Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 471
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
N Engl J Med ; 385(24): 2264-2270, 2021 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-34881838

RESUMO

Inherited junctional epidermolysis bullosa is a severe genetic skin disease that leads to epidermal loss caused by structural and mechanical fragility of the integuments. There is no established cure for junctional epidermolysis bullosa. We previously reported that genetically corrected autologous epidermal cultures regenerated almost an entire, fully functional epidermis on a child who had a devastating form of junctional epidermolysis bullosa. We now report long-term clinical outcomes in this patient. (Funded by POR FESR 2014-2020 - Regione Emilia-Romagna and others.).


Assuntos
Epiderme/transplante , Epidermólise Bolhosa Juncional/terapia , Queratinócitos/transplante , Transdução Genética , Transgenes , Autorrenovação Celular , Células Cultivadas/transplante , Criança , Células Clonais , Epiderme/patologia , Epidermólise Bolhosa Juncional/genética , Epidermólise Bolhosa Juncional/patologia , Seguimentos , Doenças Genéticas Inatas/patologia , Doenças Genéticas Inatas/terapia , Terapia Genética , Vetores Genéticos , Humanos , Queratinócitos/citologia , Queratinócitos/fisiologia , Masculino , Regeneração , Células-Tronco/fisiologia , Transplante Autólogo
2.
Cancer Immunol Immunother ; 69(5): 825-834, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32060687

RESUMO

Adjuvant cytokine-induced killer (CIK) cell immunotherapy has shown potential in improving the prognosis of hepatocellular carcinoma (HCC) patients after curative resection. However, whether an individual could obtain survival benefit from CIK cell treatment remains unknown. In the present study, we focused on the characteristics of CIK cells and aimed to identify the best predictive biomarker for adjuvant CIK cell treatment in patients with HCC after surgery. This study included 48 patients with HCC treated with postoperative adjuvant CIK cell immunotherapy. The phenotype activity and cytotoxic activity of CIK cells were determined by flow cytometry and xCELLigence™ Real-Time Cell Analysis (RTCA) system, respectively. Correlation analysis revealed that the cytotoxic activity of CIK cells was significantly negative correlated with the percentage of CD3+ CD4+ cell subsets, but significantly positive correlated with CD3-CD56+ and CD3+ CD56+ cell subsets. Survival analysis showed that there were no significant associations between patients' prognosis and the phenotype of CIK cells. By contrast, there was statistically significant improvement in recurrence-free survival (RFS) and overall survival (OS) for patients with high cytotoxic activity of CIK cells as compared with those with low cytotoxic activity of CIK cells. Univariate and multivariate analyses indicated that CIK cell cytotoxicity was an independent prognostic factor for RFS and OS. In conclusion, a high cytotoxic activity of CIK cells can serve as a valuable biomarker for adjuvant CIK cell immunotherapy of HCC patients after surgery.


Assuntos
Carcinoma Hepatocelular/terapia , Células Matadoras Induzidas por Citocinas/transplante , Citotoxicidade Imunológica , Imunoterapia/métodos , Neoplasias Hepáticas/terapia , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/mortalidade , Técnicas de Cultura de Células , Células Cultivadas/imunologia , Células Cultivadas/transplante , Terapia Combinada/métodos , Células Matadoras Induzidas por Citocinas/imunologia , Testes Imunológicos de Citotoxicidade , Feminino , Citometria de Fluxo , Hepatectomia , Humanos , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/mortalidade , Masculino , Pessoa de Meia-Idade , Período Pós-Operatório , Prognóstico , Análise de Sobrevida , Transplante Autólogo/métodos
3.
Am J Physiol Gastrointest Liver Physiol ; 317(6): G845-G852, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31589468

RESUMO

Intestinal organoid cultures provide an in vitro model system for studying pathways and mechanisms involved in epithelial damage and repair. Derived from either embryonic or induced pluripotent stem cells or adult intestinal stem cells or tissues, these self-organizing, multicellular structures contain polarized mature cells that recapitulate both the physiology and heterogeneity of the intestinal epithelium. These cultures provide a cutting-edge technology for defining regenerative pathways that are induced following radiation or chemical damage, which directly target the cycling intestinal stem cell, or damage resulting from viral, bacterial, or parasitic infection of the epithelium. Novel signaling pathways or biological mechanisms identified from organoid studies that mediate regeneration of the epithelium following damage are likely to be important targets of preventive or therapeutic modalities to mitigate intestinal injury. The evolution of these cultures to include more components of the intestinal wall and the ability to genetically modify them are key components for defining the mechanisms that modulate epithelial regeneration.


Assuntos
Células-Tronco Adultas , Enteropatias , Intestinos , Organoides , Regeneração/fisiologia , Animais , Células Cultivadas/fisiologia , Células Cultivadas/transplante , Humanos , Enteropatias/etiologia , Enteropatias/metabolismo , Enteropatias/terapia , Intestinos/efeitos dos fármacos , Intestinos/efeitos da radiação , Modelos Biológicos , Organoides/fisiologia , Organoides/transplante , Engenharia Tecidual/métodos
4.
Eur J Vasc Endovasc Surg ; 55(2): 257-265, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29208350

RESUMO

OBJECTIVE: Endothelial cells (ECs) play an important role in neovascularisation, but are too limited in number for adequate therapeutic applications. Mesenchymal stem cells (MSCs) have the potential to differentiate into endothelial lineage cells, which makes them attractive candidates for therapeutic angiogenesis. The aim of this study was to investigate efficient differentiation of MSCs into ECs by inducing medium in vitro. METHODS: MSCs were isolated from bone marrow by density gradient centrifugation. The characterisation of the MSCs was determined by their cluster of differentiation (CD) marker profile. Inducing medium containing vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), insulin like growth factor (IGF), epidermal growth factor (EGF), ascorbic acid, and heparin was applied to differentiate the MSCs into ECs. Endothelial differentiation was quantitatively evaluated using flow cytometry. Real time quantitative PCR (qRT-PCR) was used to analyse mRNA expression of endothelial markers. Tube formation assay was further performed to examine the functional status of the differentiated MSCs. RESULTS: Flow cytometry analysis demonstrated that CD31+ and CD34+ cells increased steadily from 12% at 3 days, to 40% at 7 days, and to 60% at 14 days. Immunofluorescence staining further confirmed the expression of CD31 and CD34. qRT-PCR showed that expression of von Willebrand factor (vWF), vascular endothelial cadherin (VE-cadherin) and vascular endothelial growth factor receptor-2 (VEGFR-2) were significantly higher in the induced MSCs group compared with the uninduced MSCs group. The functional behavior of the differentiated cells was tested by tube formation assay in vitro on matrigel. Induced MSCs were capable of developing capillary networks, and progressive formation of vessel like structures was associated with increased EC population. CONCLUSIONS: These results provide a method to efficiently promote differentiation of MSCs into ECs in vitro for potential application in the treatment of peripheral arterial disease.


Assuntos
Diferenciação Celular/fisiologia , Citocinas/metabolismo , Células Endoteliais/fisiologia , Células-Tronco Mesenquimais/fisiologia , Doença Arterial Periférica/terapia , Biomarcadores/metabolismo , Separação Celular/métodos , Células Cultivadas/fisiologia , Células Cultivadas/transplante , Centrifugação com Gradiente de Concentração/métodos , Meios de Cultura/metabolismo , Células Endoteliais/transplante , Citometria de Fluxo , Humanos , Neovascularização Fisiológica/fisiologia , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Fator A de Crescimento do Endotélio Vascular
5.
Adv Exp Med Biol ; 1059: 219-240, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29736576

RESUMO

Stem cell research plays a central role in the future of medicine, which is mainly dependent on the advances on regenerative medicine (RM), specifically in the disciplines of tissue engineering (TE) and cellular therapeutics. All RM strategies depend upon the harnessing, stimulation, or guidance of endogenous developmental or repair processes in which cells have an important role. Among the most clinically challenging disorders, cartilage degeneration, which also affects subchondral bone becoming an osteochondral (OC) defect, is one of the most demanding. Although primary cells have been clinically applied, stem cells are currently seen as the promising tool of RM-related research because of its availability, in vitro proliferation ability, pluri- or multipotency, and immunosuppressive features. Being the OC unit, a transition from the bone to cartilage, mesenchymal stem cells (MSCs) are the main focus for OC regeneration. Promising alternatives, which can also be obtained from the patient or at banks and have great differentiation potential toward a wide range of specific cell types, have been reported. Still, ethical concerns and tumorigenic risk are currently under discussion and assessment. In this book chapter, we revise the existing stem cell-based approaches for engineering bone and cartilage, focusing on cell therapy and TE. Furthermore, 3D OC composites based on cell co-cultures are described. Finally, future directions and challenges still to be faced are critically discussed.


Assuntos
Doenças Ósseas/terapia , Doenças das Cartilagens/terapia , Medicina Regenerativa/métodos , Transplante de Células-Tronco/métodos , Células-Tronco Adultas/transplante , Transplante de Medula Óssea , Células Cultivadas/transplante , Condrócitos/transplante , Condrogênese , Células-Tronco Embrionárias/citologia , Previsões , Humanos , Células-Tronco Pluripotentes Induzidas/transplante , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Osteogênese , Medicina Regenerativa/tendências , Engenharia Tecidual/métodos
6.
Dev Biol ; 419(2): 199-216, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27640086

RESUMO

The neural crest (NC) is a remarkable transient structure generated during early vertebrate development. The neural crest progenitors have extensive migratory capacity and multipotency, harboring stem cell-like characteristics such as self-renewal. They can differentiate into a variety of cell types from craniofacial skeletal tissues to the trunk peripheral nervous system (PNS). Multiple regulators such as signaling factors, transcription factors, and migration machinery components are expressed at different stages of NC development. Gain- and loss-of-function studies in various vertebrate species revealed epistatic relationships of these molecules that could be assembled into a gene regulatory network defining the processes of NC induction, specification, migration, and differentiation. These basic developmental studies led to the subsequent establishment and molecular validation of neural crest stem cells (NCSCs) derived by various strategies. We provide here an overview of the isolation and characterization of NCSCs from embryonic, fetal, and adult tissues; the experimental strategies for the derivation of NCSCs from embryonic stem cells, induced pluripotent stem cells, and skin fibroblasts; and recent developments in the use of patient-derived NCSCs for modeling and treating neurocristopathies. We discuss future research on further refinement of the culture conditions required for the differentiation of pluripotent stem cells into axial-specific NC progenitors and their derivatives, developing non-viral approaches for the generation of induced NC cells (NCCs), and using a genomic editing approach to correct genetic mutations in patient-derived NCSCs for transplantation therapy. These future endeavors should facilitate the therapeutic applications of NCSCs in the clinical setting.


Assuntos
Doenças do Sistema Nervoso/cirurgia , Crista Neural/transplante , Células-Tronco Neurais/transplante , Transplante de Células-Tronco , Células-Tronco Adultas/transplante , Animais , Técnicas de Cultura de Células , Linhagem da Célula , Células Cultivadas/transplante , Desenvolvimento Embrionário , Transição Epitelial-Mesenquimal , Transplante de Tecido Fetal , Previsões , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Células-Tronco Embrionárias Humanas/transplante , Humanos , Células-Tronco Pluripotentes Induzidas/transplante , Crista Neural/fisiologia , Neurogênese , Especificidade de Órgãos , Vertebrados/embriologia
7.
J Craniofac Surg ; 28(6): 1462-1467, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28708646

RESUMO

BACKGROUND: Cultured epithelial cells transplantation is a known surgical technique for vitiligo. OBJECTIVE: To evaluate the factors influencing efficacy and safety of cultured epithelial cells transplantation in 9-month follow-up. METHODS: Demographic, clinical, and repigmentation outcomes were reviewed for patients with facial segmental vitiligo who had undergone cultured epithelial cells transplantation from November 2013 to July 2015 at the clinic of the Department of Dermatology, Huashan Hospital, China. RESULTS: Twenty-eight patients who had undergone cultured epithelial cells transplantation were included. A satisfactory result (>50% repigmentation) was achieved in 79% patients with facial segmental vitiligo in 9 months. The treatment effect was significantly different in 6th month (P = 0.032), 9th month (P = 0.006) compared with 3rd month. Disease stability did significantly affect repigmentation outcome in 9th month (Z = 2.113, P = 0.035). No significant difference was observed between single segmental type versus mixed type (Z = 1.081, P = 0.280). Adverse effects were nearly absent. CONCLUSION: Cultured epithelial cells transplantation is a relatively safe and effective therapy for facial segmental stable vitiligo patients.


Assuntos
Transplante de Células/métodos , Células Cultivadas/transplante , Células Epiteliais/transplante , Face/fisiopatologia , Vitiligo , Humanos , Vitiligo/fisiopatologia , Vitiligo/terapia
8.
Int Wound J ; 14(1): 165-171, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26968430

RESUMO

The critical problem of post-burn depigmentation is the lacking normal melanocytes. Auto-skin grafting and autologous non-cultured epidermal cell suspension have been used to improve the appearance. However, a large amount of skin graft is required of donor sites in the former method, while the latter method is thought to be complicated and costly. This study is designed to generalise the experience of tiny epidermal particles graft (TEPG) for treating post-burn depigmentation. From 2012 to 2013, 30 consecutive patients with depigmentation caused by burn injuries were divided into I and II group. I group: 15 cases (11 males and 4 females) were treated by the TEPG. II group: 15 patients (10 males and 5 females) were treated by suction blister epidermal skin graft (SBEG). Imagine-Pro Plus software was used to evaluate the size of repigmentation (RP) 12 months post-surgery. SPSS software 13.0 was used to evaluate the data. The optimum rate of RP was defined as more than 75% (RP > 75%) when excellent RP was defined as more than 90% (>90%). All patients were followed up for 12 months. The mean size of RP in two groups demonstrated that there were statistically significant differences in pigmentation between the two groups (P = 0·002), while there was no significant difference in the other factors (gender, site and age). No infection occurred in the recipient site. Pathological result showed that melanocytes existed at the basal layer of resurfacing skin. Optimum RP (RP > 75%) was seen in 12 patients in I group and 9 patients in II group. Excellent RP was achieved in 14 cases in I group and 10 patients in II group. Excellent RP can be obtained by the abovementioned two surgical techniques. In contrast to SBEG, TEPG is less traumatic, and definite effects can be guaranteed. It is a preferred treatment, especially for those patients who suffer from large depigmented lesions.


Assuntos
Queimaduras/complicações , Queimaduras/cirurgia , Células Cultivadas/transplante , Melanócitos/transplante , Pigmentação da Pele/fisiologia , Transplante de Pele/métodos , Transplante Autólogo/métodos , Feminino , Humanos , Masculino , Resultado do Tratamento , Cicatrização/fisiologia
9.
Am J Hematol ; 91(5): 453-60, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26858124

RESUMO

Limited cell numbers in umbilical cord blood (UCB) grafts present a major impediment to favorable outcomes in adult transplantation, largely related to delayed or failed engraftment. The advent of UCB transplantation (UCBT) using two grafts successfully circumvents this obstacle, despite the engraftment of only one unit. Preclinical models suggested that the addition of UCB T cells at the time of transplant can enhance engraftment. We tested whether ex vivo activation by CD3/CD28 costimulation and expansion of T cells from a single UCB graft would be safe and feasible in adults with advanced hematologic malignancies, with an overall objective of optimizing engraftment in single unit UCBT. In this phase 1 study, recipients of single UCB units were eligible if the unit was stored in two adequate fractions. Dose limiting toxicity was defined as grade 3 or grade 4 GVHD within 90 days of UCBT. Four patients underwent UCBT; all were treated at the first dose level (10(5) cells/kg). At the 10(5) cells/kg dose level two subjects experienced grade 3 intestinal GVHD, thus meeting stopping criteria. For three subjects, neutrophil engraftment was early (12, 17, and 20 days), while one subject experienced primary graft failure. We observed early donor T cell trafficking and found that expanded T cells produced supraphysiologic levels of cytokines relevant to engraftment and to lymphoid differentiation and function. Taken together, these preliminary data suggest rapid engraftment in recipients of a single UCBT combined with relatively low doses of activated T cells, though potentially complicated by severe GVHD.


Assuntos
Transplante de Células-Tronco de Sangue do Cordão Umbilical/métodos , Transfusão de Linfócitos , Subpopulações de Linfócitos T/transplante , Adulto , Anemia Refratária com Excesso de Blastos/terapia , Fator Ativador de Células B/biossíntese , Preservação de Sangue , Antígenos CD28/imunologia , Complexo CD3/imunologia , Células Cultivadas/transplante , Transplante de Células-Tronco de Sangue do Cordão Umbilical/efeitos adversos , Transplante de Células-Tronco de Sangue do Cordão Umbilical/mortalidade , Criopreservação , Citocinas/análise , Relação Dose-Resposta Imunológica , Estudos de Viabilidade , Feminino , Sobrevivência de Enxerto , Doença Enxerto-Hospedeiro/etiologia , Histocompatibilidade , Humanos , Recém-Nascido , Leucemia Mieloide Aguda/terapia , Transfusão de Linfócitos/efeitos adversos , Masculino , Dose Máxima Tolerável , Proteínas de Membrana/biossíntese , Pessoa de Meia-Idade , Neutrófilos/transplante , Especificidade do Receptor de Antígeno de Linfócitos T , Condicionamento Pré-Transplante/métodos , Resultado do Tratamento
11.
Circ Res ; 113(5): 571-87, 2013 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-23948583

RESUMO

Recent advances in the burgeoning field of genome engineering are accelerating the realization of personalized therapeutics for cardiovascular disease. In the postgenomic era, sequence-specific gene-editing tools enable the functional analysis of genetic alterations implicated in disease. In partnership with high-throughput model systems, efficient gene manipulation provides an increasingly powerful toolkit to study phenotypes associated with patient-specific genetic defects. Herein, this review emphasizes the latest developments in genome engineering and how applications within the field are transforming our understanding of personalized medicine with an emphasis on cardiovascular diseases.


Assuntos
Proteínas de Bactérias/fisiologia , Doenças Cardiovasculares/genética , Desoxirribonucleases de Sítio Específico do Tipo II/fisiologia , Desoxirribonucleases/fisiologia , Engenharia Genética/métodos , Genômica , Células-Tronco Pluripotentes Induzidas/citologia , Mutagênese Sítio-Dirigida/métodos , Medicina de Precisão/tendências , Xanthomonas/enzimologia , Animais , Proteínas de Bactérias/genética , Sítios de Ligação , Doenças Cardiovasculares/terapia , Diferenciação Celular , Células Cultivadas/citologia , Células Cultivadas/transplante , DNA/genética , DNA/metabolismo , Desoxirribonucleases/genética , Desoxirribonucleases de Sítio Específico do Tipo II/genética , Previsões , Genes Reporter , Engenharia Genética/tendências , Humanos , Células-Tronco Pluripotentes Induzidas/transplante , Modelos Cardiovasculares , Modelos Genéticos , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/fisiologia , Reparo de DNA por Recombinação , Especificidade por Substrato , Xanthomonas axonopodis/enzimologia , Peixe-Zebra/genética
12.
Xenotransplantation ; 22(1): 1-6, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25041180

RESUMO

Although xenografts have always held immeasurable potential as an inexhaustible source of donor organs, immunological barriers and physiological incompatibility have proved to be formidable obstacles to clinical utility. An exciting, new regenerative medicine-based approach termed "semi-xenotransplantation" (SX) seeks to overcome these obstacles by combining the availability and reproducibility of animal organs with the biocompatibility and functionality of human allografts. Compared to conventional xenotransplantation wherein the whole organ is animal-derived, SX grafts are cleansed of their antigenic cellular compartment to produce whole-organ extracellular matrix scaffolds that retain their innate structure and vascular channels. These scaffolds are then repopulated with recipient or donor human stem cells to generate biocompatible semi-xenografts with the structure and function of native human organs. While numerous hurdles must be still overcome in order for SX to become a viable treatment option for end-stage organ failure, the immense potential of SX for meeting the urgent needs for a new source of organs and immunosuppression-free transplantation justifies the interest that the transplant community is committing to the field.


Assuntos
Bioprótese , Células Cultivadas/transplante , Matriz Extracelular/transplante , Medicina Regenerativa/tendências , Alicerces Teciduais , Animais , Diferenciação Celular , Previsões , Rejeição de Enxerto/prevenção & controle , Humanos , Tolerância Imunológica , Medicina Regenerativa/métodos , Células-Tronco/citologia , Transplantes/provisão & distribuição
13.
Xenotransplantation ; 22(1): 7-19, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25130196

RESUMO

BACKGROUND: The need for pig islet xenotransplantation in patients with type 1 diabetes is compelling; however, the ideal age at which islets should be isolated from the donor pig remains uncertain. Pig islet transplantation in primates, as a valuable pre-clinical model, has been explored using adult, neonatal, fetal pig islets, and also pancreatic primordia from pig embryos as beta cell donors. Neonatal pig islets have some advantages over adult and fetal islets, but the optimal age within the first month of life at which neonatal islets should be isolated and transplanted is as yet unclear. METHODS: In an attempt to answer this question, we carried out a literature search, but limited the search primarily to evidence in the clinically-relevant pig-to-non-human primate model. RESULTS: We identified surprisingly few studies in this model directed to this topic. Even in pig-to-rodent models, there were few definitive data. CONCLUSION: From the few data available to us, we conclude that pancreatectomy and islet isolation from neonatal pigs may have advantages over adult pigs and that isolation during the first week of life may have minor advantages over later weeks.


Assuntos
Transplante das Ilhotas Pancreáticas/métodos , Sus scrofa , Doadores de Tecidos , Transplante Heterólogo/métodos , Fatores Etários , Animais , Animais Recém-Nascidos , Células Cultivadas/transplante , Diabetes Mellitus Experimental/cirurgia , Diabetes Mellitus Tipo 1/cirurgia , Transplante de Tecido Fetal , Xenoenxertos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Omento , Veia Porta , Primatas , Roedores , Sus scrofa/embriologia , Sus scrofa/crescimento & desenvolvimento , Suínos , Transplante Heterotópico/métodos
14.
Digestion ; 92(2): 73-7, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26227553

RESUMO

The intestinal epithelium exerts multiple, indispensable functions to maintain the homeostasis of our body. However, this layer is frequently damaged by various gastrointestinal diseases; repair of such a damaged intestinal epithelium becomes difficult and the stem cell function gets lost or severely disrupted. Recent advances in the stem cell biology of the gastrointestinal tract have provided major breakthroughs, such as in vitro culture of intestinal stem cells (ISCs), and have also thrown light on the transplantation of those cells to repair the damaged intestinal mucosa. Based on such newly developed techniques, we are now coming close to apply them to treat various gastrointestinal diseases, including inflammatory bowel diseases (IBD). Accordingly, ISC-based therapy is currently under development for treating IBD patients, which may potentially provide benefits to those patients by the achievement of mucosal healing and the subsequent improvement of their prognosis. In this review, we would like to highlight studies that have provided information on the great advances made thus far in the research of ISCs. In addition, we would like to express some of our perspectives on the clinical use of ex-vivo-cultured ISCs in the treatment of IBD.


Assuntos
Células-Tronco Adultas/transplante , Células Cultivadas/transplante , Doenças Inflamatórias Intestinais/terapia , Mucosa Intestinal/citologia , Medicina Regenerativa/tendências , Técnicas de Cultura de Células , Trato Gastrointestinal , Humanos , Intestinos/fisiopatologia , Medicina Regenerativa/métodos , Cicatrização
15.
Br J Haematol ; 160(2): 123-32, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23061882

RESUMO

Gamma delta (γδ) Τ cells are non-conventional T lymphocyte effectors that can interact with and eradicate tumour cells. Several data demonstrate that these T cells, which are implicated in the first line of defence against pathogens, have anti-tumour activity against many cancers and suggest that γδ Τ cell-mediated immunotherapy is feasible and might induce objective tumour responses. Due to the importance of γδ Τ lymphocytes in the induction and control of immunity, a complete understanding of their biology is crucial for the development of a potent cancer immunotherapy. This review discusses recent advances in γδ Τ basic research and data from clinical trials on the use of γδ Τ cells in the treatment of different cancers. It analyses how this knowledge might be applied to develop new strategies for the clinical manipulation and the potentiation of γδ Τ lymphocyte activity in cancer immunotherapy.


Assuntos
Imunoterapia Adotiva , Neoplasias/terapia , Receptores de Antígenos de Linfócitos T gama-delta/análise , Subpopulações de Linfócitos T/transplante , Linfócitos T Citotóxicos/transplante , Imunidade Adaptativa , Adjuvantes Imunológicos/farmacologia , Antígenos de Neoplasias/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/imunologia , Células Cultivadas/transplante , Ensaios Clínicos como Assunto , Células Dendríticas/imunologia , Células Dendríticas/transplante , Difosfonatos/farmacologia , Difosfonatos/uso terapêutico , Rearranjo Gênico da Cadeia delta dos Receptores de Antígenos dos Linfócitos T , Rearranjo Gênico da Cadeia gama dos Receptores de Antígenos dos Linfócitos T , Humanos , Imunidade Inata , Vigilância Imunológica/imunologia , Células Matadoras Naturais/imunologia , Células-Tronco Mesenquimais/imunologia , Neoplasias/imunologia , Receptores de Antígenos de Linfócitos T alfa-beta/análise , Receptores de Antígenos de Linfócitos T gama-delta/genética , Subpopulações de Linfócitos T/química , Subpopulações de Linfócitos T/imunologia , Linfócitos T Citotóxicos/química , Linfócitos T Citotóxicos/imunologia , Linfócitos T Reguladores/imunologia , Pesquisa Translacional Biomédica , Evasão Tumoral/imunologia
16.
Osteoarthritis Cartilage ; 21(10): 1465-73, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23831631

RESUMO

BACKGROUND: An important goal of stem cell research in orthopaedics is to develop clinically relevant techniques that could be applied to heal cartilage or joint pathology. Stem cell treatment in orthopaedics for joint pathology is promising since these cells have the ability to modulate different processes in the various tissues of the joint simultaneously. The non life-threatening nature of musculoskeletal system disorders makes safety of stem cell therapy a necessary prerequisite. OBJECTIVE: To systematically review the literature and provide an overview of reported adverse events (AEs) of intra-articular treatment with culture-expanded stem cells in humans. DESIGN: A systematic literature search was performed in Pubmed, EMBASE, Web of Science and CINAHL in February 2013. AEs were reported into three categories: local/systemic, serious adverse event or AE (SAE/AE), related/unrelated. RESULTS: 3039 Potentially eligible articles were identified of which eventually eight fulfilled our inclusion criteria. In total, 844 procedures with a mean follow-up of 21 months were analysed. Autologous bone marrow-derived mesenchymal stem cells (BM-MSCs) were used for cartilage repair and osteoarthritis treatment in all included studies. Four SAEs were reported by the authors. One infection following bone marrow aspiration (BMA) was reported as probably related and resolved with antibiotics. One pulmonary embolism occurred 2 weeks after BMA and was reported as possibly related. Two tumours, both not at the site of injection, were reported as unrelated. Twenty-two other cases of possible procedure-related and seven of possible stem cell-product related adverse events (AEs) were documented. The main AEs related to the procedure were increased pain/swelling and dehydration after BMA. Increased pain and swelling was the only AE reported as related to the stem cell-product. CONCLUSIONS: Based on current literature review we conclude that application of cultured stem cells in joints appears to be safe. We believe that with continuous caution for potential side effects, it is reasonable to continue with the development of articular stem cell therapies.


Assuntos
Transplante de Células-Tronco Mesenquimais/efeitos adversos , Transplante de Células-Tronco Mesenquimais/métodos , Osteoartrite/terapia , Técnicas de Cultura de Células , Células Cultivadas/transplante , Humanos , Injeções Intra-Articulares
18.
Nat Genet ; 6(4): 335-41, 1994 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8054972

RESUMO

An ex vivo approach to gene therapy for familial hypercholesterolaemia (FH) has been developed in which the recipient is transplanted with autologous hepatocytes that are genetically corrected with recombinant retroviruses carrying the LDL receptor. We describe the treatment of a 29 year old woman with homozygous FH by ex vivo gene therapy directed to liver. She tolerated the procedures well and in situ hybridization of liver tissue four months after therapy revealed evidence for engraftment of transgene expressing cells. The patient's LDL/HDL ratio declined from 10-13 before gene therapy to 5-8 following gene therapy, improvements which have remained stable for the duration of the treatment (18 months). This represents the first report of human gene therapy in which stable correction of a therapeutic endpoint has been achieved.


Assuntos
Células Cultivadas/transplante , Terapia Genética , Hiperlipoproteinemia Tipo II/terapia , Fígado , Receptores de LDL/genética , Proteínas Recombinantes/uso terapêutico , Adulto , Biópsia , Terapia Combinada , Ponte de Artéria Coronária , Doença das Coronárias/etiologia , Doença das Coronárias/cirurgia , Feminino , Seguimentos , Regulação da Expressão Gênica/efeitos dos fármacos , Genes Sintéticos , Terapia Genética/métodos , Vetores Genéticos , Humanos , Hiperlipoproteinemia Tipo II/sangue , Hiperlipoproteinemia Tipo II/complicações , Hiperlipoproteinemia Tipo II/tratamento farmacológico , Hiperlipoproteinemia Tipo II/patologia , Hibridização in Situ Fluorescente , Lipoproteínas HDL/sangue , Lipoproteínas LDL/sangue , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Lovastatina/farmacologia , Lovastatina/uso terapêutico , Receptores de LDL/biossíntese , Receptores de LDL/deficiência , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/biossíntese , Segurança , Regulação para Cima/efeitos dos fármacos
19.
Br J Haematol ; 156(3): 383-7, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22017592

RESUMO

Patients with paroxysmal nocturnal haemoglobinuria (PNH) have expanded clonal cells bearing a somatic mutation in the PIGA gene. Our previous study on two PNH patients with chromosome 12 rearrangements demonstrated the involvement of HMGA2 expression in clonal expansion. The present study investigated HMGA2 expression in PNH patients without chromosomal abnormalities. The expression of short HMGA2 with latent exon was significantly high in peripheral blood cells from 18 of 24 patients. Over-expression of truncated HMGA2 in mouse bone marrow cells caused expansion in recipient mice. These results support the idea that deregulated expression of HMGA2 causes expansion of PNH cells.


Assuntos
Proteína HMGA2/fisiologia , Hemoglobinúria Paroxística/metabolismo , Processamento Alternativo , Anemia Aplástica/patologia , Animais , Células da Medula Óssea/patologia , Divisão Celular , Células Cultivadas/transplante , Células Clonais/patologia , Ensaio de Unidades Formadoras de Colônias , Éxons/genética , Regulação da Expressão Gênica , Proteína HMGA2/biossíntese , Proteína HMGA2/genética , Hemoglobinúria Paroxística/genética , Hemoglobinúria Paroxística/patologia , Humanos , Proteínas de Membrana/deficiência , Camundongos , Quimera por Radiação , Proteínas Recombinantes de Fusão/fisiologia , Transdução Genética
20.
Br J Haematol ; 158(6): 778-87, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22816563

RESUMO

In vitro exposure of haematopoietic stem and progenitor cells (HSPC) to cytokines in expansion or gene therapy protocols reduces homing and engraftment in vivo. We have previously reported that this is related in part to altered tissue specificity of short-term homing, leading to loss of cells in non-haematopoietic tissues. Here we demonstrate that defective engraftment persists when cultured HSPC are transplanted by intrabone injection. Changes in engraftment function occur within 24 h of cytokine exposure, and are evident when engraftment is analysed solely in the injected bone. A novel ex vivo model of the bone marrow was developed, in which the attachment of infused HSPC in rodent long bones is reduced following culture with cytokines. Finally, cultured HSPC demonstrated reduced adhesion to N-cadherin, osteopontin and vascular cell-adhesion molecule-1, ligands present in bone marrow niches. These changes in adhesive function occur rapidly, and are not related to downregulation of the relevant receptors. Our findings suggest that cytokine exposure of adult human HSPC results in altered adhesion within bone marrow niches, further leading to reduced engraftment potential in vivo.


Assuntos
Medula Óssea/patologia , Adesão Celular/fisiologia , Fatores Estimuladores de Colônias/farmacologia , Sobrevivência de Enxerto/fisiologia , Mobilização de Células-Tronco Hematopoéticas , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/patologia , Nicho de Células-Tronco/fisiologia , Adulto , Animais , Antígenos CD34/análise , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/transplante , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/farmacologia , Organismos Livres de Patógenos Específicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA