Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 227
Filtrar
1.
Stem Cell Res Ther ; 15(1): 117, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38654373

RESUMO

BACKGROUND: The detection rate of superficial non-ampullary duodenal epithelial tumors (SNADETs) has recently been increasing. Large tumors may contain malignant lesions and early therapeutic intervention is recommended. Endoscopic mucosal dissection (ESD) is considered a feasible treatment modality, however, the anatomical and physiological characteristics of the duodenum create a risk of postoperative perforation after ESD. METHODS: To explore whether myoblast sheet transplantation could prevent delayed perforation after ESD, a first-in-human (FIH) clinical trial of laparoscopic autologous myoblast sheet transplantation after duodenal ESD was launched. Autologous myoblast sheets fabricated from muscle tissue obtained seven weeks before ESD were transplanted laparoscopically onto the serous side of the ESD. The primary endpoints were the onset of peritonitis due to delayed perforation within three days after surgery and all adverse events during the follow-up period. RESULTS: Three patients with SNADETs ≥ 20 mm in size underwent transplantation of a myoblast sheet onto the serous side of the duodenum after ESD. In case 1, The patient's postoperative course was uneventful. Endoscopy and abdominal computed tomography revealed no signs of delayed perforation. Despite incomplete mucosal closure in case 2, and multiple micro perforations during ESD in case 3, cell sheet transplantation could prevent the postoperative massive perforation after ESD, and endoscopy on day 49 after transplantation revealed no stenosis. CONCLUSIONS: This clinical trial showed the safety, efficacy, and procedural operability of this novel regenerative medicine approach involving transplanting an autologous myoblast sheet laparoscopically onto the serosa after ESD in cases with a high risk of delayed perforation. This result indicates the potential application of cell sheet medicine in treating various abdominal organs and conditions with minimal invasiveness in the future. TRIAL REGISTRATION: jRCT, jRCT2073210094. Registered November 8 2021, https://jrct.niph.go.jp/latest-detail/jRCT2073210094 .


Assuntos
Laparoscopia , Mioblastos , Transplante Autólogo , Humanos , Laparoscopia/métodos , Laparoscopia/efeitos adversos , Masculino , Feminino , Mioblastos/transplante , Transplante Autólogo/métodos , Pessoa de Meia-Idade , Duodeno , Idoso , Mucosa Intestinal , Ressecção Endoscópica de Mucosa/efeitos adversos , Ressecção Endoscópica de Mucosa/métodos , Neoplasias Duodenais/cirurgia , Perfuração Intestinal/etiologia
2.
Elife ; 112022 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-35076017

RESUMO

Skeletal muscle myoblasts (iMyoblasts) were generated from human induced pluripotent stem cells (iPSCs) using an efficient and reliable transgene-free induction and stem cell selection protocol. Immunofluorescence, flow cytometry, qPCR, digital RNA expression profiling, and scRNA-Seq studies identify iMyoblasts as a PAX3+/MYOD1+ skeletal myogenic lineage with a fetal-like transcriptome signature, distinct from adult muscle biopsy myoblasts (bMyoblasts) and iPSC-induced muscle progenitors. iMyoblasts can be stably propagated for >12 passages or 30 population doublings while retaining their dual commitment for myotube differentiation and regeneration of reserve cells. iMyoblasts also efficiently xenoengrafted into irradiated and injured mouse muscle where they undergo differentiation and fetal-adult MYH isoform switching, demonstrating their regulatory plasticity for adult muscle maturation in response to signals in the host muscle. Xenograft muscle retains PAX3+ muscle progenitors and can regenerate human muscle in response to secondary injury. As models of disease, iMyoblasts from individuals with Facioscapulohumeral Muscular Dystrophy revealed a previously unknown epigenetic regulatory mechanism controlling developmental expression of the pathological DUX4 gene. iMyoblasts from Limb-Girdle Muscular Dystrophy R7 and R9 and Walker Warburg Syndrome patients modeled their molecular disease pathologies and were responsive to small molecule and gene editing therapeutics. These findings establish the utility of iMyoblasts for ex vivo and in vivo investigations of human myogenesis and disease pathogenesis and for the development of muscle stem cell therapeutics.


Muscular dystrophies are a group of inherited genetic diseases characterised by progressive muscle weakness. They lead to disability or even death, and no cure exists against these conditions. Advances in genome sequencing have identified many mutations that underly muscular dystrophies, opening the door to new therapies that could repair incorrect genes or rebuild damaged muscles. However, testing these ideas requires better ways to recreate human muscular dystrophy in the laboratory. One strategy for modelling muscular dystrophy involves coaxing skin or other cells from an individual into becoming 'induced pluripotent stem cells'; these can then mature to form almost any adult cell in the body, including muscles. However, this approach does not usually create myoblasts, the 'precursor' cells that specifically mature into muscle during development. This limits investigations into how disease-causing mutations impact muscle formation early on. As a response, Guo et al. developed a two-step protocol of muscle maturation followed by stem cell growth selection to isolate and grow 'induced myoblasts' from induced pluripotent stem cells taken from healthy volunteers and muscular dystrophy patients. These induced myoblasts can both make more of themselves and become muscle, allowing Guo et al. to model three different types of muscular dystrophy. These myoblasts also behave as stem cells when grafted inside adult mouse muscles: some formed human muscle tissue while others remained as precursor cells, which could then respond to muscle injury and start repair. The induced myoblasts developed by Guo et al. will enable scientists to investigate the impacts of different mutations on muscle tissue and to better test treatments. They could also be used as part of regenerative medicine therapies, to restore muscle cells in patients.


Assuntos
Células-Tronco Pluripotentes Induzidas/transplante , Distrofia Muscular Facioescapuloumeral/terapia , Mioblastos/transplante , Animais , Diferenciação Celular , Linhagem Celular , Linhagem da Célula , Células Cultivadas , Modelos Animais de Doenças , Proteínas de Homeodomínio/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Camundongos , Desenvolvimento Muscular , Distrofia Muscular Facioescapuloumeral/patologia , Fator de Transcrição PAX3/metabolismo , Recuperação de Função Fisiológica , Regeneração
3.
Mol Ther ; 29(4): 1425-1438, 2021 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-33429079

RESUMO

We evaluated the cardiac function recovery following skeletal myoblast cell-sheet transplantation and the long-term outcomes after applying this treatment in 23 patients with ischemic cardiomyopathy. We defined patients as "responders" when their left ventricular ejection fraction remained unchanged or improved at 6 months after treatment. At 6 months, 16 (69.6%) patients were defined as responders, and the average increase in left ventricular ejection fraction was 4.9%. The responders achieved greater improvement degrees in left ventricular and hemodynamic function parameters, and they presented improved exercise capacity. During the follow-up period (56 ± 28 months), there were four deaths and the overall 5-year survival rate was 95%. Although the responders showed higher freedom from mortality and/or heart failure admission (5-year, 81% versus 0%; p = 0.0002), both groups presented an excellent 5-year survival rate (5-year, 93% versus 100%; p = 0.297) that was higher than that predicted using the Seattle Heart Failure Model. The stepwise logistic regression analysis showed that the preoperative estimated glomerular filtration rate and the left ventricular end-systolic volume index were independently associated with the recovery progress. Approximately 70% of patients with "no-option" ischemic cardiomyopathy responded well to the cell-sheet transplantation. Preoperative renal and left ventricular function might predict the patients' response to this treatment.


Assuntos
Cardiomiopatias/terapia , Insuficiência Cardíaca/terapia , Mioblastos/transplante , Isquemia Miocárdica/terapia , Cardiomiopatias/genética , Cardiomiopatias/patologia , Feminino , Coração/crescimento & desenvolvimento , Coração/fisiopatologia , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/patologia , Ventrículos do Coração/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Isquemia Miocárdica/genética , Isquemia Miocárdica/patologia , Volume Sistólico/genética , Transplante Autólogo/efeitos adversos , Transplante Autólogo/métodos , Função Ventricular Esquerda/genética
4.
Int Urol Nephrol ; 53(5): 893-906, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33245534

RESUMO

PURPOSE: We tried to investigate the role of Schwann and satellite cells in the treatment of neurogenic bladder and bowel dysfunction; following spinal cord injury in the rabbit model. METHODS: Twelve male New Zealand rabbits underwent induction of neurogenic bladder by spinal cord injury. Rabbits underwent the fiber tractography analysis to confirm the induction of spinal cord injury. Then, animals were randomly divided into two groups. In group I (n = 4), Schwann cells were obtained from autologous peroneal nerve. In group II (n = 4), the co-culture of nerve-muscle cells was obtained from autologous peroneal nerve and quadriceps muscle. Animals in the control group (n = 4) did not undergo any rehabilitation therapy. One and 4 months after injection of cells into the external anal sphincter, electromyography, urethral pressure profiles, urodynamic studies, voiding cystourethrogram, and manometry was performed to confirm the efficacy of treatment in short- (1 month) and long-term (4 months) follow-ups. RESULTS: The investigations validated that no statistically significant difference was detected between the two experimental groups in a short-term follow-up (p-value > 0.05). However, the functional features were improved in group II in long-term follow-up. In both groups, the external anal sphincter contracted in response to electrical signals delivered to the muscle. However, more signals were detected in group II in electromyography evaluation. The immunohistochemical staining demonstrated that the histological features of the bladder and spinal cord were more satisfactory in group II in all follow-ups compared to group I, in terms of less edema, inflammation, presence of progenitor cells, and expression of muscle and nerve markes. CONCLUSION: Our results suggested that the injection of nerve-muscle co-culture cells into the external anal sphincter may be a helpful tactic for ameliorating the urological complications; following spinal cord injury induction in the rabbit model.


Assuntos
Mioblastos/transplante , Células de Schwann/transplante , Traumatismos da Medula Espinal/complicações , Bexiga Urinaria Neurogênica/etiologia , Bexiga Urinaria Neurogênica/cirurgia , Animais , Modelos Animais de Doenças , Masculino , Coelhos , Distribuição Aleatória , Engenharia Tecidual/métodos
5.
Cell Transplant ; 29: 963689720963882, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33121270

RESUMO

The recent advent of endoscopy has enabled the endoscopic submucosal dissection (ESD) of superficial nonampullary duodenal epithelial tumors. However, the substantially thin wall and presence of bile and pancreatic juice make it technically difficult to perform duodenal ESD without perforation, which leads to lethal complications. The present study evaluated the efficacy of autologous myoblast sheet transplantation for the prevention of late perforation after duodenal ESD in a porcine model. Two weeks before ESD, skeletal muscle was surgically excised from the femur of pigs, and myoblasts were isolated and seeded in temperature-responsive culture dishes to prepare sheets. Immediately after ESD, the autologous myoblast sheets were attached to the serosal surface at the ESD site with omentopexy. The pigs were divided into two groups: the autologous myoblast sheet group (n = 5), where the myoblast cell sheet was attached to the ESD ulcer part from the duodenal serous side, and the Omentum group (n = 5), where only the omentum was used. The pigs were sacrificed and analyzed macroscopically and histologically on postoperative day 3. The macroscopic examination of the abdominal cavity revealed perforation in the ESD ulcer area and leakage of bile in the Omentum group but no perforation in the Sheet group. A histopathological examination revealed that continuity of the duodenal wall at the ESD site was maintained with dense connective tissue in the Sheet group. In conclusion, autologous myoblast sheets were useful for preventing perforation after duodenal ESD.


Assuntos
Duodeno/cirurgia , Ressecção Endoscópica de Mucosa/efeitos adversos , Perfuração Intestinal/prevenção & controle , Perfuração Intestinal/terapia , Mioblastos/transplante , Animais , Modelos Animais de Doenças , Duodeno/patologia , Fibroblastos/citologia , Perfilação da Expressão Gênica , Perfuração Intestinal/sangue , Perfuração Intestinal/etiologia , Mioblastos/citologia , Necrose , Omento/patologia , Suínos , Transplante Autólogo , Resultado do Tratamento
6.
J Clin Invest ; 130(11): 5652-5664, 2020 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-32946430

RESUMO

Muscular dystrophies are a heterogeneous group of genetic diseases, characterized by progressive degeneration of skeletal and cardiac muscle. Despite the intense investigation of different therapeutic options, a definitive treatment has not been developed for this debilitating class of pathologies. Cell-based therapies in muscular dystrophies have been pursued experimentally for the last three decades. Several cell types with different characteristics and tissues of origin, including myogenic stem and progenitor cells, stromal cells, and pluripotent stem cells, have been investigated over the years and have recently entered in the clinical arena with mixed results. In this Review, we do a roundup of the past attempts and describe the updated status of cell-based therapies aimed at counteracting the skeletal and cardiac myopathy present in dystrophic patients. We present current challenges, summarize recent progress, and make recommendations for future research and clinical trials.


Assuntos
Diferenciação Celular , Distrofias Musculares , Mioblastos , Células-Tronco Pluripotentes , Transplante de Células-Tronco , Humanos , Músculo Esquelético/fisiologia , Distrofias Musculares/metabolismo , Distrofias Musculares/patologia , Distrofias Musculares/terapia , Mioblastos/metabolismo , Mioblastos/patologia , Mioblastos/transplante , Miocárdio/metabolismo , Miocárdio/patologia , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/patologia , Células-Tronco Pluripotentes/transplante , Regeneração
7.
Stem Cell Reports ; 15(1): 80-94, 2020 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-32619494

RESUMO

Duchenne muscular dystrophy (DMD) is a progressive and fatal muscle-wasting disease caused by DYSTROPHIN deficiency. Cell therapy using muscle stem cells (MuSCs) is a potential cure. Here, we report a differentiation method to generate fetal MuSCs from human induced pluripotent stem cells (iPSCs) by monitoring MYF5 expression. Gene expression profiling indicated that MYF5-positive cells in the late stage of differentiation have fetal MuSC characteristics, while MYF5-positive cells in the early stage of differentiation have early myogenic progenitor characteristics. Moreover, late-stage MYF5-positive cells demonstrated good muscle regeneration potential and produced DYSTROPHIN in vivo after transplantation into DMD model mice, resulting in muscle function recovery. The engrafted cells also generated PAX7-positive MuSC-like cells under the basal lamina of DYSTROPHIN-positive fibers. These findings suggest that MYF5-positive fetal MuSCs induced in the late stage of iPSC differentiation have cell therapy potential for DMD.


Assuntos
Células-Tronco Fetais/transplante , Distrofia Muscular de Duchenne/terapia , Mioblastos/transplante , Animais , Biomarcadores/metabolismo , Diferenciação Celular , Linhagem da Célula , Modelos Animais de Doenças , Distrofina/metabolismo , Genes Reporter , Proteínas de Fluorescência Verde/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Camundongos , Desenvolvimento Muscular , Distrofia Muscular de Duchenne/patologia , Fator Regulador Miogênico 5/metabolismo , Fator de Transcrição PAX3/metabolismo , Recuperação de Função Fisiológica , Regeneração
8.
Methods Mol Biol ; 2126: 167-175, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32112388

RESUMO

Superparamagnetic iron oxide (SPIO) nanoparticles can function as specific, long-term multimodal contrast agents for noninvasive imaging studies. Here we describe how to achieve high-resolution, long-term, serial images of single-label transplanted cells through two complementary imaging techniques: magnetic resonance imaging (MRI) and microcomputed tomography (µCT).


Assuntos
Rastreamento de Células/métodos , Meios de Contraste/química , Coração/fisiologia , Nanopartículas Magnéticas de Óxido de Ferro/química , Imageamento por Ressonância Magnética/métodos , Imagem Multimodal/métodos , Mioblastos/citologia , Microtomografia por Raio-X/métodos , Animais , Animais Recém-Nascidos , Camundongos , Mioblastos/transplante
9.
J Cereb Blood Flow Metab ; 40(2): 404-419, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-30621518

RESUMO

The myoblast-mediated delivery of angiogenic genes represents a cell-based approach for targeted induction of therapeutic collateralization. Here, we tested the superiority of myoblast-mediated co-delivery of vascular endothelial growth factor-A (VEGF) together with platelet-derived growth factor-BB (PDGF-BB) on transpial collateralization of an indirect encephalomyosynangiosis (EMS) in a model of chronic cerebral ischemia. Mouse myoblasts expressing a reporter gene alone (empty vector), VEGF, PDGF-BB or VEGF and PDGF-BB through a single bi-cistronic vector (VIP) were implanted into the temporalis muscle of an EMS following permanent ipsilateral internal carotid artery occlusion in adult, male C57BL/6N mice. Over 84 days, myoblast engraftment and gene product expression, hemodynamic impairment, transpial collateralization, angiogenesis, pericyte recruitment and post-ischemic neuroprotection were assessed. By day 42, animals that received PDGF-BB in combination with VEGF (VIP) showed superior hemodynamic recovery, EMS collateralization and ischemic protection with improved pericyte recruitment around the parenchymal vessels and EMS collaterals. Also, supplementation of PDGF-BB resulted in a striking astrocytic activation with intrinsic VEGF mobilization in the cortex below the EMS. Our findings suggest that EMS surgery together with myoblast-mediated co-delivery of VEGF/PDGF-BB may have the potential to serve as a novel treatment strategy for augmentation of collateral flow in the chronically hypoperfused brain.


Assuntos
Becaplermina , Isquemia Encefálica , Córtex Cerebral , Circulação Cerebrovascular , Vetores Genéticos , Mioblastos , Fator A de Crescimento do Endotélio Vascular , Animais , Becaplermina/biossíntese , Becaplermina/genética , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Isquemia Encefálica/fisiopatologia , Isquemia Encefálica/terapia , Córtex Cerebral/irrigação sanguínea , Córtex Cerebral/metabolismo , Doença Crônica , Masculino , Camundongos , Músculo Esquelético/metabolismo , Mioblastos/metabolismo , Mioblastos/transplante , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética
10.
Hum Gene Ther ; 31(3-4): 241-252, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31801386

RESUMO

Stem cell therapy is a promising strategy to treat muscle diseases such as Duchenne muscular dystrophy (DMD). To avoid immune rejection of donor cells or donor-derived muscle, autologous cells, which have been genetically modified to express dystrophin, are preferable to cells derived from healthy donors. Restoration of full-length dystrophin (FL-dys) using viral vectors is extremely challenging, due to the limited packaging capacity of the vectors, but we have recently shown that either a foamy viral or lentiviral vector is able to package FL-dys open-reading frame and transduce myoblasts derived from a DMD patient. Differentiated myotubes derived from these transduced cells produced FL-dys. Here, we transplanted the foamy viral dystrophin-corrected DMD myoblasts intramuscularly into mdx nude mice, and showed that the transduced cells contributed to muscle regeneration, expressing FL-dys in nearly all the muscle fibers of donor origin. Furthermore, we showed that the restored FL-dys recruited members of the dystrophin-associated protein complex and neuronal nitric oxide synthase within donor-derived muscle fibers, evidence that the restored dystrophin protein is functional. Dystrophin-expressing donor-derived muscle fibers expressed lower levels of utrophin than host muscle fibers, providing additional evidence of functional improvement of donor-derived myofibers. This is the first in vivo evidence that foamy virus vector-transduced DMD myoblasts can contribute to muscle regeneration and mediate functional dystrophin restoration following their intramuscular transplantation, representing a promising therapeutic strategy for individual small muscles in DMD.


Assuntos
Distrofina/genética , Vetores Genéticos/genética , Mioblastos/metabolismo , Mioblastos/transplante , Spumavirus/genética , Transdução Genética , Antígeno AC133/metabolismo , Animais , Biomarcadores , Transplante de Células , Células Cultivadas , Modelos Animais de Doenças , Imunofluorescência , Expressão Gênica , Vetores Genéticos/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos mdx , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Óxido Nítrico Sintase Tipo I/metabolismo , Regeneração , Sarcoglicanas/metabolismo
11.
Exp Neurol ; 323: 113086, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31639376

RESUMO

Muscular dystrophies are a group of genetic muscle disorders that cause progressive muscle weakness and degeneration. Within this group, Duchenne muscular dystrophy (DMD) is the most common and one of the most severe. DMD is an X chromosome linked disease that occurs to 1 in 3500 to 1 in 5000 boys. The cause of DMD is a mutation in the dystrophin gene, whose encoded protein provides both structural support and cell signaling capabilities. So far, there are very limited therapeutic options available and there is no cure for this disease. In this review, we discuss the existing cell therapy research, especially stem cell-based, which utilize myoblasts, satellite cells, bone marrow cells, mesoangioblasts and CD133+ cells. Finally, we focus on human pluripotent stem cells (hPSCs) which hold great potential in treating DMD. hPSCs can be used for autologous transplantation after being specified to a myogenic lineage. Over the last few years, there has been a rapid development of isolation, as well as differentiation, techniques in order to achieve effective transplantation results of myogenic cells specified from hPSCs. In this review, we summarize the current methods of hPSCs myogenic commitment/differentiation, and describe the current status of hPSC-derived myogenic cell transplantation.


Assuntos
Distrofia Muscular de Duchenne/terapia , Mioblastos/citologia , Células-Tronco Pluripotentes/citologia , Transplante de Células-Tronco/métodos , Diferenciação Celular/fisiologia , Humanos , Mioblastos/transplante
12.
Ann Thorac Surg ; 108(5): e303-e306, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30980822

RESUMO

Heart failure is a refractory condition despite remarkable medical progress in therapeutic concepts. Recently, tissue-engineered cell-sheet implantation for end-stage heart failure has been explored experimentally and clinically. We present the case of a 22-year-old woman with ischemic cardiomyopathy who underwent mitral valve replacement and coronary artery bypass graft for infective endocarditis. Ventricular assist device therapy was recommended. After cell-sheet therapy, cardiac function and clinical symptoms significantly improved, and the improvement persisted for more than 36 months without ventricular assist device therapy or heart transplantation. This regenerative treatment might be feasible and effective for severe heart failure of ischemic etiology.


Assuntos
Mioblastos/transplante , Isquemia Miocárdica/cirurgia , Feminino , Humanos , Fatores de Tempo , Engenharia Tecidual , Transplante Autólogo , Resultado do Tratamento , Adulto Jovem
13.
Cancer Radiother ; 23(2): 151-160, 2019 Apr.
Artigo em Francês | MEDLINE | ID: mdl-30898418

RESUMO

Abdominal and pelvic irradiations play a major place in the management of patients with cancer and present a risk of acute and late side effects. Radiation-induced lesions can affect kidney or urological structures. These side effects can have an impact in the quality of life of patients. The aim of this article is to describe the physiopathology, the symptomatology, and the principles of management of radiation-induced nephropathy, uretheritis, cystitis, and urethritis.


Assuntos
Radioterapia/efeitos adversos , Doenças Urológicas/etiologia , Doenças Urológicas/terapia , Antioxidantes/uso terapêutico , Estrogênios Conjugados (USP)/administração & dosagem , Humanos , Fatores Imunológicos/administração & dosagem , Terapia a Laser , Mioblastos/transplante , Neoplasias/radioterapia , Polidesoxirribonucleotídeos/administração & dosagem , Fator A de Crescimento do Endotélio Vascular/administração & dosagem
14.
Cell Prolif ; 52(3): e12602, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30891802

RESUMO

OBJECTIVES: Myoblast transfer therapy (MTT) is a technique to replace muscle satellite cells with genetically repaired or healthy myoblasts, to treat muscular dystrophies. However, clinical trials with human myoblasts were ineffective, showing almost no benefit with MTT. One important obstacle is the rapid senescence of human myoblasts. The main purpose of our study was to compare the various methods for scalable generation of proliferative human myoblasts. METHODS: We compared the immortalization of primary myoblasts with hTERT, cyclin D1 and CDK4R24C , two chemically defined methods for deriving myoblasts from pluripotent human embryonic stem cells (hESCs), and introduction of viral MyoD into hESC-myoblasts. RESULTS: Our results show that, while all the strategies above are suboptimal at generating bona fide human myoblasts that can both proliferate and differentiate robustly, chemically defined hESC-monolayer-myoblasts show the most promise in differentiation potential. CONCLUSIONS: Further efforts to optimize the chemically defined differentiation of hESC-monolayer-myoblasts would be the most promising strategy for the scalable generation of human myoblasts, for applications in MTT and high-throughput drug screening.


Assuntos
Mioblastos/citologia , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Transformação Celular Viral , Células Cultivadas , Ciclina D1/genética , Quinase 4 Dependente de Ciclina/genética , Marcadores Genéticos , Células-Tronco Embrionárias Humanas/citologia , Humanos , Desenvolvimento Muscular , Proteína MyoD/genética , Mioblastos/fisiologia , Mioblastos/transplante , Regeneração , Células Satélites de Músculo Esquelético/citologia , Telomerase/genética
15.
J Am Soc Nephrol ; 30(2): 261-276, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30635373

RESUMO

BACKGROUND: Although hepatocyte growth factor (HGF) has antifibrotic effects and is involved in angiogenesis and vasodilation, systemic administration of HGF to prevent kidney fibrosis is not a feasible strategy for suppressing interstitial fibrosis in patients with CKD. METHODS: We investigated a novel therapy involving HGF transgenic cell sheets grown in culture from human mesothelial cells and administered to rats with unilateral ureteral obstruction (UUO). We compared progression of fibrosis in rats with UUO that received one of five interventions: HGF-transgenic mesothelial cell sheets transplanted to the kidney surface, HGF-transgenic mesothelial cell sheets transplanted to thigh, mesotherial cell sheets transplanted to kidney, no sheets, or HGF injections. RESULTS: HGF transgenic cell sheets transplanted to the kidney strongly suppressed the induction of myofibroblasts and collagen in the kidney for 28 days; other interventions did not. Additionally, the HGF-secreting cell sheets ameliorated loss of peritubular capillaries and maintained renal blood flow. CONCLUSIONS: These findings suggest that cell sheet therapy is a novel and promising strategy for inhibiting progressive fibrosis in CKD.


Assuntos
Fator de Crescimento de Hepatócito/farmacologia , Insuficiência Renal Crônica/patologia , Insuficiência Renal Crônica/terapia , Obstrução Ureteral/terapia , Animais , Transplante de Células/métodos , Células Cultivadas , Modelos Animais de Doenças , Progressão da Doença , Fibrose/patologia , Fibrose/prevenção & controle , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Masculino , Mioblastos/transplante , Distribuição Aleatória , Ratos , Insuficiência Renal Crônica/metabolismo , Sensibilidade e Especificidade , Transfecção , Resultado do Tratamento , Obstrução Ureteral/patologia
16.
Tissue Eng Part C Methods ; 25(2): 103-113, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30648471

RESUMO

IMPACT STATEMENT: Methodologies for incorporation of cells into tissue-engineered grafts, particularly at the later preclinical stages, are suboptimal and non-validated, and monitoring cell fate within scaffolds cultured in bioreactors and in vivo is challenging. In this study, we demonstrate how bioluminescence imaging (BLI) can overcome these difficulties and allow quantitative cell tracking at multiple stages of the bioengineering preclinical pipeline. Our robust bioluminescence-based approach allowed reproducible longitudinal monitoring of mesoangioblast localization and survival in 2D/3D tissue culture, in organ-scale bioreactors, and in vivo. Our findings will encourage the use of BLI in tissue engineering studies, improving the overall quality of cell-scaffold interaction research.


Assuntos
Bioengenharia/métodos , Rastreamento de Células/métodos , Esôfago/fisiologia , Medições Luminescentes/métodos , Células-Tronco Mesenquimais/citologia , Músculo Esquelético/citologia , Mioblastos/citologia , Diferenciação Celular , Células Cultivadas , Criança , Humanos , Processamento de Imagem Assistida por Computador , Mioblastos/transplante , Alicerces Teciduais
17.
IUBMB Life ; 70(10): 976-984, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30212601

RESUMO

This study investigated the effect of muscle-derived stem cells (MDSCs) and adipose tissue-derived stem cells (ADSCs) in the treatment of stress urinary incontinence (SUI) and their differences in a rat model. MDSCs and ADSC were isolated from rats (n = 10), examined for their properties, and labeled with enhanced green fluorescent protein (EGFP) and ß-galactosidase (ß-gal) gene. Rats received bladder-neck and transurethral sphincter injection of EGFP-labeled MDSCs and ß-gal gene-labeled ADSC and injection of D-Hanks as a control (n = 24 each group). At 0, 15, 30, and 60 days after cells injection, urinary voiding function was assessed by urine dynamics detector. The rats were killed to harvest their urethras for tracking of MDSCs and ADSC. Western blotting and quantitative real-time reverse transcription PCR (qRT-PCR) was performed to detect smooth muscle contents. Urodynamic test showed that MDSCs and ADSC improved the function of urination in rats with intrinsic sphincter deficiency (ISD), and effect of MDSCs-treatment was more pronounced. In addition, histologic analysis showed that the MDSCs and ADSC-treated groups had significantly higher myosin and α-smooth muscle actin (α-SMA) content than the control group. Compared with ADSC-treated groups, the MDSCs-treated groups in myosin and α-SMA content showed the tendency of increase. In summary, MDSCs and ADSCs have obvious effects in the treatment and/or prevention of ISD and transplantation of MDSCs is more effective than ADSC. © 2018 IUBMB Life, 70(10):976-984, 2018.


Assuntos
Células-Tronco Mesenquimais , Músculo Esquelético/transplante , Transplante de Células-Tronco , Incontinência Urinária por Estresse/terapia , Actinas/metabolismo , Animais , Modelos Animais de Doenças , Proteínas de Fluorescência Verde/administração & dosagem , Proteínas de Fluorescência Verde/química , Humanos , Injeções , Músculo Esquelético/citologia , Músculo Liso/metabolismo , Músculo Liso/patologia , Mioblastos/citologia , Mioblastos/transplante , Miosinas/metabolismo , Ratos , Uretra/patologia , Incontinência Urinária por Estresse/genética , Incontinência Urinária por Estresse/urina
18.
J Cardiovasc Transl Res ; 11(5): 412-419, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30155598

RESUMO

Progressive cardiomyocyte loss in Duchenne muscular dystrophy (DMD) leads to cardiac fibrosis, cardiomyopathy, and eventually heart failure. In the present study, we observed that myogenic progenitor cells (MPC) carry mRNA for the dystrophin gene. We tested whether cardiac function can be improved in DMD by allograft transplantation of MPC-derived exosomes (MPC-Exo) into the heart to restore dystrophin protein expression. Exo from C2C12 cells (an MPC cell line) or vehicle were delivered locally into the hearts of MDX mice. After 2 days of treatment, we observed that MPC-Exo restored dystrophin expression in the hearts of MDX mice, which correlated with improved myocardial function in dystrophin-deficient MDX mouse hearts. In conclusion, this study demonstrated that allogeneic WT-MPC-Exo transplantation transiently restored dystrophin gene expression and improved cardiac function in MDX mice, suggesting that allogenic exosomal delivery may serve as an alternative treatment for cardiomyopathy of DMD.


Assuntos
Cardiomiopatias/cirurgia , Distrofina/metabolismo , Exossomos/transplante , Distrofia Muscular de Duchenne/complicações , Mioblastos/transplante , Miocárdio/metabolismo , Transplante de Células-Tronco/métodos , Função Ventricular Esquerda , Aloenxertos , Animais , Cardiomiopatias/etiologia , Cardiomiopatias/metabolismo , Cardiomiopatias/fisiopatologia , Linhagem Celular , Modelos Animais de Doenças , Distrofina/genética , Exossomos/metabolismo , Exossomos/ultraestrutura , Masculino , Camundongos Endogâmicos mdx , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/fisiopatologia , Mioblastos/metabolismo , Mioblastos/ultraestrutura , Miocárdio/patologia , Recuperação de Função Fisiológica
19.
Sci Rep ; 8(1): 8512, 2018 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-29855599

RESUMO

Cell microencapsulation is an attractive strategy for cell-based therapies that allows the implantation of genetically engineered cells and the continuous delivery of de novo produced therapeutic products. However, the establishment of a way to retrieve the implanted encapsulated cells in case the treatment needs to be halted or when cells need to be renewed is still a big challenge. The combination of micro and macroencapsulation approaches could provide the requirements to achieve a proper immunoisolation, while maintaining the cells localized into the body. We present the development and characterization of a porous implantable macrocapsule device for the loading of microencapsulated cells. The device was fabricated in polyamide by selective laser sintering (SLS), with controlled porosity defined by the design and the sintering conditions. Two types of microencapsulated cells were tested in order to evaluate the suitability of this device; erythropoietin (EPO) producing C2C12 myoblasts and Vascular Endothelial Growth Factor (VEGF) producing BHK fibroblasts. Results showed that, even if the metabolic activity of these cells decreased over time, the levels of therapeutic protein that were produced and, importantly, released to the media were stable.


Assuntos
Alginatos/química , Células Imobilizadas/citologia , Fibroblastos/citologia , Mioblastos/citologia , Nylons/química , Animais , Cápsulas/química , Sobrevivência Celular , Terapia Baseada em Transplante de Células e Tecidos , Células Imobilizadas/metabolismo , Células Imobilizadas/transplante , Cricetinae , Composição de Medicamentos/métodos , Eritropoetina/metabolismo , Fibroblastos/metabolismo , Fibroblastos/transplante , Camundongos , Mioblastos/metabolismo , Mioblastos/transplante , Porosidade , Impressão Tridimensional , Alicerces Teciduais/química , Fator A de Crescimento do Endotélio Vascular/metabolismo
20.
Mol Ther ; 26(4): 1093-1108, 2018 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-29503200

RESUMO

Duchenne muscular dystrophy (DMD) is a lethal muscle-wasting disease currently without cure. We investigated the use of the PiggyBac transposon for full-length dystrophin expression in murine mesoangioblast (MABs) progenitor cells. DMD murine MABs were transfected with transposable expression vectors for full-length dystrophin and transplanted intramuscularly or intra-arterially into mdx/SCID mice. Intra-arterial delivery indicated that the MABs could migrate to regenerating muscles to mediate dystrophin expression. Intramuscular transplantation yielded dystrophin expression in 11%-44% of myofibers in murine muscles, which remained stable for the assessed period of 5 months. The satellite cells isolated from transplanted muscles comprised a fraction of MAB-derived cells, indicating that the transfected MABs may colonize the satellite stem cell niche. Transposon integration site mapping by whole-genome sequencing indicated that 70% of the integrations were intergenic, while none was observed in an exon. Muscle resistance assessment by atomic force microscopy indicated that 80% of fibers showed elasticity properties restored to those of wild-type muscles. As measured in vivo, transplanted muscles became more resistant to fatigue. This study thus provides a proof-of-principle that PiggyBac transposon vectors may mediate full-length dystrophin expression as well as functional amelioration of the dystrophic muscles within a potential autologous cell-based therapeutic approach of DMD.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Elementos de DNA Transponíveis , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Distrofia Muscular de Duchenne/genética , Mioblastos/metabolismo , Mioblastos/transplante , Animais , Linhagem Celular , Terapia Baseada em Transplante de Células e Tecidos/métodos , Modelos Animais de Doenças , Distrofina/genética , Imunofluorescência , Dosagem de Genes , Expressão Gênica , Ordem dos Genes , Genes Reporter , Masculino , Camundongos , Camundongos Endogâmicos mdx , Camundongos SCID , Distrofia Muscular de Duchenne/patologia , Distrofia Muscular de Duchenne/fisiopatologia , Distrofia Muscular de Duchenne/terapia , Fenótipo , Transgenes , Transplante Autólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA