Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 209
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(29): e2110746119, 2022 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-35858361

RESUMEN

Intraneuronal inclusions of misfolded α-synuclein (α-syn) and prion-like spread of the pathologic α-syn contribute to progressive neuronal death in Parkinson's disease (PD). Despite the pathologic significance, no efficient therapeutic intervention targeting α-synucleinopathy has been developed. In this study, we provide evidence that astrocytes, especially those cultured from the ventral midbrain (VM), show therapeutic potential to alleviate α-syn pathology in multiple in vitro and in vivo α-synucleinopathic models. Regulation of neuronal α-syn proteostasis underlies the therapeutic function of astrocytes. Specifically, VM-derived astrocytes inhibited neuronal α-syn aggregation and transmission in a paracrine manner by correcting not only intraneuronal oxidative and mitochondrial stresses but also extracellular inflammatory environments, in which α-syn proteins are prone to pathologic misfolding. The astrocyte-derived paracrine factors also promoted disassembly of extracellular α-syn aggregates. In addition to the aggregated form of α-syn, VM astrocytes reduced total α-syn protein loads both by actively scavenging extracellular α-syn fibrils and by a paracrine stimulation of neuronal autophagic clearance of α-syn. Transplantation of VM astrocytes into the midbrain of PD model mice alleviated α-syn pathology and protected the midbrain dopamine neurons from neurodegeneration. We further showed that cografting of VM astrocytes could be exploited in stem cell-based therapy for PD, in which host-to-graft transmission of α-syn pathology remains a critical concern for long-term cell therapeutic effects.


Asunto(s)
Astrocitos , Trasplante de Tejido Encefálico , Enfermedad de Parkinson , Proteostasis , alfa-Sinucleína , Animales , Astrocitos/trasplante , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/metabolismo , Mesencéfalo/patología , Mesencéfalo/cirugía , Ratones , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/terapia , alfa-Sinucleína/metabolismo
3.
Int J Mol Sci ; 22(22)2021 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-34830023

RESUMEN

Transplantation of exogenous dopaminergic (DA) neurons is an alternative strategy to replenish DA neurons that have lost along the course of Parkinson's disease (PD). From the perspective of ethical acceptation, the source limitations, and the intrinsic features of PD pathology, astrocytes (AS) and mesenchymal stem cells (MSCs) are the two promising candidates of DA induction. In the present study, we induced AS or MSCs primary culture by the combination of the classical transcription-factor cocktails Mash1, Lmx1a, and Nurr1 (MLN), the chemical cocktails (S/C/D), and the morphogens SHH, FGF8, and FGF2 (S/F8/F2); the efficiency of induction into DA neurons was further analyzed by using immunostaining against the DA neuronal markers. AS could be efficiently converted into the DA neurons in vitro by the transcriptional regulation of MLN, and the combination with S/C/D or S/F8/F2 further increased the conversion efficiency. In contrast, MSCs from umbilical cord (UC-MSCs) or adipose tissue (AD-MSCs) showed moderate TH immunoreactivity after the induction with S/F8/F2 instead of with MLN or S/C/D. Our data demonstrated that AS and MSCs held lineage-specific molecular codes on the induction into DA neurons and highlighted the unique superiority of AS in the potential of cell replacement therapy for PD.


Asunto(s)
Astrocitos/trasplante , Neuronas Dopaminérgicas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Enfermedad de Parkinson/terapia , Animales , Astrocitos/metabolismo , Diferenciación Celular/genética , Dopamina/metabolismo , Neuronas Dopaminérgicas/patología , Neuronas Dopaminérgicas/trasplante , Humanos , Trasplante de Células Madre Mesenquimatosas , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Cultivo Primario de Células , Ratas , Factores de Transcripción/genética , Cordón Umbilical/metabolismo , Cordón Umbilical/trasplante
4.
Acta Neuropathol ; 142(1): 117-137, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33913039

RESUMEN

Loss-of-function variants in the low-density lipoprotein receptor-related protein 10 (LRP10) gene have been associated with autosomal-dominant Parkinson's disease (PD), PD dementia, and dementia with Lewy bodies (DLB). Moreover, LRP10 variants have been found in individuals diagnosed with progressive supranuclear palsy and amyotrophic lateral sclerosis. Despite this genetic evidence, little is known about the expression and function of LRP10 protein in the human brain under physiological or pathological conditions. To better understand how LRP10 variants lead to neurodegeneration, we first performed an in-depth characterisation of LRP10 expression in post-mortem brains and human-induced pluripotent stem cell (iPSC)-derived astrocytes and neurons from control subjects. In adult human brain, LRP10 is mainly expressed in astrocytes and neurovasculature but undetectable in neurons. Similarly, LRP10 is highly expressed in iPSC-derived astrocytes but cannot be observed in iPSC-derived neurons. In astrocytes, LRP10 is present at trans-Golgi network, plasma membrane, retromer, and early endosomes. Interestingly, LRP10 also partially co-localises and interacts with sortilin-related receptor 1 (SORL1). Furthermore, although LRP10 expression and localisation in the substantia nigra of most idiopathic PD and DLB patients and LRP10 variant carriers diagnosed with PD or DLB appeared unchanged compared to control subjects, significantly enlarged LRP10-positive vesicles were detected in a patient carrying the LRP10 p.Arg235Cys variant. Last, LRP10 was detected in Lewy bodies (LB) at late maturation stages in brains from idiopathic PD and DLB patients and in LRP10 variant carriers. In conclusion, high LRP10 expression in non-neuronal cells and undetectable levels in neurons of control subjects indicate that LRP10-mediated pathogenicity is initiated via cell non-autonomous mechanisms, potentially involving the interaction of LRP10 with SORL1 in vesicle trafficking pathways. Together with the specific pattern of LRP10 incorporation into mature LBs, these data support an important mechanistic role for disturbed vesicle trafficking and loss of LRP10 function in neurodegenerative diseases.


Asunto(s)
Encéfalo/metabolismo , Proteínas Relacionadas con Receptor de LDL/genética , Cuerpos de Lewy/metabolismo , Enfermedad por Cuerpos de Lewy/metabolismo , Proteínas de Transporte de Membrana/genética , Enfermedad de Parkinson/metabolismo , Adulto , Anciano , Astrocitos/metabolismo , Astrocitos/trasplante , Encéfalo/citología , Encéfalo/patología , Variación Genética , Humanos , Células Madre Pluripotentes Inducidas/trasplante , Cuerpos de Lewy/patología , Enfermedad por Cuerpos de Lewy/patología , Persona de Mediana Edad , Enfermedades Neurodegenerativas/patología , Neuronas/trasplante , Enfermedad de Parkinson/patología
5.
Mol Cell Biochem ; 476(2): 619-631, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33070275

RESUMEN

Parkinson's disease (PD) is a disorder characterized by a progressive loss of the dopaminergic neurons in the substantia nigra and a depletion of the neurotransmitter dopamine in the striatum. Our published results indicate that fasciculation and elongation protein zeta-1 (FEZ1) plays a role in the astrocyte-mediated protection of dopamine neurons and regulation of the neuronal microenvironment during the progression of PD. In this study, we examined the effects of engrafted type-2 astrocytes (T2As) with high expression of FEZ1 on the improvement of the symptoms and functional reconstruction of PD rats. T2As were stereotactically transplanted into the striatum of rats with PD induced by 6-hydroxydopamine (6-OHDA). An examination of apomorphine (APO)-induced rotations was performed to evaluate dopamine neuron damage and motor functions. Remarkably, the grafted cells survived in the lesion environment for six weeks or longer after implantation. In addition, the transplantation of T2As decrease the average velocity and the duration time of the APO-induced rotations, and increase the actuation time, as measured in the rotation behavioural tests. In the substantia nigra, the transplantation of T2As reduced the PD-induced GFAP, TH and FEZ1 downregulation. The grafted cells exclusively migrated to other regions near the injection site in the striatum and differentiated into GFAP+ astrocytes or TH+ neurons. Furthermore, by detecting monoamine neurotransmitters through high-performance liquid chromatography, we found that the nigrostriatal pathway had been repaired to some extent. Taken together, these results suggest that engrafted T2As with high expression of FEZ1 improved the symptoms and functional reconstruction of PD rats, providing a theoretical basis for FEZ1 as a potential target and engraftment of T2As as a therapeutic strategy in the treatment of PD.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Apomorfina/farmacología , Astrocitos/trasplante , Neuronas Dopaminérgicas/efectos de los fármacos , Enfermedad de Parkinson/terapia , Sustancia Negra/metabolismo , Adrenérgicos/administración & dosificación , Animales , Astrocitos/citología , Astrocitos/metabolismo , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/metabolismo , Masculino , Actividad Motora/efectos de los fármacos , Oxidopamina/administración & dosificación , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Ratas , Ratas Sprague-Dawley
6.
Neurotoxicology ; 83: 28-39, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33309839

RESUMEN

Exosomes play critical roles in neurogenesis. This study aims to explore the mechanism of astrocyte-derived exosomes in neonatal rats with hypoxic-ischemic brain damage (HIBD). Astrocytes were collected and astrocyte-derived exosomes were isolated and identified. Neonatal rats were pre-treated with exosomes and then subjected to HIBD induction. Then the neurobehaviors, neuronal apoptosis, inflammation and oxidative stress in rat brain were measured. Differentially expressed microRNAs (miRNAs) in rat brain before and after HI procedure were analyzed. H19-7 cells were subjected to oxygen and glucose deprivation (OGD) for in vitro studies. Target relation between miR-17-5p and BNIP2 was identified. Gain- and loss-of functions of miR-17-5p and BNIP2 were conducted to identify their roles in viability, apoptosis, oxidative stress and inflammation of OGD-treated cells. Collectively, astrocyte-derived exosomes improved neurobehaviors, and reduced cerebral infarction, neuronal apoptosis, oxidative and inflammation in vivo and in vitro. The exosomes carried miR-17-5p bound to BNIP2 and negatively regulated BNIP2 expression in OGD-treated cells. Over-expression of miR-17-5p increased viability, and decreased OGD-induced apoptosis, oxidative stress and inflammation of H19-7 cells, which were reversed by over-expression of BNIP2. Taken together, the study suggested that astrocyte-derived exosomes could carry miR-17-5p to protect neonatal rats from HIBD via inhibiting BNIP-2 expression.


Asunto(s)
Astrocitos/trasplante , Encéfalo/metabolismo , Exosomas/trasplante , Terapia Genética , Hipoxia-Isquemia Encefálica/prevención & control , MicroARNs/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Animales , Animales Recién Nacidos , Apoptosis , Astrocitos/metabolismo , Encéfalo/patología , Línea Celular , Modelos Animales de Enfermedad , Regulación hacia Abajo , Exosomas/genética , Exosomas/metabolismo , Hipoxia-Isquemia Encefálica/genética , Hipoxia-Isquemia Encefálica/metabolismo , Hipoxia-Isquemia Encefálica/patología , Mediadores de Inflamación/metabolismo , MicroARNs/genética , Estrés Oxidativo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Ratas Sprague-Dawley
7.
PLoS One ; 14(11): e0225760, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31774879

RESUMEN

Even though HIV-1 replication can be suppressed by combination antiretroviral therapy (cART) inflammatory processes still occur, contributing to comorbidities. Comorbidities are attributed to variety of factors, including HIV-1 mediated inflammation. Several HIV-1 proteins mediate central nervous system (CNS) inflammation, including Nef. Nef is an early HIV-1 protein, toxic to neurons and glia and is sufficient to cause learning impairment similar to some deficits observed in HIV-1 associated neurocognitive disorders. To determine whether hippocampal Nef expression by astrocytes contributes to comorbidities, specifically peripheral inflammation, we infused Sprague Dawley rats with GFP- (control) or Nef-transfected astrocytes into the right hippocampus. Brain, lung, and ileum were collected postmortem for the measurement of inflammatory markers. Increased blood-brain-barrier permeability and serum IL-1ß levels were detected in the Nef-treated rats. The lungs of Nef-treated rats demonstrated leukocyte infiltration, macrophage upregulation, and enhanced vascular permeability. Ileal tissue showed reactive follicular lymphoid hyperplasia, increased permeability and macrophage infiltration. The intracerebroventricular application of IL-1 receptor antagonist reduced infiltration of immune cells into ileum and lung, indicating the important role of IL-1ß in mediating the spread of inflammation from the brain to other tissues. This suggests that localized expression of a single viral protein, HIV-1 Nef, can contribute to a broader inflammatory response by upregulation of IL-1ß. Further, these results suggest that Nef contributes to the chronic inflammation seen in HIV patients, even in those whose viremia is controlled by cART.


Asunto(s)
Astrocitos/trasplante , Barrera Hematoencefálica/patología , Hipocampo/patología , Enfermedades Pulmonares Intersticiales/etiología , Neuronas/patología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Animales , Astrocitos/metabolismo , Barrera Hematoencefálica/metabolismo , Células Cultivadas , Hipocampo/metabolismo , Interleucina-1beta/metabolismo , Enfermedades Pulmonares Intersticiales/patología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética
8.
Stem Cells ; 37(10): 1344-1356, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31287930

RESUMEN

Due to the limited capacity of brain tissue to self-regenerate after traumatic brain injury (TBI), the mobilization of endogenous neural stem cells (NSCs) is a popular research topic. In the clinic, the neurogenic abilities of adults versus neonates vary greatly, which is likely related to functional differences in NSCs. Recent studies have demonstrated that the molecules secreted from astrocytes play important roles in NSC fate determination. In this study, conditioned media (CM) derived from neonatal or adult rat astrocytes, which were unstimulated or stimulated by lipopolysaccharide (LPS), were prepared to treat NSCs. Our results revealed that neonatal rat astrocytes can significantly promote the proliferation of NSCs, compared with adult rat astrocytes, regardless of whether or not they were stimulated by LPS. Furthermore, we used mass spectrometry to detect the constituents of the CM from each group. We analyzed and screened for a protein, Tenascin-C (TNC), which was highly expressed in the neonatal group but poorly expressed in the adult group. We found that TNC can bind to the NSC surface epidermal growth factor receptor and promote proliferation through the PI3K-AKT pathway in vitro. Additionally, we confirmed in vivo that TNC can promote damage repair in a rat model of TBI, through enhancing the proliferation of endogenous NSCs. We believe that these findings provide a mechanistic understanding of why neonates show better neuroregenerative abilities than adults. This also provides a potential future therapeutic target, TNC, for injury repair after TBI. Stem Cells 2019;37:1344-1356.


Asunto(s)
Astrocitos/trasplante , Lesiones Traumáticas del Encéfalo/terapia , Células-Madre Neurales/trasplante , Animales , Diferenciación Celular , Proliferación Celular , Humanos , Ratas
9.
Nanomedicine ; 21: 102048, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31271878

RESUMEN

Neurodegenerative diseases and traumatic brain injuries can destroy neurons, resulting in sensory and motor function loss. Transplantation of differentiated neurons from stem cells could help restore such lost functions. Plasmonic gold nanorods (AuNR) were integrated in growth surfaces to stimulate and modulate neural cells in order to tune cell physiology. An AuNR nanocomposite system was fabricated, characterized, and then utilized to study the differentiation of embryonic rat neural stem cells (NSCs). Results demonstrated that this plasmonic surface 1) accelerated differentiation, yielding almost twice as many differentiated neural cells as a traditional NSC culture surface coated with poly-D-lysine and laminin for the same time period; and 2) promoted differentiation of NSCs into neurons and astrocytes in a 2:1 ratio, as evidenced by the expression of relevant marker proteins. These results indicate that the design and properties of this AuNR plasmonic surface would be advantageous for tissue engineering to address neural degeneration.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Nanotubos/química , Enfermedades Neurodegenerativas/terapia , Neuronas/trasplante , Animales , Astrocitos/trasplante , Lesiones Traumáticas del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/terapia , Células Cultivadas , Células Madre Embrionarias/efectos de los fármacos , Oro/química , Oro/farmacología , Humanos , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/trasplante , Enfermedades Neurodegenerativas/patología , Neuronas/efectos de los fármacos , Ratas
10.
Mult Scler Relat Disord ; 28: 263-272, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30639828

RESUMEN

Sox2 transcription factor has been frequently used for reprograming starting cells to neural progenitor/stem cells. In vivo administration of Sox2 in the adult mouse brain reprogrammed the transduced astrocytes to neurons. In searching for adequate cell source for repairing the myelin insults, here, we studied the possible conversion of astrocytes to oligodendrocyte lineage cells by Sox2, while an extensive demyelination exists in animal brain. Lentiviral particles expressing Sox2-GFP were injected into the corpora callosa of animals fed with cuprizone diet for 12 weeks. Transduced cells were mainly astrocytes that changed their fate to oligodendrocyte lineage cells by time. For further conformation astrocytes received the vector in culture and then transplanted to the animal brains. Tracing the fate of transplanted cells showed their conversion to oligodendrocyte lineage cells. In vitro transduced cell were also maintained in the oligodendrocyte progenitor cell (OPC) induction medium. Produced OPC-like cells were positive for specific markers. This study provides a new strategy for endogenous production of myelinating cells. After optimizing the experimental conditions for safety and feasibility, this approach may contribute into future cell based therapies in patients with white matter insults as like as those with multiple sclerosis.


Asunto(s)
Astrocitos/metabolismo , Encéfalo/metabolismo , Linaje de la Célula/fisiología , Enfermedades Desmielinizantes/metabolismo , Oligodendroglía/metabolismo , Factores de Transcripción SOXB1/metabolismo , Animales , Astrocitos/patología , Astrocitos/trasplante , Encéfalo/patología , Diferenciación Celular/fisiología , Células Cultivadas , Cuprizona , Enfermedades Desmielinizantes/patología , Enfermedades Desmielinizantes/terapia , Modelos Animales de Enfermedad , Vectores Genéticos , Lentivirus/genética , Masculino , Ratones Endogámicos C57BL , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Células-Madre Neurales/trasplante , Oligodendroglía/patología , Oligodendroglía/trasplante , Regeneración/fisiología , Factores de Transcripción SOXB1/genética
11.
Regen Med ; 13(7): 803-819, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30284949

RESUMEN

AIM: It would be clinically ideal to target astrocytes in vivo for conversion into oligodendrocyte lineage cells to reduce astrogliosis and generate new myelinating cells. MATERIALS & METHODS: Here, we prepared a GFP-labeled human astrocyte cell line, treated with epigenetic modifiers trichostatin A or 5-azacytidine and transplanted them into cuprizone-induced demyelinated mice brains. The fate of the transplanted astrocytes was studied at days 7, 14 and 28 post-transplantation. RESULTS: GFP+ astrocytes were reduced over time, whereas the GFP+ oligodendrocyte lineage cells were found on days 14 and 28. Nontreated astrocytes did not convert to myelinating cells following transplantation. Cell conversion was proved in vitro by maintaining the treated cells in oligodendrocyte progenitor cell medium. CONCLUSION: These findings seem promising for the application of epigenetic modifiers, especially their targeted delivery to glial scars to treat demyelinating diseases.


Asunto(s)
Astrocitos/citología , Encéfalo/patología , Técnicas de Reprogramación Celular , Gliosis/patología , Vaina de Mielina/metabolismo , Oligodendroglía/citología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/trasplante , Azacitidina/farmacología , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Trasplante de Células , Enfermedades Desmielinizantes , Epigenómica , Humanos , Ácidos Hidroxámicos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Oligodendroglía/metabolismo
12.
Adv Mater ; 30(50): e1805209, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30285286

RESUMEN

Immunology is the next frontier of nano/biomaterial science research, with the immune system determining the degree of tissue repair. However, the complexity of the inflammatory response represents a significant challenge that is essential to understand for the development of future therapies. Cell-instructive 3D culture environments are critical to improve our understanding of the link between the behavior and morphology of inflammatory cells and to remodel their response to injury. This study has taken two recent high-profile innovations-functional peptide-based hydrogels, and the inclusion of anti-inflammatory agents via coassembly-to make a programmed anti-inflammatory nanoscaffold (PAIN) with unusual and valuable properties that allows tissue-independent switching of the inflammatory cascade. Here, extraordinary durability of the anti-inflammatory agent allows, for the first time, the development of a 3D culture system that maintains the growth and cytoskeletal reorganization of brain tissue, while also facilitating the trophic behavior of brain cells for 22 d in vitro. Notably, this behavior was confirmed within an active scar site due to the unprecedented resilience to the presence of inflammatory cells and enzymes in the brain. Efficacy of the culture system is demonstrated via novel insights about inflammatory cell behavior, which would be impossible to obtain via in vivo experimentation.


Asunto(s)
Antiinflamatorios/química , Nanotecnología/métodos , Andamios del Tejido/química , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Astrocitos/citología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/trasplante , Encéfalo/metabolismo , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Técnicas de Cultivo de Célula , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Femenino , Hidrogeles/química , Interleucina-1alfa/metabolismo , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Nanotecnología/instrumentación , Péptidos/química , Reología , Ingeniería de Tejidos
13.
Stem Cell Res Ther ; 9(1): 241, 2018 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-30257722

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a fatal paralytic disease with no cure or treatment to stop disease progression. Because ALS represents an urgent unmet medical need, a significant number of therapeutics are being tested in preclinical and clinical studies. A recent publication in Stem Cell Research & Therapy by Izrael and colleagues reports about embryonic stem cell-derived astrocytes as a potential cell therapy for ALS. Such cells behave as highly trophic "young astrocytes", being able to delay disease onset and prolong survival when injected intrathechally in murine models of ALS overexpressing the SOD1G93A mutation. The safety and therapeutic potential of these cells are currently being evaluated in a clinical trial in ALS patients. This commentary discusses the mechanisms of action and potential therapeutic effects of these "young astrocytes" in ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/terapia , Astrocitos/trasplante , Tratamiento Basado en Trasplante de Células y Tejidos , Superóxido Dismutasa-1/genética , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/fisiopatología , Animales , Astrocitos/citología , Modelos Animales de Enfermedad , Regulación Enzimológica de la Expresión Génica/genética , Humanos , Ratones , Ratones Transgénicos , Neuronas Motoras
14.
J Control Release ; 286: 114-124, 2018 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-30026082

RESUMEN

Degeneration of dopamine (DA) neurons in the substantia nigra pars compacta is the pathological hallmark of Parkinson's disease (PD). In PD multiple pathogenic mechanisms initiate and drive this neurodegenerative process, making the development of effective treatments challenging. To date, PD patients are primarily treated with dopaminergic drugs able to temporarily enhance DA levels, therefore relieving motor symptoms. However, the drawbacks of these therapies including the inability to alter disease progression are constantly supporting the search for alternative treatment approaches. Over the past years efforts have been put into the development of new therapeutic strategies based on the delivery of therapeutic genes using viral vectors or transplantation of DA neurons for cell-based DA replacement. Here, past achievements and recent advances in gene- and cell-based therapies for PD are outlined. We discuss how current gene and cell therapy strategies hold great promise for the treatment of PD and how the use of stem cells and recent developments in cellular reprogramming could contribute to open a new avenue in PD therapy.


Asunto(s)
Técnicas de Reprogramación Celular/métodos , Terapia Genética/métodos , Enfermedad de Parkinson/terapia , Animales , Astrocitos/citología , Astrocitos/metabolismo , Astrocitos/trasplante , Reprogramación Celular , Dopamina/genética , Dopamina/metabolismo , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/trasplante , Humanos , Microglía/citología , Microglía/metabolismo , Microglía/trasplante , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo
15.
Nanomedicine (Lond) ; 13(11): 1333-1348, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29949467

RESUMEN

AIM: To develop a 3D neural cell construct for encapsulated delivery of transplant cells; develop hydrogels seeded with magnetic nanoparticle (MNP)-labeled cells suitable for cell tracking by MRI. MATERIALS & METHODS: Astrocytes were exogenously labeled with MRI-compatible iron-oxide MNPs prior to intra-construct incorporation within a 3D collagen hydrogel. RESULTS: A connective, complex cellular network was clearly observable within the 3D constructs, with high cellular viability. MNP accumulation in astrocytes provided a hypointense MRI signal at 24 h & 14 days. CONCLUSION: Our findings support the concept of developing a 3D construct possessing the dual advantages of (i) support of long-term cell survival of neural populations with (ii) the potential for noninvasive MRI-tracking of intra-construct cells for neuroregenerative applications.


Asunto(s)
Astrocitos/ultraestructura , Tratamiento Basado en Trasplante de Células y Tejidos , Nanopartículas de Magnetita/administración & dosificación , Degeneración Nerviosa/diagnóstico por imagen , Neuronas/ultraestructura , Animales , Astrocitos/química , Astrocitos/trasplante , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Rastreo Celular , Humanos , Hidrogeles/química , Hidrogeles/farmacología , Imagen por Resonancia Magnética , Nanopartículas de Magnetita/química , Microscopía Electrónica de Transmisión , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/patología , Neuronas/química , Neuronas/trasplante , Polímeros/administración & dosificación , Polímeros/química , Ratas
17.
Mol Neurobiol ; 55(11): 8563-8585, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29564811

RESUMEN

Astrocytes play metabolic and structural support roles and contribute to the integrity of the blood-brain barrier (BBB), linking communication between neurons and the endothelium. Cyclin-dependent kinase 5 (CDK5) likely exerts a dual effect on the endothelium and astrocytes due to its involvement in migration and angiogenesis; the overactivation of CDK5 is associated with dysfunction in glutamate recapture and hypoxia. Recently, we proposed that CDK5-targeted astrocytes facilitate the recovery of neurological and motor function in transplanted ischemic rats. In the current study, we treated cerebral ischemic rats and endothelial cells exposed to glutamate toxicity with CDK5 knock-down (CDK5-KD) astrocytes to determine the role of CDK5 in neurovascular integrity. We found that the effects of CDK5-KD were sustained for 4 months, preventing neuronal and astrocyte loss, facilitating the recovery of the BBB via the production of BDNF by endogenous astrocytes (GFP-) surrounding vessels in the motor cortex and the corpus callosum of global ischemic rats, and improving neurological performance. These findings were supported by the in vitro findings of increased transendothelial resistance, p120-ctn+ adhesion and reduced intercellular gaps induced by a CDK5 inhibitor (roscovitine) in bEnd.3 cells in a glutamate-toxicity model. Additionally, CDK5-KD astrocytes in co-culture protected the endothelial cell viability, increased BDNF release from astrocytes, increased BDNF immunoreactivity in neighboring astrocytes and endothelial cells and enhanced cell adhesion in a glutamate-toxicity model. Altogether, these findings suggest that a CDK5 reduction in astrocytes protects the endothelium, which promotes BDNF release, endothelial adhesion, and the recovery of neurovascular unit integrity and brain function in ischemic rats.


Asunto(s)
Astrocitos/trasplante , Isquemia Encefálica/enzimología , Isquemia Encefálica/terapia , Encéfalo/irrigación sanguínea , Quinasa 5 Dependiente de la Ciclina/metabolismo , Técnicas de Silenciamiento del Gen , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Isquemia Encefálica/patología , Isquemia Encefálica/fisiopatología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Adhesión Celular , Línea Celular , Técnicas de Cocultivo , Cuerpo Calloso/metabolismo , Modelos Animales de Enfermedad , Impedancia Eléctrica , Células Endoteliales/metabolismo , Glutamatos/toxicidad , Masculino , Ratones , Actividad Motora , Corteza Motora/metabolismo , Corteza Motora/patología , Neuronas/metabolismo , Ratas Wistar , Accidente Cerebrovascular/patología , Accidente Cerebrovascular/fisiopatología
18.
J Clin Invest ; 128(1): 463-482, 2018 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-29227284

RESUMEN

Transplantation of neural progenitor cells (NPCs) is a potential therapy for treating neurodegenerative disorders, but this approach has faced many challenges and limited success, primarily because of inhospitable host brain environments that interfere with enriched neuron engraftment and function. Astrocytes play neurotrophic roles in the developing and adult brain, making them potential candidates for helping with modification of hostile brain environments. In this study, we examined whether astrocytic function could be utilized to overcome the current limitations of cell-based therapies in a murine model of Parkinson's disease (PD) that is characterized by dopamine (DA) neuron degeneration in the midbrain. We show here that cografting astrocytes, especially those derived from the midbrain, remarkably enhanced NPC-based cell therapeutic outcomes along with robust DA neuron engraftment in PD rats for at least 6 months after transplantation. We further show that engineering of donor astrocytes with Nurr1 and Foxa2, transcription factors that were recently reported to polarize harmful immunogenic glia into the neuroprotective form, further promoted the neurotrophic actions of grafted astrocytes in the cell therapeutic approach. Collectively, these findings suggest that cografting astrocytes could be a potential strategy for successful cell therapeutic outcomes in neurodegenerative disorders.


Asunto(s)
Astrocitos/trasplante , Mesencéfalo/metabolismo , Células-Madre Neurales/trasplante , Enfermedad de Parkinson Secundaria/terapia , Animales , Astrocitos/metabolismo , Astrocitos/patología , Modelos Animales de Enfermedad , Factor Nuclear 3-beta del Hepatocito/metabolismo , Mesencéfalo/patología , Ratones , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Enfermedad de Parkinson Secundaria/inducido químicamente , Enfermedad de Parkinson Secundaria/metabolismo , Enfermedad de Parkinson Secundaria/patología , Ratas , Ratas Sprague-Dawley
19.
Balkan Med J ; 35(2): 141-147, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29039346

RESUMEN

BACKGROUND: Transplantation of fetal mesencephalic tissue is a well-established concept for functional reinnervation of the dopamine-depleted rat striatum. However, there is no extensive description of the glial response of the host brain following this procedure. AIMS: The present study aimed to quantitatively and qualitatively analyse astrogliosis surrounding intrastriatal grafts and compare it to the reaction to mechanical injury with the transplantation instrument only. STUDY DESIGN: Animal experimentation. METHODS: The standard 6-hydroxydopamine-induced unilateral model of Parkinson's disease was used. The experimental animals received transplantation of a single-cell suspension of E14 ventral mesencephalic tissue. Control animals (sham-transplanted) were subjected to injury by the transplantation cannula, without injection of a cell suspension. Histological analyses were carried out 7 and 28 days following the procedure by immunohistochemistry assays for tyrosine hydroxylase and glial fibrillary acidic protein. To evaluate astrogliosis, the cell density and immunopositive area were measured in distinct zones within and surrounding the grafts or the cannula tract. RESULTS: Statistical analysis revealed that astrogliosis in the grafted striatum increased from day 7 to day 28, as shown by a significant change in both cell density and the immunopositive area. The cell density increased from 816.7±370.6 to 1403±272.1 cells/mm2 (p<0.0001) аnd from 523±245.9 to 1164±304.8 cells/mm2 (p<0.0001) in the two zones in the graft core, and from 1151±218.6 to 1485±210.6 cells/mm2 (p<0.05) for the zone in the striatum immediately adjacent to the graft. The glial fibrillary acidic protein-expressing area increased from 0.3109±0.1843 to 0.7949±0.1910 (p<0.0001) and from 0.1449±0.1240 to 0.702±0.2558 (p<0.0001) for the same zones in the graft core, and from 0.5277±0.1502 to 0.6969±0.1223 (p<0.0001) for the same area adjacent to the graft zone. However, astrogliosis caused by mechanical impact only (control) did not display such dynamics. This finding suggests an influence of the grafted cells on the host's glia, possibly through cross-talk between astrocytes and transplanted neurons. CONCLUSION: This bidirectional relationship is affected by multiple factors beyond the mechanical trauma. Elucidation of these factors might help achieve better functional outcomes after intracerebral transplantation.


Asunto(s)
Astrocitos/trasplante , Trasplante de Tejido Encefálico , Trasplante de Células , Modelos Animales de Enfermedad , Trasplante de Tejido Fetal , Enfermedad de Parkinson/terapia , Animales , Masculino , Ratas , Ratas Sprague-Dawley , Roedores
20.
Glia ; 65(12): 1867-1884, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28631858

RESUMEN

Rapid advances in Ca2+ imaging techniques enable us to simultaneously monitor the activities of hundreds of astrocytes in the intact brain, thus providing a powerful tool for understanding the functions of both host and engrafted astrocytes in sensory processing in vivo. These techniques include both improved Ca2+ indicators and advanced optical recording methods. Astrocytes in multiple cortical and sub-cortical areas are able to respond to the corresponding sensory modalities. These sensory stimuli produce astrocytic Ca2+ responses through different cellular mechanisms. In addition, it has been suggested that astrocytic gene deficiencies in various sensory systems cause impairments in sensory circuits and cognition. Therefore, glial transplantation would be a potentially interesting approach for the cell-based therapy for glia-related disorders. There are multiple cell sources for glial transplantation, including neural stem cells, glial progenitors, and pluripotent stem cells. Both in vitro and in vivo studies have shown that engrafted astrocytes derived from these cell sources are capable of responding to sensory stimulation by elevating the intracellular Ca2+ concentration. These results indicate that engrafted astrocytes not only morphologically but also functionally integrate into the host neural network. Until now, many animal studies have proven that glial transplantation would be a good choice for treating multiple glial disorders. Together, these studies on the sensory responses of host and engrafted astrocytes have provided us a novel perspective in both neuron-glia circuit functions and future treatment strategies for glial disorders.


Asunto(s)
Vías Aferentes/fisiología , Astrocitos/fisiología , Astrocitos/trasplante , Encéfalo/citología , Sensación/fisiología , Animales , Calcio/metabolismo , Humanos , Células Madre/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA