Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Cell ; 174(6): 1477-1491.e19, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-30146158

RESUMEN

Aging is a major risk factor for both genetic and sporadic neurodegenerative disorders. However, it is unclear how aging interacts with genetic predispositions to promote neurodegeneration. Here, we investigate how partial loss of function of TBK1, a major genetic cause for amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) comorbidity, leads to age-dependent neurodegeneration. We show that TBK1 is an endogenous inhibitor of RIPK1 and the embryonic lethality of Tbk1-/- mice is dependent on RIPK1 kinase activity. In aging human brains, another endogenous RIPK1 inhibitor, TAK1, exhibits a marked decrease in expression. We show that in Tbk1+/- mice, the reduced myeloid TAK1 expression promotes all the key hallmarks of ALS/FTD, including neuroinflammation, TDP-43 aggregation, axonal degeneration, neuronal loss, and behavior deficits, which are blocked upon inhibition of RIPK1. Thus, aging facilitates RIPK1 activation by reducing TAK1 expression, which cooperates with genetic risk factors to promote the onset of ALS/FTD.


Asunto(s)
Apoptosis , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Adulto , Anciano , Envejecimiento , Animales , Apoptosis/efectos de los fármacos , Axones/metabolismo , Conducta Animal , Encéfalo/citología , Encéfalo/metabolismo , Células Cultivadas , Humanos , Quinasa I-kappa B/metabolismo , Ratones , Ratones Noqueados , Microglía/citología , Microglía/efectos de los fármacos , Microglía/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/deficiencia , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Médula Espinal/metabolismo , Estaurosporina/farmacología , Factor de Necrosis Tumoral alfa/farmacología
2.
Mol Cell ; 81(2): 370-385.e7, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33271062

RESUMEN

The mechanisms of cellular energy sensing and AMPK-mediated mTORC1 inhibition are not fully delineated. Here, we discover that RIPK1 promotes mTORC1 inhibition during energetic stress. RIPK1 is involved in mediating the interaction between AMPK and TSC2 and facilitate TSC2 phosphorylation at Ser1387. RIPK1 loss results in a high basal mTORC1 activity that drives defective lysosomes in cells and mice, leading to accumulation of RIPK3 and CASP8 and sensitization to cell death. RIPK1-deficient cells are unable to cope with energetic stress and are vulnerable to low glucose levels and metformin. Inhibition of mTORC1 rescues the lysosomal defects and vulnerability to energetic stress and prolongs the survival of RIPK1-deficient neonatal mice. Thus, RIPK1 plays an important role in the cellular response to low energy levels and mediates AMPK-mTORC1 signaling. These findings shed light on the regulation of mTORC1 during energetic stress and unveil a point of crosstalk between pro-survival and pro-death pathways.


Asunto(s)
Proteína 5 Relacionada con la Autofagia/genética , Proteína de Dominio de Muerte Asociada a Fas/genética , Intestino Grueso/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Animales Recién Nacidos , Proteína 5 Relacionada con la Autofagia/deficiencia , Caspasa 8/genética , Caspasa 8/metabolismo , Muerte Celular/genética , Proteína de Dominio de Muerte Asociada a Fas/deficiencia , Regulación de la Expresión Génica , Glucosa/antagonistas & inhibidores , Glucosa/farmacología , Células HEK293 , Células HT29 , Humanos , Intestino Grueso/efectos de los fármacos , Intestino Grueso/patología , Células Jurkat , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Lisosomas/patología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Metformina/antagonistas & inhibidores , Metformina/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Proteína Serina-Treonina Quinasas de Interacción con Receptores/deficiencia , Transducción de Señal , Sirolimus/farmacología , Proteína 2 del Complejo de la Esclerosis Tuberosa/genética , Proteína 2 del Complejo de la Esclerosis Tuberosa/metabolismo
3.
Nature ; 587(7832): 133-138, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32968279

RESUMEN

Cell death in human diseases is often a consequence of disrupted cellular homeostasis. If cell death is prevented without restoring cellular homeostasis, it may lead to a persistent dysfunctional and pathological state. Although mechanisms of cell death have been thoroughly investigated1-3, it remains unclear how homeostasis can be restored after inhibition of cell death. Here we identify TRADD4-6, an adaptor protein, as a direct regulator of both cellular homeostasis and apoptosis. TRADD modulates cellular homeostasis by inhibiting K63-linked ubiquitination of beclin 1 mediated by TRAF2, cIAP1 and cIAP2, thereby reducing autophagy. TRADD deficiency inhibits RIPK1-dependent extrinsic apoptosis and proteasomal stress-induced intrinsic apoptosis. We also show that the small molecules ICCB-19 and Apt-1 bind to a pocket on the N-terminal TRAF2-binding domain of TRADD (TRADD-N), which interacts with the C-terminal domain (TRADD-C) and TRAF2 to modulate the ubiquitination of RIPK1 and beclin 1. Inhibition of TRADD by ICCB-19 or Apt-1 blocks apoptosis and restores cellular homeostasis by activating autophagy in cells with accumulated mutant tau, α-synuclein, or huntingtin. Treatment with Apt-1 restored proteostasis and inhibited cell death in a mouse model of proteinopathy induced by mutant tau(P301S). We conclude that pharmacological targeting of TRADD may represent a promising strategy for inhibiting cell death and restoring homeostasis to treat human diseases.


Asunto(s)
Apoptosis/efectos de los fármacos , Homeostasis/efectos de los fármacos , Proteína de Dominio de Muerte Asociada a Receptor de TNF/antagonistas & inhibidores , Proteína de Dominio de Muerte Asociada a Receptor de TNF/metabolismo , Animales , Autofagia/efectos de los fármacos , Proteína 3 que Contiene Repeticiones IAP de Baculovirus/metabolismo , Beclina-1/química , Beclina-1/metabolismo , Bortezomib/antagonistas & inhibidores , Bortezomib/farmacología , Línea Celular , Humanos , Proteína Huntingtina/metabolismo , Proteínas Inhibidoras de la Apoptosis/metabolismo , Masculino , Ratones , Modelos Moleculares , Ovillos Neurofibrilares/metabolismo , Proteoma/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/química , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Proteína de Dominio de Muerte Asociada a Receptor de TNF/química , Proteína de Dominio de Muerte Asociada a Receptor de TNF/deficiencia , Factor 2 Asociado a Receptor de TNF/metabolismo , Ubiquitinación , alfa-Sinucleína/metabolismo , Proteínas tau/metabolismo
4.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-33766915

RESUMEN

Microglial-derived inflammation has been linked to a broad range of neurodegenerative and neuropsychiatric conditions, including amyotrophic lateral sclerosis (ALS). Using single-cell RNA sequencing, a class of Disease-Associated Microglia (DAMs) have been characterized in neurodegeneration. However, the DAM phenotype alone is insufficient to explain the functional complexity of microglia, particularly with regard to regulating inflammation that is a hallmark of many neurodegenerative diseases. Here, we identify a subclass of microglia in mouse models of ALS which we term RIPK1-Regulated Inflammatory Microglia (RRIMs). RRIMs show significant up-regulation of classical proinflammatory pathways, including increased levels of Tnf and Il1b RNA and protein. We find that RRIMs are highly regulated by TNFα signaling and that the prevalence of these microglia can be suppressed by inhibiting receptor-interacting protein kinase 1 (RIPK1) activity downstream of the TNF receptor 1. These findings help to elucidate a mechanism by which RIPK1 kinase inhibition has been shown to provide therapeutic benefit in mouse models of ALS and may provide an additional biomarker for analysis in ongoing phase 2 clinical trials of RIPK1 inhibitors in ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/enzimología , Inflamación/enzimología , Microglía/enzimología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Proteínas de Ciclo Celular/genética , Modelos Animales de Enfermedad , Interleucina-1beta/metabolismo , Proteínas de Transporte de Membrana/genética , Ratones , Ratones Mutantes , Microglía/patología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/antagonistas & inhibidores , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Análisis de la Célula Individual , Superóxido Dismutasa-1/genética , Transcriptoma , Factor de Necrosis Tumoral alfa/metabolismo
5.
Nucleic Acids Res ; 49(10): e58, 2021 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-33693773

RESUMEN

We present barcoded oligonucleotides ligated on RNA amplified for multiplexed and parallel insitu analyses (BOLORAMIS), a reverse transcription-free method for spatially-resolved, targeted, in situ RNA identification of single or multiple targets. BOLORAMIS was demonstrated on a range of cell types and human cerebral organoids. Singleplex experiments to detect coding and non-coding RNAs in human iPSCs showed a stem-cell signature pattern. Specificity of BOLORAMIS was found to be 92% as illustrated by a clear distinction between human and mouse housekeeping genes in a co-culture system, as well as by recapitulation of subcellular localization of lncRNA MALAT1. Sensitivity of BOLORAMIS was quantified by comparing with single molecule FISH experiments and found to be 11%, 12% and 35% for GAPDH, TFRC and POLR2A, respectively. To demonstrate BOLORAMIS for multiplexed gene analysis, we targeted 96 mRNAs within a co-culture of iNGN neurons and HMC3 human microglial cells. We used fluorescence in situ sequencing to detect error-robust 8-base barcodes associated with each of these genes. We then used this data to uncover the spatial relationship among cells and transcripts by performing single-cell clustering and gene-gene proximity analyses. We anticipate the BOLORAMIS technology for in situ RNA detection to find applications in basic and translational research.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Hibridación Fluorescente in Situ/métodos , Oligonucleótidos/química , ARN/análisis , Análisis de la Célula Individual/métodos , Animales , Línea Celular , Humanos , Ratones
6.
Proc Natl Acad Sci U S A ; 114(41): E8788-E8797, 2017 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-28904096

RESUMEN

Dysfunction of microglia is known to play an important role in Alzheimer's disease (AD). Here, we investigated the role of RIPK1 in microglia mediating the pathogenesis of AD. RIPK1 is highly expressed by microglial cells in human AD brains. Using the amyloid precursor protein (APP)/presenilin 1 (PS1) transgenic mouse model, we found that inhibition of RIPK1, using both pharmacological and genetic means, reduced amyloid burden, the levels of inflammatory cytokines, and memory deficits. Furthermore, inhibition of RIPK1 promoted microglial degradation of Aß in vitro. We characterized the transcriptional profiles of adult microglia from APP/PS1 mice and identified a role for RIPK1 in regulating the microglial expression of CH25H and Cst7, a marker for disease-associated microglia (DAM), which encodes an endosomal/lysosomal cathepsin inhibitor named Cystatin F. We present evidence that RIPK1-mediated induction of Cst7 leads to an impairment in the lysosomal pathway. These data suggest that RIPK1 may mediate a critical checkpoint in the transition to the DAM state. Together, our study highlights a non-cell death mechanism by which the activation of RIPK1 mediates the induction of a DAM phenotype, including an inflammatory response and a reduction in phagocytic activity, and connects RIPK1-mediated transcription in microglia to the etiology of AD. Our results support that RIPK1 is an important therapeutic target for the treatment of AD.


Asunto(s)
Enfermedad de Alzheimer/patología , Biomarcadores/metabolismo , Microglía/patología , Presenilina-1/fisiología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/metabolismo , Fenotipo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética
7.
Cell Res ; 32(7): 621-637, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35661830

RESUMEN

RIPK1 is a master regulator of multiple cell death pathways, including apoptosis and necroptosis, and inflammation. Importantly, activation of RIPK1 has also been shown to promote the transcriptional induction of proinflammatory cytokines in cells undergoing necroptosis, in animal models of amyotrophic lateral sclerosis (ALS) and Alzheimer's disease (AD), and in human ALS and AD. Rare human genetic carriers of non-cleavable RIPK1 variants (D324V and D324H) exhibit distinct symptoms of recurrent fevers and increased transcription of proinflammatory cytokines. Multiple RIPK1 inhibitors have been advanced into human clinical trials as new therapeutics for human inflammatory and neurodegenerative diseases, such as ALS and AD. However, it is unclear whether and how RIPK1 kinase activity directly mediates inflammation independent of cell death as the nuclear function of RIPK1 has not yet been explored. Here we show that nuclear RIPK1 is physically associated with the BAF complex. Upon RIPK1 activation, the RIPK1/BAF complex is recruited by specific transcription factors to active enhancers and promoters marked by H3K4me1 and H3K27ac. Activated nuclear RIPK1 mediates the phosphorylation of SMARCC2, a key component of the BAF complex, to promote chromatin remodeling and the transcription of specific proinflammatory genes. Increased nuclear RIPK1 activation and RIPK1/BAF-mediated chromatin-remodeling activity were found in cells expressing non-cleavable RIPK1, and increased enrichment of activated RIPK1 on active enhancers and promoters was found in an animal model and human pathological samples of ALS. Our results suggest that RIPK1 kinase serves as a transcriptional coregulator in nucleus that can transmit extracellular stimuli to the BAF complex to modulate chromatin accessibility and directly regulate the transcription of specific genes involved in mediating inflammatory responses.


Asunto(s)
Esclerosis Amiotrófica Lateral , Esclerosis Amiotrófica Lateral/genética , Animales , Apoptosis , Cromatina , Ensamble y Desensamble de Cromatina , Citocinas/metabolismo , Inflamación/patología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo
8.
Int J Neuropsychopharmacol ; 14(8): 1111-26, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21138621

RESUMEN

The neuronal circuitry underlying stress- and drug-induced reinstatement of cocaine-seeking has been relatively well characterized; however, less is known regarding the long-term molecular changes following cocaine administration that may promote future reinstatement. The transcription factor cAMP response element-binding protein (CREB) is necessary for stress- but not cocaine-induced reinstatement of conditioned reward, suggesting that different molecular mechanisms may underlie these two types of reinstatement. To explore the relationship between this transcription factor and reinstatement, we utilized the place-conditioning paradigm to examine alterations in gene expression in the amygdala, a neural substrate critically involved in stress-induced reinstatement, following the development of cocaine reward and subsequent extinction. Our findings demonstrate that the amygdala transcriptome was altered by CREB deficiency more than by previous cocaine experience, with an over-representation of genes involved in the immune response. However, a subset of genes involved in stress and immune response demonstrated a drug×genotype interaction, indicating that cocaine produces different long-term alterations in gene expression depending on the presence or absence of CREB. This profile of gene expression in the context of addiction enhances our understanding of the long-term molecular changes that occur throughout the addiction cycle and identifies novel genes and pathways that might lead to the creation of better therapeutic agents.


Asunto(s)
Amígdala del Cerebelo/efectos de los fármacos , Conducta Adictiva/metabolismo , Cocaína/farmacología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Inhibidores de Captación de Dopamina/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Amígdala del Cerebelo/inmunología , Amígdala del Cerebelo/metabolismo , Amígdala del Cerebelo/patología , Animales , Conducta Adictiva/genética , Conducta Adictiva/inmunología , Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Cocaína/metabolismo , Condicionamiento Psicológico/efectos de los fármacos , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/efectos de los fármacos , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/inmunología , Inhibidores de Captación de Dopamina/metabolismo , Extinción Psicológica/efectos de los fármacos , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Terapia Molecular Dirigida , Recompensa , Factores de Transcripción/genética , Factores de Transcripción/inmunología , Transcriptoma
9.
Nat Commun ; 12(1): 4826, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34376696

RESUMEN

Loss-of-function mutations in NEK1 gene, which encodes a serine/threonine kinase, are involved in human developmental disorders and ALS. Here we show that NEK1 regulates retromer-mediated endosomal trafficking by phosphorylating VPS26B. NEK1 deficiency disrupts endosomal trafficking of plasma membrane proteins and cerebral proteome homeostasis to promote mitochondrial and lysosomal dysfunction and aggregation of α-synuclein. The metabolic and proteomic defects of NEK1 deficiency disrupts the integrity of blood-brain barrier (BBB) by promoting lysosomal degradation of A20, a key modulator of RIPK1, thus sensitizing cerebrovascular endothelial cells to RIPK1-dependent apoptosis and necroptosis. Genetic inactivation of RIPK1 or metabolic rescue with ketogenic diet can prevent postnatal lethality and BBB damage in NEK1 deficient mice. Inhibition of RIPK1 reduces neuroinflammation and aggregation of α-synuclein in the brains of NEK1 deficient mice. Our study identifies a molecular mechanism by which retromer trafficking and metabolism regulates cerebrovascular integrity, cerebral proteome homeostasis and RIPK1-mediated neuroinflammation.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Glucosa/metabolismo , Complejos Multiproteicos/metabolismo , Quinasa 1 Relacionada con NIMA/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Animales , Animales Recién Nacidos , Línea Celular , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Activación Enzimática , Células HEK293 , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/citología , Microglía/metabolismo , Quinasa 1 Relacionada con NIMA/genética , Necroptosis/genética , Fosforilación , Transporte de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
10.
Nat Rev Drug Discov ; 19(8): 553-571, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32669658

RESUMEN

Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) is a key mediator of cell death and inflammation. The unique hydrophobic pocket in the allosteric regulatory domain of RIPK1 has enabled the development of highly selective small-molecule inhibitors of its kinase activity, which have demonstrated safety in preclinical models and clinical trials. Potential applications of these RIPK1 inhibitors for the treatment of monogenic and polygenic autoimmune, inflammatory, neurodegenerative, ischaemic and acute conditions, such as sepsis, are emerging. This article reviews RIPK1 biology and disease-associated mutations in RIPK1 signalling pathways, highlighting clinical trials of RIPK1 inhibitors and potential strategies to mitigate development challenges.


Asunto(s)
Sistemas de Liberación de Medicamentos , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteína Serina-Treonina Quinasas de Interacción con Receptores/antagonistas & inhibidores , Animales , Humanos , Mutación , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Transducción de Señal/efectos de los fármacos
11.
Cell Rep ; 33(10): 108447, 2020 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-33296651

RESUMEN

The contribution and mechanism of cerebrovascular pathology in Alzheimer's disease (AD) pathogenesis are still unclear. Here, we show that venular and capillary cerebral endothelial cells (ECs) are selectively vulnerable to necroptosis in AD. We identify reduced cerebromicrovascular expression of murine N-acetyltransferase 1 (mNat1) in two AD mouse models and hNat2, the human ortholog of mNat1 and a genetic risk factor for type-2 diabetes and insulin resistance, in human AD. mNat1 deficiency in Nat1-/- mice and two AD mouse models promotes blood-brain barrier (BBB) damage and endothelial necroptosis. Decreased mNat1 expression induces lysosomal degradation of A20, an important regulator of necroptosis, and LRP1ß, a key component of LRP1 complex that exports Aß in cerebral ECs. Selective restoration of cerebral EC expression of mNAT1 delivered by adeno-associated virus (AAV) rescues cerebromicrovascular levels of A20 and LRP1ß, inhibits endothelial necroptosis and activation, ameliorates mitochondrial fragmentation, reduces Aß deposits, and improves cognitive function in the AD mouse model.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Arilamina N-Acetiltransferasa/metabolismo , Isoenzimas/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/metabolismo , Animales , Arilamina N-Acetiltransferasa/genética , Transporte Biológico/fisiología , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Proteínas de Ciclo Celular/metabolismo , Cerebro/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Femenino , Humanos , Isoenzimas/genética , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Necroptosis/fisiología , Fragmentos de Péptidos/metabolismo , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA