Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Chem Neuroanat ; 132: 102306, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37394105

RESUMEN

In multiple sclerosis (MS), activation of the astrocytes and microglia induces a cascading inflammatory response. Overexpression of the aquaporin 4 (AQP4) in the glia is a trigger for this reaction. This study aimed to block AQP4 by injecting TGN020 to alleviate the symptoms of MS. Total of 30 male mice were randomly divided into control (intact), cuprizone model of MS (fed with 0.2% cuprizone for 35 days), and TGN020-treated (received daily intraperitoneal injections of 200 mg/kg TGN020 with cuprizone intake) groups. Astrogliosis, M1-M2 microglia polarization, NLRP3 inflammasome activation, and demyelination were investigated in the corpus callosum by immunohistochemistry, real-time PCR, western blot, and luxol fast blue staining. The Rotarod test was performed for a behavior assessment. AQP4 inhibition caused a significant decrease in the expression of the astrocyte-specific marker, GFAP. It also changed the microglia polarization from M1 to M2 indicated by a significant downregulation of iNOS, CD86, MHC-ІІ, and upregulation of arginase1, CD206, and TREM-2. In addition, western blot data showed a significant decrease in the NLRP3, caspase1, and IL-1b proteins in the treatment group, which indicated inflammasome inactivation. The molecular changes following the TGN020 injection resulted in remyelination and motor recovery enhancement in the treatment group. In conclusion, the results draw the attention to the role of AQP4 in the cuprizone model of MS.


Asunto(s)
Enfermedades Desmielinizantes , Esclerosis Múltiple , Masculino , Animales , Ratones , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/metabolismo , Microglía/metabolismo , Cuprizona , Astrocitos/metabolismo , Inflamasomas/metabolismo , Acuaporina 4/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/tratamiento farmacológico , Enfermedades Desmielinizantes/metabolismo , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad
2.
J Neuroimmunol ; 360: 577707, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34507013

RESUMEN

Intrauterine inflammation affects fetal development of the nervous system and may cause prenatal brain injury in offspring. Previously, neural stem cells have been extensively used as a therapeutic choice for nervous system diseases. Recently, the therapeutic ability of conditioned medium, harvested from cultured stem cells, has captured the attention of researchers in the field. Our study aimed to compare the therapeutic effect of neural stem cells (NSCs) or NSC-conditioned medium (NSC-CM) after prenatal brain injury. The animal model was induced by intraperitoneal injection of lipopolysaccharide into the pregnant mice and NSCs or NSC-CM were transplanted into the lateral ventricle of embryos in treatment groups. Inflammation and apoptosis were evaluated postpartum in offspring via measuring the expression of NLRP3 gene and protein, the expression and the activity of caspase-3, and the expression of pro-inflammatory cytokines by real-time PCR, immunohistochemistry, western blotting, ELISA, and colorimetric assay kit. A rotarod test was performed for motor function evaluation. Data showed that although NSC-CM fought against the inflammation and apoptosis and improved the motor function, NSCs acted more efficiently. In conclusion, the results of our study contend that NSCs have a better therapeutic effect than CM in prenatal brain injury.


Asunto(s)
Lesiones Encefálicas/terapia , Medios de Cultivo Condicionados/farmacología , Encefalitis/terapia , Enfermedades Fetales/terapia , Terapias Fetales , Células-Madre Neurales/trasplante , Animales , Apoptosis , Lesiones Encefálicas/embriología , Lesiones Encefálicas/etiología , Caspasa 3/biosíntesis , Caspasa 3/genética , Células Cultivadas , Citocinas/biosíntesis , Citocinas/genética , Modelos Animales de Enfermedad , Encefalitis/etiología , Femenino , Enfermedades Fetales/etiología , Inflamasomas/fisiología , Inyecciones Intraperitoneales , Inyecciones Intraventriculares , Ventrículos Laterales , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/toxicidad , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/biosíntesis , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Embarazo , Efectos Tardíos de la Exposición Prenatal , Prueba de Desempeño de Rotación con Aceleración Constante
3.
Cell Mol Neurobiol ; 41(7): 1467-1481, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32594382

RESUMEN

Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system. The main causes of MS disease progression, demyelination, and tissue damage are oxidative stress and mitochondrial dysfunction. Hence, the latter are considered as important therapeutic targets. Recent studies have demonstrated that mesenchymal stem cells (MSCs) possess antioxidative properties and are able to target mitochondrial dysfunction. Therefore, we investigated the effect of transplanting Wharton's jelly-derived MSCs in a demyelination mouse model of MS in which mice were fed cuprizone (CPZ) for 12 weeks. CPZ is a copper chelator that impairs the activity of cytochrome oxidase, decreases oxidative phosphorylation, and produces degenerative changes in oligodendrocytes, leading to toxic demyelination similar to those found in MS patients. Results showed that MSCs caused a significant increase in the percentage of myelinated areas and in the number of myelinated fibers in the corpus callosum of the CPZ + MSC group, compared to the CPZ group, as assessed by Luxol fast blue staining and transmission electron microscopy. In addition, transplantation of MSCs significantly increased the number of oligodendrocytes while decreasing astrogliosis and microgliosis in the corpus callosum of the CPZ + MSC group, evaluated by immunofluorescence. Moreover, the mechanism by which MSCs exert these physiological effects was found to be through abolishing the effect of CPZ on oxidative stress markers and mitochondrial dysfunction. Indeed, malondialdehyde significantly decreased while glutathione and superoxide dismutase significantly increased in CPZ + MSC mice group, in comparison witth the CPZ group alone. Furthermore, cell therapy with MSC transplantation increased the expression levels of mitochondrial biogenesis transcripts PGC1α, NRF1, MFN2, and TFAM. In summary, these results demonstrate that MSCs may attenuate MS by promoting an antioxidant response, reducing oxidative stress, and improving mitochondrial homeostasis.


Asunto(s)
Células Madre Mesenquimatosas/metabolismo , Mitocondrias/efectos de los fármacos , Oligodendroglía/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Remielinización/efectos de los fármacos , Animales , Cuprizona/farmacología , Enfermedades Desmielinizantes/tratamiento farmacológico , Enfermedades Desmielinizantes/metabolismo , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Esclerosis Múltiple/metabolismo , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/metabolismo , Oligodendroglía/metabolismo
4.
J Chem Neuroanat ; 106: 101792, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32353514

RESUMEN

In multiple sclerosis, myelin sheaths around the axons are degenerated due to uncontrolled inflammation in the central nervous system. Oligodendrocytes (OLs) are myelin-forming cells that secrete trophic factors necessary for myelin protection. Beneficial features of conditioned medium (CM) derived from different stem cells are nowadays under investigation in treating neurodegenerative diseases. Here, we used the differentiation capacity of Wharton's jelly mesenchymal stem cells (WJMSCs) to obtain OLs. Then, the study aimed to evaluate the status of inflammation and myelination in male experimental autoimmune encephalomyelitis (EAE) mice after intranasal administration of CM derived from OLs (OL-CM). Inflammation was studied by evaluating gliosis, inflammatory cell infiltration and expression of inflammation indicators including NLRP3 inflammasome, interleukin-1ß, interleukin-18, glial fibrillary acidic protein, and ionized calcium binding adaptor molecule 1. Remyelination was studied by luxol fast blue staining and evaluating the expression of myelin indicators including myelin basic protein and oligodendrocyte transcription factor. In addition, we followed the trend of body weight and functional recovery during the 28-day study. ELISA assay revealed that OL-CM contained brain-derived neurotrophic factor, glial cell-derived neurotrophic factor, and ciliary neurotrophic factor. Data showed that OL-CM moderated inflammation, augmented remyelination, and gained normal body weight. Notably, these anti-inflammatory and regenerative effects of OL-CM improved neurological functions in EAE mice. In conclusion, the current study offered a new choice for treating multiple sclerosis using noninvasive intranasal administration of CM harvested from easily achievable WJMSCs-differentiated OLs.


Asunto(s)
Diferenciación Celular , Medios de Cultivo Condicionados/farmacología , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Células Madre Mesenquimatosas , Oligodendroglía , Administración Intranasal , Animales , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Inflamación/metabolismo , Ratones
5.
Neuropathology ; 40(1): 84-92, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31709666

RESUMEN

Experimental autoimmune encephalomyelitis (EAE) as an experimental model of multiple sclerosis (MS) is characterized by demyelination, infiltration of inflammatory cells into the nervous system and dysregulation of serum inflammatory cytokines. We investigated the correlation of serum cytokines and other inflammatory markers with the EAE pathogenesis. After EAE induction, the levels of different serum cytokine/inflammatory mediators were measured. Furthermore, motor functions, myelination, and lymphocyte infiltration in EAE mice were also assessed. Our results revealed that the serum concentrations of T-helper 1 (Th1) and Th17 cytokines, interleukin (IL)-6, IL-1ß, IL-1α and prostaglandin E2 in EAE mice were significantly higher than controls. The ratios of pro- to anti-inflammatory cytokines were different between the EAE and the control group. A statistically significant positive correlation was found between the IL-6/IL-10 ratio and the EAE severity, demyelination rate, and lymphocyte infiltration in EAE mice. Results indicate that the profiles of serum pro- and anti-inflammatory cytokines might be useful as biomarkers for monitoring the pathological manifestation of EAE. Furthermore, evaluating the dynamic interplay of serum cytokine levels and the correlation with pathogenic mechanisms of EAE may provide diagnostic and therapeutic insights for MS and some other inflammatory disorders.


Asunto(s)
Citocinas/sangre , Encefalomielitis Autoinmune Experimental/sangre , Encefalomielitis Autoinmune Experimental/patología , Mediadores de Inflamación/sangre , Animales , Biomarcadores/sangre , Femenino , Ratones , Ratones Endogámicos C57BL
6.
Asian Spine J ; 14(2): 131-138, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31711062

RESUMEN

STUDY DESIGN: Experimental animal study. PURPOSE: This study investigated the therapeutic effects of human breast milk stem cell (BMSC)-conditioned medium (BMSC-CM) in a model of spinal cord injury (SCI) in male Sprague-Dawley rats. OVERVIEW OF LITERATURE: SCI is one of the leading causes of disability in addition to sensory and motor impairment. So far, there have been no successful treatments for SCI. Given the positive outcomes associated with using stem cells and their derivatives as a treatment for various diseases, there is a growing interest in using them as an SCI treatment. Recent research has demonstrated that CM from stem cells has therapeutic advantages. METHODS: Human BMSCs were isolated and characterized, and CM was subsequently collected. Animals received an intrathecal administration of BMSC-CM after SCI. The activity of caspase-3 was measured to assess apoptosis, and levels of tumor necrosis factor-α and interleukin-1ß were measured to assess inflammation. Also, sensory and locomotor performances were assessed after SCI and BMSC-CM administration. RESULTS: Administration of CM from BMSC reduced apoptosis and inflammation at the site of injury in a rat model of SCI (p<0.05). Motor, sensory, locomotor, and sensorimotor performances were significantly improved in rats that received BMSC-CM after SCI. CONCLUSIONS: Intrathecal administration of BMSC-CM improved recovery in a rat model of SCI.

7.
Cell Reprogram ; 21(5): 249-259, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31596622

RESUMEN

We recently reported the application of human menstrual blood stem cells' (HuMenSCs) transplantation as a treatment modality in a rat model of premature ovarian failure (POF). We continued to investigate further in this respect. Female rats were injected intraperitoneally with 36 mg/kg busulfan. HuMenSCs were obtained, grown, and analyzed for immunophenotypic features at passage three. The cells were labeled with CM-Dil and infused into the rats. There were four groups: normal, negative control, treatment, and Sham. One month after treatment, the ovaries were collected and weighed. Histological sections were prepared from the ovary and HuMenSCs were tracking. Subsequently, we examined the changes of expression of Bax and B cell lymphoma 2 (Bcl2) genes by real-time polymerase chain reaction assay. One month after HuMenSCs transplantation, these cells were located in the ovarian interstitium and granulosa cells (GCs). The number of TUNEL-positive cells significantly decreased in the treatment group. Also the expression level of Bax genes, unlike Bcl2 gene, significantly decreased compared with negative and sham groups. In our study, HuMenSCs were tracked in ovarian tissues within 2 months after transplantation, and they differentiated into GCs. Therefore, the use of these cells can be a practical and low-cost method for the treatment of POF patients.


Asunto(s)
Células Sanguíneas/citología , Busulfano/toxicidad , Células de la Granulosa/citología , Folículo Ovárico/citología , Insuficiencia Ovárica Primaria/terapia , Trasplante de Células Madre/métodos , Células Madre/citología , Adulto , Alquilantes/toxicidad , Animales , Femenino , Humanos , Menstruación , Insuficiencia Ovárica Primaria/inducido químicamente , Insuficiencia Ovárica Primaria/patología , Ratas , Ratas Wistar , Adulto Joven
8.
J Neuroimmunol ; 333: 476958, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31103935

RESUMEN

The use of growth factors is considered to be one of the promising therapeutic strategies for multiple sclerosis (MS). Various studies have shown that platelet-rich plasma (PRP), a bioproduct of concentrated platelets, contains a variety of growth factors such as insulin-like growth factor 1 (IGF-1), platelet-derived growth factor (PDGF), epithelial growth factor (EGF), and transforming growth factor ß (TGF-ß). The therapeutic roles of PRP, with regard to a wide range of growth factors, on the nervous system have been shown in a limited number of studies. This study aimed to investigate the therapeutic effect of PRP in experimental autoimmune encephalomyelitis (EAE) mouse model of MS. PRP was prepared and intrathecally injected into the EAE mice. The EAE scoring test, the modified neurological severity score (mNSS) test, luxol fast blue and hematoxylin and eosin staining, real-time PCR, and western blotting were used for studying the effect of PRP on the motosensory function, remyelination, inflammatory cell infiltration, gliosis, and inflammatory cytokines expression. PRP administration in treated animals improved the functional abilities, remyelination, and oligodendrogenesis compared to the EAE mice. Furthermore, high numbers of microglia, astrocytes and infiltrating inflammatory cells and also the expression of proinflammatory cytokines were reversed after PRP therapy. In conclusion, these data suggest the PRP as a potential candidate for MS treatment.


Asunto(s)
Encefalomielitis Autoinmune Experimental/terapia , Plasma Rico en Plaquetas , Animales , Encefalomielitis Autoinmune Experimental/patología , Femenino , Gliosis/patología , Gliosis/prevención & control , Inflamación , Inyecciones Espinales , Interleucina-1beta/biosíntesis , Interleucina-1beta/genética , Interleucina-6/biosíntesis , Interleucina-6/genética , Ratones , Ratones Endogámicos C57BL , Proteína Básica de Mielina/biosíntesis , Proteína Básica de Mielina/genética , Plasma Rico en Plaquetas/inmunología , Médula Espinal/patología , Regulación hacia Arriba
9.
J Mol Histol ; 50(3): 263-271, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31016544

RESUMEN

Oxidative stress with mitochondrial defects has a central role in the development and deterioration of Multiple sclerosis (MS). According to new findings of the effects of metformin on mitochondrial function, has attracted a lot of attention. Furthermore, it is suggested that metformin exerts its beneficial influence through AMP-activated protein kinase (AMPK) pathway. In the current study, we investigated the possible protective effects of metformin on oxidative stress and mitochondrial function by activating the AMPK pathway in the cuprizone-induced demyelination. Mice were fed with cuprizone for 6 weeks. Animals simultaneously received metformin. After sacrificing animals, myelinations, and gliosis, changes in transcription factor and biochemical analysis were assessed. Transmission electron microscopy and luxol fast blue staining revealed that the myelinated axons within corpus callosum of cuprizone-induced demyelination animals increased after administration of metformin. Metformin also upregulated the expression of mitochondrial biogenesis genes. Furthermore, the biochemical analysis demonstrated that metformin ameliorated the oxidative stress induced by cuprizone. Immunohistochemistry analysis showed that astrogliosis and microgliosis were decreased after metformin administration while it enhanced the number of oligodendrocytes. Our data implicated that metformin exerts its therapeutic effects on MS by AMPK signaling improved mitochondrial homeostasis and protected oligodendrocytes.


Asunto(s)
Metformina/administración & dosificación , Mitocondrias/efectos de los fármacos , Esclerosis Múltiple/tratamiento farmacológico , Proteínas Quinasas/genética , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Axones/efectos de los fármacos , Axones/patología , Cuprizona/toxicidad , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/tratamiento farmacológico , Enfermedades Desmielinizantes/patología , Modelos Animales de Enfermedad , Hemostasis/efectos de los fármacos , Hemostasis/genética , Humanos , Ratones , Mitocondrias/genética , Esclerosis Múltiple/inducido químicamente , Esclerosis Múltiple/patología , Oligodendroglía/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
10.
J Cell Biochem ; 120(8): 12785-12795, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30861185

RESUMEN

Prenatal white matter injury is a serious problem due to maternal inflammation leading to postnatal disabilities. In this study, we used the periventricular leukomalacia (PVL) model as a common prenatal white matter injury by maternal administration of lipopolysaccharide (LPS). Neural stem cells (NSCs) have shown therapeutic ability in neurological disorders through a different mechanism such as immunomodulation. Here, we studied the preventive potential of NSCs following in utero transplantation into the embryonic lateral ventricle in an LPS-induced white matter injury model. Pregnant animals were divided into three groups and received phosphate buffered saline, LPS, or LPS + NSCs. The brains of offspring were obtained and evaluated by real-time polymerase chain reaction (PCR), immunohistochemy, enzyme-linked immunosorbent assay (ELISA), terminal deoxynucleotidyl transferase-mediated biotinylated-dUTP nick-end labeling (TUNEL), and caspase-3 activity assay. The LPS-induced maternal inflammation degenerated the myelin sheath in the offspring periventricular region which was associated with an increased microglial number, oligodendrocytes degeneration, proinflammatory cytokine secretion, and cell apoptosis. The transplanted NSCs homed into the brain and ameliorated the evaluated parameters. The expression of proinflammatory cytokines interleukin-1ß (IL-1ß), IL-6, and tumor necrosis factor-α (TNF-α), cell apoptosis and caspase-3 activity were inhibited by NSCs. In addition, Olig2 and myelin basic protein immunohistochemy staining showed that prenatal NSCs transplantation augmented the myelination in the periventricular white matter of offspring. In conclusion, we think that prenatal therapeutic strategies, such as in utero NSCs transplantation, may prevent prenatal white matter injury after birth.


Asunto(s)
Lesiones Encefálicas/terapia , Lipopolisacáridos/efectos adversos , Células-Madre Neurales/trasplante , Sustancia Blanca/embriología , Animales , Lesiones Encefálicas/inducido químicamente , Lesiones Encefálicas/inmunología , Caspasa 3/genética , Caspasa 3/metabolismo , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Regulación del Desarrollo de la Expresión Génica , Inyecciones Intraventriculares , Células-Madre Neurales/citología , Embarazo , Ratas , Ratas Sprague-Dawley , Sustancia Blanca/efectos de los fármacos , Sustancia Blanca/lesiones
11.
Brain Res ; 1711: 226-235, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30703369

RESUMEN

The use of neurotrophic factors is considered to be a novel therapeutic approach for restoring and/or maintaining neurological function in neurodegenerative disorders, such as multiple sclerosis (MS). Various studies have shown that conditioned medium produced by oligodendrocyte (OL-CM) contain a variety of neurotrophic factors. Here, we investigated the restorative effects of OL-CM, collected from oligodendrocytes cultured in a self-assembling peptide hydrogels scaffold (PuraMatrix), in experimental autoimmune encephalomyelitis (EAE) mouse model. Neural stem/progenitor cells, isolated from the embryonic mouse brain, were cultured and differentiated into oligodendrocyte. Cell viability and proliferation of oligodendrocytes were assessed by live/dead and MTT assays. Motor functions, myelination, cell infiltration, gliosis, and inflammatory process were assessed in EAE mice after intracranial injection of OL-CM at different concentrations. Application of OL-CM improved clinical score and neurological function in EAE mice and reduced the inflammatory cell infiltration and demyelination. Furthermore, administration of OL-CM reduced the expression of pro-inflammatory cytokines and suppressed the activation of NLRP3-inflammasome complex in EAE mice. These data suggest the potential therapeutic effect of OL-CM for MS treatment.


Asunto(s)
Técnicas de Cultivo de Célula/instrumentación , Medios de Cultivo Condicionados/farmacología , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Nanofibras , Factores de Crecimiento Nervioso/uso terapéutico , Oligodendroglía/metabolismo , Andamios del Tejido , Animales , Factor Neurotrófico Derivado del Encéfalo/análisis , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Factor Neurotrófico Ciliar/análisis , Medios de Cultivo/farmacología , Medios de Cultivo Condicionados/química , Enfermedades Desmielinizantes/prevención & control , Modelos Animales de Enfermedad , Células Madre Embrionarias/citología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Gliosis/prevención & control , Inflamasomas/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Factores de Crecimiento Nervioso/farmacología , Oligopéptidos
12.
J Chem Neuroanat ; 97: 1-8, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30703433

RESUMEN

After spinal cord injury (SCI) local inflammation is induced following secretion of interleukin-1beta (IL-1ß) and IL-18. It has been described that the secretion of IL-1ß and IL-18 is mediated by a cytoplasmic multiprotein complex, termed inflammasome. Mesenchymal stem cells (MSCs) have been extensively used for treating inflammatory diseases in which they showed immunomodulation characteristics. We utilized the anti-inflammatory potential of Wharton's jelly mesenchymal stem cells (WJ-MSCs) to target inflammasome complex in rat SCI model. Real time-polymerase chain reaction, western blotting, and ELISA assay were done one week after SCI to measure the expression of the inflammasome components including NLRP1, ASC, and active caspase-1 as well as IL-1ß, IL-18, and tumor necrosis factor-α (TNF-α). The histologic alteration and hind-limb locomotion were evaluated three weeks after injury by nissl staining and Basso, Beattie, Bresnahan (BBB), respectively. Our results showed that WJ-MSCs transplantation significantly decreased the SCI-induced expression of the evaluated factors in both mRNA and protein levels. In addition, WJ-MSCs significantly increased the number of normal-appearance neurons in the ventral horn of spinal cord. Noteworthy, these effects resulted in a significant improvement of motor function recovery. We conclude that inflammasome inhibition may be one of the mechanisms for the anti-inflammatory effect of MSCs in the SCI.


Asunto(s)
Inflamasomas/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Proteínas del Tejido Nervioso/metabolismo , Traumatismos de la Médula Espinal/fisiopatología , Animales , Modelos Animales de Enfermedad , Inyecciones Espinales , Masculino , Células Madre Mesenquimatosas , Ratas , Ratas Wistar , Recuperación de la Función , Traumatismos de la Médula Espinal/metabolismo
13.
J Mol Histol ; 50(2): 129-140, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30671880

RESUMEN

Bone marrow-derived mesenchymal stem cells (BMMSCs) transplantation has shown to be effective in treating chronic kidney disease. However, the effectiveness of this strategy is constrained by low homing and survival rate of transplanted cells in the injured organs. Therefore, developing strategies to improve homing and cell survival rate and therapeutic potential in cell-based therapies seems necessary. The purpose of this study is to evaluate the effect of pretreating BMMSCs with melatonin (MT) on the prosurvival and renoprotective of transplanted cells into the irreversible model of unilateral ureteral obstruction. Adult male Sprague-Dawley rats were randomized into four groups: Sham, UUO, UUO + BMMSCs, and UUO + BMMSCs + MT. The results of our study demonstrated that preconditioning with MT enhanced homing of BMMSCs into the injured kidney. MT reduced the number of TUNEL positive transplanted cells in the UUO + BMMSCs + MT group. The UUO + BMMSCs + MT group showed lower expressions of TGF-ß1, α-SMA and TNF-α at both gene and protein levels but higher expression of E-cadherin compared with the UUO + BMMSCs group. In addition, MT preconditioned BMMSCs ameliorated basement membrane disruption and histological status of injured renal tubules and also reduced fibrosis in damaged kidneys. In conclusion, our results show that stem cells pretreated by MT may represent a feasible approach for improving the beneficial effects of stem cell therapy and significantly enhance their survival after transplantation to the injured kidney.


Asunto(s)
Melatonina/farmacología , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/efectos de los fármacos , Premedicación/métodos , Insuficiencia Renal Crónica/terapia , Animales , Supervivencia de Injerto/efectos de los fármacos , Riñón/fisiología , Masculino , Ratas , Ratas Sprague-Dawley , Regeneración/efectos de los fármacos
14.
J Mol Histol ; 49(5): 471-479, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30143908

RESUMEN

Glucocorticoids (GC) are known as inflammatory drugs, which are used in neuroinflammatory diseases. Unlike the classic picture, recent studies have revealed that some GC drugs exacerbate inflammatory responses in their acute and prolonged administration. Multiple sclerosis (MS) is a demyelinating inflammatory disorder, in which reactive M1 microglia phenotype play a central role. Since methylprednisolone (MP), as a synthetic GC, are commonly used by MS patients, in this study, we evaluated the effect of long-term administration of MP on microglia polarization in cuprizone (CPZ)-induced MS model. The immunostaining results showed that chronic exposure to MP in the CPZ treated mice increased the number of Iba-1 positive microglia, which significantly expressed IP10 as M1 marker than arginase as M2 marker. MP treatment induced significant amplification in the transcript levels of iNOS and TNF-α (M1-related markers) in the corpus callosum of the MS mice, whereas no change detected in the expression of IL-10 (M2-related marker) between the groups. In addition, evaluation of myelin by luxol fast blue staining and transmission electron microscopy revealed that prolonged MP administration increased demyelination in comparison to the CPZ group. In conclusion, our results show that chronic MP therapy in the CPZ-induced demyelination model of MS polarized microglia to M1 pro-inflammatory phenotype.


Asunto(s)
Enfermedades Desmielinizantes/inducido químicamente , Metilprednisolona/efectos adversos , Microglía/metabolismo , Animales , Quimiocina CXCL10/análisis , Quimiocina CXCL10/metabolismo , Cuprizona/efectos adversos , Inflamación , Metilprednisolona/uso terapéutico , Ratones , Microglía/efectos de los fármacos , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/patología
15.
J Chem Neuroanat ; 94: 54-62, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29959975

RESUMEN

OBJECTIVE: Prenatal brain injury results from undesirable circumstances during the embryonic development. Current endeavors for treating this complication are basically excluded to postnatal therapeutic approaches. Neural stem cell therapy has shown great promise for treating neurodevelopmental disorders. To our knowledge, this is the first study that investigates the therapeutic effect of in utero transplantation of neural stem cells (NSCs) in inflammation model of prenatal brain injury. METHODS: To induce prenatal injury, time-mated C57BL6J mice were intraperitoneally injected with 50 µg/kg lipopolysaccharide (LPS(on the day 15 of gestation. In the treatment group, NSCs were transplanted into the lateral ventricle of embryos on day 17 of gestation. The expression of GFAP, Iba-1, Olig2, and NeuN were assessed by real time PCR and immunohistochemistry. Changes in IL-6, TNF-α and IL-10 cytokines level, and caspase 3 activity were evaluated in the cortex of pups. RESULTS: Intrauterine transplanted NSCs homed to the brain cortex of offspring. Brain levels of pro-inflammatory cytokines showed a significant downward trend in the NSCs group. Furthermore, NSCs ameliorated inflammation-induced reactive microgliosis and astrogliosis as well as cellular degeneration. Apoptosis inhibition in the treated group was demonstrated by the decline in the caspase 3 activity and dark neurons. CONCLUSION: This study suggests a promising prospect to initiate the treatment of prenatal brain injury before birth by intrauterine transplantation of NSCs.


Asunto(s)
Lesiones Encefálicas/terapia , Encéfalo/patología , Citocinas/metabolismo , Encefalitis/terapia , Células-Madre Neurales/trasplante , Trasplante de Células Madre/métodos , Animales , Encéfalo/metabolismo , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al ADN , Encefalitis/metabolismo , Encefalitis/patología , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Cooperación Internacional , Ratones , Proteínas de Microfilamentos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Factor de Transcripción 2 de los Oligodendrocitos/metabolismo , Embarazo
16.
Mol Neurobiol ; 55(12): 9122-9138, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29651746

RESUMEN

Traumatic brain injury (TBI) is a disruption in the brain functions following a head trauma. Cell therapy may provide a promising treatment for TBI. Among different cell types, human neural stem cells cultured in self-assembling peptide scaffolds have been suggested as a potential novel method for cell replacement treatment after TBI. In the present study, we accessed the effects of human neural stem/progenitor cells (hNS/PCs) derived from epileptic human brain and human adipose-derived stromal/stem cells (hADSCs) seeded in PuraMatrix hydrogel (PM) on brain function after TBI in an animal model of brain injury. hNS/PCs were isolated from patients with medically intractable epilepsy undergone epilepsy surgery. hNS/PCs and hADSCs have the potential for proliferation and differentiation into both neuronal and glial lineages. Assessment of the growth characteristics of hNS/PCs and hADSCs revealed that the hNS/PCs doubling time was significantly longer and the growth rate was lower than hADSCs. Transplantation of hNS/PCs and hADSCs seeded in PM improved functional recovery, decreased lesion volume, inhibited neuroinflammation, and reduced the reactive gliosis at the injury site. The data suggest the transplantation of hNS/PCs or hADSCs cultured in PM as a promising treatment option for cell replacement therapy in TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/terapia , Encéfalo/patología , Epilepsia/patología , Nanopartículas/química , Células-Madre Neurales/trasplante , Péptidos/química , Andamios del Tejido/química , Tejido Adiposo/citología , Adulto , Animales , Biomarcadores/metabolismo , Encéfalo/fisiopatología , Lesiones Traumáticas del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/fisiopatología , Proliferación Celular , Separación Celular , Supervivencia Celular , Células Cultivadas , Fenómenos Electrofisiológicos , Femenino , Gliosis/patología , Gliosis/fisiopatología , Humanos , Masculino , Microglía/metabolismo , Microglía/patología , Ratas , Células del Estroma/citología
17.
Gastroenterol Hepatol Bed Bench ; 8(3): 188-99, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26328041

RESUMEN

AIM: The present study investigated the differentiation potential of human Umbilical Cord Mesenchymal Stem Cells (UCMSCs) into hepatic lineage through embryonic body-like aggregate formation in the presence of IGF-1. BACKGROUND: Cells derived from Wharton's jelly have been reported to display a wide multilineage differentiation potential, showing some similarities to both embryonic (ESC) and mesenchymal stem cells (MSCs). PATIENTS AND METHODS: Human MSCs isolated from the umbilical cord were plated in 20 µL micro drops. A two-step differentiation protocol was used and the cell aggregates were exposed to the media supplemented with IGF, HGF, oncostatin M, and dexamethasone for 21 days. Immunoperoxidase and immuno-fluorescence were performed for cyrokeratins 18, 19 and albumin. Functional assays were done by periodic acid Schiff (PAS) and indocyanine green. RESULTS: The expression of cytokeratin 19 was shown to be higher in the cells derived from 3D spheroids compared to those cultured in conventional protocol. They showed a polygonal shape after being exposed to hepatogenic media. Immunostaining demonstrated the expression of cytokeratin-18, 19 and albumin by the differentiated cells. Besides, PAS staining revealed glycogen storage in differentiated cells. Also, a greater number of large size differentiated cells were found at the periphery of the expanded cell aggregates. CONCLUSION: We established a protocol for UCMSC differentiation into hepatocytes and these cells were morphologically and functionally similar to hepatocytes. Thus, hepatocyte differentiation may be facilitated by the UCMSCs aggregate formation before administration of the differentiation protocols.

18.
Iran J Med Sci ; 40(2): 143-51, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25821294

RESUMEN

BACKGROUND: Wharton's jelly is an unlimited source of stem cells that can be used in cell therapy and tissue engineering without any ethical concern. It has been revealed the cell-free extract could be effective to induce cell differentiation. The objective of this study was to induce Wharton's jelly-derived mesenchymal stem cells (MSCs) into hepatocyte-like cells by premeabilization of the cells in the presence of HepG2 cell line extract. METHODS: MSCs were isolated from the umbilical cord, CD marker profile and their differentiation potential into adipogenic and osteogenic lineages were determined. The cells were then, permeabilized by streptolysin O in the presence of HepG cell extract. The treated cells were cultured for 17 days. The cell phenotype was evaluated and the hepatocyte specific markers were detected by immunofluorescence and immunocytochemistry. The Periodic Acid Schiff (PAS) reaction and the cellular uptake of indocyanine green were performed to evaluate the functional behavior of the differentiated cells. RESULTS: The phenotype of extract-treated MSCs changed into a round or polygonal cells with few short processes and they could express high level of albumin, cytokeratin 18 and 19. The MSCs also could store glycogen and uptake and release indocyanine green. CONCLUSION: We demonstrated for the first time that Wharton's jelly-derived MSCs could differentiate into hepatocyte-like cells by premeabilization of them in the presence of HepG2 cell extract. This study suggests a feasible method to differentiate MSCs into functional hepatocyte-like cells.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...