Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
1.
Am J Physiol Heart Circ Physiol ; 320(3): H1124-H1135, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33481698

RESUMEN

Critical limb ischemia (CLI) is a severe state of peripheral artery disease with high unmet clinical needs. Further, there are no effective treatment options for patients with CLI. Based on preclinical study results, predicting the clinical efficacy of CLI treatments is typically difficult because conventional hindlimb ischemia (HLI) rodent models display spontaneous recovery from ischemia, which is not observed in patients with CLI. Therefore, we aimed to develop a novel chronic and severe HLI model to properly evaluate the therapeutic effects of drug candidates for CLI. Severe HLI mice (Type-N) were generated by increasing the excised area of blood vessels in a hindlimb of NOG mice. Immunohistochemistry and gene expression analysis at 9 wk after the Type-N operation revealed that the ischemic limb was in a steady state with impaired angiogenesis, like that observed in patients with CLI. We did selection of chronic Type-N mice based on the number of necrotic nails and blood flow rate at 2 wk after surgery because some Type-N mice showed mild symptoms. Therapeutic treatment with cilostazol, which is used for intermittent claudication, did not restore blood flow in chronic Type-N mice. In contrast, therapeutic transplantation of pericytes and vascular endothelial cells, which can form new blood vessels in vivo, significantly improved blood flow in a subset of Type-N mice. These findings suggest that this novel chronic and severe HLI model may be a valuable standard animal model for therapeutic evaluation of the angiogenic effects of CLI drug candidates.NEW & NOTEWORTHY We developed a chronic and severe hindlimb ischemia (HLI) mouse model for preclinical research on critical limb ischemia (CLI). This model partially reflects human CLI pathology in that it does not show spontaneous restoration of blood flow or expression of angiogenic genes in the ischemic limb. This novel model may be valuable for therapeutic evaluation of the angiogenic effects of CLI drug candidates.


Asunto(s)
Inductores de la Angiogénesis/farmacología , Cilostazol/farmacología , Evaluación Preclínica de Medicamentos , Isquemia/tratamiento farmacológico , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica/efectos de los fármacos , Animales , Velocidad del Flujo Sanguíneo , Células Cultivadas , Enfermedad Crónica , Modelos Animales de Enfermedad , Miembro Posterior , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/trasplante , Humanos , Isquemia/metabolismo , Isquemia/fisiopatología , Masculino , Ratones Endogámicos NOD , Ratones SCID , Pericitos/metabolismo , Pericitos/trasplante , Flujo Sanguíneo Regional , Índice de Severidad de la Enfermedad
2.
Cancer Immunol Immunother ; 69(6): 1131-1140, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32047956

RESUMEN

Hemagglutinating virus of Japan (HVJ; Sendai virus) is an RNA virus that has cell fusion activity. HVJ-envelope (HVJ-E) is a UV-irradiated HVJ particle that loses viral replication and protein synthesis activity but retains cell fusion activity. We recently reported that HVJ-E has antitumor effects on several types of tumors. Here, we describe the results of a first-in-human phase I/IIa study in patients with advanced melanoma, receiving intratumoral administration of HVJ-E. The primary aim was to evaluate the safety and tolerability of HVJ-E, and the secondary aim was to examine the objective tumor response and antitumor immunity. Six patients with stage IIIC or IV progressive malignant melanoma with skin or lymph metastasis were enrolled. Patients were separated into two groups (n = 3 each) and received low and high doses of HVJ-E. Five of the six patients completed 4 weeks of follow-up evaluation; one patient discontinued treatment owing to progressive disease. Complete or partial responses were observed in 3 of 6 (50%) injected target lesions, 7 of 15 (47%) noninjected target lesions, and 10 of 21 (48%) target lesions. Induction of antitumor immunity was observed: activation of natural killer cells, a marked increase in interferon-γ levels in the peripheral blood, and infiltration of cytotoxic T cells into both injected and noninjected tumor lesions. Thus, intratumoral injection of HVJ-E in advanced melanoma patients showed safety and tolerability with local regression of the tumor mediated by antitumor immunity. The results suggest that HVJ-E might be a new treatment approach in patients with advanced melanoma.


Asunto(s)
Vectores Genéticos/genética , Melanoma/tratamiento farmacológico , Melanoma/inmunología , Viroterapia Oncolítica/métodos , Proteínas del Envoltorio Viral/genética , Línea Celular Tumoral , Humanos , Inyecciones Intralesiones
3.
BMC Biotechnol ; 18(1): 56, 2018 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-30200961

RESUMEN

BACKGROUND: Tissue-engineered cardiac constructs have potential in the functional recovery of heart failure; however, the preservation of these constructs is crucial for the development and widespread application of this treatment. We hypothesized that tissue-engineered skeletal myoblast (SMB) constructs may be preserved by vitrification to conserve biological function and structure. RESULTS: Scaffold-free cardiac cell-sheet constructs were prepared from SMBs and immersed in a vitrification solution containing ethylene glycol, sucrose, and carboxyl poly-L-lysine. The cell sheet was wrapped in a thin film and frozen rapidly above liquid nitrogen to achieve vitrification (vitrification group, n = 8); fresh, untreated SMB sheets (fresh group, n = 8) were used as the control. The cryopreserved SMB sheets were thawed at 2 days, 1 week, 1 month, and 3 months after cryopreservation for assessment. Thawed, cryopreserved SMB sheets were transplanted into rat hearts in a myocardial infarction nude rat model, and their effects on cardiac function were evaluated. Cell viability in the cardiac constructs of the vitrification group was comparable to that of the fresh group, independent of the period of cryopreservation (p > 0.05). The structures of the cell-sheet constructs, including cell-cell junctions such as desmosomes, extracellular matrix, and cell membranes, were maintained in the vitrification group for 3 months. The expression of cytokine genes and extracellular matrix proteins (fibronectin, collagen I, N-cadherin, and integrin α5) showed similar levels in the vitrification and fresh groups. Moreover, in an in vivo experiment, the ejection fraction was significantly improved in animals treated with the fresh or cryopreserved constructs as compared to that in the sham-treated group (p < 0.05). CONCLUSIONS: Overall, these results show that the vitrification method proposed here preserves the functionality and structure of scaffold-free cardiac cell-sheet constructs using human SMBs after thawing, suggesting the potential clinical application of this method in cell-sheet therapy.


Asunto(s)
Criopreservación/métodos , Mioblastos/citología , Infarto del Miocardio/terapia , Adulto , Anciano , Animales , Supervivencia Celular , Criopreservación/instrumentación , Matriz Extracelular/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Ratas , Ratas Endogámicas F344 , Ratas Desnudas , Regeneración , Ingeniería de Tejidos , Vitrificación
4.
Surg Today ; 48(2): 200-210, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28821963

RESUMEN

BACKGROUND AND PURPOSE: Despite promising experimental results, clinically, intramyocardial myoblast injection failed to reverse remodeling and it induced arrhythmogenicity. In contrast, scaffold-free skeletal muscle-derived cell (SC) sheets attenuated cardiac dysfunction and arrhythmogenicity via paracrine effects. We report the first clinical trial of SC sheet implantation (SCSI) conducted in four patients with dilated cardiomyopathy (DCM) supported by a left ventricular assist device (LVAD). METHODS: SC sheets were made from muscle fibers and multi-layered SC sheets were applied to the left ventricular (LV) anterolateral surface via left thoracotomy. RESULTS: There were no major cardiac adverse events. Ventricular arrhythmia decreased in all except one patient, in whom global LV function did not improve. The LV volume decreased and LV ejection fraction improved in all except the same patient. Systolic wall thickening, reflecting regional wall motion, improved in the sheet-implanted areas, and vessels in the LV apex increased in all patients, suggesting angiogenesis. The LVAD was successfully removed in two patients. CONCLUSIONS: SCSI induced reverse remodeling and angiogenesis, and improved LV function, allowing LVAD removal in two patients, although functional recovery failed to improve in the one non-responder, even with angiogenesis. SCSI is a promising regenerative therapy for DCM patients responsive to this strategy, even with LVAD assistance.


Asunto(s)
Cardiomiopatía Dilatada/terapia , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Ventrículos Cardíacos/cirugía , Corazón Auxiliar , Músculo Esquelético/citología , Adulto , Cardiomiopatía Dilatada/fisiopatología , Estudios de Factibilidad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Toracotomía/métodos , Andamios del Tejido , Resultado del Tratamiento , Función Ventricular Izquierda , Remodelación Ventricular , Adulto Joven
5.
Mol Ther ; 23(2): 374-86, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25421595

RESUMEN

Cell-sheet transplantation induces angiogenesis for chronic myocardial infarction (MI), though insufficient capillary maturation and paucity of arteriogenesis may limit its therapeutic effects. Omentum has been used clinically to promote revascularization and healing of ischemic tissues. We hypothesized that cell-sheet transplantation covered with an omentum-flap would effectively establish mature blood vessels and improve coronary microcirculation physiology, enhancing the therapeutic effects of cell-sheet therapy. Rats were divided into four groups after coronary ligation; skeletal myoblast cell-sheet plus omentum-flap (combined), cell-sheet only, omentum-flap only, and sham operation. At 4 weeks after the treatment, the combined group showed attenuated cardiac hypertrophy and fibrosis, and a greater amount of functionally (CD31(+)/lectin(+)) and structurally (CD31(+)/α-SMA(+)) mature blood vessels, along with myocardial upregulation of relevant genes. Synchrotron-based microangiography revealed that the combined procedure increased vascularization in resistance arterial vessels with better dilatory responses to endothelium-dependent agents. Serial (13)N-ammonia PET showed better global coronary flow reserve in the combined group, mainly attributed to improvement in the basal left ventricle. Consequently, the combined group had sustained improvements in cardiac function parameters and better functional capacity. Cell-sheet transplantation with an omentum-flap better promoted arteriogenesis and improved coronary microcirculation physiology in ischemic myocardium, leading to potent functional recovery in the failing heart.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Circulación Coronaria , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/terapia , Neovascularización Fisiológica , Epiplón , Animales , Movimiento Celular , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Expresión Génica , Supervivencia de Injerto , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Hemodinámica , Infarto del Miocardio/complicaciones , Miocardio/metabolismo , Miocardio/patología , Ratas , Flujo Sanguíneo Regional , Trasplantes , Remodelación Vascular , Función Ventricular Izquierda
6.
Mol Cell Biochem ; 408(1-2): 191-203, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26112906

RESUMEN

Osteopontin (OPN) is involved in various physiological processes such as inflammatory and wound healing. However, little is known about the effects of OPN on these tissues. OPN is cleaved by thrombin, and cleavage of the N-terminal fragment exposes a SVVYGLR sequence on its C-terminus. In this study, we examined the effects of the thrombin-cleaved OPN fragments on fibroblasts and myocardial fibrosis, particularly the role of the SVVYGLR sequence. The recombinant thrombin-cleaved OPN fragments (N-terminal fragment [N-OPN], C-terminal fragment [C-OPN], and the N-terminal fragment lacking the SVVYGLR sequence [ΔSV N-OPN]) were added to fibroblasts, and the cellular motility, signal activity, and production of collagen were evaluated. A sustained-release gel containing an OPN fragment or SVVYGLR peptide was transplanted into a rat model of ischemic cardiomyopathy and the quantities and ratio of collagen type I (COL I) and type III (COL III) were estimated. N-OPN significantly promoted fibroblast migration. Smad signal activity, expression of smooth muscle actin (SMA), and the production of COL III were enhanced by N-OPN and SVVYGLR peptide. Conversely, ΔSV N-OPN and C-OPN had no effect. In vivo, the expression level of N-OPN was associated with COL III distribution, and the COL III/COL I ratio was significantly increased by the sustained-release gel containing N-OPN or SVVYGLR peptide. The cardiac function was also significantly improved by the N-OPN- or SVVYGLR peptide-released gel treatment. The N-terminal fragment of thrombin-cleaved OPN-induced Smad signal activation, SMA expression, and COL III production, and its SVVYGLR sequence influences this function.


Asunto(s)
Cardiomiopatías/tratamiento farmacológico , Colágeno Tipo III/metabolismo , Fibroblastos/efectos de los fármacos , Miocardio/patología , Oligopéptidos/administración & dosificación , Piel/citología , Animales , Cardiomiopatías/fisiopatología , Línea Celular , Movimiento Celular/efectos de los fármacos , Preparaciones de Acción Retardada , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Miocardio/citología , Miocardio/metabolismo , Oligopéptidos/farmacología , Osteopontina/química , Osteopontina/metabolismo , Estructura Terciaria de Proteína , Ratas , Transducción de Señal/efectos de los fármacos , Piel/metabolismo , Trombina/metabolismo
7.
Circ J ; 79(1): 169-79, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25373489

RESUMEN

BACKGROUND: Clinical prognosis is critically poor in fulminant myocarditis, while it's initiation or progression is fated, in part, by T cell-mediated autoimmunity. Adiponectin (APN) and associated adipokines were shown to be immune tolerance inducers, although the clinically relevant delivery method into target pathologies is under debate. Whether the cell sheet-based delivery system of adipokines might induce immune tolerance and functional recovery in experimental autoimmune myocarditis (EAM) was tested. METHODS AND RESULTS: Scaffold-free-induced adipocyte cell-sheet (iACS) was generated by differentiating adipose tissue-derived syngeneic stromal vascular-fraction cells into adipocytes on temperature-responsive dishes. Rats with EAM underwent iACS implantation or sham operation. Supernatants of iACS contained a high level of APN and hepatocyte growth factor (HGF), and reduced proliferation of CD4-positive T cells in vitro. Immunohistolabelling showed that the iACS implantation elevated the levels of APN and HGF in the myocardium compared to the sham operation, which attenuated the immunological response by inhibiting CD68-positive macropharges and CD4-positive T-cells and activating Foxp3-positive regulatory T cells. Consequently, left ventricular ejection fraction was significantly greater after the iACS implantation than after the sham operation, in association with less collagen accumulation. CONCLUSIONS: The targeted delivery of adipokines using tissue-engineered iACS ameliorated cardiac performance of the EAM rat model via effector T cell suppression and induction of immune tolerance. These findings might suggest a potential of this tissue-engineered drug delivery system in treating fulminant myocarditis in the clinical setting.


Asunto(s)
Adipocitos/trasplante , Adipoquinas/administración & dosificación , Enfermedades Autoinmunes/terapia , Sistemas de Liberación de Medicamentos , Isoinjertos/metabolismo , Miocarditis/terapia , Adipocitos/metabolismo , Adipoquinas/inmunología , Adipoquinas/metabolismo , Adipoquinas/uso terapéutico , Adiponectina/administración & dosificación , Adiponectina/metabolismo , Adiponectina/farmacología , Animales , Enfermedades Autoinmunes/inmunología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Células Cultivadas , Colágeno/análisis , Medios de Cultivo Condicionados/farmacología , Corazón , Factor de Crecimiento de Hepatocito/farmacología , Tolerancia Inmunológica , Activación de Linfocitos/efectos de los fármacos , Masculino , Miocarditis/inmunología , Miocarditis/fisiopatología , Miocardio/química , Miocardio/patología , Distribución Aleatoria , Ratas , Ratas Endogámicas Lew , Proteínas Recombinantes/farmacología , Volumen Sistólico , Ingeniería de Tejidos , Trasplante Heterotópico , Trasplante Isogénico , Factor A de Crecimiento Endotelial Vascular/farmacología
8.
Circulation ; 128(11 Suppl 1): S87-94, 2013 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-24030425

RESUMEN

BACKGROUND: Transplantation of cardiomyocytes that are derived from human induced pluripotent stem cell-derived cardiomyocytes (hiPS-CMs) shows promise in generating new functional myocardium in situ, whereas the survival and functionality of the transplanted cells are critical in considering this therapeutic impact. Cell-sheet method has been used to transplant many functional cells; however, potential ischemia might limit cell survival. The omentum, which is known to have rich vasculature, is expected to be a source of blood supply. We hypothesized that transplantation of hiPS-CM cell sheets combined with an omentum flap may deliver a large number of functional hiPS-CMs with enhanced blood supply. METHODS AND RESULTS: Retrovirally established human iPS cells were treated with Wnt signaling molecules to induce cardiomyogenic differentiation, followed by superparamagnetic iron oxide labeling. Cell sheets were created from the magnetically labeled hiPS-CMs using temperature-responsive dishes and transplanted to porcine hearts with or without the omentum flap (n=8 each). Two months after transplantation, the survival of superparamagnetic iron oxide-labeled hiPS-CMs, assessed by MRI, was significantly greater in mini-pigs with the omentum than in those without it; histologically, vascular density in the transplanted area was significantly greater in mini-pigs with the omentum than in those without it. The transplanted tissues contained abundant cardiac troponin T-positive cells surrounded by vascular-rich structures. CONCLUSIONS: The omentum flap enhanced the survival of hiPS-CMs after transplantation via increased angiogenesis, suggesting that this strategy is useful in clinical settings. The combination of hiPS-CMs and the omentum flap may be a promising technique for the development of tissue-engineered vascular-rich new myocardium in vivo.


Asunto(s)
Supervivencia Celular/fisiología , Células Madre Pluripotentes Inducidas/trasplante , Trasplante de Células Madre Mesenquimatosas/métodos , Miocitos Cardíacos/trasplante , Colgajos Quirúrgicos/fisiología , Regulación hacia Arriba/fisiología , Animales , Femenino , Corazón/fisiología , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Miocitos Cardíacos/fisiología , Distribución Aleatoria , Porcinos , Porcinos Enanos
9.
Circ J ; 78(1): 222-31, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24107361

RESUMEN

BACKGROUND: Because human cardiac stem cells (CSC) have regeneration potential in damaged cardiac tissue, there is increasing interest in using them in cell-based therapies for cardiac failure. However, culture conditions, by which CSCs are expanded while maintaining their therapeutic potential, have not been optimized. We hypothesized that the plating cell-density would affect proliferation activity, differentiation and therapeutic potential of CSCs through the Notch signaling pathway. METHODS AND RESULTS: Human CSCs were plated at 4 different densities. The population doubling time, C-KIT positivity, and dexamethasone-induced multidifferentiation potential were examined in vitro. The therapeutic potential of CSCs was assessed by transplanting them into a rat acute myocardial infarction (AMI) model. The low plating density (340cells/cm(2)) maintained the multidifferentiation potential with greater proliferation activity and C-KIT positivity in vitro. On the other hand, the high plating density (5,500cells/cm(2)) induced autonomous differentiation into endothelial cells by activating Notch signaling in vitro. CSCs cultured at low or high density with Notch signal inhibitor showed significantly greater therapeutic potential in vivo compared with those cultured at high density. CONCLUSIONS: CSCs cultured with reduced Notch signaling showed better cardiomyogenic differentiation and therapeutic potentials in a rat AMI model. Thus, reducing Notch signaling is important when culturing CSCs for clinical applications.


Asunto(s)
Infarto del Miocardio , Miocardio , Receptores Notch/metabolismo , Transducción de Señal , Trasplante de Células Madre , Células Madre , Adulto , Animales , Células Cultivadas , Niño , Femenino , Xenoinjertos , Humanos , Masculino , Persona de Mediana Edad , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/terapia , Miocardio/metabolismo , Miocardio/patología , Ratas , Ratas Desnudas , Células Madre/metabolismo , Células Madre/patología
10.
Artículo en Inglés | MEDLINE | ID: mdl-38376701

RESUMEN

Critical limb ischemia (CLI) is a state of severe peripheral artery disease, with no effective treatment. Cell therapy has been investigated as a therapeutic tool for CLI, and pericytes are promising therapeutic candidates based on their angiogenic properties. We firstly generated highly proliferative and immunosuppressive pericyte-like cells from embryonic stem (ES) cells. In order to enhance the angiogenic potential, we transduced the basic fibroblast growth factor (bFGF) gene into the pericyte-like cells and found a significant enhancement of angiogenesis in a Matrigel plug assay. Furthermore, we evaluated the bFGF-expressing pericyte-like cells in the previously established chronic hindlimb ischemia model in which bone marrow-derived MSCs were not effective. As a result, bFGF-expressing pericyte-like cells significantly improved blood flow in both laser Doppler perfusion imaging (LDPI) and dynamic contrast-enhanced MRI (DCE-MRI). These findings suggest that bFGF-expressing pericyte-like cells differentiated from ES cells may be a therapeutic candidate for CLI.

11.
Circulation ; 126(11 Suppl 1): S29-37, 2012 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-22965990

RESUMEN

BACKGROUND: Human induced pluripotent stem cell-derived cardiomyocytes (hiPS-CMs) are a promising source of cells for regenerating myocardium. However, several issues, especially the large-scale preparation of hiPS-CMs and elimination of undifferentiated iPS cells, must be resolved before hiPS cells can be used clinically. The cell-sheet technique is one of the useful methods for transplanting large numbers of cells. We hypothesized that hiPS-CM-sheet transplantation would be feasible, safe, and therapeutically effective for the treatment of ischemic cardiomyopathy. METHODS AND RESULTS: Human iPS cells were established by infecting human dermal fibroblasts with a retrovirus carrying Oct3/4, Sox2, Klf4, and c-Myc. Cardiomyogenic differentiation was induced by WNT signaling molecules, yielding hiPS-CMs that were almost 90% positive for α-actinin, Nkx2.5, and cardiac troponin T. hiPS-CM sheets were created using thermoresponsive dishes and transplanted over the myocardial infarcts in a porcine model of ischemic cardiomyopathy induced by ameroid constriction of the left anterior descending coronary artery (n=6 for the iPS group receiving sheet transplantation and the sham-operated group; both groups received tacrolimus daily). Transplantation significantly improved cardiac performance and attenuated left ventricular remodeling. hiPS-CMs were detectable 8 weeks after transplantation, but very few survived long term. No teratoma formation was observed in animals that received hiPS-CM sheets. CONCLUSIONS: The culture system used yields a large number of highly pure hiPS-CMs, and hiPS-CM sheets could improve cardiac function after ischemic cardiomyopathy. This newly developed culture system and the hiPS-CM sheets may provide a basis for the clinical use of hiPS cells in cardiac regeneration therapy.


Asunto(s)
Técnicas de Cultivo de Célula , Células Madre Pluripotentes Inducidas/trasplante , Infarto del Miocardio/cirugía , Miocitos Cardíacos/trasplante , Ingeniería de Tejidos/métodos , Animales , Técnicas de Cultivo de Célula/instrumentación , Diferenciación Celular/efectos de los fármacos , Electrocardiografía Ambulatoria , Estudios de Factibilidad , Femenino , Supervivencia de Injerto , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Factor 4 Similar a Kruppel , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/patología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/fisiología , Recuperación de la Función , Retroviridae/genética , Porcinos , Porcinos Enanos , Temperatura , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Trasplante Heterólogo , Ultrasonografía , Proteínas Wnt/farmacología
12.
Circ J ; 77(5): 1297-306, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23392122

RESUMEN

BACKGROUND: Induced pluripotent stem cells (iPSCs) hold promise for a new era in treating heart failure. However, the functional microstructure of iPSC-derived cardiomyocytes (iPSC-CMs) and their ability to attach to the extracellular matrix of the recipient myocardium require further elucidation. Thus, we analyzed the functional microstructure and adhesion molecules of iPSC-CM. METHODS AND RESULTS: Immunostaining analysis showed that iPSC-CMs were similar to neonatal cardiomyocytes (CMs) in expressing the cytoskeletal proteins myosin heavy chain (MHC), myosin light chain (MLC) 2a, MLC2v, and especially ß-MHC (a neonatal CM marker), as well as the adhesion molecules N-cadherin, α7-integrin, dystrophin, α-dystroglycan, α-sarcoglycan, and laminin-α2. Electron microscopy showed abundant myofibrillar bundles with transverse Z-bands and a developed mitochondrial structure in both iPSC-CMs and neonatal CMs, although the iPSC-CMs contained fewer mitochondria with lower-density cristae. When transplanted from in vitro conditions to nude rat hearts, iPSC-CMs acquired the ability to express α-MHC, a molecule specific to adult CMs. Mechanical stretch or stimulation by insulin-like growth factor-1 enhanced the α-MHC expression in iPSC-CMs in vitro. CONCLUSIONS: Our findings in vitro and in vivo indicate that CMs derived from iPSCs contain cardiac-specific organelles and adhesion systems. These results indicate that iPSC-derived CMs may be useful in new cell therapies for heart failure.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Células Madre Pluripotentes Inducidas/fisiología , Células Madre Pluripotentes Inducidas/trasplante , Miocitos Cardíacos/fisiología , Miocitos Cardíacos/trasplante , Animales , Animales Recién Nacidos , Biomarcadores/metabolismo , Moléculas de Adhesión Celular/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular , Femenino , Inmunohistoquímica , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/ultraestructura , Factor I del Crecimiento Similar a la Insulina/farmacología , Mecanotransducción Celular , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Contracción Miocárdica , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/ultraestructura , Cadenas Pesadas de Miosina/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Fenotipo , Ratas , Ratas Endogámicas F344 , Ratas Desnudas , Estrés Mecánico , Factores de Tiempo
13.
Circ J ; 77(4): 1063-72, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23291995

RESUMEN

BACKGROUND: The implantation of skeletal myoblast (SMB) cell-sheets over the damaged area of a myocardial infarction (MI) has been shown to improve global left ventricular (LV) function through a paracrine effect. However, the regeneration process has not been fully evaluated. We hypothesized that the use of tissue Doppler strain M-mode imaging to assess myocardial layer-specific strain might enable detailed visual evaluation of the regenerative ability of SMBs. METHODS AND RESULTS: SMBs were cultured on temperature-responsive culture dishes to generate cell-sheets. At 4 weeks after inducing anterior MI, the animals were divided into 2 groups: SMB cell-sheet implantation and sham operation (n=6 in each). A total of 30 cell-sheets (1.5×10(7) cells/sheet) were placed on the epicardium, covering the infarct and border regions. Subendocardial and subepicardial strain values were measured in the infarct, border, and remote regions by tissue Doppler strain analysis. SMB cell-sheet implantation produced the following major effects: progression of LV remodeling was prevented and global LV ejection fraction increased; the subendocardial strain was significantly greater than the subepicardial strain in the treated border region; vascular density in the subendocardium was significantly higher than in the subepicardium in the treated region; the expression of vascular endothelial growth factor was significantly increased. CONCLUSIONS: Tissue Doppler strain analysis allows precise evaluation of the effect of cell-sheet implantation on layer-specific myocardial function.


Asunto(s)
Mioblastos Esqueléticos/trasplante , Infarto del Miocardio , Función Ventricular Izquierda , Animales , Femenino , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/terapia , Pericardio/diagnóstico por imagen , Pericardio/fisiopatología , Porcinos , Porcinos Enanos , Ultrasonografía
14.
Surg Today ; 43(9): 970-6, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23459789

RESUMEN

PURPOSE: Myocardial infarction (MI) remains a major cause of mortality because of the limited regenerative capacity of the myocardium. Transplantation of somatic tissue-derived cells into the heart has been shown to enhance the endogenous healing process, but the magnitude of its therapeutic effects is dependent upon the cell-source or cell-delivery method. We investigated the therapeutic effects of C-Kit positive cardiac cell (CSC) cell-sheet transplantation therapy in a rat model of MI. METHODS AND RESULTS: CSCs of human origin were sorted and cultured to generate scaffold-free CSC cell-sheets. One-layered or 3-layered cell-sheets were transplanted into nude rats 1 h after left coronary artery ligation. We observed a significant increase in the left ventricular ejection fraction and a significant decrease in left ventricular systolic dimension at 2 and 4 weeks in the 3-layer group, but not in the 1-layer or sham groups. Consistently, there was less accumulation of interstitial fibrosis in the 3-layer group than in the 1-layer or sham groups. Moreover, capillary density was significantly greater in the 3-layer group than in the 1-layer or sham groups. CONCLUSIONS: The 3-layered cell-sheet improved cardiac function associated with angiogenic and anti-fibrotic effects. Thus, CSC is a promising cell-source to use with the cell-sheet method for the treatment of cardiac failure, as long as a sufficient number of cells are delivered.


Asunto(s)
Infarto del Miocardio/patología , Infarto del Miocardio/terapia , Miocardio/citología , Trasplante de Células Madre/métodos , Anciano , Animales , Diferenciación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Fibrosis , Humanos , Ratones , Ratones Desnudos , Persona de Mediana Edad , Miocardio/patología , Neovascularización Fisiológica , Ratas
15.
Mol Cell Biochem ; 368(1-2): 203-14, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22718481

RESUMEN

Myoblast sheet transplantation for cardiac failure is a promising therapy to enhance cardiac function via paracrine mechanism. However, their efficacies of treatment showed a gradual decline. The gene modification of the implanted myoblast is important in improving the long-term results of the treatment. Elastin fiber enhances the extensibility of the infarcted wall and can prevent left ventricular dilation. We therefore hypothesized that the elastin gene modification of the implanted myoblast could strengthen and maintain the long-term improvement effects of cardiac function. In this study, we evaluated long-term follow-up benefits of functional myoblast sheets that secrete elastin in an infarcted model. The animal models were divided into three groups: a group transplanted with nontransfected, wild-type, skeletal myoblast-type sheets (WT-rSkM); group transplanted with myoblast sheets that secreted elastin fragments (ELN-rSkM); and a control group (ligation only). Cardiac function was examined by echocardiography, and cardiac remodeling after infarction was evaluated by histological examination. The cardiac function was significantly improved and the left ventricle end-diastolic dimensions were significantly reduced in the ELN-rSkM group. Histological analysis showed that left ventricular remodeling was attenuated in the ELN-rSkM group and that elastic fiber was formed in the epicardial area of ELN-rSkM group. The functionalization of myoblast sheet by elastin gene transfer showed the long-term improvement of cardiac function. Expressed recombinant elastin fiber prevented the dilation of the left ventricular chamber after myocardial infarction. The functional myoblast sheet transplantation maintained the treatment effect by the paracrine effect of myoblast and the formed recombinant elastin.


Asunto(s)
Elastina/biosíntesis , Mioblastos Esqueléticos/trasplante , Infarto del Miocardio/metabolismo , Infarto del Miocardio/terapia , Miocardio , Recuperación de la Función , Animales , Ecocardiografía , Elastina/genética , Femenino , Mioblastos Esqueléticos/metabolismo , Infarto del Miocardio/diagnóstico por imagen , Ratas , Ratas Endogámicas Lew , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Trasplante Homólogo
16.
Surg Today ; 42(2): 181-4, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22200756

RESUMEN

Dilated cardiomyopathy (DCM) is a heart muscle disease characterized by progressive heart failure, and is a leading cause of mortality and morbidity. Recently, cellular therapy for end-stage heart failure has been emerging. We herein report a 56-year-old male who received a transplant of autologous myoblast sheets manufactured in temperature-responsive culture dishes. His clinical condition improved markedly, leaving him without any arrhythmia and able to discontinue using a left ventricular assist system and avoid cardiac transplantation. These findings suggest that cellular therapy using myoblast sheets is a promising new strategy for treating patients with end-stage DCM. This method might be an effective alternative to heart transplantation in the near future.


Asunto(s)
Cardiomiopatía Dilatada/terapia , Trasplante de Células/instrumentación , Corazón Auxiliar , Mioblastos Esqueléticos/trasplante , Volumen Sistólico/fisiología , Ingeniería de Tejidos , Función Ventricular/fisiología , Cardiomiopatía Dilatada/fisiopatología , Células Cultivadas , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Trasplante Autólogo
17.
Regen Ther ; 21: 19-24, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35619946

RESUMEN

In recent years, extracellular vesicles (EVs) have attracted attention as a new therapeutic tool. In Europe, the United States, and Asia, there is an accelerating trend of moving beyond basic research on clinical trials. However, treatment using EVs is still in the research and development stage, and the general public has insufficient awareness and understanding of the risks involved in ensuring safety and efficacy, the status of laws and regulations, and global research and development trends regarding their use. The Japanese Society for Regenerative Medicine, which has promoted the research and development of regenerative medicine, an innovative medical technology based on the principle of delivering it safely, effectively, and promptly, including the establishment of laws and regulations, would like to express two positions in light of the rapid development of therapies using EVs: 1) concern about treatments that are based solely on the discretion of medical practitioners, and 2) active promotion of treatments based on sound scientific evidence. Because EVs are released from cells, there are many similarities between EVs and processed cells in terms of manufacturing processes and safety hazards. As for efficacy, the mechanism of action of EVs is still unclear, as is the case with specified processed cellsb; in such cases, it is difficult to measure potency, identify efficacy-related quality attributes, and evaluate the comparability of quality before and after a change in the manufacturing process. In other words, the number of quality attributes that can be obtained for EVs is limited because of their complex characteristics, and it is difficult to grasp their quality through specifications and characterization. Therefore, while designing a quality control strategy for EVs, it is important to ensure the quality of the final product (EVs) by controlling the raw materials and manufacturing process. On the contrary, since EVs do not contain living cell components and are not classified into specified processed cells, non-commercial clinical research on treatments using EVs and individual medical treatments with EVs at the discretion of medical practitioners are out of the scope of the Act on the Safety of Regenerative Medicine of Japan. At present, there are no relevant laws or regulations for the use of EVs other than the Medical Practitioners' Act and the Medical Care Act in Japan. Therefore, there is a concern that treatment will be performed without sufficient objective evaluation of the scientific basis for safety and efficacy. Despite these concerns, the development of therapies using EVs is underway worldwide. This could potentially lead to a wide variety of new therapeutic areas if the methods needed to stably secure and mass cultivate cells as raw materials and the technologies needed for the mass production of EVs can be developed, in addition to understanding the risks involved and developing relevant laws and regulations. As part of the Japanese Society for Regenerative Medicine, we will continue to work on the development of these methods and technologies and hope that such a promising field will be promoted with a high level of safety before reaching the public.

18.
Circulation ; 122(11 Suppl): S118-23, 2010 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-20837902

RESUMEN

BACKGROUND: The safety and efficacy of myocardial regeneration using embryonic stem cells are limited by the risk of teratoma and the high rate of cell death. METHODS AND RESULTS: To address these issues, we developed a composite construct made of a sheet of adipose tissue-derived stroma cells and embryonic stem cell-derived cardiac progenitors. Ten Rhesus monkeys underwent a transient coronary artery occlusion followed, 2 weeks later, by the open-chest delivery of the composite cell sheet over the infarcted area or a sham operation. The sheet was made of adipose tissue-derived stroma cells grown from a biopsy of autologous adipose tissue and cultured onto temperature-responsive dishes. Allogeneic Rhesus embryonic stem cells were committed to a cardiac lineage and immunomagnetically sorted to yield SSEA-1(+) cardiac progenitors, which were then deposited onto the cell sheet. Cyclosporine was given for 2 months until the animals were euthanized. Preimplantation studies showed that the SSEA-1(+) progenitors expressed cardiac markers and had lost pluripotency. After 2 months, there was no teratoma in any of the 5 cell-treated monkeys. Analysis of >1500 histological sections showed that the SSEA-1(+) cardiac progenitors had differentiated into cardiomyocytes, as evidenced by immunofluorescence and real-time polymerase chain reaction. There were also a robust engraftment of autologous adipose tissue-derived stroma cells and increased angiogenesis compared with the sham animals. CONCLUSIONS: These data collected in a clinically relevant nonhuman primate model show that developmentally restricted SSEA-1(+) cardiac progenitors appear to be safe and highlight the benefit of the epicardial delivery of a construct harboring cells with a cardiomyogenic differentiation potential and cells providing them the necessary trophic support.


Asunto(s)
Tejido Adiposo/citología , Células Madre Embrionarias/trasplante , Infarto del Miocardio/terapia , Miocardio/patología , Regeneración , Trasplante de Células Madre/métodos , Tejido Adiposo/trasplante , Animales , Diferenciación Celular , Modelos Animales de Enfermedad , Humanos , Antígeno Lewis X , Macaca mulatta , Ratones , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Neovascularización Fisiológica , Células del Estroma , Trasplante Autólogo , Trasplante Homólogo
19.
Surg Today ; 41(8): 1029-36, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21773889

RESUMEN

PURPOSE: Simultaneous injection of autologous bone marrow cells and skeletal myoblasts has been demonstrated to improve cardiac function in animal models. We evaluated the potential application of this combination cell therapy in patients with severe ischemic cardiomyopathy who required left ventricular assist device (LVAD) implantation. METHODS: Four patients (age range, 43-69 years) who required LVAD implantation due to severe ischemic cardiomyopathy were studied. Skeletal myoblasts were obtained from the thigh, while bone marrow mononuclear cells were collected and purified at the time of the operation. These cells were directly injected in a serial manner into the damaged myocardium. RESULTS: No fatal arrhythmias or major complications were observed. The number of injected skeletal myoblasts ranged from 2.7 × 10(7) to 3.0 × 10(8), and their purity ranged from 25% to 96%. Two patients showed decreased brain natriuretic peptide levels and echocardiographic improvements in the transplanted areas, as well as increased perfusion revealed by H(2) (15)O positron emission tomography, of whom one was successfully weaned from LVAD. Histological findings at autopsy of the other patient showed a small amount of skeletal muscle in the injected area. Only marginal improvements were observed in the other two patients. CONCLUSIONS: Combined cell transplantation is feasible for patients with severe ischemic cardiomyopathy, and functional recovery is anticipated in selected patients.


Asunto(s)
Trasplante de Médula Ósea , Insuficiencia Cardíaca/terapia , Mioblastos Esqueléticos/trasplante , Isquemia Miocárdica/terapia , Adulto , Anciano , Estudios de Cohortes , Estudios de Factibilidad , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/mortalidad , Corazón Auxiliar , Humanos , Persona de Mediana Edad , Isquemia Miocárdica/mortalidad , Isquemia Miocárdica/patología , Tasa de Supervivencia , Trasplante Autólogo , Resultado del Tratamiento
20.
Stem Cell Res Ther ; 11(1): 69, 2020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-32070429

RESUMEN

BACKGROUND: Transplantation of skeletal myoblast sheets is a promising strategy for the treatment of heart failure, and its therapeutic effects have already been proven in both animal disease models and clinical trials. Myoblast sheets reportedly demonstrate their therapeutic effects by producing many paracrine factors. Although the quality of processed cells for transplantation can be evaluated by the positive ratio of CD56, a myoblast marker, it is unclear which cell populations from isolated cells produce paracrine factors that have an impact on therapeutic effects, and whether these therapeutic effects are closely correlated with CD56-positive cells isolated from the skeletal muscle is also unclear. Therefore, we hypothesized that CD56-negative cells as well as CD56-positive cells isolated from the skeletal muscle produce paracrine factors and have therapeutic effects in skeletal muscle-derived cell sheet therapy for heart failure. METHODS: Cell surface and intracellular markers of CD56-negative non-myogenic cells (NMCs) and CD56-positive myoblasts were evaluated. We also analyzed cytokine expression, tube formation ability, and stem cell mobilization in both cell populations. Finally, we assessed the therapeutic effects of the cell populations in a rat myocardial infarction model. RESULTS: Analysis of cell surface and intracellular markers revealed that CD56-negative NMCs expressed fibroblast markers and a higher level of mesenchymal cell markers, such as CD49b and CD140a, than myoblasts. Both NMCs and myoblasts expressed various cytokines in vitro with different expression patterns. In addition, NMCs induced tube formation (control vs. myoblasts vs. NMCs: 100 ± 11.2 vs. 142 ± 8.3 vs. 198 ± 7.4%) and stem cell mobilization (control vs. myoblasts vs. NMCs: 100 ± 6.8 vs. 210 ± 22.9 vs. 351 ± 36.0%) to a higher degree in vitro than did myoblasts. The effect of NMCs and myoblasts on the improvement of cardiac function and suppression of myocardial fibrosis in rat myocardial infarction model was comparable. CONCLUSION: These results indicate that NMCs exhibit therapeutic effects in skeletal muscle-derived cell sheet therapy for heart failure. Thus, accurate parameters correlating with therapeutic effects need to be further explored.


Asunto(s)
Ecocardiografía/métodos , Insuficiencia Cardíaca/terapia , Músculo Esquelético/metabolismo , Infarto del Miocardio/terapia , Animales , Diferenciación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Insuficiencia Cardíaca/patología , Masculino , Músculo Esquelético/citología , Infarto del Miocardio/patología , Ratas
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda