Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
J Cell Mol Med ; 28(11): e18453, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38818569

RESUMEN

As an advance laboratory model, three-dimensional (3D) organoid culture has recently been recruited to study development, physiology and abnormality of kidney tissue. Micro-tissues derived from primary renal cells are composed of 3D epithelial structures representing the main characteristics of original tissue. In this research, we presented a simple method to isolate mouse renal clonogenic mesenchymal (MLCs) and epithelial-like cells (ELCs). Then we have done a full characterization of MLCs using flow cytometry for surface markers which showed that more than 93% of cells expressed these markers (Cd44, Cd73 and Cd105). Epithelial and stem/progenitor cell markers characterization also performed for ELC cells and upregulating of these markers observed while mesenchymal markers expression levels were not significantly increased in ELCs. Each of these cells were cultured either alone (ME) or in combination with human umbilical vein endothelial cells (HUVECs) (MEH; with an approximate ratio of 10:5:2) to generate more mature kidney structures. Analysis of 3D MEH renal micro-tissues (MEHRMs) indicated a significant increase in renal-specific gene expression including Aqp1 (proximal tubule), Cdh1 (distal tubule), Umod (loop of Henle), Wt1, Podxl and Nphs1 (podocyte markers), compared to those groups without endothelial cells, suggesting greater maturity of the former tissue. Furthermore, ex ovo transplantation showed greater maturation in the constructed 3D kidney.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana , Riñón , Animales , Riñón/metabolismo , Riñón/citología , Humanos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Ratones , Organoides/metabolismo , Organoides/citología , Células Epiteliales/metabolismo , Células Epiteliales/citología , Diferenciación Celular , Biomarcadores/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Técnicas de Cultivo Tridimensional de Células/métodos
2.
Mol Pharm ; 21(6): 2637-2658, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38728585

RESUMEN

To date, the widespread implementation of therapeutic strategies for the treatment of chronic wounds, including debridement, infection control, and the use of grafts and various dressings, has been time-consuming and accompanied by many challenges, with definite success not yet achieved. Extensive studies on mesenchymal stem cells (MSCs) have led to suggestions for their use in treating various diseases. Given the existing barriers to utilizing such cells and numerous pieces of evidence indicating the crucial role of the paracrine signaling system in treatments involving MSCs, extracellular vesicles (EVs) derived from these cells have garnered significant attention in treating chronic wounds in recent years. This review begins with a general overview of current methods for chronic wound treatment, followed by an exploration of EV structure, biogenesis, extraction methods, and characterization. Subsequently, utilizing databases such as Google Scholar, PubMed, and ScienceDirect, we have explored the latest findings regarding the role of EVs in the healing of chronic wounds, particularly diabetic and burn wounds. In this context, the role and mode of action of these nanoparticles in healing chronic wounds through mechanisms such as oxygen level elevation, oxidative stress damage reduction, angiogenesis promotion, macrophage polarization assistance, etc., as well as the use of EVs as carriers for engineered nucleic acids, have been investigated. The upcoming challenges in translating EV-based treatments for healing chronic wounds, along with possible approaches to address these challenges, are discussed. Additionally, clinical trial studies in this field are also covered.


Asunto(s)
Vesículas Extracelulares , Células Madre Mesenquimatosas , Cicatrización de Heridas , Vesículas Extracelulares/trasplante , Vesículas Extracelulares/metabolismo , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Cicatrización de Heridas/fisiología , Animales , Trasplante de Células Madre Mesenquimatosas/métodos , Enfermedad Crónica , Ensayos Clínicos como Asunto , Quemaduras/terapia
3.
Arch Iran Med ; 26(2): 100-109, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-37543930

RESUMEN

BACKGROUND: We aimed to determine the effects of systemic therapy with autologous adipose tissue derived mesenchymal stem cells (AD-MSCs) on different parameters of peritoneal function and inflammation in peritoneal dialysis (PD) patients. METHODS: We enrolled nine PD patients with ultrafiltration failure (UFF). Patients received 1.2±0.1×106 cell/kg of AD-MSCs via cubital vein and were then followed for six months at time points of baseline, 3, 6, 12, 16 and 24 weeks after infusion. UNI-PET was performed for assessment of peritoneal characteristics at baseline and weeks 12 and 24. Systemic and peritoneal levels of tumor necrosis factor α (TNF-α), interleukin-6(IL-6), IL-2 and CA125 (by ELISA) and gene expression levels of transforming growth factor beta (TGF-ß), smooth muscle actin (𝛼-SMA) and fibroblast-specific protein-1 (FSP-1) in PD effluent derived cells (by quantitative real-time PCR) were measured at baseline and weeks 3, 6, 12, 16 and 24. RESULTS: Slight improvement was observed in the following UF capacity indices: free water transport (FWT, 32%), ultrafiltration - small pore (UFSP, 18%), ultrafiltration total (UFT, 25%), osmotic conductance to glucose (OCG, 25%), D/P creatinine (0.75 to 0.70), and Dt/D0 glucose (0.23 to 0.26). There was a slight increase in systemic and peritoneal levels of CA125 and a slight decrease in gene expression levels of TGF-ß, α-SMA and FSP-1 that was more prominent at week 12 and vanished by the end of the study. CONCLUSION: Our results for the first time showed the potential of MSCs for treatment of peritoneal damage in a clinical trial. Our results could be regarded as hypothesis suggestion and will need confirmation in future studies.


Asunto(s)
Fibrosis Peritoneal , Humanos , Proyectos Piloto , Soluciones para Diálisis/metabolismo , Factor de Crecimiento Transformador beta , Glucosa/metabolismo
4.
Theory Biosci ; 142(2): 167-179, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37071370

RESUMEN

In this study, we proposed a biological model explaining the progress of autoimmune activation along different stages of systemic lupus erythematosus (SLE). For any upcoming stage of SLE, any new component is introduced, when it is added to the model. Particularly, the interaction of mesenchymal stem cells, with the components of the model, is specified in a way that both the inflammatory and anti-inflammatory functions of these cells would be covered. The biological model is then recapitulated to a model with less complexity that explains the main features of the problem. Later, a 7th-order mathematical model for SLE is proposed, based on this simplified model. Finally, the range of validity of the proposed mathematical model was assessed. For this purpose, we simulated the model and analyzed the simulation results in case of some known behaviors of the disease, such as tolerance breach, the appearance of systemic inflammation, development of clinical signs, and occurrence of flares and improvements. The model was able to reproduce these events, qualitatively.


Asunto(s)
Lupus Eritematoso Sistémico , Células Madre Mesenquimatosas , Humanos , Lupus Eritematoso Sistémico/diagnóstico , Lupus Eritematoso Sistémico/tratamiento farmacológico , Modelos Teóricos
5.
Cell Tissue Bank ; 24(1): 59-66, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35635634

RESUMEN

In the use of bovine fetal serum (FBS) there is concern about the possibility of disease transmission from animal to human. Therefore, it seems necessary to create culture conditions free of animal serum, especially in cell therapy. The aim of this study was to evaluate the feasibility of replacing human umbilical cord serum (hUCS) with FBS for in vitro expansion of umbilical cord mesenchymal stromal/stem cells (UC-MSCs). Here, UC-MSCs were cultured for five days in media supplemented either by hUCS or commercial FBS (Gibco and HyClone) to compare their viability, proliferation, morphology, Immunophenotype and differentiation potential. Our data shows that use of 5% and/or 10% hUCS, resulted in a tenfold increase in the number of MSCs; While in the presence of commercial FBS, this figure reached a maximum of five times. Notably, the rate of cell proliferation in the group containing 2% hUCS was the same as the groups containing 10% commercial FBS. Furthermore, there was no significant difference between groups in terms of viability, surface markers, and multilineage differentiation potential. These results demonstrated that hUCS can efficiently replace FBS for the routine culture of MSCs and can be used ideally in manufacturing process of UC-MSCs in cell therapy industry.


Asunto(s)
Células Madre Mesenquimatosas , Albúmina Sérica Bovina , Animales , Humanos , Células Cultivadas , Albúmina Sérica Bovina/metabolismo , Cordón Umbilical , Diferenciación Celular , Proliferación Celular
6.
Bioimpacts ; 12(5): 439-448, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36381637

RESUMEN

Introduction: Inflammation is one of the most important mechanisms involved in cisplatin-induced acute kidney injury (AKI). Mesenchymal stromal/stem cells (MSCs) exhibit anti-inflammatory and immunomodulatory abilities. Human endometrial stromal/stem cells (hEnSCs) exhibit similar properties to MSCs. These cells secrete immunoregulators, so we investigated the inflammatory aspect of hEnSCs in the treatment of cisplatin-induced AKI in Wistar rats. Methods: Each group consisted of 6 male Wistar rats. Groups were as follows: sham, model (5 mg/kg cisplatin, IP), and treatment (1 million hEnSCs, IV, 3 hours after cisplatin). Renal function, histopathology, proliferation rate, infiltration of CD3+ T cell, and expression of Il-10 and cystatin c (Cst3) were assessed on day 5. DiI-labeled cells were tracked in kidney and liver on days 4 and 14. Results: HEnSC transplantation improved cisplatin-induced injuries such as renal dysfunction and tissue damage. The highest levels of pathologic scores and hyaline cast formation were observed in the model group while hEnSCs transplantation resulted in their reduction (154.00 ± 14.95, 8.00 ± 1.41 vs. 119.40 ± 5.43, 2.50 ± 1.05). The percentage of Ki-67 positive cells in the treatment group increased while cisplatin decreased proliferation (39.91 ± 5.33 vs. 23.91 ± 3.57 in glomeruli and 39.07 ± 2.95 vs. 16.61 ± 3.25 in tubules). The expression of Cst3 and Il-10 was higher in the model and treatment groups, respectively. DiI-labeled cells were observed in the renal tubules and liver lobes on days 4 and 14. Conclusion: HEnSCs may ameliorate cisplatin-induced AKI through anti-inflammatory and immunomodulatory effects and/or through paracrine effects.

7.
Drug Chem Toxicol ; 45(5): 2262-2268, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34006164

RESUMEN

Sold under the brand name of Garamycin, gentamicin (GM) is an antibiotic in the category of aminoglycoside, that although does have many antibacterial properties, owing to several side effects, its consumption is confined. The current study is aimed at gauging the protective influences of human umbilical cord blood serum (hUCBS) on nephrotoxicity which is induced by GM. In this regard, in the present experimental design, twenty-eight male Wistar rats with the weights of 220 ± 20 g were categorized randomly into 4 groups of seven. The groups included GM (100 mg/kg), control as well as hUCBS at doses of one and two percent together with GM (100 mg/kg) for ten days in an intraperitoneal manner. Blood sampling was collected from the heart directly 24 h after the final injection for obtaining blood serum; the parameters of C-reactive protein (CRP), total oxidant status (TOS), interleukin (IL)-6, lactate dehydrogenase (LDH), total antioxidant capacity (TAC), creatinine (Cr), blood urea nitrogen (BUN), blood serum glutathione (GSH) were gauged in blood serum samples to evaluate renal function. Moreover, for histology, an examination of kidney tissue was performed. In comparison to those of the GM group, in the treatment group, hUCBS significantly decreased the levels of BUN, Cr, LDH, TOS, IL-6, and the CRP levels, and significantly increased the TAC and GSH levels. It was revealed that the treatment of the animals with hUCBS culminates in the reduction of GM' toxic impacts on the kidney.


Asunto(s)
Gentamicinas , Suero , Animales , Antibacterianos/toxicidad , Antioxidantes/farmacología , Nitrógeno de la Urea Sanguínea , Creatinina , Sangre Fetal/metabolismo , Gentamicinas/metabolismo , Gentamicinas/toxicidad , Glutatión/metabolismo , Humanos , Riñón , Masculino , Oxidantes/metabolismo , Ratas , Ratas Wistar , Suero/metabolismo
8.
Cell J ; 23(5): 568-575, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34837685

RESUMEN

OBJECTIVE: Acute kidney injury (AKI) is referred to as sudden decline in the function of kidney. Human endometrial stromal/stem cells (hEnSCs) are mesenchymal stem cell (MSC)-like cells, which are suitable candidates for regenerative medicine purposes, yet the effect of hEnSCs on cisplatin-induced AKI has not been studied; therefore, the present study was conducted to investigate this gap in the literature. MATERIALS AND METHODS: In this experimental study, hEnSCs were obtained from endometrial biopsy using collagenase I and were then cultured in DMEM/F12 medium. A total of 48 male Wistar rats (150-200 g) were classified into four groups: intact -receiving no treatment, model -receiving 5 mg/kg of body weight cisplatin, as well as phosphate-buffered saline (PBS) and cell -receiving either PBS or hEnSCs for three hours after cisplatin injection, respectively. Biochemical parameters, pathologic scores, apoptosis assay, Bcl-2 and Tnf-α expression were evaluated on day 5. RESULTS: On day 5 post-transplantation we observed that HEnSCs injection has led to a decrease in both blood urea nitrogen (BUN) and serum creatinine (SCr), compared to the model and PBS groups (0.82 ± 0.03 vs. 1.42 ± 0.06, 1.09 ± 0.05 mg/dl and 61.53 ± 3.07 vs. 116.60 ± 2.12, 112.00 ± 1.35 mg/dl, respectively). The highest levels of pathologic scores were observed in model and PBS groups, while hEnSCs transplantation resulted in a decrease in pathologic scores (149.10 ± 7.03, 141.50 ± 4.68 vs. 118 ± 2.16). HEnSCs significantly decreased the percentage of TUNELpositive cells in the cell group compared with model and PBS groups (20.37 ±. 3.37 vs. 33.67 ± 1.79, 31.53 ± 1.05 in glomeruli and 15.10 ± 1.47 vs. 42.33 ± 1.72, 39.23 ± 1.61 in tubules). In addition, HEnSCs resulted in upregulation of Bcl-2 and downregulation of Tnf-α in the cisplatin-induced AKI. CONCLUSION: Our results showed that injection of hEnSCs may improve AKI through lowering the amount of apoptosis in renal cells.

10.
Cell J ; 23(1): 1-13, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33650815

RESUMEN

OBJECTIVE: In the present study, we examined the tolerance-inducing effects of human adipose-derived mesenchymal stem cells (hAD-MSCs) and bone marrow-derived MSCs (hBM-MSCs) on a nonhuman primate model of skin transplantation. MATERIALS AND METHODS: In this experimental study, allogenic and xenogeneic of immunomodulatory properties of human AD-MSCs and BM-MSCs were evaluated by mixed lymphocyte reaction (MLR) assays. Human MSCs were obtained from BM or AD tissues (from individuals of either sex with an age range of 35 to 65 years) and intravenously injected (2×106 MSCs/kg) after allogeneic skin grafting in a nonhuman primate model. The skin sections were evaluated by H and E staining for histopathological evaluations, particularly inflammation and rejection reaction of grafts after 96 hours of cell injection. At the mRNA and protein levels, cellular mediators of inflammation, such as CD4+IL-17+ (T helper 17; Th17) and CD4+INF-γ+ (T helper 1, Th1) cells, along with CD4+FoxP3+ cells (Treg), as the mediators of immunomodulation, were measured by RT-PCR and flow cytometry analyses. RESULTS: A significant Treg cells expansion was observed in MSCs-treated animals which reached the zenith at 24 hours and remained at a high concentration for 96 hours; however, Th1 and Th17 cells were significantly decreased. Our results showed that human MSCs significantly decrease Th1 and Th17 cell proliferation by decreasing interleukin-17 (IL-17) and interferon-γ (INF-γ) production and significantly increase Treg cell proliferation by increasing FoxP3 production. They also extend the allogenic skin graft survival in nonhuman primates. Histological evaluations showed no obvious presence of inflammatory cells or skin redness or even bulging after MSCs injection up to 96 hours, compared to the group without MSCs. There were no significant differences between hBM-MSCs and hAD-MSCs in terms of histopathological scores and inflammatory responses (P<0.05). CONCLUSION: It seems that MSCs could be regarded as a valuable immunomodulatory tool to reduce the use of immunosuppressive agents.

11.
Stem Cells Dev ; 30(8): 428-440, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33787359

RESUMEN

Directed differentiation of human pluripotent stem cells (hPSCs) uses a growing number of small molecules and growth factors required for in vitro generation of renal lineage cells. Although current protocols are relatively inefficient or expensive. The first objective of the present work was to establish a new differentiation protocol for generating renal precursors. We sought to determine if inducer of definitive endoderm 1 (IDE1), a cost-effective small molecule, can be used to replace activin A. Gene expression data showed significantly increased expressions of nephrogenic markers in cells differentiated with 20 nM IDE1 compared with cells differentiated with activin A. Thus, renal lineage cells could be generated by this alternative approach. Afterward, we determined whether coculture of endothelial and mesenchymal cells could increase the maturation of three-dimensional (3D) renal structures. For this purpose, we employed a 3D coculture system in which hPSC-derived kidney precursors were cocultured with endothelial cells (ECs) and mesenchymal stem cells (MSCs), hereafter named RMEM (renal microtissue derived from coculture of renal precursors with endothelial and mesenchymal stem cells). hPSC-derived kidney precursors were cultured either alone [renal microtissue (RM)] or in coculture with human umbilical vein endothelial cells and human bone marrow-derived mesenchymal stem cells at an approximate ratio of 10:7:2, respectively. Immunofluorescent staining showed expressions of kidney-specific markers synaptopodin, LTL, and E-cadherin, as well as CD31+ ECs that were distributed throughout the RMEMs. Quantitative real-time polymerase chain reaction analysis confirmed a significant increase in gene expressions of the renal-specific markers in RMEMs compared with RMs. These findings demonstrated that renal precursors cocultured with endothelial and MSCs showed greater maturity compared with RMs. Moreover, ex ovo transplantation induced further maturation in the RMEM constructs. Our novel approach enabled the generation of RMEM that could potentially be used in high-throughput drug screening and nephrotoxicology studies.


Asunto(s)
Diferenciación Celular/genética , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Riñón/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Madre Pluripotentes/metabolismo , Línea Celular , Células Cultivadas , Técnicas de Cocultivo/métodos , Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Inmunohistoquímica , Riñón/citología , Células Madre Mesenquimatosas/citología , Proteína Homeótica Nanog/genética , Proteína Homeótica Nanog/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Células Madre Pluripotentes/citología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Curr Stem Cell Res Ther ; 15(3): 263-283, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31854277

RESUMEN

Renal disease is a major worldwide public health problem that affects one in ten people. Renal failure is caused by the irreversible loss of the structural and functional units of kidney (nephrons) due to acute and chronic injuries. In humans, new nephrons (nephrogenesis) are generated until the 36th week of gestation and no new nephron develops after birth. However, in rodents, nephrogenesis persists until the immediate postnatal period. The postnatal mammalian kidney can partly repair their nephrons. The kidney uses intrarenal and extra-renal cell sources for maintenance and repair. Currently, it is believed that dedifferentiation of surviving tubular epithelial cells and presence of resident stem cells have important roles in kidney repair. Many studies have shown that stem cells obtained from extra-renal sites such as the bone marrow, adipose and skeletal muscle tissues, in addition to umbilical cord and amniotic fluid, have potential therapeutic benefits. This review discusses the main mechanisms of renal regeneration by stem cells after a kidney injury.


Asunto(s)
Riñón/fisiología , Regeneración/fisiología , Células Madre/citología , Animales , Humanos , Inmunomodulación , Enfermedades Renales/fisiopatología , Enfermedades Renales/terapia , Trasplante de Células Madre Mesenquimatosas
13.
Sci Rep ; 9(1): 15467, 2019 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-31664077

RESUMEN

Injury to podocytes is a principle cause of initiation and progression of both immune and non-immune mediated glomerular diseases that result in proteinuria and decreased function of the kidney. Current advances in regenerative medicine shed light on the therapeutic potential of cell-based strategies for treatment of such disorders. Thus, there is hope that generation and transplantation of podocytes from induced pluripotent stem cells (iPSCs), could potentially be used as a curative treatment for glomerulonephritis caused by podocytes injury and loss. Despite several reports on the generation of iPSC-derived podocytes, there are rare reports about successful use of these cells in animal models. In this study, we first generated a model of anti-podocyte antibody-induced heavy proteinuria that resembled human membranous nephropathy and was characterized by the presence of sub-epithelial immune deposits and podocytes loss. Thereafter, we showed that transplantation of functional iPSC-derived podocytes following podocytes depletion results in recruitment of iPSC-derived podocytes within the damaged glomerulus, and leads to attenuation of proteinuria and histological alterations. These results provided evidence that application of iPSCs-derived renal cells could be a possible therapeutic strategy to favorably influence glomerular diseases outcomes.


Asunto(s)
Glomerulonefritis Membranosa/terapia , Células Madre Pluripotentes Inducidas/trasplante , Proteinuria/terapia , Trasplante de Células Madre , Animales , Modelos Animales de Enfermedad , Glomerulonefritis Membranosa/complicaciones , Ratones , Proteinuria/complicaciones
14.
Iran J Kidney Dis ; 13(4): 213-224, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31422386

RESUMEN

There has been ample of preclinical and animal studies whichshowed efficacy and safety of using mesenchymal stem cells (MSCs)after transplantation for tissue repair, immunosuppression ortolerance induction. However, there has been a significant progressrecently using MSCs in small clinical trials after transplantation. Recent results using MSCs after transplantation seem to befeasible and safe. However, there are some limitations to show theeffectiveness of these cells including source, dose, timing and routeof infusions. Currently, live donor kidney transplantation has beenespecially considered and development of recent regimes includingimmunosuppression drugs and MSCs administration to kidney andother organs and deceased donor transplantation would be crucial.Therefore, in this review we focused on immunomodulatory effectsof MSCs that have been extensively studied to suppress variousinflammatory responses in kidney transplantation.


Asunto(s)
Rechazo de Injerto/prevención & control , Fallo Renal Crónico/cirugía , Trasplante de Riñón , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/inmunología , Animales , Ensayos Clínicos como Asunto , Rechazo de Injerto/inmunología , Humanos , Inmunosupresores/uso terapéutico , Trasplante Homólogo
15.
Differentiation ; 109: 1-8, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31323479

RESUMEN

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.


Asunto(s)
Diferenciación Celular , Técnicas de Cocultivo/métodos , Cuerpos Embrioides/citología , Células Madre Embrionarias Humanas/citología , Riñón/citología , Células Madre Mesenquimatosas/citología , Células Madre Embrionarias de Ratones/citología , Animales , Biomarcadores/metabolismo , Linaje de la Célula , Autorrenovación de las Células , Células Cultivadas , Cuerpos Embrioides/metabolismo , Células Madre Embrionarias Humanas/metabolismo , Humanos , Riñón/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Células Madre Embrionarias de Ratones/metabolismo
16.
J Cell Physiol ; 234(8): 12451-12470, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30644092

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) is a monogenic inherited renal cystic disease that occurs in different races worldwide. It is characterized by the development of a multitude of renal cysts, which leads to massive enlargement of the kidney and often to renal failure in adulthood. ADPKD is caused by a mutation in PKD1 or PKD2 genes encoding the proteins polycystin-1 and polycystin-2, respectively. Recent studies showed that cyst formation and growth result from deregulation of multiple cellular pathways like proliferation, apoptosis, metabolic processes, cell polarity, and immune defense. In ADPKD, intracellular cyclic adenosine monophosphate (cAMP) promotes cyst enlargement by stimulating cell proliferation and transepithelial fluid secretion. Several interventions affecting many of these defective signaling pathways have been effective in animal models and some are currently being tested in clinical trials. Moreover, the stem cell therapy can improve nephropathies and according to studies were done in this field, can be considered as a hopeful therapeutic approach in future for PKD. This study provides an in-depth review of the relevant molecular pathways associated with the pathogenesis of ADPKD and their implications in development of potential therapeutic strategies.


Asunto(s)
Predisposición Genética a la Enfermedad , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/metabolismo , Regulación de la Expresión Génica , Humanos , Canales Catiónicos TRPP
17.
Cytotherapy ; 20(5): 660-669, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29580865

RESUMEN

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.


Asunto(s)
Células de la Médula Ósea/citología , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Células Madre Mesenquimatosas/citología , Insuficiencia Renal Crónica/terapia , Adulto , Determinación de Punto Final , Femenino , Estudios de Seguimiento , Tasa de Filtración Glomerular , Humanos , Riñón/fisiopatología , Masculino , Persona de Mediana Edad , Insuficiencia Renal Crónica/fisiopatología
18.
Kidney Blood Press Res ; 43(2): 471-478, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29590654

RESUMEN

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.


Asunto(s)
Mutación del Sistema de Lectura , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Riñón Poliquístico Autosómico Dominante/genética , Canales Catiónicos TRPP/genética , Exones , Familia , Femenino , Mutación del Sistema de Lectura/genética , Secuenciación de Nucleótidos de Alto Rendimiento/economía , Humanos , Irán , Masculino , Linaje , Defectos Congénitos del Transporte Tubular Renal/genética , Factores de Tiempo , Cálculos Urinarios/genética
19.
Stem Cell Res Ther ; 8(1): 116, 2017 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-28535817

RESUMEN

BACKGROUND: Autosomal dominant polycystic kidney disease (ADPKD) is a genetic ciliopathy disease characterized by progressive formation and enlargement of cysts in multiple organs. The kidneys are particularly affected and patients may eventually develop end-stage renal disease (ESRD). We hypothesize that bone marrow mesenchymal stromal cells (BMMSCs) are renotropic and may improve kidney function via anti-apoptotic, anti-fibrotic, and anti-inflammatory effects. In this study, we aim to assess the safety and tolerability of a BMMSC infusion in ADPKD patients. METHODS: We performed a single-arm phase I clinical trial with a 12-month follow-up. This study enrolled six eligible ADPKD patients with an estimated glomerular filtration rate (eGFR) of 25-60 ml/min/1.73 m2. Patients received autologous cultured BMMSCs (2 × 106 cells/kg) through the cubital vein according to our infusion protocol. We investigated safety issues and kidney function during the follow-up visits, and compared the findings to baseline and 1 year prior to the intervention. RESULTS: There were no patients lost to follow-up. We observed no cell-related adverse events (AE) and serious adverse events (SAE) after 12 months of follow-up. The mean eGFR value of 33.8 ± 5.3 ml/min/1.73 m2 1 year before cell infusion declined to 26.7 ± 3.1 ml/min/1.73 m2 at baseline (P = 0.03) and 25.8 ± 6.2 ml/min/1.73 m2 at the 12-month follow-up visit (P = 0.62). The mean serum creatinine (SCr) level of 2 ± 0.3 mg/dl 1 year before the infusion increased to 2.5 ± 0.4 mg/dl at baseline (P = 0.04) and 2.5 ± 0.6 mg/dl at the 12-month follow-up (P = 0.96). This indicated significant changes between the differences of these two periods (12 months before infusion to baseline, and 12 months after infusion to baseline) in SCr (P = 0.05), but not eGFR (P = 0.09). CONCLUSIONS: This trial demonstrated the safety and tolerability of an intravenous transplantation of autologous BMMSCs. BMMSC efficacy in ADPKD patients should be investigated in a randomized placebo-controlled trial with a larger population, which we intend to perform. TRIAL REGISTRATION: ClinicalTrials.gov, NCT02166489 . Registered on June 14, 2014.


Asunto(s)
Células de la Médula Ósea/citología , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Células Madre Mesenquimatosas/citología , Riñón Poliquístico Autosómico Dominante/terapia , Adulto , Presión Sanguínea , Creatinina/sangre , Demografía , Determinación de Punto Final , Femenino , Estudios de Seguimiento , Tasa de Filtración Glomerular , Humanos , Riñón/patología , Riñón/fisiopatología , Masculino , Persona de Mediana Edad , Riñón Poliquístico Autosómico Dominante/sangre , Riñón Poliquístico Autosómico Dominante/fisiopatología , Trasplante Autólogo/efectos adversos
20.
Int J Urol ; 23(7): 581-6, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27062371

RESUMEN

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.


Asunto(s)
Epispadias/complicaciones , Trasplante de Células Madre , Incontinencia Urinaria de Esfuerzo/terapia , Femenino , Humanos , Masculino , Músculos/citología , Estudios Prospectivos , Resultado del Tratamiento , Uretra , Incontinencia Urinaria , Urodinámica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA