Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Int J Mol Sci ; 23(22)2022 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-36430390

RESUMO

The effective treatment of perianal fistulizing Crohn's disease is still a challenge. Local administration of mesenchymal stromal cells (MSCs) is becoming a part of accepted treatment options. However, as a fledgling technique, it still can be optimized. A new trend in translational research, which is in line with "One Health" approach, bases on exploiting parallels between naturally occurring diseases affecting humans and companion animals. Canine anal furunculosis (AF) has been indicated as condition analogous to human perianal Crohn's disease (pCD). This narrative review provides the first comprehensive comparative analysis of these two diseases based on the published data. The paper also outlines the molecular mechanisms of action of MSCs which are likely to have a role in modulating the perianal fistula niche in humans, and refers them to the current knowledge on the immunomodulatory properties of canine MSCs. Generally, the pathogenesis of both diseases shares main determinants such as the presence of genetic predispositions, dysregulation of immune response and the relation to intestine microbiota. However, we also identified many aspects which should be further specified, such as determining the frequency of true fistulas formation in AF patients, elucidating the role of TNF and Th17 pathway in the pathogenesis of AF, or clarifying the role of epithelial-to-mesenchymal transition phenomenon in the formation of canine fistulae. Nevertheless, the available data support the hypothesis that the results from testing cell therapies in dogs with anal furunculosis have a significant translational value in optimizing MSC transplants procedures in pCD patients.


Assuntos
Doença de Crohn , Furunculose , Transplante de Células-Tronco Mesenquimais , Fístula Retal , Humanos , Cães , Animais , Transplante de Células-Tronco Mesenquimais/métodos , Doença de Crohn/patologia , Furunculose/complicações , Fístula Retal/terapia , Terapia Baseada em Transplante de Células e Tecidos/efeitos adversos
2.
Int J Mol Sci ; 22(15)2021 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-34360925

RESUMO

Mesenchymal stromal cell (MSC) therapy is making its way into clinical practice, accompanied by research into strategies improving their therapeutic potential. Preconditioning MSCs with hypoxia-inducible factors-α (HIFα) stabilizers is an alternative to hypoxic priming, but there remains insufficient data evaluating its transcriptomic effect. Herein, we determined the gene expression profile of 6 human bone marrow-derived MSCs preconditioned for 6 h in 2% O2 (hypoxia) or with 40 µM Vadadustat, compared to control cells and each other. RNA-Sequencing was performed using the Illumina platform, quality control with FastQC and adapter-trimming with BBDUK2. Transcripts were mapped to the Homo_sapiens. GRCh37 genome and converted to relative expression using Salmon. Differentially expressed genes (DEGs) were generated using DESeq2 while functional enrichment was performed in GSEA and g:Profiler. Comparison of hypoxia versus control resulted in 250 DEGs, Vadadustat versus control 1071, and Vadadustat versus hypoxia 1770. The terms enriched in both phenotypes referred mainly to metabolism, in Vadadustat additionally to vesicular transport, chromatin modifications and interaction with extracellular matrix. Compared with hypoxia, Vadadustat upregulated autophagic, phospholipid metabolism, and TLR cascade genes, downregulated those of cytoskeleton and GG-NER pathway and regulated 74 secretory factor genes. Our results provide valuable insight into the transcriptomic effects of these two methods of MSCs preconditioning.


Assuntos
Hipóxia Celular , Expressão Gênica , Glicina/análogos & derivados , Células-Tronco Mesenquimais , Ácidos Picolínicos/farmacologia , Adulto , Células Cultivadas , Feminino , Glicina/farmacologia , Humanos , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Pessoa de Meia-Idade , Transcriptoma
4.
Neurourol Urodyn ; 37(5): 1612-1622, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29485209

RESUMO

AIMS: The efficacy of cell therapy in patients with stress urinary incontinence (SUI) is lower than expected. The aim of this study was to determine the injection accuracy rate both with transurethral and periurethral route. METHODS: Autologous intraurethral cell transplantation was performed in female goats. The cells were injected either periurethrally (PERI group, two depots/animal, n = 8) or transurethrally (TRANS group, eight depots/animal, n = 11). Transurethral injections were performed under endoscopic guidance. The number and distribution of cell depots in urethras were analyzed in the three-step protocol: 1) screening of whole explants by in vivo imaging system; 2) systematic microscopic analysis of raw 10 µm cross-sections; 3) immunohistochemistry. As control, four urethras collected 1 day after transurethral transplantation were used. Episodes of cell suspension leakages after needle withdrawal were noted. RESULTS: In all experimental animals depots were identified in the urethral wall 28 days after transplantation. The mean percentage of depots located in the urethral wall in relation to all performed injections amounted to 68.7% and 67.0% for PERI and TRANS groups, respectively. The mean proportions of depots which were identified in external urethral sphincter (EUS) amounted 18.8% and 17.1%, respectively. Suspension leakage was observed in 19% of transurethral injections. CONCLUSIONS: Although majority of cell depots were administrated accurately into the urethral wall, the precise delivery of cells into EUS is limited regardless of injection method. The insufficient accuracy of cell delivery into EUS and cell suspension leakage can contribute to the low efficacy of cell therapy in human patients with SUI.


Assuntos
Transplante de Células-Tronco Mesenquimais/métodos , Suspensões , Uretra/diagnóstico por imagem , Animais , Terapia Baseada em Transplante de Células e Tecidos , Endoscopia , Feminino , Hemorragia/etiologia , Injeções , Curva de Aprendizado , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Reprodutibilidade dos Testes
5.
BMC Cell Biol ; 18(1): 13, 2017 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-28214472

RESUMO

BACKGROUND: Cell-based therapy is a treatment method in tendon injuries. Bone morphogenic protein 12 (BMP-12) possesses tenogenic activity and was proposed as a differentiating factor for stem cells directed to transplantation. However, BMPs belong to pleiotropic TGF-ß superfamily and have diverse effect on cells. Therefore, the aim of this study was to determine if BMP-12 induces tenogenic differentiation of human adipose stem cells (hASCs) and how it affects other features of this population. RESULTS: Human ASCs from 6 healthy donors were treated or not with BMP-12 (50 or 100 ng/ml, 7 days) and tested for gene expression (COLL1, SCX, MKH, DCN, TNC, RUNX2), protein expression (COLL1, COLL3, MKH), proliferation, migration, secretory activity, immunomodulatory properties and susceptibility to oxidative stress. RT-PCR revealed up-regulation of SCX, MKH and RUNX2 genes in BMP-12 treated cells (2.05, 2.65 and 1.87 fold in comparison to control, respectively, p < 0.05) and Western Blot revealed significant increase of COLL1 and MHK expression after BMP-12 treatment. Addition of BMP-12 significantly enhanced secretion of VEGF, IL-6, MMP-1 and MPP-8 by hASCs while had no effect on TGF-ß, IL-10, EGF and MMP-13. Moreover, BMP-12 presence in medium attenuated inhibitory effect of hASCs on allo-activated lymphocytes proliferation. At the same time BMP-12 displayed no influence on hASCs proliferation, migration and susceptibility to oxidative stress. CONCLUSION: BMP-12 activates tenogenic pathway in hASCs but also affects secretory activity and impairs immunomodulatory potential of this population that can influence the clinical outcome after cell transplantation.


Assuntos
Tecido Adiposo/citologia , Proteínas Morfogenéticas Ósseas/farmacologia , Imunomodulação/efeitos dos fármacos , Células-Tronco/citologia , Células-Tronco/imunologia , Tendões/citologia , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Humanos , Interleucina-6/metabolismo , Metaloproteinase 1 da Matriz/metabolismo , Metaloproteinase 8 da Matriz/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
6.
Gynecol Obstet Invest ; 82(3): 267-275, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27627653

RESUMO

BACKGROUND/AIM: Mesenchymal stem cells (MSCs) are gaining rising interest in gynecology and obstetrics. MSCs immunomodulatory properties are suitable enough to reduce perinatal morbidity caused by inflammation in premature neonates. The aim of this study was to evaluate and compare the ability to inhibit allo-activated lymphocytes proliferation by MSCs derived from different sources: amniotic membrane (AM), umbilical cord (UC) and adipose tissue (AT). METHODS: MSCs were isolated from AM (n = 7) and UC (n = 6) and AT (n = 6) of healthy women. Cells were characterized by flow cytometry and differentiation assay. To evaluate the potential of fetal and adult MSCs to diminish immunological response, mixed lymphocytes reaction (MLR) was performed. RESULTS: Amnion and UC-derived cells displayed typical MSCs characteristics. Addition of MSCs to MLR significantly inhibited the proliferation of stimulated lymphocytes. The effect was observed regardless of the MSCs type used (p < 0.01 in all groups). Comparative analysis revealed no significant differences in this action between tested MSCs types. Additionally, no type of MSCs significantly stimulated allogeneic lymphocytes. CONCLUSION: The results prove the immunosuppressive capacities of fetal-derived MSCs in vitro. In the future, they may be potentially used to treat premature newborn as well as in immunomodulation in post-transplant therapy.


Assuntos
Aloenxertos/imunologia , Âmnio/citologia , Células-Tronco Mesenquimais/imunologia , Cordão Umbilical/citologia , Tecido Adiposo/citologia , Adulto , Proliferação de Células , Células Cultivadas , Feminino , Humanos , Imunomodulação/imunologia , Recém-Nascido , Ativação Linfocitária/imunologia , Linfócitos/imunologia , Gravidez
7.
J Obstet Gynaecol Res ; 43(11): 1758-1768, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28707770

RESUMO

AIM: The study was conducted to investigate secretory activity and define the paracrine potential of mesenchymal stem cells from human umbilical cord and amniotic membrane (UC-MSCs and AM-MSCs, respectively). METHODS: UC-MSCs (n = 6) were obtained from tissue explants using an adherent method after two weeks of incubation. AM-MSCs (n = 6) were obtained by digestion with tripsin and collagenase. MSC phenotype was confirmed in vitro by performing flow cytometry, differentiation assays and vimentin staining. Supernatants were collected after 48 h culturing in serum-free conditions and the following concentrations were determined: epidermal growth factor (EGF), interleukin (IL)-6, IL-10, tumor necrosis factor-α, transforming growth factor-ß (TGF-ß), vascular endothelial growth factor-α (VEGF-α) and metalloproteinase (MMP) 1, 8 and 13, using multiplex supernatant cytokine assay. Data were compared with adipose tissue derived MSCs (AD-MSCs, n = 6). RESULTS: Both UC-MSC and AM-MSC populations were positively identified as MSCs by flow cytometry and differentiation potential into bone, cartilage and adipose tissue. Using a multiple cytokine detection assay, we proved that both UC-MSCs and AM-MSCs show high secretive capacity. However, the secretion profile differed between cells from various sources. UC-MSCs showed significantly higher production of TGF-ß and lower production of VEGF-α, compared to AD-MSCs (P = 0.004) and AM-MSCs (P = 0.039) and lower levels of EGF (P = 0005). AM-MSCs showed significantly lower levels of MMP-8 than UC-MSCs (P = 0.024); however, there was no difference in levels of released cytokines compared to AD-MSCs. CONCLUSION: AM-MSCs show similar IL production as AD-MSCs, while UC-MSCs have a significantly different profile, which suggests diverse biological potential of both cell types for immunomodulative and regenerative therapy.


Assuntos
Tecido Adiposo , Âmnio , Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/metabolismo , Comunicação Parácrina , Cordão Umbilical , Tecido Adiposo/citologia , Tecido Adiposo/imunologia , Tecido Adiposo/metabolismo , Âmnio/citologia , Âmnio/imunologia , Âmnio/metabolismo , Humanos , Cordão Umbilical/citologia , Cordão Umbilical/imunologia , Cordão Umbilical/metabolismo
8.
Ginekol Pol ; 88(4): 191-197, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28509320

RESUMO

OBJECTIVES: Comparison of the ability to inhibit alloactivated lymphocytes proliferation of human Wharton Jelly (WJ) and amniotic membrane (AM) mesenchymal stem cells (MSCs) from preterm and term pregnancies. MATERIAL AND METHODS: Term-WJ-MSCs (n = 5) and Preterm-WJ-MSCs (n = 1) were obtained from tissue explants by adherent method. Term-AM-MSCs (n = 5) and Preterm-AM-MSCs (n = 1) were obtained by tripsin and collagenase digestion method. Term and Preterm MSCs phenotype was confirmed in vitro by flow cytometry. To evaluate the potential of fetal and adult MSCs to diminish immunological response mixed lymphocytes reaction (MLR) has been performed. RESULTS: Term and Preterm cells were positively identified as MSCs by the expression of CD73 and CD90 and CD105 with simultaneous absence of CD11b, CD14, CD19, CD34, CD45 and HLA-DR. The mean inhibition of allostimulated lymphocytes after addition of fetal derived MSCs amounted 64.8% for term AM-MSCs and 42.1% for term WJ-MSCs (for both populations the effect was statistically significant, p < 0.01). The addition of preterm-MSCs to MLR resulted in reduction of stimulated lymphocytes proliferation by 64.9% for AM-MSCs and 86.1% for WJ-MSCs. CONCLUSIONS: Presented results suggest that preterm fetal tissues contain MSCs which posses similar immunosuppressive capacity as those from term pregnancies. In the future MSCs from the umbilical cord and amnion can be potentially used to prevent immuno-dependent injuries in premature newborns.


Assuntos
Âmnio/citologia , Proliferação de Células , Linfócitos/citologia , Células-Tronco Mesenquimais/citologia , Nascimento Prematuro , Geleia de Wharton/citologia , Feminino , Feto/citologia , Citometria de Fluxo , Humanos , Gravidez
9.
Int J Urol ; 23(8): 701-5, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27225822

RESUMO

OBJECTIVES: To investigate the effects of propofol and isoflurane on urethral pressure profilometry of female dogs and goats, and to identify the method of anesthesia that least influences urethral pressure profilometry and to assess its reproducibility. METHODS: The effects of premedication with midazolam, propofol sedation and isoflurane anesthesia were assessed in five female dogs. The effects of propofol and isoflurane were compared in seven goats, whereas in another group of 19 goats, the state of deep propofol sedation was compared with the state of recovery from propofol sedation. The coefficient of reproducibility and within-subject coefficient of variation were calculated to evaluate test-retest reproducibility. RESULTS: In conscious female dogs, maximal urethral closure pressure and functional area were significantly higher than under propofol or isoflurane (P = 0.04), but not different from the recovery state. In six of seven goats, maximal urethral closure pressure and functional area were higher when measured under propofol sedation than under isoflurane (median maximal urethral closure pressure, 69 vs 47 cmH2 O; P = 0.03). Maximal urethral closure pressure was lower under propofol than during recovery from propofol in 17 of 19 goats (median maximal urethral closure pressure, 54 vs 66 cmH2 O; P < 0.001). The test-retest coefficient of reproducibility for goats was 28 cmH2 O, and the within-subject coefficient of variation was 16%. CONCLUSIONS: In dogs, urethral pressure profilometry should be measured in conscious animals whenever possible. In goats, urethral pressure profilometry is least affected during recovery from propofol sedation, and it shows acceptable reproducibility under this condition.


Assuntos
Anestesia , Anestésicos Intravenosos/farmacologia , Propofol/farmacologia , Uretra/efeitos dos fármacos , Animais , Cães , Feminino , Cabras , Isoflurano , Pressão , Reprodutibilidade dos Testes
10.
Mediators Inflamm ; 2014: 482352, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25242868

RESUMO

The vast majority of myoblasts transplanted into the skeletal muscle die within the first week after injection. Inflammatory response to the intramuscular cell transfer was studied in allogeneic but not in autologous model. The aim of this study was to evaluate immune reaction to autotransplantation of myogenic cells and to assess its dynamics within the first week after injection. Muscle-derived cells or medium alone was injected into the intact skeletal muscles in autologous model. Tissue samples were collected 1, 3, and 7 days after the procedure. Our analysis revealed the peak increase of the gene expression of all evaluated cytokines (Il-1α, Il-1 ß, Il-6, Tgf-ß, and Tnf-α) at day 1. The mRNA level of analyzed cytokines normalized in subsequent time points. The increase of Il- ß gene expression was further confirmed at the protein level. Analysis of the tissue sections revealed rapid infiltration of injected cell clusters with neutrophils and macrophages. The inflammatory infiltration was almost completely resolved at day 7. The survived cells were able to participate in the muscle regeneration process. Presented results demonstrate that autotransplanted muscle-derived cells induce classical early immune reaction in the site of injection which may contribute to cellular graft elimination.


Assuntos
Músculo Esquelético/imunologia , Transplante Autólogo , Animais , Células Cultivadas , Interleucina-1alfa/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , RNA Mensageiro/genética , Ratos , Fator de Crescimento Transformador beta/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
11.
Cytokine ; 64(1): 243-50, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23859809

RESUMO

Mesenchymal stem cells due to the high proliferative potential, capacity of multilineage differentiation became a hope of regenerative medicine. However, the organism's response to the transplantation of MSCs is not fully elucidated. The aim of the present study was to evaluate the acute local tissue response to syngeneic MSCs administration into the muscle. Rat syngeneic MSCs were transplanted into the skeletal muscle and the tissue surrounding the injection site was collected after 24h. Analogous samples from untreated and PBS treated muscles served as controls. The analysis of genes expression using real-time PCR revealed significant up-regulation of proinflammatory cytokines: IL-1α, IL-1ß, IL-6 in MSCs treated muscles in comparison to the PBS group. The evaluation of protein concentration (ELISA) in collected samples showed that injection of MSCs caused significant elevation of IL-1ß. Immunofluorescent assessment of the tissue revealed infiltration of leukocytes and macrophages. Quantitative analysis of the samples immunostained for CD68 and CD43 antigens revealed that the number of phagocytes was significantly higher in MSC treated muscle when compared to the samples from PBS group. To conclude, the administration of mesenchymal stem cells into the muscle in syngeneic model induces the features of acute inflammation that affects cell engraftment.


Assuntos
Interleucina-1alfa/biossíntese , Interleucina-1beta/biossíntese , Interleucina-6/biossíntese , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Adipogenia/fisiologia , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Diferenciação Celular , Inflamação/imunologia , Interleucina-1alfa/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Contagem de Leucócitos , Leucócitos/citologia , Leucossialina/metabolismo , Macrófagos/citologia , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Osteogênese/fisiologia , Ratos , Ratos Endogâmicos Lew , Regulação para Cima
12.
Biomedicines ; 11(2)2023 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-36830980

RESUMO

The ability of MSCs to modulate the inflammatory environment is well recognized, but understanding the molecular mechanisms responsible for these properties is still far from complete. Prostaglandin E2 (PGE2), a product of the cyclooxygenase 2 (COX-2) pathway, is indicated as one of the key mediators in the immunomodulatory effect of MSCs. Due to the pleiotropic effect of this molecule, determining its role in particular intercellular interactions and aspects of cell functioning is very difficult. In this article, the authors attempt to summarize the previous observations regarding the role of PGE2 and COX-2 in the immunomodulatory properties and other vital functions of MSCs. So far, the most consistent results relate to the inhibitory effect of MSC-derived PGE2 on the early maturation of dendritic cells, suppressive effect on the proliferation of activated lymphocytes, and stimulatory effect on the differentiation of macrophages into M2 phenotype. Additionally, COX-2/PGE2 plays an important role in maintaining the basic life functions of MSCs, such as the ability to proliferate, migrate and differentiate, and it also positively affects the formation of niches that are conducive to both hematopoiesis and carcinogenesis.

13.
Stem Cell Res Ther ; 14(1): 200, 2023 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-37563650

RESUMO

BACKGROUND: Advanced renal cell carcinoma (RCC) is therapeutically challenging. RCC progression is facilitated by mesenchymal stem/stromal cells (MSCs) that exert remarkable tumor tropism. The specific mechanisms mediating MSCs' migration to RCC remain unknown. Here, we aimed to comprehensively analyze RCC secretome to identify MSCs attractants. METHODS: Conditioned media (CM) were collected from five RCC-derived cell lines (Caki-1, 786-O, A498, KIJ265T and KIJ308T) and non-tumorous control cell line (RPTEC/TERT1) and analyzed using cytokine arrays targeting 274 cytokines in addition to global CM proteomics. MSCs were isolated from bone marrow of patients undergoing standard orthopedic surgeries. RCC CM and the selected recombinant cytokines were used to analyze their influence on MSCs migration and microarray-targeted gene expression. The expression of genes encoding cytokines was evaluated in 100 matched-paired control-RCC tumor samples. RESULTS: When compared with normal cells, CM from advanced RCC cell lines (Caki-1 and KIJ265T) were the strongest stimulators of MSCs migration. Targeted analysis of 274 cytokines and global proteomics of RCC CM revealed decreased DPP4 and EGF, as well as increased AREG, FN1 and MMP1, with consistently altered gene expression in RCC cell lines and tumors. AREG and FN1 stimulated, while DPP4 attenuated MSCs migration. RCC CM induced MSCs' transcriptional reprogramming, stimulating the expression of CD44, PTX3 and RAB27B. RCC cells secreted hyaluronic acid (HA), a CD44 ligand mediating MSCs' homing to the kidney. AREG emerged as an upregulator of MSCs' transcription. CONCLUSIONS: Advanced RCC cells secrete AREG, FN1 and HA to induce MSCs migration, while DPP4 loss prevents its inhibitory effect on MSCs homing. RCC secretome induces MSCs' transcriptional reprograming to facilitate their migration. The identified components of RCC secretome represent potential therapeutic targets.


Assuntos
Carcinoma de Células Renais , Neoplasias Renais , Células-Tronco Mesenquimais , Humanos , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Dipeptidil Peptidase 4/metabolismo , Secretoma , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Células-Tronco Mesenquimais/metabolismo , Citocinas/metabolismo , Meios de Cultivo Condicionados/farmacologia , Meios de Cultivo Condicionados/metabolismo
14.
Muscle Nerve ; 45(1): 32-8, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22190303

RESUMO

INTRODUCTION: The possible reason for elimination of myogenic cells after transplantation is inflammation at the injection site associated with oxidative stress. The aim of this study was to determine whether preconditioning of muscle-derived cells with an antioxidant, sodium ascorbate, can influence the fate of transplanted cells. METHODS: Autologous transplantation of muscle-derived cells was performed in rabbits. Isolated cells were identified, lipofected with ß-galactosidase, preincubated or not with sodium ascorbate, and injected intramuscularly. RESULTS: Two weeks after autologous transplantation in the edge of a previous muscle defect, donor cells formed multinucleated young myotubes. Pretreatment of cells with sodium ascorbate before injection resulted in a significant increase of donor cells at the injection site 2 weeks after transfer. CONCLUSIONS: These results show that: (1) preincubation with antioxidant can increase the efficacy of myogenic cell transplantation; and (2) oxidative stress may play a role in elimination of cells after autologous transplantation.


Assuntos
Antioxidantes/farmacologia , Ácido Ascórbico/farmacologia , Miosite , Transplante Autólogo/efeitos adversos , Animais , Sobrevivência Celular/efeitos dos fármacos , Transplante de Células/efeitos adversos , Células Cultivadas , Desmina/metabolismo , Modelos Animais de Doenças , Proteínas de Fluorescência Verde/genética , Injeções Intramusculares , Fibras Musculares Esqueléticas , Miosite/tratamento farmacológico , Miosite/etnologia , Miosite/cirurgia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Coelhos , Estatísticas não Paramétricas , Transfecção/métodos , beta-Galactosidase/genética , beta-Galactosidase/metabolismo
15.
Int J Urol ; 19(7): 676-82, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22548443

RESUMO

OBJECTIVES: To describe a novel animal model of intrinsic sphincter deficiency. METHODS: The study was carried out on 10 female pigs. Injury to the urethral sphincter was induced by distension of the urethra. This was obtained by using the balloon of an 18-F Dufour catheter for 5 min followed by its retraction through the urethra without draining the balloon. The urethral pressure profile was evaluated before injury, immediately postinjury and at day 28 postinjury in the experimental group (n = 5), and on day 1 and day 28 in the control uninjured group (n = 5). The maximal urethral closure pressure, the functional urethral length and the area under curve of the urethral pressure profile were measured. RESULTS: The mean maximal urethral closure pressure at the beginning of the experiment was 32 cmH(2) O, and the mean functional urethral length was 4.88 cm. The assessment at day 28 showed a reduction of the maximal urethral closure pressure (50% of the control, P > 0.05), the functional urethral length (52.5% of the control, P < 0.05) and the area under curve (52% of the control, P < 0.05) in injured pigs. Histologically, a fibrosis of the sphincter was detected without rupture of the muscle layer in all the samples. CONCLUSIONS: The proposed porcine model can be used to obtain intrinsic sphincter deficiency-like urodynamic findings without rupturing the sphincter. This methodology can be applied to investigate therapies for intrinsic sphincter deficiency.


Assuntos
Cateterismo , Modelos Animais de Doenças , Uretra/lesões , Uretra/fisiopatologia , Incontinência Urinária por Estresse/fisiopatologia , Animais , Área Sob a Curva , Peso Corporal , Feminino , Pressão , Estatísticas não Paramétricas , Suínos , Uretra/patologia , Urodinâmica
16.
Biomolecules ; 12(2)2022 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-35204788

RESUMO

Mesenchymal stromal cells (MSCs) are able to modulate the immune system activity and the regeneration processes mainly through the secretion of multiple soluble factors, including prostaglandin E2 (PGE2). PGE2 is produced as a result of cyclooxygenases (COX) activity. In the present study, we investigated how ibuprofen, a nonselective COX inhibitor, affects the proliferation, migration and secretion of human bone marrow MSCs (hBM-MSCs). For this purpose, six hBM-MSCs populations were treated with ibuprofen at doses which do not differ from maximum serum concentrations during standard pharmacotherapy. Ibuprofen treatment (25 or 50 µg/mL) substantially reduced the secretion of PGE2 in all tested populations. Following ibuprofen administration, MSCs were subjected to proliferation (BrdU), transwell migration, and scratch assays, while its effect on MSCs secretome was evaluated by Proteome Profiler and Luminex immunoassays. Ibuprofen did not cause statistically significant changes in the proliferation rate and migration ability of MSCs (p > 0.05). However, ibuprofen (25 µg/mL for 3 days) significantly decreased mean secretion of: CCL2 (by 44%), HGF (by 31%), IL-6 (by 22%), VEGF (by 20%) and IL-4 (by 8%) compared to secretion of control MSCs (p < 0.05). Our results indicate that ibuprofen at therapeutic concentrations may impair the pro-regenerative properties of hBM-MSCs.


Assuntos
Ibuprofeno , Células-Tronco Mesenquimais , Medula Óssea , Células da Medula Óssea , Proliferação de Células , Humanos , Ibuprofeno/farmacologia
17.
J Immunother Cancer ; 10(1)2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35078921

RESUMO

BACKGROUND: Immune checkpoint inhibitors and chimeric antigen receptor (CAR)-based therapies have transformed cancer treatment. Recently, combining these approaches into a strategy of PD-L1-targeted CAR has been proposed to target PD-L1high tumors. Our study provides new information on the efficacy of such an approach against PD-L1low targets. METHODS: New atezolizumab-based PD-L1-targeted CAR was generated and introduced into T, NK, or NK-92 cells. Breast cancer MDA-MB-231 and MCF-7 cell lines or non-malignant cells (HEK293T, HMEC, MCF-10A, or BM-MSC) were used as targets to assess the reactivity or cytotoxic activity of the PD-L1-CAR-bearing immune effector cells. Stimulation with IFNγ or with supernatants from activated CAR T cells were used to induce upregulation of PD-L1 molecule expression on the target cells. HER2-CAR T cells were used for combination with PD-L1-CAR T cells against MCF-7 cells. RESULTS: PD-L1-CAR effector cells responded vigorously with degranulation and cytokine production to PD-L1high MDA-MB-231 cells, but not to PD-L1low MCF-7 cells. However, in long-term killing assays, both MDA-MB-231 and MCF-7 cells were eliminated by the PD-L1-CAR cells, although with a delay in the case of PD-L1low MCF-7 cells. Notably, the coculture of MCF-7 cells with activated PD-L1-CAR cells led to bystander induction of PD-L1 expression on MCF-7 cells and to the unique self-amplifying effect of the PD-L1-CAR cells. Accordingly, PD-L1-CAR T cells were active not only against MDA-MD-231 and MCF-7-PD-L1 but also against MCF-7-pLVX cells in tumor xenograft models. Importantly, we have also observed potent cytotoxic effects of PD-L1-CAR cells against non-malignant MCF-10A, HMEC, and BM-MSC cells, but not against HEK293T cells that initially did not express PD-L1 and were unresponsive to the stimulation . Finally, we have observed that HER-2-CAR T cells stimulate PD-L1 expression on MCF-7 cells and therefore accelerate the functionality of PD-L1-CAR T cells when used in combination. CONCLUSIONS: In summary, our studies show that CAR-effector cells trigger the expression of PD-L1 on target cells, which in case of PD-L1-CAR results in the unique self-amplification phenomenon. This self-amplifying effect could be responsible for the enhanced cytotoxicity of PD-L1-CAR T cells against both malignant and non-malignant cells and implies extensive caution in introducing PD-L1-CAR strategy into clinical studies.


Assuntos
Neoplasias da Mama/terapia , Citotoxicidade Imunológica , Inibidores de Checkpoint Imunológico/uso terapêutico , Imunoterapia Adotiva/métodos , Receptores de Antígenos Quiméricos/imunologia , Animais , Antígeno B7-H1/análise , Antígeno B7-H1/antagonistas & inibidores , Linhagem Celular Tumoral , Feminino , Células HEK293 , Humanos , Camundongos , Receptor ErbB-2/antagonistas & inibidores , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Cell Biol Int ; 35(10): 1055-62, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21592091

RESUMO

It is generally accepted that autologous transfers, as non-immunogenic, constitute the safest approach in cellular transplantations. However, this attitude is often associated with the need for isolation and extracorporeal propagation of cells derived from aged patients. Thus the knowledge about relationship between aging and the properties of MSCs (mesenchymal stem cells) is crucial in developing new clinical strategies. The aim of this study was to perform complex comparison of MSC derived from young and aged individuals, which included phenotype, proliferating rate, osteogenic and adipogenic potential and secretory activity. Evaluated populations were isolated from bone marrow of 3-month-old and 24-month-old rats. There was no significant difference in membrane antigen expression and PDT (population doubling time). Additionally, the adipogenic and osteogenic potential did not vary between studied populations. The reaction of MSCs to either mitogen [bFGF (basic fibroblas t growth factor)] or oxidative stress (H2O2) in vitro displayed a very similar pattern in both analysed populations. There was no difference in TGFß1 (transforming growth factor ß1) and VEGF (vascular endothelial growth factor) secretion measured by ELISA test and gene expression evaluated by real-time PCR. However, the expression of the gene for IL-1α (interleukin-1α) was 8-fold lower in oMSC (MSC isolated from old rats). These results indicate that aging individuals can be considered as candidates for autologous transplantation of bone-marrow-derived MSCs.


Assuntos
Células da Medula Óssea/citologia , Células-Tronco Mesenquimais/citologia , Adipogenia , Fatores Etários , Animais , Sobrevivência Celular , Fator 2 de Crescimento de Fibroblastos/farmacologia , Peróxido de Hidrogênio/farmacologia , Imunofenotipagem , Interleucina-1alfa/metabolismo , Proteínas de Membrana/metabolismo , Células-Tronco Mesenquimais/metabolismo , Osteogênese , Ratos , Ratos Endogâmicos Lew , Fator de Crescimento Transformador beta1/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
19.
Stem Cell Res Ther ; 12(1): 386, 2021 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-34233726

RESUMO

Autoimmune hepatitis is a chronic inflammatory hepatic disorder which may cause liver fibrosis. Appropriate treatment of autoimmune hepatitis is therefore important. Adult stem cells have been investigated as therapies for a variety of disorders in latest years. Hematopoietic stem cells (HSCs) were the first known adult stem cells (ASCs) and can give rise to all of the cell types in the blood and immune system. Originally, HSC transplantation was served as a therapy for hematological malignancies, but more recently researchers have found the treatment to have positive effects in autoimmune diseases such as multiple sclerosis. Mesenchymal stem cells (MSCs) are ASCs which can be extracted from different tissues, such as bone marrow, adipose tissue, umbilical cord, and dental pulp. MSCs interact with several immune response pathways either by direct cell-to-cell interactions or by the secretion of soluble factors. These characteristics make MSCs potentially valuable as a therapy for autoimmune diseases. Both ASC and ASC-derived exosomes have been investigated as a therapy for autoimmune hepatitis. This review aims to summarize studies focused on the effects of ASCs and their products on autoimmune hepatitis.


Assuntos
Hepatite Autoimune , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Tecido Adiposo , Hepatite Autoimune/terapia , Humanos , Cordão Umbilical
20.
Pharmaceutics ; 13(11)2021 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-34834238

RESUMO

Despite intensive clinical research on the use of mesenchymal stromal cells (MSCs), further basic research in this field is still required. Herein, we compared human bone marrow MSCs (BM-MSCs, n = 6) and Wharton's jelly MSCs (WJ-MSCs, n = 6) in their ability to interact with human primary macrophages. Evaluation of secretory potential revealed that under pro-inflammatory stimulation, WJ-MSCs secreted significantly more IL-6 than BM-MSCs (2-fold). This difference did not translate into the effect of MSCs on macrophages: both types of MSCs significantly directed M1-like macrophages toward the M2 phenotype (based on CD206 expression) to a similar extent. This observation was consistent both in flow cytometry analysis and immunocytochemical assessment. The effect of MSCs on macrophages was sustained when IL-6 signaling was blocked with Tocilizumab. Macrophages, regardless of polarization status, enhanced chemotaxis of both BM-MSCs and WJ-MSCs (p < 0.01; trans-well assay), with WJ-MSCs being significantly more responsive to M1-derived chemotactic signals than BM-MSCs. Furthermore, WJ-MSCs increased their motility (scratch assay) when exposed to macrophage-conditioned medium while BM-MSCs did not. These results indicate that although both BM-MSCs and WJ-MSCs have the ability to reciprocally interact with macrophages, the source of MSCs could slightly but significantly modify the response under clinical settings.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa