1
|
Yin J, Gou H, Qi J, Xing W. Feature gene selection and functional validation of SH3KBP1 in infantile hemangioma using machine learning. Biochem Biophys Res Commun 2025; 752:151469. [PMID: 39955954 DOI: 10.1016/j.bbrc.2025.151469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 01/07/2025] [Accepted: 02/08/2025] [Indexed: 02/18/2025]
Abstract
BACKGROUND Infantile hemangioma (IH) is a prevalent vascular tumor in infancy with a complex pathogenesis that remains unclear. This study aimed to investigate the underlying mechanisms of IH using comprehensive bioinformatics analyses and in vitro experiments. METHODS Using GSE127487, we identified differentially expressed genes (DEGs) in IH patients across three age groups (6, 12, and 24 months). GO and KEGG enrichment analyses were performed to identify biological processes and pathways. Immune cell infiltration, transcription factor target genes, miRNA expression, and metabolic pathways were analyzed. WGCNA classified IH patients into clusters, and machine learning algorithms identified key genes. The role of SH3KBP1, the most abundantly expressed gene in the skin, was investigated using shRNA knockdown and functional assays. RESULTS Gene expression in IH patients exhibited dynamic changes with age. Cellular processes and signaling pathways were consistent in the early proliferative phase, with gradual resolution in the late phase. Immune infiltration analysis revealed reduced immune cells in patients, while Pericytes were increased. NR5A1 was downregulated, while ZNF112, HSF4, and multiple miRNAs were upregulated with age. Metabolic pathways confirmed differences between proliferative and involution phases. WGCNA identified two clusters: Cluster 1 (angiogenesis and signal transduction) and Cluster 2 (metabolic and synthetic processes). Key genes, including SH3KBP1, were identified using machine learning algorithms. In vitro experiments demonstrated SH3KBP1's crucial role in cell migration and invasion. CONCLUSION This study unravels the gene expression and regulatory mechanisms of IH at different stages, providing new insights into its pathophysiology. SH3KBP1 offers a potential biomarker for future diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Jiu Yin
- Suining Central Hospital, Suning, 629000, Sichuan, China.
| | - Hui Gou
- Suining First People's Hospital, Suning, 629000, Sichuan, China.
| | - Jian Qi
- Nanchong Fifth People's Hospital, Nanchong, 637199, Sichuan, China.
| | - Wenli Xing
- Suining Central Hospital, Suning, 629000, Sichuan, China.
| |
Collapse
|
2
|
Indoxyl sulfate- and P-cresol-induced monocyte adhesion and migration is mediated by integrin-linked kinase-dependent podosome formation. Exp Mol Med 2022; 54:226-238. [PMID: 35246616 PMCID: PMC8980039 DOI: 10.1038/s12276-022-00738-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 10/22/2021] [Accepted: 11/30/2021] [Indexed: 12/21/2022] Open
Abstract
Cardiovascular disease is an important cause of death in patients with chronic kidney disease (CKD). Protein-bound uremic toxins, such as p-cresyl and indoxyl sulfate (IS), are poorly removed during hemodialysis, leading to vascular endothelial dysfunction and leukocyte extravasation. These processes can be related to dynamic adhesion structures called podosomes. Several studies have indicated the role of integrin-linked kinase (ILK) in the accumulation of integrin-associated proteins in podosomes. Here, we investigated the involvement of ILK and podosome formation in the adhesion and extravasation of monocytes under p-cresol (pc) and IS exposure. Incubation of THP-1 human monocyte cells with these toxins upregulated ILK kinase activity. Together, both toxins increased cell adhesion, podosome formation, extracellular matrix degradation, and migration of THP-1 cells, whereas ILK depletion with specific small interfering RNAs suppressed these processes. Interestingly, F-actin colocalized with cortactin in podosome cores, while ILK was colocalized in podosome rings under toxin stimulation. Podosome Wiskott-Aldrich syndrome protein (WASP)-interacting protein (WIP) and AKT protein depletion demonstrated that monocyte adhesion depends on podosome formation and that the ILK/AKT signaling pathway is involved in these processes. Ex vivo experiments showed that both toxins induced adhesion and podosome formation in leukocytes from wild-type mice, whereas these effects were not observed in leukocytes of conditional ILK-knockdown animals. In summary, under pc and IS stimulation, monocytes increase podosome formation and transmigratory capacity through an ILK/AKT signaling pathway-dependent mechanism, which could lead to vascular injury. Therefore, ILK could be a potential therapeutic target for the treatment of vascular damage associated with CKD. An enzyme involved in organizing structural proteins into protrusions of the cell membrane helps facilitate the movement of white blood cells that occurs in chronic kidney disease and can lead to cardiovascular damage. Laura Calleros from the University of Alcalá, Madrid, Spain, and colleagues showed how toxic metabolites that build up in the bloodstream as kidneys fail induce the activity of an enzyme called integrin-linked kinase (ILK). This enzyme in turn spurs white blood cells to form protrusions called podosomes that make the cells more mobile, enabling them to move out of blood vessels and into surrounding tissues where they can cause inflammation -mediated injury. ILK inactivation reversed these effects. Therapies that target the enzyme could therefore help limit the cardiovascular complications of chronic kidney disease.
Collapse
|
3
|
Blachier F, Andriamihaja M. Effects of the L-tyrosine-derived bacterial metabolite p-cresol on colonic and peripheral cells. Amino Acids 2021; 54:325-338. [PMID: 34468872 DOI: 10.1007/s00726-021-03064-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 08/06/2021] [Indexed: 11/28/2022]
Abstract
Specific families of bacteria present within the intestinal luminal content produce p-cresol from L-tyrosine. Although the hosts do not synthesize p-cresol, they can metabolize this compound within their colonic mucosa and liver leading to the production of co-metabolites including p-cresyl sulfate (p-CS) and p-cresyl glucuronide (p-CG). p-Cresol and its co-metabolites are recovered in the circulation mainly conjugated to albumin, but also in their free forms that are excreted in the urine. An increased dietary protein intake raises the amount of p-cresol recovered in the feces and urine, while fecal excretion of p-cresol is diminished by a diet containing undigestible polysaccharides. p-Cresol in excess is genotoxic for colonocytes. In addition, in these cells, this bacterial metabolite decreases mitochondrial oxygen consumption, while increasing the anion superoxide production. In chronic kidney disease (CKD), marked accumulation of p-cresol and p-CS in plasma is measured, and in renal tubular cells, p-cresol and p-CS increase oxidative stress, affect mitochondrial function, and lead to cell death, strongly suggesting that these 2 compounds act as uremic toxins that aggravate CKD progression. p-Cresol and p-CS are also suspected to play a role in the CKD-associated adverse cardiovascular events, since they affect endothelial cell proliferation and migration, decrease the capacity of endothelial wound repair, and increase the senescence of endothelial cells. Finally, the fact that concentration of p-cresol is transiently increased in young autistic children biological fluids, and that intraperitoneal injection of p-cresol in animal models induces some behavioral characteristics observed in the autism spectrum disorders (ASD), raise the view that p-cresol may possibly represent one of the components involved in ASD etiology. Further pre-clinical and clinical studies are obviously needed to determine if the lowering of p-cresol and/or p-CS circulating concentrations, by dietary and/or pharmacological means, would allow, by itself or in combination with other interventions, to improve CKD progression and associated cardiovascular outcomes, as well as some neurological outcomes in children with an early diagnosis of autism.
Collapse
Affiliation(s)
- F Blachier
- Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, Paris, France.
| | - M Andriamihaja
- Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, Paris, France
| |
Collapse
|
4
|
Comprehensive understanding of anchorage-independent survival and its implication in cancer metastasis. Cell Death Dis 2021; 12:629. [PMID: 34145217 PMCID: PMC8213763 DOI: 10.1038/s41419-021-03890-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 02/07/2023]
Abstract
Detachment is the initial and critical step for cancer metastasis. Only the cells that survive from detachment can develop metastases. Following the disruption of cell-extracellular matrix (ECM) interactions, cells are exposed to a totally different chemical and mechanical environment. During which, cells inevitably suffer from multiple stresses, including loss of growth stimuli from ECM, altered mechanical force, cytoskeletal reorganization, reduced nutrient uptake, and increased reactive oxygen species generation. Here we review the impact of these stresses on the anchorage-independent survival and the underlying molecular signaling pathways. Furthermore, its implications in cancer metastasis and treatment are also discussed.
Collapse
|
5
|
Yue Y, Wang C, Benedict C, Huang G, Truongcao M, Roy R, Cimini M, Garikipati VNS, Cheng Z, Koch WJ, Kishore R. Interleukin-10 Deficiency Alters Endothelial Progenitor Cell-Derived Exosome Reparative Effect on Myocardial Repair via Integrin-Linked Kinase Enrichment. Circ Res 2019; 126:315-329. [PMID: 31815595 DOI: 10.1161/circresaha.119.315829] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Rationale: Systemic inflammation compromises the reparative properties of endothelial progenitor cell (EPC) and their exosomes on myocardial repair, although the underlying mechanism of loss of function of exosomes from inflamed EPCs is still obscure. Objective: To determine the mechanisms of IL-10 (interleukin-10) deficient-EPC-derived exosome dysfunction in myocardial repair and to investigate if modification of specific exosome cargo can rescue reparative activity. Methods and Results: Using IL-10 knockout mice mimicking systemic inflammation condition, we compared therapeutic effect and protein cargo of exosomes isolated from wild-type EPC and IL-10 knockout EPC. In a mouse model of myocardial infarction (MI), wild-type EPC-derived exosome treatment significantly improved left ventricle cardiac function, inhibited cell apoptosis, reduced MI scar size, and promoted post-MI neovascularization, whereas IL-10 knockout EPC-derived exosome treatment showed diminished and opposite effects. Mass spectrometry analysis revealed wild-type EPC-derived exosome and IL-10 knockout EPC-derived exosome contain different protein expression pattern. Among differentially expressed proteins, ILK (integrin-linked kinase) was highly enriched in both IL-10 knockout EPC-derived exosome as well as TNFα (tumor necrosis factor-α)-treated mouse cardiac endothelial cell-derived exosomes (TNFα inflamed mouse cardiac endothelial cell-derived exosome). ILK-enriched exosomes activated NF-κB (nuclear factor κB) pathway and NF-κB-dependent gene transcription in recipient endothelial cells and this effect was partly attenuated through ILK knockdown in exosomes. Intriguingly, ILK knockdown in IL-10 knockout EPC-derived exosome significantly rescued their reparative dysfunction in myocardial repair, improved left ventricle cardiac function, reduced MI scar size, and enhanced post-MI neovascularization in MI mouse model. Conclusions: IL-10 deficiency/inflammation alters EPC-derived exosome function, content and therapeutic effect on myocardial repair by upregulating ILK enrichment in exosomes, and ILK-mediated activation of NF-κB pathway in recipient cells, whereas ILK knockdown in exosomes attenuates NF-κB activation and reduces inflammatory response. Our study provides new understanding of how inflammation may alter stem cell-exosome-mediated cardiac repair and identifies ILK as a target kinase for improving progenitor cell exosome-based cardiac therapies.
Collapse
Affiliation(s)
- Yujia Yue
- From the Center for Translational Medicine (Y.Y., C.W., C.B., G.H., M.T., R.R., M.C. V.N.S.G., Z.C., W.J.K., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Chunlin Wang
- From the Center for Translational Medicine (Y.Y., C.W., C.B., G.H., M.T., R.R., M.C. V.N.S.G., Z.C., W.J.K., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Cindy Benedict
- From the Center for Translational Medicine (Y.Y., C.W., C.B., G.H., M.T., R.R., M.C. V.N.S.G., Z.C., W.J.K., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Grace Huang
- From the Center for Translational Medicine (Y.Y., C.W., C.B., G.H., M.T., R.R., M.C. V.N.S.G., Z.C., W.J.K., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - May Truongcao
- From the Center for Translational Medicine (Y.Y., C.W., C.B., G.H., M.T., R.R., M.C. V.N.S.G., Z.C., W.J.K., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Rajika Roy
- From the Center for Translational Medicine (Y.Y., C.W., C.B., G.H., M.T., R.R., M.C. V.N.S.G., Z.C., W.J.K., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Maria Cimini
- From the Center for Translational Medicine (Y.Y., C.W., C.B., G.H., M.T., R.R., M.C. V.N.S.G., Z.C., W.J.K., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Venkata Naga Srikanth Garikipati
- From the Center for Translational Medicine (Y.Y., C.W., C.B., G.H., M.T., R.R., M.C. V.N.S.G., Z.C., W.J.K., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Zhongjian Cheng
- From the Center for Translational Medicine (Y.Y., C.W., C.B., G.H., M.T., R.R., M.C. V.N.S.G., Z.C., W.J.K., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Walter J Koch
- From the Center for Translational Medicine (Y.Y., C.W., C.B., G.H., M.T., R.R., M.C. V.N.S.G., Z.C., W.J.K., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA.,Department of Pharmacology and Medicine (W.J.K., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Raj Kishore
- From the Center for Translational Medicine (Y.Y., C.W., C.B., G.H., M.T., R.R., M.C. V.N.S.G., Z.C., W.J.K., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA.,Department of Pharmacology and Medicine (W.J.K., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| |
Collapse
|
6
|
Modification of Cardiac Progenitor Cell-Derived Exosomes by miR-322 Provides Protection against Myocardial Infarction through Nox2-Dependent Angiogenesis. Antioxidants (Basel) 2019; 8:antiox8010018. [PMID: 30634641 PMCID: PMC6356993 DOI: 10.3390/antiox8010018] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 01/05/2019] [Indexed: 02/06/2023] Open
Abstract
Myocardial infarction (MI) is the primary cause of cardiovascular mortality, and therapeutic strategies to prevent or mitigate the consequences of MI are a high priority. Cardiac progenitor cells (CPCs) have been used to treat cardiac injury post-MI, and despite poor engraftment, they have been shown to inhibit apoptosis and to promote angiogenesis through poorly understood paracrine effects. We previously reported that the direct injection of exosomes derived from CPCs (CPCexo) into mouse hearts provides protection against apoptosis in a model of acute ischemia/reperfusion injury. Moreover, we and others have reported that reactive oxygen species (ROS) derived from NADPH oxidase (NOX) can enhance angiogenesis in endothelial cells (ECs). Here we examined whether bioengineered CPCexo transfected with a pro-angiogenic miR-322 (CPCexo-322) can improve therapeutic efficacy in a mouse model of MI as compared to CPCexo. Systemic administration of CPCexo-322 in mice after ischemic injury provided greater protection post-MI than control CPCexo, in part, through enhanced angiogenesis in the border zones of infarcted hearts. Mechanistically, the treatment of cultured human ECs with CPCexo-322 resulted in a greater angiogenic response, as determined by increased EC migration and capillary tube formation via increased Nox2-derived ROS. Our study reveals that the engineering of CPCexo via microRNA (miR) programing can enhance angiogenesis, and this may be an effective therapeutic strategy for the treatment of ischemic cardiovascular diseases.
Collapse
|
7
|
Boyce ST, Lalley AL. Tissue engineering of skin and regenerative medicine for wound care. BURNS & TRAUMA 2018; 6:4. [PMID: 30009192 PMCID: PMC6040609 DOI: 10.1186/s41038-017-0103-y] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 12/12/2017] [Indexed: 12/11/2022]
Abstract
Engineering of biologic skin substitutes has progressed over time from individual applications of skin cells, or biopolymer scaffolds, to combinations of cells and scaffolds for treatment, healing, and closure of acute and chronic skin wounds. Skin substitutes may be categorized into three groups: acellular scaffolds, temporary substitutes containing allogeneic skin cells, and permanent substitutes containing autologous skin cells. Combined use of acellular dermal substitutes with permanent skin substitutes containing autologous cells has been shown to provide definitive wound closure in burns involving greater than 90% of the total body surface area. These advances have contributed to reduced morbidity and mortality from both acute and chronic wounds but, to date, have failed to replace all of the structures and functions of the skin. Among the remaining deficiencies in cellular or biologic skin substitutes are hypopigmentation, absence of stable vascular and lymphatic networks, absence of hair follicles, sebaceous and sweat glands, and incomplete innervation. Correction of these deficiencies depends on regulation of biologic pathways of embryonic and fetal development to restore the full anatomy and physiology of uninjured skin. Elucidation and integration of developmental biology into future models of biologic skin substitutes promises to restore complete anatomy and physiology, and further reduce morbidity from skin wounds and scar. This article offers a review of recent advances in skin cell thrapies and discusses the future prospects in cutaneous regeneration.
Collapse
Affiliation(s)
- Steven T Boyce
- 1Department of Surgery, University of Cincinnati, P.O. Box 670558, Cincinnati, Ohio 45267-0558 USA.,2Research Department, Shriners Hospitals for Children, Cincinnati, Ohio USA
| | - Andrea L Lalley
- 2Research Department, Shriners Hospitals for Children, Cincinnati, Ohio USA
| |
Collapse
|
8
|
Baldari S, Di Rocco G, Piccoli M, Pozzobon M, Muraca M, Toietta G. Challenges and Strategies for Improving the Regenerative Effects of Mesenchymal Stromal Cell-Based Therapies. Int J Mol Sci 2017; 18:E2087. [PMID: 28974046 PMCID: PMC5666769 DOI: 10.3390/ijms18102087] [Citation(s) in RCA: 173] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 09/18/2017] [Accepted: 09/28/2017] [Indexed: 12/11/2022] Open
Abstract
Cell-based therapies have the potential to revolutionize current treatments for diseases with high prevalence and related economic and social burden. Unfortunately, clinical trials have made only modest improvements in restoring normal function to degenerating tissues. This limitation is due, at least in part, to the death of transplanted cells within a few hours after transplant due to a combination of mechanical, cellular, and host factors. In particular, mechanical stress during implantation, extracellular matrix loss upon delivery, nutrient and oxygen deprivation at the recipient site, and host inflammatory response are detrimental factors limiting long-term transplanted cell survival. The beneficial effect of cell therapy for regenerative medicine ultimately depends on the number of administered cells reaching the target tissue, their viability, and their promotion of tissue regeneration. Therefore, strategies aiming at improving viable cell engraftment are crucial for regenerative medicine. Here we review the major factors that hamper successful cell engraftment and the strategies that have been studied to enhance the beneficial effects of cell therapy. Moreover, we provide a perspective on whether mesenchymal stromal cell-derived extracellular vesicle delivery, as a cell-free regenerative approach, may circumvent current cell therapy limitations.
Collapse
Affiliation(s)
- Silvia Baldari
- Department of Research, Advanced Diagnostic, and Technological Innovation, Regina Elena National Cancer Institute, via E. Chianesi 53, Rome 00144, Italy.
| | - Giuliana Di Rocco
- Department of Research, Advanced Diagnostic, and Technological Innovation, Regina Elena National Cancer Institute, via E. Chianesi 53, Rome 00144, Italy.
| | - Martina Piccoli
- Stem Cells and Regenerative Medicine Laboratory, Foundation Institute of Pediatric Research "Città della Speranza", corso Stati Uniti 4, Padova 35127, Italy.
| | - Michela Pozzobon
- Department of Women's and Children's Health, University of Padova, Via Giustiniani 3, Padova 35128, Italy.
| | - Maurizio Muraca
- Department of Women's and Children's Health, University of Padova, Via Giustiniani 3, Padova 35128, Italy.
| | - Gabriele Toietta
- Department of Research, Advanced Diagnostic, and Technological Innovation, Regina Elena National Cancer Institute, via E. Chianesi 53, Rome 00144, Italy.
| |
Collapse
|
9
|
Der Sarkissian S, Lévesque T, Noiseux N. Optimizing stem cells for cardiac repair: Current status and new frontiers in regenerative cardiology. World J Stem Cells 2017; 9:9-25. [PMID: 28154736 PMCID: PMC5253186 DOI: 10.4252/wjsc.v9.i1.9] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 09/20/2016] [Accepted: 10/24/2016] [Indexed: 02/06/2023] Open
Abstract
Cell therapy has the potential to improve healing of ischemic heart, repopulate injured myocardium and restore cardiac function. The tremendous hope and potential of stem cell therapy is well understood, yet recent trials involving cell therapy for cardiovascular diseases have yielded mixed results with inconsistent data thereby readdressing controversies and unresolved questions regarding stem cell efficacy for ischemic cardiac disease treatment. These controversies are believed to arise by the lack of uniformity of the clinical trial methodologies, uncertainty regarding the underlying reparative mechanisms of stem cells, questions concerning the most appropriate cell population to use, the proper delivery method and timing in relation to the moment of infarction, as well as the poor stem cell survival and engraftment especially in a diseased microenvironment which is collectively acknowledged as a major hindrance to any form of cell therapy. Indeed, the microenvironment of the failing heart exhibits pathological hypoxic, oxidative and inflammatory stressors impairing the survival of transplanted cells. Therefore, in order to observe any significant therapeutic benefit there is a need to increase resilience of stem cells to death in the transplant microenvironment while preserving or better yet improving their reparative functionality. Although stem cell differentiation into cardiomyocytes has been observed in some instance, the prevailing reparative benefits are afforded through paracrine mechanisms that promote angiogenesis, cell survival, transdifferentiate host cells and modulate immune responses. Therefore, to maximize their reparative functionality, ex vivo manipulation of stem cells through physical, genetic and pharmacological means have shown promise to enable cells to thrive in the post-ischemic transplant microenvironment. In the present work, we will overview the current status of stem cell therapy for ischemic heart disease, discuss the most recurring cell populations employed, the mechanisms by which stem cells deliver a therapeutic benefit and strategies that have been used to optimize and increase survival and functionality of stem cells including ex vivo preconditioning with drugs and a novel “pharmaco-optimizer” as well as genetic modifications.
Collapse
|
10
|
Shafiei MS, Lui S, Rockey DC. Integrin-linked kinase regulates endothelial cell nitric oxide synthase expression in hepatic sinusoidal endothelial cells. Liver Int 2015; 35:1213-21. [PMID: 24906011 PMCID: PMC4258191 DOI: 10.1111/liv.12606] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2013] [Accepted: 05/26/2014] [Indexed: 12/30/2022]
Abstract
BACKGROUND & AIMS Portal hypertension results from endothelial dysfunction after liver injury caused in part by abnormal production of endothelial cell derived nitric oxide synthase (eNOS). Here, we have postulated that endothelial mechanosensing pathways involving integrin-linked kinase (ILK) may play a critical role in portal hypertension, eNOS expression and function. In this study, we investigated the role of ILK and the small GTP-binding protein, Rho, in sinusoidal endothelial cell (SEC) eNOS regulation and function. METHODS Primary liver SECs were isolated using standard techniques. Liver injury was induced by performing bile duct ligation (BDL). To examine the expression of Rho and ILK in vivo during wound healing, SECs were infected with constitutively active Rho (V14), a dominant negative Rho (N19) and constructs encoding ILK and a short hairpin-inhibiting ILK. RESULTS Integrin-linked kinase expression was increased in SECs after liver injury; endothelin-1, vascular endothelial growth factor, and transforming growth factor beta-1 stimulated ILK expression in SECs. ILK expression in turn led to eNOS upregulation and to enhance eNOS phosphorylation and NO production. ILK knockdown or ILK (kinase) inhibition reduced eNOS mRNA expression, promoter activity, eNOS expression and ultimately NO production. In contrast, ILK overexpression had the opposite effect. Inhibition of ILK activity also disrupted the actin cytoskeleton in isolated SECs. Rho overexpression suppressed phosphorylation of the serine-threonine kinase, Akt and inhibited eNOS phosphorylation. Finally, inhibition of Rho function with the RGS domain of the p115-Rho-specific GEF (p115-RGS) significantly increased eNOS phosphorylation. CONCLUSIONS Our data suggest a potential role for ILK, the cytoskeleton and ILK signalling partners including Rho in regulating intrahepatic SEC eNOS expression and function.
Collapse
Affiliation(s)
- Mahnoush S. Shafiei
- Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, TX
| | - Songling Lui
- The Department of Medicine, The Medical University of South Carolina, Charleston, SC
| | - Don C. Rockey
- The Department of Medicine, The Medical University of South Carolina, Charleston, SC
| |
Collapse
|
11
|
García-Jérez A, Luengo A, Carracedo J, Ramírez-Chamond R, Rodriguez-Puyol D, Rodriguez-Puyol M, Calleros L. Effect of uraemia on endothelial cell damage is mediated by the integrin linked kinase pathway. J Physiol 2014; 593:601-18; discussion 618. [PMID: 25398526 DOI: 10.1113/jphysiol.2014.283887] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 11/08/2014] [Indexed: 12/24/2022] Open
Abstract
KEY POINTS Patients with chronic kidney disease have a higher risk of developing cardiovascular diseases than the general population. Their vascular endothelium is dysfunctional, among other things, because it is permanently exposed to uraemic toxins, several of which have poor clearance by conventional dialysis. Recent studies have demonstrated the important role of integrin-linked kinase (ILK) in the maintenance of endothelial integrity and in this study we investigate the involvement of ILK in the mechanism underlying vascular endothelial damage that occurs in uraemia. For the first time, we demonstrate the implication of ILK in the protection against endothelial cell damage (inhibition of proliferation, toxicity, oxidative stress and programed cell death) induced by uraemic serum from chronic kidney disease patients and uraemic toxins. This molecular mechanism may have clinical relevance because it highlights the importance of maintaining high levels of ILK activity to help preserve endothelial integrity, at least in early stages of chronic kidney disease. ABSTRACT Patients with chronic kidney disease (CKD) have a higher risk of developing cardiovascular diseases. Their vascular endothelium is dysfunctional, among other things, because it is permanently exposed to uraemic toxins, several of which, mostly protein-bound compounds such as indoxyl sulfate (IS) and p-cresyl sulphate, having poor clearance by conventional dialysis, induce endothelial toxicity. However, the molecular mechanism by which uraemic toxins regulate early stages of endothelial dysfunction remains unclear. Recent studies have demonstrated the important role of integrin-linked kinase (ILK) in the maintenance of endothelial integrity. In this study, we investigate the involvement of ILK in the mechanism underlying vascular endothelial damage that occurs in uraemia. First, we show that incubation of EA.hy926 cells with human uraemic serum from CKD patients upregulates ILK activity. This ILK activation also occurs when the cells are exposed to IS (25-100 μg ml(-1)), p-cresol (10-100 μg ml(-1)) or both combined, compared to human serum control. Next, we observed that high doses of both toxins together induce a slight decrease in cell proliferation and increase apoptosis and reactive oxygen species production. Interestingly, these toxic effects displayed a strong increase when the ILK protein is knocked down by small interfering RNA, even at low doses of uraemic toxins. Abrogation of AKT has demonstrated the ILK/AKT signalling pathway involved in these processes. This study has demonstrated the implication of ILK in the protection against endothelial cell damage induced by uraemic toxins, a molecular mechanism that could play a protective role in the early stages of endothelial dysfunction observed in uraemic patients.
Collapse
Affiliation(s)
- Andrea García-Jérez
- Department of Systems Biology, Universidad de Alcalá, Madrid, Spain; IRSIN, Spain; REDinREN (Instituto de Salud Carlos III), Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
12
|
Yen CF, Wang HS, Lee CL, Liao SK. Roles of integrin-linked kinase in cell signaling and its perspectives as a therapeutic target. Gynecol Minim Invasive Ther 2014. [DOI: 10.1016/j.gmit.2014.06.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
13
|
The role of integrin α2 in cell and matrix therapy that improves perfusion, viability and function of infarcted myocardium. Biomaterials 2014; 35:4749-58. [PMID: 24631247 DOI: 10.1016/j.biomaterials.2014.02.028] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 02/16/2014] [Indexed: 12/13/2022]
Abstract
Injectable delivery matrices hold promise in enhancing engraftment and the overall efficacy of cardiac cell therapies; however, the mechanisms responsible remain largely unknown. Here we studied the interaction of a collagen matrix with circulating angiogenic cells (CACs) in a mouse myocardial infarction model. CACs + matrix treatment enhanced CAC engraftment, and improved myocardial perfusion, viability and function compared to cells or matrix alone. Integrin-linked kinase (ILK) was up-regulated in matrix-cultured CACs. Integrin α2β1 blocking prevented ILK up-regulation, significantly reduced the adhesion, proliferation, and paracrine properties of matrix-cultured CACs, and negated the benefits of CACs + matrix therapy in vivo. Furthermore, integrin α5 was essential for the angiogenic potential of CACs on matrix. These findings indicate that the synergistic therapeutic effect of CACs + matrix therapy in MI requires the matrix to enhance CAC function via α2β1 and α5 integrin signaling mechanisms, rather than simply delivering the cells.
Collapse
|
14
|
Cui K, Yan T, Luo Q, Zheng Y, Liu X, Huang X, Zou L. Ultrasound microbubble-mediated delivery of integrin-linked kinase gene improves endothelial progenitor cells dysfunction in pre-eclampsia. DNA Cell Biol 2014; 33:301-10. [PMID: 24564279 DOI: 10.1089/dna.2013.2229] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Pre-eclampsia (PE) is a specific vascular complication in pregnancy whose precise mechanism is still unclear. We hypothesized that endothelial progenitor cells (EPCs), the precursor of endothelial cells, might be impaired in patients with PE and hold a great promise for the treatment of PE. In the present study, we analyzed the EPCs number and expression of integrin-linked kinase (ILK) in PE patients. We confirmed that both EPCs number and ILK expression were diminished in PE patients. Next, we transfected EPCs with ILK gene using ultrasonic microbubble technique (UMT) for the first time, as UMT is a novel type of gene transfer technology showing promising applications in stem cells apart from EPCs. To further investigate the transfection efficiency of UMT, RT-PCR analysis and western blot were used to examine the messenger RNA (mRNA) and protein level of ILK. After transfection of the ILK gene, EPCs function was tested to illustrate the role of ILK in cell proliferation, apoptosis, migration, and secretion. The results of the in vitro study suggested that UMT, a novel gene delivery system, could be considered a potent physical method for EPCs transfection. Moreover, the growth and angiogenetic properties of EPCs are enhanced by introducing ILK. This study may afford a new trend for EPCs transfection and gene therapy in PE.
Collapse
Affiliation(s)
- Kai Cui
- 1 Department of Obstetrics and Gynecology, Union Hospital, Huazhong University of Science and Technology , Wuhan, China
| | | | | | | | | | | | | |
Collapse
|
15
|
Roura S, Gálvez-Montón C, Pujal JM, Casani L, Fernández MA, Astier L, Gastelurrutia P, Domingo M, Prat-Vidal C, Soler-Botija C, Llucià-Valldeperas A, Llorente-Cortés V, Bayes-Genis A. New insights into lipid raft function regulating myocardial vascularization competency in human idiopathic dilated cardiomyopathy. Atherosclerosis 2013; 230:354-64. [DOI: 10.1016/j.atherosclerosis.2013.08.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 07/26/2013] [Accepted: 08/06/2013] [Indexed: 12/15/2022]
|
16
|
Vascular Regeneration: Endothelial Progenitor Cell Therapy for Ischemic Diseases. Regen Med 2013. [DOI: 10.1007/978-94-007-5690-8_35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
17
|
Cho HJ, Lee HJ, Youn SW, Koh SJ, Won JY, Chung YJ, Cho HJ, Yoon CH, Lee SW, Lee EJ, Kwon YW, Lee HY, Lee SH, Ho WK, Park YB, Kim HS. Secondary sphere formation enhances the functionality of cardiac progenitor cells. Mol Ther 2012; 20:1750-66. [PMID: 22713697 DOI: 10.1038/mt.2012.109] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Loss of cardiomyocytes impairs cardiac function after myocardial infarction (MI). Recent studies suggest that cardiac stem/progenitor cells could repair the damaged heart. However, cardiac progenitor cells are difficult to maintain in terms of purity and multipotency when propagated in two-dimensional culture systems. Here, we investigated a new strategy that enhances potency and enriches progenitor cells. We applied the repeated sphere formation strategy (cardiac explant → primary cardiosphere (CS) formation → sphere-derived cells (SDCs) in adherent culture condition → secondary CS formation by three-dimensional culture). Cells in secondary CS showed higher differentiation potentials than SDCs. When transplanted into the infarcted myocardium, secondary CSs engrafted robustly, improved left ventricular (LV) dysfunction, and reduced infarct sizes more than SDCs did. In addition to the cardiovascular differentiation of transplanted secondary CSs, robust vascular endothelial growth factor (VEGF) synthesis and secretion enhanced neovascularization in the infarcted myocardium. Microarray pathway analysis and blocking experiments using E-selectin knock-out hearts, specific chemicals, and small interfering RNAs (siRNAs) for each pathway revealed that E-selectin was indispensable to sphere initiation and ERK/Sp1/VEGF autoparacrine loop was responsible for sphere maturation. These results provide a simple strategy for enhancing cellular potency for cardiac repair. Furthermore, this strategy may be implemented to other types of stem/progenitor cell-based therapy.
Collapse
Affiliation(s)
- Hyun-Jai Cho
- Cardiovascular Center & Department of Internal Medicine, Seoul National University, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Wang F, Wang Y, Zhang L, Zou L. Gene modification with integrin-linked kinase improves function of endothelial progenitor cells in pre-eclampsia in vitro. J Cell Biochem 2012; 112:3103-11. [PMID: 21688303 DOI: 10.1002/jcb.23236] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Integrin-linked kinase (ILK), a multifunctional serine-threonine protein kinase, has been shown to have implications for the treatment of ischemia vascular diseases by promoting angiogenesis in various tissues. However, whether this kinase has therapeutic potential in pre-eclampsia is not well studied. In this report, we determined the changes in the production and action of ILK on endothelial progenitor cells (EPCs) isolated from patients with pre-eclampsia. The effects of ILK transfection on proliferation, migration, and angiogenesis of EPCs were investigated. We showed that EPCs transfected with the ILK gene expressed high levels of ILK protein and mRNA. Transfection with ILK also enhanced the proliferative, migratory, and angiogenic capabilities of EPCs, and promoted the production of VEGF. These results suggest that ILK gene transfection is an effective approach to augment angiogenic properties of EPCs in vitro and providing basis for clinical cell-based gene therapy in patients with pre-eclampsia.
Collapse
Affiliation(s)
- Fang Wang
- Department of Gynecology and Obstetrics, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China
| | | | | | | |
Collapse
|
19
|
Zhang H, Han Y, Tao J, Liu S, Yan C, Li S. Cellular repressor of E1A-stimulated genes regulates vascular endothelial cell migration by the ILK/AKT/mTOR/VEGF(165) signaling pathway. Exp Cell Res 2011; 317:2904-13. [PMID: 21939655 DOI: 10.1016/j.yexcr.2011.08.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Revised: 08/19/2011] [Accepted: 08/20/2011] [Indexed: 02/07/2023]
Abstract
The migration of vascular endothelial cells plays a critical role in a variety of vascular physiological and pathological processes, such as embryonic development, angiogenesis, wound healing, re-endothelialization, and vascular remodeling. This study clarified the role and mechanism of a new vascular homeostasis regulator, Cellular repressor of E1A-stimulated genes (CREG), in the migration of primary human umbilical vein endothelial cells (HUVECs). A wound healing assay and transwell migration model showed that upregulation of CREG expression induced HUVEC migration and it was positively correlated with the expression of vascular endothelial growth factor. Furthermore, wild type integrin-linked kinase reversed the poor mobility of CREG knock-down HUVECs; in contrast, kinase-dead integrin-linked kinase weakened the migration of HUVECs. We also studied the effect of CREG on HUVEC migration by the addition of an mTOR inhibitor, recombinant vascular endothelial growth factor(165), neutralizing antibody of vascular endothelial growth factor(165) and AKT siRNA, and we concluded that CREG induces endothelial cell migration by activating the integrin-linked kinase/AKT/mTOR/VEGF(165) signaling pathway.
Collapse
Affiliation(s)
- Huimin Zhang
- Department of Cardiology, Cardiovascular Research Institute, Shenyang Northern Hospital, Shenyang, China
| | | | | | | | | | | |
Collapse
|
20
|
Alev C, Ii M, Asahara T. Endothelial progenitor cells: a novel tool for the therapy of ischemic diseases. Antioxid Redox Signal 2011; 15:949-65. [PMID: 21254837 DOI: 10.1089/ars.2010.3872] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Circulating endothelial progenitor cells (EPCs) are believed to home to sites of neovascularization, contributing to vascular regeneration either directly via incorporation into newly forming vascular structures or indirectly via the secretion of pro-angiogenic growth factors, thereby enhancing the overall vascular and hemodynamic recovery of ischemic tissues. The therapeutic application of EPCs has been shown to be effective in animal models of ischemia, and we as well as other groups involved in clinical trials have demonstrated that the use of EPCs was safe and feasible for the treatment of critical limb ischemia and cardiovascular diseases. However, many issues in the field of EPC biology, especially in regard to the proper and unambiguous molecular characterization of these cells, still remain unresolved, hampering not only basic research but also the effective therapeutic use and widespread application of these cells. Further, recent evidence suggests that several diseases and pathological conditions are correlated with a reduction in the number and biological activity of EPCs, making the development of novel strategies to overcome the current limitations and shortcomings of this promising but still limited therapeutic tool by refinement and improvement of EPC purification, expansion, and administration techniques, a rather pressing issue.
Collapse
Affiliation(s)
- Cantas Alev
- Group of Vascular Regeneration Research, Institute of Biomedical Research and Innovation/RIKEN Center for Developmental Biology, Kobe, Japan
| | | | | |
Collapse
|
21
|
Lee DW, Lee TK, Cho IS, Park HE, Jin S, Cho HJ, Kim SH, Oh S, Kim HS. Creation of myocardial fibrosis by transplantation of fibroblasts primed with survival factors. Am J Physiol Heart Circ Physiol 2011; 301:H1004-14. [PMID: 21685267 DOI: 10.1152/ajpheart.00156.2011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
One of the major obstacles in the creation of myocardial fibrosis using fibroblasts is massive cell death after cell injection. To overcome this problem, a method that delivers fibroblasts primed with survival factors was studied. Cardiac fibroblasts were isolated from wild-type male C57BL/6 mice. Female mice were randomly placed into the following three groups: 1) fibroblasts transfected with β-galactosidase-containing adenovirus (control group), 2) fibroblasts treated with a necrosis inhibitor (NI group), and 3) fibroblasts transfected with Akt-containing adenovirus (Akt group). Pretreated cells were transplanted into the recipient heart by direct injection after a thoracotomy. Quantitative real-time PCR and morphometric analysis were performed to investigate the effects of survival factor priming on the induction of cell engraftment and fibrosis. In addition, a canine model was used to investigate the development of fibrosis and conduction modification using autologous dermal fibroblasts. The NI and Akt groups showed a better engraftment rate: 13 (NI group) and 7 (Akt group) times greater at 21 days compared with the control group. Increased fibrosis and conduction delay were also observed in the NI and Akt groups compared with the control group. Survival factor priming increased cellular engraftment and enhanced the efficacy of cell transplantation. Delivery of fibroblasts primed with survival factors might be a promising approach to develop conduction modification as a novel strategy to treat arrhythmias.
Collapse
Affiliation(s)
- Dong-Won Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Inhibition of high glucose-induced VEGF and ICAM-1 expression in human retinal pigment epithelium cells by targeting ILK with small interference RNA. Mol Biol Rep 2011; 39:613-20. [PMID: 21553049 DOI: 10.1007/s11033-011-0778-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2010] [Accepted: 04/27/2011] [Indexed: 01/09/2023]
Abstract
The aim of this study was to investigate the change of Integrin-linked kinase (ILK) expression of human retinal pigment epithelium (RPE) cells in response to high glucose, and the effect of targeting ILK with small interference RNA (siRNA) on the high glucose-induced expression of vascular endothelial growth factor (VEGF) and intercellular adhesion molecule-1 (ICAM-1). The ILK mRNA and protein expression in human RPE cells were analyzed with RT-PCR and western blot after exposure to 5.5, 30, 40, 50 mM glucose, or 5.5 mM glucose+45.5 mM mannitol for 48 h. The expression of VEGF and ICAM-1 was also determined. Cells were treated with ILK siRNA, to determine the effect of ILK on VEGF and ICAM-1 expression following treatment with high glucose. High concentrations of glucose significantly up-regulated ILK mRNA and protein expression, and the ILK expression increased along with the glucose concentration. The changes of VEGF and ICAM-1 expression were similar to that of ILK expression. Knocking down ILK gene expression with siRNA inhibited the elevation of VEGF and ICAM-1 induced by high glucose treatment. These results suggested that ILK was involved in the response of RPE cells to high glucose and may therefore play a role in the pathogenesis of diabetic ophthalmology.
Collapse
|
23
|
COMP-Ang1 stimulates HIF-1α-mediated SDF-1 overexpression and recovers ischemic injury through BM-derived progenitor cell recruitment. Blood 2011; 117:4376-86. [PMID: 21200018 DOI: 10.1182/blood-2010-07-295964] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Recruitment and adhesion of bone marrow (BM)-derived circulating progenitor cells to ischemic tissue are important for vasculogenesis and tissue repair. Recently, we found cartilage oligomeric matrix protein (COMP)-Ang1 is a useful cell-priming agent to improve the therapeutic efficacy of progenitor cells. However, the effect and the underlying mechanisms of COMP-Ang1 on recruitment of BM-derived progenitor cells (BMPCs) to foci of vascular injury have not been well defined. Here, we found that COMP-Ang1 is a critical stimulator of stromal cell-derived factor 1 (SDF-1), the principal regulator of BM-cell trafficking. Furthermore, SDF-1 stimulation by COMP-Ang1 was blocked by small-interfering RNA (siRNA) against hypoxia-inducible factor-1α (HIF-1α). COMP-Ang1 increased the synthesis of HIF-1α by activating mammalian target of rapamycin (mTOR) in hypoxic endothelium. The intermediate mechanism transmitting the COMP-Ang1 signal to the downstream mTOR/HIF-1α/SDF-1 pathway was the enhanced binding of the Tie2 receptor with integrin-linked kinase (ILK), an upstream activator of mTOR. In the mouse ischemic model, local injection of COMP-Ang1 stimulated the incorporation of BMPCs into ischemic limb, thereby enhancing neovasculogenesis and limb salvage. Collectively, our findings identify the COMP-Ang1/HIF-1α/SDF-1 pathway as a novel inducer of BMPC recruitment and neovasculogenesis in ischemic disease.
Collapse
|
24
|
Kawamoto A, Ii M, Asahara T. Vascular Regeneration: Endothelial Progenitor Cell Therapy for Ischemic Diseases. Regen Med 2011. [DOI: 10.1007/978-90-481-9075-1_31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
25
|
Tan Q, Qiu L, Li G, Li C, Zheng C, Meng H, Yang W. Transplantation of healthy but not diabetic outgrowth endothelial cells could rescue ischemic myocardium in diabetic rabbits. Scandinavian Journal of Clinical and Laboratory Investigation 2010; 70:313-21. [PMID: 20470214 DOI: 10.3109/00365511003774593] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVE There are two types of endothelial progenitor cell (EPC) in circulation, early EPC and outgrowth endothelial cell (OEC). Diabetes impairs the function of EPC, but it is not clear whether transplantation of OECs can rescue ischemic myocardium in diabetes. In this study, we compared the function of diabetic and healthy OECs in vitro. Then we administered diabetic and healthy OECs intramyocardially and compared their contribution to vasculogenesis in diabetic rabbits. METHODS Outgrowth endothelial cells from diabetic and healthy rabbits were isolated and subjected to in vitro proliferation, tube-forming, angiogenic cytokine assays. Exogenous diabetic and healthy OECs were analyzed for therapeutic efficacy in an acute ischemia model of diabetic rabbits. LV function was assessed using echocardiography. The capillary density and fibrosis area were evaluated. MRNA expression of VEGF and bFGF was analyzed using relative realtime quantitive PCR. RESULTS Proliferation, tube-forming, secretion of VEGF and bFGF of diabetic OECs were significantly reduced compared with healthy OECs. In diabetic rabbits, healthy OECs transplantation could increase capillary density and improve cardiac function, decrease fibrosis area compared with diabetic OEC and the control group. Real time PCR indicated that mRNA expression of VEGF and bFGF were augmented more in the healthy OEC group than those in the control and diabetic OEC groups. CONCLUSIONS These findings suggest that diabetes impairs the function of OECs. Transplantation of healthy OECs may rescue the ischemic myocardium by neovasculogenesis and paracrine effect in diabetic rabbits. However, autologous transplantation of diabetic OEC could not enhance cardiac function.
Collapse
Affiliation(s)
- Qiang Tan
- Department of Cardiology, The First Hospital of Qinhuangdao, Hebei Medical University, Qinhuangdao, Hebei, China
| | | | | | | | | | | | | |
Collapse
|
26
|
Kim H, Cho HJ, Kim SW, Liu B, Choi YJ, Lee J, Sohn YD, Lee MY, Houge MA, Yoon YS. CD31+ cells represent highly angiogenic and vasculogenic cells in bone marrow: novel role of nonendothelial CD31+ cells in neovascularization and their therapeutic effects on ischemic vascular disease. Circ Res 2010; 107:602-14. [PMID: 20634489 DOI: 10.1161/circresaha.110.218396] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
RATIONALE Bone marrow (BM) cells play an important role in physiological and therapeutic neovascularization. However, it remains unclear whether any specific uncultured BM cell populations have higher angiogenic and vasculogenic activities. Moreover, there has been controversy regarding the vasculogenic ability of BM cells. OBJECTIVE Preliminary flow cytometric analysis showed that CD31, traditionally a marker for endothelial cells, is expressed in certain nonendothelial BM mononuclear cells in both human and mouse. Based on the conserved CD31 expression in the axis of hematopoietic stem/progenitor cells (HSC/HPCs) to endothelial cells, we further sought to determine the comprehensive vasculogenic and angiogenic characteristics of human and mouse BM-derived CD31(+) cells. METHODS AND RESULTS Flow cytometric analysis demonstrated that all CD31(+) cells derived from BM were CD45(+) and expressed markers for both HSC/HPCs and endothelial cells. Comprehensive gene expression analyses revealed that BM-CD31(+) cells expressed higher levels of angiogenic genes than CD31(-) cells. Endothelial progenitor cells, as well as HSC/HPCs, were almost exclusively confined to the CD31(+) cell fraction, and culture of CD31(+) cells under defined conditions gave rise to endothelial cells. Finally, injection of CD31(+) cells into ischemic hindlimb repaired ischemia, increased expression of angiogenic and chemoattractive factors, and, in part, directly contributed to vasculogenesis, as demonstrated by both 3D confocal microscopy and flow cytometry. CONCLUSIONS These data indicate that BM-CD31(+) cells represent highly angiogenic and vasculogenic cells and can be a novel and highly promising source of cells for cell therapy to treat ischemic cardiovascular diseases.
Collapse
Affiliation(s)
- Hyongbum Kim
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1639 Pierce Drive, Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Song H, Song BW, Cha MJ, Choi IG, Hwang KC. Modification of mesenchymal stem cells for cardiac regeneration. Expert Opin Biol Ther 2010; 10:309-19. [PMID: 20132054 DOI: 10.1517/14712590903455997] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
IMPORTANCE OF THE FIELD Mesenchymal stem cells (MSCs) have the greatest potential for use in cell-based therapy of human heart diseases, especially in myocardial infarcts. The therapeutic potential of MSCs in myocardial repair is based on the ability of MSCs to directly differentiate into cardiac tissue and on the paracrine actions of factors released from MSCs. However, the major obstacle in the clinical application of MSC-based therapy is the poor viability of the transplanted cells due to harsh microenvironments like ischemia, inflammation and/or anoikis in the infarcted myocardium. Recently, various approaches have been implemented in an effort to improve the survival of implanted MSCs through ex vivo manipulation of MSCs. AREAS COVERED IN THIS REVIEW Major obstacles in MSC-based therapy are discussed, along with recent advances for enhancing therapeutic potential of engrafted MSCs from the past decade. WHAT THE READER WILL GAIN This review focuses primarily on ex vivo manipulation of MSCs before transplantation, which includes pretreatment, preconditioning and genetic modification of MSCs, and future directions. TAKE HOME MESSAGE Modification of MSCs before transplantation has developed into a promising option for enhancing the beneficial effects of MSC-based therapy for cardiac repair after myocardial infarction.
Collapse
Affiliation(s)
- Heesang Song
- Yonsei University College of Medicine, Cardiovascular Research Institute, 250 Seongsanno,Seodaemun-gu, Seoul, Republic of Korea
| | | | | | | | | |
Collapse
|
28
|
Chavakis E, Koyanagi M, Dimmeler S. Enhancing the outcome of cell therapy for cardiac repair: progress from bench to bedside and back. Circulation 2010; 121:325-35. [PMID: 20083719 DOI: 10.1161/circulationaha.109.901405] [Citation(s) in RCA: 116] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Emmanouil Chavakis
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University, Frankfurt, Germany
| | | | | |
Collapse
|
29
|
Song SW, Chang W, Song BW, Song H, Lim S, Kim HJ, Cha MJ, Choi E, Im SH, Chang BC, Chung N, Jang Y, Hwang KC. Integrin-linked kinase is required in hypoxic mesenchymal stem cells for strengthening cell adhesion to ischemic myocardium. Stem Cells 2009; 27:1358-65. [PMID: 19489098 DOI: 10.1002/stem.47] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Mesenchymal stem cells (MSCs) therapy has limitations due to the poor viability of MSCs after cell transplantation. Integrin-mediated adhesion is a prerequisite for cell survival. As a novel anti-death strategy to improve cell survival in the infarcted heart, MSCs were genetically modified to overexpress integrin-linked kinase (ILK). The survival rate of ILK-transfected MSCs (ILK-MSCs) was augmented by about 1.5-fold and the phosphorylation of ERK1/2 and Akt in ILK-MSCs were increased by about three and twofold, respectively. ILK-MSCs demonstrated an increase of twofold in the ratio of Bcl-2/Bax and inhibited caspase-3 activation, compared with hypoxic MSCs. The adhesion rate of ILK-MSCs also had a 32.2% increase on the cardiac fibroblast-derived three-dimensional matrix and ILK-MSCs showed higher retention by about fourfold compared to unmodified MSCs. Six animals per group were used for the in vivo experiments analyzed at 1 week after occlusion of the left coronary artery. ILK-MSC transplanted rats had a 12.0% +/- 3.1% smaller infarct size than MSC-treated rats after ligation of left anterior descending coronary artery. Transplantation of ILK-MSCs not only led to a 16.0% +/- 0.4% decrease in the fibrotic heart area, but also significantly reduced the apoptotic positive index by two-thirds when compared with ligation only. The mean microvessel count per field in the infarcted myocardium of ILK-MSCs group was increased relative to the sham group and MSCs group. In conclusion, the ILK gene transduction of MSCs further assisted cell survival and adhesion, and improved myocardial damage when compared with MSC only after transplantation.
Collapse
Affiliation(s)
- Suk-Won Song
- Department of Thoracic and Cardiovascular Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
|
31
|
Sekiguchi H, Ii M, Losordo DW. The relative potency and safety of endothelial progenitor cells and unselected mononuclear cells for recovery from myocardial infarction and ischemia. J Cell Physiol 2009; 219:235-42. [PMID: 19115244 DOI: 10.1002/jcp.21672] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Endothelial progenitor cells (EPCs) are a subset of the total mononuclear cell population (tMNCs) that possess an enhanced potential for differentiation within the endothelial-cell lineage. Typically, EPCs are selected from tMNCs via the expression of both hematopoietic stem-cell markers and endothelial-cell markers, such as CD34, or by culturing tMNCs in media selective for endothelial cells. Both EPCs and tMNCs participate in vascular growth and regeneration, and their potential use for treatment of myocardial injury or disease has been evaluated in early-phase clinical studies. Direct comparisons between EPCs and tMNCs are rare, but the available evidence appears to favor EPCs, particularly CD34+ cells, and the potency of EPCs may be increased as much as 30-fold through genetic modification. However, these observations must be interpreted with caution because clinical investigations of EPC therapy are ongoing. We anticipate that with continued development, EPC therapy will become a safe and effective treatment option for patients with acute myocardial infarction or chronic ischemic disease.
Collapse
Affiliation(s)
- Haruki Sekiguchi
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | | | | |
Collapse
|
32
|
Wang YQ, Zhang XM, Wang XD, Wang BJ, Wang W. 17-AAG, a Hsp90 inhibitor, attenuates the hypoxia-induced expression of SDF-1alpha and ILK in mouse RPE cells. Mol Biol Rep 2009; 37:1203-9. [PMID: 19266313 DOI: 10.1007/s11033-009-9490-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2008] [Accepted: 02/20/2009] [Indexed: 01/08/2023]
Abstract
The aim of this study was to investigate the changes of SDF-1alpha and ILK expression in mouse retinal pigment epithelium (RPE) cells in response to hypoxia, and the effect of 17-Allylamino-17-demethoxygeldanamycin (17-AAG), a heat shock protein 90 (Hsp90) inhibitor, on the hypoxia-induced expression of SDF-1alpha and ILK. RPE cells were cultured with 200 micromol/L cobalt chloride (CoCl(2)) for different times (1, 3, 6, 12, 24, 72 h) to imitate chemical hypoxia. Pretreatment of 17-AAG was 1 h prior to hypoxic insult. Cellular viability after 17-AAG treatment was assessed by MTT assay, and the changes of SDF-1alpha and ILK expression were examined by RT-PCR and Western blot. Up-regulation of SDF-1alpha and ILK expression in response to hypoxia was observed. One hour pretreatment of 17-AAG could remarkably decreased the hypoxia-induced SDF-1alpha and ILK expression in vitro. Our results indicated that SDF-1alpha and ILK involved in the hypoxic response of RPE cells, and 1 h pretreatment of 17-AAG had an inhibitive effect on the hypoxia-induced SDF-1alpha and ILK expression.
Collapse
Affiliation(s)
- Ye Qing Wang
- Ophthalmology Hospital, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | | | | | | | | |
Collapse
|
33
|
Kang HJ, Kim HS. Safety and efficacy of intracoronary infusion of mobilized peripheral blood stem cell in patients with myocardial infarction: MAGIC Cell-1 and MAGIC Cell-3-DES-trials. Eur Heart J Suppl 2008. [DOI: 10.1093/eurheartj/sun049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
34
|
Werner C, Böhm M, Friedrich EB. Role of integrin-linked kinase for functional capacity of endothelial progenitor cells in patients with stable coronary artery disease. Biochem Biophys Res Commun 2008; 377:331-336. [DOI: 10.1016/j.bbrc.2008.09.081] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2008] [Accepted: 09/12/2008] [Indexed: 01/29/2023]
|
35
|
Jujo K, Ii M, Losordo DW. Endothelial progenitor cells in neovascularization of infarcted myocardium. J Mol Cell Cardiol 2008; 45:530-44. [PMID: 18755197 DOI: 10.1016/j.yjmcc.2008.08.003] [Citation(s) in RCA: 192] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2008] [Revised: 08/05/2008] [Accepted: 08/05/2008] [Indexed: 02/06/2023]
Abstract
Historically, revascularization of ischemic tissue was believed to occur through the migration and proliferation of endothelial cells in nearby tissues; however, evidence accumulated in recent years indicates that a subpopulation of adult, peripheral-blood cells, collectively referred to as endothelial progenitor cells (EPCs), can differentiate into mature endothelial cells. After ischemic insult, EPCs are believed to home to sites of neovascularization, where they contribute to vascular regeneration by forming a structural component of capillaries and by secreting angiogenic factors; new evidence indicates that EPCs can also differentiate into cardiomyocytes and smooth-muscle cells. These insights into the molecular and cellular processes of tissue formation suggest that cardiac function may be preserved after myocardial infarction by transplanting EPCs into ischemic heart tissue, thereby enhancing vascular and myocardial recovery. This therapeutic strategy has been effective in animal models of ischemic disorders, and results from randomized clinical trials suggest that cell-based strategies may be safe and feasible for treatment of myocardial infarction in humans and have provided early evidence of efficacy. However, the scarcity of EPCs in the peripheral blood and evidence that several disease states reduce EPC number and/or function have prompted the development of several strategies to overcome these limitations, such as the administration of genetically modified EPCs that overexpress angiogenic growth factors. To optimize therapeutic outcomes, researchers must continue to refine methods of EPC purification, expansion, and administration, and to develop techniques that overcome the intrinsic scarcity and phenotypic deficiencies of EPCs.
Collapse
Affiliation(s)
- Kentaro Jujo
- Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | | |
Collapse
|
36
|
Aranguren XL, Verfaillie CM, Luttun A. Emerging hurdles in stem cell therapy for peripheral vascular disease. J Mol Med (Berl) 2008; 87:3-16. [PMID: 18712330 DOI: 10.1007/s00109-008-0394-3] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2008] [Revised: 06/27/2008] [Accepted: 07/17/2008] [Indexed: 12/01/2022]
Abstract
Peripheral vascular disease (PVD) is a growing medical problem in Western societies and presents itself mainly in two different clinical forms. Intermittent claudication is an early moderate manifestation, while patients with critical limb ischemia suffer from severe muscle tissue loss or ulcers and are at high risk for limb amputation. Unfortunately, many patients cannot be helped with currently available surgical or endovascular revascularization procedures because of the complex anatomy of the vascular occlusion and/or the presence of other risk factors. Noninvasive stem cell therapy has been proposed as an alternative for such patients. Although pioneering clinical experience with stem cell-related therapy seems promising, it is too early for general clinical use of this technique, since many questions remain unanswered. Indeed, while questions about safety, dose, and administration route/timing/frequency are the first ones to be addressed when designing a stem cell-based clinical approach, there is accumulating evidence from recent (pre-)clinical studies that other issues may also be at stake. For instance, the choice of stem cells to be used and its precise mechanism of action, the need/possibility for concurrent tissue regeneration in case of irreversible tissue loss, the differentiation degree and specific vascular identity of the transplanted cells, and the long-term survival of engrafted cells in the absence of a normal supportive tissue environment should be well considered. Here, rather than presenting a comprehensive and extensive overview on the current literature on stem/progenitor cells and revascularization, we highlight some of the outstanding issues emerging from the recent (pre-)clinical literature that may codetermine the successful application of stem cells in a wide range of PVD patients in the future.
Collapse
Affiliation(s)
- Xabier L Aranguren
- Center for Molecular and Vascular Biology, Katholieke Universiteit Leuven (KULeuven), Campus Gasthuisberg, Herestraat 49, 3000, Leuven, Belgium
| | | | | |
Collapse
|
37
|
Hahn JY, Cho HJ, Kang HJ, Kim TS, Kim MH, Chung JH, Bae JW, Oh BH, Park YB, Kim HS. Pre-treatment of mesenchymal stem cells with a combination of growth factors enhances gap junction formation, cytoprotective effect on cardiomyocytes, and therapeutic efficacy for myocardial infarction. J Am Coll Cardiol 2008; 51:933-43. [PMID: 18308163 DOI: 10.1016/j.jacc.2007.11.040] [Citation(s) in RCA: 246] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2007] [Revised: 10/26/2007] [Accepted: 11/27/2007] [Indexed: 11/25/2022]
Abstract
OBJECTIVES The goal of this study was to investigate the effect of pre-treatment of mesenchymal stem cells (MSCs) with growth factors (GFs) on cardiomyogenic differentiation, cytoprotective action on cardiomyocytes (CMCs), and their therapeutic efficacy in myocardial infarction. BACKGROUND Mechanisms of myocardial repair with MSC transplantation have not been fully elucidated, and therapeutic efficacy needs to be enhanced. METHODS The MSCs obtained from the bone marrow of Fisher344 rats were treated with fibroblast growth factor-2, insulin-like growth factor-1, and bone morphogenetic protein-2. The expression of cardiac specific markers and the cytoprotective effect of MSCs with its mechanism were evaluated. Efficacy of MSCs transplantation was studied in rat myocardial infarction model. RESULTS Treatment of MSCs with cocktails of GFs enhanced expression of cardiac transcription factors and survival. Induction of cardiac specific markers by coculture with CMCs and gap junctional communication with CMCs was more active in GF-treated MSCs than untreated MSCs. The GF-treated MSCs reduced apoptosis of neighboring CMCs in a hypoxic condition and enhanced the phosphorylated Akt and phosphorylated c-AMP response element binding protein expression of CMCs, which was markedly reduced by gap junction blockade. In a rat myocardial infarction model, transplantation of GF-treated MSCs resulted in smaller infarct size and better cardiac function than transplantation of untreated MSCs. Additionally, GF treatment enhanced gap junction formation of transplanted MSCs, which did not aggravate arrhythmia. CONCLUSIONS Pre-treatment of MSCs with GFs enhanced cytoprotective effects on neighboring CMCs through gap junction and improved the therapeutic efficacy of MSC transplantation for myocardial repair. "Priming of MSCs with GFs" before transplantation might improve the therapeutic efficacy of cell therapy.
Collapse
Affiliation(s)
- Joo-Yong Hahn
- National Research Laboratory for Cardiovascular Stem Cells, Seoul National University College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Chavakis E, Urbich C, Dimmeler S. Homing and engraftment of progenitor cells: a prerequisite for cell therapy. J Mol Cell Cardiol 2008; 45:514-22. [PMID: 18304573 DOI: 10.1016/j.yjmcc.2008.01.004] [Citation(s) in RCA: 243] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2007] [Revised: 12/17/2007] [Accepted: 01/01/2008] [Indexed: 12/13/2022]
Abstract
Cell therapy is a promising therapeutic option for treating patients with ischemic diseases. The efficiency of cell therapy to augment recovery after ischemia depends on the sufficient recruitment of applied cells to the target tissue. Using in vivo imaging techniques the extent of homing was shown to be rather low in most experimental and clinical studies. The elucidation of the molecular mechanisms of homing of different progenitor cell subpopulation to sites of injury is essential for the development of new specific therapeutic strategies, in order to improve the efficacy of cell-based therapies. Homing to sites of active neovascularization is a complex process depending on a timely and spatially orchestrated interplay between chemokines (e.g. SDF-1), chemokine receptors, intracellular signaling, adhesion molecules (selectins and integrins) and proteases. The review will focus on the mechanisms underlying homing of adult bone marrow-derived hematopoietic cells, mesenchymal stem cells, and vasculogenic circulating cells and discuss strategies how to optimize cell engraftment.
Collapse
Affiliation(s)
- Emmanouil Chavakis
- Molecular Cardiology, Department of Internal Medicine III, University of Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | | | | |
Collapse
|
39
|
Joshi MB, Ivanov D, Philippova M, Erne P, Resink TJ. Integrin-linked kinase is an essential mediator for T-cadherin-dependent signaling via Akt and GSK3beta in endothelial cells. FASEB J 2007; 21:3083-95. [PMID: 17485554 DOI: 10.1096/fj.06-7723com] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Glycosylphosphatidylinositol-anchored T-cadherin (T-cad) influences several parameters of angiogenesis including endothelial cell (EC) differentiation, migration, proliferation, and survival. This presupposes signal transduction networking via mediatory regulators and molecular adaptors since T-cad lacks transmembrane and cytosolic domains. Here, using pharmacological inhibition of PI3K, adenoviral-mediated T-cad-overexpression, siRNA-mediated T-cad-depletion, and agonistic antibody-mediated ligation, we demonstrate signaling by T-cad through PI3K-Akt-GSK3beta pathways in EC. T-cad-overexpressing EC exhibited increased levels and nuclear accumulation of active beta-catenin, which was transcriptionally active as shown by increased Lef/Tcf reporter activity and cyclin D1 levels. Cotransduction of EC with constitutively active GSK3beta (S9A-GSK3beta) abrogated the stimulatory effects of T-cad on active beta-catenin accumulation, proliferation, and survival. Integrin-linked kinase (ILK), a membrane proximal upstream regulator of Akt and GSK3beta, was considered a candidate signaling mediator for T-cad. T-cad was present in anti-ILK immunoprecipitates, and confocal microscopy revealed colocalization of T-cad and ILK within lamellipodia of migrating cells. ILK-siRNA abolished T-cad-dependent effects on (Ser-473)Akt/(Ser-9)GSK3beta phosphorylation, active beta-catenin accumulation, and survival. We conclude ILK is an essential mediator for T-cad signaling via Akt and GSK3beta in EC. This is the first demonstration that ILK can regulate inward signaling by GPI-anchored proteins. Furthermore, ILK-GSK3beta-dependent modulation of active beta-catenin levels by GPI-anchored T-cad represents a novel mechanism for controlling cellular beta-catenin activity.
Collapse
Affiliation(s)
- Manjunath B Joshi
- Department of Research, Cardiovascular Laboratories, Basel University Hospital, Hebelstrasse 20, CH 4031 Basel, Switzerland
| | | | | | | | | |
Collapse
|
40
|
Haraguchi T, Okada K, Tabata Y, Maniwa Y, Hayashi Y, Okita Y. Controlled Release of Basic Fibroblast Growth Factor From Gelatin Hydrogel Sheet Improves Structural and Physiological Properties of Vein Graft in Rat. Arterioscler Thromb Vasc Biol 2007; 27:548-55. [PMID: 17170380 DOI: 10.1161/01.atv.0000254811.11741.2b] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objectives—
Autologous vein grafts are still widely used, but their long-term patency is suboptimal. The objective of the current study was to determine whether wrapping a vein graft in gelatin hydrogel sheet incorporating basic fibroblast growth factor improves their mechanical and physiological properties.
Methods and Results—
Autologous femoral vein was interposed into the abdominal aorta in rats. The rats were divided into 3 groups: nontreated grafts (group A), grafts wrapped in basic fibroblast growth factor-free gelatin hydrogel sheet (group B), and grafts wrapped in basic fibroblast growth factor-impregnated gelatin hydrogel sheet (group C). On day 1, endothelial desquamation was observed in group A, and the media in groups A and B were disrupted, staining positive in the terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling assay. In contrast, the media in group C remained intact and terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling–negative, associated with activation of MAPK. Graft dilation was significantly inhibited in groups B and C compared with group A, with those in group C showing the smallest degree of neointimal proliferation. At 8 weeks grafts in group C developed neointima with homogeneous elastic laminae, presence of rigid neoadventitia that displayed neovascularity, and the highest blood flow velocity.
Conclusions—
Wrapping vein grafts in basic fibroblast growth factor- impregnated gelatin hydrogel sheet improved their structural and physiological properties, and might therefore also improve long-term patency.
Collapse
MESH Headings
- Anastomosis, Surgical
- Animals
- Aorta, Abdominal/surgery
- Blood Flow Velocity
- Blotting, Western
- Delayed-Action Preparations
- Disease Models, Animal
- Endothelium, Vascular/pathology
- Femoral Vein/pathology
- Femoral Vein/transplantation
- Fibroblast Growth Factor 2/administration & dosage
- Fibroblast Growth Factor 2/metabolism
- Gelatin
- Graft Occlusion, Vascular/prevention & control
- Graft Rejection
- Graft Survival
- Hydrogels
- Immunohistochemistry
- In Situ Nick-End Labeling
- Neovascularization, Physiologic/drug effects
- Neovascularization, Physiologic/physiology
- Probability
- Rats
- Rats, Sprague-Dawley
- Reference Values
- Sensitivity and Specificity
- Transplantation, Autologous
- Vascular Patency
Collapse
Affiliation(s)
- Tomonori Haraguchi
- Division of Cardiovascular, Thoracic, and Pediatric Surgery, Kobe University Graduate School of Medicine, Kobe 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | | | | | | | | | | |
Collapse
|
41
|
Seeger FH, Zeiher AM, Dimmeler S. Cell-enhancement strategies for the treatment of ischemic heart disease. ACTA ACUST UNITED AC 2007; 4 Suppl 1:S110-3. [PMID: 17230207 DOI: 10.1038/ncpcardio0734] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2006] [Accepted: 10/09/2006] [Indexed: 01/11/2023]
Abstract
Cell therapy is a promising option for the treatment of ischemic diseases. Infusion or injection of stem or progenitor cells has improved neovascularization and heart function after ischemia in various experimental studies and clinical phase II and III trials. One potential limitation for cell therapy is a low rate of engraftment and persistence of cells in the ischemic tissue. Moreover, impairment of the number and function of patient-derived progenitor cells might limit the efficiency of autologous stem cell therapy. Therefore, strategies to augment cell function, survival, and homing could be crucial to improve success rates for cell therapy. Experimental studies have provided novel options for improving survival and function by transduction of stem or progenitor cells with prosurvival genes (e.g. Akt or telomerase). Pretreatment of cells with small molecules, such as statins, p38 inhibitors, or endothelial nitric oxide synthase enhancers, has been used to augment cell homing, integration, and functional recovery after induction of ischemia. Priming of the tissue by mechanical activation or application of growth factors might further improve recruitment and incorporation of cells. In this article we summarize the experimental studies providing novel concepts for cell-enhancement strategies to aid the treatment of peripheral artery occlusive and ischemic heart disease.
Collapse
Affiliation(s)
- Florian H Seeger
- Department of Molecular Cardiology/Cardiology, University of Frankfurt, Frankfurt, Germany
| | | | | |
Collapse
|
42
|
Abstract
Hypoxia is an integral characteristic of the tumor microenvironment, primarily due to the microvascular defects that accompany the accelerated neoplastic growth. The presence of tumor hypoxic areas correlates with negative outcome after radiotherapy, chemotherapy, and surgery, as hypoxia not only provides an environment directly facilitating chemo- and radio-resistance, but also encourages the evolution of phenotypic changes inducing permanent resistance to treatment and metastatic spread. Therefore, successful treatment of hypoxic cells has the potential to not only improve local control but also impact overall patient survival. Specific and selective targeting of hypoxic tumor areas can be achieved at all three steps of a gene therapy treatment: delivery of the therapeutic gene to the tumor, regulation of gene expression, and therapeutic efficacy. In this review the latest developments and innovations in hypoxia-targeted gene therapy are discussed. In particular, approaches such as hypoxia-conditionally replicating viruses, cellular vehicles, and gene therapy means to disrupt the hypoxia-inducible factor (HIF) signaling are outlined.
Collapse
Affiliation(s)
- Olga Greco
- Tumour Microcirculation Group, University of Sheffield, Royal Hallamshire Hospital, Sheffield, United Kingdom
| | | |
Collapse
|
43
|
Hahn JY, Cho HJ, Bae JW, Yuk HS, Kim KI, Park KW, Koo BK, Chae IH, Shin CS, Oh BH, Choi YS, Park YB, Kim HS. β-Catenin Overexpression Reduces Myocardial Infarct Size through Differential Effects on Cardiomyocytes and Cardiac Fibroblasts. J Biol Chem 2006; 281:30979-89. [PMID: 16920707 DOI: 10.1074/jbc.m603916200] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Beta-catenin is a transcriptional regulator of several genes involved in survival and proliferation. Although previous studies suggest that beta-catenin may be involved in the process of preconditioning and healing after myocardial infarction (MI), little is known regarding the role of beta-catenin in cardiomyocytes and cardiac fibroblasts. We investigated the role of beta-catenin in cardiomyocytes and cardiac fibroblasts and whether beta-catenin overexpression could reduce MI size. Adenovirus-mediated gene transfer of nonphosphorylatable constitutively active beta-catenin (Ad-catenin) decreased apoptosis in cardiomyocytes and cardiac fibroblasts with increased expression of survivin and Bcl-2. Although Ad-catenin increased the percentage of cells in the S phase with enhanced expression of cyclin D1 and E2 in both cell types, the increase in cell number was only evident in cardiac fibroblasts, whereas hypertrophy and binuclear cells were more prominent in cardiomyocytes. All of these effects of beta-catenin gene transfer were blocked by inhibition of its nuclear translocation. Furthermore, Ad-catenin enhanced the expression of vascular endothelial growth factor in both cells and induced differentiation of cardiac fibroblasts into myofibroblasts. In a rat MI model, injection of Ad-catenin into the infarct border zone resulted in a significantly decreased MI size with anti-apoptotic effect and cell cycle activation in both cardiomyocytes and myofibroblasts. beta-Catenin may play an important role in the healing process after MI by promoting survival and cell cycle not only in cardiomyocytes but also in cardiac fibroblasts with its differentiation into myofibroblasts.
Collapse
Affiliation(s)
- Joo-Yong Hahn
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 110-744, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Lee SP, Youn SW, Cho HJ, Li L, Kim TY, Yook HS, Chung JW, Hur J, Yoon CH, Park KW, Oh BH, Park YB, Kim HS. Integrin-linked kinase, a hypoxia-responsive molecule, controls postnatal vasculogenesis by recruitment of endothelial progenitor cells to ischemic tissue. Circulation 2006; 114:150-9. [PMID: 16818815 DOI: 10.1161/circulationaha.105.595918] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Recruitment and adhesion of endothelial progenitor cells (EPCs) to hypoxic endothelial cells (ECs) is essential for vasculogenesis in ischemic tissue; little is known, however, about the key signals or intracellular signaling pathways involved in orchestrating the expression of adhesion molecules by ECs in response to hypoxia and how this is related to the recruitment of EPCs to the ischemic tissue. Here, we report that endogenous integrin-linked kinase (ILK) is a novel molecule that responds to hypoxia in ECs that regulates the expression of stromal cell-derived factor-1 (SDF-1) and intercellular adhesion molecule-1 (ICAM-1) through nuclear factor-kappaB and hypoxia-inducible factor-1alpha and induces recruitment of EPCs to ischemic areas. METHODS AND RESULTS Under hypoxia, both the endogenous amount and kinase activity of ILK were time-dependently upregulated in ECs, which was associated with increased ICAM-1 and SDF-1. This upregulation of ILK was mediated by stabilization of ILK by heat shock protein 90. ILK overexpression in normoxic ECs resulted in ICAM-1 and SDF-1 upregulation through dual control by nuclear factor-kappaB and hypoxia-inducible factor-1alpha. Blockade of ILK in hypoxic ECs significantly abrogated the expression of both molecules, which led to decreased EPC incorporation into ECs. A hindlimb ischemia model showed that ILK blockade significantly reduced EPC homing to ischemic limb and consequently led to poor neovascularization. Overexpression of ILK in the Matrigel plug significantly improved neovascularization in vivo, whereas the blockade of ILK resulted in the opposite effect. CONCLUSIONS Endogenous ILK is a novel and physiological upstream responder of numerous intracellular molecules involved in hypoxic stress in ECs and may control the recruitment of EPCs to ischemic tissue.
Collapse
Affiliation(s)
- Seung-Pyo Lee
- Department of Internal Medicine, Seoul National University College of Medicine, 28 Yongon-dong, Chongro-gu, Seoul 110-744, South Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|