1
|
Zhang X, Wang H, Cai X, Zhang A, Liu E, Li Z, Jiang T, Li D, Ding W. α7nAChR Activation Combined with Endothelial Progenitor Cell Transplantation Attenuates Lung Injury in Diabetic Rats with Sepsis through the NF-κB Pathway. Inflammation 2024; 47:1344-1355. [PMID: 38302679 DOI: 10.1007/s10753-024-01980-0] [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: 10/04/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 02/03/2024]
Abstract
Chronic diabetes mellitus compromises the vascular system, which causes organ injury, including in the lung. Due to the strong compensatory ability of the lung, patients always exhibit subclinical symptoms. Once sepsis occurs, the degree of lung injury is more severe under hyperglycemic conditions. The α7 nicotinic acetylcholine receptor (α7nAChR) plays an important role in regulating inflammation and metabolism and can improve endothelial progenitor cell (EPC) functions. In the present study, lung injury caused by sepsis was compared between diabetic rats and normal rats. We also examined whether α7nAChR activation combined with EPC transplantation could ameliorate lung injury in diabetic sepsis rats. A type 2 diabetic model was induced in rats via a high-fat diet and streptozotocin. Then, a rat model of septic lung injury was established by intraperitoneal injection combined with endotracheal instillation of LPS. The oxygenation indices, wet-to-dry ratios, and histopathological scores of the lungs were tested after PNU282987 treatment and EPC transplantation. IL-6, IL-8, TNF-α, and IL-10 levels were measured. Caspase-3, Bax, Bcl-2, and phosphorylated NF-κB (p-NF-κB) levels were determined by blotting. Sepsis causes obvious lung injury, which is exacerbated by diabetic conditions. α7nAChR activation and endothelial progenitor cell transplantation reduced lung injury in diabetic sepsis rats, alleviating inflammation and decreasing apoptosis. This treatment was more effective when PNU282987 and endothelial progenitor cells were administered together. p-NF-κB levels decreased following treatment with PNU282987 and EPCs. In conclusion, α7nAChR activation combined with EPC transplantation can alleviate lung injury in diabetic sepsis rats through the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Xiaoyun Zhang
- Department of Anesthesiology, the Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, 150086, Heilongjiang, China
| | - Haixu Wang
- Department of Anesthesiology, the Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Xuemin Cai
- Department of Anesthesiology, Nanchong Central Hospital, Nanchong, Sichuan, China
| | - Aijia Zhang
- Department of Anesthesiology, the Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Enran Liu
- Department of Anesthesiology, the Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, 150086, Heilongjiang, China
| | - Zhiyuan Li
- Department of Anesthesiology, the Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, 150086, Heilongjiang, China
| | - Tao Jiang
- Department of Anesthesiology, the Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, 150086, Heilongjiang, China
| | - Dongmei Li
- Department of Anesthesiology, the Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, 150086, Heilongjiang, China
| | - Wengang Ding
- Department of Anesthesiology, the Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, 150086, Heilongjiang, China.
| |
Collapse
|
2
|
de Boer SA, Reijrink M, Abdulahad WH, Hoekstra ES, Slart RHJA, Heerspink HJL, Westra J, Mulder DJ. Angiogenic T cells are decreased in people with type 2 diabetes mellitus and recruited by the dipeptidyl peptidase-4 inhibitor Linagliptin: A subanalysis from a randomized, placebo-controlled trial (RELEASE study). Diabetes Obes Metab 2020; 22:1220-1225. [PMID: 32166899 PMCID: PMC7317866 DOI: 10.1111/dom.14024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/03/2020] [Accepted: 03/09/2020] [Indexed: 01/17/2023]
Abstract
Angiogenic T (Tang) cells are mediators of vascular repair, and are characterized by surface expression of CXCR4. This receptor for stromal cell-derived factor-1α (SDF-1α) is cleaved by dipeptidyl peptidase-4 (DPP-4). Tang cell levels were investigated in people with type 2 diabetes mellitus (T2DM) compared with matched healthy controls and after treatment with the DPP-4 inhibitor Linagliptin. People with T2DM were randomized to 5 mg/day Linagliptin (n = 20) or placebo (n = 21) for 26 weeks. Tang cell frequency was identified in peripheral blood mononuclear cells (CD3+ CD31+ CXCR4+ ) and levels of endothelial progenitor cells (EPCs) (CD34+ CD133+ KDR+ ) were also assessed in whole blood. Circulating Tang cell levels were significantly lower in people with T2DM compared with the healthy control group. SDF-1α levels increased significantly in Linagliptin-treated people with T2DM compared to placebo, and a trend was observed in change of Tang cell levels, while EPC count did not change. In conclusion, circulating Tang cell levels were considerably lower in people with T2DM, while a trend was observed in recruitment of Tang cells after 26 weeks of treatment with Linagliptin. These data suggest that DPP-4 inhibitors may potentially exert beneficial effects on bone marrow-driven vascular repair.
Collapse
Affiliation(s)
- Stefanie A. de Boer
- Department of Internal Medicine, Division of Vascular MedicineUniversity of Groningen, University Medical Centre GroningenGroningenThe Netherlands
| | - Melanie Reijrink
- Department of Internal Medicine, Division of Vascular MedicineUniversity of Groningen, University Medical Centre GroningenGroningenThe Netherlands
| | - Wayel H. Abdulahad
- Department of Rheumatology and Clinical ImmunologyUniversity of Groningen, University Medical Centre GroningenGroningenThe Netherlands
- Department of Pathology and Medical BiologyUniversity of Groningen, University Medical Centre GroningenGroningenThe Netherlands
| | - Elisa S. Hoekstra
- Department of Rheumatology and Clinical ImmunologyUniversity of Groningen, University Medical Centre GroningenGroningenThe Netherlands
| | - Riemer H. J. A. Slart
- Department of Nuclear Medicine and Molecular ImagingUniversity of Groningen, University Medical Centre GroningenGroningenThe Netherlands
- Department of Biomedical Photonic Imaging (BMPI)University of TwenteEnschedeThe Netherlands
| | - Hiddo J. L. Heerspink
- Department of Clinical Pharmacy and PharmacologyUniversity of Groningen, University Medical Centre GroningenGroningenNetherlands
| | - Johanna Westra
- Department of Rheumatology and Clinical ImmunologyUniversity of Groningen, University Medical Centre GroningenGroningenThe Netherlands
| | - Douwe J. Mulder
- Department of Internal Medicine, Division of Vascular MedicineUniversity of Groningen, University Medical Centre GroningenGroningenThe Netherlands
| |
Collapse
|
3
|
O'Loughlin A, Kulkarni M, Vaughan EE, Creane M, Liew A, Dockery P, Pandit A, O'Brien T. Autologous circulating angiogenic cells treated with osteopontin and delivered via a collagen scaffold enhance wound healing in the alloxan-induced diabetic rabbit ear ulcer model. Stem Cell Res Ther 2014; 4:158. [PMID: 24444259 PMCID: PMC4054999 DOI: 10.1186/scrt388] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2013] [Revised: 05/24/2013] [Accepted: 12/02/2013] [Indexed: 12/26/2022] Open
Abstract
INTRODUCTION Diabetic foot ulceration is the leading cause of amputation in people with diabetes mellitus. Peripheral vascular disease is present in the majority of patients with diabetic foot ulcers. Despite standard treatments there exists a high amputation rate. Circulating angiogenic cells previously known as early endothelial progenitor cells are derived from peripheral blood and support angiogenesis and vasculogenesis, providing a potential topical treatment for non-healing diabetic foot ulcers. METHODS A scaffold fabricated from Type 1 collagen facilitates topical cell delivery to a diabetic wound. Osteopontin is a matricellular protein involved in wound healing and increases the angiogenic potential of circulating angiogenic cells. A collagen scaffold seeded with circulating angiogenic cells was developed. Subsequently the effect of autologous circulating angiogenic cells that were seeded in a collagen scaffold and topically delivered to a hyperglycemic cutaneous wound was assessed. The alloxan-induced diabetic rabbit ear ulcer model was used to determine healing in response to the following treatments: collagen seeded with autologous circulating angiogenic cells exposed to osteopontin, collagen seeded with autologous circulating angiogenic cells, collagen alone and untreated wound. Stereology was used to assess angiogenesis in wounds. RESULTS The cells exposed to osteopontin and seeded on collagen increased percentage wound closure as compared to other groups. Increased angiogenesis was observed with the treatment of collagen and collagen seeded with circulating angiogenic cells. CONCLUSIONS These results demonstrate that topical treatment of full thickness cutaneous ulcers with autologous circulating angiogenic cells increases wound healing. Cells exposed to the matricellular protein osteopontin result in superior wound healing. The wound healing benefit is associated with a more efficient vascular network. This topical therapy provides a potential novel therapy for the treatment of non-healing diabetic foot ulcers in humans.
Collapse
|
4
|
Isolation and characterization of mouse bone marrow-derived Lin⁻/VEGF-R2⁺ progenitor cells. Ann Hematol 2013; 92:1461-72. [PMID: 23771478 DOI: 10.1007/s00277-013-1815-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Accepted: 06/04/2013] [Indexed: 10/26/2022]
Abstract
Circulating endothelial progenitor cells (EPCs) in the peripheral blood (PB) have physiological roles in the maintenance of the existing vascular beds and rescue of vascular injury. In this study, we have evaluated the properties of Lin⁻/VEGF-R2⁺ progenitor cells isolated from the mouse bone marrow (BM) and further studied their distribution and integration in an animal model of laser-induced retinal vascular injury. Lin⁻/VEGF-R2⁺ cells were enriched from C57BL/6 mice BM using magnetic cell sorting with hematopoietic lineage (Lin) depletion followed by VEGF-R2 positive selection. Lin⁻/VEGF-R2⁺ BM cells were characterized using flow cytometry and immunocytochemistry and further tested for colony formation during culture and tube formation on Matrigel®. Lin⁻/VEGF-R2⁺ BM cells possessed typical EPC properties such as forming cobble-stone shaped colonies after 3 to 4 weeks of culture, CD34⁺ expression, take up of Dil-acLDL and binding to Ulex europaeus agglutinin. However, they did not form tube-like structures on Matrigel®. The progenitor cells retained their phenotype over extended period of culture. After intravitreal transplantation in eyes subjected to the laser-induced retinal vascular injury, some Lin⁻/VEGF-R2⁺ cells were able to integrate into the damaged retinal vasculature but the level of cell integration seemed less efficient when compared with previous reports in which EPCs from the human PB were employed. Our results indicate that Lin⁻/VEGF-R2⁺ cells isolated from the mouse BM share some similarities to EPCs from the human PB but most of them are at a very early stage of maturation and remain quiescent during culture and after intravitreal transplantation.
Collapse
|
5
|
Ma XL, Sun XL, Wan CY, Ma JX, Tian P. Significance of circulating endothelial progenitor cells in patients with fracture healing process. J Orthop Res 2012; 30:1860-6. [PMID: 22528744 DOI: 10.1002/jor.22134] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Accepted: 03/29/2012] [Indexed: 02/04/2023]
Abstract
Fracture healing is a complex bone formation process, and neovascularization may contribute to new bone regeneration. The circulating endothelial progenitor cell (EPC) mobilization and homing could involve in neovascularization and vasculogenesis. In this study, we investigate the changes of circulating EPC during bone fracture healing, and the possible contribution of EPCs to increased neovascularization and fracture healing. The number of circulating EPCs was monitored in twenty-four patients with long bone traumatic fracture within the first 48 h and at 3, 5, 10, and 14 days post-fracture. The mononuclear cells which isolated from peripheral blood were analyzed by flow cytometry. Peripheral blood counts of leukocytes and platelets were measured by hematology analyzer. The amount of peripheral EPCs significantly increased in patients with fracture compared to age-matched healthy control subjects within the first 48 h after injury, and peaked at 3 days post-fracture. There was no significant difference in the change trend of early EPCs between male and female, but the number of early EPCs was significantly greater in younger patients compared to older patients. A comparison of the EPCs levels between patients with severe injury (ISS > 16) and patients with mild injury (ISS ≤ 16) revealed no statistically significant difference. The level of early EPCs was inverse correlation with the level of plate after fracture, but no correlation with the level of peripheral leucocytes. These findings suggest traumatic fracture may induce the mobilization of EPCs into the peripheral circulation. The increased EPCs may contribute to neovascularization and involve in fracture healing.
Collapse
|
6
|
Dysfunction of endothelial progenitor cells under diabetic conditions and its underlying mechanisms. Arch Pharm Res 2012; 35:223-34. [PMID: 22370777 DOI: 10.1007/s12272-012-0203-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Revised: 11/18/2011] [Accepted: 11/21/2011] [Indexed: 10/28/2022]
Abstract
Cardiovascular complications have been major concerns in the treatment of diabetes, and up to 80% of all deaths in diabetic patients are linked to cardiovascular problems. Impaired angiogenesis is one of the most serious symptoms associated with diabetes, resulting in delayed wound healing and lower limb amputation. Endothelial progenitor cells (EPCs), a subpopulation of adult stem cells, are recruited from bone marrow to the injured vessel to promote endothelial regeneration and neovascularization, playing an important role in angiogenesis. Interestingly, several clinical studies have showed that the number of recruited EPCs is reduced and their function is decreased under diabetic conditions, implying that diabetic EPC dysfunction may contribute to defective angiogenesis and resultant cardiovascular complications in diabetes. To recover the functional abilities of diabetic EPCs and to address possible application of EPC cell therapy to diabetic patients, some studies provided explanations for diabetic EPC dysfunction including increased oxidative stress, involvement of the inflammatory response, alteration in the nitric oxide pathway and reduced signals for EPC recruitment. This review discusses clinical evidence of impairment of EPC functions under diabetic conditions and the suggested mechanisms for diabetic EPC dysfunction.
Collapse
|
7
|
Abstract
Endothelial progenitor cells (EPCs) were first identified by Ashara et al. in 1997 (Asahara et al. Science 275:964-967, 1997) and were thought to contribute to angiogenesis and vasculogenesis. Since their discovery, circulating levels of EPCs were found to serve as biomarkers as low levels correlate with increased cardiovascular events and death from cardiovascular causes (Werner et al. N Engl J Med 353:999-1007, 2005; Fadini et al. J Am Coll Cardiol 45:1449-1457, 2005; Hill et al. N Engl J Med 348:593-600, 2003; Schmidt-Lucke et al. Circulation 111:2981-2987, 2005). Additionally, EPC dysfunction has been associated with diabetes mellitus and other disease states. However, recently there has been a great deal of controversy in the field over the exact definition and function of an EPC. To help classify EPCs, they have been divided into two distinct groups (1) circulating angiogenic cells (also referred to as early EPCs) and (2) endothelial colony forming cells (also referred to as late outgrowth EPCs). Circulating angiogenic cells are believed to represent a cell population enriched in monocytes and exert their angiogenic effects via paracrine and signaling mechanisms whereas endothelial colony forming cells are true EPCs and may enhance angiogenesis and vasculogenesis by incorporating into the newly forming vessels. Here the isolation and identification of circulating angiogenic cells are described.
Collapse
Affiliation(s)
- Erin E Vaughan
- Regenerative Medicine Institute, University College Hospital, National University of Ireland, Galway, Ireland
| | | |
Collapse
|
8
|
Bowes T, Hanley SA, Liew A, Eglon M, Mashayekhi K, O’Kennedy R, Barry F, Taylor WR, O’Brien T, Griffin MD, Finlay WJJ, Greiser U. Developing Cell-Specific Antibodies to Endothelial Progenitor Cells Using Avian Immune Phage Display Technology. ACTA ACUST UNITED AC 2011; 16:744-54. [DOI: 10.1177/1087057111407067] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
This study aims at generating immune chicken phage display libraries and single-chain antibodies (scFvs) specifically directed against cell surface markers of cultured peripheral blood mononuclear cells (PBMCs) that contain endothelial progenitor cells (EPCs). In contrast to previous approaches that use well-defined recombinant antigens attached to plastic surfaces that may alter the structure of the proteins, the authors describe a method that maintains the cell surface markers on live cells while providing the opportunity to rapidly screen entire libraries for antibodies that bind to unknown cell surface markers of progenitor/stem cells. Chickens immunized with live EPCs, consisting of a heterogeneous population of lymphocytes and monocytes, demonstrated a robust immune response. After three rounds of biopanning, the authors purified and characterized three unique scFvs called UG1-3. Codon-optimized recombinant UG1 (gUG-1) shows binding by flow cytometry to circulating CD14-positive cells in peripheral blood consistent with predominant expression of a target protein on monocyte subsets. The authors describe the successful use of immunization of chickens for the generation of scFvs against a heterogenous population of EPCs displaying unknown cell surface markers and demonstrate the strong potential of phage display technology in the development of reagents for the isolation and characterization of stem/progenitor cells.
Collapse
Affiliation(s)
- Tyrone Bowes
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland
| | - Shirley A. Hanley
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland
| | - Aaron Liew
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland
| | - Marc Eglon
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland
| | - Kaveh Mashayekhi
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland
| | - Richard O’Kennedy
- School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland
| | - Frank Barry
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland
| | - W. Robert Taylor
- Department of Medicine and Biomedical Engineering, Emory University School of Medicine, and the Atlanta VA Medical Center, Atlanta, Georgia
| | - Timothy O’Brien
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland
| | - Matthew D. Griffin
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland
| | - William J. J. Finlay
- School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland, Pfizer R&D, Dublin, Ireland
| | - Udo Greiser
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland
| |
Collapse
|
9
|
Martin DC, Semple JL, Sefton MV. Poly(methacrylic acid-co-methyl methacrylate) beads promote vascularization and wound repair in diabetic mice. J Biomed Mater Res A 2010; 93:484-92. [PMID: 19585566 DOI: 10.1002/jbm.a.32528] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Topical application of beads made from poly(methacrylic acid-co-methyl methacrylate) (45 mol % methacrylic acid, MAA) increased the number of blood vessels and improved 1.5 x 1.5 cm full thickness wound closure in a diabetic mouse (db/db) model. Three groups were compared: MAA beads, control poly(methyl methacrylate) beads (PMMA), and no bead blanks. MAA bead treatment significantly increased percent wound closure at all timepoints (7, 14, and 21 days) with MAA bead-treated wounds almost closed at day 21 (91 +/- 5.4% MAA vs. 79 +/- 3.2% PMMA or 76 +/- 4.8% no beads; p < 0.05). This was consistent with the expected significant increase in vascularity in the MAA group at days 7 and 14. For example at day 14, MAA bead-treated wounds had a vascular density of 22.7 +/- 2.6 vessels/hpf compared with 17.0 +/- 2.0 vessels/hpf in the PMMA bead group (p < 0.05). Epithelial gap and migration measurements suggested that the increased vascularity leads to enhanced epithelial cell migration as a principal means of wound closure. Although studies are underway to elucidate the mechanism of this angiogenic response, the results presented here support the notion that such materials, perhaps in other forms, may be useful in wound care or in other situations where vascularity is to be enhanced without the use of exogenous growth factors.
Collapse
Affiliation(s)
- Daniel C Martin
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | | | | |
Collapse
|