Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 399
Filtrar
1.
Cells ; 12(1)2022 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-36611914

RESUMO

Spinal Cord Injury (SCI) is a common neurological disorder with devastating psychical and psychosocial sequelae. The majority of patients after SCI suffer from permanent disability caused by motor dysfunction, impaired sensation, neuropathic pain, spasticity as well as urinary complications, and a small number of patients experience a complete recovery. Current standard treatment modalities of the SCI aim to prevent secondary injury and provide limited recovery of lost neurological functions. Stem Cell Therapy (SCT) represents an emerging treatment approach using the differentiation, paracrine, and self-renewal capabilities of stem cells to regenerate the injured spinal cord. To date, multipotent stem cells including mesenchymal stem cells (MSCs), neural stem cells (NSCs), and hematopoietic stem cells (HSCs) represent the most investigated types of stem cells for the treatment of SCI in preclinical and clinical studies. The microenvironment of SCI has a significant impact on the survival, proliferation, and differentiation of transplanted stem cells. Therefore, a deep understanding of the pathophysiology of SCI and molecular mechanisms through which stem cells act may help improve the treatment efficacy of SCT and find new therapeutic approaches such as stem-cell-derived exosomes, gene-modified stem cells, scaffolds, and nanomaterials. In this literature review, the pathogenesis of SCI and molecular mechanisms of action of multipotent stem cells including MSCs, NSCs, and HSCs are comprehensively described. Moreover, the clinical efficacy of multipotent stem cells in SCI treatment, an optimal protocol of stem cell administration, and recent therapeutic approaches based on or combined with SCT are also discussed.


Assuntos
Células-Tronco Mesenquimais , Células-Tronco Neurais , Traumatismos da Medula Espinal , Humanos , Traumatismos da Medula Espinal/patologia , Células-Tronco Multipotentes/transplante
2.
Cells ; 10(8)2021 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-34440855

RESUMO

Anal sphincter incontinence is a chronic disease, which dramatically impairs quality of life and induces high costs for the society. Surgery, considered as the best curative option, shows a disappointing success rate. Stem/progenitor cell therapy is pledging, for anal sphincter incontinence, a substitute to surgery with higher efficacy. However, the published literature is disparate. Our aim was to perform a review on the development of cell therapy for anal sphincter incontinence with critical analyses of its pitfalls. Animal models for anal sphincter incontinence were varied and tried to reproduce distinct clinical situations (acute injury or healed injury with or without surgical reconstruction) but were limited by anatomical considerations. Cell preparations used for treatment, originated, in order of frequency, from skeletal muscle, bone marrow or fat tissue. The characterization of these preparations was often incomplete and stemness not always addressed. Despite a lack of understanding of sphincter healing processes and the exact mechanism of action of cell preparations, this treatment was evaluated in 83 incontinent patients, reporting encouraging results. However, further development is necessary to establish the correct indications, to determine the most-suited cell type, to standardize the cell preparation method and to validate the route and number of cell delivery.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Incontinência Fecal/terapia , Células-Tronco Multipotentes/transplante , Tecido Adiposo/citologia , Animais , Células da Medula Óssea/citologia , Incontinência Fecal/patologia , Humanos , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Transplante de Células-Tronco , Células-Tronco/citologia , Células-Tronco/metabolismo
3.
Stem Cells Transl Med ; 10(1): 57-67, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32985793

RESUMO

Involvement of the cerebellum in the pathophysiology of hypoxic-ischemic encephalopathy (HIE) in preterm infants is increasingly recognized. We aimed to assess the neuroprotective potential of intravenously administered multipotent adult progenitor cells (MAPCs) in the preterm cerebellum. Instrumented preterm ovine fetuses were subjected to transient global hypoxia-ischemia (HI) by 25 minutes of umbilical cord occlusion at 0.7 of gestation. After reperfusion, two doses of MAPCs were administered intravenously. MAPCs are a plastic adherent bone-marrow-derived population of adult progenitor cells with neuroprotective potency in experimental and clinical studies. Global HI caused marked cortical injury in the cerebellum, histologically indicated by disruption of cortical strata, impeded Purkinje cell development, and decreased dendritic arborization. Furthermore, global HI induced histopathological microgliosis, hypomyelination, and disruption of white matter organization. MAPC treatment significantly prevented cortical injury and region-specifically attenuated white matter injury in the cerebellum following global HI. Diffusion tensor imaging (DTI) detected HI-induced injury and MAPC neuroprotection in the preterm cerebellum. This study has demonstrated in a preclinical large animal model that early systemic MAPC therapy improved structural injury of the preterm cerebellum following global HI. Microstructural improvement was detectable with DTI. These findings support the potential of MAPC therapy for the treatment of HIE and the added clinical value of DTI for the detection of cerebellar injury and the evaluation of cell-based therapy.


Assuntos
Células-Tronco Adultas/transplante , Asfixia , Cerebelo , Hipóxia-Isquemia Encefálica , Células-Tronco Multipotentes , Animais , Asfixia/terapia , Imagem de Tensor de Difusão , Modelos Animais de Doenças , Feto , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Células-Tronco Multipotentes/transplante , Ovinos
4.
Neurotherapeutics ; 18(1): 448-459, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33067748

RESUMO

The human amnion has been used for decades in wound healing, particularly burns. Amnion epithelial cells (AECs) have been the focus of extensive research based on their possible pluripotent differentiation ability. A novel, cultured cell population derived from AECs, termed human amnion-derived multipotent progenitor (AMP) cells, secrete numerous cytokines and growth factors that enhance tissue regeneration and reduce inflammation. This AMP cell secretome, termed ST266, is a unique biological solution that accumulates in eyes and optic nerves following intranasal delivery, resulting in selective suppression of optic neuritis in the experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis, but not myelitis at the administered dose. We tested the hypothesis that systemic AMP cell administration could suppress both optic neuritis and myelitis in EAE. Intravenous and intraperitoneal administration of AMP cells significantly reduced ascending paralysis and attenuated visual dysfunction in EAE mice. AMP cell treatment increased retinal ganglion cell (RGC) survival and decreased optic nerve inflammation, with variable improvement in optic nerve demyelination and spinal cord inflammation and demyelination. Results show systemic AMP cell administration inhibits RGC loss and visual dysfunction similar to previously demonstrated effects of intranasally delivered ST266. Importantly, AMP cells also promote neuroprotective effects in EAE spinal cords, marked by reduced paralysis. Protective effects of systemically administered AMP cells suggest they may serve as a potential novel treatment for multiple sclerosis.


Assuntos
Células-Tronco Multipotentes/transplante , Mielite/terapia , Neurite Óptica/terapia , Âmnio/citologia , Animais , Doenças Desmielinizantes/terapia , Encefalomielite Autoimune Experimental/terapia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Células Ganglionares da Retina/metabolismo , Medula Espinal/patologia
5.
Transplant Proc ; 52(7): 2204-2207, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32340748

RESUMO

BACKGROUND: Nonhealing wounds can be a major clinical problem. Impaired wound healing is often related to massive tissue injury, concomitant wound healing deficiencies (chronic wounds), burn injury, or congenital conditions. We propose a novel biological dressing as an alternative surgical approach. The dressing is a form of an allogenic human skin graft equivalent with further use of allogeneic stem cells classified as an advanced therapy medicinal product. This new allogenic acellular human skin graft has been specifically developed to address the clinical indications for dressing wound lesions and promoting tissue repair in specific rare genetic diseases. METHODS: This case report illustrates the use of an acellular human skin allograft seeded with multipotent stem cells in the treatment of tissue injuries (burns), congenital conditions, and chronic wounds. Donor-tissue processing yields an acellular dermal matrix with integral collagen bundling and organization, as well as an intact basement membrane complex. RESULTS: Preclinical observations show prolonged viability of acellular human skin grafts with multipotent stem cells. This was confirmed with histological and electron-microscopic evaluation of biopsies, which demonstrated host-cell infiltration and neovascularization of the biological dressing. Moreover, the dressings were characterized by low immunogenicity, as confirmed by histology exam and T-cell proliferation assays in vitro. CONCLUSION: Our data confirmed the safety and efficacy of the evaluated acellular human skin grafts, which may be used in patients with rare diseases, such as epidermolysis bullosa, burn injuries, and chronic wounds.


Assuntos
Derme Acelular , Células-Tronco Multipotentes/transplante , Transplante de Pele/métodos , Engenharia Tecidual/métodos , Cicatrização , Curativos Biológicos , Humanos , Técnicas In Vitro , Transplante Homólogo
6.
J Orthop Res ; 38(1): 128-138, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31329308

RESUMO

Intra-synovial tendon injuries are a common orthopedic problem with limited treatment options. The synovium is a specialized connective tissue forming the inner encapsulating lining of diarthrodial joints and intra-synovial tendons. It contains multipotent mesenchymal stromal cells that render it a viable source of progenitors for tendon repair. This study evaluated the effects of autologous implantation of cells derived from normal synovium (synovial membrane cells [SMCs]) in augmenting repair in an ovine model of intra-synovial tendon injury. For this purpose, synovial biopsies were taken from the right digital flexor tendon sheath following creation of a defect to the lateral deep digital flexor tendon. Mononuclear cells were isolated by partial enzymatic digestion and assessed for MSC characteristics. Cell tracking and tendon repair were assessed by implanting 5 × 106 cells into the digital flexor tendon sheath under ultrasound guidance with the effects evaluated using magnetic resonance imaging and histopathology. Synovial biopsies yielded an average 4.0 × 105 ± 2.7 × 105 SMCs that exhibited a fibroblastic morphology, variable osteogenic, and adipogenic responses but were ubiquitously strongly chondrogenic. SMCs displayed high expression of CD29 with CD271NEGATIVE and MHC-IILOW cell-surface marker profiles, and variable expression of CD73, CD90, CD105, CD166, and MHC-I. Implanted SMCs demonstrated engraftment within the synovium, though a lack of repair of the tendon lesion over 24 weeks was observed. We conclude healthy synovium is a viable source of multipotent cells, but that the heterogeneity of synovium underlies the variability between different SMC populations, which while capable of engraftment and persistence within the synovium exhibit limited capacity of influencing tendon repair. © 2019 The Authors. Journal of Orthopaedic Research® published by Wiley Periodicals, Inc. on behalf of Orthopaedic Research Society J Orthop Res 38:128-138, 2020.


Assuntos
Células-Tronco Multipotentes/transplante , Membrana Sinovial/citologia , Traumatismos dos Tendões/cirurgia , Tendões/fisiopatologia , Animais , Diferenciação Celular , Linhagem da Célula , Células Cultivadas , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Imageamento por Ressonância Magnética , Células-Tronco Multipotentes/citologia , Ovinos , Traumatismos dos Tendões/fisiopatologia
7.
Stem Cell Res Ther ; 10(1): 234, 2019 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-31376835

RESUMO

BACKGROUND: Neurovascular unit restoration is crucial for nerve regeneration, especially in critical gaps of injured peripheral nerve. Multipotent vascular stem cells (MVSCs) harvested from an adult blood vessel are involved in vascular remodeling; however, the therapeutic benefit for nerve regeneration is not clear. METHODS: MVSCs were isolated from rats expressing green fluorescence protein (GFP), expanded, mixed with Matrigel matrix, and loaded into the nerve conduits. A nerve autograft or a nerve conduit (with acellular matrigel or MVSCs in matrigel) was used to bridge a transected sciatic nerve (10-mm critical gap) in rats. The functional motor recovery and cell fate in the regenerated nerve were investigated to understand the therapeutic benefit. RESULTS: MVSCs expressed markers such as Sox 17 and Sox10 and could differentiate into neural cells in vitro. One month following MVSC transplantation, the compound muscle action potential (CMAP) significantly increased as compared to the acellular group. MVSCs facilitated the recruitment of Schwann cell to regenerated axons. The transplanted cells, traced by GFP, differentiated into perineurial cells around the bundles of regenerated myelinated axons. In addition, MVSCs enhanced tight junction formation as a part of the blood-nerve barrier (BNB). Furthermore, MVSCs differentiated into perivascular cells and enhanced microvessel formation within regenerated neurovascular bundles. CONCLUSIONS: In rats with peripheral nerve injuries, the transplantation of MVSCs into the nerve conduits improved the recovery of neuromuscular function; MVSCs differentiated into perineural cells and perivascular cells and enhanced the formation of tight junctions in perineural BNB. This study demonstrates the in vivo therapeutic benefit of adult MVSCs for peripheral nerve regeneration and provides insight into the role of MVSCs in BNB regeneration.


Assuntos
Regeneração Nervosa/fisiologia , Potenciais de Ação , Animais , Aorta/citologia , Axônios/fisiologia , Diferenciação Celular , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Células-Tronco Multipotentes/transplante , Músculos/fisiologia , Traumatismos dos Nervos Periféricos/terapia , Ratos , Ratos Sprague-Dawley , Ratos Transgênicos , Recuperação de Função Fisiológica , Fatores de Transcrição SOXF/genética , Fatores de Transcrição SOXF/metabolismo , Células de Schwann/citologia , Células de Schwann/metabolismo , Junções Íntimas/fisiologia
8.
Nanomedicine ; 21: 102045, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31255791

RESUMO

Non-viral neurotrophic factor (NF) gene therapy is a new paradigm in glaucoma treatment with the potential for neuroprotection and regeneration of damaged retinal ganglion cells (RGCs). To improve nanoparticle gene delivery systems and generate a suitable RGC cell model to facilitate in vitro investigations, we have developed mouse multipotent retinal stem cell (MRSC)-derived RGCs (XFC-3 cells) that express key RGC characteristics as demonstrated through biomarker expression profiling and stimuli-inducible neurite extension evaluation. Dicationic gemini surfactant-, single-walled carbon nanotube-, and K2-lipopolyamine polymer-based gene delivery systems were formulated and evaluated in three-dimensional (3D) A7/XFC-3 and XFC-3/XFC-3 co-cultures to validate the model for transfection efficiency (TE) and brain-derived neurotrophic factor (BDNF) bioactivity measurements, which helped identify the K2-NPs as having high TE (63.1% ±â€¯1.4%) and high cell viability (94.4% ±â€¯0.4%). Overall, XFC-3 cells are suitable for the construction of 3D in vivo-like tissue models and enable the screening of RGC-aimed gene delivery systems for neuroprotective treatment of glaucoma.


Assuntos
Técnicas de Transferência de Genes , Glaucoma/terapia , Células-Tronco Multipotentes/citologia , Nanopartículas/química , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Técnicas de Cultura de Células , Sobrevivência Celular/genética , Técnicas de Cocultura , Terapia Genética , Glaucoma/genética , Humanos , Células-Tronco Multipotentes/transplante , Nanopartículas/administração & dosagem , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/uso terapêutico , Neuritos/efeitos dos fármacos , Neuritos/metabolismo , Retina/patologia , Células Ganglionares da Retina/citologia , Células Ganglionares da Retina/transplante , Transfecção
9.
J Cell Physiol ; 234(11): 19451-19463, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31025370

RESUMO

Cardiac progenitor cells (CPCs) have the potential to differentiate into several cell lineages with the ability to restore in cardiac tissue. Multipotency and self-renewal activity are the crucial characteristics of CPCs. Also, CPCs have promising therapeutic roles in cardiac diseases such as valvular disease, thrombosis, atherosclerosis, congestive heart failure, and cardiac remodeling. Toll-like receptors (TLRs), as the main part of the innate immunity, have a key role in the development and differentiation of immune cells. Some reports are found regarding the effect of TLRs in the maturation of stem cells. This article tried to find the potential role of TLRs in the dynamics of CPCs. By showing possible crosstalk between the TLR signaling pathways and CPCs dynamics, we could achieve a better conception related to TLRs in the regeneration of cardiac tissue.


Assuntos
Aterosclerose/genética , Insuficiência Cardíaca/genética , Células-Tronco/citologia , Receptores Toll-Like/genética , Aterosclerose/patologia , Aterosclerose/terapia , Diferenciação Celular/genética , Linhagem da Célula/genética , Coração/crescimento & desenvolvimento , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/terapia , Humanos , Imunidade Inata/genética , Células-Tronco Multipotentes/transplante , Transdução de Sinais/genética , Células-Tronco/metabolismo
10.
BMJ Open Respir Res ; 6(1): e000308, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30713713

RESUMO

Introduction: Bone marrow-derived multipotent adult progenitor cells (MAPCs) are adult allogeneic adherent stem cells currently investigated clinically for use in acute respiratory distress syndrome (ARDS). To date, there is no agreement on which is the best method for stem cells delivery in ARDS. Here, we compared the efficacy of two different methods of administration and biodistribution of MAPC for the treatment of ARDS in a sheep model. Methods: MAPC were labelled with [18F] fluoro-29-deoxy-D-glucose and delivered by endobronchial (EB) or intravenous route 1 hour after lipopolysaccharide infusion in sheep mechanically ventilated. PET/CT images were acquired to determine the biodistribution and retention of the cells at 1 and 5 hours of administration. Results: The distribution and retention of the MAPC was dependent on the method of cell administration. By EB route, PET images showed that MAPC remained at the site of administration and no changes were observed after 5 hours, whereas with intravenous route, the cells had broad biodistribution to different organs, being the lung the main organ of retention at 1 and 5 hours. MAPC demonstrated an equal effect on arterial oxygenation recovery by either route of administration. Conclusion: The EB or intravenous routes of administration of MAPC are both effective for the treatment of ARDS in an acute sheep model, and the effect of MAPC therapy is not dependent of parenchymal integration or systemic biodistribution.


Assuntos
Células-Tronco Adultas/transplante , Células-Tronco Multipotentes/transplante , Síndrome do Desconforto Respiratório/terapia , Animais , Brônquios , Células Cultivadas , Modelos Animais de Doenças , Feminino , Humanos , Infusões Intravenosas , Lipopolissacarídeos/imunologia , Masculino , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Cultura Primária de Células , Síndrome do Desconforto Respiratório/diagnóstico por imagem , Síndrome do Desconforto Respiratório/imunologia , Ovinos , Resultado do Tratamento
11.
J Neurotrauma ; 36(9): 1416-1427, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30251917

RESUMO

Following spinal cord injury (SCI), inflammation amplifies damage beyond the initial insult, providing an opportunity for targeted treatments. An ideal protective therapy would reduce both edema within the lesion area and the activation/infiltration of detrimental immune cells. Previous investigations demonstrated the efficacy of intravenous injection of multipotent adult progenitor cells (MAPC®) to modulate immune response following SCI, leading to significant improvements in tissue sparing, locomotor and urological functions. Separate studies have demonstrated that tissue inhibitor of matrix metalloproteinase-3 (TIMP3) reduces blood-brain barrier permeability following traumatic brain injury in a mouse model, leading to improved functional recovery. This study examined whether TIMP3, delivered alone or in concert with MAPC cells, improves functional recovery from a contusion SCI in a rat model. The results suggest that intravenous delivery of MAPC cell therapy 1 day following acute SCI significantly improves tissue sparing and impacts functional recovery. TIMP3 treatment provided no significant benefit, and further, when co-administered with MAPC cells, it abrogated the therapeutic effects of MAPC cell therapy. Importantly, this study demonstrated for the first time that acute treatment of SCI with MAPC cells can significantly reduce the incidence of urinary tract infection (UTI) and the use of antibiotics for UTI treatment.


Assuntos
Células-Tronco Multipotentes/transplante , Recuperação de Função Fisiológica , Traumatismos da Medula Espinal , Inibidor Tecidual de Metaloproteinase-3/farmacologia , Infecções Urinárias , Células-Tronco Adultas/transplante , Animais , Feminino , Humanos , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/efeitos dos fármacos , Recuperação de Função Fisiológica/fisiologia , Traumatismos da Medula Espinal/complicações , Traumatismos da Medula Espinal/patologia , Transplante de Células-Tronco/métodos , Infecções Urinárias/epidemiologia , Infecções Urinárias/etiologia
12.
J Cell Physiol ; 234(3): 2426-2435, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30238990

RESUMO

Cell therapy is one of the important therapeutic approaches in the treatment of many diseases such as cancer, degenerative diseases, and cardiovascular diseases. Among various cell types, which could be used as cell therapies, stem cell therapy has emerged as powerful tools in the treatment of several diseases. Multipotent stem cells are one of the main classes of stem cells that could originate from different parts of the body such as bone marrow, adipose, placenta, and tooth. Among several types of multipotent stem cells, tooth-derived stem cells (TDSCs) are associated with special properties such as accessible, easy isolation, and low invasive, which have introduced them as a good source for using in the treatment of several diseases such as neural injuries, liver fibrosis, and Cohrn's disease. Here, we provided an overview of TDSCs particular stem cells from human exfoliated deciduous teeth and clinical application of them. Moreover, we highlighted molecular mechanisms involved in the regulation of dental stem cells fate.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Células-Tronco Multipotentes/transplante , Dente Decíduo/citologia , Doenças Cardiovasculares/terapia , Diferenciação Celular/genética , Terapia Baseada em Transplante de Células e Tecidos/tendências , Polpa Dentária/citologia , Polpa Dentária/transplante , Humanos , Células-Tronco Multipotentes/citologia , Neoplasias/terapia , Degeneração Neural/terapia , Dente Decíduo/transplante
13.
Macromol Biosci ; 19(3): e1800233, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30253047

RESUMO

Conventional wound therapy utilizes wound coverage to prevent infection, trauma, and fluid and thermal loss. However, this approach is often inadequate for large and/or chronic wounds, which require active intervention via therapeutic cells to promote healing. To address this need, a patch which delivers multipotent adult progenitor cells (MAPCs) is developed. Medical-grade polyurethane (PU) films are modified using plasma immersion ion implantation (PIII), which creates a radical-rich layer capable of rapidly and covalently attaching biomolecules. It is demonstrated that a short treatment duration of 400 s maximizes surface activation and wettability, minimizes reduction in gas permeability, and preserves the hydrolytic resistance of the PU film. The reactivity of PIII-treated PU is utilized to immobilize the extracellular matrix protein tropoelastin in a functional conformation that stably withstands medical-grade ethylene oxide sterilization. The PIII-treated tropoelastin-functionalized patch significantly promotes MAPC adhesion and proliferation over standard PU, while fully maintaining cell phenotype. Topical application of the MAPC-seeded patch transfers cells to a human skin model, while undelivered MAPCs repopulate the patch surface for subsequent cell transfer. The potential of this new wound patch as a reservoir for the sustained delivery of therapeutic MAPCs and cell-secreted factors for large and/or non-healing wounds is indicated in the findings.


Assuntos
Células-Tronco Adultas/transplante , Células Imobilizadas/transplante , Materiais Revestidos Biocompatíveis/química , Membranas Artificiais , Células-Tronco Multipotentes/transplante , Pele/metabolismo , Transplante de Células-Tronco , Tropoelastina/química , Adulto , Células-Tronco Adultas/metabolismo , Células Imobilizadas/metabolismo , Humanos , Células-Tronco Multipotentes/metabolismo , Poliuretanos/química
14.
Clin Interv Aging ; 14: 2261-2271, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31908436

RESUMO

BACKGROUND: Manipulating the immune inflammatory response after cerebral ischemia has been a novel therapeutic strategy for ischemic stroke. This study attempted to investigate the effects of the transplantation of lymphocytes co-cultured with human cord blood-derived multipotent stem cells (HCB-SCs) on the inflammatory response in transient middle cerebral occlusion (tMCAO) rats. METHODS: The tMCAO rats were subjected to the transplantation of lymphocytes co-cultured with HCB-SCs through tail vein injection. Infarct size and neurological deficits were measured at 48 hrs after stroke. Neurological deficits were assessed using Bederson's scoring system and tape removal test. Blood T cell flow cytometry was performed to measure the differentiation of regulatory T cells (Tregs). Western blot was used to detect the protein levels of inflammation-related molecules, apoptosis-related molecule, and signaling molecules in ischemic brain. TUNEL staining was performed to analyze cell apoptosis in ischemic cerebral cortex. RESULTS: The transplantation of lymphocytes co-cultured with HCB-SCs significantly improved the neurological defects, reduced ischemic brain damage, and increased the proportion of peripheral CD4+CD25+Foxp3+ Tregs. Meanwhile, the transplantation of co-cultured cells decreased the expression of NLRP3 inflammasome and associated factors, such as caspase-1 and IL-1ß, and inhibited the activation of NF-κB, ERK and caspase-3 in ischemic brain. The co-cultured cells significantly decreased the number of tMCAO-induced cell apoptosis. CONCLUSION: Lymphocytes co-cultured with HCB-SCs exhibit a neuroprotective effect after ischemic stroke by promoting Tregs differentiation and suppressing NLRP3 inflammasome activation and neuron apoptosis, and might be a promising therapeutic strategy for ischemic stroke.


Assuntos
Isquemia Encefálica/terapia , Inflamassomos/metabolismo , Transfusão de Linfócitos/métodos , Linfócitos/citologia , Células-Tronco Multipotentes/transplante , Transplante de Células-Tronco/métodos , Animais , Apoptose , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Células Cultivadas , Técnicas de Cocultura , Modelos Animais de Doenças , Sangue Fetal , Humanos , Masculino , Ratos , Ratos Wistar , Transdução de Sinais
15.
Theranostics ; 8(20): 5610-5624, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30555567

RESUMO

Rationale: Mesenchymal stem cell (MSC) therapy may be a novel approach to improve interstitial cystitis/bladder pain syndrome (IC/BPS), an intractable disease characterized by severe pelvic pain and urinary frequency. Unfortunately, the properties of transplanted stem cells have not been directly analyzed in vivo, which hampers elucidation of the therapeutic mechanisms of these cells and optimization of transplantation protocols. Here, we monitored the behaviors of multipotent stem cells (M-MSCs) derived from human embryonic stem cells (hESCs) in real time using a novel combination of in vivo confocal endoscopic and microscopic imaging and demonstrated their improved therapeutic potency in a chronic IC/BPS animal model. Methods: Ten-week-old female Sprague-Dawley rats were instilled with 10 mg of protamine sulfate followed by 750 µg of lipopolysaccharide weekly for 5 weeks. The sham group was instilled with phosphate-buffered saline (PBS). Thereafter, the indicated dose (0.1, 0.25, 0.5, and 1×106 cells) of M-MSCs or PBS was injected once into the outer layer of the bladder. The distribution, perivascular integration, and therapeutic effects of M-MSCs were monitored by in vivo endoscopic and confocal microscopic imaging, awake cystometry, and histological and gene expression analyses. Results: A novel combination of longitudinal intravital confocal fluorescence imaging and microcystoscopy in living animals, together with immunofluorescence analysis of bladder tissues, demonstrated that transplanted M-MSCs engrafted following differentiation into multiple cell types and gradually integrated into a perivascular-like structure until 30 days after transplantation. The beneficial effects of transplanted M-MSCs on bladder voiding function and the pathological characteristics of the bladder were efficient and long-lasting due to the stable engraftment of these cells. Conclusion: This longitudinal bioimaging study of transplanted hESC-derived M-MSCs in living animals reveals their long-term functional integration, which underlies the improved therapeutic effects of these cells on IC/BPS.


Assuntos
Cistite Intersticial/diagnóstico por imagem , Cistite Intersticial/terapia , Microscopia Intravital/métodos , Células-Tronco Mesenquimais/citologia , Bexiga Urinária/diagnóstico por imagem , Animais , Modelos Animais de Doenças , Feminino , Transplante de Células-Tronco Mesenquimais , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/transplante , Ratos , Ratos Sprague-Dawley
16.
Tissue Cell ; 54: 94-104, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30309515

RESUMO

Neural crest stem cells that located in the postnatal hair follicle (HF-NCSC) are considered a promising tool for treatment of nervous system diseases and injuries. It is well known that HF-NCSC can be used in the spinal cord and sciatic nerve reparation but their ability to restore brain structures is poorly studied. In this article we are investigating the interaction between HF-NCSC and a nerve tissue (embryonic and adult). We have found out that HF-NCSC isolated from adult mice grow and differentiate in accordance with the mouse embryo developmental stage when co-cultured with the embryonic nerve tissue. The HF-NCSC migration is slower in the late embryonic tissue co-culture system compared to the early one. This phenomenon is related to the motor function of the cells but not to their proliferation level. We have demonstrated that the embryonic nerve tissue maintains HF-NCSC an undifferentiated status, while an adult brain tissue inhibits the cell proliferation and activates the differentiation processes. Besides, HF-NCSC pre-differentiated into the neuronal direction shows a higher survival and migration rate after the transplantation into the adult brain tissue compared to the undifferentiated HF-NCSC. Thus, we have investigated the postnatal HF-NCSC response to the nerve tissue microenvironment to analyze their possible application to the brain repair processes.


Assuntos
Diferenciação Celular/fisiologia , Folículo Piloso/citologia , Células-Tronco Neurais/citologia , Transplante de Células-Tronco/métodos , Animais , Encéfalo/citologia , Proliferação de Células/fisiologia , Técnicas de Cocultura , Embrião de Mamíferos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/transplante , Crista Neural/citologia , Células-Tronco Neurais/transplante , Técnicas de Cultura de Órgãos , Medula Espinal/citologia
17.
Clin Orthop Relat Res ; 476(10): 2091-2100, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30179944

RESUMO

BACKGROUND: Achilles tendon rupture is a common injury and the best treatment option remains uncertain between surgical and nonoperative methods. Biologic approaches using multipotent stem cells such as perivascular stem cells pose a possible treatment option, although there is currently a paucity of evidence regarding their clinical therapeutic use. QUESTIONS/PURPOSES: The purpose of this study was to determine whether injected perivascular stem cells (PSCs) would (1) improve histologic signs of tendon healing (such as percent area of collagen); and (2) improve biomechanical properties (peak load or stiffness) in a rat model of Achilles tendon transection. METHODS: Two subtypes of PSCs were derived from human adipose tissue: pericytes (CD146CD34CD45CD31) and adventitial cells (CD146CD34CD45CD31). Thirty-two athymic rats underwent right Achilles transection and were randomized to receive injection with saline (eight tendons), hydrogel (four tendons), pericytes in hydrogel (four tendons), or adventitial cells in hydrogel (eight tendons) 3 days postoperatively with the left serving as an uninjured control. Additionally, a subset of pericytes was labeled with CM-diI to track cell viability and localization. At 3 weeks, the rats were euthanized, and investigators blinded to treatment group allocation evaluated tendon healing by peak load and stiffness using biomechanical testing and percent area of collagen using histologic analysis with picrosirius red staining. RESULTS: Histologic analysis showed a higher mean percent area collagen for pericytes (30%) and adventitial cells (28%) than hydrogel (21%) or saline (26%). However, a nonparametric statistical analysis yielded no statistical difference. Mechanical testing demonstrated that the pericyte group had a higher peak load than the saline group (41 ± 7 N versus 26 ± 9 N; mean difference 15 N; 95% confidence interval [CI], 4-27 N; p = 0.003) and a higher peak load than the hydrogel group (41 ± 7 N versus 25 ± 3 N; mean difference 16; 95% CI, 8-24 N; p = 0.001). The pericyte group demonstrated higher stiffness than the hydrogel group (36 ± 12 N/mm versus 17 ± 6 N/mm; mean difference 19 N/mm; 95% CI, 5-34 N/mm; p = 0.005). CONCLUSIONS: Our results suggest that injection of PSCs improves mechanical but not the histologic properties of early Achilles tendon healing. CLINICAL RELEVANCE: This is a preliminary study that provides more insight into the use of adipose-derived PSCs as a percutaneous therapy in the setting of Achilles tendon rupture. Further experiments to characterize the function of these cells may serve as a pathway to development of minimally invasive intervention aimed at improving nonoperative management while avoiding the complications associated with surgical treatment down the line.


Assuntos
Tendão do Calcâneo/cirurgia , Tecido Adiposo/citologia , Túnica Adventícia/citologia , Células-Tronco Multipotentes/transplante , Pericitos/transplante , Transplante de Células-Tronco , Traumatismos dos Tendões/cirurgia , Cicatrização , Tendão do Calcâneo/metabolismo , Tendão do Calcâneo/fisiopatologia , Animais , Biomarcadores/metabolismo , Fenômenos Biomecânicos , Células Cultivadas , Colágeno/metabolismo , Modelos Animais de Doenças , Humanos , Masculino , Células-Tronco Multipotentes/metabolismo , Pericitos/metabolismo , Fenótipo , Ratos Nus , Traumatismos dos Tendões/metabolismo , Traumatismos dos Tendões/fisiopatologia , Fatores de Tempo
18.
Transl Res ; 200: 65-80, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30016629

RESUMO

Acute-on-chronic liver failure (ACLF) is a serious life-threatening disease with high prevalence. Liver transplantation is the only efficient clinical treatment for ACLF. Because of the rapid progression and lack of liver donors, it is urgent to find an effective and safe therapeutic approach to ACLF. Recent studies showed that multipotent cell transplantation could improve the patients' liver function and enhance their preoperative condition. Cells such as mesenchymal stem cells, bone marrow mononuclear cells and autologous peripheral blood stem cells, which addressed in this study have all been used in multipotent cell transplantation for liver diseases. However, its clinical efficiency is still debatable. This systematic review and meta-analysis explored the clinical efficiency of multipotent cell transplantation as a therapeutic approach for patients with ACLF. A detailed search of the Cochrane Library, MEDLINE, and Embase databases was conducted from inception to November 2017. The outcome measures were serum albumin, prothrombin time, alanine aminotransferase, total bilirubin, platelets, hemoglobin, white blood cells, and survival time. The quality of evidence was assessed using GRADEpro and Jaded scores. A literature search resulted in 537 citations. Of these, 9 articles met the inclusion criteria. It was found that multipotent cell transplantation was able to alleviate liver damage and improve liver function. Multipotent cell transplantation can also enhance the short-term and medium-term survival rates of ACLF. All 9 research articles included in this analysis reported no statistically significant adverse events, side effects, or complications. In conclusions, this study suggested that multipotent cell transplantation could be recommended as a potential therapeutic supplementary tool in clinical practice. However, clinical trials in large-volume centers still needed.


Assuntos
Insuficiência Hepática Crônica Agudizada/cirurgia , Células-Tronco Multipotentes/transplante , Insuficiência Hepática Crônica Agudizada/sangue , Insuficiência Hepática Crônica Agudizada/mortalidade , Adulto , Alanina Transaminase/sangue , Bilirrubina/sangue , Feminino , Hemoglobinas/análise , Humanos , Contagem de Leucócitos , Transplante de Fígado , MEDLINE , Masculino , Pessoa de Meia-Idade , Contagem de Plaquetas , Tempo de Protrombina , Albumina Sérica/análise , Taxa de Sobrevida , Resultado do Tratamento
19.
J Vasc Surg ; 68(6S): 115S-125S, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29753580

RESUMO

OBJECTIVE: The purpose of our study was to investigate the effect of adipose-derived stem cells (ASCs), endothelial-differentiated ASCs (EC/ASCs), and various conditioned media (CM) on wound healing in a diabetic swine model. We hypothesized that ASC-based therapies would accelerate wound healing. METHODS: Diabetes was induced in four Yorkshire swine through intravenous injection of streptozotocin. ASCs were harvested from flank fat and cultured in either M199 or EGM-2 medium. A duplicate series of seven full-thickness dorsal wounds were surgically created on each swine. The wounds in the cellular treatment group underwent injection of low-dose or high-dose ASCs or EC/ASCs on day 0, with a repeat injection of one half of the initial dose on day 15. Wounds assigned to the topical CM therapy were covered with 2 mL of either serum-free M199 primed by ASCs or human umbilical vein endothelial cells every 3 days. Wounds were assessed at day 0, 10, 15, 20, and 28. The swine were sacrificed on day 28. ImageJ software was used to evaluate the percentage of wound healing. The wounded skin underwent histologic, reverse transcription polymerase chain reaction, and enzyme-linked immunosorbent assay examinations to evaluate markers of angiogenesis and inflammation. RESULTS: We found an increase in the percentage of wound closure rates in cell-based treatments and topical therapies at various points compared with the untreated control wounds (P < .05). The results from the histologic, messenger RNA, and protein analyses suggested the treated wounds displayed increased angiogenesis and a diminished inflammatory response. CONCLUSIONS: Cellular therapy with ASCs, EC/ASCs, and topical CM accelerated diabetic wound healing in the swine model. Enhanced angiogenesis and immunomodulation might be key contributors to this process.


Assuntos
Tecido Adiposo/citologia , Diferenciação Celular , Meios de Cultivo Condicionados/farmacologia , Diabetes Mellitus Experimental/complicações , Células Progenitoras Endoteliais/transplante , Células-Tronco Multipotentes/transplante , Pele/irrigação sanguínea , Cicatrização , Ferimentos Penetrantes/terapia , Administração Tópica , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Meios de Cultivo Condicionados/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Células Progenitoras Endoteliais/metabolismo , Regulação da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Masculino , Células-Tronco Multipotentes/metabolismo , Neovascularização Fisiológica , Fenótipo , Pele/efeitos dos fármacos , Pele/lesões , Pele/metabolismo , Sus scrofa , Fatores de Tempo , Cicatrização/efeitos dos fármacos , Ferimentos Penetrantes/etiologia , Ferimentos Penetrantes/metabolismo , Ferimentos Penetrantes/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...