Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(7)2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-38612613

RESUMEN

The clinical severity of multiple sclerosis (MS), an autoimmune disorder of the central nervous system, is thought to be determined by environmental and genetic factors that have not yet been identified. In a recent genome-wide association study (GWAS), a single nucleotide polymorphism (SNP), rs10191329, has been associated with MS severity in two large independent cohorts of patients. Different approaches were followed by the authors to prioritize the genes that are transcriptionally regulated by such an SNP. It was concluded that the identified SNP regulates a group of proximal genes involved in brain resilience and cognitive abilities rather than immunity. Here, by conducting an alternative strategy for gene prioritization, we reached the opposite conclusion. According to our re-analysis, the main target of rs10191329 is N-Acetylglucosamine Kinase (NAGK), a metabolic gene recently shown to exert major immune functions via the regulation of the nucleotide-binding oligomerization domain-containing protein 2 (NOD2) pathway. To gain more insights into the immunometabolic functions of NAGK, we analyzed the currently known list of NAGK protein partners. We observed that NAGK integrates a dense network of human proteins that are involved in glucose metabolism and are highly expressed by classical monocytes. Our findings hold potentially major implications for the understanding of MS pathophysiology.


Asunto(s)
Enfermedades Autoinmunes , Esclerosis Múltiple , Humanos , Esclerosis Múltiple/genética , Estudio de Asociación del Genoma Completo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Acetilglucosamina
2.
Int J Mol Sci ; 23(23)2022 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-36499320

RESUMEN

In a substantial share of patients suffering from multiple sclerosis (MS), neurological functions slowly deteriorate despite a lack of radiological activity. Such a silent progression, observed in either relapsing-remitting or progressive forms of MS, is driven by mechanisms that appear to be independent from plaque activity. In this context, we previously reported that, in the spinal cord of MS patients, periplaques cover large surfaces of partial demyelination characterized notably by a transforming growth factor beta (TGF-beta) molecular signature and a decreased expression of the oligodendrocyte gene NDRG1 (N-Myc downstream regulated 1). In the present work, we re-assessed a previously published RNA expression dataset in which brain periplaques were originally used as internal controls. When comparing the mRNA profiles obtained from brain periplaques with those derived from control normal white matter samples, we found that, irrespective of plaque activity, brain periplaques exhibited a TGF-beta molecular signature, an increased expression of TGFB2 (transforming growth factor beta 2) and a decreased expression of the oligodendrocyte genes NDRG1 (N-Myc downstream regulated 1) and MAG (myelin-associated glycoprotein). From these data obtained at the mRNA level, a survey of the human proteome allowed predicting a protein-protein interaction network linking TGFB2 to the down-regulation of both NDRG1 and MAG in brain periplaques. To further elucidate the role of NDRG1 in periplaque-associated partial demyelination, we then extracted the interaction network linking NDRG1 to proteins detected in human central myelin sheaths. We observed that such a network was highly significantly enriched in RNA-binding proteins that notably included several HNRNPs (heterogeneous nuclear ribonucleoproteins) involved in the post-transcriptional regulation of MAG. We conclude that both brain and spinal cord periplaques host a chronic process of tissue remodeling, during which oligodendrocyte myelinating functions are altered. Our findings further suggest that TGFB2 may fuel such a process. Overall, the present work provides additional evidence that periplaque-associated partial demyelination may drive the silent progression observed in a subset of MS patients.


Asunto(s)
Esclerosis Múltiple , Factor de Crecimiento Transformador beta , Humanos , Encéfalo/metabolismo , Esclerosis Múltiple/genética , Esclerosis Múltiple/metabolismo , Vaina de Mielina/genética , Vaina de Mielina/metabolismo , Recurrencia Local de Neoplasia/metabolismo , Placa Amiloide/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
3.
Stem Cell Res ; 60: 102723, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35247845

RESUMEN

In adult testis, the cell mobility is essential for spermatogonia differentiation and is suspected to regulate spermatogonial stem cell fate. Netrin-1 controls cell migration and/or survival according to the cellular context. Its involvement in some self-renewing lineages raises the possibility that Netrin-1 could have a role in spermatogenesis. We show that in addition to Sertoli cells, a fraction of murine undifferentiated spermatogonia express the Netrin-1 receptor UNC5c and that UNC5c contributes to spermatogonia differentiation. Receptor loss in Unc5crcm males leads to the concomitant accumulation of transit-amplifying progenitors and short syncytia of spermatogonia. Without altering cell death rates, the consequences of Unc5c loss worsen with age: the increase in quiescent undifferentiated progenitors associated with a higher spermatogonial stem cell enriched subset leads to the spermatocyte I decline. We demonstrate in vitro that Netrin-1 promotes a guidance effect as it repulses both undifferentiated and differentiating spermatogonia. Finally, we propose that UNC5c triggers undifferentiated spermatogonia adhesion/ migration and that the repulsive activity of Netrin-1 receptors could regulate spermatogonia differentiation, and maintain germ cell homeostasis.


Asunto(s)
Espermatogénesis , Espermatogonias , Animales , Diferenciación Celular/fisiología , Homeostasis , Masculino , Ratones , Receptores de Netrina/metabolismo , Netrina-1/metabolismo , Espermatogénesis/fisiología , Testículo
4.
Int J Mol Sci ; 22(19)2021 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-34638785

RESUMEN

For a yet unknown reason, a substantial share of patients suffering from COVID-19 develop long-lasting neuropsychiatric symptoms ranging from cognitive deficits to mood disorders and/or an extreme fatigue. We previously reported that in non-neural cells, angiotensin-1 converting enzyme 2 (ACE2), the gene coding for the SARS-CoV2 host receptor, harbors tight co-expression links with dopa-decarboxylase (DDC), an enzyme involved in the metabolism of dopamine. Here, we mined and integrated data from distinct human expression atlases and found that, among a wide range of tissues and cells, enterocytes of the small intestine express the highest expression levels of ACE2, DDC and several key genes supporting the metabolism of neurotransmitters. Based on these results, we performed co-expression analyses on a recently published set of RNA-seq data obtained from SARS-CoV2-infected human intestinal organoids. We observed that in SARS-CoV2-infected enterocytes, ACE2 co-regulates not only with DDC but also with a specific group of genes involved in (i) the dopamine/trace amines metabolic pathway, (ii) the absorption of microbiota-derived L-DOPA and (iii) the absorption of neutral amino acids serving as precursors to neurotransmitters. We conclude that in patients with long COVID, a chronic infection and inflammation of small intestine enterocytes might be indirectly responsible for prolonged brain alterations.


Asunto(s)
Encéfalo/patología , COVID-19/complicaciones , Regulación de la Expresión Génica , Intestino Delgado/patología , Enzima Convertidora de Angiotensina 2/genética , Descarboxilasas de Aminoácido-L-Aromático/genética , Encéfalo/metabolismo , COVID-19/genética , COVID-19/patología , Células Cultivadas , Enterocitos/metabolismo , Enterocitos/patología , Humanos , Intestino Delgado/metabolismo , SARS-CoV-2/aislamiento & purificación , Síndrome Post Agudo de COVID-19
5.
Sci Rep ; 10(1): 20023, 2020 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-33208773

RESUMEN

Glial cells are early sensors of neuronal injury and can store lipids in lipid droplets under oxidative stress conditions. Here, we investigated the functions of the RNA-binding protein, SPEN/SHARP, in the context of Parkinson's disease (PD). Using a data-mining approach, we found that SPEN/SHARP is one of many astrocyte-expressed genes that are significantly differentially expressed in the substantia nigra of PD patients compared with control subjects. Interestingly, the differentially expressed genes are enriched in lipid metabolism-associated genes. In a Drosophila model of PD, we observed that flies carrying a loss-of-function allele of the ortholog split-ends (spen) or with glial cell-specific, but not neuronal-specific, spen knockdown were more sensitive to paraquat intoxication, indicating a protective role for Spen in glial cells. We also found that Spen is a positive regulator of Notch signaling in adult Drosophila glial cells. Moreover, Spen was required to limit abnormal accumulation of lipid droplets in glial cells in a manner independent of its regulation of Notch signaling. Taken together, our results demonstrate that Spen regulates lipid metabolism and storage in glial cells and contributes to glial cell-mediated neuroprotection.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Gotas Lipídicas/química , Neuroglía/citología , Paraquat/toxicidad , Enfermedad de Parkinson/prevención & control , Proteínas de Unión al ARN/metabolismo , Animales , Proteínas de Drosophila/genética , Drosophila melanogaster/efectos de los fármacos , Drosophila melanogaster/metabolismo , Herbicidas/toxicidad , Proteínas de Homeodominio/genética , Masculino , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Proteínas de Unión al ARN/genética
6.
Front Immunol ; 10: 2704, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31824497

RESUMEN

There is circumstantial evidence that, under neurodegenerative conditions, peptides deriving from aggregated or misfolded specific proteins elicit adaptive immune responses. On another hand, several genes involved in familial forms of neurodegenerative diseases exert key innate immune functions. However, whether or not such observations are causally linked remains unknown. To start addressing this issue, we followed a systems biology strategy based on the mining of large proteomics and immunopeptidomics databases. First, we retrieved the expression patterns of common neurodegeneration-associated proteins in two professional antigen-presenting cells, namely B lymphocytes and dendritic cells. Surprisingly, we found that under physiological conditions, numerous neurodegeneration-associated proteins are abundantly expressed by human B lymphocytes. A survey of the human proteome allowed us to map a unique protein-protein interaction network linking common neurodegeneration-associated proteins and their first shell interactors in human B lymphocytes. Interestingly, network connectivity analysis identified two major hubs that both relate with inflammation and autophagy, namely TRAF6 (TNF Receptor Associated Factor 6) and SQSTM1 (Sequestosome-1). Moreover, the mapped network in B lymphocytes comprised two additional hub proteins involved in both inflammation and autoimmunity: HSPA8 (Heat Shock Protein Family A Member 8 also known as HSC70) and HSP90AA1 (Heat Shock Protein 90 Alpha Family Class A Member 1). Based on these results, we then explored the Immune Epitope Database "IEDB-AR" and actually found that a large share of neurodegeneration-associated proteins were previously reported to provide endogenous MHC class II-binding peptides in human B lymphocytes. Of note, peptides deriving from amyloid beta A4 protein, sequestosome-1 or profilin-1 were reported to bind multiple allele-specific MHC class II molecules. In contrast, peptides deriving from microtubule-associated protein tau, presenilin 2 and serine/threonine-protein kinase TBK1 were exclusively reported to bind MHC molecules encoded by the HLA-DRB1 1501 allele, a recently-identified susceptibility gene for late onset Alzheimer's disease. Finally, we observed that the whole list of proteins reported to provide endogenous MHC class II-binding peptides in human B lymphocytes is specifically enriched in neurodegeneration-associated proteins. Overall, our work indicates that immunization against neurodegeneration-associated proteins might be a physiological process which is shaped, at least in part, by B lymphocytes.


Asunto(s)
Autofagia/inmunología , Linfocitos B/inmunología , Inflamación/inmunología , Péptidos y Proteínas de Señalización Intracelular/inmunología , Enfermedades Neurodegenerativas/inmunología , Proteína Sequestosoma-1/inmunología , Humanos , Biología de Sistemas
7.
Int J Mol Sci ; 20(23)2019 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-31779094

RESUMEN

In multiple sclerosis (MS) patients with a progressive form of the disease, spinal cord (SC) functions slowly deteriorate beyond age 40. We previously showed that in the SC of these patients, large areas of incomplete demyelination extend distance away from plaque borders and are characterized by a unique progliotic TGFB1 (Transforming Growth Factor Beta 1) genomic signature. Here, we attempted to determine whether region- and age-specific physiological parameters could promote the progression of SC periplaques in MS patients beyond age 40. An analysis of transcriptomics databases showed that, under physiological conditions, a set of 10 homeobox (HOX) genes are highly significantly overexpressed in the human SC as compared to distinct brain regions. Among these HOX genes, a survey of the human proteome showed that only HOXA5 encodes a protein which interacts with a member of the TGF-beta signaling pathway, namely SMAD1 (SMAD family member 1). Moreover, HOXA5 was previously found to promote the TGF-beta pathway. Interestingly, SMAD1 is also a protein partner of the androgen receptor (AR) and an unsupervised analysis of gene ontology terms indicates that the AR pathway antagonizes the TGF-beta/SMAD pathway. Retrieval of promoter analysis data further confirmed that AR negatively regulates the transcription of several members of the TGF-beta/SMAD pathway. On this basis, we propose that in progressive MS patients, the physiological SC overexpression of HOXA5 combined with the age-dependent decline in AR ligands may favor the slow progression of TGFB1-mediated gliosis. Potential therapeutic implications are discussed.


Asunto(s)
Envejecimiento/metabolismo , Gliosis/genética , Proteínas de Homeodominio/genética , Esclerosis Múltiple/genética , Médula Espinal/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Regulación hacia Arriba , Factores de Edad , Anciano , Encéfalo/metabolismo , Minería de Datos , Bases de Datos Genéticas , Progresión de la Enfermedad , Femenino , Perfilación de la Expresión Génica/métodos , Gliosis/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Ligandos , Masculino , Persona de Mediana Edad , Esclerosis Múltiple/metabolismo , Proteómica/métodos , Receptores Androgénicos/metabolismo , Análisis de Secuencia de ARN , Transducción de Señal , Proteína Smad1/metabolismo
8.
Mol Cell Proteomics ; 17(6): 1126-1143, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29531019

RESUMEN

High grade gliomas are the most common brain tumors in adult. These tumors are characterized by a high infiltration in microglial cells and macrophages. The immunosuppressive tumor environment is known to orient immune cells toward a pro-tumoral and anti-inflammatory phenotype. Therefore, the current challenge for cancer therapy is to find a way to reorient macrophages toward an antitumoral phenotype. Previously, we demonstrated that macrophages secreted antitumoral factors when they were invalidated for the proprotein converstase 1/3 (PC1/3) and treated with LPS. However, achieving an activation of macrophages via LPS/TLR4/Myd88-dependent pathway appears yet unfeasible in cancer patients. On the contrary, the antitumor drug Paclitaxel is also known to activate the TLR4 MyD88-dependent signaling pathway and mimics LPS action. Therefore, we evaluated if PC1/3 knock-down (KD) macrophages could be activated by Paclitaxel and efficient against glioma. We report here that such a treatment of PC1/3 KD macrophages drove to the overexpression of proteins mainly involved in cytoskeleton rearrangement. In support of this finding, we found that these cells exhibited a Ca2+ increase after Paclitaxel treatment. This is indicative of a possible depolymerization of microtubules and may therefore reflect an activation of inflammatory pathways in macrophages. In such a way, we found that PC1/3 KD macrophages displayed a repression of the anti-inflammatory pathway STAT3 and secreted more pro-inflammatory cytokines. Extracellular vesicles isolated from these PC1/3 KD cells inhibited glioma growth. Finally, the supernatant collected from the coculture between glioma cells and PC1/3 KD macrophages contained more antitumoral factors. These findings unravel the potential value of a new therapeutic strategy combining Paclitaxel and PC1/3 inhibition to switch macrophages toward an antitumoral immunophenotype.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Neoplasias Encefálicas/terapia , Glioma/terapia , Paclitaxel/farmacología , Proproteína Convertasa 1/genética , Animales , Neoplasias Encefálicas/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Técnicas de Cocultivo , Citocinas/metabolismo , Glioma/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Proteómica , Ratas
9.
Int J Mol Sci ; 18(10)2017 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-28981455

RESUMEN

We previously reported that, in multiple sclerosis (MS) patients with a progressive form of the disease, spinal cord periplaques extend distance away from plaque borders and are characterized by the co-occurrence of partial demyelination, astrocytosis and low-grade inflammation. However, transcriptomic analyses did not allow providing a comprehensive view of molecular events in astrocytes vs. oligodendrocytes. Here, we re-assessed our transcriptomic data and performed co-expression analyses to characterize astrocyte vs. oligodendrocyte molecular signatures in periplaques. We identified an astrocytosis-related co-expression module whose central hub was the astrocyte gene Cx43/GJA1 (connexin-43, also named gap junction protein α-1). Such a module comprised GFAP (glial fibrillary acidic protein) and a unique set of transcripts forming a TGFB/SMAD1/SMAD2 (transforming growth factor ß/SMAD family member 1/SMAD family member 2) genomic signature. Partial demyelination was characterized by a co-expression network whose central hub was the oligodendrocyte gene NDRG1 (N-myc downstream regulated 1), a gene previously shown to be specifically silenced in the normal-appearing white matter (NAWM) of MS patients. Surprisingly, besides myelin genes, the NDRG1 co-expression module comprised a highly significant number of translation/elongation-related genes. To identify a putative cause of NDRG1 downregulation in periplaques, we then sought to identify the cytokine/chemokine genes whose mRNA levels inversely correlated with those of NDRG1. Following this approach, we found five candidate immune-related genes whose upregulation associated with NDRG1 downregulation: TGFB1(transforming growth factor ß 1), PDGFC (platelet derived growth factor C), IL17D (interleukin 17D), IL33 (interleukin 33), and IL12A (interleukin 12A). From these results, we propose that, in the spinal cord periplaques of progressive MS patients, TGFB1 may limit acute inflammation but concurrently induce astrocytosis and an alteration of the translation/elongation of myelin genes in oligodendrocytes.


Asunto(s)
Inflamación/metabolismo , Esclerosis Múltiple/metabolismo , Médula Espinal/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Astrocitos/metabolismo , Proteínas de Ciclo Celular/metabolismo , Biología Computacional , Conexina 43/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Interleucina-12/metabolismo , Subunidad p35 de la Interleucina-12/metabolismo , Interleucina-17/metabolismo , Interleucina-33/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Linfocinas/metabolismo , Vaina de Mielina/metabolismo , Oligodendroglía/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo
10.
Mol Cell Proteomics ; 16(8): 1394-1415, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28659490

RESUMEN

The therapeutic use of RhoA inhibitors (RhoAi) has been experimentally tested in spinal cord injury (SCI). In order to decipher the underlying molecular mechanisms involved in such a process, an in vitro neuroproteomic-systems biology platform was developed in which the pan-proteomic profile of the dorsal root ganglia (DRG) cell line ND7/23 DRG was assessed in a large array of culture conditions using RhoAi and/or conditioned media obtained from SCI ex vivo derived spinal cord slices. A fine mapping of the spatio-temporal molecular events of the RhoAi treatment in SCI was performed. The data obtained allow a better understanding of regeneration/degeneration induced above and below the lesion site. Results notably showed a time-dependent alteration of the transcription factors profile along with the synthesis of growth cone-related factors (receptors, ligands, and signaling pathways) in RhoAi treated DRG cells. Furthermore, we assessed in a rat SCI model the in vivo impact of RhoAi treatment administered in situ via alginate scaffold that was combined with FK506 delivery. The improved recovery of locomotion was detected only at the early postinjury time points, whereas after overall survival a dramatic increase of synaptic contacts on outgrowing neurites in affected segments was observed. We validate these results by in vivo proteomic studies along the spinal cord segments from tissue and secreted media analyses, confirming the increase of the synaptogenesis expression factors under RhoAi treatment. Taken together, we demonstrate that RhoAi treatment seems to be useful to stimulate neurite outgrowth in both in vitro as well in vivo environments. However, for in vivo experiments there is a need for sustained delivery regiment to facilitate axon regeneration and promote synaptic reconnections with appropriate target neurons also at chronic phase, which in turn may lead to higher assumption for functional improvement.


Asunto(s)
Axones/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Proyección Neuronal/efectos de los fármacos , Traumatismos de la Médula Espinal/tratamiento farmacológico , Vesículas Sinápticas/efectos de los fármacos , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Análisis de Varianza , Animales , Axones/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/uso terapéutico , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/fisiopatología , Locomoción/efectos de los fármacos , Proyección Neuronal/fisiología , Proteómica , Ratas , Regeneración/efectos de los fármacos , Traumatismos de la Médula Espinal/fisiopatología , Vesículas Sinápticas/fisiología , Tacrolimus , Factores de Tiempo , Factores de Transcripción/metabolismo
11.
Mol Nutr Food Res ; 60(12): 2691-2699, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27468128

RESUMEN

SCOPE: The aim of the study was to assess the effects of a high-fructose diet (HFrD) on skeletal muscle transcriptomic response in healthy offspring of patients with type 2 diabetes, a subgroup of individuals prone to metabolic disorders. METHODS AND RESULTS: Ten healthy normal weight first-degree relatives of type 2 diabetic patients were submitted to a HFrD (+3.5 g fructose/kg fat-free mass per day) during 7 days. A global transcriptomic analysis was performed on skeletal muscle biopsies combined with in vitro experiments using primary myotubes. Transcriptomic analysis highlighted profound effects on fatty acid oxidation and mitochondrial pathways supporting the whole-body metabolic shift with the preferential use of carbohydrates instead of lipids. Bioinformatics tools pointed out possible transcription factors orchestrating this genomic regulation, such as PPARα and NR4A2. In vitro experiments in human myotubes suggested an indirect action of fructose in skeletal muscle, which seemed to be independent from lactate, uric acid, or nitric oxide. CONCLUSION: This study shows therefore that a large cluster of genes related to energy metabolism, mitochondrial function, and lipid oxidation was downregulated after 7 days of HFrD, thus supporting the concept that overconsumption of fructose-containing foods could contribute to metabolic deterioration in humans.


Asunto(s)
Diabetes Mellitus Tipo 2/sangre , Fructosa/administración & dosificación , Fructosa/efectos adversos , Mitocondrias/efectos de los fármacos , Fibras Musculares Esqueléticas/efectos de los fármacos , Adulto , Línea Celular , Estudios Cruzados , Dieta , Metabolismo Energético , Perfilación de la Expresión Génica , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Mitocondrias/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , PPAR alfa/genética , PPAR alfa/metabolismo , Transcriptoma , Adulto Joven
12.
PLoS One ; 11(7): e0158420, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27441662

RESUMEN

Cerebral malaria (CM) caused by Plasmodium falciparum parasites often leads to the death of infected patients or to persisting neurological sequelae despite anti-parasitic treatments. Erythropoietin (EPO) was recently suggested as a potential adjunctive treatment for CM. However diverging results were obtained in patients from Sub-Saharan countries infected with P. falciparum. In this study, we measured EPO levels in the plasma of well-defined groups of P. falciparum-infected patients, from the state of Odisha in India, with mild malaria (MM), CM, or severe non-CM (NCM). EPO levels were then correlated with biological parameters, including parasite biomass, heme, tumor necrosis factor (TNF)-α, interleukin (IL)-10, interferon gamma-induced protein (IP)-10, and monocyte chemoattractant protein (MCP)-1 plasma concentrations by Spearman's rank and multiple correlation analyses. We found a significant increase in EPO levels with malaria severity degree, and more specifically during fatal CM. In addition, EPO levels were also found correlated positively with heme, TNF-α, IL-10, IP-10 and MCP-1 during CM. We also found a significant multivariate correlation between EPO, TNF-α, IL-10, IP-10 MCP-1 and heme, suggesting an association of EPO with a network of immune factors in CM patients. The contradictory levels of circulating EPO reported in CM patients in India when compared to Africa highlights the need for the optimization of adjunctive treatments according to the targeted population.


Asunto(s)
Eritropoyetina/sangre , Hemo/metabolismo , Interleucina-10/sangre , Malaria Cerebral/sangre , Factor de Necrosis Tumoral alfa/sangre , Adulto , Antígenos de Protozoos/metabolismo , Quimiocina CCL2/sangre , Femenino , Hemopexina/metabolismo , Humanos , India , Malaria Cerebral/parasitología , Masculino , Plasmodium falciparum/fisiología , Proteínas Protozoarias/metabolismo , Índice de Severidad de la Enfermedad , Adulto Joven
13.
Mol Cell Proteomics ; 15(8): 2641-70, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27250205

RESUMEN

Spinal cord injury (SCI) represents a major debilitating health issue with a direct socioeconomic burden on the public and private sectors worldwide. Although several studies have been conducted to identify the molecular progression of injury sequel due from the lesion site, still the exact underlying mechanisms and pathways of injury development have not been fully elucidated. In this work, based on OMICs, 3D matrix-assisted laser desorption ionization (MALDI) imaging, cytokines arrays, confocal imaging we established for the first time that molecular and cellular processes occurring after SCI are altered between the lesion proximity, i.e. rostral and caudal segments nearby the lesion (R1-C1) whereas segments distant from R1-C1, i.e. R2-C2 and R3-C3 levels coexpressed factors implicated in neurogenesis. Delay in T regulators recruitment between R1 and C1 favor discrepancies between the two segments. This is also reinforced by presence of neurites outgrowth inhibitors in C1, absent in R1. Moreover, the presence of immunoglobulins (IgGs) in neurons at the lesion site at 3 days, validated by mass spectrometry, may present additional factor that contributes to limited regeneration. Treatment in vivo with anti-CD20 one hour after SCI did not improve locomotor function and decrease IgG expression. These results open the door of a novel view of the SCI treatment by considering the C1 as the therapeutic target.


Asunto(s)
Biomarcadores/metabolismo , Citocinas/metabolismo , Proteómica/métodos , Traumatismos de la Médula Espinal/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Análisis por Matrices de Proteínas , Mapas de Interacción de Proteínas , Ratas , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Factores de Tiempo
15.
Mol Cell Proteomics ; 14(11): 2857-77, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26330543

RESUMEN

Macrophages provide the first line of host immune defense. Their activation triggers the secretion of pro-inflammatory cytokines and chemokines recruiting other immune cells. In cancer, macrophages present an M2 anti-inflammatory phenotype promoting tumor growth. In this way, strategies need to be develop to reactivate macrophages. Previously thought to be expressed only in cells with a neural/neuroendocrine phenotype, the proprotein convertase 1/3 has been shown to also be expressed in macrophages and regulated as a function of the Toll-like receptor immune response. Here, we investigated the intracellular impact of the down-regulation of the proprotein convertase 1/3 in NR8383 macrophages and confirmed the results on macrophages from PC1/3 deficient mice. A complete proteomic study of secretomes and intracellular proteins was undertaken and revealed that inhibition of proprotein convertase 1/3 orient macrophages toward an M1 activated phenotype. This phenotype is characterized by filopodial extensions, Toll-like receptor 4 MyD88-dependent signaling, calcium entry augmentation and the secretion of pro-inflammatory factors. In response to endotoxin/lipopolysaccharide, these intracellular modifications increased, and the secreted factors attracted naïve T helper lymphocytes to promote the cytotoxic response. Importantly, the application of these factors onto breast and ovarian cancer cells resulted in a decrease viability or resistance. Under inhibitory conditions using interleukin 10, PC1/3-knockdown macrophages continued to secrete inflammatory factors. These data indicate that targeted inhibition of proprotein convertase 1/3 could represent a novel type of immune therapy to reactivate intra-tumoral macrophages.


Asunto(s)
Inmunoterapia/métodos , Macrófagos Alveolares/inmunología , Macrófagos Peritoneales/inmunología , Proproteína Convertasa 1/antagonistas & inhibidores , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Calcio/metabolismo , Línea Celular , Línea Celular Tumoral , Citocinas/biosíntesis , Citocinas/inmunología , Regulación de la Expresión Génica , Humanos , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/patología , Lipopolisacáridos/farmacología , Macrófagos Alveolares/citología , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Peritoneales/citología , Macrófagos Peritoneales/efectos de los fármacos , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Cultivo Primario de Células , Proproteína Convertasa 1/genética , Proproteína Convertasa 1/inmunología , Análisis por Matrices de Proteínas , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Transducción de Señal , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/patología , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología
16.
J Biol Chem ; 287(35): 30014-23, 2012 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-22782894

RESUMEN

The multifunctional protein netrin-1 was initially discovered as the main attractive cue for commissural axon guidance by acting through its receptor DCC. Recently, we have shown that netrin-1 also interacts with the orphan transmembrane receptor amyloid precursor protein (APP). APP is cleaved by proteases, generating amyloid-ß peptide, the main component of the amyloid plaques that are associated with Alzheimer disease. Our previous work demonstrated that via its interaction with APP, netrin-1 is a negative regulator of amyloid-ß production in adult brain, but the biological relevance of APP/netrin-1 interaction under non-pathological conditions was unknown. We show here that during commissural axon navigation, APP, expressed at the growth cone, is part of the DCC receptor complex mediating netrin-1-dependent axon guidance. APP interacts with DCC in the presence of netrin-1 and enhances netrin-1-mediated DCC intracellular signaling, such as MAPK activation. Inactivation of APP in mice is associated with reduced commissural axon outgrowth. Thus, APP functionally acts as a co-receptor for DCC to mediate axon guidance.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Conos de Crecimiento/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Proteolisis , Proteínas Supresoras de Tumor/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Receptor DCC , Conos de Crecimiento/patología , Ratones , Ratones Mutantes , Factores de Crecimiento Nervioso/genética , Netrina-1 , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Proteínas Supresoras de Tumor/genética
17.
Lab Invest ; 91(12): 1717-26, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21876536

RESUMEN

Netrin-1, a multifunctional laminin-related protein is widely expressed in various tissues, including kidney. The pathophysiological roles of netrin-1 in toxic acute kidney injury are unknown. To determine the role of netrin-1 in cisplatin-induced nephrotoxicity, we used netrin-1 transgenic mice that overexpress netrin-1 in the proximal tubular epithelium using the fatty acid binding protein promoter. Administration of cisplatin caused severe renal injury in WT mice but not in netrin-1 transgenic mice. Functional improvement was associated with better preservation of morphology, reduced cytokine expression and oxidative stress in the kidney, and reduced serum and urine cytokine and chemokine levels of transgenic mice as compared with WT mice. Cisplatin induced an increase in neutrophil infiltration into the kidney of WT mice, which was not significantly reduced in netrin-1 transgenic mice. Interestingly, ischemia reperfusion induced a large increase in apoptosis in WT mice but not in netrin-1 transgenic mice (215 ± 40 vs 94 ± 20 cells/5 HPF ( × 400), P < 0.0001), which was associated with reduced caspase-3 and p53 activation in the transgenic kidney. These results suggest that netrin-1 protects renal tubular epithelial cells against cisplatin-induced kidney injury by suppressing apoptosis and inflammation.


Asunto(s)
Lesión Renal Aguda/inducido químicamente , Antineoplásicos/efectos adversos , Cisplatino/efectos adversos , Túbulos Renales Proximales/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Lesión Renal Aguda/metabolismo , Animales , Apoptosis/efectos de los fármacos , Biomarcadores/metabolismo , Moléculas de Adhesión Celular/metabolismo , Células Cultivadas , Citocinas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Netrina-1 , Infiltración Neutrófila , Estrés Oxidativo , Proteína p53 Supresora de Tumor/metabolismo
18.
Am J Pathol ; 175(3): 1010-8, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19700747

RESUMEN

Netrin-1, a diffusible laminin-related protein, is highly expressed in the kidney. However, the pathophysiological roles of netrin-1 in the kidney are unknown. To address this question directly, we used transgenic mice that overexpress chicken netrin-1 in the kidney. Netrin-1 overexpression was confirmed by real-time RT-PCR and Western blot analysis. Eight-week-old wild-type and transgenic mice were subjected to 26 minutes of renal ischemia followed by reperfusion for 72 hours. Wild-type mice developed more severe renal dysfunction by 24 hours than netrin-1 transgenic mice. Functional improvement was associated with better preservation of morphology, reduced cytokine expression, and reduced oxidative stress in the kidney of transgenic mice as compared with wild-type mice. In addition, both basal and reperfusion-induced cell proliferation were dramatically increased in transgenic kidneys as determined by Ki-67 staining. Interestingly, ischemia reperfusion induced a large increase in apoptosis in wild-type mice but not in netrin-1 transgenic mice that was associated with reduced caspase-3 activation in the transgenic kidney. These results suggest that netrin-1 protects renal tubular epithelial cells against ischemia reperfusion-induced injury by increasing proliferation and suppressing apoptosis.


Asunto(s)
Apoptosis , Riñón/metabolismo , Factores de Crecimiento Nervioso/biosíntesis , Daño por Reperfusión/metabolismo , Proteínas Supresoras de Tumor/biosíntesis , Animales , Proliferación Celular , Pollos , Regulación de la Expresión Génica , Riñón/irrigación sanguínea , Riñón/patología , Ratones , Ratones Transgénicos , Factores de Crecimiento Nervioso/genética , Netrina-1 , Daño por Reperfusión/patología , Daño por Reperfusión/prevención & control , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Supresoras de Tumor/genética
19.
Biochim Biophys Acta ; 1793(2): 231-8, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18948148

RESUMEN

Eph receptors have been implicated in regulating a diverse array of cellular functions in the developing nervous system. Recently, Eph receptors have been shown to promote cell death in adult germinal zones; however, their mechanisms of action remain ill-defined. In this study, we demonstrate that EphA4 is a new member of the dependence receptors family, which can initiate cell death in the absence of its ligand ephrinB3. Upon removal of its ligand, EphA4 triggers cell death that is dependent on caspase activation as caspase inhibitors prevent cell death. EphA4 itself is cleaved by caspase-3-like caspase in the intracellular domain at position D773/774, which is necessary for cell death initiation as mutation of the cleavage site abolishes apoptosis. In the adult subventricular zone, abolishing ephrinB3 results in increased cell death, while the absence of EphA4 results in excessive numbers of neuroblasts. Furthermore, infusion of soluble ephrinB3 into the lateral ventricle reduced cell death, and together these results support a dependence role for EphA4 in adult neurogenesis.


Asunto(s)
Apoptosis/efectos de los fármacos , Efrina-B3/farmacología , Neurogénesis/efectos de los fármacos , Receptor EphA4/metabolismo , Animales , Caspasa 3/metabolismo , Línea Celular , Ventrículos Cerebrales/efectos de los fármacos , Ventrículos Cerebrales/enzimología , Ventrículos Cerebrales/patología , Activación Enzimática/efectos de los fármacos , Humanos , Ligandos , Masculino , Ratones , Fosforilación/efectos de los fármacos , Células Madre/citología , Células Madre/efectos de los fármacos , Células Madre/enzimología , Especificidad por Sustrato/efectos de los fármacos
20.
EMBO J ; 24(6): 1192-201, 2005 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-15729359

RESUMEN

Netrin-1 receptors UNC5H (UNC5H1-4) were originally proposed to mediate the chemorepulsive activity of netrin-1 during axonal guidance processes. However, UNC5H receptors were more recently described as dependence receptors and, as such, able to trigger apoptosis in the absence of netrin-1. They were also proposed as putative tumor suppressors. Here, we show that UNC5H2 physically interacts with the serine/threonine kinase death-associated protein kinase (DAP-kinase) both in cell culture and in embryonic mouse brains. This interaction occurs in part through the respective death domains of UNC5H2 and DAP-kinase. Moreover, part of UNC5H2 proapoptotic activity occurs through this interaction because UNC5H2-induced cell death is partly impaired in the presence of dominant-negative mutants of DAP-kinase or in DAP-kinase mutant murine embryonic fibroblast cells. In the absence of netrin-1, UNC5H2 reduces DAP-kinase autophosphorylation on Ser308 and increases the catalytic activity of the kinase while netrin-1 blocks UNC5H2-dependent DAP-kinase activation. Thus, the pair netrin-1/UNC5H2 may regulate cell fate by controlling the proapoptotic kinase activity of DAP-kinase.


Asunto(s)
Apoptosis/fisiología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Factores de Crecimiento Nervioso/fisiología , Receptores de Superficie Celular/fisiología , Proteínas Supresoras de Tumor/fisiología , Secuencia de Aminoácidos , Animales , Proteínas Reguladoras de la Apoptosis , Encéfalo/citología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/genética , Caspasas/metabolismo , Catálisis , Células Cultivadas , Proteínas Quinasas Asociadas a Muerte Celular , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Inmunoprecipitación , Ratones , Datos de Secuencia Molecular , Factores de Crecimiento Nervioso/metabolismo , Factores de Crecimiento Nervioso/farmacología , Receptores de Netrina , Netrina-1 , Fosforilación/efectos de los fármacos , Estructura Terciaria de Proteína , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Activación Transcripcional , Proteínas Supresoras de Tumor/metabolismo , Proteínas Supresoras de Tumor/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA