Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 137
Filtrar
1.
Dermatology ; 239(6): 919-925, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37573775

RESUMO

BACKGROUND: Vitiligo is a common depigmentation skin disease associated with significant psychosocial morbidity and profound effect on the quality of life. The treatment of vitiligo is still a major challenge in the field of dermatology. Currently, topical steroids, calcineurin inhibitors, ultraviolet phototherapy, surgery, and cultured and non-cultured epidermal melanocyte transplantation are used for the treatment of vitiligo. However, the effectiveness of these treatment modalities is limited by the lack of response, long-term treatment periods, high cost, and inevitable adverse effects. OBJECTIVES: In this study, we aimed to evaluate the efficacy of intraepidermal injection of autologous non-cultured melanocytes and keratinocytes as an alternative therapy for the refractory and stable (RS) vitiligo. METHODS: The treatment procedure was performed on thirty-nine RS vitiligo patients. The autologous skin grafts obtained from the buttock area and epidermis were separated from dermis using dispase. Single-cell autologous melanocytes and keratinocytes were prepared from the epidermis by trypsin/ethylene diamine tetra acetic acid and injected at the concentration of 100-400 × 103 cells/cm2, intra-epidermally to the selected vitiligo lesions. Vitiligo re-pigmentation was monitored employing photography. Photographs were taken prior to and 2, 4, and 6 months after the cell transplantation. Improvement of the skin depigmentation was classified as follows: <25% as minimal response, 26-50% as moderate response, 51-75% as good response, and finally 76-100% as excellent response. RESULTS: Cell infusion appeared to be safe as none of the patients exhibited any adverse effects. At the end of the sixth month follow-up period, of the treated patients, 12.8% demonstrated an excellent response, 36% exhibited a good response, and 51.2% showed a moderate to minimal response to the administered therapy. Obtained significant p value for Wilcoxon test over the checkpoints at 2nd, 4th, and 6th month (p = 0.03, 0.04, and 0.039, respectively) post-cell transplantation confirmed notable growing trend in the re-pigmentation. CONCLUSION: Our findings provide a strong support for the therapeutic efficacy of autologous non-cultured melanocytes and keratinocytes in patients with RS vitiligo.


Assuntos
Vitiligo , Humanos , Vitiligo/patologia , Qualidade de Vida , Resultado do Tratamento , Queratinócitos/patologia , Melanócitos/patologia , Melanócitos/transplante
2.
J Cell Mol Med ; 25(22): 10554-10564, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34632708

RESUMO

Acute respiratory distress syndrome (ARDS) is the most common cause of death in COVID-19 patients. The cytokine storm is the main driver of the severity and magnitude of ARDS. Placenta-derived decidua stromal cells (DSCs) have a stronger immunosuppressive effect than other sources of mesenchymal stromal cells. Safety and efficacy study included 10 patients with a median age of 50 (range 14-68) years with COVID-19-induced ARDS. DSCs were administered 1-2 times at a dose of 1 × 106 /kg. End points were safety and efficacy by survival, oxygenation and effects on levels of cytokines. Oxygenation levels increased from a median of 80.5% (range 69-88) to 95% (range 78-99) (p = 0.012), and pulmonary infiltrates disappeared in all patients. Levels of IL-6 decreased from a median of 69.3 (range 35.0-253.4) to 11 (range 4.0-38.3) pg/ml (p = 0.018), and CRP decreased from 69 (range 5-169) to 6 (range 2-31) mg/ml (p = 0.028). Two patients died, one of a myocardial infarction and the other of multiple organ failure, diagnosed before the DSC therapy. The other patients recovered and left the intensive care unit (ICU) within a median of 6 (range 3-12) days. DSC therapy is safe and capable of improving oxygenation, decreasing inflammatory cytokine level and clearing pulmonary infiltrates in patients with COVID-19.


Assuntos
Tratamento Farmacológico da COVID-19 , Transplante de Células/métodos , Síndrome da Liberação de Citocina/terapia , Síndrome do Desconforto Respiratório/virologia , Células Estromais/transplante , Adolescente , Adulto , Idoso , COVID-19/complicações , COVID-19/terapia , Transplante de Células/efeitos adversos , Síndrome da Liberação de Citocina/etiologia , Citocinas/sangue , Feminino , Humanos , Tempo de Internação , Masculino , Pessoa de Meia-Idade , Placenta/citologia , Gravidez , Síndrome do Desconforto Respiratório/terapia , Células Estromais/fisiologia , Resultado do Tratamento
3.
Exp Cell Res ; 379(2): 225-234, 2019 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-30981668

RESUMO

This study aims to prepare intermediate mesoderm-like cells from mouse embryonic fibroblasts (MEFs). In the first step, intermediate mesoderm-like cells (IMLCs) and renal epithelial-like cells (RELCs) were extracted from mouse embryonic stem cells (mESCs) in a specified media that contained two small molecules, CHIR99021 and TTNPB, along with growth factors, FGF9and BMP7. Then, MEFs were directly converted into IM by genes for the pluripotency factors, which encode the transcription factors; Oct4, Sox2, Klf4, and c-Myc (OSKM). These unstable intermediate cells were quickly encouraged to form IM with the assistance of CHIR99021 and TTNPB. The results showed that exogenous expression of OSKM factors for four days was adequate to generate partially reprogrammed cells (SSEA1+/Nanog-). Real-time PCR and immunocytochemistry analysis confirmed the presence of the MEF-derived IMs. This study introduced a method for mESCs differentiation to RELCs followed by MEF conversion in an attempt to generate IM by circumventing pluripotency.


Assuntos
Reprogramação Celular/fisiologia , Células-Tronco Embrionárias/metabolismo , Fibroblastos/metabolismo , Células-Tronco Pluripotentes/citologia , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Células-Tronco Pluripotentes Induzidas/citologia , Rim/metabolismo , Fator 4 Semelhante a Kruppel , Mesoderma/metabolismo , Camundongos
4.
Differentiation ; 109: 1-8, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31323479

RESUMO

End-stage renal disease (ESRD) is a major global public health issue. In the past decade, regenerative medicine and cell-based therapies were recommended for treatment of devastating diseases like ESRD. Renal progenitor (RP) cells are essential players in such treatment approaches. The major practical difficulties in application of RP cells are generation of these cells and preservation of their self-renewal capacity; also, they should lack identified appropriate cell surface markers. To identify and isolate RP cells, two cell surface markers namely, CD133 and CD24 were recently used. In this study, we used these markers to facilitate selection and purification of RP cells from embryoid bodies (EBs), and assessed the impact of the use of bFGF on frequency of CD133+CD24+ expression in cells presented in EBs. Moreover, following isolation of CD133+CD24+ cells from EBs, we evaluated the effect of embryonic, neonatal and adult mouse kidney-derived mesenchymal stem cells (E-KMSC, N-KMSC and A-KMSC respectively) and fibronectin on further differentiation of the sorted cells. Hence, we cultured undifferentiated human embryonic stem cells (hESCs) in suspension state in the presence or absence of bFGF and determined maximum number of CD133+CD24+ cells in bFGF-treated EBs on day 7. Then, we tested the effect of E-KMSC co-culture and seeding on fibronectin-coated plated on differentiation of the sorted cells into renal epithelial cells. Results revealed down-regulation of several RP cells, markers in CD133+CD24+ cells. In contrast, renal epithelial marker gene expressions were up-regulated after 7 days of co-culture with E-KMSC. Furthermore, fibronectin resulted in higher expression of renal epithelial markers compared to the E-KMSC co-cultured cells. All in all, bFGF could enhance the number of RP cells expressing CD133 and CD24 markers, in human EBs. We suggest E-KMSC and fibronectin as a promising supplementary factor to further induce differentiation of RP cells into renal epithelial cells.


Assuntos
Diferenciação Celular , Técnicas de Cocultura/métodos , Corpos Embrioides/citologia , Células-Tronco Embrionárias Humanas/citologia , Rim/citologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Embrionárias Murinas/citologia , Animais , Biomarcadores/metabolismo , Linhagem da Célula , Autorrenovação Celular , Células Cultivadas , Corpos Embrioides/metabolismo , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Rim/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Células-Tronco Embrionárias Murinas/metabolismo
5.
Skin Pharmacol Physiol ; 33(5): 280-292, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33053562

RESUMO

The dermal papilla comprises mesenchymal cells in hair follicles, which play the main role in regulating hair growth. Maintaining the potential hair inductivity of dermal papilla cells (DPCs) and dermal sheath cells during cell culture is the main factor in in vitro morphogenesis and regeneration of hair follicles. Using common methods for the cultivation of human dermal papilla reduces the maintenance requirements of the inductive capacity of the dermal papilla and the expression of specific dermal papilla biomarkers. Optimizing culture conditions is therefore crucial for DPCs. Moreover, exosomes appear to play a key role in regulating the hair follicle growth through a paracrine mechanism and provide a functional method for treating hair loss. The present review investigated the biology of DPCs, the molecular and cell signaling mechanisms contributing to hair follicle growth in humans, the properties of the dermal papilla, and the effective techniques in maintaining hair inductivity in DPC cultures in humans as well as hair follicle bioengineering.


Assuntos
Técnicas de Cultura de Células/métodos , Derme/citologia , Derme/crescimento & desenvolvimento , Folículo Piloso/citologia , Folículo Piloso/crescimento & desenvolvimento , Derme/metabolismo , Cabelo/citologia , Cabelo/crescimento & desenvolvimento , Cabelo/metabolismo , Folículo Piloso/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Regeneração/fisiologia
6.
J Cell Physiol ; 234(2): 1534-1546, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30078203

RESUMO

BACKGROUND: The discovery of gene- and cell-based strategies has opened a new area to investigate novel approaches for the treatment of many conditions caused by cardiac cell failure. The TBX18 (T-box 18) transcription factor is considered as a prominent factor in the sinoatrial node (SAN) formation during the embryonic development. In this in vitro study, the effect of TBX18 gene expression on human-induced pluripotent-stem-cell-derived cardiomyocytes (hiPS-CMs) to induce pacemaker-like cells was examined. METHODS: The human-dermal-fibroblast-derived iPSCs were transfected using chemical, physical, and Lentiviral methods of TBX18 gene delivery during differentiation into cardiomyocytes (CMs). After the differentiation process through small-molecule-based temporal modulation of the Wnt signaling pathway, the hiPSC-CMs were analyzed using the real-time polymerase chain reaction, immunocytochemistry, immunofluorescence, whole-cell patch-clamp recording, and western blotting to investigate the accuracy of differentiation and identify the effect exerted by TBX18. RESULTS: The hiPS-CMs showed spontaneous beating and expressed specific markers of cardiac cells. The lentiviral-mediated TBX18 delivery was the most efficient method for transfection. The results showed the increment in Connexin 43 expression among untransfected hiPS-CMs, whereas this protein was significantly downregulated followed by TBX18 overexpression. TBX18-hiPSCMs were detected with pacemaker cell features. CONCLUSIONS: It was demonstrated that the TBX18 gene is able to conduct hiPSCs to differentiate into pacemaker-like cells. The TBX18 gene delivery seems to have the potential for the development of biological pacemakers; however, more investigations are still needed to assess its usefulness to fix arrhythmic conditions with SAN failure basis.


Assuntos
Potenciais de Ação , Relógios Biológicos , Diferenciação Celular , Frequência Cardíaca , Células-Tronco Pluripotentes Induzidas/metabolismo , Nó Sinoatrial/metabolismo , Proteínas com Domínio T/metabolismo , Células Cultivadas , Humanos , Fenótipo , Nó Sinoatrial/citologia , Proteínas com Domínio T/genética , Fatores de Tempo , Regulação para Cima , Via de Sinalização Wnt
7.
Biochem Biophys Res Commun ; 517(4): 684-690, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31400855

RESUMO

Worldwide, impaired wound healing leads to a large burden of morbidity and mortality. Current treatments have several limitations. Recently, nanomaterials such as copper nanoparticles (CuNPs) have attracted considerable research interest. Here, we investigated the potential therapeutic effect of various CuNPs concentrations (1 µM, 10 µM, 100 µM, 1 mM, and 10 mM) and sizes (20 nm, 40 nm, 80 nm) in wound healing. Our results revealed that the 10 µM concentration of 40 nm CuNPs and the 1 µM concentration of 80 nm CuNPs were not toxic to the cultured fibroblast, endothelial, and keratinocyte cells, and also 1 µM concentration of 80 nm CuNPs enhanced endothelial cell migration and proliferation. Extensive assessment of in vivo wound healing demonstrated that the 1 µM concentration of 80 nm CuNPs accelerated wound healing over a shorter time via formation of granulation tissue and higher new blood vessels. Importantly, serum biochemical analysis confirmed that the 40 nm CuNP (10 µM) and 80 nm CuNP (1 µM) did not show any accumulation in the liver during wound healing. Overall, our results have indicated that the 1 µM concentration of 80 nm CuNPs is a promising NP for wound healing applications without adverse side effects.


Assuntos
Movimento Celular/efeitos dos fármacos , Cobre/farmacologia , Nanopartículas Metálicas/química , Neovascularização Fisiológica/efeitos dos fármacos , Pele/citologia , Cicatrização/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Colágeno/metabolismo , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Humanos , Fígado/citologia , Nanopartículas Metálicas/ultraestrutura
8.
Cytotherapy ; 21(1): 107-112, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30409698

RESUMO

BACKGROUND: Avascular necrosis (AVN) of femoral head is a progressive bone disease due to ischemia of femoral head; patients experience pain and they can not do normal activity. There is not an effective way to treat the cause of this disease. In recent studies, treatment of this disease using pluripotent stem cell-derived mesenchyme is safe and effective, but this method needs more investigation. In this study, the safety and efficacy of CD133+ cells were evaluated as a novel method of stem cell therapy to treat AVN. METHODS: In this prospective quasi-experimental study, the participants were selected among patients with AVN who were referred to the Royan Cell Therapy Center. Autologous bone marrow-derived CD133+ cells were injected into the necrotic site of the femoral head during core decompression (CD). The Visual Analogue Scale (VAS), Harris Hip Score (HHS), Western Ontario and McMaster Universities Arthritis Index (WOMAC) and walking distance (WD) were measured before and 2, 6 and 12 months after CD. RESULTS: Overall, nine patients (six men and three women) were investigated in this study. Their mean age was 26 years old. All of them significantly improved in VAS, HHS, WOMAC and WD scores and they could do more activity without pain. Also, imaging findings demonstrated significant reductions in joint injuries. Significant complications were not seen in patients. DISCUSSION: This prospective quasi-experimental study demonstrated that, in patients with AVN, a single bone marrow-derived CD133+ cell injection into the necrotic site of the femoral head during CD is safe and effective in providing significant, clinically relevant pain relief and patients could do more activity over 2, 6 and 12 months. This pilot study suggested further clinical trials over an extended assessment period to approve bone marrow-derived CD133+ cell injection to treat AVN.


Assuntos
Antígeno AC133/metabolismo , Transplante de Medula Óssea/métodos , Descompressão Cirúrgica/métodos , Necrose da Cabeça do Fêmur/cirurgia , Transplante de Células-Tronco/métodos , Adulto , Transplante de Medula Óssea/efeitos adversos , Feminino , Seguimentos , Humanos , Imageamento por Ressonância Magnética , Masculino , Projetos Piloto , Estudos Prospectivos , Transplante de Células-Tronco/efeitos adversos , Transplante Autólogo , Resultado do Tratamento , Escala Visual Analógica , Adulto Jovem
9.
J Cell Physiol ; 234(1): 521-536, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-30071126

RESUMO

Cell death and differentiation appear to share similar cellular features. In this study, we aimed to investigate whether differentiation and mitochondrial cell death use a common pathway. We assessed the hallmarks of apoptosis during cardiomyocyte differentiation of human embryonic stem cells and found remarkable changes in P53, reactive oxygen species, apoptotic protease-activating factor 1, poly[ADP-ribose]polymerase 1, cellular adenosine triphosphate, and mitochondrial complex I activity. Furthermore, we observed reversible mitochondrial membrane permeabilization during cardiomyocyte differentiation accompanied by reversible loss of mitochondrial membrane potential, and these changes coincided with the fluctuating patterns of cytosolic cytochrome c accumulation and subsequent caspase-9 and -3/7 activation. Moreover, the use of apoptosis inhibitors (BCL2-associated X protein [BAX] inhibitor and caspase-3/7 inhibitor) during differentiation impaired cardiomyocyte development, resulting in substantial downregulation of T, MESP1, NKX2.5, and α-MHC. Additionally, although the expression of specific differentiation markers (T, MESP1, NKX2.5, MEF2C, GATA4, and SOX17) was enhanced in doxorubicin-induced human embryonic stem cells, the stemness-specific markers (OCT4 and NANOG) showed significant downregulation. With increasing doxorubicin concentration (0.03-0.6 µM; IC50 = 0.5 µM), we observed a marked increase in the expression of mesoderm and endoderm markers. In summary, we suggest that reversible mitochondrial outer membrane permeabilization promotes cardiomyocyte differentiation through an attenuated mitochondria-mediated apoptosis-like pathway.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Permeabilidade da Membrana Celular/genética , Mitocôndrias/genética , Miócitos Cardíacos/citologia , Trifosfato de Adenosina/genética , Apoptose/genética , Fator Apoptótico 1 Ativador de Proteases/genética , Caspase 9/genética , Doxorrubicina/farmacologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Humanos , Potencial da Membrana Mitocondrial/genética , Mitocôndrias/efeitos dos fármacos , Membranas Mitocondriais/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Poli(ADP-Ribose) Polimerases/genética , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/genética , Proteína X Associada a bcl-2/genética
10.
J Cell Physiol ; 233(12): 9330-9344, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29266258

RESUMO

Various somatic tissue-derived mesenchymal stromal cells (MSCs) have been considered as an attractive therapeutic tool for treatment of liver diseases in which the secretion of soluble factors or extracellular vesicles (EVs) is the most probable mechanism. The experimental application of human embryonic stem cell-derived MSC (ES-MSC) increased rapidly and showed promising results, in vitro and in vivo. However, possible therapeutic effects of human ES-MSC and their EVs on Thioacetamide (TAA)-induced chronic liver injury have not been evaluated yet. Our data indicated that human ES-MSC can significantly suppress the proliferation of peripheral blood mononuclear cells compared to bone marrow (BM)-MSC and adipose (AD)-MSC. Moreover, ES-MSC increased the secretion of anti-inflammatory cytokines (i.e., TGF-ß and IL-10) and decreased IFN-γ, compared to other MSCs. ES-MSC EVs demonstrated immunomodulatory activities comparable to parental cells and ameliorated cirrhosis in TAA-induced chronic rat liver injury, that is, reduction in fibrosis and collagen density, necrosis, caspase density, portal vein diameter, and transaminitis. The gene expression analyses also showed upregulation in collagenases (MMP9 and MMP13), anti-apoptotic gene (BCL-2) and anti-inflammatory cytokines (TGF-ß1 and IL-10) and down-regulation of major contributors to fibrosis (Col1α, αSMA, and TIMP1), pro-apoptotic gene (BAX) and pro-inflammatory cytokines (TNFα and IL-2) following treatment with ES-MSC and ES-MSC-EV. These results demonstrated that human ES-MSC and ES-MSC EV as an off-the-shelf product, that needs further assessment to be suggested as an allogeneic product for therapeutic applications for liver fibrosis.


Assuntos
Vesículas Extracelulares/metabolismo , Células-Tronco Embrionárias Humanas/metabolismo , Cirrose Hepática/terapia , Fígado/lesões , Células-Tronco Mesenquimais/metabolismo , Tecido Adiposo/citologia , Animais , Apoptose , Células da Medula Óssea/citologia , Linhagem Celular , Sobrevivência Celular , Doença Crônica , Citocinas/metabolismo , Modelos Animais de Doenças , Vesículas Extracelulares/ultraestrutura , Hepatócitos/metabolismo , Hepatócitos/patologia , Células-Tronco Embrionárias Humanas/citologia , Humanos , Imunomodulação , Fígado/patologia , Cirrose Hepática/patologia , Masculino , Células-Tronco Mesenquimais/citologia , Ratos Wistar , Tioacetamida
11.
J Cell Biochem ; 119(5): 4150-4160, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29243842

RESUMO

Exosomes are required for the regenerative effects of human cardiosphere-derived cells (CDCs). Studies show that they mimic the cardioprotective benefits of CDCs in rodents and porcine myocardial infarction (MI) models. Hypoxic preconditioning of stem cells increases the cardioprotective effects of exosomes in MI models by enhancing angiogenesis. Several exosomal microRNAs (miRNAs) up-regulate in response to hypoxia and play a role in cardioprotective and pro-angiogenic effects. In this study, we have demonstrated that human CDCs secreted exosomes under hypoxic conditions (1% O2 for 2 days) enhanced tube formation by human umbilical vein endothelial cells (HUVECs) at a concentration of 25 µg/mL. Pro-angiogenic exosomal miRNAs including miR-126, miR-130a, and miR-210 showed a substantial increase (>2-, >2-, and >4-fold, respectively) in the hypoxic exosomes compared to normoxic CDC-derived exosomes. Our study suggested a significant benefit of hypoxic CDC exosomes for the treatment of cardiac diseases by induction of angiogenesis via enrichment of pro-angiogenic exosomal miRNAs.


Assuntos
Exossomos/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , MicroRNAs/metabolismo , Miócitos Cardíacos/metabolismo , Neovascularização Fisiológica , Esferoides Celulares/metabolismo , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Miócitos Cardíacos/citologia , Esferoides Celulares/citologia
12.
Cytotherapy ; 20(10): 1238-1246, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30318332

RESUMO

BACKGROUND: The intra-articular implantation of mesenchymal stromal cells (MSCs) as a treatment for knee osteoarthritis (OA) is an emerging new therapy. In this study, patients with knee OA received intra-articular implantations of autologous bone marrow-derived MSCs. We sought to assess the safety and efficacy of this implantation. MATERIALS AND METHODS: This was a phase 1/2 single-center, triple-blind, randomized controlled trial (RCT) with a placebo control. The subjects consisted of patients with knee OA randomly assigned to either an intra-articular implantation of MSCs (40 × 106 cells) or 5 mL normal saline (placebo). Patients were followed up for 6 months after the implantations. The pain level and function improvements for patient-reported outcomes were assessed based on a visual analog scale (VAS), Western Ontario and McMaster Universities Arthritis Index (WOMAC) and its subscales, walking distance, painless walking distance, standing time and knee flexion compared with the placebo group at 3 and 6 months following the implantations. RESULTS: Overall, 43 patients (Kellgren-Lawrence grades 2, 3 and 4) were assigned to either the MSCs (n = 19) or placebo (n = 24) group. Patients who received MSCs experienced significantly greater improvements in WOMAC total score, WOMAC pain and physical function subscales and painless walking distance compared with patients who received placebo. There were no major adverse events attributed to the MSC therapy. CONCLUSION: This randomized, triple-blind, placebo-controlled RCT demonstrated the safety and efficacy of a single intra-articular implantation of 40 × 106 autologous MSCs in patients with knee OA. Intra-articular implantation of MSCs provided significant and clinically relevant pain relief over 6 months versus placebo and could be considered a promising novel treatment for knee OA. We propose that further investigations should be conducted over an extended assessment period and with a larger cohort.


Assuntos
Transplante de Células-Tronco Mesenquimais/métodos , Osteoartrite do Joelho/terapia , Adulto , Medula Óssea , Feminino , Humanos , Injeções Intra-Articulares , Masculino , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Pessoa de Meia-Idade , Placebos , Transplante Autólogo/métodos , Resultado do Tratamento , Escala Visual Analógica , Caminhada
13.
Cytotherapy ; 20(5): 660-669, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29580865

RESUMO

BACKGROUND: Chronic kidney disease (CKD) is a progressive loss of kidney function and structure that affects approximately 13% of the population worldwide. A recent meta-analysis revealed that cell-based therapies improve impaired renal function and structure in preclinical models of CKD. We assessed the safety and tolerability of bone marrow-mesenchymal stromal cell (MSC) infusion in patients with CKD. METHODS: A single-arm study was carried out at one center with 18-month follow-up in seven eligible patients with CKD due to different etiologies such as hypertension, nephrotic syndrome (NS) and unknown etiology. We administered an intravenous infusion (1-2 × 106 cells/kg) of autologous cultured MSCs. The primary endpoint was safety, which was measured by number and severity of adverse events. The secondary endpoint was decrease in the rate of decrease in estimated glomerular filtration rate (eGFR). We compared kidney function during the follow-up visits to baseline and 18 months prior to the intervention. RESULTS: Follow-up visits of all seven patients were completed; however, we have not observed any cell-related adverse events during the trial. Changes in eGFR (P = 0.10) and serum creatinine (P = 0.24) from 18 months before cell infusion to baseline in comparison with baseline to 18 months were not statistically significant. CONCLUSIONS: We showed safety and tolerability of a single-dose infusion of autologous MSCs in patients with CKD.


Assuntos
Células da Medula Óssea/citologia , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Células-Tronco Mesenquimais/citologia , Insuficiência Renal Crônica/terapia , Adulto , Determinação de Ponto Final , Feminino , Seguimentos , Taxa de Filtração Glomerular , Humanos , Rim/fisiopatologia , Masculino , Pessoa de Meia-Idade , Insuficiência Renal Crônica/fisiopatologia
14.
Cytotherapy ; 20(4): 499-506, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29428486

RESUMO

BACKGROUND: In this study, we intend to assess the safety and tolerability of intra-articular knee implantation of autologous bone marrow-derived mesenchymal stromal cells (MSCs) in patients with rheumatoid arthritis (RA) and to determine the preliminary clinical efficacy data in this population. The trial registration numbers are as follows: Royan Institute Ethics Committee: AC/91/1133; NCT01873625. METHODS: This single-center, randomized, triple-blind, placebo-controlled phase 1/2 clinical trial randomized RA patients with knee involvement to receive either an intra-articular knee implantation of 40 million autologous bone marrow-derived MSCs per joint or normal saline (placebo). Patients were followed up for 12 months to assess therapy outcomes. RESULTS: A total of 30 patients, 15 in the MSC group and 15 in the placebo group, enrolled in this study. There were no adverse effects reported after MSC administration or during follow-up. Patients who received MSCs had superior findings according to the Western Ontario and McMaster Universities Arthritis Index (WOMAC), visual analogue scale (VAS), time to jelling and pain-free walking distance. However, this improvement could not be significantly sustained beyond 12 months. The MSC group exhibited improved standing time (P = 0.01). In addition, the MSCs appeared to contribute to reductions in methotrexate and prednisolone use. CONCLUSION: Intra-articular knee implantation of MSCs appeared to be safe and well tolerated. In addition, we observed a trend toward clinical efficacy. These results, in our opinion, have justified the need for further investigations over an extended assessment period with larger numbers of RA patients who have knee involvement.


Assuntos
Artrite Reumatoide/terapia , Células da Medula Óssea/citologia , Transplante de Medula Óssea/métodos , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Osteoartrite do Joelho/terapia , Adulto , Idoso , Artrite Reumatoide/complicações , Método Duplo-Cego , Feminino , Humanos , Injeções Intra-Articulares , Articulação do Joelho , Masculino , Pessoa de Meia-Idade , Osteoartrite do Joelho/complicações , Placebos , Resultado do Tratamento
15.
Kidney Blood Press Res ; 43(2): 471-478, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29590654

RESUMO

BACKGROUND/AIMS: Autosomal dominant polycystic kidney disease (ADPKD) is one of the most common inherited cystic kidney diseases caused by mutations in two large multi-exon genes, PKD1 and PKD2. High allelic heterogeneity and duplication of PKD1 exons 1-32 as six pseudo genes on chromosome 16 complicate molecular analysis of this disease. METHODS: We applied targeted next-generation sequencing (NGS) in 9 non-consanguineous unrelated Iranian families with ADPKD to identify the genes hosting disease-causing mutations. This approach was confirmed by Sanger sequencing. RESULTS: Here, we determined three different novel frameshift mutations and four previously reported nonsense mutations in the PKD1 gene encoding polycystin1 in heterozygotes. CONCLUSION: This study demonstrates the effectiveness of NGS in significantly reducing the cost and time for simultaneous sequence analysis of PKD1 and PKD2, simplifying the genetic diagnostics of ADPKD. Although a probable correlation between the mutation types and phenotypic outcome is possible, however for more extensive studies in future, the consideration of renal hypouricemia (RHUC) and PKD1 coexistence may be helpful. The novel frameshift mutations reported by this study are p. Q1997X, P. D73X and p. V336X.


Assuntos
Mutação da Fase de Leitura , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Rim Policístico Autossômico Dominante/genética , Canais de Cátion TRPP/genética , Éxons , Família , Feminino , Mutação da Fase de Leitura/genética , Sequenciamento de Nucleotídeos em Larga Escala/economia , Humanos , Irã (Geográfico) , Masculino , Linhagem , Erros Inatos do Transporte Tubular Renal/genética , Fatores de Tempo , Cálculos Urinários/genética
16.
Clin Lab ; 63(7): 1261-1267, 2017 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-28792715

RESUMO

BACKGROUND: Autosomal Dominant Polycystic Kidney Disease (ADPKD) is the fourth most frequent cause of endstage renal disease (ESDR), occurring at a varying frequency of 1/400 to 1/800 persons. The disease affects all ethnic groups worldwide, and there is a need for population based studies to be carried out for better diagnostic, genetic counselling, and treatment purpose. METHODS: Eighteen unrelated probands (10 males and 8 females) with a familial history of ADPKD were selected for the study. Their clinical evaluation was performed to diagnose and assess disease progression. PKD1 and PKD2 genes were genotyped in each proband by next generation sequencing (NGS). RESULTS: Mutational analysis of PKD1 and PKD2 genes using NGS in eighteen unrelated Iranian ADPKD families revealed a total of eighteen heterozygous variations. PKD1 genotype revealed eight frameshift deletion mutations, two frameshift insertion mutations, five nonsense mutations and one splice mutation and PKD2 showed one frameshift deletion mutation and one frameshift insertion mutation. Four of the variants reported were novel and were present in the PKD1 gene. Further, PKD1 truncating mutations reached ESRD earlier than patients with non-truncating PKD1 mutations (52 ± 3.2 years vs. 58 ± 10.8 years, p = 0.01). CONCLUSIONS: The PKD1 and PKD2 genotyping of ADPKD Iranian patients with familial history showed no mutational hotspot. The screening has given four novel variants that will contribute to diagnosis, genetic counselling, and treatment of ADPKD patients in general.


Assuntos
Rim Policístico Autossômico Dominante/genética , Canais de Cátion TRPP/genética , Adulto , Idoso , Análise Mutacional de DNA , Feminino , Humanos , Irã (Geográfico) , Masculino , Pessoa de Meia-Idade , Mutação
18.
Cytokine ; 85: 51-60, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27288632

RESUMO

Mesenchymal stem cells (MSCs), as cells with potential clinical utilities, have demonstrated preferential incorporation into inflammation sites. Immunophenotype and immunomodulatory functions of MSCs could alter by inflamed-microenvironments due to the local pro-inflammatory cytokine milieu. A major cellular mediator with specific function in promoting inflammation and pathogenicity of autoimmunity are IL-17-producing T helper 17 (Th17) cells that polarize in inflamed sites in the presence of pro-inflammatory cytokines such as Interleukin-1ß (IL-1ß), IL-6 and IL-23. Since MSCs are promising candidate for cell-based therapeutic strategies in inflammatory and autoimmune diseases, Th17 cell polarizing factors may alter MSCs phenotype and function. In this study, human bone-marrow-derived MSCs (BM-MSC) and adipose tissue-derived MSCs (AD-MSC) were cultured with or without IL-1ß, IL-6 and IL-23 as pro-inflammatory cytokines. The surface markers and their differentiation capacity were measured in cytokine-untreated and cytokine-treated MSCs. MSCs-mediated immunomodulation was analyzed by their regulatory effects on mixed lymphocyte reaction (MLR) and the level of IL-10, TGF-ß, IL-4, IFN-γ and TNF-α production as immunomodulatory cytokines. Pro-inflammatory cytokines showed no effect on MSCs morphology, immunophenotype and co-stimulatory molecules except up-regulation of CD45. Adipogenic and osteogenic differentiation capacity increased in CD45+ MSCs. Moreover, cytokine-treated MSCs preserved the suppressive ability of allogeneic T cell proliferation and produced higher level of TGF-ß and lower level of IL-4. We concluded pro-inflammatory cytokines up-regulate the efficacy of MSCs in cell-based therapy of degenerative, inflammatory and autoimmune disorders.


Assuntos
Diferenciação Celular/imunologia , Citocinas/imunologia , Imunomodulação/imunologia , Inflamação/imunologia , Células-Tronco Mesenquimais/imunologia , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Biomarcadores/metabolismo , Células Cultivadas , Humanos , Imunofenotipagem/métodos , Inflamação/metabolismo , Antígenos Comuns de Leucócito/imunologia , Teste de Cultura Mista de Linfócitos/métodos , Linfócitos/imunologia , Linfócitos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Células Th17/imunologia , Células Th17/metabolismo , Regulação para Cima/imunologia
19.
Int J Urol ; 23(7): 581-6, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27062371

RESUMO

OBJECTIVES: To evaluate the effect of autologous muscle-derived cells injection in the treatment of complicated stress urinary incontinence in female patients. METHODS: Female patients presenting with severe and complicated stress urinary incontinence secondary to the bladder neck and/or urethral trauma or congenital epispadias (with or without exstrophy) were enrolled in this prospective study. They underwent transurethral injection of autologous muscle-derived cells. In selected cases, another injection was given after 6 months, as per the surgeon's assessment. All patients were monitored for 1 year, and the effect of autologous muscle-derived cells was evaluated by cough stress test, 1-h pad test and Incontinence Impact Questionnaire-short form score. A multichannel urodynamic study and maximum urethral closure pressure were carried out before and 12 months after the last treatment session. Cough stress test, 1-h pad test and uroflowmetry were repeated 36 months after the last injection. Severity and occurrence of complications were recorded at each visit. RESULTS: All 10 patients who completed the study were monitored for 36 months. Three patients were cured, four had improved and three did not respond to the treatment. There was no major adverse effect related to the treatment. CONCLUSIONS: Muscle-derived cell therapy might represent a minimally-invasive and a safe procedure in the treatment of patients with severe and complicated stress urinary incontinence.


Assuntos
Epispadia/complicações , Transplante de Células-Tronco , Incontinência Urinária por Estresse/terapia , Feminino , Humanos , Masculino , Músculos/citologia , Estudos Prospectivos , Resultado do Tratamento , Uretra , Incontinência Urinária , Urodinâmica
20.
Biochem Biophys Res Commun ; 463(4): 699-705, 2015 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-26047705

RESUMO

Recent advances in the direct conversion of fibroblasts to cardiomyocytes suggest this process as a novel promising approach for cardiac cell-based therapies. Here, by screening the effects of 10 candidate small molecules along with transient overexpression of Yamanaka factors, we show ascorbic acid (AA), also known as vitamin C, enhances reprogramming of mouse fibroblasts into beating cardiomyocytes. Immunostaining and gene expression analyses for pluripotency and cardiac lineage markers confirmed beating patches were derived from non-cardiac lineage cells without passing through a pluripotent intermediate. Further analysis revealed that AA also increased the size of the beating areas and the number of cardiac progenitors. Immunostaining for cardiac markers, as well as electrophysiological analysis confirmed the functionality of directly converted cardiomyocytes. These results illustrate the importance of AA in direct conversion of fibroblasts to cardiomyocytes and may open new insights into future biomedical applications for induced cardiomyocytes.


Assuntos
Ácido Ascórbico/farmacologia , Diferenciação Celular/efeitos dos fármacos , Miócitos Cardíacos/citologia , Animais , Células Cultivadas , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA