Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Mol Biol Rep ; 51(1): 734, 2024 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-38874773

RESUMO

BACKGROUND: Liver cirrhosis, a prevalent chronic liver disease, is characterized by liver fibrosis as its central pathological process. Recent advancements highlight the clinical efficacy of umbilical cord mesenchymal stem cell (UC-MSC) therapy in the treatment of liver cirrhosis. METHODS AND RESULTS: We investigated the pharmacodynamic effects of UC-MSCs and MSC conditional medium (MSC-CM) in vivo, utilizing a carbon tetrachloride (CCl4)-induced fibrotic rat model. Concurrently, we assessed the in vitro impact of MSCs and MSC-CM on various cellular process of hepatic stellate cells (HSCs), including proliferation, apoptosis, activation, immunomodulatory capabilities, and inflammatory factor secretion. Our results indicate that both MSCs and MSC-CM significantly ameliorate the pathological extent of fibrosis in animal tissues, reducing the collagen content, serum biochemical indices and fibrosis biomarkers. In vitro, MSC-CM significantly inhibited the activation of the HSC line LX-2. Notably, MSC-CM modulated the expression of type I procollagen and TGFß-1 while increasing MMP1 expression. This modulation restored the MMP1/TIMP1 ratio imbalance and extracellular matrix deposition in TGFß-1 induced fibrosis. Both MSCs and MSC-CM not only induced apoptosis in HSCs but also suppressed proliferation and inflammatory cytokine release from activated HSCs. Furthermore, MSCs and MSC-CM exerted a suppressive effect on total lymphocyte activation. CONCLUSIONS: UC-MSCs and MSC-CM primarily modulate liver fibrosis severity by regulating HSC activation. This study provides both in vivo and in vitro pharmacodynamic evidence supporting the use of MSCs in liver fibrosis treatment.


Assuntos
Apoptose , Proliferação de Células , Células Estreladas do Fígado , Cirrose Hepática , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Cordão Umbilical , Células Estreladas do Fígado/metabolismo , Células-Tronco Mesenquimais/metabolismo , Animais , Humanos , Cirrose Hepática/patologia , Cirrose Hepática/terapia , Cirrose Hepática/metabolismo , Cordão Umbilical/citologia , Ratos , Transplante de Células-Tronco Mesenquimais/métodos , Masculino , Tetracloreto de Carbono , Modelos Animais de Doenças , Meios de Cultivo Condicionados/farmacologia , Ratos Sprague-Dawley , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Linhagem Celular , Citocinas/metabolismo
2.
Eur J Orthop Surg Traumatol ; 33(3): 669-675, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35699826

RESUMO

BACKGROUND: Outcomes of the current management of posterior cruciate ligament (PCL) rupture are still unsatisfactory. Recent literature demonstrated the efficacy of the paracrine action of mesenchymal stem cells (MSC) in ligament rupture healing. This study aimed to evaluate the outcome of arthroscopic administration of allogeneic umbilical cord-derived MSC (UC-MSC) conditioned medium (secretome) for the treatment of PCL rupture. PATIENTS AND METHODS: This is a prospective study including 12 individuals with PCL rupture grade 1 or 2 who were performed arthroscopy and secretome administrations. The functional and radiologic outcome of the knee was examined one year following intervention. RESULTS: Preoperatively, posterior drawer test revealed three cases of grade 2+ and nine cases of grade 1+, whereas the final follow-up revealed two cases of grade 2+ and ten cases of grade 1+ PCL rupture. At final follow-up, the mean scores for the IKDC, modified Cincinnati, and Lysholm were 90.58 ± 4.30, 90.90 ± 2.15, and 89.42 ± 3.16, respectively. The means of the serial hop tests were 90.33, 94.16, 93.66, and 95.33 for single, triple, crossover, and time hop tests, respectively. Five patients were able to resume competitive sport after an average of 25.8 weeks (25-38). The final MRI analysis revealed that six knees (50%) regained PCL continuity with low signal intensity, five knees (41.6%) returned near-normal PCL continuity, and one knee (8.3%) regained PCL continuity but with deformed outlines. CONCLUSIONS: Short-term follow-up indicated that the secretome generated from allogenic UC-MSC produces excellent functional and radiographic results in grade I-II PCL rupture.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Instabilidade Articular , Traumatismos do Joelho , Procedimentos de Cirurgia Plástica , Ligamento Cruzado Posterior , Humanos , Ligamento Cruzado Posterior/cirurgia , Estudos Prospectivos , Meios de Cultivo Condicionados , Secretoma , Resultado do Tratamento , Seguimentos , Articulação do Joelho , Traumatismos do Joelho/cirurgia , Artroscopia/métodos , Instabilidade Articular/cirurgia
3.
Cytotherapy ; 24(2): 205-212, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34799271

RESUMO

BACKGROUND AIMS: Despite the great advances in immunosuppressive therapy for severe aplastic anemia (SAA), most patients are not completely cured. Haploidentical hematopoietic stem cell transplantation (haplo-HSCT) has been recommended as an alternative treatment in adult SAA patients. However, haplo-HSCT presents a higher incidence of graft failure and graft-versus-host disease (GVHD). The authors designed a combination of haplo-HSCT and umbilical cord-derived mesenchymal stem cells (UC-MSCs) for treatment of SAA in adult patients and evaluated its effects. METHODS: Adult patients (≥18 years) with SAA (N = 25) were given HLA-haploidentical hematopoietic stem cells (HSCs) combined with UC-MSCs after a conditioning regimen consisting of busulfan, cyclophosphamide, fludarabine and anti-thymocyte globulin and intensive GVHD prophylaxis, including cyclosporine, basiliximab, mycophenolate mofetil and short-term methotrexate. Additionally, the effects of the protocol in adult SSA patients were compared with those observed in juvenile SAA patients (N = 75). RESULTS: All patients achieved myeloid engraftment after haplo-HSCT at a median of 16.12 days (range, 11-26). The median time of platelet engraftment was 28.30 days (range, 13-143). The cumulative incidence of grade II acute GVHD (aGVHD) at day +100 was 32.00 ± 0.91%. No one had grade III-IV aGVHD at day +100. The cumulative incidence of total chronic GVHD was 28.00 ± 0.85%. The overall survival was 71.78 ± 9.05% at a median follow-up of 42.08 months (range, 2.67-104). Promisingly, the protocol yielded a similar curative effect in both young and adult SAA patients. CONCLUSIONS: The authors' data suggest that co-transplantation of HLA-haploidentical HSCs and UC-MSCs may provide an effective and safe treatment for adult SAA.


Assuntos
Anemia Aplástica , Doença Enxerto-Hospedeiro , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Mesenquimais , Anemia Aplástica/terapia , Células-Tronco Hematopoéticas , Humanos , Condicionamento Pré-Transplante
4.
Immunopharmacol Immunotoxicol ; 40(2): 168-172, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29355065

RESUMO

BACKGROUND: The objective of this study is to investigate the effect of combination of umbilical cord-derived mesenchymal stem cell (UC-MSC) and vitamin E (VitE) on inflammation in mice with acute kidney injury (AKI). METHODS: UC-MSCs were isolated from pregnant wistar mice and cultured. A total of 90 female wistar mice were randomly divided into control group, AKI group, AKI + VitE group, AKI + UC-MSC group, and AKI + VitE + UC-MSC group (18 mice in each group) which were given no treatment, normal saline, VitE, UC-MSC, and VitE + UC-MSC, respectively. The renal pedicles on both sides were clipped for 50 min with micro-artery clips to induce AKI. Six mice were sacrificed at days 1, 3, and 7, while blood and kidney tissues were collected to detect levels of blood urea nitrogen (BUN) and creatinine (Scr). Kidney tissues were stained by HE staining to observe pathological changes; levels of interleukin-lß, TNF-α, interleukin-10, and ß-FGF were measured by ELISA. RESULTS: Compared with the control group, AKI mice showed higher levels of serum BUN and Scr, tubular swelling and necrosis suggesting that AKI model was successfully established. Mice in AKI + VitE group, AKI + UC-MSC group, and AKI + VitE + UC-MSC presented better renal function than mice of AKI group. Mice from AKI + VitE + UC-MSC group showed the best renal function with the least renal tubular injury (p < .05). ELISA detection revealed that pro-inflammatory cytokines were significantly increased and anti-inflammatory cytokine levels were significantly decreased in all time points (p < .05). VitE, UC-MSC, and VitE + UC-MSC resulted in the increase of anti-inflammatory cytokine levels and reduction of pro-inflammatory cytokine levels and the combination of VitE and UC-MSC performed favorable effect in the suppression of inflammation in AKI mice (p < .05). CONCLUSIONS: Combination of UC-MSC and VitE significantly inhibited inflammatory reaction in kidney through the regulation of inflammatory cytokines in the microenvironment of kidney with AKI. Combination of UC-MSC and VitE presented therapeutic effect on AKI than the single use of UC-MSC or VitE.


Assuntos
Injúria Renal Aguda/terapia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Cordão Umbilical/metabolismo , Vitamina E/farmacologia , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Aloenxertos , Animais , Feminino , Inflamação/metabolismo , Inflamação/patologia , Inflamação/terapia , Células-Tronco Mesenquimais/patologia , Camundongos , Cordão Umbilical/patologia
5.
In Vitro Cell Dev Biol Anim ; 60(3): 307-319, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38421574

RESUMO

Human umbilical cord-mesenchymal stem cells (hUC-MSCs) have been widely investigated as a new therapeutic agent to treat injuries and inflammatory-mediated and autoimmune diseases. Previous studies have reported on the safety of low-dose infusion of hUC-MSCs, but information on the cell behaviour at higher doses and frequency of injection of the cells remains uncertain. The aim of the present study was to demonstrate the safety and efficacy of hUC-MSCs by Cytopeutics® (Selangor, Malaysia) from low to an extremely high dose in different monitoring periods in healthy BALB/c mice as well as assessing the tumorigenicity of the cells in B-NDG SCID immunocompromised mice. Umbilical cord from two healthy human newborns was obtained and the isolation of the hUC-MSCs was performed based on previous established method. Assessment of the cells at different doses of single or multiple administrations was performed on healthy BALB/c mice in dose range finding, sub-acute (7 d and 28 d) and sub-chronic periods (90 d). Tumorigenicity potential of Cytopeutics® hUC-MSCs was also evaluated on B-NDG immunocompromised mice for 26 wk. Single or multiple administrations of Cytopeutics® hUC-MSCs up to 40 × 106 cells per kilogramme of body weight (kg BW) were found to have no adverse effect in terms of clinical symptoms, haematology and other laboratory parameters, and histology examination in healthy BALB/c mice. hUC-MSCs were also found to reduce pro-inflammatory cytokines (IL-6 and TNF-α) in a dose-dependent manner. No sign of tumor formation was observed in B-NDG mice in the 26-wk tumorigenicity assessment. Single or multiple administration of allogenic Cytopeutics® hUC-MSCs was safe even at very high doses, is non-tumorigenic and did not cause adverse effects in mice throughout the evaluation periods. In addition, Cytopeutics® hUC-MSCs exhibited immunomodulatory effect in a dose-dependent manner.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Humanos , Camundongos , Animais , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Transplante de Células-Tronco Mesenquimais/métodos , Camundongos SCID , Cordão Umbilical
6.
Front Cell Dev Biol ; 11: 1146835, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37274737

RESUMO

This century's first major epidemic of a new coronavirus illness (2019-nCoV) was a tremendous shock to the healthcare system. The onset of the pandemic has caused severe economic and health shortages. At this time, there are no viable treatments for COVID-19. Several clinical studies using cell-based therapies, such as umbilical cord mesenchymal stem cells, have showed promising results (UC-MSCs). UC-MSCs have been the focus of much study because to their potential as a treatment option for COVID-19 patients. Cytokine release syndrome, often called cytokine storm, increases the risk of morbidity and mortality from COVID-19. It has been established that UC-MSCs may suppress and control both the adaptive and innate immune responses by modulating the release of immunostimulatory cytokines. The purpose of this study is to assess and clarify the use of UC-MSCs for the treatment of ARDS caused by COVID-19.

7.
Front Med (Lausanne) ; 10: 1051831, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36744151

RESUMO

Stem cell therapy, which has promising results in acute disorders such as stroke, supports treatment by providing rehabilitation in the chronic stage patients. In acute stroke, thrombolytic medical treatment protocols are clearly defined in neurologic emergencies, but in neurologic patients who miss the "thrombolytic treatment intervention window," or in cases of hypoxic-ischemic encephalopathy, our hands are tied, and we are still unfortunately faced with hopeless clinical implementations. We consider mesenchymal stem cell therapy a viable option in these cases. In recent years, novel research has focused on neuro-stimulants and supportive and combined therapies for stroke. Currently, available treatment options are limited, and only certain patients are eligible for acute treatment. In the scope of our experience, five stroke patients were evaluated in this study, who was treated with a single dose of 1-2 × 106 cells/kg allogenic umbilical cord-mesenchymal stem cells (UC-MSCs) with the official confirmation of the Turkish Ministry of Health Stem Cell Commission. The patients were followed up for 12 months, and clinical outcomes are recorded. NIH Stroke Scale/Scores (NIHSS) decreased significantly (p = 0.0310), and the Rivermead Assessment Scale (RMA) increased significantly (p = 0.0234) for all patients at the end of the follow-up. All the patients were followed up for 1 year within a rehabilitation program. Major clinical outcome improvements were observed in the overall clinical conditions of the UC-MSC treatment patients. We observed improvement in the patients' upper extremity and muscle strength, spasticity, and fine motor functions. Considering recent studies in the literature together with our results, allogenic stem cell therapies are introduced as promising novel therapies in terms of their encouraging effects on physiological motor outcomes.

8.
Narra J ; 3(3): e225, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38455624

RESUMO

Hypoxia-inducible factor-1 alpha (HIF-1α) is a transcription factor that plays a crucial role in cellular responses to hypoxia, such as in the development of intimal hyperplasia, a common complication in arteriovenous fistula (AVF) creation. While the application of umbilical cord mesenchymal stem cells (UC-MSCs) has shown promise in various regenerative medicine applications, including tissue repair and angiogenesis, the effect of UC-MSCs on HIF-1α level in the AVF has not been tested. Therefore, the aim of this study was to evaluate the effect of UC-MSCs administration on HIF-1α levels in the AVF animal model. An experimental study was conducted on 28 local male rabbits (Lepus domestica) using a post-test-only design. The rabbits were divided randomly into four groups: normal rabbit group (negative control), placebo-treated AVF rabbit group (positive control), AVF rabbits treated with in-situ UC-MSCs injection (one dose, 106 UC-MSCs/kg body weight), and AVF rabbits treated with intravenous UC-MSCs (one dose, 106 UC-MSCs/kg body weight (BW). HIF-1α level was measured using ELISA method after 28 days post-treatment. All data were analyzed using the one-way analysis of variance (ANOVA) and continued with the Duncan's post-hoc test. The data indicated that the levels of HIF-1α were different among all four groups (p<0.001). The post-hoc analysis revealed that the HIF-1α levels in both UC-MSC treated groups were significantly lower compared to untreated AVF rabbits (p<0.05). This study suggests that UC-MSCs could be a promising therapy to prevent and reduce intimal hyperplasia in AVF.

9.
Front Cell Dev Biol ; 11: 1089817, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36875761

RESUMO

Type 1 diabetes mellitus (T1DM) is an autoimmune disorder specifically targeting pancreatic islet beta cells. Despite many efforts focused on identifying new therapies able to counteract this autoimmune attack and/or stimulate beta cells regeneration, TD1M remains without effective clinical treatments providing no clear advantages over the conventional treatment with insulin. We previously postulated that both the inflammatory and immune responses and beta cell survival/regeneration must be simultaneously targeted to blunt the progression of disease. Umbilical cord-derived mesenchymal stromal cells (UC-MSC) exhibit anti-inflammatory, trophic, immunomodulatory and regenerative properties and have shown some beneficial yet controversial effects in clinical trials for T1DM. In order to clarify conflicting results, we herein dissected the cellular and molecular events derived from UC-MSC intraperitoneal administration (i.p.) in the RIP-B7.1 mouse model of experimental autoimmune diabetes. Intraperitoneal (i.p.) transplantation of heterologous mouse UC-MSC delayed the onset of diabetes in RIP-B7.1 mice. Importantly, UC-MSC i. p. transplantation led to a strong peritoneal recruitment of myeloid-derived suppressor cells (MDSC) followed by multiple T-, B- and myeloid cells immunosuppressive responses in peritoneal fluid cells, spleen, pancreatic lymph nodes and the pancreas, which displayed significantly reduced insulitis and pancreatic infiltration of T and B Cells and pro-inflammatory macrophages. Altogether, these results suggest that UC-MSC i. p. transplantation can block or delay the development of hyperglycemia through suppression of inflammation and the immune attack.

10.
J Clin Med ; 12(22)2023 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-38002750

RESUMO

(1) Background: The current treatment for osteoarthritis is ineffective due to its focus on pain relief and lack of cartilage repair. Viscosupplementation such as hyaluronic acid improves symptoms but remains unnoticed for several months. Researchers are exploring cell-based therapies such as mesenchymal stem cells secretome and mesenchymal stem cells, which can repair cartilage damage. The objective of the research is to evaluate and compare the effectiveness of the secretome derived from umbilical cord mesenchymal stem cells (UC-MSCs) with hyaluronic acid (HA). (2) Methods: An open-label clinical trial involving 30 knee osteoarthritis patients divided into two groups received UC-MSC secretome and hyaluronic acid doses. The study assessed clinical outcomes using VAS and WOMAC and measured MMP-3 and TGF-ß1 levels before and after treatment. (3) Results: A study of 30 subjects found that the UC-MSC secretome group showed a decrease in pain in the OA knee compared to the HA group. The therapy was most effective after the third injection, and the group showed a decrease in the MMP-3 ratio and an increase in TGF-ß1 compared to the hyaluronic acid group. (4) Conclusions: UC-MSC secretome intra-articular injections showed superior clinical improvement, biomarker changes, and no side effects compared to hyaluronic acid over a 5-week interval.

11.
Bioengineering (Basel) ; 10(8)2023 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-37627815

RESUMO

Mesenchymal stromal cell (MSC)-derived products, such as trophic factors (MTFs), have anti-inflammatory properties that make them attractive for cell-free treatment. Three-dimensional (3D) culture can enhance these properties, and large-scale expansion using a bioreactor can reduce manufacturing costs. Three lots of MTFs were obtained from umbilical cord MSCs produced by either monolayer culture (Monol MTF) or using a 3D microcarrier in a spinner flask dynamic system (Bioreactor MTF). The resulting MTFs were tested and compared using anti-inflammatory potency assays in two different systems: (1) a phytohemagglutinin-activated peripheral blood mononuclear cell (PBMNC) system and (2) a lipopolysaccharide (LPS)-activated macrophage system. Cytokine expression by macrophages was measured via RT-PCR. The production costs of hypothetical units of anti-inflammatory effects were calculated using the percentage of TNF-α inhibition by MTF exposure. Bioreactor MTFs had a higher inhibitory effect on TNF (p < 0.01) than monolayer MTFs (p < 0.05). The anti-inflammatory effect of Bioreactor MTFs on IL-1ß, TNF-α, IL-8, IL-6, and MIP-1 was significantly higher than that of monolayer MTFs. The production cost of 1% inhibition of TNF-α was 11-40% higher using monolayer culture compared to bioreactor-derived MTFs. A 3D dynamic culture was, therefore, able to produce high-quality MTFs, with robust anti-inflammatory properties, more efficiently than monolayer static systems.

12.
Semin Perinatol ; 47(3): 151731, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36990922

RESUMO

Over the last 20 years, stem cells of varying origin and their associated secretome have been investigated as a therapeutic option for a myriad of neonatal models of disease, with very promising results. Despite the devastating nature of some of these disorders, translation of the preclinical evidence to the bedside has been slow. In this review, we explore the existing clinical evidence for stem cell therapies in neonates, highlight the barriers faced by researchers and suggest potential solutions to move the field forward.


Assuntos
Displasia Broncopulmonar , Doenças do Recém-Nascido , Recém-Nascido , Humanos , Transplante de Células-Tronco/métodos , Displasia Broncopulmonar/terapia , Doenças do Recém-Nascido/terapia
13.
Med Arch ; 76(1): 4-11, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35422561

RESUMO

Background: A Erectile dysfunction (ED) is one of the well-known comorbidities in males with diabetes mellitus (DM), whose pathogenesis might be induced by dysregulation of corpus cavernosum smooth muscle cells. UC-MSCs are multipotent cells that attract considerable interest due to immunoregulatory properties and might be a potential strategy to regulate and recover the functional cells and tissues, including tissue improvement in DMED. Objective: This study aims to determine the efficacy of UC-MSCs in improving the erectile function of DMED rats through analyzing the expression of TGF-ß, α-SMA, and collagen. Methods: Total number of 30 male Sprague-Dawley rats (6 to 8 weeks old) were randomly divided into four groups (negative control group, positive control group, T1 group, and T2 group). After 16 h fast, 24 rats were randomly selected and intraperitoneally injected with streptozotocin to induce DM. At 8 weeks after STZ injection, rats with DMED were identified by unresponsive erectile stimulation within 30 min. PC group received 500 µL; T1 rats treated with 500 µL PBS containing 1x106 UC-MSCs; T2 rats treated with 500 µL PBS containing 3x106 UC-MSCs. After MSCs treatment, the rats were sacrificed and the corpus cavernosum tissues were prepared for histological observations. Results: This study resulted in the administration of UC-MSCs could downregulate the expression of TGF-ß, α-SMA, and collagen leading to the improvement of DMED. Conclusion: UC-MSCs improve the expression of TGF-ß, α-SMA, and collagen on erectile dysfunction in streptozotocin-induced diabetic rats.


Assuntos
Diabetes Mellitus Experimental , Disfunção Erétil , Células-Tronco Mesenquimais , Animais , Masculino , Ratos , Colágeno , Diabetes Mellitus Experimental/complicações , Disfunção Erétil/etiologia , Disfunção Erétil/terapia , Células-Tronco Mesenquimais/metabolismo , Ratos Sprague-Dawley , Estreptozocina , Fator de Crescimento Transformador beta , Cordão Umbilical/citologia
14.
Regen Ther ; 21: 73-80, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35785041

RESUMO

Current trends indicate a growing interest among healthcare specialists and the public in the use of regenerative medicine-based approaches for skin regeneration. The approaches are categorised in either cell-based or cell-free therapies and are reportedly safe and effective. Cell-based therapies include mesenchymal stem cells (MSCs), tissue induced pluripotent stem cells (iPSCs), fibroblast-based products, and blood-derived therapies, such as those employing platelet-rich plasma (PRP) products. Cell-free therapies primarily involve the use of MSC-derived extracellular vesicles/exosomes. MSCs are isolated from various tissues, such as fat, bone marrow, umbilical cord, menstrual blood, and foetal skin, and expanded ex vivo before transplantation. In cell-free therapies, MSC exosomes, MSC-derived cultured media, and MSC-derived extracellular vesicles are collected from MSC-conditioned media or supernatant. In this review, a literature search of the Cochrane Library, MEDLINE (PubMed), EMBASE, and Scopus was conducted using several combinations of terms, such as 'stem', 'cell', 'aging', 'wrinkles', 'nasolabial folds', 'therapy', 'mesenchymal stem cells', and 'skin', to identify relevant articles providing a comprehensive update on the different regenerative medicine-based therapies and their application to skin regeneration. In addition, the regulatory perspectives on the clinical application of some of these therapies in Japan are highlighted.

15.
J Extracell Vesicles ; 10(8): e12094, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34136108

RESUMO

Extracellular vesicles (EVs) derived from the secretome of human mesenchymal stromal cells (MSC) contain numerous factors that are known to exert anti-inflammatory effects. MSC-EVs may serve as promising cell-based therapeutics for the inner ear to attenuate inflammation-based side effects from cochlear implantation which represents an unmet clinical need. In an individual treatment performed on a 'named patient basis', we intraoperatively applied allogeneic umbilical cord-derived MSC-EVs (UC-MSC-EVs) produced according to good manufacturing practice. A 55-year-old patient suffering from Menière's disease was treated with intracochlear delivery of EVs prior to the insertion of a cochlear implant. This first-in-human use of UC-MSC-EVs demonstrates the feasibility of this novel adjuvant therapeutic approach. The safety and efficacy of intracochlear EV-application to attenuate side effects of cochlea implants have to be determined in controlled clinical trials.


Assuntos
Implante Coclear/métodos , Vesículas Extracelulares/transplante , Transplante de Células-Tronco Mesenquimais/métodos , Diferenciação Celular , Implantes Cocleares/efeitos adversos , Citocinas/metabolismo , Orelha Interna/citologia , Vesículas Extracelulares/metabolismo , Humanos , Masculino , Células-Tronco Mesenquimais/fisiologia , Pessoa de Meia-Idade , Projetos Piloto , Cordão Umbilical/metabolismo
16.
AMB Express ; 10(1): 132, 2020 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-32725504

RESUMO

Mesenchymal stem cells (MSC) are a popular candidate in cellular therapy for many diseases. MSCs are well known by their feature of self-renewal and their differentiation potential. NESTIN is a cytoskeletal protein expressed in MSC that functions directly in cell proliferation and differentiation. Here, we demonstrated that adding UltraGRO, a medium supplement, could maintain and partially recover the expression of NESTIN in human umbilical cord derived MSCs (UC-MSCs). Furthermore, the UC-MSCs cultured with UltraGRO showed a better immunomodulation ability in a colitis mouse model compared with those cultured in other types of media. This indicates that the use of novel culture medium benefits the maintenance of NESTIN expression and NESTIN may be one of the vital factors that regulates the performance of MSCs.

17.
Front Immunol ; 11: 575488, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33117373

RESUMO

Mesenchymal stromal cells (MSC) have been used in over 800 clinical trials with encouraging results in the field of transplant medicine and chronic inflammatory diseases. Today, Umbilical Cord (UC)-derived MSC are the second leading source used for clinical purposes, mainly due to its easy access and superior immune modulatory effects. Although the underlying molecular mechanisms of immune suppressive activities have not been fully understood, research over the last decade strongly suggests that MSC-mediated benefits are closely related to activation of secretome networks. Nevertheless, recent findings also point to cytokine-independent mechanisms as key players of MSC-mediated immune modulation. Here, we set up a robust in vitro immune assay using phytohemagglutinin- or anti-CD3/CD28-treated human peripheral blood mononuclear cells in cell-to-cell interaction or in cell-contact independent format with UC-MSC and conducted integrated transcriptome and secretome analyses to dissect molecular pathways driving UC-MSC-mediated immune modulation. Under inflammatory stimuli, multiparametric analyses of the secretome led us to identify cytokine/chemokine expression patterns associated with the induction of MSC-reprogrammed macrophages and T cell subsets ultimately leading to immune suppression. UC-MSC transcriptome analysis under inflammatory challenge allowed the identification of 47 differentially expressed genes, including chemokines, anti- and pro-inflammatory cytokines and adhesion molecules found also in UC-MSC-immunosupressive secretomes, including the novel candidate soluble IL-2R. This study enabled us to track functionally activated UC-MSC during immune suppression and opened an opportunity to explore new pathways involved in immunity control by UC-MSC. We propose that identified immunomodulatory molecules and pathways could potentially be translated into clinical settings in order to improve UC-MSC-therapy quality and efficacy.


Assuntos
Inflamação/metabolismo , Células-Tronco Mesenquimais/metabolismo , Comunicação Parácrina , Linfócitos T/metabolismo , Transcriptoma , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Citocinas/genética , Citocinas/metabolismo , Sangue Fetal/citologia , Redes Reguladoras de Genes , Humanos , Inflamação/genética , Inflamação/imunologia , Ativação Linfocitária , Células-Tronco Mesenquimais/imunologia , Fenótipo , Via Secretória , Transdução de Sinais , Linfócitos T/imunologia
18.
Clin Transl Med ; 10(8): e262, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33377658

RESUMO

The lack of approved anti-inflammatory and neuroprotective therapies in otology has been acknowledged in the last decades and recent approaches are heralding a new era in the field. Extracellular vesicles (EVs) derived from human multipotent (mesenchymal) stromal cells (MSC) can be enriched in vesicular secretome fractions, which have been shown to exert effects (eg, neuroprotection and immunomodulation) of their parental cells. Hence, MSC-derived EVs may serve as novel drug candidates for several inner ear diseases. Here, we provide first evidence of a strong neuroprotective potential of human stromal cell-derived EVs on inner ear physiology. In vitro, MSC-EV preparations exerted immunomodulatory activity on T cells and microglial cells. Moreover, local application of MSC-EVs to the inner ear significantly attenuated hearing loss and protected auditory hair cells from noise-induced trauma in vivo. Thus, EVs derived from the vesicular secretome of human MSC may represent a next-generation biological drug that can exert protective therapeutic effects in a complex and nonregenerating organ like the inner ear.

19.
Stem Cells Cloning ; 13: 117-136, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33364786

RESUMO

INTRODUCTION: Osteoarthritis causes a progressive deterioration to the protective cartilage between the joints leading to chronic pain and disability. This review focuses on the intrinsic potential of MSCs to stabilize and repair the cartilage tissue of the knee joint in knee osteoarthritis (KOA) patients. METHODS: An online search through the PubMed database was conducted, limiting the search to the English language and human clinical trials within the past 5 years. Twenty-one clinical trials passed the inclusion criteria. Combined, those trials involved the participation of 589 patients where the progress of the treatments was monitored between a 4-month to 7-years period. The cartilage volume and defects were observed through an MRI to provide an objective assessment. While the pain and knee function were monitored using KOOS, VAS, and WOMAC scoring scales providing a subjective assessment. RESULTS: MRI scans obtained from clinical trials demonstrate a slowed progression of cartilage degeneration and early signs of cartilage regeneration in KOA patients at the 12-month follow-up period. No major adverse effects were observed post-intervention. The overall KOOS, WOMAC, and VAS scores in patients receiving MSC treatment were reduced, suggesting subjective improvements in knee function and pain reduction when compared to patients in the placebo group. CONCLUSION: The use of MSC therapy is a valid form of treatment for KOA as it targets the disease itself rather than the symptoms. We found MSC therapy in KOA patients to be safe, effective, and feasible in its execution.

20.
Mayo Clin Proc Innov Qual Outcomes ; 4(4): 443-459, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32793872

RESUMO

Glioblastoma is the most aggressive malignant primary brain tumor, with a dismal prognosis and a devastating overall survival. Despite aggressive surgical resection and adjuvant treatment, average survival remains approximately 14.6 months. The brain tumor microenvironment is heterogeneous, comprising multiple populations of tumor, stromal, and immune cells. Tumor cells evade the immune system by suppressing several immune functions to enable survival. Gliomas release immunosuppressive and tumor-supportive soluble factors into the microenvironment, leading to accelerated cancer proliferation, invasion, and immune escape. Mesenchymal stem cells (MSCs) isolated from bone marrow, adipose tissue, or umbilical cord are a promising tool for cell-based therapies. One crucial mechanism mediating the therapeutic outcomes often seen in MSC application is their tropism to sites of injury. Furthermore, MSCs interact with host immune cells to regulate the inflammatory response, and data points to the possibility of using MSCs to achieve immunomodulation in solid tumors. Interleukin 1ß, interleukin 6, tumor necrosis factor α, transforming growth factor ß, and stromal cell-derived factor 1 are notably up-regulated in glioblastoma and dually promote immune and MSC trafficking. Mesenchymal stem cells have widely been regarded as hypoimmunogenic, enabling this cell-based administration across major histocompatibility barriers. In this review, we will highlight (1) the bidirectional communication of glioma cells and tumor-associated immune cells, (2) the inflammatory mediators enabling leukocytes and transplantable MSC migration, and (3) review preclinical and human clinical trials using MSCs as delivery vehicles. Mesenchymal stem cells possess innate abilities to migrate great distances, cross the blood-brain barrier, and communicate with surrounding cells, all of which make them desirable "Trojan horses" for brain cancer therapy.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa