Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 12: 697588, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34305934

RESUMO

The Toll-interleukin-1 Receptor (TIR) domain-containing adaptor protein (TIRAP) represents a key intracellular signalling molecule regulating diverse immune responses. Its capacity to function as an adaptor molecule has been widely investigated in relation to Toll-like Receptor (TLR)-mediated innate immune signalling. Since the discovery of TIRAP in 2001, initial studies were mainly focused on its role as an adaptor protein that couples Myeloid differentiation factor 88 (MyD88) with TLRs, to activate MyD88-dependent TLRs signalling. Subsequent studies delineated TIRAP's role as a transducer of signalling events through its interaction with non-TLR signalling mediators. Indeed, the ability of TIRAP to interact with an array of intracellular signalling mediators suggests its central role in various immune responses. Therefore, continued studies that elucidate the molecular basis of various TIRAP-protein interactions and how they affect the signalling magnitude, should provide key information on the inflammatory disease mechanisms. This review summarizes the TIRAP recruitment to activated receptors and discusses the mechanism of interactions in relation to the signalling that precede acute and chronic inflammatory diseases. Furthermore, we highlighted the significance of TIRAP-TIR domain containing binding sites for several intracellular inflammatory signalling molecules. Collectively, we discuss the importance of the TIR domain in TIRAP as a key interface involved in protein interactions which could hence serve as a therapeutic target to dampen the extent of acute and chronic inflammatory conditions.


Assuntos
Inflamação/imunologia , Glicoproteínas de Membrana/imunologia , Receptores de Interleucina-1/imunologia , Tirosina Quinase da Agamaglobulinemia/imunologia , Tirosina Quinase da Agamaglobulinemia/metabolismo , Animais , Proteínas de Transporte/imunologia , Proteínas de Transporte/metabolismo , Classe Ia de Fosfatidilinositol 3-Quinase/imunologia , Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Humanos , Imunidade Inata , Inflamação/metabolismo , Glicoproteínas de Membrana/metabolismo , Modelos Biológicos , Mapas de Interação de Proteínas , Proteína Quinase C-delta/imunologia , Proteína Quinase C-delta/metabolismo , Receptor para Produtos Finais de Glicação Avançada/imunologia , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Receptores de Interleucina-1/metabolismo , Transdução de Sinais/imunologia
2.
FASEB J ; 34(2): 2497-2510, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31908004

RESUMO

Sepsis is a leading cause of morbidity and mortality in intensive care units. Previously, we identified Protein Kinase C-delta (PKCδ) as an important regulator of the inflammatory response in sepsis. An important issue in development of anti-inflammatory therapeutics is the risk of immunosuppression and inability to effectively clear pathogens. In this study, we investigated whether PKCδ inhibition prevented organ dysfunction and improved survival without compromising pathogen clearance. Sprague Dawley rats underwent sham surgery or cecal ligation and puncture (CLP) to induce sepsis. Post-surgery, PBS or a PKCδ inhibitor (200µg/kg) was administered intra-tracheally (IT). At 24 hours post-CLP, there was evidence of lung and kidney dysfunction. PKCδ inhibition decreased leukocyte influx in these organs, decreased endothelial permeability, improved gas exchange, and reduced blood urea nitrogen/creatinine ratios indicating organ protection. PKCδ inhibition significantly decreased bacterial levels in the peritoneal cavity, spleen and blood but did not exhibit direct bactericidal properties. Peritoneal chemokine levels, neutrophil numbers, or macrophage phenotypes were not altered by PKCδ inhibition. Peritoneal macrophages isolated from PKCδ inhibitor-treated septic rats demonstrated increased bacterial phagocytosis. Importantly, PKCδ inhibition increased survival. Thus, PKCδ inhibition improved survival and improved survival was associated with increased phagocytic activity, enhanced pathogen clearance, and decreased organ injury.


Assuntos
Bactérias/imunologia , Inibidores Enzimáticos/farmacologia , Macrófagos Peritoneais , Neutrófilos , Proteína Quinase C-delta/antagonistas & inibidores , Sepse , Animais , Quimiocinas , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/patologia , Masculino , Neutrófilos/imunologia , Neutrófilos/patologia , Fagocitose/efeitos dos fármacos , Proteína Quinase C-delta/imunologia , Ratos , Ratos Sprague-Dawley , Sepse/tratamento farmacológico , Sepse/imunologia , Sepse/microbiologia , Sepse/patologia
3.
Fish Shellfish Immunol ; 84: 1100-1107, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30408601

RESUMO

A primitive adaptive immune system has recently been suggested to be present in a basal chordate amphioxus (Branchiostoma belcheri, Bb), making it an ideal model for studying the origin of adaptive immune. The novel protein kinase C isoform PKC-θ, but not its closest isoform PKC-δ, plays a critical role for mammalian T-cell activation via translocation to immunological synapse (IS) mediated by a unique PKC-θ V3 domain containing one PxxP motif. To understand the evolution of this unique PKC-θ V3 domain and the primitive adaptive immune system in amphioxus, we comparatively studied the orthologs of PKC-δ and -θ from amphioxus and other species. Phylogenetic analysis showed BbPKC-δ/θ to be the common ancestor of vertebrate PKC-δ and PKC-θ, with a V3 domain containing two PxxP motifs. One motif is conserved in both zebrafish and mammalian PKC-θ but is absent in PKC-δ V3 domain of these species, and has already emerged in drosophila PKC-δ. The other non-conserved motif emerged in BbPKC-δ/θ, and only retained in Danio rerio PKC-δ (DrPKC-δ) but lost in mammalian PKC-δ and -θ. Comparative analyses of the sequence and function of BbPKC-δ/θ, DrPKC-δ, DrPKC-θ and Homo sapiens PKC-θ (HsPKC-θ) in IS translocation and T-cell receptor (TCR)-induced NF-κB activation revealed that retention of the conserved PxxP motif and loss of the non-conserved PxxP motif in mammalian PKC-θ and loss of both PxxP motifs in mammalian PKC-δ accomplish the unique function of PKC-θ in T cells. Together, this study suggests an evolutionary mechanism for PKC-θ unique V3 and reveals BbPKC-δ/θ is the common ancestor of PKC-δ and -θ with a functional proto-V3 domain, supplying new evidence for the existence of primitive adaptive immune system in amphioxus.


Assuntos
Imunidade Adaptativa/genética , Doenças dos Peixes/imunologia , Regulação da Expressão Gênica/imunologia , Anfioxos/genética , Anfioxos/imunologia , Proteína Quinase C-delta/genética , Proteína Quinase C-delta/imunologia , Proteína Quinase C-theta/genética , Proteína Quinase C-theta/imunologia , Sequência de Aminoácidos , Animais , Perfilação da Expressão Gênica/veterinária , Anfioxos/enzimologia , Filogenia , Proteína Quinase C-delta/química , Proteína Quinase C-theta/química , Alinhamento de Sequência/veterinária
4.
Eur Rev Med Pharmacol Sci ; 22(13): 4228-4237, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-30024612

RESUMO

OBJECTIVE: To investigate the effect of PKC δ gene on the anti-tuberculosis activity of macrophages and the mechanism. MATERIALS AND METHODS: Bone marrow cells of PKC δ knockout mice and wild-type mice were cultured and L929 cells were induced to differentiate into macrophages. Lipopolysaccharide (LPS) and trehalose 6,6'-dimycolate (TDM) were used to stimulate macrophages respectively. After 24 and 96 hours, cells and the supernatant were collected to evaluate the inflammatory cytokines produced by macrophages using ELISA method. Real-time PCR was performed to detect the expression of macrophage mRNA level and nitric oxide (NO) production of macrophages was measured by NO assay. RESULTS: The results showed that, after TDB stimulation, IL-1ß, IL-6, and other cytokines, as well as NO produced by macrophages of PKC δ knockout mice, were significantly decreased (p < 0.01) compared with the wild-type mice. In PKC δ knockout macrophages, the above protein-coding genes were also decreased significantly at the transcriptional level (p < 0.01). CONCLUSIONS: PKC δ can enhance the anti-tuberculosis capacity of macrophages by inducing to the release of inflammatory factors by macrophages.


Assuntos
Mediadores da Inflamação/metabolismo , Macrófagos/imunologia , Mycobacterium tuberculosis/imunologia , Proteína Quinase C-delta/metabolismo , Tuberculose/imunologia , Animais , Linhagem Celular , Modelos Animais de Doenças , Glicolipídeos/imunologia , Humanos , Mediadores da Inflamação/imunologia , Lipopolissacarídeos/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Cultura Primária de Células , Proteína Quinase C-delta/genética , Proteína Quinase C-delta/imunologia , Tuberculose/microbiologia
5.
Int Immunopharmacol ; 48: 211-218, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28528205

RESUMO

Inflammation could be described as a physiological response of the body to tissue injury, pathogen invasion, and irritants. During the inflammatory phase, cells of both the innate as well as adaptive immune system are activated and recruited to the site of inflammation. These mediators are downstream targets for the transcription factors; activator protein-1 (AP1), nuclear factor kappa-light-chain-enhancer (NF-κB), signal transducers and activators of transcription factors (STAT1), as well as interferon regulatory factors (IRFs), which control the expression of most immunomodulatory genes. There is a significant increase in active p38 mitogen-activated protein kinase (p38MAK) immediately after lipopolysaccharide (LPS) stimulation, which results in the activation of AP-1 transcription factor and expression of proinflammatory cytokines, IL-12 and IL-23. We studied the novel mechanism of p38 MAPK activation through the formation of a heterotrimeric complex of Protein kinase C delta type (PKCδ), Toll-Interleukin 1 Receptor (TIR) Domain Containing Adaptor Protein (TIRAP), and p38 proteins. TIRAP serves as an adaptor molecule which brings PKCδ and p38 in close proximity. The complex facilitates the activation of p38MAPK by PKCδ. Therefore, we propose that disruption of the heterotrimeric complex may be a good strategy to dampen the inflammatory response. Structure-based design of small molecules or peptides targetting PKCδ-TIRAP or TIRAP-p38 interfaces would be beneficial for therapy in AP1 mediated inflammatory diseases.


Assuntos
Inflamação/imunologia , Glicoproteínas de Membrana/imunologia , Proteína Quinase C-delta/imunologia , Receptores de Interleucina-1/imunologia , Fator de Transcrição AP-1/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/imunologia , Animais , Inflamação/induzido quimicamente , Lipopolissacarídeos , Macrófagos/imunologia , Camundongos Endogâmicos C57BL
6.
J Leukoc Biol ; 100(5): 919-926, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27154354

RESUMO

This review summarizes the current state of knowledge regarding the roles of the signaling adapter protein tumor necrosis factor receptor (TNFR)-associated factor 3 in regulating the functions of B and T lymphocytes. In B lymphocytes, TNFR-associated factor 3 inhibits signaling by TNFR superfamily receptors, Toll-like receptors, and interleukin-6R. In contrast, signaling to B cells by the virally encoded oncogenic protein latent membrane protein 1 is promoted by TNFR-associated factor 3. An important B cell-specific role for TNFR-associated factor 3 is the inhibition of homeostatic survival, directly relevant to the common occurrence of TNFR-associated factor 3 mutations in human B cell malignancies. TNFR-associated factor 3 was recently found to be a resident nuclear protein in B cells, where it interacts with and inhibits gene expression mediated by the cAMP response element-binding protein transcription complex, including expression of the prosurvival protein myeloid leukemia cell differentiation protein 1. In T lymphocytes, TNFR-associated factor 3 is required for normal signaling by the T cell antigen receptor, while inhibiting signaling by the interleukin-2 receptor. Cytoplasmic TNFR -associated factor 3 restrains nuclear factor-κB2 activation in both T and B cells. Clinical implications and future directions for the study of this context-dependent signaling regulator are discussed.


Assuntos
Linfócitos B/imunologia , Subpopulações de Linfócitos/imunologia , Linfócitos T/imunologia , Fator 3 Associado a Receptor de TNF/imunologia , Animais , Formação de Anticorpos , Antígenos CD40/imunologia , Linhagem Celular , Sobrevivência Celular , Previsões , Humanos , Imunidade Inata , Camundongos , Camundongos Knockout , NF-kappa B/metabolismo , Proteínas Nucleares/imunologia , Proteína Quinase C-delta/imunologia , Complexo Receptor-CD3 de Antígeno de Linfócitos T/imunologia , Receptores de Citocinas/imunologia , Fator 3 Associado a Receptor de TNF/deficiência , Proteínas da Matriz Viral/imunologia
7.
Mol Cell Biol ; 36(2): 320-9, 2016 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-26552704

RESUMO

Sphingosine-1-phosphate (S1P) is a bioactive lipid mediator that regulates many processes in inflammation and cancer. S1P is a ligand for five G-protein-coupled receptors, S1PR1 to -5, and also has important intracellular actions. Previously, we showed that intracellular S1P is involved in tumor necrosis factor alpha (TNF)-induced NF-κB activation in melanoma cell lines that express filamin A (FLNA). Here, we show that extracellular S1P activates NF-κB only in melanoma cells that lack FLNA. In these cells, S1P, but not TNF, promotes IκB kinase (IKK) and p65 phosphorylation, IκBα degradation, p65 nuclear translocation, and NF-κB reporter activity. NF-κB activation induced by S1P was mediated via S1PR1 and S1PR2. Exogenous S1P enhanced the phosphorylation of protein kinase Cδ (PKCδ), and its downregulation reduced S1P-induced the phosphorylation of IKK and p65. In addition, silencing of Bcl10 also inhibited S1P-induced IKK phosphorylation. Surprisingly, S1P reduced Akt activation in melanoma cells that express FLNA, whereas in the absence of FLNA, high phosphorylation levels of Akt were maintained, enabling S1P-mediated NF-κB signaling. In accord, inhibition of Akt suppressed S1P-mediated IKK and p65 phosphorylation and degradation of IκBα. Hence, these results support a negative role of FLNA in S1P-mediated NF-κB activation in melanoma cells through modulation of Akt.


Assuntos
Filaminas/imunologia , Lisofosfolipídeos/imunologia , Melanoma/imunologia , NF-kappa B/imunologia , Proteínas Proto-Oncogênicas c-akt/imunologia , Transdução de Sinais , Esfingosina/análogos & derivados , Linhagem Celular Tumoral , Humanos , Proteína Quinase C-delta/imunologia , Receptores de Lisoesfingolipídeo/imunologia , Esfingosina/imunologia , Receptores de Esfingosina-1-Fosfato
8.
Immunology ; 144(4): 611-20, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25322815

RESUMO

Monocytes, key components of the immune system, are a heterogeneous population comprised of classical monocytes (CD16(-) ) and non-classical monocytes (CD16(+) ). Monocytes are short lived and undergo spontaneous apoptosis, unless stimulated. Dysregulation of monocyte numbers contribute to the pathophysiology of inflammatory diseases, yet the contribution of each subset remains poorly characterized. Protein kinase C (PKC) family members are central to monocyte biology; however, their role in regulating lifespan and immune function of CD16(-) and CD16(+) monocytes has not been studied. Here, we evaluated the contribution of PKCδ and PKCε in the lifespan and immune response of both monocyte subsets. We showed that CD16(+) monocytes are more susceptible to spontaneous apoptosis because of the increased caspase-3, -8 and -9 activities accompanied by higher kinase activity of PKCδ. Silencing of PKCδ reduced apoptosis in both CD16(+) and CD16(-) monocytes. CD16(+) monocytes express significantly higher levels of PKCε and produce more tumour necrosis factor-α in CD16(+) compared with CD16(-) monocytes. Silencing of PKCε affected the survival and tumour necrosis factor-α production. These findings demonstrate a complex network with similar topography, yet unique regulatory characteristics controlling lifespan and immune response in each monocyte subset, helping define subset-specific coordination programmes controlling monocyte function.


Assuntos
Monócitos/enzimologia , Monócitos/imunologia , Proteína Quinase C-delta/imunologia , Proteína Quinase C-delta/metabolismo , Proteína Quinase C-épsilon/imunologia , Proteína Quinase C-épsilon/metabolismo , Apoptose , Caspase 3/metabolismo , Caspase 8/metabolismo , Caspase 9/metabolismo , Sobrevivência Celular , Células Cultivadas , Proteínas Ligadas por GPI/deficiência , Proteínas Ligadas por GPI/imunologia , Humanos , Monócitos/classificação , Monócitos/patologia , Proteína Quinase C-delta/genética , Proteína Quinase C-épsilon/genética , Interferência de RNA , Receptores de IgG/deficiência , Receptores de IgG/imunologia , Transdução de Sinais , Fatores de Tempo , Transfecção , Fator de Necrose Tumoral alfa/sangue
9.
PLoS One ; 9(10): e109304, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25275584

RESUMO

In vitro expanded bone marrow stromal cells contain at least two populations of fibroblasts, a CD146/MCAM positive population, previously reported to be critical for establishing the stem cell niche and a CD146-negative population that expresses CUB domain-containing protein 1 (CDCP1)/CD318. Immunohistochemistry of marrow biopsies shows that clusters of CDCP1+ cells are present in discrete areas distinct from areas of fibroblasts expressing CD146. Using a stromal cell line, HS5, which approximates primary CDCP1+ stromal cells, we show that binding of an activating antibody against CDCP1 results in tyrosine-phosphorylation of CDCP1, paralleled by phosphorylation of Src Family Kinases (SFKs) Protein Kinase C delta (PKC-δ). When CDCP1 expression is knocked-down by siRNA, the expression and secretion of myelopoietic cytokines is increased. These data suggest CDCP1 expression can be used to identify a subset of marrow fibroblasts functionally distinct from CD146+ fibroblasts. Furthermore the CDCP1 protein may contribute to the defining function of these cells by regulating cytokine expression.


Assuntos
Antígenos CD/análise , Células da Medula Óssea/imunologia , Antígeno CD146/análise , Moléculas de Adesão Celular/análise , Fibroblastos/imunologia , Proteínas de Neoplasias/análise , Adulto , Antígenos CD/genética , Antígenos CD/imunologia , Antígenos de Neoplasias , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Antígeno CD146/imunologia , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/imunologia , Células Cultivadas , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Imunofenotipagem , Masculino , Pessoa de Meia-Idade , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/imunologia , Proteína Quinase C-delta/análise , Proteína Quinase C-delta/imunologia , Interferência de RNA , RNA Interferente Pequeno/genética , Células Estromais/citologia , Células Estromais/imunologia , Células Estromais/metabolismo , Quinases da Família src/análise , Quinases da Família src/imunologia
10.
Inflamm Res ; 63(7): 581-9, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24682410

RESUMO

OBJECTIVE AND DESIGN: Activations of the complement C5a (C5a) and the urokinase-type plasminogen activator (uPA) are commonly seen together during sepsis. However, the mechanism linking these two important pathways remains elusive. MATERIAL, METHODS AND TREATMENT: We used the C57BL/6 J mice model of sepsis induced by cecal ligation puncture (CLP) procedure, injected anti-C5aR or rottlerin through the tail vein to neutralize C5aR or PKC-δ, and then isolated peritoneal macrophages. Total RNA was isolated from the cells and analyzed by quantitative PCR. RESULTS: Our study revealed that neutralizing C5aR markedly inhibited sepsis-induced uPA receptor (uPAR) expression and its downstream signaling in macrophage. Similarly, neutralizing uPAR suppressed sepsis activation of C5a signaling. Importantly, inhibition of PKC-δ largely blocked sepsis-induced expression of C5aR and uPAR. CONCLUSIONS: Our study demonstrates a crosstalk between the complement C5a signaling and the fibrinolytic uPA pathways, which may depend on each other to maintain their expression and signaling, and reveals a central role of PKC-δ in mediating sepsis-induced activation of these pathways.


Assuntos
Complemento C5a/imunologia , Macrófagos Peritoneais/imunologia , Proteína Quinase C-delta/imunologia , Sepse/imunologia , Ativador de Plasminogênio Tipo Uroquinase/imunologia , Animais , Células Cultivadas , Complemento C5a/genética , Feminino , Masculino , Camundongos Endogâmicos C57BL , Transdução de Sinais , Ativador de Plasminogênio Tipo Uroquinase/genética
11.
Mol Cell Biol ; 34(8): 1474-85, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24515435

RESUMO

Protein kinase Cδ (PKCδ) deficiency causes autoimmune pathology in humans and mice and is crucial for the maintenance of B cell homeostasis. However, the mechanisms underlying autoimmune disease in PKCδ deficiency remain poorly defined. Here, we address the antigen-dependent and -independent roles of PKCδ in B cell development, repertoire selection, and antigen responsiveness. We demonstrate that PKCδ is rapidly phosphorylated downstream of both the B cell receptor (BCR) and the B cell-activating factor (BAFF) receptor. We found that PKCδ is essential for antigen-dependent negative selection of splenic transitional B cells and is required for activation of the proapoptotic Ca(2+)-Erk pathway that is selectively activated during B cell-negative selection. Unexpectedly, we also identified a previously unrecognized role for PKCδ as a proximal negative regulator of BCR signaling that substantially impacts survival and proliferation of mature follicular B cells. As a consequence of these distinct roles, PKCδ deficiency leads to the survival and development of a B cell repertoire that is not only aberrantly autoreactive but also hyperresponsive to antigen stimulation.


Assuntos
Linfócitos B/imunologia , Tolerância Imunológica/imunologia , Células Precursoras de Linfócitos B/citologia , Proteína Quinase C-delta/metabolismo , Receptores de Antígenos de Linfócitos B/metabolismo , Transdução de Sinais , Animais , Fator Ativador de Células B/imunologia , Fator Ativador de Células B/metabolismo , Linfócitos B/citologia , Linfócitos B/metabolismo , Diferenciação Celular/imunologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação/fisiologia , Células Precursoras de Linfócitos B/imunologia , Proteína Quinase C-delta/deficiência , Proteína Quinase C-delta/imunologia , Receptores de Antígenos de Linfócitos B/imunologia , Transdução de Sinais/imunologia
12.
Immunol Res ; 58(1): 61-9, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24385089

RESUMO

Looking to the sustained psoriatic arthritis (PsA) joint as a model of local human inflammation, this study was designed to assess the T lymphocyte signal transduction pathways potentially involved in this chronic immune-mediated inflammatory process, as characterized by direct ex vivo analysis of T helper (Th)-17 T effector (Teff) cell phenotypes in synovial fluid (SF) and peripheral blood (PB) of clinically active PsA patients. The reverse-phase protein arrays (RPPA) technique was employed to identify STAT3, STAT1, JAK1, JAK2, PKCδ and ERK1/2 phosphoprotein levels on total T cell lysates in SF samples of PsA patients. Frequencies of T CD4(+)IL-17A-F(+) and T CD4(+)IL-23R(+) Th17 cells were quantified in SF and matched PB of PsA patients by flow cytometry and compared with PB of healthy controls (HC). Increased levels of JAK1, STAT3, STAT1 and PKCδ phosphoproteins were found in SF T cells of PsA patients, compared with PB of HC. The expansion of T CD4(+)IL-17A-F(+) cells, as well as of T CD4(+) cells expressing IL-23Rp19 (T CD4(+) IL-23R(+)), considered as the pathogenic phenotype of effector Th17 cells, was found to be confined to the joints of PsA patients, as the frequencies of both populations were significantly higher in SF than in matched PB, or in PB of HC. In conclusion, T lymphocyte signal transduction pathway mapping revealed an enhanced activation of JAK1/STAT3/STAT1 and PKCδ phosphoproteins that may drive the local inflammatory process, characterized by the in vivo expansion of T CD4(+)IL-17A-F(+) and T CD4(+)IL-23R(+) Th17 Teff cells in SF of clinically active joints of PsA patients.


Assuntos
Artrite Psoriásica/imunologia , Líquido Sinovial/imunologia , Células Th17/imunologia , Adulto , Artrite Psoriásica/enzimologia , MAP Quinases Reguladas por Sinal Extracelular/imunologia , Feminino , Citometria de Fluxo , Humanos , Janus Quinases/imunologia , Leucócitos Mononucleares/imunologia , Sistema de Sinalização das MAP Quinases , Masculino , Pessoa de Meia-Idade , Análise Serial de Proteínas , Proteína Quinase C-delta/imunologia , Fatores de Transcrição STAT/imunologia , Estatísticas não Paramétricas , Líquido Sinovial/citologia , Líquido Sinovial/enzimologia , Células Th17/citologia , Células Th17/enzimologia
13.
Curr Opin Pediatr ; 25(6): 722-9, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24240292

RESUMO

PURPOSE OF REVIEW: Several autoimmune lymphoproliferative syndromes have been described lately. We review here the main clinical and laboratory findings of these new disorders. RECENT FINDINGS: The prototypical autoimmune lymphoproliferative syndrome (ALPS) has had its diagnostic criteria modified, somatic mutations in RAS genes were found to cause an ALPS-like syndrome in humans, and mutations in a gene encoding a protein kinase C (PRKCD) were discovered to cause a syndrome of lymphoproliferation, autoimmunity and natural killer cell defect. SUMMARY: The recent discoveries shed light on the molecular pathways governing lymphocyte death, proliferation and immune tolerance in humans.


Assuntos
Apoptose/imunologia , Síndrome Linfoproliferativa Autoimune/diagnóstico , Genes ras , Tolerância Imunológica/imunologia , Inflamação/imunologia , Apoptose/genética , Síndrome Linfoproliferativa Autoimune/genética , Síndrome Linfoproliferativa Autoimune/imunologia , Autoimunidade/genética , Proliferação de Células , Proteína de Domínio de Morte Associada a Fas/imunologia , Feminino , Genes ras/genética , Genes ras/imunologia , Humanos , Tolerância Imunológica/genética , Inflamação/genética , Inflamação/patologia , Masculino , Mutação/imunologia , Proteína Quinase C-delta/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia
14.
PLoS One ; 8(8): e70815, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23951014

RESUMO

Bone remodeling is intrinsically regulated by cell signaling molecules. The Protein Kinase C (PKC) family of serine/threonine kinases is involved in multiple signaling pathways including cell proliferation, differentiation, apoptosis and osteoclast biology. However, the precise involvement of individual PKC isoforms in the regulation of osteoclast formation and bone homeostasis remains unclear. Here, we identify PKC-δ as the major PKC isoform expressed among all PKCs in osteoclasts; including classical PKCs (-α, -ß and -γ), novel PKCs (-δ, -ε, -η and -θ) and atypical PKCs (-ι/λ and -ζ). Interestingly, pharmacological inhibition and genetic ablation of PKC-δ impairs osteoclastic bone resorption in vitro. Moreover, disruption of PKC-δ activity protects against LPS-induced osteolysis in mice, with osteoclasts accumulating on the bone surface failing to resorb bone. Treatment with the PKC-δ inhibitor Rottlerin, blocks LPS-induced bone resorption in mice. Consistently, PKC-δ deficient mice exhibit increased trabeculae bone containing residual cartilage matrix, indicative of an osteoclast-rich osteopetrosis phenotype. Cultured ex vivo osteoclasts derived from PKC-δ null mice exhibit decreased CTX-1 levels and MARKS phosphorylation, with enhanced formation rates. This is accompanied by elevated gene expression levels of cathepsin K and PKC -α, -γ and -ε, as well as altered signaling of pERK and pcSrc416/527 upon RANKL-induction, possibly to compensate for the defects in bone resorption. Collectively, our data indicate that PKC-δ is an intrinsic regulator of osteoclast formation and bone resorption and thus is a potential therapeutic target for pathological osteolysis.


Assuntos
Lipopolissacarídeos/imunologia , Osteoclastos/patologia , Osteólise/genética , Osteólise/imunologia , Proteína Quinase C-delta/antagonistas & inibidores , Proteína Quinase C-delta/genética , Acetofenonas/uso terapêutico , Animais , Benzopiranos/uso terapêutico , Reabsorção Óssea/tratamento farmacológico , Reabsorção Óssea/genética , Reabsorção Óssea/imunologia , Reabsorção Óssea/patologia , Catepsina K/genética , Células Cultivadas , Inibidores Enzimáticos/uso terapêutico , Feminino , Deleção de Genes , Regulação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoclastos/efeitos dos fármacos , Osteoclastos/imunologia , Osteoclastos/metabolismo , Osteólise/tratamento farmacológico , Osteólise/patologia , Fosforilação , Proteína Quinase C-delta/imunologia , Transdução de Sinais
15.
Arthritis Rheum ; 65(8): 2161-71, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23666743

RESUMO

OBJECTIVE: Systemic lupus erythematosus (SLE) is a prototype autoimmune disease that is assumed to occur via a complex interplay of environmental and genetic factors. Rare causes of monogenic SLE have been described, providing unique insights into fundamental mechanisms of immune tolerance. The aim of this study was to identify the cause of an autosomal-recessive form of SLE. METHODS: We studied 3 siblings with juvenile-onset SLE from 1 consanguineous kindred and used next-generation sequencing to identify mutations in the disease-associated gene. We performed extensive biochemical, immunologic, and functional assays to assess the impact of the identified mutations on B cell biology. RESULTS: We identified a homozygous missense mutation in PRKCD, encoding protein kinase δ (PKCδ), in all 3 affected siblings. Mutation of PRKCD resulted in reduced expression and activity of the encoded protein PKCδ (involved in the deletion of autoreactive B cells), leading to resistance to B cell receptor- and calcium-dependent apoptosis and increased B cell proliferation. Thus, as for mice deficient in PKCδ, which exhibit an SLE phenotype and B cell expansion, we observed an increased number of immature B cells in the affected family members and a developmental shift toward naive B cells with an immature phenotype. CONCLUSION: Our findings indicate that PKCδ is crucial in regulating B cell tolerance and preventing self-reactivity in humans, and that PKCδ deficiency represents a novel genetic defect of apoptosis leading to SLE.


Assuntos
Apoptose , Linfócitos B/patologia , Lúpus Eritematoso Sistêmico/enzimologia , Lúpus Eritematoso Sistêmico/genética , Mutação de Sentido Incorreto , Proteína Quinase C-delta/deficiência , Proteína Quinase C-delta/genética , Adolescente , Adulto , Linfócitos B/imunologia , Linfócitos B/metabolismo , Proliferação de Células , Criança , Feminino , Variação Genética , Homozigoto , Humanos , Hiperplasia , Tolerância Imunológica , Lúpus Eritematoso Sistêmico/patologia , Masculino , Polimorfismo de Nucleotídeo Único , Proteína Quinase C-delta/imunologia , Adulto Jovem
16.
J Autoimmun ; 41: 175-81, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23478041

RESUMO

Sjögren's syndrome (SS) is an autoimmune exocrinopathy characterized by an epithelium injury with dense lymphocytic infiltrates, mainly composed of activated T and B cells. Present at the interface of genetic and environmental risk factors, DNA methylation is suspected to play a key role in SS. To clarify this point, global DNA methylation was tested within salivary gland epithelial cells (SGEC), peripheral T cells and B cells from SS patients. Global DNA methylation was reduced in SGEC from SS patients, while no difference was observed in T and B cells. SGEC demethylation in SS patients was associated with a 7-fold decrease in DNA methyl transferase (DNMT) 1 and a 2-fold increase in Gadd45-alpha expression. The other DNA methylation/demethylation partners, tested by real time PCR (DNMT3a/b, PCNA, UHRF1, MBD2, and MBD4), were not different. Interestingly, SGEC demethylation may be attributed in part to the infiltrating B cells as suspected in patients treated with anti-CD20 antibodies to deplete B cells. Such hypothesis was confirmed using co-culture experiments with human salivary gland cells and B cells. Furthermore, B cell-mediated DNA demethylation could be ascribed to an alteration of the PKC delta/ERK/DNMT1 pathway. As a consequence, part of the SGEC dysfunction in SS may be linked to epigenetic modifications, thus opening new therapeutic perspectives in SS.


Assuntos
Linfócitos B/imunologia , Epigênese Genética/imunologia , Glândulas Salivares/imunologia , Síndrome de Sjogren/imunologia , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Antígenos CD20/imunologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/imunologia , Linhagem Celular , Células Cultivadas , Técnicas de Cocultura , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/imunologia , Metilação de DNA/imunologia , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/imunologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Expressão Gênica/imunologia , Humanos , Molécula 1 de Adesão Intercelular/imunologia , Molécula 1 de Adesão Intercelular/metabolismo , Microscopia de Fluorescência , Proteínas Nucleares/genética , Proteínas Nucleares/imunologia , Proteína Quinase C-delta/imunologia , Proteína Quinase C-delta/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Glândulas Salivares/metabolismo , Glândulas Salivares/patologia , Transdução de Sinais/imunologia , Síndrome de Sjogren/genética , Síndrome de Sjogren/patologia
17.
Clin Immunol ; 147(1): 1-8, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23454274

RESUMO

Post-translational modification of proteins by deamidation or transamidation by tissue transglutaminase (tTG) has been suggested as a possible mechanism for the development of autoimmunity. Sequence analysis of protein kinase C delta (PKCδ) identified an amino acid motif that suggested the possibility that PKCδ was a glutamine substrate of tTG and MALDI-TOF analysis of synthesised peptides from PKCδ proved that this was the case. Polymerisation experiments using recombinant tTG and biotinylated hexapeptide substrate incorporation assays demonstrated that PKCδ is a substrate for tTG-mediated transamidation. Elevated levels of anti-PKCδ antibodies were detected in sera from patients with coeliac disease (p<0.0001) but not from patients with other autoimmune disorders. These data suggest that a subset of patients with coeliac disease produce autoantibodies against PKCδ and that this response may stem from a tTG-PKCδ substrate interaction.


Assuntos
Autoantígenos/imunologia , Doença Celíaca/imunologia , Proteínas de Ligação ao GTP/imunologia , Proteína Quinase C-delta/imunologia , Transglutaminases/imunologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Autoanticorpos/sangue , Autoanticorpos/imunologia , Autoantígenos/sangue , Autoantígenos/metabolismo , Western Blotting , Doença Celíaca/metabolismo , Criança , Ensaio de Imunoadsorção Enzimática , Feminino , Proteínas de Ligação ao GTP/metabolismo , Glutamina/genética , Glutamina/imunologia , Glutamina/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Peptídeos/química , Peptídeos/imunologia , Peptídeos/metabolismo , Ligação Proteica , Proteína 2 Glutamina gama-Glutamiltransferase , Proteína Quinase C-delta/genética , Proteína Quinase C-delta/metabolismo , Processamento de Proteína Pós-Traducional/imunologia , Proteínas Recombinantes/química , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Especificidade por Substrato , Transglutaminases/metabolismo , Adulto Jovem
18.
Blood ; 121(16): 3117-25, 2013 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-23430113

RESUMO

Defective lymphocyte apoptosis results in chronic lymphadenopathy and/or splenomegaly associated with autoimmune phenomena. The prototype for human apoptosis disorders is the autoimmune lymphoproliferative syndrome (ALPS), which is caused by mutations in the FAS apoptotic pathway. Recently, patients with an ALPS-like disease called RAS-associated autoimmune leukoproliferative disorder, in which somatic mutations in NRAS or KRAS are found, also were described. Despite this progress, many patients with ALPS-like disease remain undefined genetically. We identified a homozygous, loss-of-function mutation in PRKCD (PKCδ) in a patient who presented with chronic lymphadenopathy, splenomegaly, autoantibodies, elevated immunoglobulins and natural killer dysfunction associated with chronic, low-grade Epstein-Barr virus infection. This mutation markedly decreased protein expression and resulted in ex vivo B-cell hyperproliferation, a phenotype similar to that of the PKCδ knockout mouse. Lymph nodes showed intense follicular hyperplasia, also mirroring the mouse model. Immunophenotyping of circulating lymphocytes demonstrated expansion of CD5+CD20+ B cells. Knockdown of PKCδ in normal mononuclear cells recapitulated the B-cell hyperproliferative phenotype in vitro. Reconstitution of PKCδ in patient-derived EBV-transformed B-cell lines partially restored phorbol-12-myristate-13-acetate-induced cell death. In summary, homozygous PRKCD mutation results in B-cell hyperproliferation and defective apoptosis with consequent lymphocyte accumulation and autoantibody production in humans, and disrupts natural killer cell function.


Assuntos
Síndrome Linfoproliferativa Autoimune/genética , Linfócitos B/patologia , Mutação , Proteína Quinase C-delta/genética , Animais , Apoptose , Síndrome Linfoproliferativa Autoimune/complicações , Síndrome Linfoproliferativa Autoimune/imunologia , Síndrome Linfoproliferativa Autoimune/patologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Linhagem Celular , Proliferação de Células , Criança , Citocinas/imunologia , Infecções por Vírus Epstein-Barr/complicações , Expressão Gênica , Técnicas de Silenciamento de Genes , Herpesvirus Humano 4/isolamento & purificação , Homozigoto , Humanos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/patologia , Linfonodos/imunologia , Linfonodos/patologia , Doenças Linfáticas/complicações , Masculino , Camundongos , Proteína Quinase C-delta/imunologia , Esplenomegalia/complicações
19.
Blood ; 121(16): 3112-6, 2013 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-23319571

RESUMO

Primary B-cell disorders comprise a heterogeneous group of inherited immunodeficiencies, often associated with autoimmunity causing significant morbidity. The underlying genetic etiology remains elusive in the majority of patients. In this study, we investigated a patient from a consanguineous family suffering from recurrent infections and severe lupuslike autoimmunity. Immunophenotyping revealed progressive decrease of CD19(+) B cells, a defective class switch indicated by low numbers of IgM- and IgG-memory B cells, as well as increased numbers of CD21(low) B cells. Combined homozygosity mapping and exome sequencing identified a biallelic splice-site mutation in protein C kinase δ (PRKCD), causing the absence of the corresponding protein product. Consequently, phosphorylation of myristoylated alanine-rich C kinase substrate was decreased, and mRNA levels of nuclear factor interleukin (IL)-6 and IL-6 were increased. Our study uncovers human PRKCD deficiency as a novel cause of common variable immunodeficiency-like B-cell deficiency with severe autoimmunity.


Assuntos
Autoimunidade , Linfócitos B/patologia , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/patologia , Mutação , Proteína Quinase C-delta/genética , Adulto , Antígenos CD19/imunologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Criança , Feminino , Humanos , Imunoglobulina G/imunologia , Imunoglobulina M/imunologia , Síndromes de Imunodeficiência/imunologia , Síndromes de Imunodeficiência/terapia , Imunofenotipagem , Masculino , Linhagem , Polimorfismo de Nucleotídeo Único , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Proteína Quinase C-delta/imunologia
20.
Cell Adh Migr ; 6(5): 413-8, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23076052

RESUMO

Enteroaggregative Escherichia coli (EAEC) is an important cause of endemic and epidemic diarrheal disease worldwide. Although not classically considered an inflammatory pathogen in the style of Shigella and Salmonella species, clinical data from patients suggests that inflammatory responses may play an important role during EAEC disease. However, the specific role of inflammation during EAEC pathogenesis has not been investigated in detail. To better understand how EAEC may induce inflammation, we have focused our attention on the intimate interactions between EAEC and the host epithelium and the subsequent induction of host cell signaling events leading to innate immune responses. Here, we discuss our recent findings on the signaling pathway by which EAEC promotes transepithelial migration of polymorphonuclear leukocytes (PMNs), the role of aggregative adherence fimbriae in triggering this event and the implementation of human intestinal xenografts in immunodeficient mice for studying EAEC pathogenesis in vivo. Our findings suggest that EAEC shares conserved mechanisms of inducing PMN recruitment with other intestinal pathogens, providing new insight into the potential pathological consequences of EAEC-induced inflammation.


Assuntos
Infecções por Escherichia coli/imunologia , Escherichia coli/patogenicidade , Interações Hospedeiro-Patógeno , Inflamação/microbiologia , Adesinas de Escherichia coli/imunologia , Adesinas de Escherichia coli/metabolismo , Araquidonato 12-Lipoxigenase/metabolismo , Ácido Araquidônico/metabolismo , Aderência Bacteriana , Membrana Celular/metabolismo , Movimento Celular , Escherichia coli/imunologia , Escherichia coli/metabolismo , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/microbiologia , Fímbrias Bacterianas/imunologia , Humanos , Imunidade Inata , Inflamação/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Proteína 2 Associada à Farmacorresistência Múltipla , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Neutrófilos/imunologia , Neutrófilos/metabolismo , Proteína Quinase C-delta/imunologia , Proteína Quinase C-delta/metabolismo , Transdução de Sinais , Migração Transendotelial e Transepitelial
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...