Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Acta Neuropathol Commun ; 10(1): 37, 2022 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-35305685

RESUMEN

Machado-Joseph disease (MJD) or Spinocerebellar ataxia type 3 (SCA3) is the most common form of dominant SCA worldwide. Magnetic Resonance Imaging (MRI) and Proton Magnetic Resonance Spectroscopy (1H-MRS) provide promising non-invasive diagnostic and follow-up tools, also serving to evaluate therapies efficacy. However, pre-clinical studies showing relationship between MRI-MRS based biomarkers and functional performance are missing, which hampers an efficient clinical translation of therapeutics. This study assessed motor behaviour, neurochemical profiles, and morphometry of the cerebellum of MJD transgenic mice and patients aiming at establishing magnetic-resonance-based biomarkers. 1H-MRS and structural MRI measurements of MJD transgenic mice were performed with a 9.4 Tesla scanner, correlated with motor performance on rotarod and compared with data collected from human patients. We found decreased cerebellar white and grey matter and enlargement of the fourth ventricle in both MJD mice and human patients as compared to controls. N-acetylaspartate (NAA), NAA + N-acetylaspartylglutamate (NAA + NAAG), Glutamate, and Taurine, were significantly decreased in MJD mouse cerebellum regardless of age, whereas myo-Inositol (Ins) was increased at early time-points. Lower neurochemical ratios levels (NAA/Ins and NAA/total Choline), previously correlated with worse clinical status in SCAs, were also observed in MJD mice cerebella. NAA, NAA + NAAG, Glutamate, and Taurine were also positively correlated with MJD mice motor performance. Importantly, these 1H-MRS results were largely analogous to those found for MJD in human studies and in our pilot data in human patients. We have established a magnetic resonance-based biomarker approach to monitor novel therapies in preclinical studies and human clinical trials.


Asunto(s)
Enfermedad de Machado-Joseph , Animales , Biomarcadores , Cerebelo/diagnóstico por imagen , Cerebelo/patología , Ácido Glutámico , Humanos , Enfermedad de Machado-Joseph/patología , Ratones , Ratones Transgénicos , Taurina
2.
Cytokine Growth Factor Rev ; 58: 114-133, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33397585

RESUMEN

The devastating global impact of the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has prompted scientists to develop novel strategies to fight Coronavirus Disease of 2019 (COVID-19), including the examination of pre-existing treatments for other viral infections in COVID-19 patients. This review provides a reasoned discussion of the possible use of Mesenchymal Stromal Cells (MSC) or their products as a treatment in SARS-CoV-2-infected patients. The main benefits and concerns of using this cellular therapy, guided by preclinical and clinical data obtained from similar pathologies will be reviewed. MSC represent a highly immunomodulatory cell population and their use may be safe according to clinical studies developed in other pathologies. Notably, four clinical trials and four case reports that have already been performed in COVID-19 patients obtained promising results. The clinical application of MSC in COVID-19 is very preliminary and further investigational studies are required to determine the efficacy of the MSC therapy. Nevertheless, these preliminary studies were important to understand the therapeutic potential of MSC in COVID-19. Based on these encouraging results, the United States Food and Drug Administration (FDA) authorized the compassionate use of MSC, but only in patients with Acute Respiratory Distress Syndrome (ARDS) and a poor prognosis. In fact, patients with severe SARS-CoV-2 can present infection and tissue damage in different organs, such as lung, heart, liver, kidney, gut and brain, affecting their function. MSC may have pleiotropic activities in COVID-19, with the capacity to fight inflammation and repair lesions in several organs.


Asunto(s)
COVID-19/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/fisiología , COVID-19/epidemiología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Tratamiento Basado en Trasplante de Células y Tejidos/tendencias , Humanos , Trasplante de Células Madre Mesenquimatosas/métodos , Trasplante de Células Madre Mesenquimatosas/tendencias , Insuficiencia Multiorgánica/prevención & control , Insuficiencia Multiorgánica/terapia , SARS-CoV-2/patogenicidad
3.
Adv Exp Med Biol ; 1049: 439-466, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29427116

RESUMEN

Polyglutamine (polyQ) diseases are a family of neurodegenerative disorders with very heterogeneous clinical presentations, although with common features such as progressive neuronal death. Thus, at the time of diagnosis patients might present an extensive and irreversible neuronal death demanding cell replacement or support provided by cell-based therapies. For this purpose stem cells, which include diverse populations ranging from embryonic stem cells (ESCs), to fetal stem cells, mesenchymal stromal cells (MSCs) or induced pluripotent stem cells (iPSCs) have remarkable potential to promote extensive brain regeneration and recovery in neurodegenerative disorders. This regenerative potential has been demonstrated in exciting pre and clinical assays. However, despite these promising results, several drawbacks are hampering their successful clinical implementation. Problems related to ethical issues, quality control of the cells used and the lack of reliable models for the efficacy assessment of human stem cells. In this chapter the main advantages and disadvantages of the available sources of stem cells as well as their efficacy and potential to improve disease outcomes are discussed.


Asunto(s)
Trastornos Heredodegenerativos del Sistema Nervioso/terapia , Trasplante de Células Madre/métodos , Células Madre , Animales , Encéfalo/fisiología , Trastornos Heredodegenerativos del Sistema Nervioso/genética , Trastornos Heredodegenerativos del Sistema Nervioso/metabolismo , Humanos , Péptidos/genética , Péptidos/metabolismo , Regeneración
4.
Neurobiol Dis ; 66: 92-103, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24607884

RESUMEN

In Krabbe's disease (KD), a leukodystrophy caused by ß-galactosylceramidase deficiency, demyelination and a myelin-independent axonopathy contributes to the severe neuropathology. Beyond axonopathy, we show that in Twitcher mice, a model of KD, a decreased number of axons both in the PNS and in the CNS, and of neurons in dorsal root ganglia (DRG), occurred before the onset of demyelination. Despite the early axonal loss, and although in vitro Twitcher neurites degenerated over time, Twitcher DRG neurons displayed an initial neurite overgrowth and, following sciatic nerve injury, Twitcher axons were regeneration-competent, at a time point where axonopathy was already ongoing. Psychosine, the toxic substrate that accumulates in KD, induced lipid raft clustering. At the mechanistic level, TrkA recruitment to lipid rafts was dysregulated in Twitcher neurons, and defective activation of the ERK1/2 and AKT pathways was identified. Besides defective recruitment of signaling molecules to lipid rafts, the early steps of endocytosis and the transport of endocytic and synaptic vesicles were impaired in Twitcher DRG neurons. Defects in axonal transport, specifically in the retrograde component, correlated with decreased levels of dynein, abnormal levels of post-translational tubulin modifications and decreased microtubule stability. The identification of the axonal defects that precede demyelination in KD, together with the finding that Twitcher axons are regeneration-competent when axonopathy is already installed, opens new windows of action to effectively correct the neuropathology that characterizes this disorder.


Asunto(s)
Transporte Axonal/fisiología , Axones/fisiología , Endocitosis/fisiología , Leucodistrofia de Células Globoides/fisiopatología , Microtúbulos/metabolismo , Animales , Axones/patología , Células Cultivadas , Modelos Animales de Enfermedad , Dineínas/metabolismo , Femenino , Ganglios Espinales/patología , Ganglios Espinales/fisiopatología , Leucodistrofia de Células Globoides/patología , Masculino , Microdominios de Membrana/patología , Microdominios de Membrana/fisiología , Ratones , Ratones Mutantes Neurológicos , Neuronas Motoras/patología , Neuronas Motoras/fisiología , Neuritas/patología , Neuritas/fisiología , Neuronas/patología , Neuronas/fisiología , Nervio Ciático/lesiones , Nervio Ciático/patología , Nervio Ciático/fisiopatología , Vesículas Sinápticas/patología , Vesículas Sinápticas/fisiología , Vesículas Transportadoras/patología , Vesículas Transportadoras/fisiología , Tubulina (Proteína)/metabolismo
5.
Cell Transplant ; 23(2): 239-52, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23809254

RESUMEN

Krabbe's disease (KD) is a demyelinating disorder caused by the deficiency of lysosomal galactocerebrosidase (GALC), affecting both the central (CNS) and the peripheral nervous system (PNS). A current therapy, hematopoietic stem cell transplantation (HSCT), is ineffective at correcting the PNS pathology. We have previously shown that systemic delivery of immortalized bone marrow-derived murine mesenchymal stromal cells (BM-MSCs) diminishes the neuropathology of transplanted Twitcher mice, a murine model of KD. In this study, to move one step closer to clinical application, the effectiveness of a systematic delivery of primary BM-MSCs to promote recovery of the Twitcher PNS was assessed. Primary BM-MSCs grafted to the Twitcher sciatic nerve led to increased GALC activity that was not correlated to decreased psychosine (the toxic GALC substrate) accumulation. Nevertheless, BM-MSC transplantation rescued the axonal phenotype of Twitcher mice in the sciatic nerve, with an increased density of both myelinated and unmyelinated axons in transplanted animals. Whereas no increase in myelination was observed, upon transplantation an increased proliferation of Schwann cell precursors occurred. Supporting these findings, in vitro, BM-MSCs promoted neurite outgrowth of Twitcher sensory neurons and proliferation of Twitcher Schwann cells. Moreover, BM-MSCs expressed nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) and promoted increased BDNF synthesis by neighboring Schwann cells. Besides their action in neurons and glia, BM-MSCs led to macrophage activation in Twitcher sciatic nerves. In summary, primary BM-MSCs diminish the neuropathology of Twitcher sciatic nerves by coordinately affecting neurons, glia, and macrophages.


Asunto(s)
Células Madre Mesenquimatosas/citología , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Femenino , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones Transgénicos , Factor de Crecimiento Nervioso/metabolismo , Psicosina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
6.
Cell Transplant ; 22(2): 189-204, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23006656

RESUMEN

Leukodystrophies are a group of disorders characterized by myelin dysfunction, either at the level of myelin formation or maintenance, that affect the central nervous system (CNS) and also in some cases, to a lesser extent, the peripheral nervous system (PNS). Although these genetic-based disorders are generally rare, all together they have a significant impact in the society, with an estimated overall incidence of 1 in 7,663 live births. Currently, there is no cure for leukodystrophies, and the development of effective treatments remains challenging. Not only leukodystrophies generally progress very fast, but also most are multifocal needing the simultaneous targeting at multiple sites. Moreover, as the CNS is affected, the blood-brain barrier (BBB) limits the efficacy of treatment. Recently, interest on cell therapy has increased, and the leukodystrophies for which metabolic correction is needed have become first-choice candidates for cell-based clinical trials. In this review, we present and discuss the available cell transplantation therapies in metabolic leukodystrophies including fucosidosis, X-linked adrenoleukodystrophy, metachromatic leukodystrophy, Canavan disease, and Krabbe's disease. We will discuss the latest advances of cell therapy and its pitfalls in this group of disorders, taking into account, among others, the limitations imposed by reduced cell migration in multifocal conditions, the need to achieve corrective enzyme threshold levels, and the growing awareness that not only myelin but also the associated axonopathy needs to be targeted in some leukodystrophies.


Asunto(s)
Encefalopatías/terapia , Trasplante de Células/tendencias , Tratamiento Basado en Trasplante de Células y Tejidos/tendencias , Leucodistrofia de Células Globoides/terapia , Leucodistrofia Metacromática/terapia , Animales , Encefalopatías/metabolismo , Encefalopatías/patología , Trasplante de Células/efectos adversos , Tratamiento Basado en Trasplante de Células y Tejidos/efectos adversos , Humanos , Leucodistrofia de Células Globoides/fisiopatología , Leucodistrofia Metacromática/metabolismo , Leucodistrofia Metacromática/patología
7.
Stem Cells ; 29(11): 1738-51, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21898691

RESUMEN

In Krabbe's disease, a demyelinating disorder, add-on strategies targeting the peripheral nervous system (PNS) are needed, as it is not corrected by bone-marrow (BM) transplantation. To circumvent this limitation of BM transplantation, we assessed whether i.v. delivery of immortalized EGFP(+) BM-derived murine mesenchymal stromal cells (BM-MSC(TERT-EGFP) ) targets the PNS of a Krabbe's disease model, the Twitcher mouse. In vitro, BM-MSC(TERT-EGFP) retained the phenotype of primary BM-MSC and did not originate tumors upon transplantation in nude mice. In vivo, undifferentiated EGFP(+) cells grafted the Twitcher sciatic nerve where an increase in Schwann cell precursors and axonal number was detected. The same effect was observed on BM-MSC(TERT-EGFP) i.v. delivery following sciatic nerve crush, a model of axonal regeneration. Reiterating the in vivo findings, in a coculture system, BM-MSC(TERT-EGFP) induced the proliferation of Twitcher-derived Schwann cells and the neurite outgrowth of both Twitcher-derived neurons and wild-type neurons grown in the presence of psychosine, the toxic substrate that accumulates in Krabbe's disease. In vitro, this neuritogenic effect was blocked by K252a, an antagonist of Trk receptors, and by antibody blockage of brain derived neurotrophic factor, a neurotrophin secreted by BM-MSC(TERT-EGFP) and induced in neighboring Schwann cells. In vivo, BM-MSC(TERT-EGFP) surmounted the effect of K252a, indicating their ability to act through a neurotrophin-independent mechanism. In summary, i.v. delivery of BM-MSC(TERT-EGFP) exerts a multilevel effect targeting neurons and Schwann cells, coordinately diminishing neuropathology. Therefore, to specifically target the PNS, MSC should be considered an add-on option to BM transplantation in Krabbe's disease and in other disorders where peripheral axonal loss occurs.


Asunto(s)
Células de la Médula Ósea/citología , Leucodistrofia de Células Globoides/metabolismo , Leucodistrofia de Células Globoides/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Adipogénesis/efectos de los fármacos , Adipogénesis/genética , Animales , Western Blotting , Carbazoles/farmacología , Línea Celular , Inhibidores Enzimáticos/farmacología , Citometría de Flujo , Inmunohistoquímica , Alcaloides Indólicos/farmacología , Leucodistrofia de Células Globoides/genética , Ratones , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Nervio Ciático/lesiones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA