Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
1.
Mol Cell Neurosci ; 118: 103694, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34954382

RESUMEN

Rupture and stretching of spinal roots are common incidents that take place in high-energy accidents. The proximal axotomy of motoneurons by crushing of ventral roots is directly related to the degeneration of half of the lesioned population within the first two weeks. Moreover, only a small percentage of surviving motoneurons can successfully achieve regeneration after such a proximal lesion, and new treatments are necessary to improve this scenario. In this sense, mesenchymal stem cells (MSC) are of great interest once they secrete a broad spectrum of bioactive molecules that are immunomodulatory and can restore the environment after a lesion. The present work aimed at studying the effects of human mesenchymal stem cells (hMSC) therapy after ventral root crush (VRC) in mice. We evaluated motoneuron survival, glial reaction, and synapse preservation at the ventral horn. For this purpose, C57BL/6 J were submitted to a crush procedure of L4 to L6 ventral roots and treated with a single intravenous injection of adipose-derived hMSC. Evaluation of the results was carried out at 7, 14, and 28 days after injury. Analysis of motoneuron survival and astrogliosis showed that hMSC treatment resulted in higher motoneuron preservation (motoneuron survival ipsi/contralateral ratio: VRC group = 53%, VRC + hMSC group = 66%; p < 0.01), combined with reduction of astrogliosis (ipsi/contralateral GFAP immunolabeling: VRC group = 470%, VRC + hMSC group = 250%; p < 0.001). The morphological classification and Sholl analysis of microglial activation revealed that hMSC treatment reduced type V and increased type II profiles, indicating an enhancement of surveying over activated microglial cells. The glial reactivity modulation directly influenced synaptic inputs in apposition to axotomized motoneurons. In the hMSC-treated group, synaptic maintenance was increased (ipsi/contralateral synaptophysin immunolabeling: VRC group = 53%, VRC + hMSC group = 64%; p < 0.05). Overall, the present data show that intravenous injection of hMSC has neuroprotective and anti-inflammatory effects, decreasing reactive astrogliosis, and microglial reaction. Also, such cell therapy results in motoneuron preservation, combined with significant maintenance of spinal cord circuits, in particular those related to the ventral horn.


Asunto(s)
Gliosis , Células Madre Mesenquimatosas , Animales , Gliosis/terapia , Humanos , Ratones , Ratones Endogámicos C57BL , Neuroprotección , Médula Espinal , Raíces Nerviosas Espinales/lesiones , Raíces Nerviosas Espinales/patología
2.
J Neuroinflammation ; 19(1): 139, 2022 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-35690757

RESUMEN

BACKGROUND: Therapeutic hypothermia significantly improves outcomes after moderate-severe hypoxic-ischemic encephalopathy (HIE), but it is partially effective. Although hypothermia is consistently associated with reduced microgliosis, it is still unclear whether it normalizes microglial morphology and phenotype. METHODS: Near-term fetal sheep (n = 24) were randomized to sham control, ischemia-normothermia, or ischemia-hypothermia. Brain sections were immunohistochemically labeled to assess neurons, microglia and their interactions with neurons, astrocytes, myelination, and gitter cells (microglia with cytoplasmic lipid granules) 7 days after cerebral ischemia. Lesions were defined as areas with complete loss of cells. RNAscope® was used to assess microglial phenotype markers CD86 and CD206. RESULTS: Ischemia-normothermia was associated with severe loss of neurons and myelin (p < 0.05), with extensive lesions, astrogliosis and microgliosis with a high proportion of gitter cells (p < 0.05). Microglial wrapping of neurons was present in both the ischemia groups. Hypothermia improved neuronal survival, suppressed lesions, gitter cells and gliosis (p < 0.05), and attenuated the reduction of myelin area fraction. The "M1" marker CD86 and "M2" marker CD206 were upregulated after ischemia. Hypothermia partially suppressed CD86 in the cortex only (p < 0.05), but did not affect CD206. CONCLUSIONS: Hypothermia prevented lesions after cerebral ischemia, but only partially suppressed microglial wrapping and M1 marker expression. These data support the hypothesis that persistent upregulation of injurious microglial activity may contribute to partial neuroprotection after hypothermia, and that immunomodulation after rewarming may be an important therapeutic target.


Asunto(s)
Hipotermia Inducida , Hipotermia , Hipoxia-Isquemia Encefálica , Sustancia Blanca , Animales , Gliosis/terapia , Hipoxia-Isquemia Encefálica/metabolismo , Inflamación/terapia , Isquemia , Ovinos , Sustancia Blanca/patología
3.
Metab Brain Dis ; 37(7): 2315-2329, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35778625

RESUMEN

Therapeutic hypothermia (TH) is the standard treatment for neonatal hypoxia-ischemia (HI) with a time window limited up to 6 h post injury. However, influence of sexual dimorphism in the therapeutic window for TH has not yet been elucidated in animal models of HI. Therefore, the aim of this study was to investigate the most effective time window to start TH in male and female rats submitted to neonatal HI. Wistar rats (P7) were divided into the following groups: NAÏVE and SHAM (control groups), HI (submitted to HI) and TH (submitted to HI and TH; 32ºC for 5 h). TH was started at 2 h (TH-2 h group), 4 h (TH-4 h group), or 6 h (TH-6 h group) after HI. At P14, animals were subjected to behavioural tests, volume of lesion and reactive astrogliosis assessments. Male and female rats from the TH-2 h group showed reduction in the latency of behavioral tests, and decrease in volume of lesion and intensity of GFAP immunofluorescence. TH-2 h females also showed reduction of degenerative cells and morphological changes in astrocytes. Interestingly, females from the TH-6 h group showed an increase in volume of lesion and in number of degenerative hippocampal cells, associated with worse behavioral performance. Together, these results indicate that TH neuroprotection is time- and sex-dependent. Moreover, TH started later (6 h) can worsen volume of brain lesion in females. These data indicate the need to develop specific therapeutic protocols for each sex and reinforce the importance of early onset of the hypothermic treatment.


Asunto(s)
Hipotermia Inducida , Hipoxia-Isquemia Encefálica , Animales , Masculino , Femenino , Ratas , Hipoxia-Isquemia Encefálica/terapia , Hipoxia-Isquemia Encefálica/patología , Gliosis/terapia , Gliosis/patología , Ratas Wistar , Animales Recién Nacidos , Encéfalo , Isquemia/patología , Isquemia/terapia , Modelos Animales de Enfermedad
4.
Cerebellum ; 20(3): 420-429, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33394333

RESUMEN

Spinocerebellar ataxia type 1 (SCA1) is a fatal neurodegenerative disease caused by an abnormal expansion of CAG repeats in the Ataxin1 (ATXN1) gene. SCA1 is characterized by motor deficits, cerebellar neurodegeneration, and gliosis and gene expression changes. Expression of brain-derived neurotrophic factor (BDNF), growth factor important for the survival and function of cerebellar neurons, is decreased in ATXN1[82Q] mice, the Purkinje neuron specific transgenic mouse model of SCA1. As this decrease in BDNF expression may contribute to cerebellar neurodegeneration, we tested whether delivery of extrinsic human BDNF via osmotic ALZET pumps has a beneficial effect on disease severity in this mouse model of SCA1. Additionally, to test the effects of BDNF on established and progressing cerebellar pathogenesis and motor deficits, we delivered BDNF post-symptomatically. We have found that post-symptomatic delivery of extrinsic BDNF ameliorated motor deficits and cerebellar pathology (i.e., dendritic atrophy of Purkinje cells, and astrogliosis) indicating therapeutic potential of BDNF even after the onset of symptoms in SCA1. However, BDNF did not alter Purkinje cell gene expression changes indicating that certain aspects of disease pathogenesis cannot be ameliorated/slowed down with BDNF and that combinational therapies may be needed.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/uso terapéutico , Ataxias Espinocerebelosas/terapia , Animales , Cerebelo/patología , Dendritas/patología , Femenino , Regulación de la Expresión Génica , Técnicas de Transferencia de Gen , Terapia Genética , Gliosis/patología , Gliosis/terapia , Humanos , Masculino , Ratones , Ratones Transgénicos , Células de Purkinje/patología , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/patología
5.
Int J Neurosci ; 131(6): 571-579, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32241216

RESUMEN

PURPOSE: Clinical research suggests that transcranial direct current stimulation (tDCS) at bilateral supraorbital foramen and inferior orbital rim and nose intersections may facilitate rehabilitation after stroke. However, the underlying neurobiological mechanisms of tDCS remain poorly understood, impeding its clinical application. Here, we investigated the effect of tDCS applied after stroke on neural cells. MATERIALS AND METHODS: Middle cerebral arterial occlusion (MCAO) reperfusion was induced in rats. Animals with comparable infarcts were randomly divided into MCAO group and MCAO + tDCS group. Recovery of neurological function was assessed behaviorally by modified neurological severity score (mNSS). Ischemic tissue damage verified histologically by TTC and HE staining. Immunohistochemical staining, real-time qPCR, and western blot were applied to determine the changes of neural cells in ischemic brains. RESULTS: The results reveal that tDCS treated by multilead brain reflex instrument can promote the recovery of neurological function, remarkably reduce cerebral infarct volume, promote brain tissue rehabilitation, and can effectively inhibit astrocytosis and enhance neuronal survival and synaptic function in ischemic brains. CONCULSIONS: Our study suggests that tDCS treated by multilead brain reflex instrument could be prospectively developed into a clinical treatment modality.


Asunto(s)
Gliosis/terapia , Infarto de la Arteria Cerebral Media/rehabilitación , Accidente Cerebrovascular Isquémico/rehabilitación , Neuronas , Recuperación de la Función , Rehabilitación de Accidente Cerebrovascular , Estimulación Transcraneal de Corriente Directa , Animales , Supervivencia Celular/fisiología , Modelos Animales de Enfermedad , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/fisiopatología , Accidente Cerebrovascular Isquémico/etiología , Accidente Cerebrovascular Isquémico/patología , Accidente Cerebrovascular Isquémico/fisiopatología , Masculino , Neuronas/metabolismo , Neuronas/patología , Ratas , Ratas Sprague-Dawley , Recuperación de la Función/fisiología , Índice de Severidad de la Enfermedad
6.
Crit Care Med ; 48(4): e290-e298, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32205619

RESUMEN

OBJECTIVES: Survivors of sepsis are frequently left with significant cognitive and behavioral impairments. These complications derive from nonresolving inflammation that persists following hospital discharge. To date, no study has investigated the effects of mesenchymal stromal cell therapy on the blood-brain barrier, astrocyte activation, neuroinflammation, and cognitive and behavioral alterations in experimental sepsis. DESIGN: Prospective, randomized, controlled experimental study. SETTING: Government-affiliated research laboratory. SUBJECTS: Male Swiss Webster mice (n = 309). INTERVENTIONS: Sepsis was induced by cecal ligation and puncture; sham-operated animals were used as control. All animals received volume resuscitation (1 mL saline/mouse subcutaneously) and antibiotics (meropenem 10 mg/kg intraperitoneally at 6, 24, and 48 hours). Six hours after surgery, mice were treated with mesenchymal stromal cells IV (1 × 10 cells in 0.05 mL of saline/mouse) or saline (0.05 mL IV). MEASUREMENTS AND MAIN RESULTS: At day 1, clinical score and plasma levels of inflammatory mediators were increased in cecal ligation and puncture mice. Mesenchymal stromal cells did not alter clinical score or survival rate, but reduced levels of systemic interleukin-1ß, interleukin-6, and monocyte chemoattractant protein-1. At day 15, survivor mice completed a battery of cognitive and behavioral tasks. Cecal ligation and puncture mice exhibited spatial and aversive memory deficits and anxiety-like behavior. These effects may be related to increased blood-brain barrier permeability, with altered tight-junction messenger RNA expression, increased brain levels of inflammatory mediators, and astrogliosis (induced at day 3). Mesenchymal stromal cells mitigated these cognitive and behavioral alterations, as well as reduced blood-brain barrier dysfunction, astrocyte activation, and interleukin-1ß, interleukin-6, tumor necrosis factor-α, and interleukin-10 levels in vivo. In cultured primary astrocytes stimulated with lipopolysaccharide, conditioned media from mesenchymal stromal cells reduced astrogliosis, interleukin-1ß, and monocyte chemoattractant protein-1, suggesting a paracrine mechanism of action. CONCLUSIONS: In mice who survived experimental sepsis, mesenchymal stromal cell therapy protected blood-brain barrier integrity, reduced astrogliosis and neuroinflammation, as well as improved cognition and behavior.


Asunto(s)
Barrera Hematoencefálica , Trastornos del Conocimiento , Gliosis , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Sepsis , Animales , Masculino , Ratones , Conducta Animal , Barrera Hematoencefálica/metabolismo , Trastornos del Conocimiento/prevención & control , Modelos Animales de Enfermedad , Gliosis/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Estudios Prospectivos , Sepsis/terapia
7.
Mol Ther ; 27(7): 1313-1326, 2019 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-31043342

RESUMEN

Mutations within over 250 known genes are associated with inherited retinal degeneration. Clinical success following gene-replacement therapy for congenital blindness due to RPE65 mutations establishes a platform for the development of downstream treatments targeting other forms of inherited ocular disease. Unfortunately, several challenges relevant to complex disease pathology and limitations of current gene-transfer technologies impede the development of related strategies for each specific form of inherited retinal degeneration. Here, we describe a gene-augmentation strategy that delays retinal degeneration by stimulating features of anabolic metabolism necessary for survival and structural maintenance of photoreceptors. We targeted two critical points of regulation in the canonical insulin/AKT/mammalian target of rapamycin (mTOR) pathway with AAV-mediated gene augmentation in a mouse model of retinitis pigmentosa. AAV vectors expressing the serine/threonine kinase, AKT3, promote dramatic preservation of photoreceptor numbers, structure, and partial visual function. This protective effect was associated with successful reprogramming of photoreceptor metabolism toward pathways associated with cell growth and survival. Collectively, these findings underscore the importance of AKT activity and downstream pathways associated with anabolic metabolism in photoreceptor survival and maintenance.


Asunto(s)
Terapia Genética/métodos , Neuroprotección/genética , Células Fotorreceptoras de Vertebrados/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Retinitis Pigmentosa/terapia , Transducción de Señal/genética , Transducción Genética , Animales , Supervivencia Celular/genética , Células Cultivadas , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/genética , Modelos Animales de Enfermedad , Vectores Genéticos , Gliosis/genética , Gliosis/terapia , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Mutación Puntual , Degeneración Retiniana/terapia , Retinitis Pigmentosa/genética , Serina-Treonina Quinasas TOR/metabolismo , Agudeza Visual/genética
8.
Int J Mol Sci ; 21(24)2020 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-33339379

RESUMEN

A systemic inflammatory response induces multiple organ dysfunction and results in poor long-term neurological outcomes in neonatal sepsis. However, there is no effective therapy for treating or preventing neonatal sepsis besides antibiotics and supportive care. Therefore, a novel strategy to improve neonatal sepsis-related morbidity and mortality is desirable. Recently, we reported that prophylactic therapy with human amniotic stem cells (hAFSCs) improved survival in a rat model of lipopolysaccharide (LPS)-induced neonatal sepsis through immunomodulation. Besides improving the mortality, increasing survival without major morbidities is an important goal of neonatal intensive care for neonatal sepsis. This study investigated long-term neurological outcomes in neonatal sepsis survivors treated with hAFSCs using the LPS-induced neonatal sepsis model in rats. We found that prophylactic therapy with hAFSCs improved spatial awareness and memory-based behavior in neonatal sepsis survivors at adolescence in rats. The treatment suppressed acute reactive gliosis and subsequently reduced astrogliosis in the hippocampal region over a long period of assessment. To the best of our knowledge, this is the first report that proves the concept that hAFSC treatment improves cognitive impairment in neonatal sepsis survivors. We demonstrate the efficacy of hAFSC therapy in improving the mortality and morbidity associated with neonatal sepsis.


Asunto(s)
Disfunción Cognitiva/prevención & control , Gliosis/prevención & control , Trasplante de Células Madre Mesenquimatosas/métodos , Sepsis Neonatal/complicaciones , Líquido Amniótico/citología , Animales , Células Cultivadas , Disfunción Cognitiva/etiología , Disfunción Cognitiva/terapia , Gliosis/etiología , Gliosis/terapia , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratas , Ratas Sprague-Dawley
9.
Brain ; 141(1): 99-116, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29186350

RESUMEN

Mucopolysaccharidosis IIIB is a paediatric lysosomal storage disease caused by deficiency of the enzyme α-N-acetylglucosaminidase (NAGLU), involved in the degradation of the glycosaminoglycan heparan sulphate. Absence of NAGLU leads to accumulation of partially degraded heparan sulphate within lysosomes and the extracellular matrix, giving rise to severe CNS degeneration with progressive cognitive impairment and behavioural problems. There are no therapies. Haematopoietic stem cell transplant shows great efficacy in the related disease mucopolysaccharidosis I, where donor-derived monocytes can transmigrate into the brain following bone marrow engraftment, secrete the missing enzyme and cross-correct neighbouring cells. However, little neurological correction is achieved in patients with mucopolysaccharidosis IIIB. We have therefore developed an ex vivo haematopoietic stem cell gene therapy approach in a mouse model of mucopolysaccharidosis IIIB, using a high-titre lentiviral vector and the myeloid-specific CD11b promoter, driving the expression of NAGLU (LV.NAGLU). To understand the mechanism of correction we also compared this with a poorly secreted version of NAGLU containing a C-terminal fusion to IGFII (LV.NAGLU-IGFII). Mucopolysaccharidosis IIIB haematopoietic stem cells were transduced with vector, transplanted into myeloablated mucopolysaccharidosis IIIB mice and compared at 8 months of age with mice receiving a wild-type transplant. As the disease is characterized by increased inflammation, we also tested the anti-inflammatory steroidal agent prednisolone alone, or in combination with LV.NAGLU, to understand the importance of inflammation on behaviour. NAGLU enzyme was substantially increased in the brain of LV.NAGLU and LV.NAGLU-IGFII-treated mice, with little expression in wild-type bone marrow transplanted mice. LV.NAGLU treatment led to behavioural correction, normalization of heparan sulphate and sulphation patterning, reduced inflammatory cytokine expression and correction of astrocytosis, microgliosis and lysosomal compartment size throughout the brain. The addition of prednisolone improved inflammatory aspects further. Substantial correction of lysosomal storage in neurons and astrocytes was also achieved in LV.NAGLU-IGFII-treated mice, despite limited enzyme secretion from engrafted macrophages in the brain. Interestingly both wild-type bone marrow transplant and prednisolone treatment alone corrected behaviour, despite having little effect on brain neuropathology. This was attributed to a decrease in peripheral inflammatory cytokines. Here we show significant neurological disease correction is achieved using haematopoietic stem cell gene therapy, suggesting this therapy alone or in combination with anti-inflammatories may improve neurological function in patients.


Asunto(s)
Encefalitis/etiología , Encefalitis/terapia , Terapia Genética/métodos , Macrófagos/enzimología , Mucopolisacaridosis III , Células Madre/fisiología , Animales , Encéfalo/enzimología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Gliosis/terapia , Glicosaminoglicanos/genética , Glicosaminoglicanos/metabolismo , Humanos , Hígado/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mucopolisacaridosis III/complicaciones , Mucopolisacaridosis III/genética , Mucopolisacaridosis III/patología , Mucopolisacaridosis III/terapia , Prednisolona/uso terapéutico , Bazo/enzimología , Sulfatasas/genética , Sulfatasas/metabolismo
10.
Glia ; 66(1): 191-205, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29024008

RESUMEN

Neuroinflammation mediated by chronically activated microglia, largely caused by abnormal accumulation of misfolded α-synuclein (αSyn) protein, is known to contribute to the pathophysiology of Parkinson's disease (PD). In this work, based on the immunomodulatory activities displayed by particular heat-shock proteins (HSPs), we tested a novel vaccination strategy that used a combination of αSyn and Grp94 (HSPC4 or Gp96) chaperone and a murine PD model. We used two different procedures, first, the adoptive transfer of splenocytes from αSyn/Grp94-immunized mice to recipient animals, and second, direct immunization with αSyn/Grp94, to study the effects in a chronic mouse MPTP-model of parkinsonism. We found that both approaches promoted a distinct profile in the peripheral system-supported by humoral and cellular immunity-consisting of a Th1-shifted αSyn-specific response accompanied by an immune-regulatory/Th2-skewed general phenotype. Remarkably, this mixed profile sustained by αSyn/Grp94 immunization led to strong suppression of microglial activation in the substantia nigra and striatum, pointing to a newly described positive effect of anti-αSyn Th1-responses in the context of PD. This strategy is the first to target αSyn and report the suppression of PD-associated microgliosis. Overall, we show that the αSyn/Grp94 combination supports a distinct and long-lasting immune profile in the peripheral system, which has an impact at the CNS level by suppressing chronic microglial activation in an MPTP model of PD. Furthermore, our study demonstrates that reshaping peripheral immunity by vaccination with appropriate misfolding protein/HSP combinations could be highly beneficial as a treatment for neurodegenerative misfolding diseases.


Asunto(s)
Gliosis/etiología , Gliosis/terapia , Inmunización/métodos , Intoxicación por MPTP , Glicoproteínas de Membrana/inmunología , alfa-Sinucleína/inmunología , Traslado Adoptivo , Análisis de Varianza , Animales , Antígenos CD4/metabolismo , Enfermedad Crónica , Citocinas/metabolismo , Modelos Animales de Enfermedad , Intoxicación por MPTP/inducido químicamente , Intoxicación por MPTP/complicaciones , Intoxicación por MPTP/inmunología , Intoxicación por MPTP/terapia , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/patología , Sustancia Negra/metabolismo , Sustancia Negra/patología , Linfocitos T Reguladores/metabolismo
11.
Exp Eye Res ; 169: 1-12, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29355737

RESUMEN

Photobiomodulation (PBM) with 670 nm light has been shown to accelerate wound healing in soft tissue injuries, and also to protect neuronal tissues. However, little data exist on its effects on the non-neuronal components of the retina, such as Müller cells (MCs), which are the principal macroglia of the retina that play a role in maintaining retinal homeostasis. The aim of this study was to explore the effects of 670 nm light on activated MCs using in vivo and in vitro stress models. Adult Sprague-Dawley rats were exposed to photo-oxidative damage (PD) for 24 h and treated with 670 nm light at 0, 3 and 14 days after PD. Tissue was collected at 30 days post-PD for analysis. Using the in vitro scratch model with a human MC line (MIO-M1), area coverage and cellular stress were analysed following treatment with 670 nm light. We showed that early treatment with 670 nm light after PD reduced MC activation, lowering the retinal expression of GFAP and FGF-2. 670 nm light treatment mitigated the production of MC-related pro-inflammatory cytokines (including IL-1ß), and reduced microglia/macrophage (MG/MΦ) recruitment into the outer retina following PD. This subsequently decreased photoreceptor loss, slowing the progression of retinal degeneration. In vitro, we showed that 670 nm light directly modulated MC activation, reducing rates of area coverage by suppressing cellular proliferation and spreading. This study indicates that 670 nm light treatment post-injury may have therapeutic benefit when administered shortly after retinal damage, and could be useful for retinal degenerations where MC gliosis is a feature of disease progression.


Asunto(s)
Células Ependimogliales/efectos de la radiación , Gliosis/terapia , Fototerapia/métodos , Traumatismos Experimentales por Radiación/terapia , Traumatismos por Radiación/terapia , Retina/efectos de la radiación , Degeneración Retiniana/terapia , Animales , Línea Celular , Movimiento Celular , Supervivencia Celular , Citocinas/metabolismo , Modelos Animales de Enfermedad , Células Ependimogliales/metabolismo , Células Ependimogliales/patología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente Indirecta , Proteína Ácida Fibrilar de la Glía/metabolismo , Gliosis/etiología , Gliosis/metabolismo , Gliosis/patología , Humanos , Luz/efectos adversos , Estrés Oxidativo , Traumatismos por Radiación/etiología , Traumatismos por Radiación/metabolismo , Traumatismos por Radiación/patología , Traumatismos Experimentales por Radiación/etiología , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/patología , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Retina/metabolismo , Retina/patología , Degeneración Retiniana/etiología , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología
12.
Anesthesiology ; 129(2): 343-366, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29462012

RESUMEN

Chronic pain is maintained in part by central sensitization, a phenomenon of synaptic plasticity, and increased neuronal responsiveness in central pain pathways after painful insults. Accumulating evidence suggests that central sensitization is also driven by neuroinflammation in the peripheral and central nervous system. A characteristic feature of neuroinflammation is the activation of glial cells, such as microglia and astrocytes, in the spinal cord and brain, leading to the release of proinflammatory cytokines and chemokines. Recent studies suggest that central cytokines and chemokines are powerful neuromodulators and play a sufficient role in inducing hyperalgesia and allodynia after central nervous system administration. Sustained increase of cytokines and chemokines in the central nervous system also promotes chronic widespread pain that affects multiple body sites. Thus, neuroinflammation drives widespread chronic pain via central sensitization. We also discuss sex-dependent glial/immune signaling in chronic pain and new therapeutic approaches that control neuroinflammation for the resolution of chronic pain.


Asunto(s)
Sensibilización del Sistema Nervioso Central/fisiología , Dolor Crónico/metabolismo , Dolor Crónico/terapia , Mediadores de Inflamación/metabolismo , Manejo del Dolor/métodos , Analgésicos/farmacología , Analgésicos/uso terapéutico , Animales , Sensibilización del Sistema Nervioso Central/efectos de los fármacos , Dolor Crónico/patología , Ensayos Clínicos como Asunto/métodos , Gliosis/metabolismo , Gliosis/patología , Gliosis/terapia , Humanos , Inflamación/metabolismo , Inflamación/patología , Inflamación/terapia , Mediadores de Inflamación/antagonistas & inhibidores
13.
J Stroke Cerebrovasc Dis ; 27(9): 2453-2465, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30029838

RESUMEN

INTRODUCTION: Stroke represents an attractive target for cell therapy. Although different types of cells have been employed in animal models with variable results, the human adipose-derived stem cells (hASCs) have demonstrated favorable characteristics in the treatment of diseases with inflammatory substrate, but experience in their intracerebral administration is lacking. The purpose of this study is to evaluate the effect and safety of the intracerebral application of hASCs in a stroke model. METHODS: A first group of Athymic Nude mice after stroke received a stereotactic injection of hASCs at a concentration of 4 × 104/µL at the penumbra area, a second group without stroke received the same cell concentration, and a third group had only stroke and no cells. After 7, 15, and 30 days, the animals underwent fluorodeoxyglucose-positron emission tomography and magnetic resonance imaging; subsequently, they were sacrificed for histological evaluation (HuNu, GFAP, IBA-1, Ki67, DCX) of the penumbra area and ipsilateral subventricular zone (iSVZ). RESULTS: The in vitro studies found no alterations in the molecular karyotype, clonogenic capacity, and expression of 62 kDa transcription factor and telomerase. Animals implanted with cells showed no adverse events. The implanted cells showed no evidence of proliferation or differentiation. However, there was an increase of capillaries, less astrocytes and microglia, and increased bromodeoxyuridine and doublecortin-positive cells in the iSVZ and in the vicinity of ischemic injury. CONCLUSIONS: These results suggest that hASCs in the implanted dose modulate inflammation, promote endogenous neurogenesis, and do not proliferate or migrate in the brain. These data confirm the safety of cell therapy with hASCs.


Asunto(s)
Isquemia Encefálica/terapia , Trasplante de Células Madre , Tejido Adiposo/citología , Animales , Encéfalo/irrigación sanguínea , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Encéfalo/patología , Isquemia Encefálica/diagnóstico por imagen , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Proliferación Celular , Modelos Animales de Enfermedad , Proteína Doblecortina , Gliosis/diagnóstico por imagen , Gliosis/metabolismo , Gliosis/patología , Gliosis/terapia , Humanos , Masculino , Ratones Desnudos , Microglía/metabolismo , Microglía/patología , Actividad Motora , Neovascularización Patológica/diagnóstico por imagen , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Neovascularización Patológica/terapia , Neuronas/metabolismo , Neuronas/patología , Distribución Aleatoria , Trasplante de Células Madre/efectos adversos , Células Madre/citología , Trasplante Heterólogo
14.
J Neuroinflammation ; 13(1): 230, 2016 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-27590010

RESUMEN

BACKGROUND: Although electroconvulsive therapy (ECT) is regarded as one of the efficient treatments for intractable psychiatric disorders, the mechanism of therapeutic action remains unclear. Recently, many studies indicate that ECT affects the immune-related cells, such as microglia, astrocytes, and lymphocytes. Moreover, microglial activation and astrocytic activation have been implicated in the postmortem brains of schizophrenia patients. We previously demonstrated that Gunn rats showed schizophrenia-like behavior and microglial activation in their brains. The present study examined the effects of electroconvulsive shock (ECS), an animal counterpart of ECT, on schizophrenia-like behavior, microgliosis, and astrogliosis in the brain of Gunn rats. METHODS: The rats were divided into four groups, i.e., Wistar sham, Wistar ECS, Gunn sham, and Gunn ECS. ECS groups received ECS once daily for six consecutive days. Subsequently, prepulse inhibition (PPI) test was performed, and immunohistochemistry analysis was carried out to determine the activation degree of microglia and astrocytes in the hippocampus by using anti-CD11b and anti-glial fibrillary acidic protein (GFAP) antibody, respectively. RESULTS: We found PPI deficit in Gunn rats compared to Wistar rats, and it was significantly improved by ECS. Immunohistochemistry analysis revealed that immunoreactivity of CD11b and GFAP was significantly increased in Gunn rats compared to Wistar rats. ECS significantly attenuated the immunoreactivity of both CD11b and GFAP in Gunn rats. CONCLUSIONS: ECS ameliorated schizophrenia-like behavior of Gunn rats and attenuated microgliosis and astrogliosis in the hippocampus of Gunn rats. Accordingly, therapeutic effects of ECT may be exerted, at least in part, by inhibition of glial activation. These results may provide crucial information to elucidate the role of activated glia in the pathogenesis of schizophrenia and to determine whether future therapeutic interventions should attempt to up-regulate or down-regulate glial functions.


Asunto(s)
Electrochoque , Gliosis/terapia , Hipocampo/patología , Esquizofrenia/patología , Estimulación Acústica , Animales , Astrocitos/patología , Astrocitos/fisiología , Antígeno CD11b/metabolismo , Modelos Animales de Enfermedad , Proteína Ácida Fibrilar de la Glía/metabolismo , Gliosis/etiología , Trastornos de la Audición/genética , Masculino , Microglía/patología , Inhibición Prepulso/fisiología , Psicoacústica , Ratas , Ratas Gunn , Ratas Wistar , Esquizofrenia/complicaciones , Esquizofrenia/genética
15.
Stem Cells ; 33(10): 3006-16, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26175331

RESUMEN

A variety of diseases lead to degeneration of retinal ganglion cells (RGCs) and their axons within the optic nerve resulting in loss of visual function. Although current therapies may delay RGC loss, they do not restore visual function or completely halt disease progression. Regenerative medicine has recently focused on stem cell therapy for both neuroprotective and regenerative purposes. However, significant problems remain to be addressed, such as the long-term impact of reactive gliosis occurring in the host retina in response to transplanted stem cells. The aim of this work was to investigate retinal glial responses to intravitreally transplanted bone marrow mesenchymal stem cells (BM-MSCs) to help identify factors able to modulate graft-induced reactive gliosis. We found in vivo that intravitreal BM-MSC transplantation is associated with gliosis-mediated retinal folding, upregulation of intermediate filaments, and recruitment of macrophages. These responses were accompanied by significant JAK/STAT3 and MAPK (ERK1/2 and JNK) cascade activation in retinal Muller glia. Lipocalin-2 (Lcn-2) was identified as a potential new indicator of graft-induced reactive gliosis. Pharmacological inhibition of STAT3 in BM-MSC cocultured retinal explants successfully reduced glial fibrillary acidic protein expression in retinal Muller glia and increased BM-MSC retinal engraftment. Inhibition of stem cell-induced reactive gliosis is critical for successful transplantation-based strategies for neuroprotection, replacement, and regeneration of the optic nerve.


Asunto(s)
Gliosis/terapia , Trasplante de Células Madre Mesenquimatosas , Neuroglía/patología , Medicina Regenerativa , Animales , Axones/patología , Células de la Médula Ósea/citología , Células Ependimogliales/patología , Gliosis/patología , Humanos , Células Madre Mesenquimatosas , Ratones , Nervio Óptico/patología , Retina/crecimiento & desarrollo , Retina/patología , Células Ganglionares de la Retina/patología
16.
Exp Brain Res ; 234(11): 3225-3232, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27377070

RESUMEN

We have reported previously that intracranial application of near-infrared light (NIr) reduces clinical signs and offers neuroprotection in a subacute MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) monkey model of Parkinson's disease. In this study, we explored whether NIr reduces the gliosis in this animal model. Sections of midbrain (containing the substantia nigra pars compacta; SNc) and striatum were processed for glial fibrillary acidic protein (to label astrocytes; GFAP) and ionised calcium-binding adaptor molecule 1 (to label microglia; IBA1) immunohistochemistry. Cell counts were undertaken using stereology, and cell body sizes were measured using ImageJ. Our results showed that NIr treatment reduced dramatically (~75 %) MPTP-induced astrogliosis in both the SNc and striatum. Among microglia, however, NIr had a more limited impact in both nuclei; although there was a reduction in overall cell size, there were no changes in the number of microglia in the MPTP-treated monkeys after NIr treatment. In summary, we showed that NIr treatment influenced the glial response, particularly that of the astrocytes, in our monkey MPTP model of Parkinson's disease. Our findings raise the possibility of glial cells as a future therapeutic target using NIr.


Asunto(s)
1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Gliosis/etiología , Gliosis/terapia , Rayos Infrarrojos/uso terapéutico , Intoxicación por MPTP/complicaciones , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/farmacología , Análisis de Varianza , Animales , Proteínas de Unión al Calcio , Cuerpo Estriado/metabolismo , Cuerpo Estriado/patología , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Terapia por Luz de Baja Intensidad , Intoxicación por MPTP/patología , Macaca fascicularis , Masculino , Proteínas de Microfilamentos , Neuroglía/efectos de los fármacos , Neuroglía/efectos de la radiación , Neurotoxinas/toxicidad , Sustancia Negra/efectos de los fármacos , Sustancia Negra/metabolismo , Sustancia Negra/patología
17.
J Nanobiotechnology ; 14: 3, 2016 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-26753543

RESUMEN

BACKGROUND: Electrodes for neural stimulation and recording are used for the treatment of neurological disorders. Their features critically depend on impedance and interaction with brain tissue. The effect of surface modification on electrode impedance was examined in vitro and in vivo after intracranial implantation in rats. Electrodes coated by electrophoretic deposition with platinum nanoparticles (NP; <10 and 50 nm) as well as uncoated references were implanted into the rat's subthalamic nucleus. After postoperative recovery, rats were electrostimulated for 3 weeks. Impedance was measured before implantation, after recovery and then weekly during stimulation. Finally, local field potential was recorded and tissue-to-implant reaction was immunohistochemically studied. RESULTS: Coating with NP significantly increased electrode's impedance in vitro. Postoperatively, the impedance of all electrodes was temporarily further increased. This effect was lowest for the electrodes coated with particles <10 nm, which also showed the most stable impedance dynamics during stimulation for 3 weeks and the lowest total power of local field potential during neuronal activity recording. Histological analysis revealed that NP-coating did not affect glial reactions or neural cell-count. CONCLUSIONS: Coating with NP <10 nm may improve electrode's impedance stability without affecting biocompatibility. Increased impedance after NP-coating may improve neural recording due to better signal-to-noise ratio.


Asunto(s)
Impedancia Eléctrica/uso terapéutico , Gliosis/terapia , Nanopartículas/administración & dosificación , Nanopartículas/química , Neuronas/efectos de los fármacos , Platino (Metal)/administración & dosificación , Platino (Metal)/química , Animales , Materiales Biocompatibles/administración & dosificación , Encéfalo/efectos de los fármacos , Diseño de Equipo/métodos , Ligandos , Masculino , Microelectrodos , Ratas , Ratas Sprague-Dawley
18.
Glia ; 63(2): 194-205, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25139829

RESUMEN

Astrogliosis occurs at the lesion site within days to weeks after spinal cord injury (SCI) and involves the proliferation and hypertrophy of astrocytes, leading to glia scar formation. Changes in gene expression by deregulated microRNAs (miRNAs) are involved in the process of central nervous system neurodegeneration. Here, we report that mir-145, a miRNA enriched in rat spinal neurons and astrocytes, was downregulated at 1 week and 1 month after SCI. Our in vitro studies using astrocytes prepared from neonatal spinal cord tissues indicated that potent inflammagen lipopolysaccharide downregulated mir-145 expression in astrocytes, suggesting that SCI-triggered inflammatory signaling pathways could play the inhibitory role in astrocytic mir-145 expression. To induce overexpression of mir-145 in astrocytes at the spinal cord lesion site, we developed a lentivirus-mediated pre-miRNA delivery system using the promoter of glial fibrillary acidic protein (GFAP), an astrocyte-specific intermediate filament. The results indicated that astrocyte-specific overexpression of mir-145 reduced astrocytic cell density at the lesion border of the injured spinal cord. In parallel, overexpression of mir-145 reduced the size of astrocytes and the number of related cell processes, as well as cell proliferation and migration. Through a luciferase reporter system, we found that GFAP and c-myc were the two potential targets of mir-145 in astrocytes. Together, the findings demonstrate the novel role of mir-145 in the regulation of astrocytic dynamics, and reveal that the downregulation of mir-145 in astrocytes is a critical factor inducing astrogliosis after SCI. GLIA 2015;63:194-205.


Asunto(s)
Gliosis/etiología , Gliosis/terapia , MicroARNs/uso terapéutico , Traumatismos de la Médula Espinal/complicaciones , Animales , Animales Recién Nacidos , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Células Cultivadas , Citocinas/farmacología , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Femenino , Regulación de la Expresión Génica/fisiología , Proteína Ácida Fibrilar de la Glía/metabolismo , Sustancia Gris/metabolismo , Sustancia Gris/patología , Lipopolisacáridos/farmacología , MicroARNs/genética , MicroARNs/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/terapia
19.
Mol Ther ; 22(3): 498-510, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24394188

RESUMEN

There is emerging evidence that the misfolding of superoxide dismutase 1 (SOD1) may represent a common pathogenic event in both familial and sporadic amyotrophic lateral sclerosis (ALS). To reduce the burden of misfolded SOD1 species in the nervous system, we have tested a novel therapeutic approach based on adeno-associated virus (AAV)-mediated tonic expression of a DNA construct encoding a secretable single-chain fragment variable (scFv) antibody composed of the variable heavy and light chain regions of a monoclonal antibody (D3H5) binding specifically to misfolded SOD1. A single intrathecal injection of the AAV encoding the single-chain antibody in SOD1(G93A) mice at 45 days of age resulted in sustained expression of single-chain antibodies in the spinal cord, and it delayed disease onset and extension of life span by up to 28%, in direct correlation with scFv titers in the spinal cord. The treatment caused attenuation of neuronal stress signals and reduction in levels of misfolded SOD1 in the spinal cord of SOD1(G93A) mice. From these results, we propose that an immunotherapy based on intrathecal inoculation of AAV encoding a secretable scFv against misfolded SOD1 should be considered as potential treatment for ALS, especially for individuals carrying SOD1 mutations.


Asunto(s)
Esclerosis Amiotrófica Lateral/terapia , Dependovirus/genética , Anticuerpos de Cadena Única/inmunología , Médula Espinal/inmunología , Superóxido Dismutasa/inmunología , Esclerosis Amiotrófica Lateral/inmunología , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Terapia Genética , Vectores Genéticos/administración & dosificación , Gliosis/patología , Gliosis/terapia , Células HEK293 , Humanos , Inmunoterapia , Inyecciones Espinales , Ratones , Pliegue de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Anticuerpos de Cadena Única/farmacología , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1
20.
J Neurosci ; 33(16): 7027-37, 2013 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-23595760

RESUMEN

Active amyloid-ß (Aß) immunotherapy is under investigation to prevent or treat early Alzheimer's disease (AD). In 2002, a Phase II clinical trial (AN1792) was halted due to meningoencephalitis in ∼6% of the AD patients, possibly caused by a T-cell-mediated immunological response. Thus, generating a vaccine that safely generates high anti-Aß antibody levels in the elderly is required. In this study, MER5101, a novel conjugate of Aß1-15 peptide (a B-cell epitope fragment) conjugated to an immunogenic carrier protein, diphtheria toxoid (DT), and formulated in a nanoparticular emulsion-based adjuvant, was administered to 10-month-old APPswe/PS1ΔE9 transgenic (Tg) and wild-type (Wt) mice. High anti-Aß antibody levels were observed in both vaccinated APPswe/PS1ΔE9 Tg and Wt mice. Antibody isotypes were mainly IgG1 and IgG2b, suggesting a Th2-biased response. Restimulation of splenocytes with the Aß1-15:DT conjugate resulted in a strong proliferative response, whereas proliferation was absent after restimulation with Aß1-15 or Aß1-40/42 peptides, indicating a cellular immune response against DT while avoiding an Aß-specific T-cell response. Moreover, significant reductions in cerebral Aß plaque burden, accompanied by attenuated microglial activation and increased synaptic density, were observed in MER5101-vaccinated APPswe/PS1ΔE9 Tg mice compared with Tg adjuvant controls. Last, MER5101-immunized APPswe/PS1ΔE9 Tg mice showed improvement of cognitive deficits in both contextual fear conditioning and the Morris water maze. Our novel, highly immunogenic Aß conjugate vaccine, MER5101, shows promise for improving Aß vaccine safety and efficacy and therefore, may be useful for preventing and/or treating early AD.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Péptidos beta-Amiloides/inmunología , Trastornos del Conocimiento/patología , Trastornos del Conocimiento/terapia , Toxoide Diftérico/inmunología , Inmunización/métodos , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animales , Formación de Anticuerpos/inmunología , Células CHO/química , Proliferación Celular/efectos de los fármacos , Corteza Cerebral/inmunología , Corteza Cerebral/metabolismo , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/inmunología , Condicionamiento Clásico/fisiología , Cricetinae , Medios de Cultivo Condicionados/farmacología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta Inmunológica , Ensayo de Inmunoadsorción Enzimática , Mapeo Epitopo , Miedo , Proteína Ácida Fibrilar de la Glía/metabolismo , Gliosis/inmunología , Gliosis/terapia , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Inmunoprecipitación , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Transgénicos , Mutación/genética , Fragmentos de Péptidos/inmunología , Presenilina-1/genética , Bazo/citología , Estadísticas no Paramétricas , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Transfección
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda