Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Neurosci ; 26(1): 12-26, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36536241

RESUMEN

Iron dysregulation has been implicated in multiple neurodegenerative diseases, including Parkinson's disease (PD). Iron-loaded microglia are frequently found in affected brain regions, but how iron accumulation influences microglia physiology and contributes to neurodegeneration is poorly understood. Here we show that human induced pluripotent stem cell-derived microglia grown in a tri-culture system are highly responsive to iron and susceptible to ferroptosis, an iron-dependent form of cell death. Furthermore, iron overload causes a marked shift in the microglial transcriptional state that overlaps with a transcriptomic signature found in PD postmortem brain microglia. Our data also show that this microglial response contributes to neurodegeneration, as removal of microglia from the tri-culture system substantially delayed iron-induced neurotoxicity. To elucidate the mechanisms regulating iron response in microglia, we performed a genome-wide CRISPR screen and identified novel regulators of ferroptosis, including the vesicle trafficking gene SEC24B. These data suggest a critical role for microglia iron overload and ferroptosis in neurodegeneration.


Asunto(s)
Ferroptosis , Células Madre Pluripotentes Inducidas , Sobrecarga de Hierro , Enfermedad de Parkinson , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Hierro/metabolismo , Sobrecarga de Hierro/metabolismo , Microglía/metabolismo , Enfermedad de Parkinson/genética
2.
Nat Neurosci ; 24(11): 1522-1533, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34675436

RESUMEN

Coronavirus disease 2019 (COVID-19) can damage cerebral small vessels and cause neurological symptoms. Here we describe structural changes in cerebral small vessels of patients with COVID-19 and elucidate potential mechanisms underlying the vascular pathology. In brains of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-infected individuals and animal models, we found an increased number of empty basement membrane tubes, so-called string vessels representing remnants of lost capillaries. We obtained evidence that brain endothelial cells are infected and that the main protease of SARS-CoV-2 (Mpro) cleaves NEMO, the essential modulator of nuclear factor-κB. By ablating NEMO, Mpro induces the death of human brain endothelial cells and the occurrence of string vessels in mice. Deletion of receptor-interacting protein kinase (RIPK) 3, a mediator of regulated cell death, blocks the vessel rarefaction and disruption of the blood-brain barrier due to NEMO ablation. Importantly, a pharmacological inhibitor of RIPK signaling prevented the Mpro-induced microvascular pathology. Our data suggest RIPK as a potential therapeutic target to treat the neuropathology of COVID-19.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Proteasas 3C de Coronavirus/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Microvasos/metabolismo , SARS-CoV-2/metabolismo , Animales , Barrera Hematoencefálica/patología , Encéfalo/patología , Chlorocebus aethiops , Proteasas 3C de Coronavirus/genética , Cricetinae , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Mesocricetus , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microvasos/patología , SARS-CoV-2/genética , Células Vero
3.
Cell Rep ; 35(6): 109112, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33979622

RESUMEN

Receptor interacting protein kinase 1 (RIPK1) mediates cell death and inflammatory signaling and is increased in multiple sclerosis (MS) brain samples. Here, we investigate the role of glial RIPK1 kinase activity in mediating MS pathogenesis. We demonstrate RIPK1 levels correlate with MS disease progression. We find microglia are susceptible to RIPK1-mediated cell death and identify an inflammatory gene signature that may contribute to the neuroinflammatory milieu in MS patients. We uncover a distinct role for RIPK1 in astrocytes in regulating inflammatory signaling in the absence of cell death and confirm RIPK1-kinase-dependent regulation in human glia. Using a murine MS model, we show RIPK1 inhibition attenuates disease progression and suppresses deleterious signaling in astrocytes and microglia. Our results suggest RIPK1 kinase activation in microglia and astrocytes induces a detrimental neuroinflammatory program that contributes to the neurodegenerative environment in progressive MS.


Asunto(s)
Microglía/metabolismo , Esclerosis Múltiple/genética , Enfermedades Neuroinflamatorias/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Ratones , Esclerosis Múltiple/patología , Transducción de Señal
4.
Cell Death Dis ; 11(10): 904, 2020 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-33097690

RESUMEN

Microglia serve as the innate immune cells of the central nervous system (CNS) by providing continuous surveillance of the CNS microenvironment and initiating defense mechanisms to protect CNS tissue. Upon injury, microglia transition into an activated state altering their transcriptional profile, transforming their morphology, and producing pro-inflammatory cytokines. These activated microglia initially serve a beneficial role, but their continued activation drives neuroinflammation and neurodegeneration. Multiple sclerosis (MS) is a chronic, inflammatory, demyelinating disease of the CNS, and activated microglia and macrophages play a significant role in mediating disease pathophysiology and progression. Colony-stimulating factor-1 receptor (CSF1R) and its ligand CSF1 are elevated in CNS tissue derived from MS patients. We performed a large-scale RNA-sequencing experiment and identified CSF1R as a key node of disease progression in a mouse model of progressive MS. We hypothesized that modulating microglia and infiltrating macrophages through the inhibition of CSF1R will attenuate deleterious CNS inflammation and reduce subsequent demyelination and neurodegeneration. To test this hypothesis, we generated a novel potent and selective small-molecule CSF1R inhibitor (sCSF1Rinh) for preclinical testing. sCSF1Rinh blocked receptor phosphorylation and downstream signaling in both microglia and macrophages and altered cellular functions including proliferation, survival, and cytokine production. In vivo, CSF1R inhibition with sCSF1Rinh attenuated neuroinflammation and reduced microglial proliferation in a murine acute LPS model. Furthermore, the sCSF1Rinh attenuated a disease-associated microglial phenotype and blocked both axonal damage and neurological impairments in an experimental autoimmune encephalomyelitis (EAE) model of MS. While previous studies have focused on microglial depletion following CSF1R inhibition, our data clearly show that signaling downstream of this receptor can be beneficially modulated in the context of CNS injury. Together, these data suggest that CSF1R inhibition can reduce deleterious microglial proliferation and modulate microglial phenotypes during neuroinflammatory pathogenesis, particularly in progressive MS.


Asunto(s)
Inflamación/patología , Esclerosis Múltiple/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/patología , Humanos , Macrófagos/efectos de los fármacos , Ratones , Microglía/patología , Esclerosis Múltiple/patología , Transducción de Señal/efectos de los fármacos
5.
Cell Mol Gastroenterol Hepatol ; 9(2): 295-312, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31606566

RESUMEN

BACKGROUND AND AIMS: Tumor necrosis factor (TNF) is a major pathogenic effector and a therapeutic target in inflammatory bowel disease (IBD), yet the basis for TNF-induced intestinal epithelial cell (IEC) death is unknown, because TNF does not kill normal IECs. Here, we investigated how chronic nuclear factor (NF)- κB activation, which occurs in human IBD, promotes TNF-dependent IEC death in mice. METHODS: Human IBD specimens were stained for p65 and cleaved caspase-3. C57BL/6 mice with constitutively active IKKß in IEC (Ikkß(EE)IEC), Ripk1D138N/D138N knockin mice, and Ripk3-/- mice were injected with TNF or lipopolysaccharide. Enteroids were also isolated from these mice and challenged with TNF with or without RIPK1 and RIPK3 inhibitors or butylated hydroxyanisole. Ripoptosome-mediated caspase-8 activation was assessed by immunoprecipitation. RESULTS: NF-κB activation in human IBD correlated with appearance of cleaved caspase-3. Congruently, unlike normal mouse IECs that are TNF-resistant, IECs in Ikkß(EE)IEC mice and enteroids were susceptible to TNF-dependent apoptosis, which depended on the protein kinase function of RIPK1. Constitutively active IKKß facilitated ripoptosome formation, a RIPK1 signaling complex that mediates caspase-8 activation by TNF. Butylated hydroxyanisole treatment and RIPK1 inhibitors attenuated TNF-induced and ripoptosome-mediated caspase-8 activation and IEC death in vitro and in vivo. CONCLUSIONS: Contrary to common expectations, chronic NF-κB activation induced intestinal crypt apoptosis after TNF stimulation, resulting in severe mucosal erosion. RIPK1 kinase inhibitors selectively inhibited TNF destructive properties while preserving its survival and proliferative properties, which do not require RIPK1 kinase activity. RIPK1 kinase inhibition could be a potential treatment for IBD.


Asunto(s)
Apoptosis/inmunología , Colitis Ulcerosa/inmunología , Enfermedad de Crohn/inmunología , Mucosa Intestinal/patología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Factor de Transcripción ReIA/metabolismo , Adulto , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Células Cultivadas , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/patología , Colon/efectos de los fármacos , Colon/inmunología , Colon/patología , Colonoscopía , Enfermedad de Crohn/tratamiento farmacológico , Enfermedad de Crohn/patología , Modelos Animales de Enfermedad , Células Epiteliales/inmunología , Células Epiteliales/patología , Técnicas de Sustitución del Gen , Humanos , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Íleon/inmunología , Íleon/patología , Imidazoles/farmacología , Imidazoles/uso terapéutico , Indoles/farmacología , Indoles/uso terapéutico , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Ratones Noqueados , Organoides , Cultivo Primario de Células , RNA-Seq , Proteína Serina-Treonina Quinasas de Interacción con Receptores/antagonistas & inhibidores , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Factor de Necrosis Tumoral alfa/metabolismo
6.
Methods Mol Biol ; 1857: 125-134, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30136236

RESUMEN

The serine/threonine kinase RIPK1 has numerous biological and pathological functions, mediating prosurvival as well as prodeath apoptotic and necroptotic signaling pathways downstream of various receptors, including death receptors and Toll-like receptors (TLRs). RIPK1 has been implicated in various diseases, including ischemia-reperfusion injury and inflammatory bowel disease (IBD). The recent generation of RIPK1 kinase inactive mice has enabled us to genetically interrogate the role of RIPK1 kinase-mediated necroptosis in disease models. Here, we describe procedures utilizing kinase inactive Ripk1D138N/D138N mice to analyze necroptosis induction in vitro in bone-marrow derived macrophages (BMDMs) and in vivo in a murine model of TNF-induced shock.


Asunto(s)
Apoptosis , Modelos Animales de Enfermedad , Macrófagos/patología , Necrosis , Proteína Serina-Treonina Quinasas de Interacción con Receptores/antagonistas & inhibidores , Choque Séptico/patología , Factor de Necrosis Tumoral alfa/toxicidad , Animales , Células Cultivadas , Femenino , Macrófagos/efectos de los fármacos , Masculino , Ratones , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Choque Séptico/inducido químicamente
7.
J Clin Invest ; 128(5): 2064-2075, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29664014

RESUMEN

Receptor interacting protein kinase 1 (RIPK1) has important kinase-dependent and kinase-independent scaffolding functions that activate or prevent apoptosis or necroptosis in a cell context-dependent manner. The kinase activity of RIPK1 mediates hypothermia and lethality in a mouse model of TNF-induced shock, reflecting the hyperinflammatory state of systemic inflammatory response syndrome (SIRS), where the proinflammatory "cytokine storm" has long been viewed as detrimental. Here, we demonstrate that cytokine and chemokine levels did not predict survival and, importantly, that kinase-inactive Ripk1D138N/D138N hematopoietic cells afforded little protection from TNF- or TNF/zVAD-induced shock in reconstituted mice. Unexpectedly, RIPK1 kinase-inactive mice transplanted with WT hematopoietic cells remained resistant to TNF-induced shock, revealing that a nonhematopoietic lineage mediated protection. TNF-treated Ripk1D138N/D138N mice exhibited no significant increases in intestinal or vascular permeability, nor did they activate the clotting cascade. We show that TNF administration damaged the liver vascular endothelium and induced phosphorylated mixed lineage kinase domain-like (phospho-MLKL) reactivity in endothelial cells isolated from TNF/zVAD-treated WT, but not Ripk1D138N/D138N, mice. These data reveal that the tissue damage present in this SIRS model is reflected, in part, by breaks in the vasculature due to endothelial cell necroptosis and thereby predict that RIPK1 kinase inhibitors may provide clinical benefit to shock and/or sepsis patients.


Asunto(s)
Endotelio Vascular/enzimología , Hígado/enzimología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Síndrome de Respuesta Inflamatoria Sistémica/enzimología , Clorometilcetonas de Aminoácidos/toxicidad , Animales , Endotelio Vascular/lesiones , Endotelio Vascular/patología , Células Madre Hematopoyéticas , Hígado/patología , Ratones , Ratones de la Cepa 129 , Ratones Mutantes , Necrosis , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Síndrome de Respuesta Inflamatoria Sistémica/inducido químicamente , Síndrome de Respuesta Inflamatoria Sistémica/genética , Síndrome de Respuesta Inflamatoria Sistémica/patología , Factor de Necrosis Tumoral alfa/toxicidad
9.
J Immunol ; 200(2): 737-748, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29212904

RESUMEN

Necroptosis is a form of cell death associated with inflammation; however, the biological consequences of chronic necroptosis are unknown. Necroptosis is mediated by RIPK1, RIPK3, and MLKL kinases but in hematopoietic cells RIPK1 has anti-inflammatory roles and functions to prevent necroptosis. Here we interrogate the consequences of chronic necroptosis on immune homeostasis by deleting Ripk1 in mouse dendritic cells. We demonstrate that deregulated necroptosis results in systemic inflammation, tissue fibrosis, and autoimmunity. We show that inflammation and autoimmunity are prevented upon expression of kinase inactive RIPK1 or deletion of RIPK3 or MLKL. We provide evidence that the inflammation is not driven by microbial ligands, but depends on the release of danger-associated molecular patterns and MyD88-dependent signaling. Importantly, although the inflammation is independent of type I IFN and the nucleic acid sensing TLRs, blocking these pathways rescues the autoimmunity. These mouse genetic studies reveal that chronic necroptosis may underlie human fibrotic and autoimmune disorders.


Asunto(s)
Autoinmunidad , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Inmunidad , Inflamación/etiología , Inflamación/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Animales , Autoanticuerpos/inmunología , Autoinmunidad/genética , Linfocitos B/inmunología , Linfocitos B/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Perfilación de la Expresión Génica , Inflamación/patología , Inflamación/prevención & control , Linfadenopatía/genética , Linfadenopatía/inmunología , Linfadenopatía/metabolismo , Linfadenopatía/patología , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Necrosis/genética , Necrosis/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/deficiencia , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Transducción de Señal , Receptores Toll-Like/metabolismo
10.
J Immunol ; 198(11): 4435-4447, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28461567

RESUMEN

The innate immune response is a central element of the initial defense against bacterial and viral pathogens. Macrophages are key innate immune cells that upon encountering pathogen-associated molecular patterns respond by producing cytokines, including IFN-ß. In this study, we identify a novel role for RIPK1 and RIPK3, a pair of homologous serine/threonine kinases previously implicated in the regulation of necroptosis and pathologic tissue injury, in directing IFN-ß production in macrophages. Using genetic and pharmacologic tools, we show that catalytic activity of RIPK1 directs IFN-ß synthesis induced by LPS in mice. Additionally, we report that RIPK1 kinase-dependent IFN-ß production may be elicited in an analogous fashion using LPS in bone marrow-derived macrophages upon inhibition of caspases. Notably, this regulation requires kinase activities of both RIPK1 and RIPK3, but not the necroptosis effector protein, MLKL. Mechanistically, we provide evidence that necrosome-like RIPK1 and RIPK3 aggregates facilitate canonical TRIF-dependent IFN-ß production downstream of the LPS receptor TLR4. Intriguingly, we also show that RIPK1 and RIPK3 kinase-dependent synthesis of IFN-ß is markedly induced by avirulent strains of Gram-negative bacteria, Yersinia and Klebsiella, and less so by their wild-type counterparts. Overall, these observations identify unexpected roles for RIPK1 and RIPK3 kinases in the production of IFN-ß during the host inflammatory responses to bacterial infection and suggest that the axis in which these kinases operate may represent a target for bacterial virulence factors.


Asunto(s)
Interferón beta/biosíntesis , Lipopolisacáridos/inmunología , Macrófagos/inmunología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Animales , Apoptosis/inmunología , Bacterias Gramnegativas/inmunología , Interferón beta/inmunología , Klebsiella/inmunología , Macrófagos/microbiología , Ratones , Necrosis/inmunología , Fosforilación , Receptor Toll-Like 4/inmunología , Yersinia/inmunología
11.
Immunity ; 45(1): 46-59, 2016 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-27396959

RESUMEN

Macrophages are a crucial component of the innate immune system in sensing pathogens and promoting local and systemic inflammation. RIPK1 and RIPK3 are homologous kinases, previously linked to activation of necroptotic death. In this study, we have described roles for these kinases as master regulators of pro-inflammatory gene expression induced by lipopolysaccharide, independent of their well-documented cell death functions. In primary macrophages, this regulation was elicited in the absence of caspase-8 activity, required the adaptor molecule TRIF, and proceeded in a cell autonomous manner. RIPK1 and RIPK3 kinases promoted sustained activation of Erk, cFos, and NF-κB, which were required for inflammatory changes. Utilizing genetic and pharmacologic tools, we showed that RIPK1 and RIPK3 account for acute inflammatory responses induced by lipopolysaccharide in vivo; notably, this regulation did not require exogenous manipulation of caspases. These findings identified a new pharmacologically accessible pathway that may be relevant to inflammatory pathologies.


Asunto(s)
Inmunidad Innata , Inflamación/inmunología , Macrófagos/inmunología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Caspasa 8/genética , Caspasa 8/metabolismo , Células Cultivadas , Femenino , Lipopolisacáridos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Necrosis , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Transducción de Señal , Transcriptoma
12.
Cell Rep ; 15(11): 2449-61, 2016 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-27264187

RESUMEN

Tumor necrosis factor (TNF) induces necroptosis, a RIPK3/MLKL-dependent form of inflammatory cell death. In response to infection by Gram-negative bacteria, multiple receptors on macrophages, including TLR4, TNF, and type I IFN receptors, are concurrently activated, but it is unclear how they crosstalk to regulate necroptosis. We report that TLR4 activates CASPASE-8 to cleave and remove the deubiquitinase cylindromatosis (CYLD) in a TRIF- and RIPK1-dependent manner to disable necroptosis in macrophages. Inhibiting CASPASE-8 leads to CYLD-dependent necroptosis caused by the TNF produced in response to TLR4 ligation. While lipopolysaccharides (LPS)-induced necroptosis was abrogated in Tnf(-/-) macrophages, a soluble TNF antagonist was not able to do so in Tnf(+/+) macrophages, indicating that necroptosis occurs in a cell-autonomous manner. Surprisingly, TNF-mediated auto-necroptosis of macrophages requires type I IFN, which primes the expression of key necroptosis-signaling molecules, including TNFR2 and MLKL. Thus, the TNF necroptosis pathway is regulated by both negative and positive crosstalk.


Asunto(s)
Apoptosis , Cisteína Endopeptidasas/metabolismo , Citoprotección , Interferón Tipo I/metabolismo , Macrófagos/metabolismo , Proteolisis , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Caspasa 8/metabolismo , Enzima Desubiquitinante CYLD , Lipopolisacáridos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , Necrosis , Proteínas Quinasas/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal , Receptor Toll-Like 4/metabolismo
13.
J Gen Virol ; 96(Pt 2): 351-359, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25381055

RESUMEN

Several members of the family Circoviridae have been shown to encode proteins with apoptotic activity. For example, both porcine circovirus type 2 (PCV2) and chicken anemia virus (CAV) encode a third viral protein (VP3) that has been shown to be cytotoxic. Interestingly, in the case of the CAV protein (designated apoptin), apoptosis is specific to transformed cell types. Similarities in genome structure and organization suggest that PCV type 1 (PCV1) may also contain a third ORF, which codes for a protein with homologous activity. To investigate this, ORF prediction followed by gene expression analyses were conducted on a gene found to be homologous to CAV and PCV2 VP3. Our data presented herein elucidate a putative ORF3 that codes for a viral protein with functional similarity to that of apoptin and PCV2 VP3. Unlike its homologues, sequence analysis revealed a highly hydrophobic, extended C-terminal domain in PCV1 VP3, which harbours a strong nuclear export signal. Subcellular localization analysis demonstrated divergent PCV1 VP3 localization patterns compared with that of CAV VP3. Interestingly, cytotoxicity studies revealed evidence that apoptosis may be selective to transformed cell types, similar to apoptin; however, PCV1 VP3 induced a dramatic G1 cell cycle arrest as opposed to the G2/M arrest observed with apoptin. These results indicate that nuclear localization of PCV1 VP3 is necessary neither for induction of apoptosis nor for transformed cell selectivity, and suggest a mechanism of action distinct from that of apoptin.


Asunto(s)
Apoptosis , Circovirus/fisiología , Interacciones Huésped-Patógeno , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Animales , Ciclo Celular , Línea Celular , Supervivencia Celular , Perfilación de la Expresión Génica , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido , Porcinos , Proteínas Virales/genética
14.
Proc Natl Acad Sci U S A ; 111(40): 14436-41, 2014 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-25246544

RESUMEN

Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) is recruited to the TNF receptor 1 to mediate proinflammatory signaling and to regulate TNF-induced cell death. RIPK1 deficiency results in postnatal lethality, but precisely why Ripk1(-/-) mice die remains unclear. To identify the lineages and cell types that depend on RIPK1 for survival, we generated conditional Ripk1 mice. Tamoxifen administration to adult RosaCreER(T2)Ripk1(fl/fl) mice results in lethality caused by cell death in the intestinal and hematopoietic lineages. Similarly, Ripk1 deletion in cells of the hematopoietic lineage stimulates proinflammatory cytokine and chemokine production and hematopoietic cell death, resulting in bone marrow failure. The cell death reflected cell-intrinsic survival roles for RIPK1 in hematopoietic stem and progenitor cells, because Vav-iCre Ripk1(fl/fl) fetal liver cells failed to reconstitute hematopoiesis in lethally irradiated recipients. We demonstrate that RIPK3 deficiency partially rescues hematopoiesis in Vav-iCre Ripk1(fl/fl) mice, showing that RIPK1-deficient hematopoietic cells undergo RIPK3-mediated necroptosis. However, the Vav-iCre Ripk1(fl/fl) Ripk3(-/-) progenitors remain TNF sensitive in vitro and fail to repopulate irradiated mice. These genetic studies reveal that hematopoietic RIPK1 deficiency triggers both apoptotic and necroptotic death that is partially prevented by RIPK3 deficiency. Therefore, RIPK1 regulates hematopoiesis and prevents inflammation by suppressing RIPK3 activation.


Asunto(s)
Apoptosis/fisiología , Médula Ósea/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/deficiencia , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Médula Ósea/patología , Células Cultivadas , Citocinas/sangre , Células Epiteliales/citología , Células Epiteliales/metabolismo , Antagonistas de Estrógenos/farmacología , Citometría de Flujo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Mediadores de Inflamación/sangre , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Necrosis , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Bazo/citología , Bazo/metabolismo , Tamoxifeno/farmacología , Timo/citología , Timo/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
15.
Nature ; 513(7516): 90-4, 2014 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-25132550

RESUMEN

Necroptosis has emerged as an important pathway of programmed cell death in embryonic development, tissue homeostasis, immunity and inflammation. RIPK1 is implicated in inflammatory and cell death signalling and its kinase activity is believed to drive RIPK3-mediated necroptosis. Here we show that kinase-independent scaffolding RIPK1 functions regulate homeostasis and prevent inflammation in barrier tissues by inhibiting epithelial cell apoptosis and necroptosis. Intestinal epithelial cell (IEC)-specific RIPK1 knockout caused IEC apoptosis, villus atrophy, loss of goblet and Paneth cells and premature death in mice. This pathology developed independently of the microbiota and of MyD88 signalling but was partly rescued by TNFR1 (also known as TNFRSF1A) deficiency. Epithelial FADD ablation inhibited IEC apoptosis and prevented the premature death of mice with IEC-specific RIPK1 knockout. However, mice lacking both RIPK1 and FADD in IECs displayed RIPK3-dependent IEC necroptosis, Paneth cell loss and focal erosive inflammatory lesions in the colon. Moreover, a RIPK1 kinase inactive knock-in delayed but did not prevent inflammation caused by FADD deficiency in IECs or keratinocytes, showing that RIPK3-dependent necroptosis of FADD-deficient epithelial cells only partly requires RIPK1 kinase activity. Epidermis-specific RIPK1 knockout triggered keratinocyte apoptosis and necroptosis and caused severe skin inflammation that was prevented by RIPK3 but not FADD deficiency. These findings revealed that RIPK1 inhibits RIPK3-mediated necroptosis in keratinocytes in vivo and identified necroptosis as a more potent trigger of inflammation compared with apoptosis. Therefore, RIPK1 is a master regulator of epithelial cell survival, homeostasis and inflammation in the intestine and the skin.


Asunto(s)
Apoptosis , Células Epiteliales/citología , Células Epiteliales/patología , Homeostasis , Necrosis , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Animales , Caspasa 8/metabolismo , Supervivencia Celular , Células Epiteliales/metabolismo , Proteína de Dominio de Muerte Asociada a Fas/deficiencia , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Femenino , Inflamación/metabolismo , Inflamación/patología , Mucosa Intestinal/metabolismo , Intestinos/citología , Intestinos/patología , Queratinocitos/metabolismo , Queratinocitos/patología , Masculino , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/metabolismo , Células de Paneth/metabolismo , Células de Paneth/patología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/deficiencia , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Receptores Tipo I de Factores de Necrosis Tumoral/deficiencia , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Piel/citología , Piel/metabolismo , Piel/patología
16.
J Immunol ; 193(4): 1539-1543, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25015821

RESUMEN

The serine/threonine kinase RIPK1 is recruited to TNFR1 to mediate proinflammatory signaling and to regulate TNF-induced cell death. A RIPK1 deficiency results in perinatal lethality, impaired NFκB and MAPK signaling, and sensitivity to TNF-induced apoptosis. Chemical inhibitor and in vitro-reconstitution studies suggested that RIPK1 displays distinct kinase activity-dependent and -independent functions. To determine the contribution of RIPK1 kinase to inflammation in vivo, we generated knock-in mice endogenously expressing catalytically inactive RIPK1 D138N. Unlike Ripk1(-/-) mice, which die shortly after birth, Ripk1(D138N/D138N) mice are viable. Cells expressing RIPK1 D138N are resistant to TNF- and polyinosinic-polycytidylic acid-induced necroptosis in vitro, and Ripk1(D138N/D138N) mice are protected from TNF-induced shock in vivo. Moreover, Ripk1(D138N/D138N) mice fail to control vaccinia virus replication in vivo. This study provides genetic evidence that the kinase activity of RIPK1 is not required for survival but is essential for TNF-, TRIF-, and viral-initiated necroptosis.


Asunto(s)
Apoptosis/inmunología , Hipotermia/mortalidad , Necrosis/inmunología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Factor de Necrosis Tumoral alfa/farmacología , Proteínas Adaptadoras del Transporte Vesicular/inmunología , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Técnicas de Sustitución del Gen , Hipotermia/inducido químicamente , Inflamación/genética , Inflamación/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , FN-kappa B/inmunología , Necrosis/inducido químicamente , Poli I-C/farmacología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/inmunología , Receptores Tipo I de Factores de Necrosis Tumoral/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Vaccinia/inmunología , Virus Vaccinia/crecimiento & desarrollo , Virus Vaccinia/inmunología , Replicación Viral/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...