Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Exp Cell Res ; 434(2): 113875, 2024 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-38092345

RESUMO

The mammalian renal organ represents a pinnacle of complexity, housing functional filtering units known as nephrons. During embryogenesis, the depletion of niches containing renal progenitor cells (RPCs) and the subsequent incapacity of adult kidneys to generate new nephrons have prompted the formulation of protocols aimed at isolating residual RPCs from mature kidneys and inducing their generation from diverse cell sources, notably pluripotent stem cells. Recent strides in the realm of regenerative medicine and the repair of tissues using stem cells have unveiled critical signaling pathways essential for the maintenance and generation of human RPCs in vitro. These findings have ushered in a new era for exploring novel strategies for renal protection. The present investigation delves into potential transcription factors and signaling cascades implicated in the realm of renal progenitor cells, focusing on their protection and differentiation. The discourse herein elucidates contemporary research endeavors dedicated to the acquisition of progenitor cells, offering crucial insights into the developmental mechanisms of these cells within the renal milieu and paving the way for the formulation of innovative treatment modalities.


Assuntos
Rim , Células-Tronco Pluripotentes , Animais , Humanos , Diferenciação Celular , Medicina Regenerativa , Transdução de Sinais , Mamíferos
2.
J Cell Mol Med ; 28(11): e18453, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38818569

RESUMO

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.


Assuntos
Células Endoteliais da Veia Umbilical Humana , Rim , Animais , Rim/metabolismo , Rim/citologia , Humanos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Camundongos , Organoides/metabolismo , Organoides/citologia , Células Epiteliais/metabolismo , Células Epiteliais/citologia , Diferenciação Celular , Biomarcadores/metabolismo , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/citologia , Técnicas de Cultura de Células em Três Dimensões/métodos
3.
Mol Pharm ; 21(6): 2637-2658, 2024 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-38728585

RESUMO

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.


Assuntos
Vesículas Extracelulares , Células-Tronco Mesenquimais , Cicatrização , Vesículas Extracelulares/transplante , Vesículas Extracelulares/metabolismo , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Cicatrização/fisiologia , Animais , Transplante de Células-Tronco Mesenquimais/métodos , Doença Crônica , Ensaios Clínicos como Assunto , Queimaduras/terapia
4.
Biogerontology ; 24(2): 293-301, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36648709

RESUMO

Tissue fibrosis is associated with the aging process of most of our organs, and organ aging correlates with the chronic accumulation of senescent cells. Fibrosis occurs when fibroblasts proliferate and deposit pathological amounts of extracellular matrix (ECM), leading to progressive tissue scarring and organ dysfunction. Fibroblasts play a key role in fibrosis, especially in the skin where fibroblasts are the most abundant cell type in the dermis and are mainly responsible for the synthesis of ECM. This study aims to investigate how senescent fibroblasts and their secretome influence dermal fibrosis. Here we used human dermal fibroblasts (HDFs) treated with doxorubicin (doxo) to induce senescence. The senescent phenotype of these stress-induced premature senescent (SIPS) cells was confirmed with several markers. The expression of pro-fibrotic genes was quantified and finally, the impact of their secretome on the fibrotic response of non-senescent fibroblasts was assessed. Doxorubicin treatment, induced senescence in fibroblasts which has been confirmed with elevated senescence-associated ß- galactosidase (SA-ß-gal) activity, absence of BrdU incorporation, upregulation of p21, and loss of Lamin b1. Expression levels of the pro-fibrotic genes ACTA2 and FN1 increased in SIPS cells, but in contrast to studies using lung fibroblasts the secretome of these cells failed to induce a paracrine fibrotic response in non-senescent cells. In general, these results suggest that these senescent cells are potentially profibrotic, and their accumulation can trigger fibrosis in organs.


Assuntos
Secretoma , Pele , Humanos , Células Cultivadas , Pele/metabolismo , Senescência Celular , Fibrose , Fibroblastos/metabolismo
5.
Differentiation ; 124: 28-42, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35158252

RESUMO

The extracellular matrix (ECM) is an essential network entity surrounding and supporting cells to guarantee their physiological function and homeostasis. It is important to choose the most appropriate ECM for in vitro experiments of mammalian cells for tissue engineering and functional analyses. Although most studies have examined optimization of the correct ECM that can be used to assess mammalian cells, a comprehensive study has not been conducted. In this review, we present the factors to be taken into consideration when designing or optimizing an appropriate ECM for development and maintenance of kidney tissues and compare the previously reported ECMs.


Assuntos
Matriz Extracelular , Engenharia Tecidual , Animais , Homeostase , Rim , Mamíferos
6.
Cell Tissue Bank ; 24(1): 59-66, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35635634

RESUMO

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.


Assuntos
Células-Tronco Mesenquimais , Soroalbumina Bovina , Animais , Humanos , Células Cultivadas , Soroalbumina Bovina/metabolismo , Cordão Umbilical , Diferenciação Celular , Proliferação de Células
7.
Cell Tissue Res ; 387(2): 207-224, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35088178

RESUMO

The kidney is a highly complex organ in the human body. Although creating an in vitro model of the human kidney is challenging, tremendous advances have been made in recent years. Kidney organoids are in vitro kidney models that are generated from stem cells in three-dimensional (3D) cultures. They exhibit remarkable degree of similarities with the native tissue in terms of cell type, morphology, and function. The establishment of 3D kidney organoids facilitates a mechanistic study of cell communications, and these organoids can be used for drug screening, disease modeling, and regenerative medicine applications. This review discusses the cellular complexity during in vitro kidney generation. We intend to highlight recent progress in kidney organoids and the applications of these relatively new technologies.


Assuntos
Rim , Organoides , Humanos , Medicina Regenerativa/métodos
8.
Drug Chem Toxicol ; 45(5): 2262-2268, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34006164

RESUMO

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.


Assuntos
Gentamicinas , Soro , Animais , Antibacterianos/toxicidade , Antioxidantes/farmacologia , Nitrogênio da Ureia Sanguínea , Creatinina , Sangue Fetal/metabolismo , Gentamicinas/metabolismo , Gentamicinas/toxicidade , Glutationa/metabolismo , Humanos , Rim , Masculino , Oxidantes/metabolismo , Ratos , Ratos Wistar , Soro/metabolismo
9.
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
10.
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
11.
J Cell Physiol ; 234(8): 12451-12470, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30644092

RESUMO

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.


Assuntos
Predisposição Genética para Doença , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/metabolismo , Regulação da Expressão Gênica , Humanos , Canais de Cátion TRPP
12.
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
13.
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
14.
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
16.
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
17.
Cytotherapy ; 17(11): 1495-505, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26341479

RESUMO

BACKGROUND AIMS: Chronic kidney disease (CKD) attributed to cisplatin is well documented. Mesenchymal stromal cells (MSCs) are proven to be renotropic. Although they have been shown to improve function in CKD and reduce fibrosis in different experimental rodent models, their efficiency in primates is unknown. The present study aimed to evaluate the prevention of CKD and reduction of fibrosis in monkeys treated with MSCs after cisplatin nephrotoxicity. METHODS: We induced CKD in adult rhesus Macaca mulatta monkeys by means of intravenous administration of cisplatin. Autologous MSCs were transplanted by means of intrarenal arterial injections to assess the adverse effects of cisplatin in two CKD models: preventative and stable. Preventative CKD monkeys (n = 3) underwent cell transplantation 4 days after the cisplatin injection. The stable CKD monkeys (n = 2) underwent cell transplantation 6 months after the cisplatin injection. Non-treated (n = 4) and normal saline-injected animals (n = 3) comprised the control and vehicle groups, respectively. We followed the animals for survival rate, serum biochemistry, urine analysis and histopathological indices. RESULTS: In the preventive CKD model, MSC transplantation tended to improve some renal functions but significantly reduced the histopathologic score compared with the vehicle and control groups. In the stable CKD model, MSCs did not ameliorate renal function or pathological score. CONCLUSIONS: These results suggest that MSCs tend to delay progression of CKD and fibrosis but do not reduce established interstitial fibrosis in this unique primate model of cisplatin-induced nephrotoxicity.


Assuntos
Transplante de Células-Tronco Mesenquimais/métodos , Insuficiência Renal Crônica/prevenção & controle , Animais , Cisplatino/efeitos adversos , Modelos Animais de Doenças , Fibrose/prevenção & controle , Fibrose/terapia , Rim/patologia , Macaca mulatta , Masculino , Células-Tronco Mesenquimais/citologia , Insuficiência Renal Crônica/induzido quimicamente , Insuficiência Renal Crônica/mortalidade , Insuficiência Renal Crônica/terapia , Taxa de Sobrevida , Transplante Autólogo
18.
Cytotherapy ; 16(6): 734-49, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24801377

RESUMO

BACKGROUND: Clinically, acute kidney injury (AKI) is a potentially devastating condition for which no specific therapy improves efficacy of the repair process. Bone marrow mesenchymal stromal cells (BM-MSCs) are proven to be beneficial for the renal repair process after AKI in different experimental rodent models, but their efficacy in large animals and humans remains unknown. This study aims to assess the effect of autologous rhesus Macaque mulatta monkey BM-MSC transplantation in cisplatin-induced AKI. METHODS: We chose a model of AKI induced by intravenous administration of 5 mg/kg cisplatin. BM-MSCs were transplanted through intra-arterial injection. The animals were followed for survival, biochemistry analysis and pathology. RESULTS: Transplantation of 5 × 10(6) cells/kg ameliorated renal function during the first week, as shown by significantly lower serum creatinine and urea values and higher urine creatinine and urea clearance without hyponatremia, hyperkalemia, proteinuria and polyuria up to 84 d compared with the vehicle and control groups. The superparamagnetic iron oxide nanoparticle-labeled cells were found in both the glomeruli and tubules. BM-MSCs markedly accelerated Foxp3+ T-regulatory cells in response to cisplatin-induced damage, as revealed by higher numbers of Foxp3+ cells within the tubuli of these monkeys compared with cisplatin-treated monkeys in the control and vehicle groups. CONCLUSIONS: These data demonstrate that BM-MSCs in this unique large-animal model of cisplatin-induced AKI exhibited recovery and protective properties.


Assuntos
Injúria Renal Aguda/terapia , Terapia Baseada em Transplante de Células e Tecidos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/tratamento farmacológico , Transplante de Células-Tronco Mesenquimais , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/patologia , Animais , Células da Medula Óssea/citologia , Cisplatino/administração & dosagem , Cisplatino/efeitos adversos , Humanos , Injeções , Macaca mulatta , Células-Tronco Mesenquimais/citologia , Artéria Renal
19.
Regen Ther ; 27: 506-518, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38745839

RESUMO

Introduction: Acute kidney injury (AKI) is a common health problem that leads to high morbidity and potential mortality. The failure of conventional treatments to improve forms of this condition highlights the need for innovative and effective treatment approaches. Regenerative therapies with Renal Progenitor Cells (RPCs) have been proposed as a promising new strategy. A growing body of evidence suggests that progenitor cells differentiated from different sources, including human embryonic stem cells (hESCs), can effectively treat AKI. Methods: Here, we describe a method for generating RPCs and directed human Embryoid Bodies (EBs) towards CD133+CD24+ renal progenitor cells and evaluate their functional activity in alleviating AKI. Results: The obtained results show that hESCs-derived CD133+CD24+ RPCs can engraft into damaged renal tubules and restore renal function and structure in mice with gentamicin-induced kidney injury, and significantly decrease blood urea nitrogen levels, suppress oxidative stress and inflammation, and attenuate histopathological disturbances, including tubular necrosis, tubular dilation, urinary casts, and interstitial fibrosis. Conclusion: The results suggest that RPCs have a promising regenerative potential in improving renal disease and can lay the foundation for future cell therapy and disease modeling.

20.
J Craniofac Surg ; 24(4): 1292-7, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23851791

RESUMO

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.


Assuntos
Transplante Ósseo/métodos , Doenças Mandibulares/cirurgia , Transplante de Células-Tronco Mesenquimais/métodos , Procedimentos de Cirurgia Plástica/métodos , Engenharia Tecidual/métodos , Alicerces Teciduais , Adolescente , Adulto , Aloenxertos/transplante , Densidade Óssea/fisiologia , Reabsorção Óssea/etiologia , Separação Celular/métodos , Estudos de Viabilidade , Feminino , Sobrevivência de Enxerto , Humanos , Masculino , Reconstrução Mandibular/métodos , Osseointegração/fisiologia , Projetos Piloto , Radiografia Panorâmica/métodos , Tomografia Computadorizada Espiral/métodos , Cicatrização/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA