Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 175
Filtrar
1.
Cell Rep ; 43(4): 114095, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38613787

RESUMEN

Interferon (IFN) contributes to the host's antiviral response by inducing IFN-stimulated genes (ISGs). However, their functional targets and the mechanism of action remain elusive. Here, we report that one such ISG, TRIM21, interacts with and degrades the TRPV2 channel in myeloid cells, reducing its expression and providing host protection against viral infections. Moreover, viral infection upregulates TRIM21 in paracrine and autocrine manners, downregulating TRPV2 in neighboring cells to prevent viral spread to uninfected cells. Consistently, the Trim21-/- mice are more susceptible to HSV-1 and VSV infection than the Trim21+/+ littermates, in which viral susceptibility is rescued by inhibition or deletion of TRPV2. Mechanistically, TRIM21 catalyzes the K48-linked ubiquitination of TRPV2 at Lys295. TRPV2K295R is resistant to viral-infection-induced TRIM21-dependent ubiquitination and degradation, promoting viral infection more profoundly than wild-type TRPV2 when reconstituted into Lyz2-Cre;Trpv2fl/fl myeloid cells. These findings characterize targeting the TRIM21-TRPV2 axis as a conducive strategy to control viral spread to bystander cells.


Asunto(s)
Ribonucleoproteínas , Canales Catiónicos TRPV , Ubiquitinación , Virosis , Animales , Humanos , Ratones , Regulación hacia Abajo , Células HEK293 , Herpesvirus Humano 1/fisiología , Interferones/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/metabolismo , Ribonucleoproteínas/metabolismo , Canales Catiónicos TRPV/metabolismo , Canales Catiónicos TRPV/genética , Virosis/metabolismo
2.
Cell Res ; 33(12): 923-939, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37932447

RESUMEN

Combination therapy with PD-1 blockade and IL-2 substantially improves anti-tumor efficacy comparing to monotherapy. The underlying mechanisms responsible for the synergistic effects of the combination therapy remain enigmatic. Here we show that PD-1 ligation results in BATF-dependent transcriptional induction of the membrane-associated E3 ubiquitin ligase MARCH5, which mediates K27-linked polyubiquitination and lysosomal degradation of the common cytokine receptor γ chain (γc). PD-1 ligation also activates SHP2, which dephosphorylates γcY357, leading to impairment of γc family cytokine-triggered signaling. Conversely, PD-1 blockade restores γc level and activity, thereby sensitizing CD8+ T cells to IL-2. We also identified Pitavastatin Calcium as an inhibitor of MARCH5, which combined with PD-1 blockade and IL-2 significantly improves the efficacy of anti-tumor immunotherapy in mice. Our findings uncover the mechanisms by which PD-1 signaling antagonizes γc family cytokine-triggered immune activation and demonstrate that the underlying mechanisms can be exploited for increased efficacy of combination immunotherapy of cancer.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico , Subunidad gamma Común de Receptores de Interleucina , Neoplasias , Receptor de Muerte Celular Programada 1 , Animales , Ratones , Linfocitos T CD8-positivos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Interleucina-2 , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Ubiquitinación , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas de la Membrana/metabolismo
3.
Cell Mol Immunol ; 20(12): 1403-1412, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37932533

RESUMEN

Various cellular stress conditions trigger mitochondrial DNA (mtDNA) release from mitochondria into the cytosol. The released mtDNA is sensed by the cGAS-MITA/STING pathway, resulting in the induced expression of type I interferon and other effector genes. These processes contribute to the innate immune response to viral infection and other stress factors. The deregulation of these processes causes autoimmune diseases, inflammatory metabolic disorders and cancer. Therefore, the cGAS-MITA/STING pathway is a potential target for intervention in infectious, inflammatory and autoimmune diseases as well as cancer. In this review, we focus on the mechanisms underlying the mtDNA-triggered activation of the cGAS-MITA/STING pathway, the effects of the pathway under various physiological and pathological conditions, and advances in the development of drugs that target cGAS and MITA/STING.


Asunto(s)
Enfermedades Autoinmunes , Neoplasias , Humanos , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Transducción de Señal , Inmunidad Innata , Nucleotidiltransferasas/metabolismo , Mitocondrias/metabolismo , Enfermedades Autoinmunes/patología , Neoplasias/patología
4.
Nat Cell Biol ; 25(8): 1208-1222, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37443289

RESUMEN

Evasion of antitumour immunity is a hallmark of cancer. STING, a putative innate immune signalling adaptor, has a pivotal role in mounting antitumour immunity by coordinating innate sensing and adaptive immune surveillance in myeloid cells. STING is markedly silenced in various human malignancies and acts as a cell-intrinsic tumour suppressor. How STING exerts intrinsic antitumour activity remains unclear. Here, we report that STING restricts aerobic glycolysis independent of its innate immune function. Mechanistically, STING targets hexokinase II (HK2) to block its hexokinase activity. As such, STING inhibits HK2 to restrict tumour aerobic glycolysis and promote antitumour immunity in vivo. In human colorectal carcinoma samples, lactate, which can be used as a surrogate for aerobic glycolysis, is negatively correlated with STING expression level and antitumour immunity. Taken together, this study reveals that STING functions as a cell-intrinsic metabolic checkpoint that restricts aerobic glycolysis to promote antitumour immunity. These findings have important implications for the development of STING-based therapeutic modalities to improve antitumour immunotherapy.


Asunto(s)
Neoplasias Colorrectales , Hexoquinasa , Humanos , Hexoquinasa/genética , Hexoquinasa/metabolismo , Fosforilación , Transducción de Señal , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Glucólisis
5.
J Mol Cell Biol ; 2023 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-37442610

RESUMEN

Estrogen receptor α (ERα) is an important driver and therapeutic target in approximately 70% of breast cancers. How ERα drives breast carcinogenesis is not fully understood. In this study, we show that ERα is a negative regulator of type I interferon (IFN) response, which is critical for breast carcinogenesis. Activation of ERα by its natural ligand estradiol inhibits IFN-ß-induced transcription of downstream IFN-stimulated genes (ISGs), whereas deficiency of ERα or stimulation with its antagonist fulvestrant has opposite effects. Mechanistically, ERα inhibits type I IFN response by two distinct mechanisms. ERα induces expression of the histone 2A variant H2A.Z, which restricts engagement of the IFN-stimulated gene factor 3 (ISGF3) complex at the ISG promoters. ERα also interacts with STAT2, which leads to disruption of the ISGF3 complex. These two events mutually lead to transcriptional inhibition of ISGs induced by type I IFNs. In a xenograft mouse tumor model, fulvestrant enhances the ability of IFN-ß to suppress ERα+ breast tumor growth. Consistently, clinical data suggests that ERα+ breast cancer patients with higher levels of ISGs exhibit an increased survival rate. Our findings suggest that ERα inhibits type I IFN response via two distinct mechanisms to promote breast cancer.

6.
Cell Mol Immunol ; 20(2): 175-188, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36600052

RESUMEN

Upon viral infection, cytoplasmic pattern recognition receptors detect viral nucleic acids and activate the adaptor protein VISA/MAVS- or MITA/STING-mediated innate antiviral response. Whether and how the innate antiviral response is regulated by neuronal endocrine functions is unclear. Here, we show that viral infection reduced the serum levels of the ß-adrenergic hormones epinephrine and norepinephrine as well as the cellular levels of their receptors ADRB1 and ADRB2. We further show that an increase in epinephrine/norepinephrine level inhibited the innate antiviral response in an ADRB1-/2-dependent manner. Mechanistically, epinephrine/norepinephrine stimulation activated the downstream kinase PKA, which catalyzed the phosphorylation of MITA at S241, S243 and T263, inhibiting MITA activation and suppressing the innate immune response to DNA virus. In addition, phosphorylation of VISA at T54 by PKA antagonized the innate immune response to RNA virus. These findings reveal the regulatory mechanisms of innate antiviral responses by epinephrine/norepinephrine and provide a possible explanation for increased host susceptibility to viral infection in stressful and anxiety-promoting situations.


Asunto(s)
Proteínas de la Membrana , Virosis , Humanos , Antivirales , Epinefrina/farmacología , Inmunidad Innata/genética , Proteínas de la Membrana/genética , Norepinefrina/farmacología , Receptores Adrenérgicos beta 1/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Activación Enzimática , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo
7.
Curr Opin Immunol ; 78: 102248, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36193584

RESUMEN

Mediator of IRF3 activation (MITA, also known as stimulator of interferon genes (STING) and endoplasmic reticulum interferon stimulator (ERIS)) is an ER-associated protein that senses cellular and bacterium-derived cyclic dinucleotide (CDN), leading to induction of type-I interferons (IFNs) and innate immune responses against viruses and bacteria. Recently, it has become clear that sensing of CDN and induction of autophagy are two evolutionarily conserved functions of MITA, predating its role in mediating type-I IFN induction. Studies have shown that MITA-mediated signaling promotes a number of autoimmune disorders caused by gene mutations in human. Here, we summarize the most recent progress on MITA-mediated signaling in a view of evolution and highlight the roles of MITA in human inflammatory disorders caused by gene mutations and in genetically modified mouse models. We also briefly introduce the chemicals targeting MITA and discuss their potential in treatment of MITA-mediated inflammatory diseases. Finally, we propose several key questions that should be addressed for targeting MITA for treatment of related autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes , Interferón Tipo I , Proteínas de la Membrana , Animales , Humanos , Ratones , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Autoinmunidad , Inmunidad Innata , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Virosis/inmunología , Virosis/metabolismo
8.
Proc Natl Acad Sci U S A ; 119(43): e2207280119, 2022 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-36252040

RESUMEN

The current view of nucleic acid-mediated innate immunity is that binding of intracellular sensors to nucleic acids is sufficient for their activation. Here, we report that endocytosis of virus or foreign DNA initiates a priming signal for the DNA sensor cyclic GMP-AMP synthase (cGAS)-mediated innate immune response. Mechanistically, viral infection or foreign DNA transfection triggers recruitment of the spleen tyrosine kinase (SYK) and cGAS to the endosomal vacuolar H+ pump (V-ATPase), where SYK is activated and then phosphorylates human cGASY214/215 (mouse cGasY200/201) to prime its activation. Upon binding to DNA, the primed cGAS initiates robust cGAMP production and mediator of IRF3 activation/stimulator of interferon genes-dependent innate immune response. Consistently, blocking the V-ATPase-SYK axis impairs DNA virus- and transfected DNA-induced cGAMP production and expression of antiviral genes. Our findings reveal that V-ATPase-SYK-mediated tyrosine phosphorylation of cGAS following endocytosis of virus or other cargos serves as a priming signal for cGAS activation and innate immune response.


Asunto(s)
Endocitosis , Inmunidad Innata , Nucleotidiltransferasas , Quinasa Syk , ATPasas de Translocación de Protón Vacuolares , Animales , Humanos , Ratones , ADN , Interferones/metabolismo , Proteínas de la Membrana/metabolismo , Nucleotidiltransferasas/metabolismo , Transducción de Señal/genética , Quinasa Syk/metabolismo , Tirosina , ATPasas de Translocación de Protón Vacuolares/metabolismo
9.
Cell Mol Immunol ; 19(10): 1117-1129, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35982175

RESUMEN

Interleukin 5 (IL-5) plays crucial roles in type 2-high asthma by mediating eosinophil maturation, activation, chemotaxis and survival. Inhibition of IL-5 signaling is considered a strategy for asthma treatment. Here, we identified MARCH2 and MARCH3 as critical negative regulators of IL-5-triggered signaling. MARCH2 and MARCH3 associate with the IL-5 receptor α chain (IL-5Rα) and mediate its K27-linked polyubiquitination at K379 and K383, respectively, and its subsequent lysosomal degradation. Deficiency of MARCH2 or MARCH3 modestly increases the level of IL-5Rα and enhances IL-5-induced signaling, whereas double knockout of MARCH2/3 has a more dramatic effect. March2/3 double knockout markedly increases the proportions of eosinophils in the bone marrow and peripheral blood in mice. Double knockout of March2/3 aggravates ovalbumin (OVA)-induced eosinophilia and causes increased inflammatory cell infiltration, peribronchial mucus secretion and production of Th2 cytokines. Neutralization of Il-5 attenuates OVA-induced airway inflammation and the enhanced effects of March2/3 double deficiency. These findings suggest that MARCH2 and MARCH3 play redundant roles in targeting IL-5Rα for degradation and negatively regulating allergic airway inflammation.


Asunto(s)
Asma , Eosinofilia , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Líquido del Lavado Bronquioalveolar , Citocinas/metabolismo , Eosinófilos , Inflamación/metabolismo , Interleucina-5/metabolismo , Interleucina-5/farmacología , Subunidad alfa del Receptor de Interleucina-5/metabolismo , Ligasas/metabolismo , Ligasas/farmacología , Ratones , Ratones Endogámicos BALB C , Ovalbúmina/farmacología , Ubiquitina/metabolismo
10.
Signal Transduct Target Ther ; 7(1): 137, 2022 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-35468896

RESUMEN

Whether and how innate antiviral response is regulated by humoral metabolism remains enigmatic. We show that viral infection induces progesterone via the hypothalamic-pituitary-adrenal axis in mice. Progesterone induces downstream antiviral genes and promotes innate antiviral response in cells and mice, whereas knockout of the progesterone receptor PGR has opposite effects. Mechanistically, stimulation of PGR by progesterone activates the tyrosine kinase SRC, which phosphorylates the transcriptional factor IRF3 at Y107, leading to its activation and induction of antiviral genes. SARS-CoV-2-infected patients have increased progesterone levels, and which are co-related with decreased severity of COVID-19. Our findings reveal how progesterone modulates host innate antiviral response, and point to progesterone as a potential immunomodulatory reagent for infectious and inflammatory diseases.


Asunto(s)
COVID-19 , SARS-CoV-2 , Animales , Antivirales , COVID-19/genética , Humanos , Sistema Hipotálamo-Hipofisario , Inmunidad Innata/genética , Ratones , Sistema Hipófiso-Suprarrenal , Progesterona/farmacología
11.
Nat Commun ; 13(1): 1700, 2022 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-35361799

RESUMEN

Anti-PD-1/PD-L1 immunotherapy has achieved impressive therapeutic outcomes in patients with multiple cancer types. However, the underlined molecular mechanism(s) for moderate response rate (15-25%) or resistance to PD-1/PD-L1 blockade remains not completely understood. Here, we report that inhibiting the deubiquitinase, USP8, significantly enhances the efficacy of anti-PD-1/PD-L1 immunotherapy through reshaping an inflamed tumor microenvironment (TME). Mechanistically, USP8 inhibition increases PD-L1 protein abundance through elevating the TRAF6-mediated K63-linked ubiquitination of PD-L1 to antagonize K48-linked ubiquitination and degradation of PD-L1. In addition, USP8 inhibition also triggers innate immune response and MHC-I expression largely through activating the NF-κB signaling. Based on these mechanisms, USP8 inhibitor combination with PD-1/PD-L1 blockade significantly activates the infiltrated CD8+ T cells to suppress tumor growth and improves the survival benefit in several murine tumor models. Thus, our study reveals a potential combined therapeutic strategy to utilize a USP8 inhibitor and PD-1/PD-L1 blockade for enhancing anti-tumor efficacy.


Asunto(s)
Endopeptidasas , Complejos de Clasificación Endosomal Requeridos para el Transporte , Inmunoterapia , Neoplasias , Microambiente Tumoral , Ubiquitina Tiolesterasa , Animales , Linfocitos T CD8-positivos , Línea Celular Tumoral , Endopeptidasas/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Humanos , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Ubiquitina Tiolesterasa/genética
12.
Proc Natl Acad Sci U S A ; 119(10): e2116279119, 2022 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-35238669

RESUMEN

The warning cytokine interleukin-33 receptor (IL-33R) mediates local inflammatory responses and plays crucial roles in the pathogenesis of immune diseases such as pulmonary fibrosis and rheumatoid arthritis. Whether and how IL-33R is regulated remain enigmatic. Here, we identified ubiquitin-specific protease 38 (USP38) as a negative regulator of IL-33R­mediated signaling. USP38 deficiency promotes interleukin-33 (IL-33)­induced downstream proinflammatory responses in vitro and in vivo. Usp38−/− mice are more susceptible to inflammatory damage and death and developed more serious pulmonary fibrosis after bleomycin treatment. USP38 is constitutively associated with IL-33R and deconjugates its K27-linked polyubiquitination at K511, resulting in its autophagic degradation. We further show that the E3 ubiquitin ligase tumor necrosis factor receptor­associated factor 6 (TRAF6) catalyzes K27-linked polyubiquitination of IL-33R at K511, and that deficiency of TRAF6 inhibits IL-33­mediated signaling. Our findings suggest that K27-linked polyubiquitination and deubiquitination of IL-33R by TRAF6 and USP38 reciprocally regulate IL-33R level and signaling, which represents a critical mechanism in the regulation of IL-33­triggered lung inflammatory response and pulmonary fibrosis.


Asunto(s)
Inflamación/fisiopatología , Interleucina-33/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Fibrosis Pulmonar/fisiopatología , Proteasas Ubiquitina-Específicas/metabolismo , Autofagia , Regulación hacia Abajo , Humanos , Inflamación/metabolismo , Interleucina-33/metabolismo , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Transducción de Señal , Proteasas Ubiquitina-Específicas/genética , Ubiquitinación
13.
Signal Transduct Target Ther ; 7(1): 21, 2022 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-35075102

RESUMEN

Interleukin-3 (IL-3) is a hematopoietic growth factor and critical regulator of inflammatory response such as sepsis. IL-3 binds to IL-3 receptor α (IL-3Rα), which is then associated with IL-3Rß to initiate signaling. How IL-3-triggered physiological and pathological effects are regulated at the receptor level is unclear. Here, we show that the plasma membrane-associated E3 ubiquitin ligase MARCH3 negatively regulates IL-3-triggered signaling. MARCH3 is associated with IL-3Rα, mediates its K48-linked polyubiquitination at K377 and promotes its proteasomal degradation. MARCH3-deficiency promotes IL-3-triggered transcription of downstream effector genes and IL-3-induced expansion of myeloid cells. In the cecal ligation and puncture (CLP) model of sepsis, MARCH3-deficiency aggravates IL-3-ampified expression of inflammatory cytokines, organ damage and inflammatory death. Our findings suggest that regulation of IL-3Rα by MARCH3 plays an important role in IL-3-triggered physiological functions and inflammatory diseases.


Asunto(s)
Subunidad alfa del Receptor de Interleucina-3/inmunología , Interleucina-3/inmunología , Péptidos y Proteínas de Señalización Intracelular/inmunología , Proteolisis , Ubiquitinación/inmunología , Animales , Células HEK293 , Humanos , Inflamación/genética , Inflamación/inmunología , Interleucina-3/genética , Subunidad alfa del Receptor de Interleucina-3/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ubiquitinación/genética
14.
Cell Insight ; 1(1): 100006, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37192985
15.
Cell Mol Immunol ; 18(12): 2648-2659, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34785732

RESUMEN

The IL-6-STAT3 axis is critically involved in inflammation-associated carcinogenesis (IAC). How this axis is regulated to modulate IAC remains unknown. Here, we show that the plasma membrane-associated E3 ubiquitin ligase MARCH3 negatively regulates STAT3 activation triggered by IL-6, as well as another IL-6 subfamily member, Oncostatin M (OSM). MARCH3 is associated with the IL-6 receptor α-chain (IL-6Rα) and its coreceptor gp130. Biochemical experiments indicated that MARCH3 mediates the polyubiquitination of IL-6Rα at K401 and gp130 at K849 following IL-6 stimulation, leading to their translocation to and degradation in lysosomes. MARCH3 deficiency increases IL-6- and OSM-triggered activation of STAT3 and induction of downstream effector genes in various cell types. MARCH3 deficiency enhances dextran sulfate sodium (DSS)-induced STAT3 activation, increases the expression of inflammatory cytokines, and exacerbates colitis, as well as azoxymethane (AOM)/DSS-induced colitis-associated cancer in mice. In addition, MARCH3 is downregulated in human colorectal cancer tissues and associated with poor survival across different cancer types. Our findings suggest that MARCH3 is a pivotal negative regulator of IL-6-induced STAT3 activation, inflammation, and inflammation-associated carcinogenesis.


Asunto(s)
Colitis , Ubiquitina-Proteína Ligasas , Animales , Carcinogénesis/metabolismo , Colitis/inducido químicamente , Colitis/complicaciones , Colitis/metabolismo , Sulfato de Dextran/efectos adversos , Modelos Animales de Enfermedad , Interleucina-6/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptores de Interleucina-6 , Transducción de Señal , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
16.
J Mol Cell Biol ; 13(10): 721-727, 2021 12 30.
Artículo en Inglés | MEDLINE | ID: mdl-34609492

RESUMEN

The cyclic guanosine monophosphate‒adenosine monophosphate synthase (cGAS)‒mediator of interferon response factor 3 activation/stimulator of interferon genes (MITA/STING) axis has emerged as a major pathway, which senses microbial or mislocated cellular DNA in the cytosol to trigger innate immune responses. cGAS senses cytosolic DNA without a preference of self- or nonself-DNA. How the cGAS‒MITA/STING axis is inactivated upon nuclear envelope breakdown (NEBD) at mitotic entry in vertebrate cells to avoid self-DNA sensing remains unclear until very recently. In this review, we summarize the recent advances on how cGAS responds to chromosomes upon NEBD and the mechanisms involved in the inactivation of the cGAS‒MITA/STING pathways in mitosis.


Asunto(s)
Proteínas de la Membrana , Transducción de Señal , Citosol/metabolismo , Inmunidad Innata/fisiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo
18.
Adv Sci (Weinh) ; 8(19): e2100779, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34363353

RESUMEN

In eukaryote cells, core components of chromatin, such as histones and DNA, are packaged in nucleus. Leakage of nuclear materials into cytosol will induce pathological effects. However, the underlying mechanisms remain elusive. Here, cytoplasmic localization of nuclear materials induced by chromatin dysregulation (CLIC) in mammalian cells is reported. H3K9me3 inhibition by small chemicals, HP1α knockdown, or knockout of H3K9 methylase SETDB1, induces formation of cytoplasmic puncta containing histones H3.1, H4 and cytosolic DNA, which in turn activates inflammatory genes and autophagic degradation. Autophagy deficiency rescues H3 degradation, and enhances the activation of inflammatory genes. MRE11, a subunit of MRN complex, enters cytoplasm after heterochromatin dysregulation. Deficiency of MRE11 or NBS1, but not RAD50, inhibits CLIC puncta in cytosol. MRE11 depletion represses tumor growth enhanced by HP1α deficiency, suggesting a connection between CLIC and tumorigenesis. This study reveals a novel pathway that heterochromatin dysregulation induces translocation of nuclear materials into cytoplasm, which is important for inflammatory diseases and cancer.


Asunto(s)
Citoplasma/genética , Citoplasma/metabolismo , Epigénesis Genética/genética , Histonas/genética , Histonas/metabolismo , Animales , Masculino , Ratones , Ratones Endogámicos BALB C , Modelos Animales , Factores de Transcripción/genética
19.
Nat Commun ; 12(1): 4227, 2021 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-34244482

RESUMEN

Glycine decarboxylase (GLDC) is a key enzyme of glycine cleavage system that converts glycine into one-carbon units. GLDC is commonly up-regulated and plays important roles in many human cancers. Whether and how GLDC is regulated by post-translational modifications is unknown. Here we report that mechanistic target of rapamycin complex 1 (mTORC1) signal inhibits GLDC acetylation at lysine (K) 514 by inducing transcription of the deacetylase sirtuin 3 (SIRT3). Upon inhibition of mTORC1, the acetyltransferase acetyl-CoA acetyltransferase 1 (ACAT1) catalyzes GLDC K514 acetylation. This acetylation of GLDC impairs its enzymatic activity. In addition, this acetylation of GLDC primes for its K33-linked polyubiquitination at K544 by the ubiquitin ligase NF-X1, leading to its degradation by the proteasomal pathway. Finally, we find that GLDC K514 acetylation inhibits glycine catabolism, pyrimidines synthesis and glioma tumorigenesis. Our finding reveals critical roles of post-translational modifications of GLDC in regulation of its enzymatic activity, glycine metabolism and tumorigenesis, and provides potential targets for therapeutics of cancers such as glioma.


Asunto(s)
Carcinogénesis/genética , Glioma/genética , Glicina-Deshidrogenasa (Descarboxilante)/metabolismo , Glicina/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Acetil-CoA C-Acetiltransferasa/metabolismo , Acetilación , Animales , Carcinogénesis/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Glioma/metabolismo , Glioma/patología , Células HEK293 , Humanos , Masculino , Ratones , Complejo de la Endopetidasa Proteasomal/metabolismo , Procesamiento Proteico-Postraduccional , Proteolisis , Pirimidinas/biosíntesis , Proteínas Represoras/metabolismo , Sirtuina 3/genética , Sirtuina 3/metabolismo , Activación Transcripcional , Ubiquitinación/genética , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Cell Discov ; 7(1): 46, 2021 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-34155193

RESUMEN

MITA (also known as STING) is an ER-located adaptor protein, which mediates DNA-triggered innate immune response and is critically involved in autoimmune diseases and tumorigenesis. MITA is regulated by post-translational modifications, but how post-transcriptional mechanisms are involved in the regulation of MITA is still largely unknown. Here, we identified the RNA-binding protein LUC7L2 as a negative regulator of DNA virus-triggered innate immune response. LUC7L2-deficient mice exhibited resistance to lethal herpes simplex virus 1 (HSV-1) infection and reduced HSV-1 loads in the brain. Mechanistically, LUC7L2 directly bound to intron 3 of MITA precursor messenger RNA, inhibited its splicing and promoted its nonsense-mediated decay, leading to its downregulation at protein level. LUC7L2-deficient cells had markedly increased MITA level, leading to heightened innate antiviral response. Finally, LUC7L2 was induced following HSV-1 infection. Our findings reveal a feedback negative post-transcriptional regulatory mechanism for regulation of MITA-mediated innate immune response to viral and aberrant cellular DNA.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...