Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
J Pathol ; 244(3): 323-333, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29214629

RESUMEN

Chronic muscle inflammation is a critical feature of Duchenne muscular dystrophy and contributes to muscle fibre injury and disease progression. Although previous studies have implicated T cells in the development of muscle fibrosis, little is known about their role during the early stages of muscular dystrophy. Here, we show that T cells are among the first cells to infiltrate mdx mouse dystrophic muscle, prior to the onset of necrosis, suggesting an important role in early disease pathogenesis. Based on our comprehensive analysis of the kinetics of the immune response, we further identify the early pre-necrotic stage of muscular dystrophy as the relevant time frame for T-cell-based interventions. We focused on protein kinase C θ (PKCθ, encoded by Prkcq), a critical regulator of effector T-cell activation, as a potential target to inhibit T-cell activity in dystrophic muscle. Lack of PKCθ not only reduced the frequency and number of infiltrating T cells but also led to quantitative and qualitative changes in the innate immune cell infiltrate in mdx/Prkcq-/- muscle. These changes were due to the inhibition of T cells, since PKCθ was necessary for T-cell but not for myeloid cell infiltration of acutely injured muscle. Targeting T cells with a PKCθ inhibitor early in the disease process markedly diminished the size of the inflammatory cell infiltrate and resulted in reduced muscle damage. Moreover, diaphragm necrosis and fibrosis were also reduced following treatment. Overall, our findings identify the early T-cell infiltrate as a therapeutic target and highlight the potential of PKCθ inhibition as a therapeutic approach to muscular dystrophy. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Quimiotaxis de Leucocito/efectos de los fármacos , Diafragma/efectos de los fármacos , Distrofia Muscular Animal/prevención & control , Proteína Quinasa C-theta/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Linfocitos T/efectos de los fármacos , Animales , Diafragma/enzimología , Diafragma/inmunología , Diafragma/patología , Modelos Animales de Enfermedad , Fibrosis , Inmunidad Innata/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Ratones Noqueados , Distrofia Muscular Animal/enzimología , Distrofia Muscular Animal/inmunología , Distrofia Muscular Animal/patología , Necrosis , Proteína Quinasa C-theta/deficiencia , Proteína Quinasa C-theta/genética , Proteína Quinasa C-theta/metabolismo , Índice de Severidad de la Enfermedad , Transducción de Señal/efectos de los fármacos , Linfocitos T/enzimología , Linfocitos T/inmunología , Factores de Tiempo
2.
Cancers (Basel) ; 15(5)2023 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-36900414

RESUMEN

BACKGROUND: Pituitary adenoma (PA) bone invasion results in adverse outcomes, such as reduced rates of complete surgical resection and biochemical remission as well as increased recurrence rates, though few studies have been conducted. METHODS: We collected clinical specimens of PAs for staining and statistical analysis. Evaluation of the ability of PA cells to induce monocyte-osteoclast differentiation by coculturing PA cells with RAW264.7 in vitro. An in vivo model of bone invasion was used to simulate the process of bone erosion and evaluate the effect of different interventions in alleviating bone invasion. RESULTS: We found an overactivation of osteoclasts in bone-invasive PAs and concomitant aggregation of inflammatory factors. Furthermore, activation of PKCθ in PAs was established as a central signaling promoting PA bone invasion through the PKCθ/NF-κB/IL-1ß pathway. By inhibiting PKCθ and blocking IL1ß, we were able to significantly reverse bone invasion in an in vivo study. Meanwhile, we also found that celastrol, as a natural product, can obviously reduce the secretion of IL-1ß as well as alleviate the progression of bone invasion. CONCLUSIONS: By activating the PKCθ/NF-κB/IL-1ß pathway, pituitary tumors are able to induce monocyte-osteoclast differentiation in a paracrine manner and promote bone invasion, which can be alleviated by celastrol.

3.
Cells ; 11(22)2022 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-36428957

RESUMEN

The underlying mechanisms for statin-induced myopathy (SIM) are still equivocal. In this study, we employ Drosophila melanogaster to dissect possible underlying mechanisms for SIM. We observe that chronic fluvastatin treatment causes reduced general locomotion activity and climbing ability. In addition, transmission microscopy of dissected skeletal muscles of fluvastatin-treated flies reveals strong myofibrillar damage, including increased sarcomere lengths and Z-line streaming, which are reminiscent of myopathy, along with fragmented mitochondria of larger sizes, most of which are round-like shapes. Furthermore, chronic fluvastatin treatment is associated with impaired lipid metabolism and insulin signalling. Mechanistically, knockdown of the statin-target Hmgcr in the skeletal muscles recapitulates fluvastatin-induced mitochondrial phenotypes and lowered general locomotion activity; however, it was not sufficient to alter sarcomere length or elicit myofibrillar damage compared to controls or fluvastatin treatment. Moreover, we found that fluvastatin treatment was associated with reduced expression of the skeletal muscle chloride channel, ClC-a (Drosophila homolog of CLCN1), while selective knockdown of skeletal muscle ClC-a also recapitulated fluvastatin-induced myofibril damage and increased sarcomere lengths. Surprisingly, exercising fluvastatin-treated flies restored ClC-a expression and normalized sarcomere lengths, suggesting that fluvastatin-induced myofibrillar phenotypes could be linked to lowered ClC-a expression. Taken together, these results may indicate the potential role of ClC-a inhibition in statin-associated muscular phenotypes. This study underlines the importance of Drosophila melanogaster as a powerful model system for elucidating the locomotion and muscular phenotypes, promoting a better understanding of the molecular mechanisms underlying SIM.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas , Enfermedades Musculares , Animales , Humanos , Drosophila melanogaster/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/efectos adversos , Canales de Cloruro/metabolismo , Fluvastatina/efectos adversos , Enfermedades Musculares/genética , Drosophila/metabolismo , Locomoción , Fenotipo
4.
Biomolecules ; 11(2)2021 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-33562506

RESUMEN

Protein Kinase C theta (PKCθ) is a serine/threonine kinase that belongs to the novel PKC subfamily. In normal tissue, its expression is restricted to skeletal muscle cells, platelets and T lymphocytes in which PKCθ controls several essential cellular processes such as survival, proliferation and differentiation. Particularly, PKCθ has been extensively studied for its role in the immune system where its translocation to the immunological synapse plays a critical role in T cell activation. Beyond its physiological role in immune responses, increasing evidence implicates PKCθ in the pathology of various diseases, especially autoimmune disorders and cancers. In this review, we discuss the implication of PKCθ in various types of cancers and the PKCθ-mediated signaling events controlling cancer initiation and progression. In these types of cancers, the high PKCθ expression leads to aberrant cell proliferation, migration and invasion resulting in malignant phenotype. The recent development and application of PKCθ inhibitors in the context of autoimmune diseases could benefit the emergence of treatment for cancers in which PKCθ has been implicated.


Asunto(s)
Enfermedades Autoinmunes/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias/genética , Proteína Quinasa C-theta/genética , Subunidades de Proteína/genética , Transducción de Señal/genética , Animales , Enfermedades Autoinmunes/enzimología , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Movimiento Celular , Proliferación Celular , Humanos , FN-kappa B/genética , FN-kappa B/metabolismo , Invasividad Neoplásica , Metástasis de la Neoplasia , Neoplasias/enzimología , Neoplasias/inmunología , Neoplasias/patología , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteína Quinasa C-theta/química , Proteína Quinasa C-theta/inmunología , Subunidades de Proteína/química , Subunidades de Proteína/inmunología , Células Th17/inmunología , Células Th17/patología , Células Th2/inmunología , Células Th2/patología
5.
Elife ; 102021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-34110283

RESUMEN

The nuclear pore complex (NPC) is the sole and selective gateway for nuclear transport, and its dysfunction has been associated with many diseases. The metazoan NPC subcomplex RanBP2, which consists of RanBP2 (Nup358), RanGAP1-SUMO1, and Ubc9, regulates the assembly and function of the NPC. The roles of immune signaling in regulation of NPC remain poorly understood. Here, we show that in human and murine T cells, following T-cell receptor (TCR) stimulation, protein kinase C-θ (PKC-θ) directly phosphorylates RanGAP1 to facilitate RanBP2 subcomplex assembly and nuclear import and, thus, the nuclear translocation of AP-1 transcription factor. Mechanistically, TCR stimulation induces the translocation of activated PKC-θ to the NPC, where it interacts with and phosphorylates RanGAP1 on Ser504 and Ser506. RanGAP1 phosphorylation increases its binding affinity for Ubc9, thereby promoting sumoylation of RanGAP1 and, finally, assembly of the RanBP2 subcomplex. Our findings reveal an unexpected role of PKC-θ as a direct regulator of nuclear import and uncover a phosphorylation-dependent sumoylation of RanGAP1, delineating a novel link between TCR signaling and assembly of the RanBP2 NPC subcomplex.


Asunto(s)
Proteínas Activadoras de GTPasa , Chaperonas Moleculares , Proteínas de Complejo Poro Nuclear , Receptores de Antígenos de Linfocitos T/metabolismo , Proteína SUMO-1 , Enzimas Ubiquitina-Conjugadoras , Animales , Proteínas Activadoras de GTPasa/química , Proteínas Activadoras de GTPasa/metabolismo , Humanos , Ratones , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Poro Nuclear/química , Poro Nuclear/metabolismo , Proteínas de Complejo Poro Nuclear/química , Proteínas de Complejo Poro Nuclear/metabolismo , Fosforilación , Proteína Quinasa C-theta/química , Proteína Quinasa C-theta/metabolismo , Proteína SUMO-1/química , Proteína SUMO-1/metabolismo , Transducción de Señal/fisiología , Enzimas Ubiquitina-Conjugadoras/química , Enzimas Ubiquitina-Conjugadoras/metabolismo
6.
Front Immunol ; 7: 24, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26870040

RESUMEN

B7 proteins CD80 (B7-1) and CD86 (B7-2) are expressed on most antigen-presenting cells and provide critical co-stimulatory or inhibitory input to T cells via their T-cell-expressed receptors: CD28 and CTLA-4. CD28 is expressed on effector T cells and regulatory T cells (Tregs), and CD28-dependent signals are required for optimum activation of effector T cell functions. CD28 ligation on effector T cells leads to formation of distinct molecular patterns and induction of cytoskeletal rearrangements at the immunological synapse (IS). CD28 plays a critical role in recruitment of protein kinase C (PKC)-θ to the effector T cell IS. CTLA-4 is constitutively expressed on the surface of Tregs, but it is expressed on effector T cells only after activation. As CTLA-4 binds to B7 proteins with significantly higher affinity than CD28, B7 ligand recognition by cells expressing both receptors leads to displacement of CD28 and PKC-θ from the IS. In Tregs, B7 ligand recognition leads to recruitment of CTLA-4 and PKC-η to the IS. CTLA-4 plays a role in regulation of T effector and Treg IS stability and cell motility. Due to their important roles in regulating T-cell-mediated responses, B7 receptors are emerging as important drug targets in oncology. In this review, we present an integrated summary of current knowledge about the role of B7 family receptor-ligand interactions in the regulation of spatial and temporal IS dynamics in effector and Tregs.

7.
Front Immunol ; 6: 562, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26594212

RESUMEN

Alternative splicing of nuclear pre-mRNA is essential for generating protein diversity and regulating gene expression. While many immunologically relevant genes undergo alternative splicing, the role of regulated splicing in T cell immune responses is largely unexplored, and the signaling pathways and splicing factors that regulate alternative splicing in T cells are poorly defined. Here, we show using a combination of Jurkat T cells, human primary T cells, and ex vivo naïve and effector virus-specific T cells isolated after influenza A virus infection that SC35 phosphorylation is induced in response to stimulatory signals. We show that SC35 colocalizes with RNA polymerase II in activated T cells and spatially overlaps with H3K27ac and H3K4me3, which mark transcriptionally active genes. Interestingly, SC35 remains coupled to the active histone marks in the absence of continuing stimulatory signals. We show for the first time that nuclear PKC-θ co-exists with SC35 in the context of the chromatin template and is a key regulator of SC35 in T cells, directly phosphorylating SC35 peptide residues at RNA recognition motif and RS domains. Collectively, our findings suggest that nuclear PKC-θ is a novel regulator of the key splicing factor SC35 in T cells.

8.
Front Immunol ; 3: 260, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22969762

RESUMEN

We recently provided the first description of a nuclear mechanism used by Protein Kinase C-theta (PKC-θ) to mediate T cell gene expression. In this mode, PKC-θ tethers to chromatin to form an active nuclear complex by interacting with proteins including RNA polymerase II, the histone kinase MSK-1, the demethylase LSD1, and the adaptor molecule 14-3-3ζ at regulatory regions of inducible immune response genes. Moreover, our genome-wide analysis identified many novel PKC-θ target genes and microRNAs implicated in T cell development, differentiation, apoptosis, and proliferation. We have expanded our ChIP-on-chip analysis and have now identified a transcription factor motif containing NF-κB binding sites that may facilitate recruitment of PKC-θ to chromatin at coding genes. Furthermore, NF-κB association with chromatin appears to be a prerequisite for the assembly of the PKC-θ active complex. In contrast, a distinct NF-κB-containing module appears to operate at PKC-θ targeted microRNA genes, and here NF-κB negatively regulates microRNA gene transcription. Our efforts are also focusing on distinguishing between the nuclear and cytoplasmic functions of PKCs to ascertain how these kinases may synergize their roles as both cytoplasmic signaling proteins and their functions on the chromatin template, together enabling rapid induction of eukaryotic genes. We have identified an alternative sequence within PKC-θ that appears to be important for nuclear translocation of this kinase. Understanding the molecular mechanisms used by signal transduction kinases to elicit specific and distinct transcriptional programs in T cells will enable scientists to refine current therapeutic strategies for autoimmune diseases and cancer.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA