Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Drug Resist Updat ; 42: 35-45, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30877905

RESUMEN

Glioblastoma multiforme (GBM) is among the most incurable cancers. GBMs survival rate has not markedly improved, despite new radical surgery protocols, the introduction of new anticancer drugs, new treatment protocols, and advances in radiation techniques. The low efficacy of therapy, and short interval between remission and recurrence, could be attributed to the resistance of a small fraction of tumorigenic cells to treatment. The existence and importance of cancer stem cells (CSCs) is perceived by some as controversial. Experimental evidences suggest that the presence of therapy-resistant glioblastoma stem cells (GSCs) could explain tumor recurrence and metastasis. Some scientists, including most of the authors of this review, believe that GSCs are the driving force behind GBM relapses, whereas others however, question the existence of GSCs. Evidence has accumulated indicating that non-tumorigenic cancer cells with high heterogeneity, could undergo reprogramming and become GSCs. Hence, targeting GSCs as the "root cells" initiating malignancy has been proposed to eradicate this devastating disease. Most standard treatments fail to completely eradicate GSCs, which can then cause the recurrence of the disease. To effectively target GSCs, a comprehensive understanding of the biology of GSCs as well as the mechanisms by which these cells survive during treatment and develop into new tumor, is urgently needed. Herein, we provide an overview of the molecular features of GSCs, and elaborate how to facilitate their detection and efficient targeting for therapeutic interventions. We also discuss GBM classifications based on the molecular stem cell subtypes with a focus on potential therapeutic approaches.


Asunto(s)
Antineoplásicos/uso terapéutico , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Humanos , Metástasis de la Neoplasia/tratamiento farmacológico , Metástasis de la Neoplasia/patología , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/patología
2.
Int J Mol Sci ; 21(7)2020 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-32218162

RESUMEN

Natural killer (NK) cell therapy is one of the most promising treatments for Glioblastoma Multiforme (GBM). However, this emerging technology is limited by the availability of sufficient numbers of fully functional cells. Here, we investigated the efficacy of NK cells that were expanded and treated by interleukin-2 (IL-2) and heat shock protein 70 (HSP70), both in vitro and in vivo. Proliferation and cytotoxicity assays were used to assess the functionality of NK cells in vitro, after which treated and naïve NK cells were administrated intracranially and systemically to compare the potential antitumor activities in our in vivo rat GBM models. In vitro assays provided strong evidence of NK cell efficacy against C6 tumor cells. In vivo tracking of NK cells showed efficient homing around and within the tumor site. Furthermore, significant amelioration of the tumor in rats treated with HSP70/Il-2-treated NK cells as compared to those subjected to nontreated NK cells, as confirmed by MRI, proved the efficacy of adoptive NK cell therapy. Moreover, results obtained with systemic injection confirmed migration of activated NK cells over the blood brain barrier and subsequent targeting of GBM tumor cells. Our data suggest that administration of HSP70/Il-2-treated NK cells may be a promising therapeutic approach to be considered in the treatment of GBM.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Glioblastoma/patología , Proteínas HSP70 de Choque Térmico/farmacología , Interleucina-2/farmacología , Animales , Línea Celular Tumoral , Técnicas de Cocultivo , Glioblastoma/metabolismo , Inmunofenotipificación , Células Asesinas Naturales/inmunología , Masculino , Ratas
3.
J Cell Physiol ; 234(6): 8249-8258, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30378105

RESUMEN

Mesenchymal stromal cells (MSCs) can effectively contribute to tissue regeneration inside the inflammatory microenvironment mostly through modulating immune responses. MSC-derived extracellular vesicles (MSC-EVs) display immunoregulatory functions similar to parent cells. Interactions between MSC-EVs and immune cells make them an ideal therapeutic candidate for infectious, inflammatory, and autoimmune diseases. These properties of MSC-EVs have encouraged researchers to perform extensive studies on multiple factors that mediate MSC-EVs immunomodulatory effects. Investigation of proteins involved in the complex interplay of MSC-EVs and immune cells may help us to better understand their functions. Here, we performed a comprehensive proteomic analysis of MSC-EVs that was previously reported by ExoCarta database. A total of 938 proteins were identified as MSC-EV proteome using quantitative proteomics techniques. Kyoto Encyclopedia of Genes and Genomes analysis demonstrates that ECM-receptor interaction, focal adhesion, and disease-specific pathways are enriched in MSC-EVs. By detail analysis of proteins presence in immune system process, we found that expression of some cytokines, chemokines, and chemokine receptors such as IL10, HGF, LIF, CCL2, VEGFC, and CCL20, which leads to migration of MSC-EVs to injured sites, suppression of inflammation and promotion of regeneration in inflammatory and autoimmune diseases. Also, some chemoattractant proteins such as CXCL2, CXCL8, CXCL16, DEFA1, HERC5, and IFITM2 were found in MSC-EV proteome. They may actively recruit immune cells to the proximity of MSC or MSC-EVs, may result in boosting immune response under specific circumstances, and may have protective role in infectious diseases. In this review, we summarize available information about immunomodulation of MSC-EVs with particular emphasis on their proteomics analysis.


Asunto(s)
Vesículas Extracelulares/genética , Inmunidad Celular/genética , Inflamación/genética , Diferenciación Celular/genética , Quimiocinas/genética , Citocinas/genética , Vesículas Extracelulares/metabolismo , Regulación de la Expresión Génica/genética , Humanos , Inmunidad Celular/inmunología , Inmunomodulación/genética , Inflamación/metabolismo , Células Madre Mesenquimatosas/metabolismo , Proteómica , Receptores de Quimiocina/genética
4.
J Cell Physiol ; 234(6): 9564-9576, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30362564

RESUMEN

Organoids can be regarded as a beneficial tool for discovery of new therapeutics for diabetes and/or maturation of pancreatic progenitors (PP) towards ß cells. Here, we devised a strategy to enhance maturation of PP by assembly of three-dimensional (3D) pancreatic organoids (PO) containing human embryonic stem (ES) cell derivatives including ES-derived pancreatic duodenal homeobox 1 (PDX1) + early PP, mesenchymal stem cells, and endothelial cells at an optimized cell ratio, on Matrigel. The PO was placed in a 3D-printed tissue trapper and heterotopically implanted into the peritoneal cavity of immunodeficient mice where it remained for 90 days. Our results indicated that, in contrast to corresponding early PP transplants, 3D PO developed more vascularization as indicated by greater area and number of vessels, a higher number of insulin-positive cells and improvement of human C-peptide secretions. Based on our findings, PO-derived ß cells could be considered a novel strategy to study human ß-cell development, novel therapeutics, and regenerative medicine for diabetes.


Asunto(s)
Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/trasplante , Organoides/citología , Páncreas/citología , Impresión Tridimensional , Ingeniería de Tejidos , Animales , Diferenciación Celular , Línea Celular , Células Endoteliales/citología , Humanos , Células Secretoras de Insulina/citología , Células Madre Mesenquimatosas/citología , Ratones , Organoides/trasplante , Cavidad Peritoneal/citología , Trasplante Heterotópico
5.
J Cell Physiol ; 234(12): 22493-22504, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31120149

RESUMEN

Glioblastoma multiforme (GBM) is a unique aggressive tumor and mostly develops in the brain, while rarely spreading out of the central nervous system. It is associated with a high mortality rate; despite tremendous efforts having been made for effective therapy, tumor recurrence occurs with high prevalence. To elucidate the mechanisms that lead to new drug discovery, animal models of tumor progression is one of the oldest and most beneficial approaches to not only investigating the aggressive nature of the tumor, but also improving preclinical research. It is also a useful tool for predicting novel therapies' effectiveness as well as side effects. However, there are concerns that must be considered, such as the heterogeneity of tumor, biological properties, pharma dynamic, and anatomic shapes of the models, which have to be similar to humans as much as possible. Although several methods and various species have been used for this approach, the real recapitulation of the human tumor has been left under discussion. The GBM model, which has been verified in this study, has been established by using the Rat C6 cell line. By exploiting bioinformatic tools, the similarities between aberrant gene expression and pathways have been predicted. In this regard, 610 common genes and a number of pathways have been detected. Moreover, while magnetic resonance imaging analysis enables us to compare tumor features between these two specious, pathological findings provides most of the human GBM characteristics. Therefore, the present study provides genomics, pathologic, and imaging evidence for showing the similarities between human and rat GBM models.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/fisiología , Genómica , Glioblastoma/genética , Glioblastoma/patología , Animales , Línea Celular Tumoral , Biología Computacional , Regulación hacia Abajo , Humanos , Neoplasias Experimentales , Mapas de Interacción de Proteínas , Ratas , Especificidad de la Especie , Transcriptoma , Regulación hacia Arriba
6.
J Cell Physiol ; 233(12): 9330-9344, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29266258

RESUMEN

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.


Asunto(s)
Vesículas Extracelulares/metabolismo , Células Madre Embrionarias Humanas/metabolismo , Cirrosis Hepática/terapia , Hígado/lesiones , Células Madre Mesenquimatosas/metabolismo , Tejido Adiposo/citología , Animales , Apoptosis , Células de la Médula Ósea/citología , Línea Celular , Supervivencia Celular , Enfermedad Crónica , Citocinas/metabolismo , Modelos Animales de Enfermedad , Vesículas Extracelulares/ultraestructura , Hepatocitos/metabolismo , Hepatocitos/patología , Células Madre Embrionarias Humanas/citología , Humanos , Inmunomodulación , Hígado/patología , Cirrosis Hepática/patología , Masculino , Células Madre Mesenquimatosas/citología , Ratas Wistar , Tioacetamida
7.
J Cell Biochem ; 119(10): 8048-8073, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29377241

RESUMEN

Mesenchymal stem cells (MSCs) are adult multipotent cells that due to their ability to homing to damaged tissues and differentiate into specialized cells, are remarkable cells in the field of regenerative medicine. It's suggested that the predominant mechanism of MSCs in tissue repair might be related to their paracrine activity. The utilization of MSCs for tissue repair is initially based on the differentiation ability of these cells; however now it has been revealed that only a small fraction of the transplanted MSCs actually fuse and survive in host tissues. Indeed, MSCs supply the microenvironment with the secretion of soluble trophic factors, survival signals and the release of extracellular vesicles (EVs) such as exosome. Also, the paracrine activity of EVs could mediate the cellular communication to induce cell-differentiation/self-renewal. Recent findings suggest that EVs released by MSCs may also be critical in the physiological function of these cells. This review provides an overview of MSC-derived extracellular vesicles as a hopeful opportunity to advance novel cell-free therapy strategies that might prevail over the obstacles and risks associated with the use of native or engineered stem cells. EVs are very stable; they can pass the biological barriers without rejection and can shuttle bioactive molecules from one cell to another, causing the exchange of genetic information and reprogramming of the recipient cells. Moreover, extracellular vesicles may provide therapeutic cargo for a wide range of diseases and cancer therapy.


Asunto(s)
Vesículas Extracelulares/metabolismo , Células Madre Mesenquimatosas/citología , Medicina Regenerativa/métodos , Animales , Comunicación Celular/fisiología , Humanos
8.
Cytotherapy ; 20(4): 499-506, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29428486

RESUMEN

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.


Asunto(s)
Artritis Reumatoide/terapia , Células de la Médula Ósea/citología , Trasplante de Médula Ósea/métodos , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Osteoartritis de la Rodilla/terapia , Adulto , Anciano , Artritis Reumatoide/complicaciones , Método Doble Ciego , Femenino , Humanos , Inyecciones Intraarticulares , Articulación de la Rodilla , Masculino , Persona de Mediana Edad , Osteoartritis de la Rodilla/complicaciones , Placebos , Resultado del Tratamiento
9.
Cytotherapy ; 17(2): 232-41, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25593079

RESUMEN

BACKGROUND AIMS: Recent studies have proposed that cellular transplantation may have some regenerative and functional efficacy in the treatment of cerebral palsy (CP); however, much remains to be understood regarding its safety, feasibility and efficacy. This study was initiated to evaluate the safety of autologous bone marrow-derived CD133(+) cell intrathecal injection. METHODS: Children (n = 12), aged 4 to 12 years, who were diagnosed with different types of CP underwent BM aspiration. CD133(+) cells were enriched from the BM samples and intrathecally injected. The Gross Motor Function Measure (GMFM-66), Gross Motor Function Classification System (GMFCS), UK FIM+FAM, Functional Independence Measure (FIM) and Functional Assessment Measure (FAM) were assessed at baseline and 6 months after the procedure. Patients' ability to balance was measured by the Berg Balance Scale (BBS), and severity of spasticity was evaluated by the Modified Ashworth Scale. Magnetic resonance imaging was done at baseline and 6 months after therapy. This study was registered in ClinicalTrials.gov (NCT01404663). RESULTS: There were no adverse events detected by clinical and laboratory tests or imaging studies, with the exception of a seizure in 1 patient. A significant improvement was observed 6 months after cell transplantation versus baseline according to GMFM, GMFCS, FIM+FAM, Ashworth Scale, and BBS outcomes. CONCLUSIONS: Subarachnoid injection of CD133-positive enriched bone marrow progenitor cells in children with CP is a safe approach. The results suggest a possible short-term improvement in neurological function.


Asunto(s)
Antígenos CD/metabolismo , Parálisis Cerebral/terapia , Glicoproteínas/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Destreza Motora/fisiología , Espasticidad Muscular/terapia , Péptidos/metabolismo , Antígeno AC133 , Células de la Médula Ósea , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Niño , Preescolar , Femenino , Células Madre Hematopoyéticas , Humanos , Inyecciones Espinales , Masculino , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Células Madre Mesenquimatosas/citología , Seguridad
10.
J Craniofac Surg ; 24(4): 1292-7, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23851791

RESUMEN

Mandibular continuity defects occur after tumor resection, maxillofacial injury, or osteomyelitis. In this clinical pilot study, we report a novel method for reconstruction of mandibular continuity defect by in vivo tissue engineering. In 3 patients with critical-size mandibular bone defects, the allogenic mandibular bone scaffold was customized, loaded by ex vivo expanded mesenchymal stem cells, and transplanted into the surgical defect site. According to the bone scintigraphy, vascularized bone was identified in 2 cases. In spiral computed tomography, normal bone healing without significant bone resorption was seen at the 2 viable grafts, but at the failed construction, there was a lack of osteointegration to the adjacent host bone and a higher density in the medullary bone. According to the serial panoramic imaging, the patients with viable bone grafts had normal bone healing, whereas the other patient had progressive overall bone resorption. Our results demonstrate the feasibility of allogenic bone scaffold loaded by mesenchymal stem cells in the reconstruction of mandibular continuity defects. Although long-term results are not yet available, it may be a novel method of reconstruction and a basis for further studies.


Asunto(s)
Trasplante Óseo/métodos , Enfermedades Mandibulares/cirugía , Trasplante de Células Madre Mesenquimatosas/métodos , Procedimientos de Cirugía Plástica/métodos , Ingeniería de Tejidos/métodos , Andamios del Tejido , Adolescente , Adulto , Aloinjertos/trasplante , Densidad Ósea/fisiología , Resorción Ósea/etiología , Separación Celular/métodos , Estudios de Factibilidad , Femenino , Supervivencia de Injerto , Humanos , Masculino , Reconstrucción Mandibular/métodos , Oseointegración/fisiología , Proyectos Piloto , Radiografía Panorámica/métodos , Tomografía Computarizada Espiral/métodos , Cicatrización de Heridas/fisiología
11.
Cell J ; 25(6): 363-371, 2023 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-37434453

RESUMEN

OBJECTIVE: Mesenchymal stromal cells (MSCs) play immunomodulatory role in various autoimmune diseases. Previous pre-clinical and clinical studies have shown that MSCs could be a therapeutic modality for psoriasis. However, the mechanisms of treatment and its possible side effects are under investigation. In this study, the safety and probable efficacy of injecting allogeneic adipose-derived mesenchymal stromal cells (ADSCs) in psoriatic patients were evaluated. MATERIALS AND METHODS: In this phase I clinical study with six months of follow-up, total number of 1×106 or 3×106 cells/cm2 of ADSCs were injected into the subcutaneous tissue of each plaque as a single dose in three males and two females (3M/2F) with a mean age of 32.8 ± 8.18. The primary outcome was safety. Changes in clinical and histological indexes, the number of B and T lymphocytes in local and peripheral blood, and serum levels of inflammatory cytokines were assessed. Paired t test was used to compare variables at two time points (baseline and six months after injection) and repeated measures ANOVA test was utilized for variables at three time points in follow-up visits. RESULTS: No major adverse effects such as burning, pain, itching, or any systemic side effects were observed following ADSCs injection, and the lesions showed slight to considerable improvement after injection. The mRNA expression levels of pro-inflammatory factors were reduced in the dermis of the patients after injection. The increased expression level of Foxp3 transcription factor in the patient blood samples suggested modulation of inflammation after ADMSCs administration. Six months after the intervention, no major side effects were reported, but skin thickness, erythema, and scaling of the plaques, as well as the PASI score, were decreased in majority of patients. CONCLUSION: Our study suggested that ADSC injection could be considered as a safe and effective therapeutic approach for psoriatic plaques (registration number: IRCT20080728001031N24).

12.
Mult Scler Relat Disord ; 78: 104895, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37515913

RESUMEN

Multiple sclerosis (MS) is a progressive, demyelinating neurodegenerative disease of the central nervous system. MS is immune-mediated and leads to disability especially in young adults. Even though 18 MS therapy drugs were approved, they slightly inhibit disease progression and do not induce regeneration and repair in the nervous system. Mesenchymal stromal cells (MSCs) have emerged as a new therapeutic modality in regenerative medicine and tissue engineering due to their immunomodulation and bio regenerative properties. We have designed a randomized, controlled clinical trial to assess safety and possible efficacy of MSC application in MS patients. Twenty-one MS patients were enrolled. Patients were allocated in two distinct groups: treatment group, which received systemic transplantation of autologous bone marrow-derived MSCs, and control group, which received placebo at the first injections. Patients in control group received MSCs at the second injection while the treatment group received placebo. All the patients were followed for 18 months. Follow-ups included regular visits, laboratory evaluation, and imaging analysis. Control patients received MSCs six month after treatment group. No severe immediate or late adverse events were observed in both groups after interventions. We did not find any significant differences in the rate of relapses, Expanded Disability Status Scale (EDSS) score, cognitive condition, Magnetic Resonance Imaging (MRI) findings, or any biomarkers of cerebrospinal fluid between the two groups and in each group before and after cell infusion. Transplantation of autologous bone marrow-derived mesenchymal stromal cells is safe and feasible. The efficacy of transplantation of these cells should be evaluated through designing randomized clinical trials with larger sample sizes, different administration routes, other cell types (allogeneic adipose derived MSCs, allogeneic Wharton's jelly derived MSCs …), repeated injections, and longer follow-up periods.

13.
Cell J ; 24(2): 62-68, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35279961

RESUMEN

Objective: Perianal fistulas in Crohn's disease (CD) are the main challenges in inflammatory bowel diseases (IBDs). Some of the fistulas are refractory to any therapeutic strategy. The aim of this study was to evaluate the therapeutic effects of mesenchymal stromal cells (MSCs) as a novel promising modality for the treatment of fistulizing CD. Materials and Methods: This case series clinical interventional study was conducted from 2014 to 2017 at Shariati Hospital, an IBD referral center in Tehran, Iran. Refractory adult patients with CD who had draining perianal fistulas were enrolled in this study. All patients were examined by a colorectal surgeon and the fistula imaging studies were performed by pelvic magnetic resonance imaging (MRI). After autologous bone marrow (BM) aspiration and MSCs isolation, the cells were cultured and passaged under current good manufacturing practice (cGMP) conditions. Four intra-fistula injections of cells, each containing 40×106 MSCs suspended in fibrin glue, were administered by an expert surgeon every 4 weeks. Procedure safety, feasibility and closure of the perianal fistulas at week 24 were assessed. Clinical examination and MRI findings were considered as the primary end points. Results: In total, 5 patients (2 males and 3 females) were enrolled in this study. No adverse events were observed during the six-month follow-up in these patients. Both the Crohn's Disease Activity Index (CDAI) and Perianal Disease Activity Index (PDAI) scores decreased in all patients after cell injections and one patient achieved complete remission with closure of fistulas, discontinuation of fistula discharge, and closure of the external opening. Conclusion: Local injection of MSCs combined with fibrin glue is potentially a safe and effective therapeutic approach for complex perianal fistulas in patients with CD.

14.
Cancer Lett ; 501: 200-209, 2021 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-33220334

RESUMEN

Despite recent advances in cancer immunotherapy, there have been limitations in cancer treatment and patient survival due to a lack of antigen recognition and immunosuppressive tumor microenvironment. To overcome this issue, we have shown that miRNA modified tumor-derived Extracellular Vesicles (mt-EVs) would be an advantageous prospect since they are tumor specific and associated antigen sources which cause increase in maturation and antigen-presenting function of dendritic cells. Also, miRNAs are promising candidates for cancer therapy because of their ability to control several host immune subsets to respond against cancer cells as well as tumor microenvironment remodeling. Here, we report that mt-EVs containing tumor specific antigens loaded with miRNAs (Let-7i, miR-142 and, miR-155) could increase the survival rate of tumor-bearing mice and induce reduction in tumor growth. Importantly, the administration of mt-EVs elicited cytotoxic T cells with increasing in IFNγ and Granzyme B production ability. Notably, intramuscular (IM) injection of let7i, miR142-EVs had a significant effect on dendritic cell (DC) maturation and T cell activation along with tumor shrinkage. Collectively, our findings suggest that administration of miRNA containing EVs may be effective immunotherapy against solid tumors.


Asunto(s)
Vesículas Extracelulares/trasplante , Neoplasias Mamarias Experimentales/terapia , MicroARNs/genética , Animales , Antígenos de Neoplasias/genética , Células Dendríticas/metabolismo , Vesículas Extracelulares/genética , Femenino , Granzimas/metabolismo , Inyecciones Intramusculares , Interferón gamma/metabolismo , Activación de Linfocitos , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/inmunología , Ratones , Linfocitos T/inmunología , Microambiente Tumoral
15.
Cell J ; 23(7): 772-778, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34979067

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder with very limited treatment options. Stem cells have been raised as a new treatment modality for these patients. We have designed a single-center, prospective, open-label, and single arm clinical trial to assess the safety, feasibility, and rather efficacy of administrating allogeneic adipose-derived mesenchymal stromal cells (Ad-MSCs) in ALS patients. We enrolled 17 patients with confirmed ALS diagnosis with ALS Functional Rating Scale-Revised (ALSFRS-R) ≥24 and predicted forced vital capacity (FVC) ≥40%. Allogeneic Ad-MSCs were transplanted intravenously for all patients. Follow-ups were done at 24 hours, 2, 4, 6, and 12 months after cell infusion by checking adverse events, laboratory tests, and clinically by ALSFRS-R and FVC. Patients were also followed five years later and ALSFRS-R score was recorded in the survived individuals. There was no report of severe adverse events related to cell infusion. Two patients experienced dyspnea and chest pain 36 and 65 days after cell infusion due to pulmonary emboli. The progressive decrease in ALSFRS-R and FVC levels was recorded and three patients died in the first year. During five years follow up, despite a notable decrease in functional scores, 5 patients survived. Intravenous (IV) infusion of allogeneic Ad-MSCs in ALS patients is safe and feasible. The survival rate of the patients is more than IV autologous MSCs (Registration number: IRCT20080728001031N26).

16.
Front Immunol ; 11: 221, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32210954

RESUMEN

Exosomes are nano vesicles from the larger family named Extracellular Vesicle (EV)s which are released by various cells including tumor cells, mast cells, dendritic cells, B lymphocytes, neurons, adipocytes, endothelial cells, and epithelial cells. They are considerable messengers that can exchange proteins and genetic materials between the cells. Within the past decade, Tumor derived exosomes (TEX) have been emerged as important mediators in cancer initiation, progression and metastasis as well as host immune suppression and drug resistance. Although tumor derived exosomes consist of tumor antigens and several Heat Shock Proteins such as HSP70 and HSP90 to stimulate immune response against tumor cells, they contain inhibitory molecules like Fas ligand (Fas-L), Transforming Growth Factor Beta (TGF-ß) and Prostaglandin E2 (PGE2) leading to decrease the cytotoxicity and establish immunosuppressive tumor microenvironment (TME). To bypass this problem and enhance immune response, some macromolecules such as miRNAs, HSPs and activatory ligands have been recognized as potent immune inducers that could be used as anti-tumor agents to construct a nano sized tumor vaccine. Here, we discussed emerging engineered exosomes as a novel therapeutic strategy and considered the associated challenges.


Asunto(s)
Vesículas Extracelulares/metabolismo , Inmunoterapia/métodos , Neoplasias/metabolismo , Animales , Bioingeniería , Carcinogénesis , Humanos , Tolerancia Inmunológica , Metástasis de la Neoplasia , Neoplasias/inmunología , Neoplasias/terapia , Microambiente Tumoral
17.
ACS Appl Mater Interfaces ; 11(41): 37421-37433, 2019 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-31525863

RESUMEN

Extracellular vesicles derived from mesenchymal stem cells (MSC-EVs) have been widely reported as promising cell-free products that show therapeutic effects of the parental cells but not their limitations. Due to the intrinsic liver tropism of MSC-EVs, they have been widely used as therapeutics or drug carriers for treatment of liver diseases. However, rapid clearance from the target site may attenuate the efficiency of systemically administered MSC-EVs. Herein, sustained release into the peritoneum has been proposed as a new strategy to prolong the bioavailability of the MSC-EVs in the target liver. During intraperitoneal injection, clickable polyethylene glycol (PEG) macromeres were mixed with MSC-EVs to form EV-encapsulated PEG hydrogels via a fast, biocompatible click reaction. Upon biodegradation, the EV-laden hydrogels were swollen gradually to release EVs in a sustained manner over 1 month. In vivo tracking of the labeled EVs revealed that the accumulation of EVs in the liver was extended by hydrogel-mediated delivery for 1 month. Four weeks after injection in a rat model of chronic liver fibrosis, the physical and histopathological investigations of the harvested liver showed superior antifibrosis, anti-apoptosis, and regenerative effects of the EVs when delivered by the sustained systemic release (Gel-EV) to the conventional bolus injection (Free-EV). Specifically, the Gel-EV system improved the antifibrosis, anti-inflammation, anti-apoptosis, and regenerative effects of the EVs to nearly 40, 50, 40, and 50% compared to Free-EV, respectively, as was specified by quantification of the fibrotic area, α-SMA density, and caspase-3 density in the harvested tissues and ALT enzyme in serum. This study may potentiate the use of MSC-EVs as cell-free therapeutics for chronic liver failure. The sustained systemic delivery strategy may open a new paradigm to extend the effects of disease-targeting EVs over time.


Asunto(s)
Vesículas Extracelulares/trasplante , Regeneración Hepática , Células Madre Mesenquimatosas/metabolismo , Animales , Modelos Animales de Enfermedad , Enfermedad Hepática en Estado Terminal , Vesículas Extracelulares/metabolismo , Humanos , Masculino , Polietilenglicoles/química , Polietilenglicoles/farmacología , Ratas , Ratas Wistar
18.
Cell J ; 20(4): 592-598, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30124008

RESUMEN

OBJECTIVE: Amyotrophic lateral sclerosis (ALS) is the most severe disorder within the spectrum of motor neuron diseases (MND) that has no effective treatment and a progressively fatal outcome. We have conducted two clinical trials to assess the safety and feasibility of intravenous (IV) and intrathecal (IT) injections of bone marrow derived mesenchymal stromal cells (BM-MSCs) in patients with ALS. MATERIALS AND METHODS: This is an interventional/experimental study. We enrolled 14 patients that met the following inclusion criteria: definitive diagnosis of sporadic ALS, ALS Functional Rating Scale (ALS-FRS) ≥24, and ≥40% predicted forced vital capacity (FVC). All patients underwent bone marrow (BM) aspiration to obtain an adequate sample for cell isolation and culture. Patients in group 1 (n=6) received an IV and patients in group 2 (n=8) received an IT injection of the cell suspension. All patients in both groups were followed at 24 hours and 2, 4, 6, and 12 months after the injection with ALS-FRS, FVC, laboratory tests, check list of side effects and brain/spinal cord magnetic resonance imaging (MRI). In each group, one patient was lost to follow up one month after cell injection and one patient from IV group died due to severe respiratory insufficiency and infection. RESULTS: During the follow up there were no reports of adverse events in terms of clinical and laboratory assessments. In MRI, there was not any new abnormal finding. The ALS-FRS score and FVC percentage significantly reduced in all patients from both groups. CONCLUSION: This study has shown that IV and IT transplantation of BM-derived stromal cells is safe and feasible (Registration numbers: NCT01759797 and NCT01771640).

20.
Cell J ; 20(2): 267-277, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29633605

RESUMEN

OBJECTIVES: The regenerative potential of bone marrow-derived mononuclear cells (MNCs) and CD133+ stem cells in the heart varies in terms of their pro-angiogenic effects. This phase II/III, multicenter and double-blind trial is designed to compare the functional effects of intramyocardial autologous transplantation of both cell types and placebo in patients with recent myocardial infarction (RMI) post-coronary artery bypass graft. MATERIALS AND METHODS: This was a phase II/III, randomized, double-blind, placebo-controlled trial COMPARE CPM-RMI (CD133, Placebo, MNCs - recent myocardial infarction) conducted in accordance with the Declaration of Helsinki that assessed the safety and efficacy of CD133 and MNCs compared to placebo in patients with RMI. We randomly assigned 77 eligible RMI patients selected from 5 hospitals to receive CD133+ cells, MNC, or a placebo. Patients underwent gated single photon emission computed tomography assessments at 6 and 18 months post-intramyocardial transplantation. We tested the normally distributed efficacy outcomes with a mixed analysis of variance model that used the entire data set of baseline and between-group comparisons as well as within subject (time) and group×time interaction terms. RESULTS: There were no related serious adverse events reported. The intramyocardial transplantation of both cell types increased left ventricular ejection fraction by 9% [95% confidence intervals (CI): 2.14% to 15.78%, P=0.01] and improved decreased systolic wall thickening by -3.7 (95% CI: -7.07 to -0.42, P=0.03). The CD133 group showed significantly decreased non-viable segments by 75% (P=0.001) compared to the placebo and 60% (P=0.01) compared to the MNC group. We observed this improvement at both the 6- and 18-month time points. CONCLUSIONS: Intramyocardial injections of CD133+ cells or MNCs appeared to be safe and efficient with superiority of CD133+ cells for patients with RMI. Although the sample size precluded a definitive statement about clinical outcomes, these results have provided the basis for larger studies to confirm definitive evidence about the efficacy of these cell types (Registration Number: NCT01167751).

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA