1
|
Ebadi M, Jonart LM, Ostergaard J, Gordon PM. CD99 antibody disrupts T-cell acute lymphoblastic leukemia adhesion to meningeal cells and attenuates chemoresistance. Sci Rep 2021; 11:24374. [PMID: 34934147 PMCID: PMC8692434 DOI: 10.1038/s41598-021-03929-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 12/06/2021] [Indexed: 12/02/2022] Open
Abstract
Central nervous system (CNS) relapse is a significant cause of treatment failure among patients with acute lymphoblastic leukemia. In prior work we found that the meninges, the thin layer of tissue that covers the brain and spinal cord, harbor leukemia cells in the CNS. Importantly, direct interactions between leukemia and meningeal cells enabled leukemia chemoresistance. Herein, we show that an antibody targeting CD99, a transmembrane protein expressed on meningeal cells and many leukemia cells, disrupts adhesion between leukemia and meningeal cells and restores sensitivity of the leukemia cells to chemotherapy. This work identifies a mechanism regulating critical intercellular interactions within the CNS leukemia niche and may lead to novel therapeutic approaches for overcoming niche-mediated chemoresistance.
Collapse
Affiliation(s)
- Maryam Ebadi
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Minnesota, 420 Delaware St SE, MMC 366, Minneapolis, MN, 55455, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Leslie M Jonart
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Minnesota, 420 Delaware St SE, MMC 366, Minneapolis, MN, 55455, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Jason Ostergaard
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Minnesota, 420 Delaware St SE, MMC 366, Minneapolis, MN, 55455, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Peter M Gordon
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Minnesota, 420 Delaware St SE, MMC 366, Minneapolis, MN, 55455, USA. .,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
2
|
Song F, Wang S, Pang X, Fan Z, Zhang J, Chen X, He L, Ma B, Pei X, Li Y. An Active Fraction of Trillium tschonoskii Promotes the Regeneration of Intestinal Epithelial Cells After Irradiation. Front Cell Dev Biol 2021; 9:745412. [PMID: 34796175 PMCID: PMC8593212 DOI: 10.3389/fcell.2021.745412] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 10/11/2021] [Indexed: 12/29/2022] Open
Abstract
Despite significant scientific advances toward the development of safe and effective radiation countermeasures, no drug has been approved for use in the clinic for prevention or treatment of radiation-induced acute gastrointestinal syndrome (AGS). Thus, there is an urgent need to develop potential drugs to accelerate the repair of injured intestinal tissue. In this study, we investigated that whether some fractions of Traditional Chinese Medicine (TCM) have the ability to regulate intestinal crypt cell proliferation and promotes crypt regeneration after radiation. By screening the different supplements from a TCM library, we found that an active fraction of the rhizomes of Trillium tschonoskii Maxim (TT), TT-2, strongly increased the colony-forming ability of irradiated rat intestinal epithelial cell line 6 (IEC-6) cells. TT-2 significantly promoted the proliferation and inhibited the apoptosis of irradiated IEC-6 cells. Furthermore, in a small intestinal organoid radiation model, TT-2 promoted irradiated intestinal organoid growth and increased Lgr5+ intestinal stem cell (ICS) numbers. More importantly, the oral administration of TT-2 remarkably enhanced intestinal crypt cell proliferation and promoted the repair of the intestinal epithelium of mice after abdominal irradiation (ABI). Mechanistically, TT-2 remarkably activated the expression of ICS-associated and proliferation-promoting genes and inhibited apoptosis-related gene expression. Our data indicate that active fraction of TT can be developed into a potential oral drug for improving the regeneration and repair of intestinal epithelia that have intestinal radiation damage.
Collapse
Affiliation(s)
- Feiling Song
- Experimental Hematology and Biochemistry Lab, Beijing Institute of Radiation Medicine, Beijing, China
| | - Sihan Wang
- Stem Cells and Regenerative Medicine Lab, Institute of Health Service and Transfusion Medicine, Beijing, China.,South China Research Center for Stem Cell & Regenerative Medicine, SCIB, Guangzhou, China
| | - Xu Pang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Zeng Fan
- Stem Cells and Regenerative Medicine Lab, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Jie Zhang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Xiaojuan Chen
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Lijuan He
- Stem Cells and Regenerative Medicine Lab, Institute of Health Service and Transfusion Medicine, Beijing, China.,South China Research Center for Stem Cell & Regenerative Medicine, SCIB, Guangzhou, China
| | - Baiping Ma
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Xuetao Pei
- Stem Cells and Regenerative Medicine Lab, Institute of Health Service and Transfusion Medicine, Beijing, China.,South China Research Center for Stem Cell & Regenerative Medicine, SCIB, Guangzhou, China
| | - Yanhua Li
- Experimental Hematology and Biochemistry Lab, Beijing Institute of Radiation Medicine, Beijing, China.,South China Research Center for Stem Cell & Regenerative Medicine, SCIB, Guangzhou, China
| |
Collapse
|
3
|
Wang S, Han Y, Zhang J, Yang S, Fan Z, Song F, He L, Yue W, Li Y, Pei X. Me6TREN targets β-catenin signaling to stimulate intestinal stem cell regeneration after radiation. Theranostics 2020; 10:10171-10185. [PMID: 32929341 PMCID: PMC7481405 DOI: 10.7150/thno.46415] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/30/2020] [Indexed: 01/08/2023] Open
Abstract
Background: Acute gastrointestinal syndrome (AGS) is one of the most severe clinical manifestations after exposure to high doses of radiation, and is life-threatening in radiological emergency scenarios. However, an unmet challenge is lacking of an FDA-approved drug that can ameliorate the damage of radiation-exposed intestinal tissues and accelerate the regeneration of injured epithelia. In this study, we investigated whether the small molecule Me6TREN (Me6) can regulate intestinal stem cell (ISC) proliferation and promote crypt regeneration after irradiation. Methods: Lethally irradiated mice were administered with Me6 or PBS to study the survival rate, and sections of their small intestine were subjected to immunostaining to evaluate epithelial regeneration. An intestinal organoid culture system was employed to detect the role of Me6 in organoid growth and ISC proliferation. We further investigated the key signaling pathways associated with Me6 using microarray, western blotting, and RNA interference techniques. Results: We identified the small molecule Me6 as a potent intestinal radiation countermeasure. Systemic administration of Me6 significantly improved ISC and crypt cell regeneration and enhanced the survival of mice after high doses of radiation. Using an in vitro intestinal organoid culture system, we found that Me6 not only induced ISC proliferation but also increased the budding rate of intestinal organoids under unirradiated and irradiated conditions. Me6 remarkably activated the expression of ISC-associated and proliferation-promoting genes, such as Ascl2, Lgr5, Myc, and CyclinD1. Mechanistically, Me6 strongly stimulated the phosphorylation of β-catenin at the S552 site and increased the transcriptional activity of β-catenin, a key signaling pathway for ISC self-renewal and proliferation. This is further evidenced by the fact that knockdown of β-catenin abolished the effect of Me6 on intestinal organoid growth in vitro and crypt regeneration in irradiated mice. Conclusion: The small molecule Me6TREN induced ISC proliferation, enhanced intestinal organoid growth in vitro, and promoted intestinal tissue regeneration after radiation injury by activating β-catenin signaling.
Collapse
|
4
|
Beltran-Camacho L, Jimenez-Palomares M, Rojas-Torres M, Sanchez-Gomar I, Rosal-Vela A, Eslava-Alcon S, Perez-Segura MC, Serrano A, Antequera-González B, Alonso-Piñero JA, González-Rovira A, Extremera-García MJ, Rodriguez-Piñero M, Moreno-Luna R, Larsen MR, Durán-Ruiz MC. Identification of the initial molecular changes in response to circulating angiogenic cells-mediated therapy in critical limb ischemia. Stem Cell Res Ther 2020; 11:106. [PMID: 32143690 PMCID: PMC7060566 DOI: 10.1186/s13287-020-01591-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 01/10/2020] [Accepted: 02/06/2020] [Indexed: 12/18/2022] Open
Abstract
Background Critical limb ischemia (CLI) constitutes the most aggressive form of peripheral arterial occlusive disease, characterized by the blockade of arteries supplying blood to the lower extremities, significantly diminishing oxygen and nutrient supply. CLI patients usually undergo amputation of fingers, feet, or extremities, with a high risk of mortality due to associated comorbidities. Circulating angiogenic cells (CACs), also known as early endothelial progenitor cells, constitute promising candidates for cell therapy in CLI due to their assigned vascular regenerative properties. Preclinical and clinical assays with CACs have shown promising results. A better understanding of how these cells participate in vascular regeneration would significantly help to potentiate their role in revascularization. Herein, we analyzed the initial molecular mechanisms triggered by human CACs after being administered to a murine model of CLI, in order to understand how these cells promote angiogenesis within the ischemic tissues. Methods Balb-c nude mice (n:24) were distributed in four different groups: healthy controls (C, n:4), shams (SH, n:4), and ischemic mice (after femoral ligation) that received either 50 μl physiological serum (SC, n:8) or 5 × 105 human CACs (SE, n:8). Ischemic mice were sacrificed on days 2 and 4 (n:4/group/day), and immunohistochemistry assays and qPCR amplification of Alu-human-specific sequences were carried out for cell detection and vascular density measurements. Additionally, a label-free MS-based quantitative approach was performed to identify protein changes related. Results Administration of CACs induced in the ischemic tissues an increase in the number of blood vessels as well as the diameter size compared to ischemic, non-treated mice, although the number of CACs decreased within time. The initial protein changes taking place in response to ischemia and more importantly, right after administration of CACs to CLI mice, are shown. Conclusions Our results indicate that CACs migrate to the injured area; moreover, they trigger protein changes correlated with cell migration, cell death, angiogenesis, and arteriogenesis in the host. These changes indicate that CACs promote from the beginning an increase in the number of vessels as well as the development of an appropriate vascular network. Graphical abstract ![]()
Collapse
Affiliation(s)
- Lucia Beltran-Camacho
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain.,Institute of Biomedical Research Cadiz (INIBICA), Cadiz, Spain
| | - Margarita Jimenez-Palomares
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain.,Institute of Biomedical Research Cadiz (INIBICA), Cadiz, Spain
| | - Marta Rojas-Torres
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain.,Institute of Biomedical Research Cadiz (INIBICA), Cadiz, Spain
| | - Ismael Sanchez-Gomar
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain.,Institute of Biomedical Research Cadiz (INIBICA), Cadiz, Spain
| | - Antonio Rosal-Vela
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain.,Institute of Biomedical Research Cadiz (INIBICA), Cadiz, Spain
| | - Sara Eslava-Alcon
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain.,Institute of Biomedical Research Cadiz (INIBICA), Cadiz, Spain
| | | | - Ana Serrano
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain
| | - Borja Antequera-González
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain.,Institute of Biomedical Research Cadiz (INIBICA), Cadiz, Spain
| | - Jose Angel Alonso-Piñero
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain.,Institute of Biomedical Research Cadiz (INIBICA), Cadiz, Spain
| | - Almudena González-Rovira
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain.,Institute of Biomedical Research Cadiz (INIBICA), Cadiz, Spain
| | - Mª Jesús Extremera-García
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain.,Institute of Biomedical Research Cadiz (INIBICA), Cadiz, Spain
| | | | - Rafael Moreno-Luna
- Laboratory of Neuroinflammation, Hospital Nacional de Paraplejicos, SESCAM, Toledo, Spain
| | - Martin Røssel Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Mª Carmen Durán-Ruiz
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain. .,Institute of Biomedical Research Cadiz (INIBICA), Cadiz, Spain.
| |
Collapse
|
5
|
Yu G, Li S, Liu P, Shi Y, Liu Y, Yang Z, Fan Z, Zhu W. LncRNA TUG1 functions as a ceRNA for miR-6321 to promote endothelial progenitor cell migration and differentiation. Exp Cell Res 2020; 388:111839. [PMID: 31935381 DOI: 10.1016/j.yexcr.2020.111839] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 12/18/2022]
|
6
|
Jonart LM, Ebadi M, Basile P, Johnson K, Makori J, Gordon PM. Disrupting the leukemia niche in the central nervous system attenuates leukemia chemoresistance. Haematologica 2019; 105:2130-2140. [PMID: 31624109 PMCID: PMC7395284 DOI: 10.3324/haematol.2019.230334] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 10/14/2019] [Indexed: 12/21/2022] Open
Abstract
Protection from acute lymphoblastic leukemia relapse in the central nervous system (CNS) is crucial to survival and quality of life for leukemia patients. Current CNS-directed therapies cause significant toxicities and are only partially effective. Moreover, the impact of the CNS microenvironment on leukemia biology is poorly understood. In this study we showed that leukemia cells associated with the meninges of xenotransplanted mice, or co-cultured with meningeal cells, exhibit enhanced chemoresistance due to effects on both apoptosis balance and quiescence. From a mechanistic standpoint, we found that leukemia chemoresistance is primarily mediated by direct leukemia-meningeal cell interactions and overcome by detaching the leukemia cells from the meninges. Next, we used a co-culture adhesion assay to identify drugs that disrupted leukemia-meningeal adhesion. In addition to identifying several drugs that inhibit canonical cell adhesion targets we found that Me6TREN (Tris[2-(dimethylamino)ethyl]amine), a novel hematopoietic stem cell-mobilizing compound, also disrupted leukemia-meningeal adhesion and enhanced the efficacy of cytarabine in treating CNS leukemia in xenotransplanted mice. This work demonstrates that the meninges exert a critical influence on leukemia chemoresistance, elucidates mechanisms of relapse beyond the well-described role of the blood-brain barrier, and identifies novel therapeutic approaches for overcoming chemoresistance.
Collapse
Affiliation(s)
- Leslie M Jonart
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Minnesota.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Maryam Ebadi
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Minnesota.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Patrick Basile
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Minnesota.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Kimberly Johnson
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Minnesota.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Jessica Makori
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Minnesota.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Peter M Gordon
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Minnesota .,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
7
|
Li Y, Li X, Han S, Lian W, Cheng J, Xie X, Li M. Exogenous FGF-2 improves biological activity of endothelial progenitor cells exposed to high glucose conditions. J Interv Med 2019; 1:9-14. [PMID: 34805825 PMCID: PMC8586578 DOI: 10.19779/j.cnki.2096-3602.2018.01.04] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Purpose: To investigate the effects of exogenous basic fibroblast growth factor -2 (FGF-2) on the biological activity of endothelial progenitor cells (EPCs) exposed to high glucose conditions. Materials and Methods: 1) Bone marrow EPCs from C57BL/6 mice were isolated and cultured in vitro. EPC purity was identified by flow cytometry and immunofluorescence staining. 2) Apoptosis was detected by TUNEL assay. Migration and tube formation ability was detected by Transwell chamber and Matrigel assays, respectively. The expression and activation of β-catenin was detected by Western blot. 3) Doppler flowmetry was used to detect the effect of FGF2 on blood flow recovery in ischemic hind limbs of mice. Results: 1) FGF-2 treatment reversed high glucose induced growth inhibition of EPCs. FGF-2 treatment also increased migration and tube formation ability of EPCs even in high glucose conditions. 2) Western blot analysis demonstrated that the percentage of activated β-catenin/total β-catenin in the high glucose group were significantly lower than that in the control group, while FGF-2 treatment reversed high glucose induced β-catenin inhibition. 3) In vivo experiments demonstrated that the blood flow recovery in ischemic hind limbs of mice was significantly improved after FGF-2 treatment. Conclusion: Exogenous FGF-2 could play a role in the functional repair of damaged EPC exposed to high glucose conditions, via the activation of the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Yang Li
- Department of Interventional and Vascular surgery, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China.,Institute of Intervention Radiology and Vascular Surgery, Tongji University, Shanghai 200072, China
| | - Xue Li
- Department of Interventional and Vascular surgery, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China.,Institute of Intervention Radiology and Vascular Surgery, Tongji University, Shanghai 200072, China
| | - Shilong Han
- Department of Interventional and Vascular surgery, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China.,Institute of Intervention Radiology and Vascular Surgery, Tongji University, Shanghai 200072, China
| | - Weishuai Lian
- Department of Interventional and Vascular surgery, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China.,Institute of Intervention Radiology and Vascular Surgery, Tongji University, Shanghai 200072, China
| | - Jie Cheng
- Department of Interventional and Vascular surgery, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China.,Institute of Intervention Radiology and Vascular Surgery, Tongji University, Shanghai 200072, China
| | - Xiaoyun Xie
- Department of Interventional and Vascular surgery, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China.,Institute of Intervention Radiology and Vascular Surgery, Tongji University, Shanghai 200072, China
| | - Maoquan Li
- Department of Interventional and Vascular surgery, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China.,Institute of Intervention Radiology and Vascular Surgery, Tongji University, Shanghai 200072, China
| |
Collapse
|
8
|
Kwon YW, Lee SJ, Heo SC, Lee TW, Park GT, Yoon JW, Kim SC, Shin HJ, Lee SC, Kim JH. Role of CXCR2 in the Ac-PGP-Induced Mobilization of Circulating Angiogenic Cells and its Therapeutic Implications. Stem Cells Transl Med 2018; 8:236-246. [PMID: 30474937 PMCID: PMC6392381 DOI: 10.1002/sctm.18-0035] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 09/14/2018] [Indexed: 02/06/2023] Open
Abstract
Circulating angiogenic cells (CACs) have been implicated in the repair of ischemic tissues, and their mobilization from bone marrow is known to be regulated by the activations of chemokine receptors, including CXCR2 and CXCR4. This study was conducted to investigate the role of N‐acetylated proline‐glycine‐proline (Ac‐PGP; a collagen‐derived chemotactic tripeptide) on CAC mobilization and its therapeutic potential for the treatment of peripheral artery diseases. Ac‐PGP was administered daily to a murine hind limb ischemia model, and the effects of Ac‐PGP on blood perfusion and CAC mobilization (Sca1+Flk1+ cells) into peripheral blood were assessed. Intramuscular administration of Ac‐PGP significantly improved ischemic limb perfusion and increased limb salvage rate by increasing blood vessel formation, whereas Ac‐PGP‐induced blood perfusion and angiogenesis in ischemic limbs were not observed in CXCR2‐knockout mice. In addition, Ac‐PGP‐induced CAC mobilization was found to occur in wild‐type mice but not in CXCR2‐knockout mice. Transplantation of bone marrow from green fluorescent protein (GFP) transgenic mice to wild‐type mice showed bone marrow‐derived cells homed to ischemic limbs after Ac‐PGP administration and that GFP‐positive cells contributed to the formation of ILB4‐positive capillaries and α smooth muscle actin (α‐SMA)‐positive arteries. These results suggest CXCR2 activation in bone marrow after Ac‐PGP administration improves blood perfusion and reduces tissue necrosis by inducing CAC mobilization. These findings suggest a new pharmaceutical basis for the treatment of critical limb ischemia. stem cells translational medicine2019;8:236&246
Collapse
Affiliation(s)
- Yang Woo Kwon
- Department of Physiology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Seung Jun Lee
- Department of Orthopaedic Surgery, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Soon Chul Heo
- Department of Physiology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Tae Wook Lee
- Department of Physiology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Gyu Tae Park
- Department of Physiology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Jung Won Yoon
- Department of Physiology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Seung-Chul Kim
- Department of Obstetrics and Gynecology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Ho Jin Shin
- Division of Hematology-Oncology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Sang Chul Lee
- Functional Genomics Research Center, KRIBB, Daejeon, Republic of Korea
| | - Jae Ho Kim
- Department of Physiology, Pusan National University School of Medicine, Yangsan, Republic of Korea.,Research Institute of Convergence Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| |
Collapse
|
9
|
Ebrahimi B. In vivo reprogramming for heart regeneration: A glance at efficiency, environmental impacts, challenges and future directions. J Mol Cell Cardiol 2017; 108:61-72. [PMID: 28502796 DOI: 10.1016/j.yjmcc.2017.05.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 05/08/2017] [Indexed: 02/08/2023]
Abstract
Replacing dying or diseased cells of a tissue with new ones that are converted from patient's own cells is an attractive strategy in regenerative medicine. In vivo reprogramming is a novel strategy that can circumvent the hurdles of autologous/allogeneic cell injection therapies. Interestingly, studies have demonstrated that direct injection of cardiac transcription factors or specific miRNAs into the infarct border zone of murine hearts following myocardial infarction converts resident cardiac fibroblasts into functional cardiomyocytes. Moreover, in vivo cardiac reprogramming not only drives cardiac tissue regeneration, but also improves cardiac function and survival rate after myocardial infarction. Thanks to the influence of cardiac microenvironment and the same developmental origin, cardiac fibroblasts seem to be more amenable to reprogramming toward cardiomyocyte fate than other cell sources (e.g. skin fibroblasts). Thus, reprogramming of cardiac fibroblasts to functional induced cardiomyocytes in the cardiac environment holds great promises for induced regeneration and potential clinical purposes. Application of small molecules in future studies may represent a major advancement in this arena and pharmacological reprogramming would convey reprogramming technology to the translational medicine paradigm. This study reviews accomplishments in the field of in vitro and in vivo mouse cardiac reprogramming and then deals with strategies for the enhancement of the efficiency and quality of the process. Furthermore, it discusses challenges ahead and provides suggestions for future research. Human cardiac reprogramming is also addressed as a foundation for possible application of in vivo cardiac reprogramming for human heart regeneration in the future.
Collapse
Affiliation(s)
- Behnam Ebrahimi
- Yazd Cardiovascular Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
10
|
He S, Zhao T, Guo H, Meng Y, Qin G, Goukassian DA, Han J, Gao X, Zhu Y. Coordinated Activation of VEGF/VEGFR-2 and PPARδ Pathways by a Multi-Component Chinese Medicine DHI Accelerated Recovery from Peripheral Arterial Disease in Type 2 Diabetic Mice. PLoS One 2016; 11:e0167305. [PMID: 27930695 PMCID: PMC5145164 DOI: 10.1371/journal.pone.0167305] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 11/12/2016] [Indexed: 12/26/2022] Open
Abstract
Diabetic mellitus (DM) patients are at an increased risk of developing peripheral arterial disease (PAD). Danhong injection (DHI) is a Chinese patent medicine widely used for several cardiovascular indications but the mechanism of action is not well-understood. We investigated the therapeutic potential of DHI on experimental PAD in mice with chemically induced as well as genetic (KKAy) type 2 DM and the overlapping signaling pathways regulating both therapeutic angiogenesis and glucose homeostasis. Compared with normal genetic background wild type (WT) mice, both DM mice showed impaired perfusion recovery in hind-limb ischemia (HLI) model. DHI treatment significantly accelerated perfusion recovery, lowered blood glucose and improved glucose tolerance in both DM models. Bioluminescent imaging demonstrated a continuous ischemia-induced vascular endothelial growth factor receptor 2 (VEGFR-2) gene expressions with a peak time coincident with the maximal DHI stimulation. Flow cytometry analysis showed a DHI-mediated increase in endothelial progenitor cell (EPC) mobilization from bone marrow to circulating peripheral blood. DHI administration upregulated the expression of vascular endothelial growth factor A (VEGF-A) and VEGF receptor-2 (VEGFR-2) in ischemic muscle. A cross talk between ischemia-induced angiogenesis and glucose tolerance pathways was analyzed by Ingenuity Pathway Analysis (IPA) which suggested an interaction of VEGF-A/VEGFR-2 and peroxisome proliferator-activated receptor δ (PPARδ)/peroxisome proliferator-activated receptor γ (PPARγ) genes. We confirmed that upregulation of VEGF-A/VEGFR-2 by DHI promoted PPARδ gene expression in both type 2 diabetic mice. Our findings demonstrated that a multi-component Chinese medicine DHI effectively increased blood flow recovery after tissue ischemia in diabetic mice by promoting angiogenesis and improving glucose tolerance through a concomitant activation of VEGF-A/VEGFR-2 and PPARδ signaling pathways.
Collapse
Affiliation(s)
- Shuang He
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, Tianjin, China
| | - Tiechan Zhao
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, Tianjin, China
| | - Hao Guo
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, Tianjin, China
| | - Yanzhi Meng
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, Tianjin, China
| | - Gangjian Qin
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Department of Medicine-Cardiology and Department of Molecular Pharmacology and Biological Chemistry, Northwestern University Feinberg School of Medicine, Chicago, United States of America
| | - David A. Goukassian
- Center of Biomedical Research, Tufts University School of Medicine, Boston, United States of America
| | - Jihong Han
- State Key Laboratory of Medicinal Chemical Biology, and Collaborative Innovation Center for Biotherapy, Nankai University, Tianjin, China
| | - Xuimei Gao
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yan Zhu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, Tianjin, China
- Molecular Cardiology Research Institute, Tufts Medical Center and Tufts University School of Medicine, Boston, United States of America
- * E-mail:
| |
Collapse
|
11
|
Li Y, Kinzenbaw DA, Modrick ML, Pewe LL, Faraci FM. Context-dependent effects of SOCS3 in angiotensin II-induced vascular dysfunction and hypertension in mice: mechanisms and role of bone marrow-derived cells. Am J Physiol Heart Circ Physiol 2016; 311:H146-56. [PMID: 27106041 DOI: 10.1152/ajpheart.00204.2016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 04/18/2016] [Indexed: 11/22/2022]
Abstract
Carotid artery disease is a major contributor to stroke and cognitive deficits. Angiotensin II (Ang II) promotes vascular dysfunction and disease through mechanisms that include the IL-6/STAT3 pathway. Here, we investigated the importance of suppressor of cytokine signaling 3 (SOCS3) in models of Ang II-induced vascular dysfunction. We examined direct effects of Ang II on carotid arteries from SOCS3-deficient (SOCS3(+/-)) mice and wild-type (WT) littermates using organ culture and then tested endothelial function with acetylcholine (ACh). A low concentration of Ang II (1 nmol/l) did not affect ACh-induced vasodilation in WT but reduced that of SOCS3(+/-) mice by ∼50% (P < 0.05). In relation to mechanisms, effects of Ang II in SOCS3(+/-) mice were prevented by inhibitors of STAT3, IL-6, NF-κB, or superoxide. Systemic Ang II (1.4 mg/kg per day for 14 days) also reduced vasodilation to ACh in WT. Surprisingly, SOCS3 deficiency prevented most of the endothelial dysfunction. To examine potential underlying mechanisms, we performed bone marrow transplantation. WT mice reconstituted with SOCS3(+/-) bone marrow were protected from Ang II-induced endothelial dysfunction, whereas reconstitution of SOCS3(+/-) mice with WT bone marrow exacerbated Ang II-induced effects. The SOCS3 genotype of bone marrow-derived cells did not influence direct effects of Ang II on vascular function. These data provide new mechanistic insight into the influence of SOCS3 on the vasculature, including divergent effects depending on the source of Ang II. Bone marrow-derived cells deficient in SOCS3 protect against systemic Ang II-induced vascular dysfunction.
Collapse
Affiliation(s)
- Ying Li
- Department of Pharmacology, Francois M. Abboud Cardiovascular Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Dale A Kinzenbaw
- Department of Internal Medicine, Francois M. Abboud Cardiovascular Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Mary L Modrick
- Department of Internal Medicine, Francois M. Abboud Cardiovascular Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Lecia L Pewe
- Department of Microbiology, Francois M. Abboud Cardiovascular Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Frank M Faraci
- Department of Pharmacology, Francois M. Abboud Cardiovascular Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa; Department of Internal Medicine, Francois M. Abboud Cardiovascular Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa; Iowa City Veterans Affairs Healthcare System, Iowa City, Iowa
| |
Collapse
|
12
|
Zhao H, Huang H, Lin S. Chemical approaches to angiogenesis in development and regeneration. Methods Cell Biol 2016; 134:369-76. [PMID: 27312498 DOI: 10.1016/bs.mcb.2016.03.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Vascular endothelial cells are essential building blocks of angiogenesis, which is required for normal embryonic development and tissue regeneration. In this chapter, we describe how to use transgenic zebrafish embryos expressing vascular-specific green fluorescent protein to evaluate differentiation, growth, and morphogenesis of endothelial cells. When combined with instrument automation and computational analysis, this method allows high-throughput screening for biologically active small chemical molecules that are effective in promoting angiogenesis. These molecules can be validated in mammalian endothelial cell differentiation and proliferation assays. These studies provide new reagents and therapeutic candidates for regenerative medicine studies.
Collapse
Affiliation(s)
- H Zhao
- University of California Los Angeles, Los Angeles, CA, United States; Jinan University, Guangzhou, China
| | - H Huang
- University of California Los Angeles, Los Angeles, CA, United States
| | - S Lin
- University of California Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
13
|
Liu Y, Xu Y, Fang F, Zhang J, Guo L, Weng Z. Therapeutic Efficacy of Stem Cell-based Therapy in Peripheral Arterial Disease: A Meta-Analysis. PLoS One 2015; 10:e0125032. [PMID: 25923119 PMCID: PMC4414514 DOI: 10.1371/journal.pone.0125032] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 03/19/2015] [Indexed: 11/18/2022] Open
Abstract
Several cell-based therapies for peripheral arterial disease (PAD) have been studied in multiple clinical trials; however, the outcome of this treatment remains controversial. The aim of this study was to perform a meta-analysis of the clinical trials on stem cell-based therapy after PAD. We searched for clinical trials that investigated the effect of stem cell-based therapy on patients with PAD and were published between January 2000 and October 2014. The outcomes of interest comprised all-cause mortality, amputation rate, ulcer healing, and ankle-brachial index (ABI). In addition, pooled odds ratios (ORs) and 95% confidence intervals (CIs) were calculated to assess the safety and efficacy of the stem cell-based therapies for PAD. Thirteen studies were retrieved from 261 citations for the analysis, and in total, 527 patients (mean age: 64.2 years; median follow up: 6 months) were included in the analysis. After synthesizing data, the meta-analysis showed significant improvement in the amputation rate (OR=0.33, 95%CI=0.22-0.51; P<0.001), ulcer healing (OR=6.11, 95%CI=3.04-12.28; P<0.001), and ABI (SMD=0.65, 95%CI=0.33-0.97; P<0.001) for the stem cell-based therapy group compared with the controls. Moreover, significant improvement in the amputation rate, ulcer healing, and ABI were also found based on the time point and stem cell source. In addition, no significant difference was found in the all-cause mortality (OR=0.80, 95%CI=0.39-1.641; P=0.546) between the stem cell-based therapy and control groups. Therefore, according to the results of our meta-analysis, stem cell-based therapy is safe and shows a beneficial outcome for patients with PAD, especially in the short term.
Collapse
Affiliation(s)
- Yumeng Liu
- Department of Radiology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yunyun Xu
- Institute of Pediatrics, Children’s Hospital Affiliated to Soochow University, Suzhou, Jiangsu, China
| | - Fang Fang
- Institute of Pediatrics, Children’s Hospital Affiliated to Soochow University, Suzhou, Jiangsu, China
| | - Jianting Zhang
- Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, Jiangsu, China
| | - Liang Guo
- Department of Radiology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- * E-mail: (ZW); (LG)
| | - Zhen Weng
- Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, Jiangsu, China
- * E-mail: (ZW); (LG)
| |
Collapse
|