Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27.703
Filtrar
1.
Methods Mol Biol ; 2848: 187-196, 2025.
Artigo em Inglês | MEDLINE | ID: mdl-39240524

RESUMO

In several ocular diseases, degeneration of retinal neurons can lead to permanent blindness. Transplantation of stem cell (SC)-derived RGCs has been proposed as a potential therapy for RGC loss. Although there are reports of successful cases of SC-derived RGC transplantation, achieving long-distance regeneration and functional connectivity remains a challenge. To address these hurdles, retinal organoids are being used to study the regulatory mechanism of stem cell transplantation. Here we present a modified protocol for differentiating human embryonic stem cells (ESCs) into retinal organoids and transplanting organoid-derived RGCs into the murine eyes.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias Humanas , Células Ganglionares da Retina , Humanos , Animais , Camundongos , Células-Tronco Embrionárias Humanas/citologia , Células Ganglionares da Retina/citologia , Transplante de Células-Tronco/métodos , Organoides/citologia , Organoides/transplante , Técnicas de Cultura de Células/métodos , Terapia Baseada em Transplante de Células e Tecidos/métodos , Retina/citologia , Células-Tronco Embrionárias/citologia
2.
Methods Mol Biol ; 2848: 59-71, 2025.
Artigo em Inglês | MEDLINE | ID: mdl-39240516

RESUMO

Glaucoma is one of the leading causes of irreversible blindness. Stem cell therapy has shown promise in the treatment of primary open-angle glaucoma in animal models. Stem cell-free therapy using stem cell-derived trophic factors might be in demand in patients with high-risk conditions or religious restrictions. In this chapter, we describe methods for trabecular meshwork stem cell (TMSC) cultivation, secretome harvesting, and protein isolation, as well as assays to ensure the health of TMSC post-secretome harvesting and for secretome periocular injection into mice for therapeutic purposes.


Assuntos
Células-Tronco , Malha Trabecular , Malha Trabecular/metabolismo , Malha Trabecular/citologia , Animais , Camundongos , Humanos , Células-Tronco/citologia , Células-Tronco/metabolismo , Regeneração , Glaucoma/terapia , Transplante de Células-Tronco/métodos , Secretoma , Modelos Animais de Doenças , Glaucoma de Ângulo Aberto/terapia , Células Cultivadas , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Técnicas de Cultura de Células/métodos
3.
Medicine (Baltimore) ; 103(22): e38399, 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-39259078

RESUMO

Premature ovarian failure (POF), a condition influenced by genetic and immune factors, remains incurable despite years of intensive research and significant efforts. This persisting challenge underscores the urgency to address this escalating health concern. Fortunately, stem cell regenerative medicine has emerged as a promising avenue for developing therapeutic strategies and innovative treatments for POF. Bibliometric analysis, renowned for its objectivity, systematic approach, and comprehensive coverage of a given field, has yet to be applied to the study of stem cell research in POF. This study used CiteSpace software to assess contributions and co-occurrence relationships among various countries/regions, institutes, journals, and authors. This approach also allowed us to identify research hotspots and promising future trends within this field. Additionally, we generated visualizing maps utilizing the Web of Science Core Collection (WOSCC) and PubMed publications. By providing valuable information and references, we aim to enhance the understanding of the challenges involved in translating stem cell regeneration into clinical therapeutic potential for POF. Furthermore, our analysis and findings guide researchers and clinicians, facilitating future collaborative research and clinical intervention efforts.


Assuntos
Bibliometria , Insuficiência Ovariana Primária , Insuficiência Ovariana Primária/terapia , Feminino , Humanos , Células-Tronco , Pesquisa com Células-Tronco , Transplante de Células-Tronco/métodos , Medicina Regenerativa/métodos
4.
Stem Cell Res Ther ; 15(1): 291, 2024 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-39256865

RESUMO

BACKGROUND: Stem-cell-derived therapy is a promising option for tissue regeneration. Human iPSC-derived progenitors of smooth muscle cells (pSMCs) exhibit limited proliferation and differentiation, which minimizes the risk of tumor formation while restoring smooth muscle cells (SMCs). Up to 29% of women suffer from recurrence of vaginal prolapse after prolapse surgery. Therefore, there is a need for therapies that can restore vaginal function. SMCs contribute to vaginal tone and contractility. We sought to examine whether human pSMCs can restore vaginal function in a rat model. METHODS: Female immunocompromised RNU rats were divided into 5 groups: intact controls (n = 12), VSHAM (surgery + saline injection, n = 35), and three cell-injection groups (surgery + cell injection using pSMCs from three patients, n = 14/cell line). The surgery to induce vaginal injury was analogous to prolapse surgery. Menopause was induced by surgical ovariectomy. The vagina, urethra, bladder were harvested 10 weeks after surgery (5 weeks after cell injection). Organ bath myography was performed to evaluate the contractile function of the vagina, and smooth muscle thickness was examined by tissue immunohistochemistry. Collagen I, collagen III, and elastin mRNA and protein expressions in tissues were assessed. RESULTS: Vaginal smooth muscle contractions induced by carbachol and KCl in the cell-injection groups were significantly greater than those in the VSHAM group. Collagen I protein expression in the vagina of the cell-injections groups was significantly higher than in the VSHAM group. Vaginal elastin protein expression was similar between the cell-injection and VSHAM groups. In the urethra, gene expression levels of collagen I, III, and elastin were all significantly greater in the cell-injection groups than in the VSHAM group. Collagen I, III, and elastin protein expression of the urethra did not show a consistent trend between cell-injection groups and the VSHAM group. CONCLUSIONS: Human iPSC-derived pSMCs transplantation appears to be associated with improved contractile function of the surgically injured vagina in a rat model. This is accompanied by changes in extracellular protein expression the vagina and urethra. These observations support further efforts in the translation of pSMCs into a treatment for regenerating the surgically injured vagina in women who suffer recurrent prolapse after surgery.


Assuntos
Modelos Animais de Doenças , Miócitos de Músculo Liso , Vagina , Animais , Feminino , Ratos , Humanos , Miócitos de Músculo Liso/metabolismo , Transplante de Células-Tronco/métodos , Elastina/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Contração Muscular , Diferenciação Celular
6.
Stem Cell Res Ther ; 15(1): 290, 2024 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-39256845

RESUMO

BACKGROUND: This comprehensive systematic review and meta-analysis investigated the mid- to long-term efficacy and safety of stem cell therapy in patients with acute myocardial infarction (AMI). METHODS: The study encompassed 79 randomized controlled trials with 7103 patients, rendering it the most up-to-date and extensive analysis in this field. This study specifically focused on the impact of stem cell therapy on left ventricular ejection fraction (LVEF), major adverse cardiac events (MACE), and infarct size. RESULTS: Stem cell therapy significantly improved LVEF at 6, 12, 24, and 36 months post-transplantation compared to control values, indicating its potential for long-term cardiac function enhancement. A trend toward reduced MACE occurrence was observed in the intervention groups, suggesting the potential of stem cell therapy to lower the risk of cardiovascular death, reinfarction, and stroke. Significant LVEF improvements were associated with long cell culture durations exceeding 1 week, particularly when combined with high injected cell quantities (at least 108 cells). No significant reduction in infarct size was observed. CONCLUSIONS: This review highlights the potential of stem cell therapy as a promising therapeutic approach for patients with AMI, offering sustained LVEF improvement and a potential reduction in MACE risk. However, further research is required to optimize cell culture techniques, determine the optimal timing and dosage, and investigate procedural variations to maximize the efficacy and safety of stem cell therapy in this context.


Assuntos
Infarto do Miocárdio , Transplante de Células-Tronco , Humanos , Infarto do Miocárdio/terapia , Transplante de Células-Tronco/métodos , Ensaios Clínicos Controlados Aleatórios como Assunto , Volume Sistólico , Resultado do Tratamento , Função Ventricular Esquerda/fisiologia
7.
Cells ; 13(17)2024 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-39273075

RESUMO

Spinal cord injury (SCI) is a serious condition accompanied by severe adverse events that affect several aspects of the patient's life, such as motor, sensory, and functional impairment. Despite its severe consequences, definitive treatment for these injuries is still missing. Therefore, researchers have focused on developing treatment strategies aimed at ensuring full recovery post-SCI. Accordingly, attention has been drawn toward cellular therapy using mesenchymal stem cells. Considering their wide availability, decreased immunogenicity, wide expansion capacity, and impressive effectiveness in many therapeutic approaches, adipose-derived stem cell (ADSC) injections in SCI cases have been investigated and showed promising results. In this review, SCI pathophysiology and ADSC transplantation benefits are discussed independently, together with SCI animal models and adipose stem cell preparation and application techniques. The mechanisms of healing in an SCI post-ADSC injection, the outcomes of this therapeutic approach, and current clinical trials are also deliberated, in addition to the challenges and future perspectives, aiming to encourage further research in this field.


Assuntos
Tecido Adiposo , Traumatismos da Medula Espinal , Transplante de Células-Tronco , Traumatismos da Medula Espinal/terapia , Humanos , Animais , Tecido Adiposo/citologia , Transplante de Células-Tronco/métodos , Células-Tronco/citologia , Terapia Baseada em Transplante de Células e Tecidos/métodos , Transplante de Células-Tronco Mesenquimais/métodos , Modelos Animais de Doenças
8.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 49(6): 989-997, 2024 Jun 28.
Artigo em Inglês, Chinês | MEDLINE | ID: mdl-39311795

RESUMO

The dental pulp is the only soft tissue structure within the tooth, serving functions such as sensation and nutrition. However, the dental pulp is highly susceptible to necrosis due to external factors. Currently, root canal therapy is the most commonly used treatment for pulp necrosis. Nevertheless, teeth treated with root canal therapy are prone to secondary infections and adverse outcomes like vertical root fractures. Regenerative endodontic therapy has emerged as a solution, aiming to replace damaged tooth structures, including dentin, root structure, and the pulp-dentin complex cells. This approach demonstrates significant advantages in addressing clinical symptoms and achieving regeneration of the root and even the pulp. Since the discovery of dental pulp stem cells, regenerative endodontic therapy has gained new momentum. Advances in cell transplantation and cell homing techniques have rapidly developed, showing promising potential for clinical applications.


Assuntos
Polpa Dentária , Regeneração , Transplante de Células-Tronco , Polpa Dentária/fisiologia , Polpa Dentária/citologia , Humanos , Regeneração/fisiologia , Transplante de Células-Tronco/métodos , Endodontia Regenerativa/métodos , Células-Tronco/citologia , Tratamento do Canal Radicular/métodos , Engenharia Tecidual/métodos , Necrose da Polpa Dentária/terapia
9.
Nat Commun ; 15(1): 8132, 2024 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-39284802

RESUMO

Mucopolysaccharidoses are inherited metabolic disorders caused by the deficiency in lysosomal enzymes required to break down glycosaminoglycans. Accumulation of glycosaminoglycans leads to progressive, systemic degenerative disease. The central nervous system is particularly affected, resulting in developmental delays, neurological regression, and early mortality. Current treatments fail to adequately address neurological defects. Here we explore the potential of human induced pluripotent stem cell (hiPSC)-derived microglia progenitors as a one-time, allogeneic off-the-shelf cell therapy for several mucopolysaccharidoses (MPS). We show that hiPSC-derived microglia progenitors, possessing normal levels of lysosomal enzymes, can deliver functional enzymes into four subtypes of MPS knockout cell lines through mannose-6-phosphate receptor-mediated endocytosis in vitro. Additionally, our findings indicate that a single administration of hiPSC-derived microglia progenitors can reduce toxic glycosaminoglycan accumulation and prevent behavioral deficits in two different animal models of MPS. Durable efficacy is observed for eight months after transplantation. These results suggest a potential avenue for treating MPS with hiPSC-derived microglia progenitors.


Assuntos
Modelos Animais de Doenças , Glicosaminoglicanos , Células-Tronco Pluripotentes Induzidas , Microglia , Mucopolissacaridoses , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Animais , Microglia/metabolismo , Humanos , Mucopolissacaridoses/terapia , Camundongos , Glicosaminoglicanos/metabolismo , Camundongos Knockout , Diferenciação Celular , Transplante de Células-Tronco/métodos , Lisossomos/metabolismo
10.
J Med Life ; 17(6): 582-587, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-39296433

RESUMO

Follicular unit hair extraction (FUE) is effective for hair restoration but is less successful on scarred tissue due to reduced vascularity and altered tissue architecture. Stem cell therapy can enhance tissue regeneration, possibly improving FUE outcomes on scarred tissue. This study investigated the impact of stem cell therapy prior to FUE on scarred tissue. Sixty patients with scalp scars from trauma or previous surgeries were divided into two groups. Group A (n = 30) received autologous stem cell therapy followed by FUE, while Group B (n = 30) underwent FUE without prior stem cell treatment. Autologous stem cells were harvested from patients' adipose tissue and injected into the scarred area four weeks before FUE. Outcomes were assessed at 3-, 6-, and 12-months post-transplantation, focusing on hair density, graft survival rate, and patient satisfaction. Histological examinations evaluated tissue regeneration. Group A showed significantly higher hair density (mean increase of 45%) and graft survival rates (87%) compared to Group B (mean increase of 25%, graft survival rate of 60%) at all follow-up points (P < 0.05). Histological analysis revealed enhanced neovascularization and reduced fibrosis in the stem cell-treated group, with 70% more new blood vessels and 50% less fibrotic tissue compared to the control group. Patient satisfaction scores were higher in Group A (average score of 8.5 out of 10) versus Group B (6.0), indicating better aesthetic outcomes and reduced scar visibility. Pre-treatment with autologous stem cell therapy significantly improved FUE effectiveness on scarred tissue, enhancing graft survival, hair density, and patient satisfaction. Further research is recommended to optimize this therapeutic strategy.


Assuntos
Cicatriz , Folículo Piloso , Transplante de Células-Tronco , Humanos , Cicatriz/terapia , Cicatriz/patologia , Folículo Piloso/transplante , Feminino , Adulto , Transplante de Células-Tronco/métodos , Masculino , Pessoa de Meia-Idade , Transplante Autólogo , Alopecia/terapia , Couro Cabeludo , Cabelo/transplante , Adulto Jovem
11.
Chin Clin Oncol ; 13(Suppl 1): AB079, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39295397

RESUMO

BACKGROUND: Whole brain radiotherapy (WBRT) is commonly used as consolidation therapy in primary central nervous system lymphoma (PCNSL). However, high-dose chemotherapy followed by autologous stem cell transplantation (HD-ASCT) has emerged as an alternative approach for PCNSL. This systematic review aims to assess the efficacy and safety of both treatment modalities. METHODS: The systematic review follows PRISMA guidelines. A comprehensive search strategy identified relevant studies from PubMed, Europe PMC, and Cochrane Library. The following search terms were used: "primary central nervous system lymphoma", "Autologous Stem Cell Transplantation", and "whole-brain radiotherapy". We included randomized controlled trials (RCTs) cohort studies evaluating the use of whole-brain radiotherapy and high-dose chemotherapy followed by autologous stem cell transplantation in the treatment of histologically-confirmed PCNSL. Publications included were limited to English language full texts that were published in the past 10 years. Data extraction & manuscript quality assessment was done by two independent reviewers with a third reviewer to resolve any discrepancy. Primary outcomes include overall survival (OS), progression-free survival (PFS) & treatment related toxicity (TRT). Secondary outcomes were clinical neurological function and performance score assessments. Individual studies were assessed using the Jadad Scale and the Newcastle-Ottawa Scale for observational studies. RESULTS: We identified 5 studies, consisting of 2 RCTs and 3 cohort studies. After all studies considered, analysis revealed that consolidation therapy with HD-ASCT had a better overall PFS and OS compared to whole-brain radiotherapy (P<0.005). Both groups showed similar TRT with mostly haematological toxicity. Holistically clinical cognitive functions are found to be improved in HD-ASCT Patients and poorer results are exhibited by WBRT patients primarily in executive functions. Performance statuses are scored differently across all studies with slightly preferable results shown in patients treated with HDC-ASCT. CONCLUSIONS: Based on the findings of this systematic review, HDC-ASCT might be a preferable choice of consolidative therapy as shown with better OS, PFS with similar TRT. While WBRT are more feasible and cost-efficient, risks of cognitive impairment and reduced performance status after WBRT should be considered for further treatment choices. Further randomized clinical trials with a similar scoring system are needed.


Assuntos
Neoplasias do Sistema Nervoso Central , Linfoma , Transplante Autólogo , Humanos , Transplante Autólogo/métodos , Neoplasias do Sistema Nervoso Central/terapia , Linfoma/terapia , Transplante de Células-Tronco Hematopoéticas/métodos , Transplante de Células-Tronco/métodos
13.
Exp Parasitol ; 265: 108823, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39187057

RESUMO

Trichinosis is a common parasitic disease that affects the striated skeletal muscles, causing apoptotic and degenerative changes associated with myogenin expression in the affected myocytes. Hence, this study aimed to assess the ameliorative effects of stem cells and atorvastatin added to ivermectin on the infected myocytes during the muscular phase of murine trichinosis. 120 laboratory Swiss albino male mice were divided into 10 groups, and each group was subdivided into intestinal and muscular phases (each n = 6); uninfected control; untreated infected control; infected received ivermectin monotherapy; infected received atorvastatin monotherapy; infected received stem cells monotherapy; infected received ivermectin and atorvastatin dual therapy; infected received ivermectin and stem cells dual therapy; infected received atorvastatin and stem cells dual therapy; infected received ivermectin 0.2, atorvastatin 40, and stem cells triple therapy; and infected received ivermectin 0.1, atorvastatin 20, and stem cells triple therapy. Intestinal phase mice were sacrificed on the 5th day post-infection, while those of the muscular phase were sacrificed on the 35th day post-infection. Parasitological, histopathological, ultrastructural, histochemical, biochemical, and myogenin gene expression assessments were performed. The results revealed that mice that received ivermectin, atorvastatin, and stem cell triple therapies showed the maximum reduction in the adult worm and larvae burden, marked improvement in the underlying muscular degenerative changes (as was noticed by histopathological, ultrastructural, and histochemical Feulgen stain assessment), lower biochemical levels of serum NK-κB and tissue NO, and lower myogenin expression. Accordingly, the combination of stem cells, atorvastatin, and ivermectin affords a potential synergistic activity against trichinosis with considerable healing of the underlying degenerative sequel.


Assuntos
Apoptose , Atorvastatina , Ivermectina , Miogenina , Triquinelose , Animais , Atorvastatina/farmacologia , Atorvastatina/uso terapêutico , Masculino , Camundongos , Ivermectina/farmacologia , Ivermectina/uso terapêutico , Triquinelose/tratamento farmacológico , Triquinelose/parasitologia , Apoptose/efeitos dos fármacos , Miogenina/genética , Miogenina/metabolismo , Músculo Esquelético/parasitologia , Músculo Esquelético/patologia , Músculo Esquelético/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Microscopia Eletrônica de Transmissão , Transplante de Células-Tronco , Trichinella spiralis/genética , Trichinella spiralis/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos
14.
Stem Cell Res Ther ; 15(1): 253, 2024 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-39135088

RESUMO

Stem cell therapy (SCT) is a promising solution for addressing health challenges in Africa, particularly non-communicable diseases (NCDs). With their regenerative potential, stem cells have the inherent capacity to differentiate into numerous cell types for tissue repair. Despite infrastructural, ethical, and legal challenges, SCT holds immense promise for managing chronic illnesses and deep-seated tissue injuries. The rising prevalence of NCDs in Africa highlights the need for innovative strategies and treatment options. SCT offers hope in combating conditions like burns, osteoarthritis, diabetes, Alzheimer's disease, stroke, heart failure and cancer, potentially reducing the burden of NCDs on the continent. Despite SCT's opportunities in Africa, there are significant obstacles. However, published research on SCT in Africa is scarce, but recent initiatives such as the Basic School on Neural Stem Cells (NSC) express interest in developing NSC research in Africa. SCT research in African regions, notably on neurogenesis, demonstrates a concentration on studying neurological processes in indigenous settings. While progress has been made in South Africa and Nigeria, issues such as brain drain and impediments to innovation remain. Clinical trials have investigated the efficacy of stem cell treatments, emphasising both potential benefits and limitations in implementing these therapies efficiently. Financing research, developing regulatory frameworks, and resolving affordability concerns are critical steps toward realizing the potential of stem cell treatment in Africa.


Assuntos
Doenças não Transmissíveis , Transplante de Células-Tronco , Humanos , Doenças não Transmissíveis/terapia , África/epidemiologia , Transplante de Células-Tronco/métodos , Terapia Baseada em Transplante de Células e Tecidos/métodos
15.
Nat Commun ; 15(1): 7223, 2024 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-39174514

RESUMO

Electrical stimulation holds promise for enhancing neuronal differentiation of neural stem cells to treat traumatic brain injury. However, once the stem cells leave the stimulating material and migrate post transplantation, electrical stimulation on them is diminished. Here, we wrap the stem cells with wireless electrical nanopatches, the conductive graphene nanosheets. Under electromagnetic induction, electrical stimulation can thus be applied in-situ to individual nanopatch-wrapped stem cells on demand, stimulating their neuronal differentiation through a MAPK/ERK signaling pathway. Consequently, 41% of the nanopatch-wrapped stem cells differentiate into functional neurons in 5 days, as opposed to only 16.3% of the unwrapped ones. The brain injury male mice implanted with the nanopatch-wrapped stem cells and exposed to a rotating magnetic field 30 min/day exhibit significant recovery of brain tissues, behaviors, and cognitions, within 28 days. This study opens up an avenue to individualized electrical stimulation of transplanted stem cells for treating neurodegenerative diseases.


Assuntos
Lesões Encefálicas Traumáticas , Diferenciação Celular , Células-Tronco Neurais , Transplante de Células-Tronco , Animais , Lesões Encefálicas Traumáticas/terapia , Lesões Encefálicas Traumáticas/patologia , Masculino , Camundongos , Células-Tronco Neurais/transplante , Células-Tronco Neurais/citologia , Transplante de Células-Tronco/métodos , Grafite/química , Estimulação Elétrica , Tecnologia sem Fio , Neurônios , Humanos , Encéfalo , Nanoestruturas/química
17.
Neurobiol Dis ; 200: 106650, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39197536

RESUMO

The human body is a complex, integral whole, and disruptions in one organ can lead to dysfunctions in other parts of the organ network. The facial nerve, as the seventh cranial nerve, arises from the brainstem, controls facial expression muscles and plays a crucial role in brain-body communication. This vulnerable nerve can be damaged by trauma, inflammation, tumors, and congenital diseases, often impairing facial expression. Stem cells have gained significant attention for repairing peripheral nerve injuries due to their multidirectional differentiation potential. Additionally, various biomaterials have been used in tissue engineering for regeneration and repair. However, the therapeutic potential of stem cells and biomaterials in treating facial nerve injuries requires further exploration. In this review, we summarize the roles of stem cells and biomaterials in the regeneration and repair of damaged facial nerves, providing a theoretical basis for the recovery and reconstruction of body-brain crosstalk between the brain and facial expression muscles.


Assuntos
Materiais Biocompatíveis , Nervo Facial , Regeneração Nervosa , Humanos , Regeneração Nervosa/fisiologia , Animais , Nervo Facial/fisiologia , Encéfalo/fisiologia , Traumatismos do Nervo Facial/terapia , Traumatismos do Nervo Facial/fisiopatologia , Células-Tronco/fisiologia , Transplante de Células-Tronco/métodos , Engenharia Tecidual/métodos
18.
Int J Mol Sci ; 25(16)2024 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-39201431

RESUMO

Recent studies have highlighted the therapeutic potential of stem cells for various diseases. However, unlike other tissues, brain tissue has a specific structure, consisting of synapses. These synapses not only transmit but also process and refine information. Therefore, synaptic regeneration plays a key role in therapy of neurodegenerative disorders. Neurexins (NRXNs) and neuroligins (NLGNs) are synaptic cell adhesion molecules that connect pre- and postsynaptic neurons at synapses, mediate trans-synaptic signaling, and shape neural network properties by specifying synaptic functions. In this study, we investigated the synaptic regeneration effect of human neural stem cells (NSCs) overexpressing NRXNs (F3.NRXN) and NLGNs (F3.NLGN) in a spinal cord injury model. Overexpression of NRXNs and NLGNs in the neural stem cells upregulated the expression of synaptophysin, PSD95, VAMP2, and synapsin, which are synaptic markers. The BMS scores indicated that the transplantation of F3.NRXN and F3.NLGN enhanced the recovery of locomotor function in adult rodents following spinal cord injury. Transplanted F3.NRXN and F3.NLGN differentiated into neurons and formed a synapse with the host cells in the spinal cord injury mouse model. In addition, F3.NRXN and F3.NLGN cells restored growth factors (GFs) and neurotrophic factors (NFs) and induced the proliferation of host cells. This study suggested that NSCs overexpressing NRXNs and NLGNs could be candidates for cell therapy in spinal cord injuries by facilitating synaptic regeneration.


Assuntos
Moléculas de Adesão Celular Neuronais , Modelos Animais de Doenças , Células-Tronco Neurais , Traumatismos da Medula Espinal , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/terapia , Traumatismos da Medula Espinal/genética , Células-Tronco Neurais/metabolismo , Animais , Humanos , Moléculas de Adesão Celular Neuronais/metabolismo , Moléculas de Adesão Celular Neuronais/genética , Camundongos , Sinapses/metabolismo , Transplante de Células-Tronco/métodos , Diferenciação Celular , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/genética , Feminino , Neuroliginas
19.
Int J Mol Sci ; 25(16)2024 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-39201545

RESUMO

Stargardt disease, one of the most common forms of inherited retinal diseases, affects individuals worldwide. The primary cause is mutations in the ABCA4 gene, leading to the accumulation of toxic byproducts in the retinal pigment epithelium (RPE) and subsequent photoreceptor cell degeneration. Over the past few years, research on Stargardt disease has advanced significantly, focusing on clinical and molecular genetics. Recent studies have explored various innovative therapeutic approaches, including gene therapy, stem cell therapy, and pharmacological interventions. Gene therapy has shown promise, particularly with adeno-associated viral (AAV) vectors capable of delivering the ABCA4 gene to retinal cells. However, challenges remain due to the gene's large size. Stem cell therapy aims to replace degenerated RPE and photoreceptor cells, with several clinical trials demonstrating safety and preliminary efficacy. Pharmacological approaches focus on reducing toxic byproduct accumulation and modulating the visual cycle. Precision medicine, targeting specific genetic mutations and pathways, is becoming increasingly important. Novel techniques such as clustered regularly interspaced palindromic repeats (CRISPR)/Cas9 offer potential for directly correcting genetic defects. This review aims to synthesize recent advancements in understanding and treating Stargardt disease. By highlighting breakthroughs in genetic therapies, stem cell treatments, and novel pharmacological strategies, it provides a comprehensive overview of emerging therapeutic options.


Assuntos
Transportadores de Cassetes de Ligação de ATP , Terapia Genética , Doença de Stargardt , Doença de Stargardt/genética , Humanos , Terapia Genética/métodos , Transportadores de Cassetes de Ligação de ATP/genética , Mutação , Transplante de Células-Tronco/métodos , Animais , Degeneração Macular/terapia , Degeneração Macular/genética , Degeneração Macular/congênito , Epitélio Pigmentado da Retina/metabolismo , Sistemas CRISPR-Cas
20.
Int Braz J Urol ; 50(6): 703-713, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39133793

RESUMO

OBJECTIVE: To describe the evidence of Platelet Rich Plasma (PRP), Stem cells therapy (SCT) and Extracorporeal shockwave therapy (ESWL) for the treatment of Peyronies disease (PD), including information from the main urological society guidelines. MATERIALS AND METHODS: A literature review of PubMed articles published between 2000 and 2023 was conducted, utilizing keywords such as "Peyronie's Disease", "Penile curvature", "Platelet Rich Plasma", "Stem cells", and "Extracorporeal shockwave therapy". Only full-text articles in English were included, excluding case reports and opinions. RESULTS: A considerable number of clinical trials were conducted using PRP penile injections for therapy of PD, showing reduction of curvature, plaque size and improvement in quality of life. Preclinical studies in rats have shown the potential benefit of adipose-derived stem cells, with improvements in erectile function and fibrosis. Human studies with mesenchymal stem cells demonstrated promising results, with reduction of curvature and plaque size. ESWL effects on PD were investigated in randomized clinical trials and demonstrated no significant impact in curvature or plaque size, but reasonable effect on pain control. CONCLUSION: Restorative therapies has emerged as an innovative treatment option for PD and the results from current studies appear to be promising and demonstrated good safety profile. Unfortunately, due to scarce evidence, PRP and SCT are still considered experimental by American Urological Association (AUA) and European Association of Urology (EAU) guidelines. ESWT is recommended, by the same guidelines, for pain control only. More high-quality studies with long-term follow-up outcomes are needed to evaluate efficacy and reproducibility of those therapies.


Assuntos
Tratamento por Ondas de Choque Extracorpóreas , Induração Peniana , Plasma Rico em Plaquetas , Transplante de Células-Tronco , Induração Peniana/terapia , Humanos , Masculino , Tratamento por Ondas de Choque Extracorpóreas/métodos , Transplante de Células-Tronco/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA