucki
- Beckman Institute for Advanced Science and Technology, Urbana, IL, USA
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics and Central Bank of Frozen Tissues & Genetic Specimens, Medical University of Gdansk, Gdansk, Poland
- Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.PL), Gdansk, Poland
| | - Lawrence W Dobrucki
- Beckman Institute for Advanced Science and Technology, Urbana, IL, USA.
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Department of Medical Laboratory Diagnostics and Central Bank of Frozen Tissues & Genetic Specimens, Medical University of Gdansk, Gdansk, Poland.
- Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.PL), Gdansk, Poland.
| |
Collapse
|
40
|
MacAskill MG, Saif J, Condie A, Jansen MA, MacGillivray TJ, Tavares AAS, Fleisinger L, Spencer HL, Besnier M, Martin E, Biglino G, Newby DE, Hadoke PWF, Mountford JC, Emanueli C, Baker AH. Robust Revascularization in Models of Limb Ischemia Using a Clinically Translatable Human Stem Cell-Derived Endothelial Cell Product. Mol Ther 2018; 26:1669-1684. [PMID: 29703701 PMCID: PMC6035339 DOI: 10.1016/j.ymthe.2018.03.017] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 03/26/2018] [Accepted: 03/26/2018] [Indexed: 12/21/2022] Open
Abstract
Pluripotent stem cell-derived differentiated endothelial cells offer high potential in regenerative medicine in the cardiovascular system. With the aim of translating the use of a human stem cell-derived endothelial cell product (hESC-ECP) for treatment of critical limb ischemia (CLI) in man, we report a good manufacturing practice (GMP)-compatible protocol and detailed cell tracking and efficacy data in multiple preclinical models. The clinical-grade cell line RC11 was used to generate hESC-ECP, which was identified as mostly endothelial (60% CD31+/CD144+), with the remainder of the subset expressing various pericyte/mesenchymal stem cell markers. Cell tracking using MRI, PET, and qPCR in a murine model of limb ischemia demonstrated that hESC-ECP was detectable up to day 7 following injection. Efficacy in several murine models of limb ischemia (immunocompromised/immunocompetent mice and mice with either type I/II diabetes mellitus) demonstrated significantly increased blood perfusion and capillary density. Overall, we demonstrate a GMP-compatible hESC-ECP that improved ischemic limb perfusion and increased local angiogenesis without engraftment, paving the way for translation of this therapy.
Collapse
Affiliation(s)
- Mark G MacAskill
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK; Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
| | - Jaimy Saif
- Experimental Cardiovascular Medicine Division, Bristol Heart Institute, University of Bristol, Bristol, UK
| | - Alison Condie
- Scottish National Blood Transfusion Service, Edinburgh, UK
| | - Maurits A Jansen
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK; Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
| | | | - Adriana A S Tavares
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK; Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
| | - Lucija Fleisinger
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Helen L Spencer
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Marie Besnier
- Experimental Cardiovascular Medicine Division, Bristol Heart Institute, University of Bristol, Bristol, UK
| | - Ernesto Martin
- Experimental Cardiovascular Medicine Division, Bristol Heart Institute, University of Bristol, Bristol, UK
| | - Giovanni Biglino
- Experimental Cardiovascular Medicine Division, Bristol Heart Institute, University of Bristol, Bristol, UK
| | - David E Newby
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Patrick W F Hadoke
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Joanne C Mountford
- Scottish National Blood Transfusion Service, Edinburgh, UK; Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Costanza Emanueli
- Experimental Cardiovascular Medicine Division, Bristol Heart Institute, University of Bristol, Bristol, UK; National Heart and Lung Institute, Imperial College London, London, UK
| | - Andrew H Baker
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
41
|
|
42
|
|
43
|
Sultan MT, Lee OJ, Kim SH, Ju HW, Park CH. Silk Fibroin in Wound Healing Process. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1077:115-126. [DOI: 10.1007/978-981-13-0947-2_7] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
44
|
Barrett EJ, Liu Z, Khamaisi M, King GL, Klein R, Klein BEK, Hughes TM, Craft S, Freedman BI, Bowden DW, Vinik AI, Casellini CM. Diabetic Microvascular Disease: An Endocrine Society Scientific Statement. J Clin Endocrinol Metab 2017; 102:4343-4410. [PMID: 29126250 PMCID: PMC5718697 DOI: 10.1210/jc.2017-01922] [Citation(s) in RCA: 327] [Impact Index Per Article: 40.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 08/29/2017] [Indexed: 01/18/2023]
Abstract
Both type 1 and type 2 diabetes adversely affect the microvasculature in multiple organs. Our understanding of the genesis of this injury and of potential interventions to prevent, limit, or reverse injury/dysfunction is continuously evolving. This statement reviews biochemical/cellular pathways involved in facilitating and abrogating microvascular injury. The statement summarizes the types of injury/dysfunction that occur in the three classical diabetes microvascular target tissues, the eye, the kidney, and the peripheral nervous system; the statement also reviews information on the effects of diabetes and insulin resistance on the microvasculature of skin, brain, adipose tissue, and cardiac and skeletal muscle. Despite extensive and intensive research, it is disappointing that microvascular complications of diabetes continue to compromise the quantity and quality of life for patients with diabetes. Hopefully, by understanding and building on current research findings, we will discover new approaches for prevention and treatment that will be effective for future generations.
Collapse
Affiliation(s)
- Eugene J. Barrett
- Division of Endocrinology, Department of Medicine, University of Virginia, Charlottesville, Virginia 22908
| | - Zhenqi Liu
- Division of Endocrinology, Department of Medicine, University of Virginia, Charlottesville, Virginia 22908
| | - Mogher Khamaisi
- Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts 02215
| | - George L. King
- Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts 02215
| | - Ronald Klein
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53705
| | - Barbara E. K. Klein
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53705
| | - Timothy M. Hughes
- Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Suzanne Craft
- Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Barry I. Freedman
- Divisions of Nephrology and Endocrinology, Department of Internal Medicine, Centers for Diabetes Research, and Center for Human Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Donald W. Bowden
- Divisions of Nephrology and Endocrinology, Department of Internal Medicine, Centers for Diabetes Research, and Center for Human Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Aaron I. Vinik
- EVMS Strelitz Diabetes Center, Eastern Virginia Medical Center, Norfolk, Virginia 23510
| | - Carolina M. Casellini
- EVMS Strelitz Diabetes Center, Eastern Virginia Medical Center, Norfolk, Virginia 23510
| |
Collapse
|
45
|
Tsui HY, Liu YC, Yan X, Lin Y, Xu Y, Tan Q. Combined effects of artificial dermis and vascular endothelial growth factor concentration gradient on wound healing in diabetic porcine model. Growth Factors 2017; 35:216-224. [PMID: 29447490 DOI: 10.1080/08977194.2018.1435532] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Wounds in patients with diabetes mellitus are one of the most prevalent impaired wounds in the world. Vascular endothelial growth factor (VEGF) is one of the most important proangiogenic mediators. Artificial dermal (AD) such as Pelnac® has been shown, in humans and animal models, a great therapeutic potential in full-thickness skin wounds. We attempt to promote the wound healing in diabetic porcine models through combined use of AD and constant concentration of VEGF or VEGF concentration gradient. We created full-thickness excisional wounds in diabetic animal models. Analyzed the healing process through images, histology and immunohistochemistry. Results show that the combination of AD and concentration gradient of VEGF could provide an appropriate angiogenesis, improve granulation formation, increase epithelization and maintain the VEGF levels of the wound bed. Eventually accelerate the direct healing of diabetic wounds or make good preparation for secondary skin graft.
Collapse
Affiliation(s)
- Hok Yin Tsui
- a Department of Burns and Plastic Surgery , Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School , Nanjing , China
| | - You Chen Liu
- a Department of Burns and Plastic Surgery , Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School , Nanjing , China
| | - Xin Yan
- a Department of Burns and Plastic Surgery , Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School , Nanjing , China
| | - Yue Lin
- a Department of Burns and Plastic Surgery , Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School , Nanjing , China
| | - Ye Xu
- a Department of Burns and Plastic Surgery , Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School , Nanjing , China
| | - Qian Tan
- a Department of Burns and Plastic Surgery , Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School , Nanjing , China
| |
Collapse
|
46
|
Zhao K, Li X, Lin B, Yang D, Zhou Y, Li Z, Guo Q, Lu N. Oroxyloside inhibits angiogenesis through suppressing internalization of VEGFR2/Flk-1 in endothelial cells. J Cell Physiol 2017; 233:3454-3464. [PMID: 28926106 DOI: 10.1002/jcp.26198] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 09/14/2017] [Indexed: 12/12/2022]
Abstract
Increasing flavonoids have been reported to possess anti-angiogenic effects. Inhibition of angiogenesis plays a critical role in the treatment of cancer, especially in advanced metastatic cancer. In this study, we assessed the effect of Oroxylin A-7-glucuronide (Oroxyloside), a main metabolite of Oroxylin A, on angiogenesis in human endothelial cell-like EA.hy926 cells. Oroxyloside suppressed the migration and tube formation of EA.hy926 cells. Meanwhile, microvessels sprouting from aortic rings and new blood vessels on the chicken chorioallantoic membrane (CAM) were also inhibited. Mechanism studies showed that Oroxyloside reduced the autophosphorylation of vascular endothelial growth factor receptor 2 (VEGFR2/Flk-1) while it up-regulated the expression of R-Ras and VE-cadherin. In consequence, Oroxyloside inhibited the downstream Akt/MAPK/NF-κB pathways and then decreased the nuclear translocation and DNA binding ability of NF-κB. Furthermore, in vivo study showed that Oroxyloside exhibited a potential anti-angiogenic effect in Matrigel plug assay and inhibited growth of xenografted tumors with low systemic toxicity, which could be ascribed to the inhibition of VEGFR2 internalization. Taken together, these results suggested that Oroxyloside could inhibit angiogenesis in vitro and in vivo via suppressing the internalization of VEGFR2 and might serve as a potential antitumor agent.
Collapse
Affiliation(s)
- Kai Zhao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Basic Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Xiaorui Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Basic Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Binyan Lin
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Basic Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Dawei Yang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Basic Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Yuxin Zhou
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Basic Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Zhiyu Li
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, P.R. China
| | - Qinglong Guo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Basic Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Na Lu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Basic Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| |
Collapse
|
47
|
Paquin-Veillette J, Lizotte F, Robillard S, Béland R, Breton MA, Guay A, Despatis MA, Geraldes P. Deletion of AT2 Receptor Prevents SHP-1-Induced VEGF Inhibition and Improves Blood Flow Reperfusion in Diabetic Ischemic Hindlimb. Arterioscler Thromb Vasc Biol 2017; 37:2291-2300. [PMID: 29074590 DOI: 10.1161/atvbaha.117.309977] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 10/05/2017] [Indexed: 01/15/2023]
Abstract
OBJECTIVE Ischemia caused by narrowing of femoral artery is a major cause of peripheral arterial disease and morbidity affecting patients with diabetes mellitus. We have previously reported that the inhibition of the angiogenic response to VEGF (vascular endothelial growth factor) in diabetic mice was associated with the increased expression of SHP-1 (SH2 domain-containing phosphatase 1), a protein that can be activated by the AT2 (angiotensin II type 2) receptor. Deletion of AT2 receptor has been shown to promote angiogenesis within the ischemic muscle. However, the relative impact of AT2 receptor in diabetic condition remains unknown. APPROACH AND RESULTS Nondiabetic and diabetic AT2 null (Atgr2-/Y) mice underwent femoral artery ligation after 2 months of diabetes mellitus. Blood perfusion was measured every week ≤4 weeks post-surgery. Expression of the VEGF, SHP-1, and renin-angiotensin pathways was evaluated. Blood flow in the ischemic muscle of diabetic Atgr2-/Y mice recovered faster and ≤80% after 4 weeks compared with 51% recovery in diabetic control littermates. Diabetic Atgr2-/Y had reduced apoptotic endothelial cells and elevated small vessel formation compared with diabetic Atgr2+/Y mice, as well as increased SHP-1 expression and reduced VEGF receptor activity. In endothelial cells, high glucose levels and AT2 agonist treatment did not change SHP-1, VEGF, and VEGF receptor expression. However, the activity of SHP-1 and its association with the VEGF receptors were increased, causing inhibition of the VEGF action in endothelial cell proliferation and migration. CONCLUSIONS Our results suggest that the deletion of AT2 receptor reduced SHP-1 activity and restored VEGF actions, leading to an increased blood flow reperfusion after ischemia in diabetes mellitus.
Collapse
MESH Headings
- Animals
- Blood Glucose/metabolism
- Cattle
- Cell Movement
- Cell Proliferation
- Cells, Cultured
- Diabetes Mellitus/genetics
- Diabetes Mellitus/metabolism
- Diabetes Mellitus/physiopathology
- Diabetic Angiopathies/genetics
- Diabetic Angiopathies/metabolism
- Diabetic Angiopathies/physiopathology
- Disease Models, Animal
- Endothelial Cells/metabolism
- Gene Deletion
- Genotype
- Hindlimb
- Ischemia/genetics
- Ischemia/metabolism
- Ischemia/physiopathology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Skeletal/blood supply
- Neovascularization, Physiologic
- Phenotype
- Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism
- Receptor, Angiotensin, Type 2/deficiency
- Receptor, Angiotensin, Type 2/genetics
- Receptors, Vascular Endothelial Growth Factor/metabolism
- Recovery of Function
- Regional Blood Flow
- Renin-Angiotensin System
- Signal Transduction
- Time Factors
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- Judith Paquin-Veillette
- From the Research Center of the Centre Hospitalier Universitaire de Sherbrooke (J.P.-V., F.L., S.R., R.B., M.-A.B., A.G., P.G.), Québec, Canada; and Departments of Surgery (M.-A.D.) and Medicine (P.G.), Division of Endocrinology, Université de Sherbrooke, Québec, Canada
| | - Farah Lizotte
- From the Research Center of the Centre Hospitalier Universitaire de Sherbrooke (J.P.-V., F.L., S.R., R.B., M.-A.B., A.G., P.G.), Québec, Canada; and Departments of Surgery (M.-A.D.) and Medicine (P.G.), Division of Endocrinology, Université de Sherbrooke, Québec, Canada
| | - Stéphanie Robillard
- From the Research Center of the Centre Hospitalier Universitaire de Sherbrooke (J.P.-V., F.L., S.R., R.B., M.-A.B., A.G., P.G.), Québec, Canada; and Departments of Surgery (M.-A.D.) and Medicine (P.G.), Division of Endocrinology, Université de Sherbrooke, Québec, Canada
| | - Raphaël Béland
- From the Research Center of the Centre Hospitalier Universitaire de Sherbrooke (J.P.-V., F.L., S.R., R.B., M.-A.B., A.G., P.G.), Québec, Canada; and Departments of Surgery (M.-A.D.) and Medicine (P.G.), Division of Endocrinology, Université de Sherbrooke, Québec, Canada
| | - Marc-André Breton
- From the Research Center of the Centre Hospitalier Universitaire de Sherbrooke (J.P.-V., F.L., S.R., R.B., M.-A.B., A.G., P.G.), Québec, Canada; and Departments of Surgery (M.-A.D.) and Medicine (P.G.), Division of Endocrinology, Université de Sherbrooke, Québec, Canada
| | - Andréanne Guay
- From the Research Center of the Centre Hospitalier Universitaire de Sherbrooke (J.P.-V., F.L., S.R., R.B., M.-A.B., A.G., P.G.), Québec, Canada; and Departments of Surgery (M.-A.D.) and Medicine (P.G.), Division of Endocrinology, Université de Sherbrooke, Québec, Canada
| | - Marc-Antoine Despatis
- From the Research Center of the Centre Hospitalier Universitaire de Sherbrooke (J.P.-V., F.L., S.R., R.B., M.-A.B., A.G., P.G.), Québec, Canada; and Departments of Surgery (M.-A.D.) and Medicine (P.G.), Division of Endocrinology, Université de Sherbrooke, Québec, Canada
| | - Pedro Geraldes
- From the Research Center of the Centre Hospitalier Universitaire de Sherbrooke (J.P.-V., F.L., S.R., R.B., M.-A.B., A.G., P.G.), Québec, Canada; and Departments of Surgery (M.-A.D.) and Medicine (P.G.), Division of Endocrinology, Université de Sherbrooke, Québec, Canada.
| |
Collapse
|
48
|
Hedhli J, Konopka CJ, Schuh S, Bouvin H, Cole JA, Huntsman HD, Kilian KA, Dobrucki IT, Boppart MD, Dobrucki LW. Multimodal Assessment of Mesenchymal Stem Cell Therapy for Diabetic Vascular Complications. Theranostics 2017; 7:3876-3888. [PMID: 29109784 PMCID: PMC5667411 DOI: 10.7150/thno.19547] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 06/07/2017] [Indexed: 12/26/2022] Open
Abstract
Peripheral arterial disease (PAD) is a debilitating complication of diabetes mellitus (DM) that leads to thousands of injuries, amputations, and deaths each year. The use of mesenchymal stem cells (MSCs) as a regenerative therapy holds the promise of regrowing injured vasculature, helping DM patients live healthier and longer lives. We report the use of muscle-derived MSCs to treat surgically-induced hindlimb ischemia in a mouse model of type 1 diabetes (DM-1). We serially evaluate several facets of the recovery process, including αVβ3-integrin expression (a marker of angiogenesis), blood perfusion, and muscle function. We also perform microarray transcriptomics experiments to characterize the gene expression states of the MSC-treated is- chemic tissues, and compare the results with those of non-ischemic tissues, as well as ischemic tissues from a saline-treated control group. The results show a multifaceted impact of mMSCs on hindlimb ischemia. We determined that the angiogenic activity one week after mMSC treatment was enhanced by approximately 80% relative to the saline group, which resulted in relative increases in blood perfusion and muscle strength of approximately 42% and 1.7-fold, respectively. At the transcriptomics level, we found that several classes of genes were affected by mMSC treatment. The mMSCs appeared to enhance both pro-angiogenic and metabolic genes, while suppressing anti-angiogenic genes and certain genes involved in the inflammatory response. All told, mMSC treatment appears to exert far-reaching effects on the microenvironment of ischemic tissue, enabling faster and more complete recovery from vascular occlusion.
Collapse
|
49
|
Sigrist S, Mechine-Neuville A, Mandes K, Calenda V, Braun S, Legeay G, Bellocq JP, Pinget M, Kessler L. Influence of VEGF on the Viability of Encapsulated Pancreatic Rat Islets after Transplantation in Diabetic Mice. Cell Transplant 2017; 12:627-35. [PMID: 14579931 DOI: 10.3727/000000003108747109] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
After pancreatic islet transplantation, insufficient blood supply is responsible for the loss of islet viability. The aim of our study was: 1) to determine the influence of vascular endothelial growth factor (VEGF) on the survival of encapsulated rat islets transplanted into healthy and diabetic mice and 2) to evaluate the metabolic efficiency of the VEGF-supplemented grafts. Twenty-four hours after culture, 50 rat islets immobilized into collagen in the presence of VEGF (100 ng/ml) and encapsulated (AN69 membrane, HOSPAL) were grafted in the peritoneal cavity of healthy or streptozotocin-induced diabetic mice (n = 6). Seven, 14, and 28 days after implantation, the encapsulation device and tissue surrounding the device were removed and the following parameters were analyzed: the number and the diameter of buds, the distance between devices and buds, the amount of cellular adhesion on the capsule surface, and the level of insulin secreted by encapsulated islet. For reversal of diabetes, 1000 rat islets encapsulated in the presence of VEGF were implanted in the peritoneal cavity of diabetic mice and fasting glycemia was analyzed. After 7 days of islet implantation in the absence of VEGF, the bud diameter was 16.1 ± 6.9 μm in diabetic mice and 34.4 ± 3.9 μm in healthy mice. However, the number of buds increased by a factor 2.5 in the presence of VEGF in both types of mice. Furthermore, when islets were transplanted in the presence of VEGF, the distance between the device and the buds was significantly decreased in both types of mice (p < 0.001) after 7, 14, and 28 days of islet implantation. Capsule analysis showed a decrease in cellular adhesion when the islets were encapsulated in the presence of VEGF. Insulin secretion of the islets was higher in the presence of VEGF compared with islets alone at all steps of the study. When 1000 rat islets were transplanted in the presence of VEGF, the glycemia level decreased to 6.2 ± 0.8 mmol/L after 3 days and remained stable until at least 28 days. In contrast, in the absence of VEGF, the initial decrease in the glucose level was rapidly followed by a relapse in hyperglycemia. In summary, VEGF increased the viability of engrafted encapsulated islets, increasing the duration of a normalized glycemia in diabetic mice following transplantation. Local adjunction of VEGF may therefore improve the clinical outcome of islet transplantation.
Collapse
Affiliation(s)
- S Sigrist
- Centre européen d'étude du Diabète, Faculté de Médecine, 11, rue Humann, 67 000, Strasbourg, France
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
López-Díez R, Shen X, Daffu G, Khursheed M, Hu J, Song F, Rosario R, Xu Y, Li Q, Xi X, Zou YS, Li H, Schmidt AM, Yan SF. Ager Deletion Enhances Ischemic Muscle Inflammation, Angiogenesis, and Blood Flow Recovery in Diabetic Mice. Arterioscler Thromb Vasc Biol 2017. [PMID: 28642238 DOI: 10.1161/atvbaha.117.309714] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Diabetic subjects are at higher risk of ischemic peripheral vascular disease. We tested the hypothesis that advanced glycation end products (AGEs) and their receptor (RAGE) block angiogenesis and blood flow recovery after hindlimb ischemia induced by femoral artery ligation through modulation of immune/inflammatory mechanisms. APPROACH AND RESULTS Wild-type mice rendered diabetic with streptozotocin and subjected to unilateral femoral artery ligation displayed increased accumulation and expression of AGEs and RAGE in ischemic muscle. In diabetic wild-type mice, femoral artery ligation attenuated angiogenesis and impaired blood flow recovery, in parallel with reduced macrophage content in ischemic muscle and suppression of early inflammatory gene expression, including Ccl2 (chemokine [C-C motif] ligand-2) and Egr1 (early growth response gene-1) versus nondiabetic mice. Deletion of Ager (gene encoding RAGE) or transgenic expression of Glo1 (reduces AGEs) restored adaptive inflammation, angiogenesis, and blood flow recovery in diabetic mice. In diabetes mellitus, deletion of Ager increased circulating Ly6Chi monocytes and augmented macrophage infiltration into ischemic muscle tissue after femoral artery ligation. In vitro, macrophages grown in high glucose display inflammation that is skewed to expression of tissue damage versus tissue repair gene expression. Further, macrophages grown in high versus low glucose demonstrate blunted macrophage-endothelial cell interactions. In both settings, these adverse effects of high glucose were reversed by Ager deletion in macrophages. CONCLUSIONS These findings indicate that RAGE attenuates adaptive inflammation in hindlimb ischemia; underscore microenvironment-specific functions for RAGE in inflammation in tissue repair versus damage; and illustrate that AGE/RAGE antagonism may fill a critical gap in diabetic peripheral vascular disease.
Collapse
Affiliation(s)
- Raquel López-Díez
- From the Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine (R.L.D., X.S., G.D., M.K., F.S., R.R., Y.X., Q.L., X.X., Y.S.Z., A.M.S., S.F.Y.), Department of Population Health (J.H., H.L.), and Department of Environmental Science (H.L.), New York University School of Medicine, New York
| | - Xiaoping Shen
- From the Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine (R.L.D., X.S., G.D., M.K., F.S., R.R., Y.X., Q.L., X.X., Y.S.Z., A.M.S., S.F.Y.), Department of Population Health (J.H., H.L.), and Department of Environmental Science (H.L.), New York University School of Medicine, New York
| | - Gurdip Daffu
- From the Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine (R.L.D., X.S., G.D., M.K., F.S., R.R., Y.X., Q.L., X.X., Y.S.Z., A.M.S., S.F.Y.), Department of Population Health (J.H., H.L.), and Department of Environmental Science (H.L.), New York University School of Medicine, New York
| | - Md Khursheed
- From the Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine (R.L.D., X.S., G.D., M.K., F.S., R.R., Y.X., Q.L., X.X., Y.S.Z., A.M.S., S.F.Y.), Department of Population Health (J.H., H.L.), and Department of Environmental Science (H.L.), New York University School of Medicine, New York
| | - Jiyuan Hu
- From the Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine (R.L.D., X.S., G.D., M.K., F.S., R.R., Y.X., Q.L., X.X., Y.S.Z., A.M.S., S.F.Y.), Department of Population Health (J.H., H.L.), and Department of Environmental Science (H.L.), New York University School of Medicine, New York
| | - Fei Song
- From the Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine (R.L.D., X.S., G.D., M.K., F.S., R.R., Y.X., Q.L., X.X., Y.S.Z., A.M.S., S.F.Y.), Department of Population Health (J.H., H.L.), and Department of Environmental Science (H.L.), New York University School of Medicine, New York
| | - Rosa Rosario
- From the Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine (R.L.D., X.S., G.D., M.K., F.S., R.R., Y.X., Q.L., X.X., Y.S.Z., A.M.S., S.F.Y.), Department of Population Health (J.H., H.L.), and Department of Environmental Science (H.L.), New York University School of Medicine, New York
| | - Yunlu Xu
- From the Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine (R.L.D., X.S., G.D., M.K., F.S., R.R., Y.X., Q.L., X.X., Y.S.Z., A.M.S., S.F.Y.), Department of Population Health (J.H., H.L.), and Department of Environmental Science (H.L.), New York University School of Medicine, New York
| | - Qing Li
- From the Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine (R.L.D., X.S., G.D., M.K., F.S., R.R., Y.X., Q.L., X.X., Y.S.Z., A.M.S., S.F.Y.), Department of Population Health (J.H., H.L.), and Department of Environmental Science (H.L.), New York University School of Medicine, New York
| | - Xiangmei Xi
- From the Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine (R.L.D., X.S., G.D., M.K., F.S., R.R., Y.X., Q.L., X.X., Y.S.Z., A.M.S., S.F.Y.), Department of Population Health (J.H., H.L.), and Department of Environmental Science (H.L.), New York University School of Medicine, New York
| | - Yu Shan Zou
- From the Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine (R.L.D., X.S., G.D., M.K., F.S., R.R., Y.X., Q.L., X.X., Y.S.Z., A.M.S., S.F.Y.), Department of Population Health (J.H., H.L.), and Department of Environmental Science (H.L.), New York University School of Medicine, New York
| | - Huilin Li
- From the Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine (R.L.D., X.S., G.D., M.K., F.S., R.R., Y.X., Q.L., X.X., Y.S.Z., A.M.S., S.F.Y.), Department of Population Health (J.H., H.L.), and Department of Environmental Science (H.L.), New York University School of Medicine, New York
| | - Ann Marie Schmidt
- From the Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine (R.L.D., X.S., G.D., M.K., F.S., R.R., Y.X., Q.L., X.X., Y.S.Z., A.M.S., S.F.Y.), Department of Population Health (J.H., H.L.), and Department of Environmental Science (H.L.), New York University School of Medicine, New York
| | - Shi Fang Yan
- From the Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine (R.L.D., X.S., G.D., M.K., F.S., R.R., Y.X., Q.L., X.X., Y.S.Z., A.M.S., S.F.Y.), Department of Population Health (J.H., H.L.), and Department of Environmental Science (H.L.), New York University School of Medicine, New York.
| |
Collapse
|