Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 235
Filtrar
1.
Sci Rep ; 11(1): 19798, 2021 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-34611276

RESUMO

Autosomal Dominant Polycystic Kidney Disease (ADPKD) is the most common inherited renal disorder, characterized by renal cyst development leading to end-stage renal disease. Although the appropriate choice of suitable reference is critical for quantitative RNA analysis, no comparison of frequently used "housekeeping" genes is available. Here, we determined the validity of 7 candidate housekeeping genes (Actb, Actg1, B2m, Gapdh, Hprt, Pgam1 and Ppia) in kidney tissues from mouse models orthologous to ADPKD, including a cystic mice (CY) 10-12 weeks old (Pkd1flox/flox:Nestincre/Pkd1flox/-:Nestincre, n = 10) and non-cystic (NC) controls (Pkd1flox/flox/Pkd1flox/-, n = 10), Pkd1-haploinsufficient (HT) mice (Pkd1+/-, n = 6) and wild-type (WT) controls (Pkd1+/+, n = 6) and a severely cystic (SC) mice 15 days old (Pkd1V/V, n = 7) and their controls (CO, n = 5). Gene expression data were analyzed using six distinct statistical softwares. The estimation of the ideal number of genes suggested the use of Ppia alone as sufficient, although not ideal, to analyze groups altogether. Actb, Hprt and Ppia expression profiles were correlated in all samples. Ppia was identified as the most stable housekeeping gene, while Gapdh was the least stable for all kidney samples. Stat3 expression level was consistent with upregulation in SC compared to CO when normalized by Ppia expression. In conclusion, present findings identified Ppia as the best housekeeping gene for CY + NC and SC + CO groups, while Hprt was the best for the HT + WT group.


Assuntos
Genes Essenciais , Rim/metabolismo , Peptidilprolil Isomerase/genética , Proteína Quinase C/deficiência , Animais , Biomarcadores , Modelos Animais de Doenças , Expressão Gênica , Camundongos , Camundongos Knockout , RNA Mensageiro , Reação em Cadeia da Polimerase em Tempo Real , Fator de Transcrição STAT3/genética
2.
J Neurosci ; 41(35): 7329-7339, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34290081

RESUMO

Post-tetanic potentiation (PTP) is a form of short-term plasticity that lasts for tens of seconds following a burst of presynaptic activity. It has been proposed that PTP arises from protein kinase C (PKC) phosphorylation of Munc18-1, an SM (Sec1/Munc-18 like) family protein that is essential for release. To test this model, we made a knock-in mouse in which all Munc18-1 PKC phosphorylation sites were eliminated through serine-to-alanine point mutations (Munc18-1SA mice), and we studied mice of either sex. The expression of Munc18-1 was not altered in Munc18-1SA mice, and there were no obvious behavioral phenotypes. At the hippocampal CA3-to-CA1 synapse and the granule cell parallel fiber (PF)-to-Purkinje cell (PC) synapse, basal transmission was largely normal except for small decreases in paired-pulse facilitation that are consistent with a slight elevation in release probability. Phorbol esters that mimic the activation of PKC by diacylglycerol still increased synaptic transmission in Munc18-1SA mice. In Munc18-1SA mice, 70% of PTP remained at CA3-to-CA1 synapses, and the amplitude of PTP was not reduced at PF-to-PC synapses. These findings indicate that at both CA3-to-CA1 and PF-to-PC synapses, phorbol esters and PTP enhance synaptic transmission primarily by mechanisms that are independent of PKC phosphorylation of Munc18-1.SIGNIFICANCE STATEMENT A leading mechanism for a prevalent form of short-term plasticity, post-tetanic potentiation (PTP), involves protein kinase C (PKC) phosphorylation of Munc18-1. This study tests this mechanism by creating a knock-in mouse in which Munc18-1 is replaced by a mutated form of Munc18-1 that cannot be phosphorylated. The main finding is that most PTP at hippocampal CA3-to-CA1 synapses or at cerebellar granule cell-to-Purkinje cell synapses does not rely on PKC phosphorylation of Munc18-1. Thus, mechanisms independent of PKC phosphorylation of Munc18-1 are important mediators of PTP.


Assuntos
Proteínas Munc18/metabolismo , Plasticidade Neuronal/fisiologia , Proteína Quinase C/metabolismo , Processamento de Proteína Pós-Traducional , Substituição de Aminoácidos , Animais , Feminino , Técnicas de Introdução de Genes , Hipocampo/fisiologia , Masculino , Camundongos , Camundongos Knockout , Potenciais Pós-Sinápticos em Miniatura/efeitos dos fármacos , Potenciais Pós-Sinápticos em Miniatura/fisiologia , Proteínas Munc18/deficiência , Mutação de Sentido Incorreto , Ésteres de Forbol/farmacologia , Fosforilação , Mutação Puntual , Proteína Quinase C/deficiência , Células de Purkinje/fisiologia , Proteínas Recombinantes/metabolismo , Transmissão Sináptica/efeitos dos fármacos
3.
Dev Cell ; 56(1): 95-110.e10, 2021 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-33207226

RESUMO

Cancer-associated fibroblasts (CAFs) promote tumor malignancy, but the precise transcriptional mechanisms regulating the acquisition of the CAF phenotype are not well understood. We show that the upregulation of SOX2 is central to this process, which is repressed by protein kinase Cζ (PKCζ). PKCζ deficiency activates the reprogramming of colonic fibroblasts to generate a predominant SOX2-dependent CAF population expressing the WNT regulator Sfrp2 as its top biomarker. SOX2 directly binds the Sfrp1/2 promoters, and the inactivation of Sox2 or Sfrp1/2 in CAFs impaired the induction of migration and invasion of colon cancer cells, as well as their tumorigenicity in vivo. Importantly, recurrence-free and overall survival of colorectal cancer (CRC) patients negatively correlates with stromal PKCζ levels. Also, SOX2 expression in the stroma is associated with CRC T invasion and worse prognosis of recurrence-free survival. Therefore, the PKCζ-SOX2 axis emerges as a critical step in the control of CAF pro-tumorigenic potential.


Assuntos
Fibroblastos Associados a Câncer/metabolismo , Carcinogênese/metabolismo , Neoplasias Colorretais/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Membrana/metabolismo , Proteína Quinase C/deficiência , Fatores de Transcrição SOXB1/metabolismo , Animais , Fibroblastos Associados a Câncer/patologia , Carcinogênese/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Progressão da Doença , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Invasividade Neoplásica/genética , Organoides/metabolismo , Organoides/patologia , Ligação Proteica , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , RNA-Seq , Recidiva , Fatores de Transcrição SOXB1/genética , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Análise de Célula Única , Regulação para Cima , beta Catenina/genética , beta Catenina/metabolismo
4.
Biochim Biophys Acta Mol Basis Dis ; 1867(1): 165987, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33039594

RESUMO

BACKGROUND: Protein kinase D (PKD) family, which includes PKD/PKD1, PKD2, and PKD3, has been increasingly implicated in the regulation of multiple cellular functions and human diseases. We recently reported that pharmacologic inhibition of PKD ameliorated the pathologic responses and severity of pancreatitis. However, to further investigate the importance of PKD family members in pancreatitis, it is necessary to explore the effects of pancreas-specific genetic inhibition of PKD isoform on pathology of pancreatitis. METHODS: We generated a mouse model (referred as PKD3Δpanc mice) with pancreas-specific deletion of PKD3, the predominant PKD isoform in mouse pancreatic acinar cells, by crossing Pkd3flox/flox mice with Pdx1-Cre transgenic mice which express Cre recombinase under the control of the mouse Pdx1 promoter. Pancreas-specific deletion of the PKD3 gene and PKD3 protein was confirmed by PCR and Western blot analysis. Experimental pancreatitis was induced in PKD3Δpanc and Pkd3flox/flox (control mice) littermates by intraperitoneal injections of cerulein or L-arginine. RESULTS: Compared to the control mice, PKD3Δpanc mice displayed significant attenuation in inflammation, necrosis, and severity of pancreatitis in both experimental models. PKD3Δpanc mice had markedly decreased NF-κB and trypsinogen activation, pancreatic mRNA expression of multiple inflammatory molecules, and the receptor-interacting protein kinase 1 (RIP1) activation in pancreatitis. PKD3Δpanc mice also had less pancreatic ATP depletion, increased pro-survival Bcl-2 family protein expression, and autophagy promotion. CONCLUSION: With PKD3Δpanc mouse model, we further demonstrated that PKD plays a critical role in pathobiological process of pancreatitis and PKD constitutes a novel therapeutic target to treat this disorder.


Assuntos
Deleção de Genes , Pâncreas/metabolismo , Pancreatite/metabolismo , Proteína Quinase C/deficiência , Animais , Modelos Animais de Doenças , Inflamação/genética , Inflamação/metabolismo , Camundongos , Camundongos Knockout , Necrose , Especificidade de Órgãos , Pâncreas/patologia , Pancreatite/genética , Pancreatite/patologia , Proteína Quinase C/metabolismo , Índice de Gravidade de Doença
5.
Mol Neurobiol ; 58(2): 617-630, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32996086

RESUMO

The brain encodes, stores, and retrieves relevant information in the form of memories that are classified as short-term (STM) and long-term memories (LTM) depending on the interval between acquisition and retrieval. It is classically accepted that STM undergo a consolidation process to form LTM, but the molecular determinants involved are not well understood. Among the molecular components relevant for memory formation, we focused our attention on the protein kinase C (PKC) family of enzymes since they control key aspects of the synaptic plasticity and memory. Within the different PKC isoforms, PKC-gamma has been specifically associated with learning and memory since mice lacking this isoform (PKC-gamma KO mice) showed mild cognitive impairment and deficits in hippocampal synaptic plasticity. We now reveal that PKC-gamma KO mice present a severe impairment in hippocampal-dependent STM using different memory tests including the novel object-recognition and novel place-recognition, context fear conditioning and trace fear conditioning. In contrast, no differences between genotypes were observed in an amygdala-dependent test, the delay fear conditioning. Strikingly, all LTM tasks that could be assessed 24 h after acquisition were not perturbed in the KO mice. The analysis of c-Fos expression in several brain areas after trace fear conditioning acquisition showed a blunted response in the dentate gyrus of PKC-gamma KO mice compared with WT mice, but such differences between genotypes were absent when the amygdala or the prefrontal cortex were examined. In the hippocampus, PKC-gamma was found to translocate to the membrane after auditory trace, but not after delay fear conditioning. Together, these results indicate that PKC-gamma dysfunction affects specifically hippocampal-dependent STM performance and disclose PKC-gamma as a molecular player differentially involved in STM and LTM processes.


Assuntos
Hipocampo/enzimologia , Memória de Longo Prazo , Memória de Curto Prazo , Proteína Quinase C/deficiência , Animais , Condicionamento Clássico , Giro Denteado/patologia , Medo , Isoenzimas/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Memória Espacial
6.
Proc Natl Acad Sci U S A ; 117(46): 29001-29012, 2020 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-33122431

RESUMO

Autosomal-dominant polycystic kidney disease (ADPKD) is the most common genetic renal disease, primarily caused by germline mutation of PKD1 or PKD2, leading to end-stage renal disease. The Hippo signaling pathway regulates organ growth and cell proliferation. Herein, we demonstrate the regulatory mechanism of cystogenesis in ADPKD by transcriptional coactivator with PDZ-binding motif (TAZ), a Hippo signaling effector. TAZ was highly expressed around the renal cyst-lining epithelial cells of Pkd1-deficient mice. Loss of Taz in Pkd1-deficient mice reduced cyst formation. In wild type, TAZ interacted with PKD1, which inactivated ß-catenin. In contrast, in PKD1-deficient cells, TAZ interacted with AXIN1, thus increasing ß-catenin activity. Interaction of TAZ with AXIN1 in PKD1-deficient cells resulted in nuclear accumulation of TAZ together with ß-catenin, which up-regulated c-MYC expression. Our findings suggest that the PKD1-TAZ-Wnt-ß-catenin-c-MYC signaling axis plays a critical role in cystogenesis and might be a potential therapeutic target against ADPKD.


Assuntos
Doenças Renais Policísticas/genética , Doenças Renais Policísticas/metabolismo , Rim Policístico Autossômico Dominante/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Transativadores/metabolismo , Via de Sinalização Wnt/fisiologia , beta Catenina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteína Axina , Proliferação de Células , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Humanos , Rim/metabolismo , Rim/patologia , Camundongos , Camundongos Knockout , Doenças Renais Policísticas/patologia , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/patologia , Proteína Quinase C/deficiência , Proteína Quinase C/genética , Canais de Cátion TRPP/genética , Transcriptoma
7.
Gastroenterology ; 159(3): 1019-1035.e22, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32446697

RESUMO

BACKGROUND & AIMS: Pancreatic tumor cells release small extracellular vesicles (sEVs, exosomes) that contain lipids and proteins, RNA, and DNA molecules that might promote formation of metastases. It is not clear what cargo these vesicles contain and how they are released. Protein kinase D1 (PRKD1) inhibits cell motility and is believed to be dysregulated in pancreatic ductal adenocarcinomas. We investigated whether it regulates production of sEVs in pancreatic cancer cells and their ability to form premetastatic niches for pancreatic cancer cells in mice. METHODS: We analyzed data from UALCAN and human pancreatic tissue microarrays to compare levels of PRKD1 between tumor and nontumor tissues. We studied mice with pancreas-specific disruption of Prkd1 (PRKD1KO mice), mice that express oncogenic KRAS (KC mice), and KC mice with disruption of Prkd1 (PRKD1KO-KC mice). Subcutaneous xenograft tumors were grown in NSG mice from Panc1 cells; some mice were then given injections of sEVs. Pancreata and lung tissues from mice were analyzed by histology, immunohistochemistry, and/or quantitative polymerase chain reaction; we performed nanoparticle tracking analysis of plasma sEVs. The Prkd1 gene was disrupted in Panc1 cells using CRISPR-Cas9 or knocked down with small hairpin RNAs, or PRKD1 activity was inhibited with the selective inhibitor CRT0066101. Pancreatic cancer cell lines were analyzed by gene-expression microarray, quantitative polymerase chain reaction, immunoblot, and immunofluorescence analyses. sEVs secreted by Panc1 cell lines were analyzed by flow cytometry, transmission electron microscopy, and mass spectrometry. RESULTS: Levels of PRKD1 were reduced in human pancreatic ductal adenocarcinoma tissues compared with nontumor tissues. PRKD1KO-KC mice developed more pancreatic intraepithelial neoplasia, at a faster rate, than KC mice, and had more lung metastases and significantly shorter average survival time. Serum from PRKD1KO-KC mice had increased levels of sEVs compared with KC mice. Pancreatic cancer cells with loss or inhibition of PRKD1 increased secretion of sEVs; loss of PRKD1 reduced phosphorylation of its substrate, cortactin, resulting in increased F-actin levels at the plasma membrane. sEVs from cells with loss or reduced expression of PRKD1 had altered content, and injection of these sEVs into mice increased metastasis of xenograft tumors to lung, compared with sEVs from pancreatic cells that expressed PRKD1. PRKD1-deficient pancreatic cancer cells showed increased loading of integrin α6ß4 into sEVs-a process that required CD82. CONCLUSIONS: Human pancreatic ductal adenocarcinoma has reduced levels of PRKD1 compared with nontumor pancreatic tissues. Loss of PRKD1 results in reduced phosphorylation of cortactin in pancreatic cancer cell lines, resulting in increased in F-actin at the plasma membrane and increased release of sEVs, with altered content. These sEVs promote metastasis of xenograft and pancreatic tumors to lung in mice.


Assuntos
Carcinoma Ductal Pancreático/secundário , Vesículas Extracelulares/metabolismo , Neoplasias Pulmonares/secundário , Neoplasias Pancreáticas/patologia , Proteína Quinase C/deficiência , Animais , Carcinogênese/patologia , Carcinoma Ductal Pancreático/sangue , Linhagem Celular Tumoral , Movimento Celular , Conjuntos de Dados como Assunto , Regulação para Baixo , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Pulmão/patologia , Neoplasias Pulmonares/sangue , Camundongos , Camundongos Knockout , Invasividade Neoplásica/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Pâncreas/patologia , Neoplasias Pancreáticas/sangue , Fosforilação , Cultura Primária de Células , Proteína Quinase C/genética , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Sci Rep ; 10(1): 3902, 2020 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-32127582

RESUMO

PRK1 and PRK2 are two closely related AGC-family serine/threonine protein kinases. Here we demonstrate novel roles for them at cilia and in cancer biology. In both instances serum withdrawal leads to increased activating PRK1 and PRK2 phosphorylation (pPRK1/pPRK2) and their depletion results in reduced spheroid growth. pPRK1/pPRK2 localise to the transition zone of cilia and their co-depletion results in reduced cilia size, impaired planer polarity and impaired cilia associated signalling. High PRK2 (but not PRK1) expression correlates with poor outcome in patients with basal-like/Triple Negative (TN) Breast Cancer (BC) where there is also higher expression relative to other BC tumour subtypes. In agreement, depletion of PRK1 and PRK2 in mouse TNBC cells, or CRISPR/Cas9 mediated deletion of PRK2 alone, significantly reduces cell proliferation and spheroid growth. Finally proteomic analysis to identify PRK2 binding partners in mouse TNBC cells revealed proteins that are important for both cilia and BC biology. Taken together these data demonstrate novel roles for PRK1 and PRK2 at cilia and in BC biology and in the case of PRK2 in particular, identifies it as a novel TNBC therapeutic target.


Assuntos
Cílios/enzimologia , Proteína Quinase C/metabolismo , Neoplasias de Mama Triplo Negativas/enzimologia , Neoplasias de Mama Triplo Negativas/patologia , Animais , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Proteína Quinase C/deficiência , Proteína Quinase C/genética , Transdução de Sinais , Neoplasias de Mama Triplo Negativas/genética
9.
J Mol Med (Berl) ; 98(5): 659-671, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32185407

RESUMO

Polycystic kidney disease (PKD) leads to continuous decline of renal function by growth of renal cysts. Enhanced proliferation and transepithelial chloride secretion through cystic fibrosis transmembrane conductance regulator (CFTR) and Ca2+-activated TMEM16A Cl- channels is thought to cause an increase in cyst volume. Recent work shows the pro-proliferative role of the Ca2+ activated Cl- channel TMEM16A (anoctamin 1), and demonstrates the essential contribution of TMEM16A to CFTR-dependent Cl- secretion. The present data demonstrate an increase in intracellular Ca2+ ([Ca2+]i) signals and Cl- secretion by TMEM16A, in renal collecting duct principle cells from dog (MDCK) and mouse (M1) as well as primary tubular epithelial cells from PKD1-/- knockout mice. M1 organoids proliferated, increased expression of TMEM16A, and secreted Cl- upon knockdown of endogenous polycystin 1 or 2 (PKD1,2), by retroviral transfection with shPKD1 and shPKD2, respectively. Knockdown of PKD1 or PKD2 increased basal intracellular Ca2+ levels and enhanced purinergic Ca2+ release from endoplasmic reticulum. In contrast, ryanodine receptors were found not to be expressed in mouse renal epithelial cells and caffeine had no effects on [Ca2+]i. Ca2+ signals, proliferation, and Cl- secretion were largely reduced by knockdown or blockade of TMEM16A. TMEM16A may be therefore important for enhanced Ca2+ release from IP3-sensitive Ca2+ stores in polycystic kidney disease. KEY MESSAGES: • ADPKD leads to continuous decline of renal function by growth of renal cysts. • Knockdown of PKD1 or PKD2 increases TMEM16A expression. • TMEM16A enhanced intracellular Ca2+ signals, Cl- secretion, and proliferation. • TMEM16A contributes to cyst growth in ADPKD.


Assuntos
Anoctamina-1/genética , Anoctamina-1/metabolismo , Sinalização do Cálcio , Cálcio/metabolismo , Suscetibilidade a Doenças , Doenças Renais Policísticas/genética , Doenças Renais Policísticas/metabolismo , Potenciais de Ação , Trifosfato de Adenosina/metabolismo , Animais , Linhagem Celular , Proliferação de Células , Cloretos/metabolismo , Modelos Animais de Doenças , Retículo Endoplasmático/metabolismo , Espaço Intracelular/metabolismo , Transporte de Íons , Camundongos , Camundongos Knockout , Doenças Renais Policísticas/patologia , Proteína Quinase C/deficiência , Canais de Cátion TRPP/deficiência
10.
J Immunol ; 204(9): 2439-2446, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32198145

RESUMO

We reported that protein kinase C-η (PKCη) forms a novel (to our knowledge) signaling complex with the checkpoint inhibitory protein CTLA-4 in regulatory T cells (Tregs). This complex is required for the contact-dependent suppressive activity of Tregs, including suppression of antitumor immunity. However, the importance of PKCη in protective immunity mediated by T effector cells remains unclear. We used mice with germline or conditional Treg-specific deletion of Prkch, the PKCη-encoding gene, to explore CD8+ T cell-dependent antiviral immunity using the lymphocytic choriomeningitis virus Armstrong strain acute infection model as well as the in vitro activation of murine or human CD8+ T cells. Five days following infection, germline Prkch -/- mice displayed enhanced viral clearance compared with control mice. Similarly, Prkch Treg-specific conditional knockout mice also showed improved viral clearance and displayed enhanced expression of granzyme B and IFN-γ by both virus-specific and total CD8+ T cells, demonstrating that enhanced viral clearance in germline Prkch -/- mice is caused by PKCη deficiency in Tregs and the resulting functional defect of Prkch -/- Tregs. In addition, purified Prkch -/- mouse CD8+ T cells as well as PRKCH knockdown human CD8+ T cells displayed intact, or even enhanced, T cell activation in vitro as measured by proliferation and expression of granzyme B and IFN-γ. Thus, global PKCη deletion does not impair overall CD8+ T cell-mediated immunity, including antiviral immunity, implying that selective pharmacological PKCη inhibition could be safely used in vivo to inhibit undesired contact-dependent suppression by Tregs and, thus, enhance tumor-specific and, likely, virus-specific immunity.


Assuntos
Linfócitos T CD8-Positivos , Ativação Linfocitária , Proteína Quinase C , Linfócitos T Reguladores , Viroses , Animais , Humanos , Camundongos , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Antígeno CTLA-4/imunologia , Granzimas/imunologia , Células HEK293 , Imunidade Celular/efeitos dos fármacos , Imunidade Celular/imunologia , Interferon gama/imunologia , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Camundongos Knockout , Proteína Quinase C/deficiência , Proteína Quinase C/imunologia , Inibidores de Proteínas Quinases/imunologia , Inibidores de Proteínas Quinases/farmacologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Viroses/imunologia
11.
Hepatology ; 72(5): 1717-1734, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32048304

RESUMO

BACKGROUND AND AIMS: Liver fibrosis (LF) is a central pathological process that occurs in most types of chronic liver diseases. Advanced LF causes cirrhosis, hepatocellular carcinoma, and liver failure. However, the exact molecular mechanisms underlying the initiation and progression of LF remain largely unknown. APPROACH AND RESULTS: This study was designed to investigate the role of protein kinase D3 (PKD3; gene name Prkd3) in the regulation of liver homeostasis. We generated global Prkd3 knockout (Prkd3-/- ) mice and myeloid-cell-specific Prkd3 knockout (Prkd3∆LysM ) mice, and we found that both Prkd3-/- mice and Prkd3∆LysM mice displayed spontaneous LF. PKD3 deficiency also aggravated CCl4 -induced LF. PKD3 is highly expressed in hepatic macrophages (HMs), and PKD3 deficiency skewed macrophage polarization toward a profibrotic phenotype. Activated profibrotic macrophages produced transforming growth factor beta that, in turn, activates hepatic stellate cells to become matrix-producing myofibroblasts. Moreover, PKD3 deficiency decreased the phosphatase activity of SH2-containing protein tyrosine phosphatase-1 (a bona-fide PKD3 substrate), resulting in sustained signal transducer and activator of transcription 6 activation in macrophages. In addition, we observed that PKD3 expression in HMs was down-regulated in cirrhotic human liver tissues. CONCLUSIONS: PKD3 deletion in mice drives LF through the profibrotic macrophage activation.


Assuntos
Cirrose Hepática Experimental/patologia , Cirrose Hepática/patologia , Proteína Quinase C/deficiência , Animais , Tetracloreto de Carbono/toxicidade , Células Cultivadas , Progressão da Doença , Células Estreladas do Fígado/metabolismo , Humanos , Fígado/citologia , Fígado/patologia , Cirrose Hepática Experimental/induzido quimicamente , Cirrose Hepática Experimental/diagnóstico , Cirrose Hepática Experimental/genética , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Miofibroblastos/metabolismo , Cultura Primária de Células , Proteína Quinase C/genética , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Índice de Gravidade de Doença , Análise Serial de Tecidos , Fator de Crescimento Transformador beta/metabolismo
12.
Sci Rep ; 10(1): 231, 2020 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-31937827

RESUMO

Polycystic kidney disease is a complex clinical entity which comprises a group of genetic diseases that leads to renal cyst development. We evaluated the most suitable housekeeping genes for microRNA expression by RT-qPCR analyses of kidney tissues in Pkd1-deficient mouse models from a panel of five candidates genes (miR-20a, miR-25, miR-26a, miR-191 and U6) and 3 target genes (miR-17, miR-21 and let-7a) using samples from kidneys of cystic mice (Pkd1flox/flox:Nestincre, CY), non-cystic controls (Pkd1flox/flox, NC), Pkd1-haploinsufficient (Pkd1+/-, HT), wild-type controls (Pkd1+/+, WT), severely cystic mice (Pkd1V/V, SC), wild-type controls (CO). The stability of the candidate genes was investigated using NormFinder, GeNorm, BestKeeper, DataAssist, and RefFinder software packages and the comparative ΔCt method. The analyses identified miR-26a as the most stable housekeeping gene for all kidney samples, miR-20a for CY and NC, miR-20a and miR-26a for HT and WT, and miR-25 and miR-26a for SC and CO. Expression of miR-21 was upregulated in SC compared to CO and trends of miR-21 upregulation and let-7a downregulation in CY and HT compared to its control kidneys, when normalized by different combinations of miR-20a, miR-25 and miR-26a. Our findings established miR-20a, miR-25, and miR-26a as the best housekeeping genes for miRNA expression analyses by RT-qPCR in kidney tissues of Pkd1-deficient mouse models.


Assuntos
Perfilação da Expressão Gênica , Genes Essenciais/genética , Rim/metabolismo , MicroRNAs/genética , Proteína Quinase C/deficiência , Animais , Haploinsuficiência , Camundongos , Proteína Quinase C/genética
13.
Biochem Biophys Res Commun ; 523(2): 398-404, 2020 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-31870546

RESUMO

The role of protein kinase N1 (PKN1) in cell aggregation and spheroid formation was investigated using mouse embryonic fibroblasts (MEFs) deficient in kinase activity caused by a point mutation (T778A) in the activation loop. Wild type (WT) MEFs formed cell aggregates within a few hours in suspension cultures placed in poly-2-hydroxyethylmethacrylate (poly-HEMA) coated flat-bottom dishes. By contrast, PKN1[T778A] (PKN1 T778A/T778A homozygous knock-in) MEFs showed significantly delayed aggregate formation and higher susceptibility to cell death. Video analysis of suspension cultures revealed decreased cell motility and lesser frequency of cell-cell contact in PKN1[T778A] MEFs compared to that in WT MEFs. Aggregate formation of PKN1[T778A] MEFs was compensated by shaking the cell suspension. When cultured in U-shaped ultra-low attachment well plates, initially larger-sized and loosely packed aggregates of WT MEFs underwent compaction resulting in a single round spheroid. On the other hand, image-based quantitative analysis of PKN1[T778A] MEFs revealed irregular compaction with decreased roundness, solidity, and sphericity within 24 h. Flow cytometry of PKN1[T778A] MEFs revealed decreased surface-expression of N-cadherin and integrins α5 and αV. These results suggest that kinase activity of PKN1 controls cell aggregation and spheroid compaction in MEF suspension culture, possibly by regulating the cell migration and cell-surface expression of N-cadherin and integrins.


Assuntos
Proteína Quinase C/metabolismo , Animais , Caderinas/metabolismo , Agregação Celular/fisiologia , Membrana Celular/metabolismo , Sobrevivência Celular/fisiologia , Células Cultivadas , Fibroblastos/citologia , Fibroblastos/enzimologia , Técnicas de Introdução de Genes , Integrina alfa5/metabolismo , Integrina alfaV/metabolismo , Camundongos , Camundongos Mutantes , Mutação Puntual , Proteína Quinase C/deficiência , Proteína Quinase C/genética , Esferoides Celulares/citologia , Esferoides Celulares/enzimologia
14.
Circulation ; 140(21): 1737-1752, 2019 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-31564129

RESUMO

BACKGROUND: Heart failure is a complex syndrome that results from structural or functional impairment of ventricular filling or blood ejection. Protein phosphorylation is a major and essential intracellular mechanism that mediates various cellular processes in cardiomyocytes in response to extracellular and intracellular signals. The RHOA-associated protein kinase (ROCK/Rho-kinase), an effector regulated by the small GTPase RHOA, causes pathological phosphorylation of proteins, resulting in cardiovascular diseases. RHOA also activates protein kinase N (PKN); however, the role of PKN in cardiovascular diseases remains unclear. METHODS: To explore the role of PKNs in heart failure, we generated tamoxifen-inducible, cardiomyocyte-specific PKN1- and PKN2-knockout mice by intercrossing the αMHC-CreERT2 line with Pkn1flox/flox and Pkn2flox/flox mice and applied a mouse model of transverse aortic constriction- and angiotensin II-induced heart failure. To identify a novel substrate of PKNs, we incubated GST-tagged myocardin-related transcription factor A (MRTFA) with recombinant GST-PKN-catalytic domain or GST-ROCK-catalytic domain in the presence of radiolabeled ATP and detected radioactive GST-MRTFA as phosphorylated MRTFA. RESULTS: We demonstrated that RHOA activates 2 members of the PKN family of proteins, PKN1 and PKN2, in cardiomyocytes of mice with cardiac dysfunction. Cardiomyocyte-specific deletion of the genes encoding Pkn1 and Pkn2 (cmc-PKN1/2 DKO) did not affect basal heart function but protected mice from pressure overload- and angiotensin II-induced cardiac dysfunction. Furthermore, we identified MRTFA as a novel substrate of PKN1 and PKN2 and found that MRTFA phosphorylation by PKN was considerably more effective than that by ROCK in vitro. We confirmed that endogenous MRTFA phosphorylation in the heart was induced by pressure overload- and angiotensin II-induced cardiac dysfunction in wild-type mice, whereas cmc-PKN1/2 DKO mice suppressed transverse aortic constriction- and angiotensin II-induced phosphorylation of MRTFA. Although RHOA-mediated actin polymerization accelerated MRTFA-induced gene transcription, PKN1 and PKN2 inhibited the interaction of MRTFA with globular actin by phosphorylating MRTFA, causing increased serum response factor-mediated expression of cardiac hypertrophy- and fibrosis-associated genes. CONCLUSIONS: Our results indicate that PKN1 and PKN2 activation causes cardiac dysfunction and is involved in the transition to heart failure, thus providing unique targets for therapeutic intervention for heart failure.


Assuntos
Actinas/metabolismo , Insuficiência Cardíaca/enzimologia , Miócitos Cardíacos/enzimologia , Proteína Quinase C/metabolismo , Transativadores/metabolismo , Animais , Modelos Animais de Doenças , Regulação da Expressão Gênica , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos Cardíacos/patologia , Fosforilação , Ligação Proteica , Proteína Quinase C/deficiência , Proteína Quinase C/genética , Transdução de Sinais , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
15.
Cancer Cell ; 36(2): 156-167.e7, 2019 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-31378680

RESUMO

We report that mouse LSL-KrasG12D;Trp53fl/fl (KP)-mediated lung adenocarcinoma (LADC) tumorigenesis can proceed through both PKCι-dependent and PKCι-independent pathways. The predominant pathway involves PKCι-dependent transformation of bronchoalveolar stem cells (BASCs). However, KP mice harboring conditional knock out Prkci alleles (KPI mice) develop LADC tumors through PKCι-independent transformation of Axin2+ alveolar type 2 (AT2) stem cells. Transformed growth of KPI, but not KP, tumors is blocked by Wnt pathway inhibition in vitro and in vivo. Furthermore, a KPI-derived genomic signature predicts sensitivity of human LADC cells to Wnt inhibition, and identifies a distinct subset of primary LADC tumors exhibiting a KPI-like genotype. Thus, LADC can develop through both PKCι-dependent and PKCι-independent pathways, resulting in tumors exhibiting distinct oncogenic signaling and pharmacologic vulnerabilities.


Assuntos
Adenocarcinoma de Pulmão/enzimologia , Transformação Celular Neoplásica/metabolismo , Genes ras , Isoenzimas/metabolismo , Neoplasias Pulmonares/enzimologia , Proteína Quinase C/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Via de Sinalização Wnt , Adenocarcinoma de Pulmão/tratamento farmacológico , Adenocarcinoma de Pulmão/genética , Adenocarcinoma de Pulmão/patologia , Células Epiteliais Alveolares/metabolismo , Células Epiteliais Alveolares/patologia , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Feminino , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Isoenzimas/deficiência , Isoenzimas/genética , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Proteína Quinase C/deficiência , Proteína Quinase C/genética , Inibidores de Proteínas Quinases/farmacologia , Carga Tumoral , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética , beta Catenina/genética , beta Catenina/metabolismo
16.
Biochem Biophys Res Commun ; 514(2): 428-435, 2019 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-31053300

RESUMO

Excessive oxidative stress induces significant injury and cytotoxicity to neuronal cells. The current study tested expression and the potential function of the circular RNA PRKCI (circPRKCI) in oxidative stress-injured neuronal cells. In cultured SH-SY5Y neuronal cells, hydrogen peroxide (H2O2) downregulated circPRKCI expression, causing accumulation of miR-545 and miR-589, but reduction of their target, the transcription factor E2F7. Importantly, ectopic overexpression of circPRKCI in SH-SY5Y cells significantly attenuated H2O2-induced cytotoxicity. Conversely, siRNA-mediated knockdown of circPRKCI induced SH-SY5Y cell death and apoptosis. Further studies demonstrated that H2O2-induced cytotoxicity in SH-SY5Y cells was inhibited by miR-545/589 inhibitors, but mimicked by miR-545/589 mimics. Importantly, CRISPR/Cas9-mediated knockout (KO) of E2F7 induced potent SH-SY5Y cell death and apoptosis. Furthermore, transfection of circPRKCI siRNA or miR-545/589 mimics were ineffective in E2F7 KO cells. In the primary human neurons, H2O2 stimulation similarly induced circPRKCI downregulation, miR-545/589 accumulation and E2F7 reduction. Moreover, H2O2-induced death and apoptosis in the primary neurons were significantly inhibited by circPRKCI overexpression or miR-545/589 inhibitors. Taken together, our results show that dysregulation of circPRKCI-miR-545/589-E2F7 axis mediated H2O2-induced neuronal cell injury. Targeting this novel cascade could be a fine strategy to protect neurons from oxidative stress.


Assuntos
Fator de Transcrição E2F7/genética , Peróxido de Hidrogênio/toxicidade , Isoenzimas/genética , MicroRNAs/genética , Neurônios/efeitos dos fármacos , Neurônios/patologia , Proteína Quinase C/genética , Apoptose/efeitos dos fármacos , Apoptose/genética , Linhagem Celular , Células Cultivadas , Regulação para Baixo/efeitos dos fármacos , Fator de Transcrição E2F7/deficiência , Técnicas de Inativação de Genes , Humanos , Isoenzimas/deficiência , MicroRNAs/metabolismo , Neurônios/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Proteína Quinase C/deficiência , RNA Interferente Pequeno/genética , Regulação para Cima
17.
J Neurosci ; 39(29): 5773-5793, 2019 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-31101759

RESUMO

Chronic pain is a serious debilitating disease for which effective treatment is still lacking. Acid-sensing ion channel 1a (ASIC1a) has been implicated in nociceptive processing at both peripheral and spinal neurons. However, whether ASIC1a also contributes to pain perception at the supraspinal level remains elusive. Here, we report that ASIC1a in ACC is required for thermal and mechanical hypersensitivity associated with chronic pain. ACC-specific genetic deletion or pharmacological blockade of ASIC1a reduced the probability of cortical LTP induction and attenuated inflammatory thermal hyperalgesia and mechanical allodynia in male mice. Using cell type-specific manipulations, we demonstrate that ASIC1a in excitatory neurons of ACC is a major player in cortical LTP and pain behavior. Mechanistically, we show that ASIC1a tuned pain-related cortical plasticity through protein kinase C λ-mediated increase of membrane trafficking of AMPAR subunit GluA1 in ACC. Importantly, postapplication of ASIC1a inhibitors in ACC reversed previously established nociceptive hypersensitivity in both chronic inflammatory pain and neuropathic pain models. These results suggest that ASIC1a critically contributes to a higher level of pain processing through synaptic potentiation in ACC, which may serve as a promising analgesic target for treatment of chronic pain.SIGNIFICANCE STATEMENT Chronic pain is a debilitating disease that still lacks effective therapy. Ion channels are good candidates for developing new analgesics. Here, we provide several lines of evidence to support an important role of cortically located ASIC1a channel in pain hypersensitivity through promoting long-term synaptic potentiation in the ACC. Our results indicate a promising translational potential of targeting ASIC1a to treat chronic pain.


Assuntos
Canais Iônicos Sensíveis a Ácido/biossíntese , Giro do Cíngulo/metabolismo , Isoenzimas/deficiência , Neuralgia/metabolismo , Plasticidade Neuronal/fisiologia , Medição da Dor/métodos , Proteína Quinase C/deficiência , 6-Ciano-7-nitroquinoxalina-2,3-diona/administração & dosagem , Canais Iônicos Sensíveis a Ácido/genética , Animais , Células Cultivadas , Giro do Cíngulo/efeitos dos fármacos , Isoenzimas/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microinjeções/métodos , Neuralgia/genética , Neuralgia/prevenção & controle , Plasticidade Neuronal/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Medição da Dor/efeitos dos fármacos , Proteína Quinase C/genética
18.
Exp Cell Res ; 380(2): 188-197, 2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-31026442

RESUMO

Although serine/threonine-protein kinases are found to participate in a wide range of cancer progression, the involvement of protein kinase D3 (PRKD3) in gastric cancer has not been explored. Here, we investigated the role of PRKD3 in gastric cancer (GC) and its potential mechanisms. PRKD3 was over-expressed in gastric cancer tissues and cells. In vitro, PRKD3 ectopic expression accelerated the proliferation and growth of GES-1, SGC7901 and MKN-28 cells. By contrast, PRKD3 knockdown suppressed the proliferation of SGC7901 and MKN-28 GC cells. In vivo, xenograted tumorigenesis was blunted by PRKD3 silencing. Mechanistically, PRKD3 up-regulated 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) and activated glycolysis as shown by increased glucose consumption and lactate production. Knockdown of PFKFB3 suppressed the glycolysis in gastric cancer cells with highly expressed PRKD3 but not in PRKD3 silenced cells. PRKD3 over-expression induced phosphorylation of p65 at serine 536 was critical for the up-regulation of glycolytic enzyme PFKFB3. Furthermore, PRKD and PFKFB3 inhibitor suppressed the viability of GC cells. Our results suggest that targeting PRKD3/p65/PFKFB3 cascade maybe a promising therapeutic strategy for gastric cancer.


Assuntos
Glicólise , Fosfofrutoquinase-2/metabolismo , Proteína Quinase C/metabolismo , Neoplasias Gástricas/metabolismo , eIF-2 Quinase/metabolismo , Animais , Proliferação de Células , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Proteína Quinase C/deficiência , Neoplasias Gástricas/patologia
19.
Cancer Cell ; 35(3): 385-400.e9, 2019 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-30827887

RESUMO

Increasingly effective therapies targeting the androgen receptor have paradoxically promoted the incidence of neuroendocrine prostate cancer (NEPC), the most lethal subtype of castration-resistant prostate cancer (PCa), for which there is no effective therapy. Here we report that protein kinase C (PKC)λ/ι is downregulated in de novo and during therapy-induced NEPC, which results in the upregulation of serine biosynthesis through an mTORC1/ATF4-driven pathway. This metabolic reprogramming supports cell proliferation and increases intracellular S-adenosyl methionine (SAM) levels to feed epigenetic changes that favor the development of NEPC characteristics. Altogether, we have uncovered a metabolic vulnerability triggered by PKCλ/ι deficiency in NEPC, which offers potentially actionable targets to prevent therapy resistance in PCa.


Assuntos
Carcinoma Neuroendócrino/patologia , Regulação para Baixo , Isoenzimas/deficiência , Neoplasias da Próstata/patologia , Proteína Quinase C/deficiência , Serina/metabolismo , Fator 4 Ativador da Transcrição/metabolismo , Vias Biossintéticas , Carcinoma Neuroendócrino/genética , Carcinoma Neuroendócrino/metabolismo , Linhagem Celular Tumoral , Metilação de DNA , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , S-Adenosilmetionina/metabolismo
20.
Neuropsychopharmacology ; 43(12): 2390-2398, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30111812

RESUMO

The constitutively active, atypical protein kinase C, protein kinase M-ζ (PKMζ), is exclusively expressed in the brain and its expression increases following exposure to drugs of abuse. However, the limitations of currently available tools have made it difficult to examine the role of PKMζ in cocaine self-administration and relapse. The current study demonstrates that constitutive deletion of PKMζ potentiates cue-induced reinstatement of cocaine seeking and increases both food and cocaine self-administration, without affecting cue-driven food seeking in both male and female mice. Conditional deletion of PKMζ within the nucleus accumbens recapitulated the increase in cocaine taking and seeking seen in the constitutive knockout mice, but only in male animals. Site-specific knockdown of PKMζ in the nucleus accumbens had no effect on cocaine taking or seeking in female mice. Additionally, neither male nor female mice exhibited any alterations in food self-administration or cue-induced reinstatement of food seeking following accumbal deletion of PKMζ. Taken together these results indicate that PKMζ may act to dampen cocaine taking and seeking. Furthermore, these results indicate that PKMζ is playing divergent roles in reward seeking in males and females.


Assuntos
Cocaína/administração & dosagem , Deleção de Genes , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/enzimologia , Proteína Quinase C/deficiência , Animais , Inibidores da Captação de Dopamina/administração & dosagem , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Quinase C/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA