Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell ; 187(4): 831-845.e19, 2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-38301645

RESUMO

The paraneoplastic Ma antigen (PNMA) proteins are associated with cancer-induced paraneoplastic syndromes that present with an autoimmune response and neurological symptoms. Why PNMA proteins are associated with this severe autoimmune disease is unclear. PNMA genes are predominantly expressed in the central nervous system and are ectopically expressed in some tumors. We show that PNMA2, which has been co-opted from a Ty3 retrotransposon, encodes a protein that is released from cells as non-enveloped virus-like capsids. Recombinant PNMA2 capsids injected into mice induce autoantibodies that preferentially bind external "spike" PNMA2 capsid epitopes, whereas a capsid-assembly-defective PNMA2 protein is not immunogenic. PNMA2 autoantibodies in cerebrospinal fluid of patients with anti-Ma2 paraneoplastic disease show similar preferential binding to spike capsid epitopes. PNMA2 capsid-injected mice develop learning and memory deficits. These observations suggest that PNMA2 capsids act as an extracellular antigen, capable of generating an autoimmune response that results in neurological deficits.


Assuntos
Antígenos de Neoplasias , Neoplasias , Proteínas do Tecido Nervoso , Síndromes Paraneoplásicas do Sistema Nervoso , Animais , Humanos , Camundongos , Autoanticorpos , Capsídeo/metabolismo , Epitopos , Neoplasias/complicações , Síndromes Paraneoplásicas do Sistema Nervoso/metabolismo , Síndromes Paraneoplásicas do Sistema Nervoso/patologia , Antígenos de Neoplasias/metabolismo , Proteínas do Tecido Nervoso/metabolismo
2.
Cell Mol Immunol ; 21(3): 260-274, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38233562

RESUMO

Metabolic flexibility has emerged as a critical determinant of CD8+ T-cell antitumor activity, yet the mechanisms driving the metabolic flexibility of T cells have not been determined. In this study, we investigated the influence of the nuclear cap-binding complex (CBC) adaptor protein ARS2 on mature T cells. In doing so, we discovered a novel signaling axis that endows activated CD8+ T cells with flexibility of glucose catabolism. ARS2 upregulation driven by CD28 signaling reinforced splicing factor recruitment to pre-mRNAs and affected approximately one-third of T-cell activation-induced alternative splicing events. Among these effects, the CD28-ARS2 axis suppressed the expression of the M1 isoform of pyruvate kinase in favor of PKM2, a key determinant of CD8+ T-cell glucose utilization, interferon gamma production, and antitumor effector function. Importantly, PKM alternative splicing occurred independently of CD28-driven PI3K pathway activation, revealing a novel means by which costimulation reprograms glucose metabolism in CD8+ T cells.


Assuntos
Processamento Alternativo , Antígenos CD28 , Antígenos CD28/metabolismo , Processamento Alternativo/genética , Fosfatidilinositol 3-Quinases/metabolismo , Linfócitos T CD8-Positivos , Glucose/metabolismo
3.
Connect Tissue Res ; 65(1): 26-40, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37898909

RESUMO

PURPOSE/AIM: Cartilage injury and subsequent osteoarthritis (OA) are debilitating conditions affecting millions worldwide. As there are no cures for these ailments, novel therapies are needed to suppress disease pathogenesis. Given that joint injuries are known to produce damage-associated molecular patterns (DAMPs), our central premise is that the Toll-like receptor 4 (TLR4) pathway is a principal driver in the early response to cartilage damage and subsequent pathology. We postulate that TLR4 activation is initiated/perpetuated by DAMPs released following joint damage. Thus, antagonism of the TLR4 pathway immediately after injury may suppress the development of joint surface defects. MATERIALS AND METHODS: Two groups were utilized: (1) 8-week-old, male C57BL6 mice treated systemically with a known TLR4 antagonist and (2) mice injected with vehicle control. A full-depth cartilage lesion on the midline of the patellofemoral groove was created in the right knee of each mouse. The left knee was used as a sham surgery control. Gait changes were evaluated over 4 weeks using a quantitative gait analysis system. At harvest, knee joints were processed for pathologic assessment, Nanostring® transcript expression, and immunohistochemistry (IHC). RESULTS: Short-term treatment with a TLR4 antagonist at 14-days significantly improved relevant gait parameters; improved cartilage metrics and modified Mankin scores were also seen. Additionally, mRNA expression and IHC showed reduced expression of inflammatory mediators in animals treated with the TLR4 antagonist. CONCLUSIONS: Collectively, this work demonstrates that systemic treatment with a TLR4 antagonist is protective to further cartilage damage 14-days post-injury in a murine model of induced disease.


Assuntos
Doenças das Cartilagens , Cartilagem Articular , Osteoartrite do Joelho , Osteoartrite , Camundongos , Masculino , Animais , Receptor 4 Toll-Like , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Osteoartrite/patologia , Cartilagem/patologia , Doenças das Cartilagens/patologia , Cartilagem Articular/patologia , Osteoartrite do Joelho/patologia
4.
Elife ; 122023 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-36951542

RESUMO

Amyotrophic Lateral Sclerosis (ALS) is a fatal neurodegenerative disease characterized by progressive motor neuron dysfunction and loss. A portion of ALS cases are caused by mutation of the proteasome shuttle factor Ubiquilin 2 (UBQLN2), but the molecular pathway leading from UBQLN2 dysfunction to disease remains unclear. Here, we demonstrate that UBQLN2 regulates the domesticated gag-pol retrotransposon 'paternally expressed gene 10 (PEG10)' in human cells and tissues. In cells, the PEG10 gag-pol protein cleaves itself in a mechanism reminiscent of retrotransposon self-processing to generate a liberated 'nucleocapsid' fragment, which uniquely localizes to the nucleus and changes the expression of genes involved in axon remodeling. In spinal cord tissue from ALS patients, PEG10 gag-pol is elevated compared to healthy controls. These findings implicate the retrotransposon-like activity of PEG10 as a contributing mechanism in ALS through the regulation of gene expression, and restraint of PEG10 as a primary function of UBQLN2.


Assuntos
Esclerose Amiotrófica Lateral , Doenças Neurodegenerativas , Humanos , Esclerose Amiotrófica Lateral/genética , Retroelementos , Doenças Neurodegenerativas/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Neurônios Motores/metabolismo , Mutação , Proteínas Relacionadas à Autofagia/metabolismo , Ubiquitinas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo
5.
Immunity ; 54(12): 2772-2783.e5, 2021 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-34788602

RESUMO

Humoral immunity is essential for protection against pathogens, emphasized by the prevention of 2-3 million deaths worldwide annually by childhood immunizations. Long-term protective immunity is dependent on the continual production of neutralizing antibodies by the subset of long-lived plasma cells (LLPCs). LLPCs are not intrinsically long-lived, but require interaction with LLPC niche stromal cells for survival. However, it remains unclear which and how these interactions sustain LLPC survival and long-term humoral immunity. We now have found that the immunosuppressive enzyme indoleamine 2,3- dioxygenase 1 (IDO1) is required to sustain antibody responses and LLPC survival. Activation of IDO1 occurs upon the engagement of CD80/CD86 on the niche dendritic cells by CD28 on LLPC. Kynurenine, the product of IDO1 catabolism, activates the aryl hydrocarbon receptor in LLPC, reinforcing CD28 expression and survival signaling. These findings expand the immune function of IDO1 and uncover a novel pathway for sustaining LLPC survival and humoral immunity.


Assuntos
Células Dendríticas/imunologia , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Plasmócitos/imunologia , Animais , Anticorpos Neutralizantes/metabolismo , Antígeno B7-1/metabolismo , Antígenos CD28/metabolismo , Autorrenovação Celular , Sobrevivência Celular , Células Cultivadas , Feminino , Imunidade Humoral , Memória Imunológica , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Camundongos , Camundongos Knockout
6.
Cell Rep ; 31(12): 107815, 2020 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-32579940

RESUMO

Durable humoral immunity against epidemic infectious disease requires the survival of long-lived plasma cells (LLPCs). LLPC longevity is dependent on metabolic programs distinct from short-lived plasma cells (SLPCs); however, the mechanistic basis for this difference is unclear. We have previously shown that CD28, the prototypic T cell costimulatory receptor, is expressed on both LLPCs and SLPCs but is essential only for LLPC survival. Here we show that CD28 transduces pro-survival signaling specifically in LLPCs through differential SLP76 expression. CD28 signaling in LLPCs increased glucose uptake, mitochondrial mass/respiration, and reactive oxygen species (ROS) production. Unexpectedly, CD28-mediated regulation of mitochondrial respiration, NF-κB activation, and survival was ROS dependent. IRF4, a target of NF-κB, was upregulated by CD28 activation in LLPCs and decreased IRF4 levels correlated with decreased glucose uptake, mitochondrial mass, ROS, and CD28-mediated survival. Altogether, these data demonstrate that CD28 signaling induces a ROS-dependent metabolic program required for LLPC survival.


Assuntos
Antígenos CD28/metabolismo , Plasmócitos/citologia , Plasmócitos/metabolismo , Animais , Células da Medula Óssea/citologia , Respiração Celular , Sobrevivência Celular , Feminino , Glucose/metabolismo , Humanos , Fatores Reguladores de Interferon/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Baço/citologia
7.
Stem Cell Res ; 43: 101710, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31986485

RESUMO

The RNA binding protein ARS2 is highly expressed in hematopoietic progenitor populations and is required for adult hematopoiesis. Recent molecular studies found that ARS2 coordinates interactions between nascent RNA polymerase II transcripts and downstream RNA processing machineries, yet how such interactions influence hematopoiesis remains largely unknown. Techniques to differentiate embryonic stem cells (ESC) to hematopoietic progenitor cells (HPC) and mature blood cells have increased molecular understanding of hematopoiesis. Taking such an in vitro approach to examine the influence of ARS2 on hematopoiesis, we found that ARS2 suppresses expression of some HSC signature genes and differentiation of ESC to a HPC population (CSMD-HPC) identified by markers expressed on bone marrow resident hematopoietic stem cells. In line with ARS2's ability to promote proliferation of cultured cells, ARS2 knockout ESC showed limited expansion and yielded less CSMD-HPC than wild-type ESC. In contrast, transient ARS2 knockdown led to doubling the number of CSMD-HPC generated per ESC without affecting further differentiation into mature T-cells. Overall, data indicate that ARS2 negatively regulates early hematopoietic differentiation of ESC, in stark contrast to its supportive role in adult hematopoiesis. Consequently, manipulation of ARS2 expression and/or function has potential utility in hematopoietic cell engineering and regenerative medicine.


Assuntos
Proteínas de Ligação a DNA/genética , Células-Tronco Embrionárias Murinas/metabolismo , Proteínas de Ligação a RNA/metabolismo , Fatores de Transcrição/genética , Animais , Diferenciação Celular , Células Cultivadas , Camundongos
8.
Exp Hematol ; 64: 45-58.e9, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29775646

RESUMO

Recent biochemical characterization of arsenic resistance protein 2 (Ars2) has established it as central in determining the fate of nascent ribonucleic acid (RNA) polymerase II (RNAPII) transcripts. Through interactions with the nuclear 5'-7-methylguanosine cap-binding complex, Ars2 promotes cotranscriptional processing coupled with nuclear export or degradation of several classes of RNAPII transcripts, allowing for gene expression programs that facilitate rapid and sustained proliferation of immortalized cells in culture. However, rapidly dividing cells in culture do not represent the physiological condition of the vast majority of cells in an adult mammal. To examine functions of Ars2 in a physiological setting, we generated inducible Ars2 knockout mice and found that deletion of Ars2 from adult mice resulted in defective hematopoiesis in bone marrow and thymus. Importantly, only some of this defect could be explained by the requirement of Ars2 for rapid proliferation, which we found to be cell-type specific in vivo. Rather, Ars2 was required for survival of developing thymocytes and for limiting differentiation of bone marrow resident long-term hematopoietic stem cells. As a result, Ars2 knockout led to rapid thymic involution and loss of the ability of mice to regenerate peripheral blood after myeloablation. These in vivo data demonstrate that Ars2 expression is important at several steps of hematopoiesis, likely because Ars2 acts on gene expression programs underlying essential cell fate decisions such as the decision to die,proliferate, or differentiate.


Assuntos
Hematopoese/fisiologia , Proteínas Nucleares/fisiologia , Fatores de Transcrição/fisiologia , Animais , Apoptose , Divisão Celular Assimétrica , Medula Óssea/patologia , Medula Óssea/fisiologia , Autorrenovação Celular , Deleção Clonal , Ensaio de Unidades Formadoras de Colônias , Proteínas de Ligação a DNA , Regulação da Expressão Gênica , Hematopoese/genética , Células-Tronco Hematopoéticas/patologia , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Proteínas Nucleares/deficiência , Especificidade de Órgãos , Quimera por Radiação , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Recombinantes/metabolismo , Células Estromais/fisiologia , Timócitos/citologia , Timo/patologia , Fatores de Transcrição/deficiência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...