Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
BMC Med ; 21(1): 386, 2023 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-37798633

RESUMEN

BACKGROUND: We previously demonstrated that CD34 + cell transplantation in animals healed intractable fractures via osteogenesis and vasculogenesis; we also demonstrated the safety and efficacy of this cell therapy in an earlier phase I/II clinical trial conducted on seven patients with fracture nonunion. Herein, we present the results of a phase III clinical trial conducted to confirm the results of the previous phase studies using a larger cohort of patients. METHODS: CD34 + cells were mobilized via administration of granulocyte colony-stimulating factor, harvested using leukapheresis, and isolated using magnetic cell sorting. Autologous CD34 + cells were transplanted in 15 patients with tibia nonunion and 10 patients with femur nonunion, who were followed up for 52 weeks post transplantation. The main outcome was a reduction in time to heal the tibia in nonunion patients compared with that in historical control patients. We calculated the required number of patients as 15 based on the results of the phase I/II study. An independent data monitoring committee performed the radiographic assessments. Adverse events and medical device failures were recorded. RESULTS: All fractures healed during the study period. The time to radiological fracture healing was 2.8 times shorter in patients with CD34 + cell transplantation than in the historical control group (hazard ratio: 2.81 and 95% confidence interval 1.16-6.85); moreover, no safety concerns were observed. CONCLUSIONS: Our findings strongly suggest that autologous CD34 + cell transplantation is a novel treatment option for fracture nonunion. TRIAL REGISTRATION: UMIN-CTR, UMIN000022814. Registered on 22 June 2016.


Asunto(s)
Fracturas Óseas , Fracturas no Consolidadas , Humanos , Trasplante de Células , Curación de Fractura , Fracturas Óseas/terapia , Fracturas no Consolidadas/terapia , Factor Estimulante de Colonias de Granulocitos , Trasplante Autólogo , Resultado del Tratamiento
2.
J Med Virol ; 93(5): 2694-2704, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33368358

RESUMEN

The pandemic of novel coronavirus disease (COVID-19) is not yet close to being over, more than 8 months after the first cases, but researchers are making great progress in fighting the disease. We have conducted a brief review of the geographic differences in the prevalence of COVID-19, the updated pathological findings, prognostic factors, and treatments for disease prevention and improvement of prognosis. Although hydroxychloroquine and tocilizumab have been recommended by some researchers, many clinical trials have failed to confirm any beneficial effect of these and other drugs on COVID-19, in terms of improved clinical status or reduced patient mortality. Currently, glucocorticoid is the only drug that reduces the mortality of COVID-19 in a randomized controlled trial; however, it is still necessary to establish the optimal timing of administration. It is also urgent to set up an international or national cohort to address the risk factors associated with infection, the natural history of COVID-19, including the disease type, surrogate markers for critically ill, long-term sequelae, and reinfection after exposure, identify responders to glucocorticoid, and establish optimal treatment strategies for disease control.


Asunto(s)
COVID-19/diagnóstico , COVID-19/epidemiología , COVID-19/terapia , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antivirales/uso terapéutico , COVID-19/transmisión , Humanos , Hidroxicloroquina/uso terapéutico , Pandemias , Prevalencia , Pronóstico , Factores de Riesgo , SARS-CoV-2/patogenicidad , Resultado del Tratamiento
3.
J Wound Care ; 30(8): 666-676, 2021 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-34382845

RESUMEN

In consultation with academia and the Pharmaceuticals and Medical Devices Agency (PMDA), we have developed guidance for drafting protocols for clinical trials concerning medical devices for the healing of hard-to-heal wounds without ischaemia. The guidance summarises the validity of single-arm trials for hard-to-heal wounds, the definition of hard-to-heal wounds without ischaemia, methods of patient enrolment and clinical endpoints. This review focuses on the logical thinking process that was used when establishing the guidance for improving the efficiency of clinical trials concerning medical devices for hard-to-heal wounds. We particularly focused on the feasibility of conducting single-arm trials and also tried to clarify the definition of hard-to-heal wounds. If the feasibility of randomised control trials is low, conducting single-arm trials should be considered for the benefit of patients. In addition, hard-to-heal wounds were defined as meeting the following two conditions: wounds with a wound area reduction <50% at four weeks despite appropriate standards of care; and wounds which cannot be closed by a relatively simple procedure (for example, suture, skin graft and small flaps). Medical devices for hard-to-heal wound healing are classified into two types: (1) devices for promoting re-epithelialisation; and (2) devices for improving the wound bed. For medical devices for promoting re-epithelialisation, we suggest setting complete wound closure, percent wound area reduction or distance moved by the wound edge as the primary endpoint in single-arm trials for hard-to-heal wounds. For medical devices for improving the wound bed, we suggest setting the period in which wounds can be closed by secondary intention or a simple procedure, such as the primary endpoint.


Asunto(s)
Trasplante de Piel , Cicatrización de Heridas , Humanos , Japón , Literatura de Revisión como Asunto
4.
Circ J ; 83(7): 1422-1430, 2019 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-31178469

RESUMEN

In 1997, the seminal manuscript by Asahara, Murohara, Isner et al outlined the evidence for the existence of circulating, bone marrow-derived cells capable of stimulating and contributing to the formation of new blood vessels. Consistent with the paradigm shift that this work represented, it triggered much scientific debate and controversy, some of which persists 2 decades later. In contrast, the clinical application of autologous CD34 cell therapy has been marked by a track record of consistent safety and clinical benefit in multiple ischemic conditions. In this review, we summarize the preclinical and clinical evidence from over 700 patients in clinical trials of CD34 cell therapy.


Asunto(s)
Antígenos CD34/inmunología , Trastornos Cerebrovasculares/cirugía , Células Progenitoras Endoteliales/trasplante , Extremidad Inferior/irrigación sanguínea , Isquemia Miocárdica/cirugía , Neovascularización Fisiológica , Enfermedad Arterial Periférica/cirugía , Regeneración , Trasplante de Células Madre , Animales , Trastornos Cerebrovasculares/diagnóstico , Trastornos Cerebrovasculares/inmunología , Trastornos Cerebrovasculares/fisiopatología , Células Progenitoras Endoteliales/inmunología , Humanos , Isquemia Miocárdica/diagnóstico , Isquemia Miocárdica/inmunología , Isquemia Miocárdica/fisiopatología , Enfermedad Arterial Periférica/diagnóstico , Enfermedad Arterial Periférica/inmunología , Enfermedad Arterial Periférica/fisiopatología , Recuperación de la Función , Trasplante de Células Madre/efectos adversos , Resultado del Tratamiento
5.
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
6.
Circ J ; 78(2): 490-501, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24257136

RESUMEN

BACKGROUND: A prior phase I/IIa clinical trial provided evidence for safety, feasibility and potential efficacy of i.m. injection of granulocyte colony-stimulating factor (G-CSF)-mobilized CD34+ cells in patients with critical limb ischemia (CLI). METHODS AND RESULTS: A phase II trial of CD34+ cell therapy was conducted in patients with CLI to explore endpoint selection and timing. No-option CLI patients (n=11) underwent i.m. transplantation of G-CSF-mobilized CD34+ cells isolated by magnetic sorting. Ischemic rest pain scales improved from week 2 vs. baseline (P<0.05). Skin perfusion pressure (P=0.0175), transcutaneous partial oxygen pressure (P=0.0446) and pain-free walking distance (P=0.0056) improved from week 2, total walking distance from week 8 (P=0.0182) and toe brachial pressure index from week 12 (P=0.0174) vs. baseline. These parameters peaked at week 36 or 52. Rutherford's category improved from week 24 vs. baseline (P=0.0065). CLI-free ratio serially increased and peaked (85.7%) at week 36. Serial change in Rutherford's category correlated with that in Rest Pain Scale (P=0.0374), but not with that in any physiological parameters. CONCLUSIONS: Ischemic rest pain scales and physiological parameters improved relatively early after cell therapy, then plateaued later accompanied by recovery from the CLI state. Rutherford's category and CLI-free ratio at week 36 or later may be suitable endpoints in cell therapy clinical trials for CLI. Functional parameters should be evaluated independently of such clinical endpoints for ischemia severity. ( CLINICAL TRIAL REGISTRATION: URL: https://dbcentre3.jmacct.med.or.jp/jmactr/Default.aspx. Unique identifier: JMA-IIA00022)


Asunto(s)
Antígenos CD34 , Isquemia , Extremidad Inferior , Manejo del Dolor , Dolor/fisiopatología , Trasplante de Células Madre , Células Madre , Adulto , Anciano , Autoinjertos , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Femenino , Humanos , Isquemia/fisiopatología , Isquemia/terapia , Extremidad Inferior/irrigación sanguínea , Extremidad Inferior/fisiopatología , Masculino , Persona de Mediana Edad
7.
J Gastroenterol Hepatol ; 29(10): 1830-8, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24731186

RESUMEN

BACKGROUND AND AIM: Preclinical studies in rodent models of chronic liver fibrosis have shown that transplantation of peripheral blood (PB) CD34(+) cells leads to hepatic regeneration and a reduction of liver fibrosis by suppressing hepatic stellate cell activity and increasing matrix metalloproteinase activity. The aim of this study was to examine the safety and clinical efficacy of intrahepatic transplantation of autologous granulocyte colony-stimulating factor (G-CSF)-mobilized PB-CD34(+) cells in patients with decompensated liver cirrhosis. METHODS: PB-CD34(+) cells were isolated from G-CSF-mobilized apheresis products. Ten patients were treated with G-CSF-mobilized PB-CD34(+) cells (treatment group) and seven patients were treated with standard medical therapy. For mobilization, patients in the treatment group received subcutaneous injections of 10 µg G-CSF/kg/day for 5 days. The cells were then injected at three different doses (5 × 10(5) , 1 × 10(6) and 2 × 10(6) cells/kg) through the hepatic artery. Thereafter, all patients were followed up for 24 months. RESULTS: G-CSF treatment and leukapheresis were well tolerated, and no serious adverse events were observed. Patients in the treatment group had a significant but transient splenomegaly. After 24 weeks, serum albumin was significantly increased in patients who had received middle or high doses of CD34(+) cells compared with baseline. Doppler ultrasound showed a significant increase in hepatic blood flow velocity and blood flow volume after CD34(+) cell therapy. The hepatic vein pressure gradient decreased in two patients who received high-dose CD34(+) cells at week 16. CONCLUSIONS: CD34(+) cell therapy is feasible, safe and effective in slowing the decline of hepatic reserve function.


Asunto(s)
Antígenos CD34 , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Cirrosis Hepática/terapia , Trasplante de Células Madre de Sangre Periférica/métodos , Anciano , Autoinjertos , Estudios de Factibilidad , Femenino , Estudios de Seguimiento , Factor Estimulante de Colonias de Granulocitos/farmacología , Arteria Hepática , Células Estrelladas Hepáticas/parasitología , Venas Hepáticas/fisiopatología , Humanos , Inyecciones Subcutáneas , Circulación Hepática , Cirrosis Hepática/enzimología , Cirrosis Hepática/patología , Cirrosis Hepática/fisiopatología , Regeneración Hepática , Masculino , Metaloproteinasas de la Matriz/metabolismo , Persona de Mediana Edad , Estudios Prospectivos , Terapéutica , Factores de Tiempo , Presión Venosa
8.
Regen Ther ; 27: 455-463, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38737403

RESUMEN

Introduction: In this multicenter clinical study, we aimed to investigate the efficacy and safety of the transhepatic arterial administration of granulocyte-colony stimulating factor (G-CSF)-mobilized autologous peripheral blood (PB)-CD34+ cells compared with standard therapy in patients with decompensated cirrhosis type C. Methods: Patients were randomly assigned (2:1) to the CD34+ cell transplant (CD34+ cell) or standard-of-care (SOC) group and followed up for 52 weeks. The primary endpoints were the non-progression rate of Child-Pugh (CP) scores at 24 weeks post-enrollment and the safety of the protocol treatment. Results: Fourteen patients (CD34+ cell group: 10; SOC group: 4) were enrolled. CP scores at 24 weeks had a non-progression rate of 90% in the CD34+ cell group and 100% in the SOC group, with no significant difference between groups. Importantly, 4 out of 10 patients in the CD34+ cell group exhibited an improvement from decompensated to compensated cirrhosis, whereas all patients in the SOC group remained in decompensated cirrhosis. With regard to secondary endpoints, a trend toward increased serum albumin levels in the CD34+ cell group was noted. Serious adverse events (SAEs) occurred in three patients in the CD34+ cell group and in one patient in the SOC group. No causal relationship was observed between all SAEs and G-CSF, leukapheresis, or cell transplantation in the CD34+ cell group. No patients died and no hepatocellular carcinoma occurred within the study period. Conclusions: PB-CD34+ cell infusion therapy may have the potential to circumvent the decompensated stage of cirrhosis, thus avoiding the need for liver transplantation.

9.
Lab Invest ; 93(9): 1036-53, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23897412

RESUMEN

Lnk, an intracellular adapter protein, is expressed in hematopoietic cell lineages, which has recently been proved as an essential inhibitory signaling molecule for stem cell self-renewal in the stem cell factor-c-Kit signaling pathway with enhanced hematopoietic and osteogenic reconstitution in Lnk-deficient mice. Moreover, the therapeutic potential of hematopoietic stem/endothelial progenitor cells (EPCs) for fracture healing has been demonstrated with mechanistic insight into vasculogenesis/angiogenesis and osteogenesis enhancement in the fracture sites. We report here, Lnk siRNA-transfected endothelial commitment of c-kit+/Sca-1+/lineage- subpopulations of bone marrow cells have high EPC colony-forming capacity exhibiting endothelial markers, VE-Cad, VEGF and Ang-1. Lnk siRNA-transfected osteoblasts also show highly osteoblastic capacity. In vivo, locally transfected Lnk siRNA could successfully downregulate the expression of Lnk at the fracture site up to 1 week, and radiological and histological examination showed extremely accelerated fracture healing in Lnk siRNA-transfected mice. Moreover, Lnk siRNA-transfected mice exhibited sufficient therapeutic outcomes with intrinstic enhancement of angiogenesis and osteogenesis, specifically, the mice demonstrated better blood flow recovery in the sites of fracture. In our series of experiments, we clarified that a negatively regulated Lnk system contributed to a favorable circumstance for fracture healing by enhancing vasculogenesis/angiogenesis and osteogenesis. These findings suggest that downregulation of Lnk system may have the clinical potential for faster fracture healing, which contributes to the reduction of delayed unions or non-unions.


Asunto(s)
Fracturas Óseas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neovascularización Fisiológica/fisiología , ARN Interferente Pequeño/metabolismo , Cicatrización de Heridas/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Células de la Médula Ósea/metabolismo , Proliferación Celular , Distribución de Chi-Cuadrado , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Histocitoquímica , Péptidos y Proteínas de Señalización Intracelular/análisis , Péptidos y Proteínas de Señalización Intracelular/genética , Flujometría por Láser-Doppler , Masculino , Proteínas de la Membrana , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica/genética , Osteoblastos/citología , Osteoblastos/metabolismo , Osteogénesis/genética , Osteogénesis/fisiología , Fenotipo , ARN Interferente Pequeño/genética , Flujo Sanguíneo Regional , Estadísticas no Paramétricas , Transfección , Cicatrización de Heridas/genética , Microtomografía por Rayos X
10.
Nutrients ; 15(12)2023 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-37375642

RESUMEN

Type 2 diabetes mellitus is associated with an increased risk of dementia, potentially through multifactorial pathologies, including neuroinflammation. Therefore, there is a need to identify novel agents that can suppress neuroinflammation and prevent cognitive impairment in diabetes. In the present study, we demonstrated that a high-glucose (HG) environment elevates the intracellular reactive oxygen species (ROS) levels and triggers inflammatory responses in the mouse microglial cell line BV-2. We further found that thioredoxin-interacting protein (TXNIP), a ROS-responsive positive regulator of the nucleotide-binding oligomerization domain (NOD)-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome, was also upregulated, followed by NLRP3 inflammasome activation and subsequent interleukin-1beta (IL-1ß) production in these cells. Conversely, caspase-1 was not significantly activated, suggesting the involvement of noncanonical pathways in these inflammatory responses. Moreover, our results demonstrated that taxifolin, a natural flavonoid with antioxidant and radical scavenging activities, suppressed IL-1ß production by reducing the intracellular ROS levels and inhibiting the activation of the TXNIP-NLRP3 axis. These findings suggest the novel anti-inflammatory effects of taxifolin on microglia in an HG environment, which could help develop novel strategies for suppressing neuroinflammation in diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2 , Inflamasomas , Ratones , Animales , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Microglía/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Enfermedades Neuroinflamatorias , Glucosa/farmacología , Interleucina-1beta/metabolismo
11.
Nutrients ; 15(2)2023 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-36678220

RESUMEN

The molecular pathogenesis of nonalcoholic steatohepatitis (NASH) includes a complex interaction of metabolic stress and inflammatory stimuli. Considering the therapeutic goals of NASH, it is important to determine whether the treatment can prevent the progression from NASH to hepatocellular carcinoma. Taxifolin, also known as dihydroquercetin, is a natural bioactive flavonoid with antioxidant and anti-inflammatory properties commonly found in various foods and health supplement products. In this study, we demonstrated that Taxifolin treatment markedly prevented the development of hepatic steatosis, chronic inflammation, and liver fibrosis in a murine model of NASH. Its mechanisms include a direct action on hepatocytes to inhibit lipid accumulation. Taxifolin also increased brown adipose tissue activity and suppressed body weight gain through at least two distinct pathways: direct action on brown adipocytes and indirect action via fibroblast growth factor 21 production in the liver. Notably, the Taxifolin treatment after NASH development could effectively prevent the development of liver tumors. Collectively, this study provides evidence that Taxifolin shows pleiotropic effects for the treatment of the NASH continuum. Our data also provide insight into the novel mechanisms of action of Taxifolin, which has been widely used as a health supplement with high safety.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Humanos , Animales , Ratones , Enfermedad del Hígado Graso no Alcohólico/etiología , Hígado/metabolismo , Obesidad/metabolismo , Carcinogénesis/metabolismo , Transformación Celular Neoplásica/patología , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad
12.
J Cell Physiol ; 227(4): 1538-52, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21678408

RESUMEN

We investigated whether transplantation of purified human peripheral blood CD34(+) cells could reduce established liver fibrosis and up-regulate therapeutic regeneration. Human peripheral blood CD34(+) cells were isolated from total mononuclear cells of healthy volunteers by magnetic cell sorting. Recipient nude rats were injected intraperitoneally with carbon tetrachloride (CCl(4)) twice weekly for 3 weeks before single administration of CD34(+) cells. CCl(4) was then re-administered twice weekly for 3 more weeks, and the nude rats were sacrificed. Saline (control group), 1 × 10(5) (low-dose group), 5 × 10(5) (middle-dose group), or 2 × 10(6) (high-dose group) CD34(+) cells/kg body weight were intrasplenically transplanted after CCl(4) treatment for 3 weeks. Reverse transcriptase-polymerase chain reaction analysis of the freshly isolated CD34(+) cells revealed the expression of CD31, keratin19, α-smooth muscle actin (α-SMA), and epithelial growth factor, but not other liver related markers. The transplanted cells differentiated into vascular and sinusoidal endothelial cells, and vascular smooth muscle cells. CD34(+) cell transplantation reduced liver fibrosis in a dose-dependent fashion, with decreased collagen type-I and α-SMA-positive cells after 6 weeks of CCl(4) treatment by Mallory's Azan and immunohistochemical staining. Gelatin zymography showed that the expression levels of active matrix metalloproteinase-2 and -9 in CD34(+) cell transplanted livers were significantly stronger than those in saline-infused livers. In recipients of high-doses of CD34(+) cells, the number of PCNA-positive hepatocyte increased 6 weeks after CCl(4) treatment compared with saline-infused livers. We conclude that human peripheral blood CD34(+) cell transplantation halts established liver fibrosis and promotes hepatic regeneration in CCl(4)-induced chronic liver injury.


Asunto(s)
Enfermedad Hepática Crónica Inducida por Sustancias y Drogas/terapia , Regeneración Hepática , Trasplante de Células Madre de Sangre Periférica , Animales , Antígenos CD34/metabolismo , Secuencia de Bases , Tetracloruro de Carbono/toxicidad , Diferenciación Celular , Supervivencia Celular , Enfermedad Hepática Crónica Inducida por Sustancias y Drogas/genética , Enfermedad Hepática Crónica Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Crónica Inducida por Sustancias y Drogas/patología , Cartilla de ADN/genética , Células Endoteliales/patología , Expresión Génica , Células Estrelladas Hepáticas/patología , Humanos , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/patología , Cirrosis Hepática/terapia , Regeneración Hepática/genética , Regeneración Hepática/fisiología , Masculino , Miocitos del Músculo Liso/patología , Ratas , Ratas Endogámicas F344 , Ratas Desnudas , Trasplante Heterólogo
13.
J Neurosci Res ; 90(12): 2281-92, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22996658

RESUMEN

Spinal cord injury causes initial mechanical damage, followed by ischemia-induced, secondary degeneration, worsening the tissue damage. Although endothelial progenitor cells (EPCs) have been reported to play an important role for pathophysiological neovascularization in various ischemic tissues, the EPC kinetics following spinal cord injury have never been elucidated. In this study, we therefore assessed the in vivo kinetics of bone marrow-derived EPCs by EPC colony-forming assay and bone marrow transplantation from Tie2/lacZ transgenic mice into wild-type mice with spinal cord injury. The number of circulating mononuclear cells and EPC colonies formed by the mononuclear cells peaked at day 3 postspinal cord injury. Bone marrow transplantation study revealed that bone marrow-derived EPCs recruited into the injured spinal cord markedly increased at day 7, when neovascularization and astrogliosis drastically occurred in parallel with axon growth in the damaged tissue. To elucidate further the contribution of EPCs to recovery after spinal cord injury, exogenous EPCs were systemically infused immediately after the injury. The administered EPCs were incorporated into the injured spinal cord and accelerated neovascularization and astrogliosis. These findings suggest that bone marrow-derived EPCs may contribute to the tissue repair by augmenting neovascularization and astrogliosis following spinal cord injury.


Asunto(s)
Astrocitos/patología , Trasplante de Médula Ósea , Células Endoteliales/fisiología , Gliosis/etiología , Células Madre Hematopoyéticas/fisiología , Neovascularización Fisiológica , Traumatismos de la Médula Espinal/cirugía , Cicatrización de Heridas/fisiología , Animales , Linaje de la Célula , Movimiento Celular , Ensayo de Unidades Formadoras de Colonias , Genes Reporteros , Gliosis/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Quimera por Radiación , Recuperación de la Función , Traumatismos de la Médula Espinal/patología , Isquemia de la Médula Espinal/patología
14.
Stem Cells ; 29(11): 1650-5, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21948649

RESUMEN

Endothelial progenitor cells (EPCs) have been isolated and shown to be effective in animal models of ischemia, and many groups involved in clinical trials have demonstrated that EPC therapy is safe and feasible for the treatment of critical limb ischemia and cardiovascular diseases. However, many issues in the field of EPC biology, especially in regards 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. In this review, we introduce the recent concept of EPC identification in terms of hematopoietic and nonhematopoietic EPCs along with the development of EPC biology research. Furthermore, we define the role of circulating EPCs in postnatal neovascularization to illustrate the future direction of EPC therapeutic applications. Next, we review on-going medical applications of EPC for cardiovascular and peripheral vascular diseases, introduce the practical example of therapeutic application of EPCs to patients with ischemic disease, and discuss about the feedback of clinical researches.


Asunto(s)
Células Endoteliales/citología , Células Madre/citología , Animales , Humanos , Isquemia/terapia , Neovascularización Fisiológica/fisiología , Trasplante de Células Madre
15.
Nat Med ; 11(11): 1197-204, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16244652

RESUMEN

Sonic hedgehog (Shh) is a crucial regulator of organ development during embryogenesis. We investigated whether intramyocardial gene transfer of naked DNA encoding human Shh (phShh) could promote a favorable effect on recovery from acute and chronic myocardial ischemia in adult animals, not only by promoting neovascularization, but by broader effects, consistent with the role of this morphogen in embryogenesis. After Shh gene transfer, the hedgehog pathway was upregulated in mammalian fibroblasts and cardiomyocytes. This resulted in preservation of left ventricular function in both acute and chronic myocardial ischemia by enhanced neovascularization, and reduced fibrosis and cardiac apoptosis. Shh gene transfer also enhanced the contribution of bone marrow-derived endothelial progenitor cells to myocardial neovascularization. These data suggest that Shh gene therapy may have considerable therapeutic potential in individuals with acute and chronic myocardial ischemia by triggering expression of multiple trophic factors and engendering tissue repair in the adult heart.


Asunto(s)
Terapia Genética , Corazón/embriología , Miocardio/metabolismo , Transducción de Señal , Transactivadores/uso terapéutico , Enfermedad Aguda , Animales , Células COS , Células Cultivadas , Chlorocebus aethiops , Enfermedad Crónica , Modelos Animales de Enfermedad , Ecocardiografía , Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog , Humanos , Ratones , Ratones Mutantes , Isquemia Miocárdica/etiología , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/fisiopatología , Isquemia Miocárdica/terapia , Miocardio/citología , Miocitos Cardíacos/metabolismo , Neovascularización Fisiológica , ARN Mensajero/metabolismo , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Porcinos , Función Ventricular Izquierda/fisiología
16.
Ann Vasc Dis ; 15(4): 241-252, 2022 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-36644256

RESUMEN

CD34 is a cell surface marker, which is expressed in various somatic stem/progenitor cells such as bone marrow (BM)-derived hematopoietic stem cells and endothelial progenitor cells (EPCs), skeletal muscle satellite cells, epithelial hair follicle stem cells, and adipose tissue mesenchymal stem cells. CD34+ cells in BM and peripheral blood are known as a rich source of EPCs. Thus, vascular regeneration therapy using granulocyte colony stimulating factor (G-CSF) mobilized- or BM CD34+ cells has been carried out in patients with various vascular diseases such as chronic severe lower limb ischemia, acute myocardial infarction, refractory angina, ischemic cardiomyopathy, and dilated cardiomyopathy as well as ischemic stroke. Pilot and randomized clinical trials demonstrated the safety, feasibility, and effectiveness of the CD34+ cell therapy in peripheral arterial, cardiovascular, and cerebrovascular diseases. This review provides an overview of the preclinical and clinical reports of CD34+ cell therapy for vascular regeneration.

17.
Artículo en Inglés | MEDLINE | ID: mdl-35415075

RESUMEN

Background/objective: The purpose of this study was to report the outcomes of a clinical trial conducted in Japan to assess the safety and effectiveness of third-generation autologous chondrocyte implantation (ACI) using IK-01 (CaReS™), which does not require flap coverage, in the treatment of patients with focal cartilage injury of the knee. Methods: This was an open label, exploratory clinical trial. Patients were enrolled between June 2012 and September 2016. The primary endpoint of the study was the International Knee Documentation Committee (IKDC) score at 52 weeks after implantation. The IKDC, Lysholm, and visual analog scale (VAS) scores were evaluated at the time of screening and at 4, 12, 24, 36, and 52 weeks after implantation. Improvements from the baseline scores were evaluated using the equation "(postoperative score) - (preoperative score)." Magnetic resonance imaging (MRI) was performed at 2, 12, 24, and 52 weeks after implantation, and MRI measurements were evaluated using T1 rho and T2 mapping. Results: Nine patients were enrolled in this study and were examined for safety. Product quality did not satisfy the specification in one patient, and bacterial joint infection occurred in one patient. As a result, seven patients were included in the outcome analyses. The mean IKDC score significantly improved from 36.4 preoperatively to 74.1% at 52 weeks after implantation (p < 0.0001). The mean Lysholm and VAS scores also significantly improved from 39.6 to 57.4 to 89.6 and 22.9, respectively, after surgery. In the MRI evaluation, the T1 rho and T2 values of the implanted area were similar to those of the surrounding cartilage at 52 weeks after implantation. Conclusions: Third generation ACI (IK-01) can be an effective treatment option for focal cartilage defects of the knee; however, surgeons must pay careful attention to the risk of postoperative joint infection.

18.
Front Med (Lausanne) ; 9: 681316, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35360717

RESUMEN

This study was conducted to evaluate the safety and efficacy of human peripheral blood CD34 positive (CD34+) cells transplanted into a murine chronic stroke model to obtain pre-clinical proof of concept, prior to clinical testing. Granulocyte colony stimulating factor (G-CSF) mobilized human CD34+ cells [1 × 104 cells in 50 µl phosphate-buffered saline (PBS)] were intravenously (iv) or intra-carotid arterially (ia) transplanted 4 weeks after the induction of stroke (chronic stage), and neurological function was evaluated. In this study, severe combined immune deficiency (SCID) mice were used to prevent excessive immune response after cell therapy. Two weeks post cell therapy, the ia CD34+ cells group demonstrated a significant improvement in neurological functions compared to the PBS control. The therapeutic effect was maintained 8 weeks after the treatment. Even after a single administration, ia transplantation of CD34+ cells had a significant therapeutic effect on chronic stroke. Based on the result of this pre-clinical proof of concept study, a future clinical trial of autologous peripheral blood CD34+ cells administration in the intra-carotid artery for chronic stroke patients is planned.

19.
J Mol Cell Cardiol ; 51(3): 308-17, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21557947

RESUMEN

Although endothelial progenitor cells (EPCs) differentiate from minor populations of stem cells in bone marrow (BM), the differential role of hematopoietic stem cell (HSC) subpopulations in EPC development is largely unclear. Morphological characterization of EPC colonies has revealed that c-kit+/Sca-1+/lineage (Lin)-(KSL) cells mainly develop small EPC-colony forming units (CFUs) not large EPC-CFUs. In contrast, c-kit+/Sca-1-/Lin- (KL) cells develop large EPC-CFUs not small EPC-CFUs. Neither c-kit-/Sca-1+/Lin- (SL) cells nor c-kit-/Sca-1-/Lin- (L) cells develop EPC-CFUs to an appreciable extent. Hindlimb ischemia enhances formation of large EPC-CFUs from all HSC subpopulations, suggesting an important role for ischemia in functional EPC development. Real time RT-PCR analysis shows that KSL, KL and SL cells but not L cells express various factors at high levels, maintaining a BM-EPC pool. In hindlimb ischemia, transplanted KSL, KL and SL cells efficiently differentiate into endothelial lineage cells in situ and augment capillary density. The percentage of Ki-67+ cycling cells among transplanted cells in ischemic tissue was also greater for KSL, KL and SL cells than L cells. Moreover, the frequency of VEGF- or SDF-1-expressing cells was higher transplanted KSL, KL or SL cells than L cells. Thus, KSL, KL and SL cells are not different in their angiogenic competence under ischemic conditions. In conclusion, although KSL cells are clearly the most potent contributors to EPC development, KL and SL cells may also contribute to neovascularization via both autocrine and paracrine mechanisms in response to ischemic signals.


Asunto(s)
Células de la Médula Ósea/metabolismo , Células Endoteliales/metabolismo , Células Madre Hematopoyéticas/metabolismo , Isquemia/metabolismo , Neovascularización Fisiológica , Animales , Células de la Médula Ósea/citología , Diferenciación Celular , Hipoxia de la Célula/genética , Proliferación Celular , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/citología , Regulación del Desarrollo de la Expresión Génica , Células Madre Hematopoyéticas/citología , Isquemia/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neovascularización Fisiológica/genética
20.
Lab Invest ; 91(4): 539-52, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21135814

RESUMEN

Human multipotent adipose-derived stem cells (hMADSCs) have recently been isolated featuring extensive expansion capacity ex vivo. We tested the hypothesis that hMADSC transplantation might contribute to cardiac functional recovery by its direct or indirect effect on myocardial infarction (MI). Nude rats were either transplanted with hMADSCs or PBS (control) in ischemic myocardium immediately following MI. Echocardiographical assessment of cardiac function after MI with hMADSCs showed significant improvement of each parameter compared to that with PBS. Histological analysis also showed significantly reduced infarct size and increased capillary density in peri-infarct myocardium by hMADSC treatment. However, remarkable transdifferentiation of hMADSCs into cardiac or vascular lineage cells was not observed. Despite the less transdifferentiation capacity, hMADSCs produced robust multiple pro-angiogenic growth factors and chemokines, such as vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), and stromal cell-derived factor-1α (SDF-1α). Specifically, hMADSC-derived SDF-1α had a crucial role for cooperative angiogenesis, with the paracrine effect of hMADSCs and Tie2-positive bone marrow (BM) progenitor recruitment in ischemic myocardium. hMADSCs exhibit a therapeutic effect on cardiac preservation following MI, with the production of VEGF, bFGF, and SDF-1α showing paracrine effects and endogenous BM stem/progenitor recruitment to ischemic myocardium rather than its direct contribution to tissue regeneration.


Asunto(s)
Adipocitos/trasplante , Isquemia Miocárdica/cirugía , Trasplante de Células Madre , Animales , Células de la Médula Ósea/patología , Trasplante de Médula Ósea , Capilares/patología , Diferenciación Celular , Quimiocina CXCL12 , Vasos Coronarios/patología , Ecocardiografía , Factores de Crecimiento de Fibroblastos , Corazón/fisiopatología , Humanos , Infarto del Miocardio/complicaciones , Infarto del Miocardio/patología , Isquemia Miocárdica/diagnóstico , Isquemia Miocárdica/etiología , Isquemia Miocárdica/fisiopatología , Miocardio/patología , Neovascularización Fisiológica , Comunicación Paracrina , Ratas , Ratas Desnudas , Células Madre/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA