1
|
Liu F, Xu X, Sun T. Vascular endothelial growth factor accelerates healing of foot ulcers in diabetic rats via promoting M2 macrophage polarization. Diabet Med 2024; 41:e15388. [PMID: 38934613 DOI: 10.1111/dme.15388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 05/16/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024]
Abstract
AIM The objective was to investigate the specific role and the regulatory mechanism of vascular endothelial growth factor (VEGF) during wound healing in diabetic foot ulcer (DFU). METHODS Streptozotocin-induced diabetic rats were used to establish a DFU animal model. VEGF and Axitinib (a specific inhibitor of VEGFR) were used for treatment in vivo. The wounds at different time points were imaged and histological analysis of the wounds were performed by haematoxylin and eosin (H&E) staining and Masson's trichrome staining. Immunohistochemical staining was conducted to examine CD31 and eNOS expression in the wounds. Immunofluorescence assay and quantitative real-time PCR were performed to examine macrophage markers. In addition, THP-1 was differentiated to macrophages, and then treated with interleukin (IL)-4 to induce M2 macrophages, followed by VEGF treatment. The conditional medium (CM) from VEGF-mediated macrophages were collected to culture human dermal fibroblasts (HDFs). Cell viability and migration were measured by Cell Counting Kit (CCK)-8, wound-healing and Transwell assays, respectively. RESULTS VEGF treatment remarkably accelerated wound healing of DFU rats. VEGF promoted collagen deposition and elevated CD31 and eNOS expression, confirming the pro-angiogenesis of VEGF around diabetic wound in rats. Meanwhile, VEGF restricted pro-inflammatory cytokines and increased F4/80 and CD206 expression, highlighting the activated macrophages and enhanced M2 macrophages following VEGF treatment in diabetic wounds of DFU rats. However, Axitinib exerted an opposite function to VEGF in DFU rats. Moreover, VEGF directly promoted macrophage polarization toward M2 phenotype in vitro, and the CM from VEGF-mediated M2 macrophages markedly promoted HDFs proliferation, migration and collagen deposition. CONCLUSION VEGF might accelerate the wound healing of DFU through promoting M2 macrophage polarization and fibroblast migration.
Collapse
Affiliation(s)
- Fei Liu
- Department of Hand, Foot and Ankle Surgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Xianrui Xu
- Department of Neurology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Tao Sun
- Department of Burn and Plastic Surgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| |
Collapse
|
2
|
van Rhijn-Brouwer FCCC, Wever KE, Kiffen R, van Rhijn JR, Gremmels H, Fledderus JO, Vernooij RWM, Verhaar MC. Systematic review and meta-analysis of the effect of bone marrow-derived cell therapies on hind limb perfusion. Dis Model Mech 2024; 17:dmm050632. [PMID: 38616715 PMCID: PMC11139036 DOI: 10.1242/dmm.050632] [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: 11/27/2023] [Accepted: 04/03/2024] [Indexed: 04/16/2024] Open
Abstract
Preclinical and clinical studies on the administration of bone marrow-derived cells to restore perfusion show conflicting results. We conducted a systematic review and meta-analysis on preclinical studies to assess the efficacy of bone marrow-derived cells in the hind limb ischemia model and identify possible determinants of therapeutic efficacy. In vivo animal studies were identified using a systematic search in PubMed and EMBASE on 10 January 2022. 85 studies were included for systematic review and meta-analysis. Study characteristics and outcome data on relative perfusion were extracted. The pooled mean difference was estimated using a random effects model. Risk of bias was assessed for all included studies. We found a significant increase in perfusion in the affected limb after administration of bone marrow-derived cells compared to that in the control groups. However, there was a high heterogeneity between studies, which could not be explained. There was a high degree of incomplete reporting across studies. We therefore conclude that the current quality of preclinical research is insufficient (low certainty level as per GRADE assessment) to identify specific factors that might improve human clinical trials.
Collapse
Affiliation(s)
| | - Kimberley Elaine Wever
- Department of Anaesthesiology, Pain and Palliative Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Romy Kiffen
- Department of Anaesthesiology, Pain and Palliative Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Jon-Ruben van Rhijn
- Institute of Life Sciences and Chemistry, HU University of Applied Sciences Utrecht, 3584 CS Utrecht, The Netherlands
| | - Hendrik Gremmels
- Department of Medical Microbiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Joost Ougust Fledderus
- Department of Nephrology and Hypertension, Regenerative Medicine Center Utrecht, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Robin Wilhelmus Maria Vernooij
- Department of Nephrology and Hypertension, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Marianne Christina Verhaar
- Department of Nephrology and Hypertension, Regenerative Medicine Center Utrecht, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
3
|
Nishibata T, Amino N, Tanaka-Kado R, Tsujimoto S, Kawashima T, Konagai S, Suzuki T, Takeuchi M. Blockade of EP4 by ASP7657 Modulates Myeloid Cell Differentiation In Vivo and Enhances the Antitumor Effect of Radiotherapy. BIOMED RESEARCH INTERNATIONAL 2023; 2023:7133726. [PMID: 38058393 PMCID: PMC10697779 DOI: 10.1155/2023/7133726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 10/20/2023] [Accepted: 10/31/2023] [Indexed: 12/08/2023]
Abstract
The tumor microenvironment (TME) is thought to influence the antitumor efficacy of immuno-oncology agents through various products of both tumor and stromal cells. One immune-suppressive factor is prostaglandin E2 (PGE2), a lipid mediator whose biosynthesis is regulated by ubiquitously expressed cyclooxygenase- (COX-) 1 and inducible COX-2. By activating its receptors, PGE2 induces immune suppression to modulate differentiation of myeloid cells into myeloid-derived suppressor cells (MDSCs) rather than dendritic cells (DCs). Pharmacological blockade of prostaglandin E receptor 4 (EP4) causes a decrease in MDSCs, reprogramming of macrophage polarization, and increase in tumor-infiltrated T cells, leading to enhancement of antitumor immunity in preclinical models. Here, we report the effects of the highly potent EP4 antagonist ASP7657 on the DC population in tumor and antitumor immune activation in an immunocompetent mouse tumor model. Oral administration of ASP7657 inhibited tumor growth, which was accompanied by an increase in intratumor DC and CD8+ T cell populations and a decrease in the M-MDSC population in a CT26 immunocompetent mouse model. The antitumor activity of ASP7657 was dependent on CD8+ T cells and enhanced when combined with an antiprogrammed cell death-1 (PD-1) antibody. Notably, ASP7657 also significantly enhanced the antitumor efficacy of radiotherapy in an anti-PD-1 antibody refractory model. These results indicate that the therapeutic potential of ASP7657 arises via upregulation of DCs and subsequent CD8+ T cell activation in addition to suppression of MDSCs in mouse models and that combining EP4 antagonists with radiotherapy or an anti-PD-1 antibody can improve antitumor efficacy.
Collapse
Affiliation(s)
- Toshihide Nishibata
- Immuno-oncology, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - Nobuaki Amino
- Immuno-oncology, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - Ruriko Tanaka-Kado
- Immuno-oncology, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - Susumu Tsujimoto
- Immuno-oncology, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - Tomoko Kawashima
- Immuno-oncology, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - Satoshi Konagai
- Immuno-oncology, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - Tomoyuki Suzuki
- Immuno-oncology, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - Masahiro Takeuchi
- Immuno-oncology, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| |
Collapse
|
4
|
Pu Z, Shimizu Y, Hayashi T, Che Y, Suzuki J, Tsuzuki K, Narita S, Shibata R, Calvert JW, Murohara T. Cardiac Lymphatic Insufficiency Leads to Diastolic Dysfunction Via Myocardial Morphologic Change. JACC Basic Transl Sci 2023; 8:958-972. [PMID: 37719433 PMCID: PMC10504400 DOI: 10.1016/j.jacbts.2023.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 01/18/2023] [Accepted: 01/18/2023] [Indexed: 09/19/2023]
Abstract
Although cardiac lymphatic vessels have received increasing attention in recent years, there is still a knowledge gap between cardiac lymphatics and heart homeostasis in a normal heart. In the present study, we established a mouse model of cardiac lymphatic insufficiency ablating cardiac lymphatic collector vessels to reveal the crucial role of cardiac lymphatic vessels in maintaining cardiac homeostasis and the impact on cardiac function both in physiological and pathologic settings. Furthermore, therapeutic lymphangiogenesis improved the adverse effect on cardiac morphologic changes and functions. These findings suggest that the cardiac lymphatic system would be a novel therapeutic target for heart disease.
Collapse
Affiliation(s)
- Zhongyue Pu
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuuki Shimizu
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takumi Hayashi
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yiyang Che
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Junya Suzuki
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuhito Tsuzuki
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shingo Narita
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Rei Shibata
- Department of Advanced Cardiovascular Therapeutics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - John W. Calvert
- Carlyle Fraser Heart Center, Division of Cardiothoracic Surgery, Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
5
|
Therapeutic angiogenesis for patients with no-option critical limb ischemia by adipose-derived regenerative cells: TACT-ADRC multicenter trial. Angiogenesis 2022; 25:535-546. [PMID: 35802311 PMCID: PMC9263817 DOI: 10.1007/s10456-022-09844-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 06/19/2022] [Indexed: 12/24/2022]
Abstract
Background Patients with critical limb ischemia (CLI) still have a high rate of lower limb amputation, which is associated with not only a decrease in quality of life but also poor life prognosis. Implantation of adipose-derived regenerative cells (ADRCs) has an angiogenic potential for patients with limb ischemia. Objectives We investigated safety, feasibility, and efficacy of therapeutic angiogenesis by cell transplantation (TACT) of ADRCs for those patients in multicenter clinical trial in Japan. Methods The TACT-ADRC multicenter trial is a prospective, interventional, open-labeled study. Patients with CLI (Fontaine class III–IV) who have no other option for standard revascularization therapy were enrolled in this study. Thirty-four target ischemic limbs of 29 patients were received freshly isolated autologous ADRCs implantation. Results The overall survival rate at a post-operative period and at 6 months follow-up was 100% at any time points. As a primary endpoint for efficacy evaluation, 32 limbs out of 34 (94.1%) were free from major amputation for 6 months. Numerical rating scale (from 6 to 1) as QOL score, ulcer size (from 317 mm2 at to 109 mm2), and 6-min walking distance (from 255 to 369 m) improved in 90.6%, 83.3%, and 72.2% patients, respectively. Conclusions Implantation of autologous ADRCs could be safe and effective for the achievement of therapeutic angiogenesis in the multicenter settings, as a result in no major adverse event, optimal survival rate, and limb salvage for patients with no-conventional option against critical limb ischemia. TRN: jRCTb040190118; Date: Nov. 24th, 2015. Supplementary Information The online version contains supplementary material available at 10.1007/s10456-022-09844-7.
Collapse
|
6
|
Long Term Response to Circulating Angiogenic Cells, Unstimulated or Atherosclerotic Pre-Conditioned, in Critical Limb Ischemic Mice. Biomedicines 2021; 9:biomedicines9091147. [PMID: 34572333 PMCID: PMC8469527 DOI: 10.3390/biomedicines9091147] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/31/2021] [Accepted: 08/31/2021] [Indexed: 01/05/2023] Open
Abstract
Critical limb ischemia (CLI), the most severe form of peripheral artery disease, results from the blockade of peripheral vessels, usually correlated to atherosclerosis. Currently, endovascular and surgical revascularization strategies cannot be applied to all patients due to related comorbidities, and even so, most patients require re-intervention or amputation within a year. Circulating angiogenic cells (CACs) constitute a good alternative as CLI cell therapy due to their vascular regenerative potential, although the mechanisms of action of these cells, as well as their response to pathological conditions, remain unclear. Previously, we have shown that CACs enhance angiogenesis/arteriogenesis from the first days of administration in CLI mice. Also, the incubation ex vivo of these cells with factors secreted by atherosclerotic plaques promotes their activation and mobilization. Herein, we have evaluated the long-term effect of CACs administration in CLI mice, whether pre-stimulated or not with atherosclerotic factors. Remarkably, mice receiving CACs and moreover, pre-stimulated CACs, presented the highest blood flow recovery, lower progression of ischemic symptoms, and decrease of immune cells recruitment. In addition, many proteins potentially involved, like CD44 or matrix metalloproteinase 9 (MMP9), up-regulated in response to ischemia and decreased after CACs administration, were identified by a quantitative proteomics approach. Overall, our data suggest that pre-stimulation of CACs with atherosclerotic factors might potentiate the regenerative properties of these cells in vivo.
Collapse
|
7
|
Tsuzuki K, Shimizu Y, Suzuki J, Pu Z, Yamaguchi S, Fujikawa Y, Kato K, Ohashi K, Takefuji M, Bando YK, Ouchi N, Calvert JW, Shibata R, Murohara T. Adverse Effect of Circadian Rhythm Disorder on Reparative Angiogenesis in Hind Limb Ischemia. J Am Heart Assoc 2021; 10:e020896. [PMID: 34348468 PMCID: PMC8475022 DOI: 10.1161/jaha.121.020896] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 06/21/2021] [Indexed: 12/13/2022]
Abstract
Background Circadian rhythm disorders, often seen in modern lifestyles, are a major social health concern. The aim of this study was to examine whether circadian rhythm disorders would influence angiogenesis and blood perfusion recovery in a mouse model of hind limb ischemia. Methods and Results A jet-lag model was established in C57BL/6J mice using a light-controlled isolation box. Control mice were kept at a light/dark 12:12 (12-hour light and 12-hour dark) condition. Concentrations of plasma vascular endothelial growth factor and circulating endothelial progenitor cells in control mice formed a circadian rhythm, which was diminished in the jet-lag model (P<0.05). The jet-lag condition deteriorated tissue capillary formation (P<0.001) and tissue blood perfusion recovery (P<0.01) in hind limb ischemia, which was associated with downregulation of vascular endothelial growth factor expression in local ischemic tissue and in the plasma. Although the expression of clock genes (ie, Clock, Bmal1, and Cry) in local tissues was upregulated after ischemic injury, the expression levels of cryptochrome (Cry) 1 and Cry2 were inhibited by the jet-lag condition. Next, Cry1 and Cry2 double-knockout mice were examined for blood perfusion recoveries and a reparative angiogenesis. Cry1 and Cry2 double-knockout mice revealed suppressed capillary density (P<0.001) and suppressed tissue blood perfusion recovery (P<0.05) in the hind limb ischemia model. Moreover, knockdown of CRY1/2 in human umbilical vein endothelial cells was accompanied by increased expression of WEE1 and decreased expression of HOXC5. This was associated with decreased proliferative capacity, migration ability, and tube formation ability of human umbilical vein endothelial cells, respectively, leading to impairment of angiogenesis. Conclusions Our data suggest that circadian rhythm disorder deteriorates reparative ischemia-induced angiogenesis and that maintenance of circadian rhythm plays an important role in angiogenesis.
Collapse
Affiliation(s)
- Kazuhito Tsuzuki
- Department of CardiologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Yuuki Shimizu
- Department of CardiologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Junya Suzuki
- Department of CardiologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Zhongyue Pu
- Department of CardiologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Shukuro Yamaguchi
- Department of CardiologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Yusuke Fujikawa
- Department of CardiologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Katsuhiro Kato
- Department of CardiologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Koji Ohashi
- Department of CardiologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Mikito Takefuji
- Department of CardiologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Yasuko K. Bando
- Department of CardiologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Noriyuki Ouchi
- Department of CardiologyNagoya University Graduate School of MedicineNagoyaJapan
| | - John W. Calvert
- Division of Cardiothoracic SurgeryDepartment of SurgeryCarlyle Fraser Heart CenterEmory University School of MedicineAtlantaGA
| | - Rei Shibata
- Department of Advanced Cardiovascular TherapeuticsNagoya University Graduate School of MedicineNagoyaJapan
| | - Toyoaki Murohara
- Department of CardiologyNagoya University Graduate School of MedicineNagoyaJapan
| |
Collapse
|
8
|
Lei Y, Cui Q, Yang G, Piao L, Inoue A, Wu H, Li X, Kuzuya M, Cheng XW. Statins Mitigate Stress-Related Vascular Aging and Atherosclerosis in apoE-Deficient Mice Fed High Fat-Diet: The Role of Glucagon-Like Peptide-1/Adiponectin Axis. Front Cell Dev Biol 2021; 9:687868. [PMID: 34368136 PMCID: PMC8335539 DOI: 10.3389/fcell.2021.687868] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/04/2021] [Indexed: 01/30/2023] Open
Abstract
Objectives Exposure to chronic psychosocial stress is a risk factor for atherosclerotic cardiovascular diseases. Given that the 3-hydroxy-3-methylglutaryl-coenzyme reductase inhibitor statins prevent atherogenesis, we evaluated whether pitavastatin prevents chronic stress- and high fat diet-induced vascular senescence and atherogenesis in apolipoprotein E-deficient (ApoE–/–) mice, with a special focus on glucagon-like peptide-1 (GLP-1)/adiponectin (APN) axis. Methods and Results 6-week-old ApoE–/– mice loaded a high-fat diet were randomly assigned into non-stress (n = 12) and stress (n = 13) groups for 12 weeks. Non-stress control mice were left undisturbed. Chronic stress accelerated high fat diet-induce arterial senescence and atherosclerotic plaque growth. The chronic stress lowered the levels of circulating GLP-1 as well as adipose and plasma APN. As compared with the stress alone mice, the pitavastatin-treated mice had reduced macrophage infiltration, elastin fragments, and increased plaque collagen volume, and lowered levels of osteopontin, toll-like receptor-2/-4, macrophage chemoattractant protein-1, C-X-C chemokine receptor-4, p47phox, p47phox, gp91phox, cathepsins S, p16, and p21, mRNAs and/or proteins. Pitavastatin increased plasma GLP-1 and APN levels and suppressed matrix metalloproteinase-2/-9 gene expressions and activities in the aortas. Finally, the protective effect of pitavastatin was abrogated by APN blocking. Conclusion These findings suggested that the pitavastatin-mediated pleiotropic vasculoprotective effects are likely attributable, at least in part, to the elevation of GLP-1 and APN levels and the inhibition of diet-induced plaque inflammation, oxidative stress, and proteolysis in ApoE–/– mice received chronic stress conditions.
Collapse
Affiliation(s)
- Yanna Lei
- Department of Intensive Care Unit, Yanbian University Hospital, Yanjin, China
| | - Qingsong Cui
- Department of Intensive Care Unit, Yanbian University Hospital, Yanjin, China
| | - Guang Yang
- Department of Cardiology and Hypertension, Yanbian University Hospital, Yanjin, China
| | - Limei Piao
- Department of Cardiology and Hypertension, Yanbian University Hospital, Yanjin, China
| | - Aiko Inoue
- Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hongxian Wu
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiang Li
- Department of Intensive Care Unit, Yanbian University Hospital, Yanjin, China
| | - Masafumi Kuzuya
- Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Xian Wu Cheng
- Department of Intensive Care Unit, Yanbian University Hospital, Yanjin, China.,Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
9
|
Magenta A, Florio MC, Ruggeri M, Furgiuele S. Autologous cell therapy in diabetes‑associated critical limb ischemia: From basic studies to clinical outcomes (Review). Int J Mol Med 2021; 48:173. [PMID: 34278463 DOI: 10.3892/ijmm.2021.5006] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 09/30/2020] [Indexed: 01/13/2023] Open
Abstract
Cell therapy is becoming an attractive alternative for the treatment of patients with no‑option critical limb ischemia (CLI). The main benefits of cell therapy are the induction of therapeutic angiogenesis and neovascularization that lead to an increase in blood flow in the ischemic limb and tissue regeneration in non‑healing cutaneous trophic lesions. In the present review, the current state of the art of strategies in the cell therapy field are summarized, focusing on intra‑operative autologous cell concentrates in diabetic patients with CLI, examining different sources of cell concentrates and their mechanisms of action. The present study underlined the detrimental effects of the diabetic condition on different sources of autologous cells used in cell therapy, and also in delaying wound healing capacity. Moreover, relevant clinical trials and critical issues arising from cell therapy trials are discussed. Finally, the new concept of cell therapy as an adjuvant therapy to increase wound healing in revascularized diabetic patients is introduced.
Collapse
Affiliation(s)
| | - Maria Cristina Florio
- Laboratory of Cardiovascular Science, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, MD 21224, USA
| | - Massimo Ruggeri
- Department of Vascular Surgery, San Camillo de Lellis Hospital, I‑02100 Rieti, Italy
| | | |
Collapse
|
10
|
Pu Z, Shimizu Y, Tsuzuki K, Suzuki J, Hayashida R, Kondo K, Fujikawa Y, Unno K, Ohashi K, Takefuji M, Bando YK, Ouchi N, Calvert JW, Shibata R, Murohara T. Important Role of Concomitant Lymphangiogenesis for Reparative Angiogenesis in Hindlimb Ischemia. Arterioscler Thromb Vasc Biol 2021; 41:2006-2018. [PMID: 33910373 DOI: 10.1161/atvbaha.121.316191] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Zhongyue Pu
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (Z.P., Y.S., K.T., J.S., R.H., K.K., Y.F., K.U., K.O., M.T., Y.K.B., N.O., R.S., T.M.)
| | - Yuuki Shimizu
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (Z.P., Y.S., K.T., J.S., R.H., K.K., Y.F., K.U., K.O., M.T., Y.K.B., N.O., R.S., T.M.)
| | - Kazuhito Tsuzuki
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (Z.P., Y.S., K.T., J.S., R.H., K.K., Y.F., K.U., K.O., M.T., Y.K.B., N.O., R.S., T.M.)
| | - Junya Suzuki
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (Z.P., Y.S., K.T., J.S., R.H., K.K., Y.F., K.U., K.O., M.T., Y.K.B., N.O., R.S., T.M.)
| | - Ryo Hayashida
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (Z.P., Y.S., K.T., J.S., R.H., K.K., Y.F., K.U., K.O., M.T., Y.K.B., N.O., R.S., T.M.)
| | - Kazuhisa Kondo
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (Z.P., Y.S., K.T., J.S., R.H., K.K., Y.F., K.U., K.O., M.T., Y.K.B., N.O., R.S., T.M.)
| | - Yusuke Fujikawa
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (Z.P., Y.S., K.T., J.S., R.H., K.K., Y.F., K.U., K.O., M.T., Y.K.B., N.O., R.S., T.M.)
| | - Kazumasa Unno
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (Z.P., Y.S., K.T., J.S., R.H., K.K., Y.F., K.U., K.O., M.T., Y.K.B., N.O., R.S., T.M.)
| | - Koji Ohashi
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (Z.P., Y.S., K.T., J.S., R.H., K.K., Y.F., K.U., K.O., M.T., Y.K.B., N.O., R.S., T.M.)
| | - Mikito Takefuji
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (Z.P., Y.S., K.T., J.S., R.H., K.K., Y.F., K.U., K.O., M.T., Y.K.B., N.O., R.S., T.M.)
| | - Yasuko K Bando
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (Z.P., Y.S., K.T., J.S., R.H., K.K., Y.F., K.U., K.O., M.T., Y.K.B., N.O., R.S., T.M.)
| | - Noriyuki Ouchi
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (Z.P., Y.S., K.T., J.S., R.H., K.K., Y.F., K.U., K.O., M.T., Y.K.B., N.O., R.S., T.M.)
| | - John W Calvert
- Division of Cardiothoracic Surgery, Department of Surgery, Carlyle Fraser Heart Center, Emory University School of Medicine, Atlanta, GA (J.W.C.)
| | - Rei Shibata
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (Z.P., Y.S., K.T., J.S., R.H., K.K., Y.F., K.U., K.O., M.T., Y.K.B., N.O., R.S., T.M.)
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (Z.P., Y.S., K.T., J.S., R.H., K.K., Y.F., K.U., K.O., M.T., Y.K.B., N.O., R.S., T.M.)
| |
Collapse
|
11
|
Current Status of Angiogenic Cell Therapy and Related Strategies Applied in Critical Limb Ischemia. Int J Mol Sci 2021; 22:ijms22052335. [PMID: 33652743 PMCID: PMC7956816 DOI: 10.3390/ijms22052335] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/20/2021] [Accepted: 02/23/2021] [Indexed: 02/06/2023] Open
Abstract
Critical limb ischemia (CLI) constitutes the most severe form of peripheral arterial disease (PAD), it is characterized by progressive blockade of arterial vessels, commonly correlated to atherosclerosis. Currently, revascularization strategies (bypass grafting, angioplasty) remain the first option for CLI patients, although less than 45% of them are eligible for surgical intervention mainly due to associated comorbidities. Moreover, patients usually require amputation in the short-term. Angiogenic cell therapy has arisen as a promising alternative for these "no-option" patients, with many studies demonstrating the potential of stem cells to enhance revascularization by promoting vessel formation and blood flow recovery in ischemic tissues. Herein, we provide an overview of studies focused on the use of angiogenic cell therapies in CLI in the last years, from approaches testing different cell types in animal/pre-clinical models of CLI, to the clinical trials currently under evaluation. Furthermore, recent alternatives related to stem cell therapies such as the use of secretomes, exosomes, or even microRNA, will be also described.
Collapse
|
12
|
Niu Y, Wang Z, Shi Y, Dong L, Wang C. Modulating macrophage activities to promote endogenous bone regeneration: Biological mechanisms and engineering approaches. Bioact Mater 2021; 6:244-261. [PMID: 32913932 PMCID: PMC7451865 DOI: 10.1016/j.bioactmat.2020.08.012] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/07/2020] [Accepted: 08/11/2020] [Indexed: 02/08/2023] Open
Abstract
A coordinated interaction between osteogenesis and osteoimmune microenvironment is essential for successful bone healing. In particular, macrophages play a central regulatory role in all stages of bone repair. Depending on the signals they sense, these highly plastic cells can mediate the host immune response against the exterior signals of molecular stimuli and implanted scaffolds, to exert regenerative potency to a varying extent. In this article, we first encapsulate the immunomodulatory functions of macrophages during bone regeneration into three aspects, as sweeper, mediator and instructor. We introduce the phagocytic role of macrophages in different bone healing periods ('sweeper') and overview a variety of paracrine cytokines released by macrophages either mediating cell mobilisation, vascularisation and matrix remodelling ('mediator'), or directly driving the osteogenic differentiation of bone progenitors and bone repair ('instructor'). Then, we systematically classify and discuss the emerging engineering strategies to recruit, activate and modulate the phenotype transition of macrophages, to exploit the power of endogenous macrophages to enhance the performance of engineered bone tissue.
Collapse
Affiliation(s)
- Yiming Niu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Macau SAR, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210093, China
| | - Zhenzhen Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Macau SAR, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210093, China
| | - Yuchen Shi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Macau SAR, China
| | - Lei Dong
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210093, China
| | - Chunming Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Macau SAR, China
| |
Collapse
|
13
|
Suzuki J, Shimizu Y, Tsuzuki K, Pu Z, Narita S, Yamaguchi S, Katagiri T, Iwata E, Masutomi T, Fujikawa Y, Shibata R, Murohara T. No influence on tumor growth by intramuscular injection of adipose-derived regenerative cells: safety evaluation of therapeutic angiogenesis with cell therapy. Am J Physiol Heart Circ Physiol 2021; 320:H447-H457. [PMID: 33185457 DOI: 10.1152/ajpheart.00564.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 11/11/2020] [Accepted: 11/11/2020] [Indexed: 12/12/2022]
Abstract
Therapeutic angiogenesis with autologous stem/progenitor cells is a promising novel strategy for treatment of severe ischemic diseases. Human clinical trials utilizing autologous adipose-derived regenerative cells (ADRCs) have not reported treatment-related critical adverse effects thus far. However, there is still a large knowledge gap regarding whether treatment of ischemic diseases with angiogenic therapy using ADRCs would promote unfavorable angiogenesis associated with tumors in vivo. Herein, we addressed this clinical question using a mouse hindlimb ischemia (HLI) and simultaneous remote tumor implantation model. C57BL/6J background wild-type mice were injected with murine B16F10 melanoma cells on their back, 1 day before ischemic surgery. These mice were subjected to surgical unilateral hindlimb ischemia, followed by ADRC implantation or PBS injection into the hindlimb ischemic muscles on the next day. Intramuscular implantation of ADRCs enhanced tissue capillary density and blood flow examined by a laser Doppler blood perfusion analysis in hind limb. However, this therapeutic regimen for ischemic limb using ADRCs did not affect remote melanoma growth nor the density of its feeder artery, angiogenesis, and lymphatic vessels compared with the PBS group. In addition, no distant metastases were detected in any of the mice regardless of the group. In conclusion, local implantation of ADRCs promotes angiogenesis in response to tissue ischemia in the hindlimb without promoting remote tumor growth and related angio/lymphangiogenesis. Therapeutic angiogenesis to the ischemic hindlimb using ADRCs seems to be safe regarding remote tumor growth.NEW & NOTEWORTHY In this study, we demonstrated that local injection of ADRCs can promote angiogenesis in response to tissue ischemia without promoting remote tumor growth in a mouse model. Our findings indicate that therapeutic angiogenesis to the ischemic hindlimb using ADRCs seems to be safe regarding remote tumor growth.
Collapse
Affiliation(s)
- Junya Suzuki
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuuki Shimizu
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuhito Tsuzuki
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Zhongyue Pu
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shingo Narita
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shukuro Yamaguchi
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takeshi Katagiri
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Etsuo Iwata
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomohiro Masutomi
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yusuke Fujikawa
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Rei Shibata
- Department of Advanced Cardiovascular Therapeutics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
14
|
Sun X, Wang X, Zhao Z, Chen J, Li C, Zhao G. Paeoniflorin accelerates foot wound healing in diabetic rats though activating the Nrf2 pathway. Acta Histochem 2020; 122:151649. [PMID: 33166863 DOI: 10.1016/j.acthis.2020.151649] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 10/25/2020] [Accepted: 10/26/2020] [Indexed: 12/26/2022]
Abstract
As one of the most frequent diabetic complications, diabetic foot ulcer (DFU) can cause limb ischemia or even amputation. Paeoniflorin (PF) has been reported to possess many kinds of biological functions, such as antioxidant and anti-inflammatory effects. However, the role of PF in DFU remains unknown. In this study, streptozotocin (STZ)-induced diabetic rat models and high glucose (HG)-treated Human immortalized keratinocytes (HaCaT) cells were established. Histological analysis, immunohistochemistry, Electrophoretic mobility shift assay, MTT assay, TUNEL assay, oxidative stress analysis, ELISA assay and western blot were used to investigate the role and underlying mechanisms of PF on healing in DFU. Our results showed that the STZ-induced diabetic rats had delayed wound healing compared with the normal rats, exhibited by intense oxidative DNA damage, low vascular endothelial growth factor (VEGF) and transforming growth factor β1 (TGF-β1) expression, as well as increased apoptosis. PF treatment activated the expression of nuclear factor-E2-related factor 2 (Nrf2) and improved wound healing in DFU rats. Our in vitro experiments confirmed that PF accelerated wound healing through the Nrf2 pathway under hyperglycemic conditions, with alleviated oxidative stress, increased cell proliferation and migration, decreased apoptosis, and increased the expression of VEGF and TGF-β1. Our study demonstrates the therapeutic benefits of PF in diabetic wound healing, which provides a reference for future clinical trials using PF in DFU treatment.
Collapse
Affiliation(s)
- Xiaolong Sun
- The Second Department of Surgery, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, People's Republic of China.
| | - Xu Wang
- The Second Department of Surgery, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, People's Republic of China
| | - Zhenyu Zhao
- The Second Department of Surgery, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, People's Republic of China
| | - Jing Chen
- The Second Department of Surgery, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, People's Republic of China
| | - Cheng Li
- The Second Department of Surgery, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, People's Republic of China
| | - Gang Zhao
- Department of Peripheral Vascular Disease, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, People's Republic of China
| |
Collapse
|
15
|
Katagiri T, Kondo K, Shibata R, Hayashida R, Shintani S, Yamaguchi S, Shimizu Y, Unno K, Kikuchi R, Kodama A, Takanari K, Kamei Y, Komori K, Murohara T. Therapeutic angiogenesis using autologous adipose-derived regenerative cells in patients with critical limb ischaemia in Japan: a clinical pilot study. Sci Rep 2020; 10:16045. [PMID: 32994527 PMCID: PMC7525513 DOI: 10.1038/s41598-020-73096-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 09/09/2020] [Indexed: 12/12/2022] Open
Abstract
Adipose-derived regenerative cell (ADRC) is a promising alternative source of autologous somatic stem cells for the repair of damaged tissue. This study aimed to assess the safety and feasibility of autologous ADRC implantation for therapeutic angiogenesis in patients with critical limb ischaemia (CLI). A clinical pilot study—Therapeutic Angiogenesis by Cell Transplantation using ADRCs (TACT-ADRC) study—was initiated in Japan. Adipose tissue was obtained by ordinary liposuction method. Isolated ADRCs were injected into the ischaemic limb. We performed TACT-ADRC procedure in five patients with CLI. At 6 months, no adverse events related to the TACT-ADRC were observed. No patients required major limb amputation, and ischaemic ulcers were partly or completely healed during the 6-month follow-up. In all cases, significant clinical improvements were seen in terms of rest pain and 6-min walking distance. Numbers of circulating CD34+ and CD133+ cells markers of progenitor cell persistently increased after ADRC implantation. The ratio of VEGF-A165b (an anti-angiogenic isoform of VEGF) to total VEGF-A in plasma significantly decreased after ADRC implantation. In vitro experiments, cultured with ADRC-conditioned media (CM) resulted in increased total VEGF-A and decreased VEGF-A165b in C2C12 cells, but not in macrophages. ADRC-CM also increased CD206+ cells expression and decreased TNF-α in macrophages. Autologous ADRC implantation was safe and effective in patients with CLI and could repair damaged tissue via its ability to promote angiogenesis and suppress tissue inflammation.
Collapse
Affiliation(s)
- Takeshi Katagiri
- Department of Cardiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Kazuhisa Kondo
- Department of Cardiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Rei Shibata
- Department of Advanced Cardiovascular Therapeutics, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya, 466-8550, Japan.
| | - Ryo Hayashida
- Department of Cardiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Satoshi Shintani
- Department of Cardiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Shukuro Yamaguchi
- Department of Cardiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Yuuki Shimizu
- Department of Cardiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Kazumasa Unno
- Department of Cardiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Ryosuke Kikuchi
- Department of Medical Technique, Nagoya University Hospital, Nagoya, Japan
| | - Akio Kodama
- Department of Vascular Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Keisuke Takanari
- Department of Plastic and Reconstructive Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuzuru Kamei
- Department of Plastic and Reconstructive Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kimihiro Komori
- Department of Vascular Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| |
Collapse
|
16
|
Vidal L, Brennan MÁ, Krissian S, De Lima J, Hoornaert A, Rosset P, Fellah BH, Layrolle P. In situ production of pre-vascularized synthetic bone grafts for regenerating critical-sized defects in rabbits. Acta Biomater 2020; 114:384-394. [PMID: 32688088 DOI: 10.1016/j.actbio.2020.07.030] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 06/16/2020] [Accepted: 07/14/2020] [Indexed: 12/13/2022]
Abstract
Reconstructing large bone defects caused by severe trauma or resection of tumors remains a challenge for surgeons. A fibula free flap and its vascularized bed can be transplanted to the reconstruction site to achieve healing. However, this technique adds morbidity, and requires microsurgery and sculpting of the bone tissue to adapt the graft to both the vasculature and the anatomy of the defect. The aim of the current study was to evaluate an alternative approach consisting of the in situ production of a pre-vascularized synthetic bone graft and its subsequent transplantation to a critical-sized bone defect. 3D printed chambers containing biphasic calcium phosphate (BCP) granules, perfused by a local vascular pedicle, with or without the addition of stromal vascular fraction (SVF), were subcutaneously implanted into New Zealand White female rabbits. SVF was prepared extemporaneously from autologous adipose tissue, the vascular pedicle was isolated from the inguinal site, while BCP granules alone served as a control group. After 8 weeks, the constructs containing a vascular pedicle exhibited abundant neovascularization with blood vessels sprouting from the pedicle, leading to significantly increased vascularization compared to BCP controls. Pre-vascularized synthetic bone grafts were then transplanted into 15 mm critical-sized segmental ulnar defects for a further 8 weeks. Micro-CT and decalcified histology revealed that pre-vascularization of synthetic bone grafts led to enhanced bone regeneration. This pre-clinical study demonstrates the feasibility and efficacy of the in situ production of pre-vascularized synthetic bone grafts for regenerating large bone defects, thereby addressing an important clinical need. STATEMENT OF SIGNIFICANCE: The current gold standard in large bone defect regeneration is vascularized fibula grafting. An alternative approach consisting of in situ production of a pre-vascularized synthetic bone graft and its subsequent transplantation to a bone defect is presented here. 3D printed chambers were filled with biphasic calcium phosphate granules, supplemented with autologous stromal vascular fraction and an axial vascular pedicle and subcutaneously implanted in inguinal sites. These pre-vascularized synthetic grafts were then transplanted into critical-sized segmental ulnar defects. Micro-CT and decalcified histology revealed that the pre-vascularized synthetic bone grafts led to higher bone regeneration than non-vascularized constructs. An alternative to vascularized fibula grafting is provided and may address an important clinical need for large bone defect reconstruction.
Collapse
|
17
|
Zhao X, Guo J, Zhang F, Zhang J, Liu D, Hu W, Yin H, Jin L. Therapeutic application of adipose-derived stromal vascular fraction in diabetic foot. Stem Cell Res Ther 2020; 11:394. [PMID: 32928305 PMCID: PMC7488783 DOI: 10.1186/s13287-020-01825-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/15/2020] [Accepted: 07/10/2020] [Indexed: 02/06/2023] Open
Abstract
Diabetic foot is one of the severest complications of diabetes. In severe cases, this disease may be lead to amputation or even death due to secondary infection and ischemic necrosis. Since the ineffectiveness of traditional therapy, autologous stem cell transplantation has been used to treat diabetic foot. This simple, safe, and effective therapy is expected to be applied and promoted in the future.In this review, we described the detailed pathogenesis of diabetic foot and the common clinical treatments currently used. We also revealed vascular remodeling as the potential mechanism of therapeutic functions of adipose-derived stromal vascular fraction (SVF) in treating diabetic foot.
Collapse
Affiliation(s)
- Xiansheng Zhao
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, Jiangsu Province, China
| | - Jiamin Guo
- Irell & Manella Graduate School of Biological Sciences, City of Hope National Medical Center, California, 91010, USA
| | - Fangfang Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, Jiangsu Province, China
| | - Jue Zhang
- Department of Endocrinology, The Affiliated ZhongDa Hospital of Southeast University, Nanjing, 210009, Jiangsu Province, China
| | - Delin Liu
- Department of Endocrinology, The Affiliated ZhongDa Hospital of Southeast University, Nanjing, 210009, Jiangsu Province, China
| | - Wenjun Hu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, Jiangsu Province, China.
| | - Han Yin
- Department of Endocrinology, The Affiliated ZhongDa Hospital of Southeast University, Nanjing, 210009, Jiangsu Province, China.
| | - Liang Jin
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, Jiangsu Province, China.
| |
Collapse
|
18
|
Azimi MS, Motherwell JM, Dutreil M, Fishel RL, Nice M, Hodges NA, Bunnell BA, Katz A, Murfee WL. A novel tissue culture model for evaluating the effect of aging on stem cell fate in adult microvascular networks. GeroScience 2020; 42:515-526. [PMID: 32206968 PMCID: PMC7205973 DOI: 10.1007/s11357-020-00178-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 03/04/2020] [Indexed: 12/18/2022] Open
Abstract
In vitro models of angiogenesis are valuable tools for understanding the underlying mechanisms of pathological conditions and for the preclinical evaluation of therapies. Our laboratory developed the rat mesentery culture model as a new tool for investigating mechanistic cell-cell interactions at specific locations across intact blood and lymphatic microvascular networks ex vivo. The objective of this study was to report a method for evaluating the effect of aging on human stem cell differentiation into pericytes during angiogenesis in cultured microvascular networks. DiI labeled exogenous stem cells were seeded onto harvested adult Wistar rat mesenteric tissues and cultured in alpha-MEM + 1% serum for up to 5 days according to four experimental groups: (1) adult human adipose-derived stem cells (hASCs), (2) aged hASCs, (3) adult human bone marrow-derived stem cells (hBMSCs), and (4) aged hBMSCs. Angiogenesis per experimental group was supported by observation of increased vessel density and capillary sprouting. For each tissue per experimental group, a subset of cells was observed in typical pericyte location wrapped along blood vessels. Stem cell differentiation into pericytes was supported by the adoption of elongated pericyte morphology along endothelial cells and positive NG2 labeling. The percentage of cells in pericyte locations was not significantly different across the experimental groups, suggesting that aged mesenchymal stem cells are able to retain their differentiation capacity. Our results showcase an application of the rat mesentery culture model for aging research and the evaluation of stem cell fate within intact microvascular networks.
Collapse
Affiliation(s)
- Mohammad S Azimi
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, 70118, USA
| | - Jessica M Motherwell
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, 70118, USA
- J. Crayton Pruitt Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611, USA
| | - Maria Dutreil
- Tulane Center for Stem Cell Research & Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Ryan L Fishel
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, 70118, USA
| | - Matthew Nice
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, 70118, USA
| | - Nicholas A Hodges
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, 70118, USA
- J. Crayton Pruitt Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611, USA
| | - Bruce A Bunnell
- Tulane Center for Stem Cell Research & Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, 70112, USA
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Adam Katz
- Depart of Surgery, University of Florida School of Medicine, Gainesville, FL, 32611, USA
| | - Walter L Murfee
- J. Crayton Pruitt Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611, USA.
| |
Collapse
|
19
|
Sun Y, Chen S, Zhang X, Pei M. Significance of Cellular Cross-Talk in Stromal Vascular Fraction of Adipose Tissue in Neovascularization. Arterioscler Thromb Vasc Biol 2020; 39:1034-1044. [PMID: 31018663 DOI: 10.1161/atvbaha.119.312425] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Adult stem cell-based therapy has been regarded as a promising treatment for tissue ischemia because of its ability to promote new blood vessel formation. Bone marrow-derived mesenchymal stem cells are the most used angiogenic cells for therapeutic neovascularization, yet the side effects and low efficacy have limited their clinical application. Adipose stromal vascular fraction is an easily accessible, heterogeneous cell system comprised of endothelial, stromal, and hematopoietic cell lineages, which has been shown to spontaneously form robust, patent, and functional vasculatures in vivo. However, the characteristics of each cell population and their specific roles in neovascularization remain an area of ongoing investigation. In this review, we summarize the functional capabilities of various stromal vascular fraction constituents during the process of neovascularization and attempt to analyze whether the cross-talk between these constituents generates a synergetic effect, thus contributing to the development of new potential therapeutic strategies to promote neovascularization.
Collapse
Affiliation(s)
- Yuan Sun
- From the Department of Vascular Surgery, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Jiangsu, China (Y.S., X.Z.); Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics (Y.S., M.P.), Exercise Physiology (M.P.), and WVU Cancer Institute, Robert C. Byrd Health Sciences Center (M.P.), West Virginia University, Morgantown; and Department of Orthopaedics, Chengdu Military General Hospital, Chengdu, Sichuan, China (S.C.)
| | - Song Chen
- From the Department of Vascular Surgery, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Jiangsu, China (Y.S., X.Z.); Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics (Y.S., M.P.), Exercise Physiology (M.P.), and WVU Cancer Institute, Robert C. Byrd Health Sciences Center (M.P.), West Virginia University, Morgantown; and Department of Orthopaedics, Chengdu Military General Hospital, Chengdu, Sichuan, China (S.C.)
| | - Xicheng Zhang
- From the Department of Vascular Surgery, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Jiangsu, China (Y.S., X.Z.); Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics (Y.S., M.P.), Exercise Physiology (M.P.), and WVU Cancer Institute, Robert C. Byrd Health Sciences Center (M.P.), West Virginia University, Morgantown; and Department of Orthopaedics, Chengdu Military General Hospital, Chengdu, Sichuan, China (S.C.)
| | - Ming Pei
- From the Department of Vascular Surgery, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Jiangsu, China (Y.S., X.Z.); Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics (Y.S., M.P.), Exercise Physiology (M.P.), and WVU Cancer Institute, Robert C. Byrd Health Sciences Center (M.P.), West Virginia University, Morgantown; and Department of Orthopaedics, Chengdu Military General Hospital, Chengdu, Sichuan, China (S.C.)
| |
Collapse
|
20
|
Murohara T. Therapeutic Angiogenesis with Somatic Stem Cell Transplantation. Korean Circ J 2020; 50:12-21. [PMID: 31854154 PMCID: PMC6923231 DOI: 10.4070/kcj.2019.0288] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 09/18/2019] [Indexed: 12/19/2022] Open
Abstract
Therapeutic angiogenesis is an important strategy to rescue ischemic tissues in patients with critical limb ischemia having no other treatment option such as endovascular angioplasty or bypass surgery. Studies indicated so far possibilities of therapeutic angiogenesis using autologous bone marrow mononuclear cells, CD34⁺ cells, peripheral blood mononuclear cells, adipose-derived stem/progenitor cells, and etc. Recent studies indicated that subcutaneous adipose tissue contains stem/progenitor cells that can give rise to several mesenchymal lineage cells. Moreover, these mesenchymal progenitor cells release a variety of angiogenic growth factors including vascular endothelial growth factor, fibroblast growth factor, hepatocyte growth factor and chemokine stromal cell-derived factor-1. Subcutaneous adipose tissues can be harvested by less invasive technique. These biological properties of adipose-derived regenerative cells (ADRCs) implicate that autologous subcutaneous adipose tissue would be a useful cell source for therapeutic angiogenesis in humans. In this review, I would like to discuss biological properties and future perspective of ADRCs-mediated therapeutic angiogenesis.
Collapse
Affiliation(s)
- Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| |
Collapse
|
21
|
Li X, Ma T, Sun J, Shen M, Xue X, Chen Y, Zhang Z. Harnessing the secretome of adipose-derived stem cells in the treatment of ischemic heart diseases. Stem Cell Res Ther 2019; 10:196. [PMID: 31248452 PMCID: PMC6598280 DOI: 10.1186/s13287-019-1289-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Adipose-derived stem cells (ASCs) are promising therapeutic cells for ischemic heart diseases, due to the ease and efficiency of acquisition, the potential of myocardial lineage differentiation, and the paracrine effects. Recently, many researchers have claimed that the ASC-based myocardial repair is mainly attributed to its paracrine effects, including the anti-apoptosis, pro-angiogenesis, anti-inflammation effects, and the inhibition of fibrosis, rather than the direct differentiation into cardiovascular lineage cells. However, the usage of ASCs comes with the problems of low cardiac retention and survival after transplantation, like other stem cells, which compromises the effectiveness of the therapy. To overcome these drawbacks, researchers have proposed various strategies for improving survival rate and ensuring sustained paracrine secretion. They also investigated the safety and efficacy of phase I and II clinical trials of ASC-based therapy for cardiovascular diseases. In this review, we will discuss the characterization and paracrine effects of ASCs on myocardial repair, followed by the strategies for stimulating the paracrine secretion of ASCs, and finally their clinical usage.
Collapse
Affiliation(s)
- Xiaoting Li
- Department of Cardiology, The Second Affiliated Hospital of Soochow University, No.1055, Sanxiang Road, Suzhou, 215004, China
| | - Teng Ma
- Department of Cardiovascular Surgery, The First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, No.899, Pinghai Road, Suzhou, 215006, China
| | - Jiacheng Sun
- Department of Cardiovascular Surgery, The First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, No.899, Pinghai Road, Suzhou, 215006, China
| | - Mingjing Shen
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Soochow University, No.1055, Sanxiang Road, Suzhou, 215004, China
| | - Xiang Xue
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Soochow University, No.1055, Sanxiang Road, Suzhou, 215004, China
| | - Yongbing Chen
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Soochow University, No.1055, Sanxiang Road, Suzhou, 215004, China.
| | - Zhiwei Zhang
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Soochow University, No.1055, Sanxiang Road, Suzhou, 215004, China.
| |
Collapse
|
22
|
Chen L, Wang ZC, Ma JJ, Sun WJ, Wang SW, Gu ZC, Yang X. Autologous nanofat transplantation accelerates foot wound healing in diabetic rats. Regen Med 2019; 14:231-241. [PMID: 30810491 DOI: 10.2217/rme-2018-0169] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Aim: This study explored the effects of local transplantation of autologous nanofat in the treatment of rats with diabetic foot wounds. Materials & methods: Nanofat was transplanted into the left foot wound of diabetic rats. Phosphate-buffered saline injection in the right served as control. We measured wound size, the extent of epithelization, microvessel density and the expression levels of cytokines at six different time-points postoperation. Results: Compared with the control feet, nanofat-treated feet had significantly smaller wound areas at 7 and 9 days after grafting and showed better re-epithelialization, a greater number of microvessels and higher levels of angiogenic factor expression. Conclusion: This research shows that autologous nanofat transplantation can promote diabetic foot wound healing in rats.
Collapse
Affiliation(s)
- Li Chen
- Department of Plastic & Reconstructive Surgery, Sir Run Run Shaw Hospital, Medical College, Zhejiang University, Hang Zhou 310016, PR China
| | - Zheng-Cai Wang
- Department of Plastic & Reconstructive Surgery, Sir Run Run Shaw Hospital, Medical College, Zhejiang University, Hang Zhou 310016, PR China
| | - Jing-Jing Ma
- Department of Plastic & Reconstructive Surgery, Sir Run Run Shaw Hospital, Medical College, Zhejiang University, Hang Zhou 310016, PR China
| | - Wen-Jia Sun
- Department of Plastic & Reconstructive Surgery, Sir Run Run Shaw Hospital, Medical College, Zhejiang University, Hang Zhou 310016, PR China
| | - Shao-Wen Wang
- Department of General Surgery, Sir Run Run Shaw Hospital, Medical College, Zhejiang University, Hang Zhou 310016, PR China
| | - Zi-Chun Gu
- Department of Plastic & Reconstructive Surgery, Sir Run Run Shaw Hospital, Medical College, Zhejiang University, Hang Zhou 310016, PR China
| | - Xuan Yang
- Department of Plastic & Reconstructive Surgery, Sir Run Run Shaw Hospital, Medical College, Zhejiang University, Hang Zhou 310016, PR China
| |
Collapse
|
23
|
Endothelial Progenitor Cells Biology in Diabetes Mellitus and Peripheral Arterial Disease and their Therapeutic Potential. Stem Cell Rev Rep 2018; 15:157-165. [DOI: 10.1007/s12015-018-9863-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
24
|
Foubert P, Liu M, Anderson S, Rajoria R, Gutierrez D, Zafra D, Tenenhaus M, Fraser JK. Preclinical assessment of safety and efficacy of intravenous delivery of autologous adipose-derived regenerative cells (ADRCs) in the treatment of severe thermal burns using a porcine model. Burns 2018; 44:1531-1542. [PMID: 29958745 DOI: 10.1016/j.burns.2018.05.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 04/10/2018] [Accepted: 05/16/2018] [Indexed: 02/06/2023]
Abstract
OBJECTIVE A number of studies have reported that application of autologous adipose-derived cell populations leads to improved outcome in different preclinical models of thermal burn injury. However, these studies were limited to assessment of relatively small injuries amounting to only ∼2% of total body surface area (TBSA) in which the complications associated with large burns (e.g.: systemic inflammation and the need for fluid resuscitation) are absent. In anticipation of translating this approach to a clinical trial in which these complications would be present we applied a preclinical model that more closely resembles a patient with large thermal burn injury requiring skin grafting. Thus, the present study used a porcine model to investigate safety and efficacy of intravenous delivery of ADRCs in the treatment of a complex burn injury comprising ∼20% TBSA and including both moderately deep (44%) partial and full thickness burns, and the injury associated with skin graft harvest. METHODS Two pairs of full thickness and partial thickness burns involving in total ∼20% TBSA were created on the back of Yorkshire pigs (n=15). Three days post-burn, full thickness wounds were excised and grafted with a 3:1 meshed autologous split thickness skin graft (STSG). Partial thickness wounds were not treated other than with dressings. Animals were then randomized to receive intravenous delivery of ADRCs (n=8) or vehicle control (n=7). Safety was assessed by monitoring systemic parameters (blood gases, hematology, and clinical chemistry) throughout the course of the study. Wound healing for both types of burn wound and for the skin graft donor sites was followed for 18days using wound imaging, histology, and trans-epidermal water loss (TEWL; skin barrier function assessment). RESULTS No serious adverse events related to ADRC infusion were noted in any of the animals. Delivery of ADRCs appeared to be safe with none of the systemic safety parameters worsened compared to the control group. TEWL and histological analyses revealed that ADRC treatment was associated with significantly accelerated healing of skin graft (27.1% vs. 1.1% on Day 5 post-grafting), donor site (52.8% vs. 33.1% on Day 5 post-excision) and partial thickness burn (81.8% vs. 59.8% on Day 18 post-treatment). Data also suggested that ADRC treatment improved parameters associated with skin graft elasticity. CONCLUSIONS This study demonstrated that intravenous delivery of autologous ADRCs appears to be a safe and feasible approach to the treatment of large burns and supports the use of ADRCs as an adjunct therapy to skin grafting in patients with severe burns.
Collapse
Affiliation(s)
| | - Mike Liu
- Cytori Therapeutics Inc, San Diego, CA, USA
| | | | | | | | | | - Mayer Tenenhaus
- UCSD Medical Center, University of California, San Diego, CA, USA
| | | |
Collapse
|
25
|
Jing X, Yin W, Tian H, Chen M, Yao X, Zhu W, Guo F, Ye Y. Icariin doped bioactive glasses seeded with rat adipose-derived stem cells to promote bone repair via enhanced osteogenic and angiogenic activities. Life Sci 2018; 202:52-60. [PMID: 29471105 DOI: 10.1016/j.lfs.2018.02.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Revised: 02/01/2018] [Accepted: 02/17/2018] [Indexed: 12/29/2022]
Abstract
AIMS Cell communication between mesenchymal stem cells and blood vessel cells are crucial for bone repair. We have previously shown that the phyto-molecule icariin significantly promoted osteogenic differentiation of rat adipose-derived stem cells (ASCs). In the present study, we aimed to investigate the relationship between icariin induced osteogenic differentiation of ASCs and angiogenesis of rat endothelial progenitor cells (EPCs). Besides, we used icariin doped 45S5 Bioglass seeded with ASCs to promote bone healing in rat calvarial bone defect models. MAIN METHODS The conditioned medium from undifferentiated ASCs (ASCs-CM) and icariin induced ASCs (Icariin-ASCs-CM) was obtained and the vascular endothelial growth factor (VEGF) protein secretion level was measured. The angiogenic capacity and molecular mechanism of ASC-CM and Icariin-ASCs-CM on rat EPCs was analyzed. Rat calvarial bone defect models were established and treated with scaffolds implantation. Micro-CT imaging, histological and immunohistological staining were performed on the isolated specimens at 12 weeks post-surgery. KEY FINDINGS VEGF protein expression was significantly increased after icariin treatment with the highest expression in the 10-7 M icariin group. Icariin-ASCs-CM obviously increased the angiogenesis of rat EPCs and this capacity was inhibited by a VEGF/VEGF receptor-specific binding inhibitor bevacizumab. Results of the in vivo investigations showed that all scaffolds promoted bone healing compared to the Control group. Icariin significantly improved the healing capacity of 45S5 Bioglass seeded with ASCs. SIGNIFICANCE Implantation of Icariin/45S5 Bioglass seeded with rat ASCs could obviously promote both osteogenesis and angiogenesis and therefore represents an ideal candidate bone substitutes for bone repair and regeneration.
Collapse
Affiliation(s)
- Xingzhi Jing
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Weifeng Yin
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Hongtao Tian
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Mengcun Chen
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xudong Yao
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Wentao Zhu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Fengjing Guo
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.
| | - Yaping Ye
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.
| |
Collapse
|
26
|
Foubert P, Zafra D, Liu M, Rajoria R, Gutierrez D, Tenenhaus M, Fraser JK. Autologous adipose-derived regenerative cell therapy modulates development of hypertrophic scarring in a red Duroc porcine model. Stem Cell Res Ther 2017; 8:261. [PMID: 29141687 PMCID: PMC5688645 DOI: 10.1186/s13287-017-0704-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 10/18/2017] [Accepted: 10/19/2017] [Indexed: 12/18/2022] Open
Abstract
Background Effective prevention and treatment of hypertrophic scars (HTSs), a common consequence of deep-partial thickness injury, remain a significant clinical challenge. Previous studies from our group have shown that autologous adipose-derived regenerative cells (ADRCs) represent a promising approach to improve wound healing and, thereby, impact HTS development. The purpose of this study was to assess the influence of local delivery of ADRCs immediately following deep-partial thickness cutaneous injury on HTS development in the red Duroc (RD) porcine model. Methods Bilateral pairs of deep-partial thickness excisional wounds (2 mm depth; 58 cm2 area) were created using an electric dermatome on RD pigs (n = 12). Autologous ADRCs were isolated from the inguinal fat pad and then sprayed directly onto the wound at a dose of 0.25 × 106 viable cells/cm2. The paired contralateral wound received vehicle control. Wound healing and development of HTS were assessed over 6 months using digital imaging, quantitative measurement of skin hardness and pigmentation, and histology. Results Data showed that ADRC treatment led to reduced scar hyperpigmentation compared to control (p < 0.05). Using the Durometer, at 2 and 6 months post-injury, skin hardness was 10–20% lower in ADRCs-treated wounds compared to control vehicle (p < 0.05). A similar trend was observed with the skin fibrometer. ADRC treatment promoted more normal collagen organization, improvement in the number of rete ridges (p < 0.01), longer elastic fiber length (p < 0.01), and reduced hypervascularity (blood vessel density; p < 0.05). ADRC treatment was associated with modulation of IL-6 expression within the wound/scar with upregulation 2 weeks after injury (wound healing phase) and downregulation at 2 months (early scarring phase) post-treatment compared to control Conclusions These findings support the potential therapeutic value of autologous ADRC administration for reduction of HTS development following deep-partial cutaneous injury. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0704-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Philippe Foubert
- Cytori Therapeutics Inc, 3020 Callan Road, 92121, San Diego, CA, USA.
| | - Diana Zafra
- Cytori Therapeutics Inc, 3020 Callan Road, 92121, San Diego, CA, USA
| | - Mike Liu
- Cytori Therapeutics Inc, 3020 Callan Road, 92121, San Diego, CA, USA
| | | | | | - Mayer Tenenhaus
- UCSD Medical Center, University of California, San Diego, CA, USA
| | - John K Fraser
- Cytori Therapeutics Inc, 3020 Callan Road, 92121, San Diego, CA, USA
| |
Collapse
|
27
|
Yang G, Lei Y, Inoue A, Piao L, Hu L, Jiang H, Sasaki T, Wu H, Xu W, Yu C, Zhao G, Ogasawara S, Okumura K, Kuzuya M, Cheng XW. Exenatide mitigated diet-induced vascular aging and atherosclerotic plaque growth in ApoE-deficient mice under chronic stress. Atherosclerosis 2017; 264:1-10. [PMID: 28734203 DOI: 10.1016/j.atherosclerosis.2017.07.014] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 07/03/2017] [Accepted: 07/12/2017] [Indexed: 01/30/2023]
Abstract
BACKGROUND AND AIMS Exposure to psychosocial stress is a risk factor for cardiovascular disorders. Because the glucagon-like peptide-1 (GLP-1) receptor (GLP-1R) agonist prevents cardiovascular injury, we investigated the beneficial effects and mechanism of the GLP-1 analogue exenatide on stress-related vascular senescence and atherosclerosis in apolipoprotein E-deficient (ApoE-/-) mice fed a high-fat (HF) diet. METHODS ApoE-/- mice fed the HF diet were assigned to non-stressed and immobilized-stress groups for 12 weeks. Mice fed the HF diet were divided into 2 groups and administered vehicle or exenatide for 12 weeks under stress conditions. RESULTS Chronic stress enhanced vascular endothelial senescence and atherosclerotic plaque growth. The stress increased the levels of plasma depeptidyl peptidase-4 activity and decreased the levels of plasma GLP-1 and both plasma and adipose adiponectin (APN). As compared with the mice subjected to stress alone, the exenatide-treated mice had decreased plaque microvessel density, macrophage accumulation, broken elastin, and enhanced plaque collagen volume, and lowered levels of peroxisome proliferator-activated receptor-α, gp91phox osteopontin, C-X-C chemokine receptor-4, toll-like receptor-2 (TLR2), TLR4, and cathepsins K, L, and S mRNAs and/or proteins. Exenatide reduced aortic matrix metalloproteinase-9 (MMP-9) and MMP-2 gene expression and activities. Exenatide also stimulated APN expression of preadipocytes and inhibited ox-low density lipoprotein-induced foam cell formation of monocytes in stressed mice. CONCLUSIONS These results indicate that the exenatide-mediated beneficial vascular actions are likely attributable, at least in part, to the enhancement of APN production and the attenuation of plaque oxidative stress, inflammation, and proteolysis in ApoE-/- mice under chronic stress.
Collapse
Affiliation(s)
- Guang Yang
- Department of Cardiology and ICU, Yanbian University Hospital, Yanjin, Jilin PR., 13000, China
| | - Yanna Lei
- Department of Cardiology and ICU, Yanbian University Hospital, Yanjin, Jilin PR., 13000, China
| | - Aiko Inoue
- Institute of Innovation for Future Society, Nagoya University, Nagoya, 4668550, Japan; Department of Community Health & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, 4668550, Japan
| | - Limei Piao
- Department of Cardiology and ICU, Yanbian University Hospital, Yanjin, Jilin PR., 13000, China; Institute of Innovation for Future Society, Nagoya University, Nagoya, 4668550, Japan
| | - Lina Hu
- Department of Public Health, Guilin Medical College, Guilin, Guangxi P. R., 541004, China
| | - Haiying Jiang
- Department of Cardiology and ICU, Yanbian University Hospital, Yanjin, Jilin PR., 13000, China
| | - Takeshi Sasaki
- Department of Anatomy and Neuroscience, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, 4313192, Japan
| | - Hongxian Wu
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Wenhu Xu
- Department of Cardiology and ICU, Yanbian University Hospital, Yanjin, Jilin PR., 13000, China; Institute of Innovation for Future Society, Nagoya University, Nagoya, 4668550, Japan
| | - Chenglin Yu
- Department of Cardiology and ICU, Yanbian University Hospital, Yanjin, Jilin PR., 13000, China; Institute of Innovation for Future Society, Nagoya University, Nagoya, 4668550, Japan
| | - Guangxian Zhao
- Department of Cardiology and ICU, Yanbian University Hospital, Yanjin, Jilin PR., 13000, China
| | - Shinyu Ogasawara
- Institute of Innovation for Future Society, Nagoya University, Nagoya, 4668550, Japan
| | - Kenji Okumura
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Masafumi Kuzuya
- Institute of Innovation for Future Society, Nagoya University, Nagoya, 4668550, Japan; Department of Community Health & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, 4668550, Japan
| | - Xian-Wu Cheng
- Department of Cardiology and ICU, Yanbian University Hospital, Yanjin, Jilin PR., 13000, China; Institute of Innovation for Future Society, Nagoya University, Nagoya, 4668550, Japan; Department of Community Health & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, 4668550, Japan; Department of Internal Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
28
|
Im GI. Bone marrow-derived stem/stromal cells and adipose tissue-derived stem/stromal cells: Their comparative efficacies and synergistic effects. J Biomed Mater Res A 2017; 105:2640-2648. [PMID: 28419760 DOI: 10.1002/jbm.a.36089] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 03/13/2017] [Accepted: 04/11/2017] [Indexed: 12/20/2022]
Abstract
Mesenchymal stem cells (MSCs) are heterogeneous cell populations that serve as reserves for tissue regeneration in the presence of disease or injury. Although MSCs are found in various tissues, bone marrow-derived stem/stromal cells (BMSCs) and adipose tissue-derived stem/stromal cells (ADSCs) have been most thoroughly investigated. Furthermore, ADSCs have recently emerged as an attractive source of MSCs due to their abundance and availability. BMSCs and ADSCs demonstrate similar morphological characteristics, but their in vitro characteristics and differentiation abilities appear to differ. In this review, the author summarizes and compares current knowledge on BMSCs and ADSCs with particular emphasis on in vitro expansion and osteogenic/angiogenic potential, and reviews knowledge of their synergistic effects when co-applied. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 2640-2648, 2017.
Collapse
Affiliation(s)
- Gun-Il Im
- Department of Orthopaedics, Dongguk University Ilsan Hospital, Goyang, Republic of Korea
| |
Collapse
|
29
|
Extracellular vesicles of ETV2 transfected fibroblasts stimulate endothelial cells and improve neovascularization in a murine model of hindlimb ischemia. Cytotechnology 2017; 69:801-814. [PMID: 28466428 DOI: 10.1007/s10616-017-0095-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 04/07/2017] [Indexed: 01/08/2023] Open
Abstract
Ischemia are common conditions related to lack of blood supply to tissues. Depending on the ischemic sites, ischemia can cause different diseases, such as hindlimb ischemia, heart infarction and stroke. This study aims to evaluate how extracellular vesicles (EVs) derived from ETV2 transfected fibroblasts affect endothelial cell proliferation and neovascularization in a murine model of hindlimb ischemia. Human fibroblasts were isolated and cultured under standard conditions and expanded to the 3th passage before use in experiments. Human fibroblasts were transduced with a viral vector containing the ETV2 gene. Transduced cells were selected by puromycin treatment. These cells were further cultured for collection of EVs, which were isolated from culture supernatant. Following co-culture with endothelial cells, EVs were evaluated for their effect on endothelial cell proliferation and were directly injected into ischemic tissues of a murine model of hindlimb ischemia. The results showed that EVs could induce endothelial cell proliferation in vitro and improved neovascularization in a murine model of hindlimb ischemia. Our results suggest that EVs derived from ETV2-transfected fibroblasts can be promising non-cellular products for the regeneration of blood vessels.
Collapse
|
30
|
Dubský M, Jirkovská A, Bem R, Nemcová A, Fejfarová V, Jude EB. Cell therapy of critical limb ischemia in diabetic patients - State of art. Diabetes Res Clin Pract 2017; 126:263-271. [PMID: 28288436 DOI: 10.1016/j.diabres.2017.02.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 12/19/2016] [Accepted: 02/22/2017] [Indexed: 01/15/2023]
Abstract
In this review we report on the state of cell therapy of critical limb ischemia (CLI) with respect to differences between diabetic and non-diabetic patients mainly from the clinical point of view. CLI is the most severe form of peripheral arterial disease and its diagnosis and treatment in diabetic patients is very difficult. The therapeutic effect of standard methods of CLI treatment is only partial - more than one third of diabetic patients are not eligible for standard revascularization; therefore, new therapeutic techniques such as cell therapy have been studied in clinical trials. Presence of CLI in patients with diabetic foot disease is associated with worse clinical outcomes such as lack of healing of foot ulcers, major amputations and premature mortality. A revascularization procedure cannot be successful as the only method in contrast to patients without diabetes, but it must always be part of a complex therapy focused not only on ischemia, but also on treatment of infection, off-loading, metabolic control of diabetes and nutrition, local therapy, etc. Therefore, the main criteria for cell therapy may vary in diabetic patients and non-diabetic persons and results of this treatment method should always be assessed in the context of ensuring comprehensive therapy. This review carries out an analysis of the source of precursor cells, route of administration and brings a brief report of published data with respect to diabetic and non-diabetic patients and our experience with autologous cell therapy of diabetic patients with CLI. Analysis of the studies in terms of diabetes is difficult, because in most of them sub-analysis for diabetic patients is not performed separately. The other problem is that it is not clear if diabetic patients received adequate complex treatment for their foot ulcers which can strongly affect the rate of major amputation as an outcome of CLI treatment.
Collapse
Affiliation(s)
- Michal Dubský
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| | | | - Robert Bem
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Andrea Nemcová
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | | | - Edward B Jude
- Diabetes Centre, Tameside Hospital NHS Foundation Trust and University of Manchester, Lancashire, UK
| |
Collapse
|
31
|
Joo HJ, Kim JH, Hong SJ. Adipose Tissue-Derived Stem Cells for Myocardial Regeneration. Korean Circ J 2017; 47:151-159. [PMID: 28382066 PMCID: PMC5378017 DOI: 10.4070/kcj.2016.0207] [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: 05/18/2016] [Revised: 07/09/2016] [Accepted: 07/15/2016] [Indexed: 12/14/2022] Open
Abstract
Over the past decade, stem cell therapy has been extensively studied for clinical application for heart diseases. Among various stem cells, adipose tissue-derived stem cell (ADSC) is still an attractive stem cell resource due to its abundance and easy accessibility. In vitro studies showed the multipotent differentiation potentials of ADSC, even differentiation into cardiomyocytes. Many pre-clinical animal studies have also demonstrated promising therapeutic results of ADSC. Furthermore, there were several clinical trials showing the positive results in acute myocardial infarction using ADSC. The present article covers the brief introduction, the suggested therapeutic mechanisms, application methods including cell dose and delivery, and human clinical trials of ADSC for myocardial regeneration.
Collapse
Affiliation(s)
- Hyung Joon Joo
- Department of Cardiology, Cardiovascular Center, Korea University Anam Hospital, Seoul, Korea
| | - Jong-Ho Kim
- Department of Cardiology, Cardiovascular Center, Korea University Anam Hospital, Seoul, Korea
| | - Soon Jun Hong
- Department of Cardiology, Cardiovascular Center, Korea University Anam Hospital, Seoul, Korea
| |
Collapse
|
32
|
Rybalko V, Hsieh PL, Ricles LM, Chung E, Farrar RP, Suggs LJ. Therapeutic potential of adipose-derived stem cells and macrophages for ischemic skeletal muscle repair. Regen Med 2017; 12:153-167. [PMID: 28244825 DOI: 10.2217/rme-2016-0094] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AIM Progressive ischemia due to peripheral artery disease causes muscle damage and reduced strength of the lower extremities. Autologous cell therapy is an attractive treatment to restore perfusion and improve muscle function. Adipose-derived stem cells (ASCs) have therapeutic potential in tissue repair, including polarizing effects on macrophages (MPs). MATERIALS & METHODS Co-culture systems of ASCs and MPs were analyzed for gene and protein expression modifications in ASC-conditioned MPs. Co-transplantation of MPs/ASCs in vivo led to improved skeletal muscle regeneration in a mouse model of peripheral artery disease. RESULTS ASCs/MPs therapy restored muscle function, increased perfusion and reduced inflammatory infiltrate. CONCLUSION Combined MPs/ASCs cell therapy is a promising approach to restore muscle function and stimulate local angiogenesis in the ischemic limb.
Collapse
Affiliation(s)
- Viktoriya Rybalko
- Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, 107 W Dean Keeton, Austin, TX 78712, USA
| | - Pei-Ling Hsieh
- Department of Kinesiology, The University of Texas at Austin, 1 University Station D3700, Austin, TX 78712, USA
| | - Laura M Ricles
- Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, 107 W Dean Keeton, Austin, TX 78712, USA
| | - Eunna Chung
- Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, 107 W Dean Keeton, Austin, TX 78712, USA
| | - Roger P Farrar
- Department of Kinesiology, The University of Texas at Austin, 1 University Station D3700, Austin, TX 78712, USA
| | - Laura J Suggs
- Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, 107 W Dean Keeton, Austin, TX 78712, USA
| |
Collapse
|
33
|
Carstens MH, Gómez A, Cortés R, Turner E, Pérez C, Ocon M, Correa D. Non-reconstructable peripheral vascular disease of the lower extremity in ten patients treated with adipose-derived stromal vascular fraction cells. Stem Cell Res 2017; 18:14-21. [DOI: 10.1016/j.scr.2016.12.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 11/04/2016] [Accepted: 12/02/2016] [Indexed: 12/22/2022] Open
|
34
|
Foubert P, Doyle-Eisele M, Gonzalez A, Berard F, Weber W, Zafra D, Alfonso Z, Zhao S, Tenenhaus M, Fraser JK. Development of a combined radiation and full thickness burn injury minipig model to study the effects of uncultured adipose-derived regenerative cell therapy in wound healing. Int J Radiat Biol 2016; 93:340-350. [DOI: 10.1080/09553002.2017.1242814] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
| | | | | | - Felipe Berard
- Lovelace Respiratory Research Institute, Albuquerque, NM, USA
| | - Waylon Weber
- Lovelace Respiratory Research Institute, Albuquerque, NM, USA
| | | | | | | | - Mayer Tenenhaus
- UCSD Medical Center, University of California, San Diego, CA, USA
| | | |
Collapse
|
35
|
Foubert P, Gonzalez AD, Teodosescu S, Berard F, Doyle-Eisele M, Yekkala K, Tenenhaus M, Fraser JK. Adipose-Derived Regenerative Cell Therapy for Burn Wound Healing: A Comparison of Two Delivery Methods. Adv Wound Care (New Rochelle) 2016; 5:288-298. [PMID: 27366590 DOI: 10.1089/wound.2015.0672] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Objective: The use of noncultured autologous stromal vascular fraction or clinical grade adipose-derived regenerative cells (ADRCs) is a promising strategy to promote wound healing and tissue repair. Nevertheless, issues regarding the optimal mode of administration remain unclear. The purpose of this study was to compare the effects of local injection and topical spray delivery of ADRCs in a porcine model of thermal burns. Approach: Full-thickness thermal burns were created on the dorsum of 10 Gottingen minipigs. Two days following injury, wounds underwent fascial excision and were randomized to receive control vehicle or freshly isolated autologous ADRCs delivered by either multiple injections into or surrounding the wound bed, or by spray onto the wound surface (0.25 × 106 viable cells/cm2). Healing was evaluated by planimetry, histopathology, and immunohistochemistry at day 7, 12, 16, 21, and 28 posttreatment. Results:In vitro analysis demonstrated that there was no substantial loss of cell number or viability attributable to the spray procedure. Planimetric assessment revealed that delivery of ADRCs by either local injection or topical spray increased wound reepithelialization relative to control at day 14. No significant difference in wound reepithelialization was observed between both delivery approaches. In addition, on day 7 posttreatment, blood vessel density was greater in wounds receiving local or topical spray ADRCs than in the wounds treated with vehicle control. Histopathologic analysis suggests that ADRC treatment may modulate the inflammatory response by reducing neutrophil infiltration at day 7 and 12 posttreatment, irrespective of the route of administration. Conclusions: These data demonstrate that local injection and spray delivery of ADRCs modulate inflammation and improve wound angiogenesis and epithelialization. Importantly, both delivery routes exhibited similar effects on wound healing. Given the greater ease-of-use associated with topical spray delivery, these data support the use of a spray system for autologous ADRC delivery.
Collapse
Affiliation(s)
| | | | | | - Felipe Berard
- Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | | | - Krishna Yekkala
- Department of Pathology, Toxikon Corporation, Bedford, Massachusetts
| | - Mayer Tenenhaus
- UCSD Medical Center, University of California, San Diego, California
| | | |
Collapse
|
36
|
The Effect of Adipose-Derived Stem Cells on Full-Thickness Skin Grafts. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1464725. [PMID: 27413735 PMCID: PMC4931067 DOI: 10.1155/2016/1464725] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 05/03/2016] [Accepted: 05/11/2016] [Indexed: 11/25/2022]
Abstract
Background. The purpose of this study was to evaluate the effects of ASCs on full-thickness skin grafts. Specifically, we investigated the anti-inflammatory effects of ASCs that are mediated via regulation of the phenotypes of activated macrophages. Methods. ASCs were isolated, cultured, and injected under full-thickness skin grafts in 15 rats (ASC group). An additional 15 rats served as controls (PBS group). Skin graft survival assessment and vascularization detection were assessed with H&E staining and laser Doppler blood flowmetry (LDF). The effects of ASCs on angiogenesis, anti-inflammation, collagen accumulation-promoting, and antiscarring were assessed. Results. We found that the skin graft survival rate was significantly increased in the ASC group. The neovascularization, collagen deposition, collagen type I to type III ratio, and levels of VEGF and TGF-β3 in the ASC group were markedly higher than those in the PBS group at day 14. Additionally, in the ASC group, the levels of iNOS, IL-1β, and TNF-α were remarkably decreased, whereas the levels of IL-10 and Arg-1 were substantially increased. Conclusions. Our results confirm that ASCs transplantation can effectively improve full-thickness skin graft survival. Additionally, the anti-inflammatory role of ASCs may indirectly contribute to skin graft survival via its effect on macrophage polarization.
Collapse
|
37
|
Jensen AR, Manning MM, Khaneki S, Drucker NA, Markel TA. Harvest tissue source does not alter the protective power of stromal cell therapy after intestinal ischemia and reperfusion injury. J Surg Res 2016; 204:361-370. [PMID: 27565072 DOI: 10.1016/j.jss.2016.05.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Revised: 04/29/2016] [Accepted: 05/03/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND Transplantation of mesenchymal stromal cells (MSCs) may be a novel treatment for intestinal ischemia. The optimal stromal cell source that could yield maximal protection after injury, however, has not been identified. We hypothesized that (1) MSCs would increase survival and mesenteric perfusion, preserve intestinal histologic architecture, and limit inflammation after intestinal ischemia and reperfusion (I/R) injury, and (2) MSCs harvested from different sources of tissue would have equivalent protective properties to the intestine after I/R inury. METHODS Adult male mice were anesthetized, and a midline laparotomy was performed. The intestines were eviscerated, the small bowel mesenteric root was identified, and baseline intestinal perfusion was determined using laser Doppler imaging. Intestinal ischemia was established by temporarily occluding the superior mesenteric artery for 60 min with a noncrushing clamp. After ischemia, the clamp was removed and the intestines were allowed to recover. Before abdominal closure, 2 × 10(6) human umbilical cord-derived MSCs, bone marrow-derived MSCs, or keratinocytes in 250 μL of phosphate-buffered saline vehicle were injected into the peritoneum. Animals were allowed to recover for 12 or 24 h (perfusion, histology, and inflammatory studies) or 7 d (survival studies). Survival data was analyzed using the log-rank test. Perfusion was expressed as a percentage of the baseline, and 12- and 24-h data was analyzed using one-way analysis of variance and the Student t-test. Nonparametric data was compared using the Mann-Whitney U-test. A P value of <0.05 was considered statistically significant. RESULTS All MSCs increased 7-d survival after I/R injury and were superior to vehicle and keratinocytes (P < 0.05). All MSCs increased mesenteric perfusion more than vehicle at 12 and 24 h after injury (P < 0.05). All MSCs provided superior perfusion compared with keratinocytes at 24 h after injury (P < 0.05). Administration of each MSC line improved intestinal histology after I/R injury (P < 0.05). Multiple proinflammatory chemokines were downregulated after the application of MSCs, suggesting a decreased inflammatory response after MSC therapy. CONCLUSIONS Transplantation of MSCs after intestinal I/R injury, irrespective of a tissue source, significantly increases survival and mesenteric perfusion and at the same time limits intestinal damage and inflammation. Further studies are needed to identify the mechanism that these cells use to promote improved outcomes after injury.
Collapse
Affiliation(s)
- Amanda R Jensen
- Department of Surgery, Section of Pediatric Surgery, Indianapolis, Indiana; Indiana University School of Medicine, Indianapolis, Indiana
| | - Morenci M Manning
- Department of Surgery, Section of Pediatric Surgery, Indianapolis, Indiana; Indiana University School of Medicine, Indianapolis, Indiana
| | - Sina Khaneki
- Department of Surgery, Section of Pediatric Surgery, Indianapolis, Indiana
| | - Natalie A Drucker
- Department of Surgery, Section of Pediatric Surgery, Indianapolis, Indiana; Indiana University School of Medicine, Indianapolis, Indiana
| | - Troy A Markel
- Department of Surgery, Section of Pediatric Surgery, Indianapolis, Indiana; Indiana University School of Medicine, Indianapolis, Indiana; Riley Hospital for Children at Indiana University Health, Indianapolis, Indiana.
| |
Collapse
|
38
|
Silvestre JS, Planat-Benard V, Casteilla L. Adipose tissue-derived therapeutic cells for peripheral artery diseases: the fatty blessing. Expert Opin Biol Ther 2016; 16:735-8. [PMID: 27097154 DOI: 10.1080/14712598.2016.1181170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Jean-Sébastien Silvestre
- a Inserm UMRS 970, Paris Cardiovascular Research Center , Paris Descartes University , Paris , France
| | - Valérie Planat-Benard
- b STROMALab , Toulouse University, EFS, ENVT, INSERM U1031, ERL CNRS 5311 , Toulouse , France
| | - Louis Casteilla
- b STROMALab , Toulouse University, EFS, ENVT, INSERM U1031, ERL CNRS 5311 , Toulouse , France
| |
Collapse
|
39
|
Prologo JD, Hawkins M, Gilliland C, Chinnadurai R, Harkey P, Chadid T, Lee Z, Brewster L. Interventional stem cell therapy. Clin Radiol 2016; 71:307-11. [PMID: 26874660 DOI: 10.1016/j.crad.2016.01.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 11/26/2015] [Accepted: 01/04/2016] [Indexed: 12/13/2022]
Abstract
The ability to deliver cells in appropriate doses to their targeted site of action is a well-known obstacle to optimising stem cell therapy. Systemic administration of cells results in pulmonary "trapping," which significantly decreases the number of available circulating cells to impact underlying disorders. Directed delivery of stem cells in interventional radiology may provide an additional option for bypassing the lungs, as well as introduce novel potential avenues for decreasing doses required to effect cellular therapy, efficiently obtain local paracrine effects, and/or to simplify targeting strategies.
Collapse
Affiliation(s)
- J D Prologo
- Division of Interventional Radiology and Image Guided Medicine, Emory University School of Medicine, 1364 Clifton Rd NE, Suite AG05, Atlanta, GA 30322, USA.
| | - M Hawkins
- Division of Interventional Radiology and Image Guided Medicine, Emory University School of Medicine, 1364 Clifton Rd NE, Suite AG05, Atlanta, GA 30322, USA
| | - C Gilliland
- Division of Interventional Radiology and Image Guided Medicine, Emory University School of Medicine, 1364 Clifton Rd NE, Suite AG05, Atlanta, GA 30322, USA
| | - R Chinnadurai
- Department of Hematology and Oncology, Emory University School of Medicine, Winship Cancer Institute, 1365B Clifton Rd NE, Suite B506, Atlanta, GA 30322, USA
| | - P Harkey
- Division of Musculoskeletal Radiology, Emory University School of Medicine, 1364 Clifton Rd NE, Suite AG05, Atlanta, GA 30322, USA
| | - T Chadid
- Department of Surgery, Emory University School of Medicine, 1364 Clifton Rd NE, Suite H100, Atlanta, GA 30322, USA
| | - Z Lee
- Department of Radiology, Case Western Reserve University College of Medicine, 11100 Euclid Avenue, Cleveland, OH 44106, USA
| | - Luke Brewster
- Department of Surgery, Emory University School of Medicine, 1364 Clifton Rd NE, Suite H100, Atlanta, GA 30322, USA; Department of Surgical and Research Services, Atlanta Veterans Medical Center, 1670 Clairmont Road, Decatur, GA 30033, USA
| |
Collapse
|
40
|
Foubert P, Barillas S, Gonzalez AD, Alfonso Z, Zhao S, Hakim I, Meschter C, Tenenhaus M, Fraser JK. Uncultured adipose-derived regenerative cells (ADRCs) seeded in collagen scaffold improves dermal regeneration, enhancing early vascularization and structural organization following thermal burns. Burns 2015; 41:1504-16. [PMID: 26059048 DOI: 10.1016/j.burns.2015.05.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 05/05/2015] [Accepted: 05/08/2015] [Indexed: 11/18/2022]
Abstract
OBJECTIVE Advances in tissue engineering have yielded a range of both natural and synthetic skin substitutes for burn wound healing application. Long-term viability of tissue-engineered skin substitutes requires the formation and maturation of neo-vessels to optimize survival and biointegration after implantation. A number of studies have demonstrated the capacity of Adipose Derived Regenerative Cells (ADRCs) to promote angiogenesis and modulate inflammation. On this basis, it was hypothesized that adding ADRCs to a collagen-based matrix (CBM) (i.e. Integra) would enhance formation and maturation of well-organized wound tissue in the setting of acute thermal burns. The purpose of this study was to evaluate whether seeding uncultured ADRCs onto CBM would improve matrix properties and enhance healing of the grafted wound. METHODS Full thickness thermal burns were created on the backs of 8 Gottingen mini-swine. Two days post-injury wounds underwent fascial excision and animals were randomized to receive either Integra seeded with either uncultured ADRCs or control vehicle. Wound healing assessment was performed by digital wound imaging, histopathological and immunohistochemical analyses. RESULTS In vitro analysis demonstrated that freshly isolated ADRCs adhered and propagated on the CBM. Histological scoring revealed accelerated maturation of wound bed tissue in wounds receiving ADRCs-loaded CBM compared to vehicle-loaded CBM. This was associated with a significant increase in depth of the wound bed tissue and collagen deposition (p<0.05). Blood vessel density in the wound bed was 50% to 69.6% greater in wounds receiving ADRCs-loaded CBM compared to vehicle-loaded CBM (p=0.05) at day 14 and 21. In addition, ADRCs delivered with CBM showed increased blood vessel lumen area and blood vessel maturation at day 21(p=0.05). Interestingly, vascularity and overall cellularity within the CBM were 50% and 45% greater in animals receiving ADRC loaded scaffolds compared to CBM alone (p<0.05). CONCLUSIONS These data demonstrate that seeding uncultured ADRCs onto CBM dermal substitute enhances wound angiogenesis, blood vessel maturation and matrix remodeling.
Collapse
Affiliation(s)
| | | | | | | | | | - Isaac Hakim
- Comparative Biosciences Inc., Sunnyvale, CA, USA
| | | | - Mayer Tenenhaus
- UCSD Medical Center, University of California, San Diego, CA, USA
| | | |
Collapse
|