Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
PLoS One ; 16(9): e0256738, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34506510

RESUMO

Neurodegenerative disease (ND) is a growing health burden worldwide, but its causes and treatments remain elusive. Although most cases of ND are sporadic, rare familial cases have been attributed to single genes, which can be investigated in animal models. We have generated a new mutation in the calcium-independent phospholipase A2 (iPLA2) VIA gene CG6718, the Drosophila melanogaster ortholog of human PLA2G6/PARK14, mutations in which cause a suite of NDs collectively called PLA2G6-associated neurodegeneration (PLAN). Our mutants display age-related loss of climbing ability, a symptom of neurodegeneration in flies. Although phospholipase activity commonly is presumed to underlie iPLA2-VIA function, locomotor decline in our mutants is rescued by a transgene carrying a serine-to-alanine mutation in the catalytic residue, suggesting that important functional aspects are independent of phospholipase activity. Additionally, we find that iPLA2-VIA knockdown in either muscle or neurons phenocopies locomotor decline with age, demonstrating its necessity in both neuronal and non-neuronal tissues. Furthermore, RNA in situ hybridization shows high endogenous iPLA2-VIA mRNA expression in adult germ cells, and transgenic HA-tagged iPLA2-VIA colocalizes with mitochondria there. Mutant males are fertile with normal spermatogenesis, while fertility is reduced in mutant females. Mutant female germ cells display age-related mitochondrial aggregation, loss of mitochondrial potential, and elevated cell death. These results suggest that iPLA2-VIA is critical for mitochondrial integrity in the Drosophila female germline, which may provide a novel context to investigate its functions with parallels to PLAN.


Assuntos
Proteínas de Drosophila , Fosfolipases A2 do Grupo X , Músculos , Doenças Neurodegenerativas , Neurônios , Animais , Animais Geneticamente Modificados , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiologia , Drosophila melanogaster , Feminino , Células Germinativas/metabolismo , Células Germinativas/patologia , Fosfolipases A2 do Grupo X/genética , Fosfolipases A2 do Grupo X/fisiologia , Masculino , Mitocôndrias/metabolismo , Músculos/metabolismo , Músculos/patologia , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Neurônios/metabolismo , Neurônios/patologia
2.
Theranostics ; 10(24): 10874-10891, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33042259

RESUMO

Rationale: Men and postmenopausal women are more prone to developing non-alcoholic fatty liver disease/steatohepatitis (NAFLD/NASH) than premenopausal women. However, the pathological links and underlying mechanisms of this disparity are still elusive. The sex-difference in hepatic very low-density lipoprotein (VLDL) assembly and secretion may contribute to NAFLD development. Estrogen-related receptor alpha (ERRα) is a key regulator of several metabolic processes. We hypothesized that ERRα plays a role contributing to the sex-difference in hepatic VLDL assembly and secretion. Methods: VLDL secretion and essential genes governing said process were assessed in male and female mice. Liver-specific ERRα-deficient (ERRαLKO) mice were generated to assess the rate of hepatic VLDL secretion and alteration in target gene expression. Overexpression of either microsomal triglyceride transfer protein (Mttp) or phospholipase A2 G12B (Pla2g12b) by adenovirus was performed to test if the fatty liver phenotype in male ERRαLKO mice was due to defects in hepatic VLDL secretion. Female ERRαLKO mice were put on a diet high in saturated fat, fructose and cholesterol (HFHC) to promote NASH development. Wild type female mice were either ovariectomized or treated with tamoxifen to induce a state of estrogen deficiency or disruption in estrogen signaling. Adenovirus was used to overexpress ERRα in these mice to test if ERRα was sufficient to rescue the suppressed VLDL secretion due to estrogen dysfunction. Finally, wild type male mice on a high-fat diet (HFD) were treated with an ERRα inverse agonist to assess if suppressing ERRα activity pharmacologically would lead to fatty liver development. Results: ERRα is an indispensable mediator modulating hepatic triglyceride-rich very low-density lipoprotein (VLDL-TG) assembly and secretion through coordinately controlling target genes apolipoprotein B (Apob), Mttp and Pla2g12b in a sex-different manner. Hepatic VLDL-TG secretion is blunted in ERRαLKO mice, leading to hepatosteatosis which exacerbates endoplasmic reticulum stress and inflammation paving ways for NASH development. Importantly, ERRα acts downstream of estrogen/ERα signaling in contributing to the sex-difference in hepatic VLDL secretion effecting hepatic lipid homeostasis. Conclusions: Our results highlight ERRα as a key mediator which contributes to the sex disparity in NAFLD development, suggesting that selectively restoring ERRα activity in the liver may be a novel strategy for treating NAFLD/NASH.


Assuntos
Disparidades nos Níveis de Saúde , Lipoproteínas VLDL/metabolismo , Fígado/patologia , Hepatopatia Gordurosa não Alcoólica/metabolismo , Receptores de Estrogênio/metabolismo , Triglicerídeos/metabolismo , Animais , Apolipoproteínas B/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Feminino , Fosfolipases A2 do Grupo X/genética , Fosfolipases A2 do Grupo X/metabolismo , Células HEK293 , Células Hep G2 , Hepatócitos , Humanos , Masculino , Camundongos , Camundongos Knockout , Nitrilas/farmacologia , Hepatopatia Gordurosa não Alcoólica/etiologia , Hepatopatia Gordurosa não Alcoólica/patologia , Cultura Primária de Células , Receptores de Estrogênio/antagonistas & inibidores , Receptores de Estrogênio/genética , Fatores Sexuais , Tiazóis/farmacologia , Receptor ERRalfa Relacionado ao Estrogênio
3.
Biochem Biophys Res Commun ; 527(2): 525-531, 2020 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-32423798

RESUMO

Soft tissue leiomyosarcoma (STLMS) is a major histological subtype of adult sarcoma. Although the molecular mechanisms ofLMS have been gradually revealed, no valid therapeutic targets have been identified. In this study, we performed a systematic screening to explore relapse-associated genes in STLMS, using data from The Cancer Genome Atlas-Sarcoma (TCGA-SARC). Then, we investigated the functional role of the gene with the best relapse-prediction value in STLMS by both in-vitro and in-vivo studies. Results showed that AMH and PLA2G10 were two genes with area under curve (AUC) values higher than 0.80 in ROC analysis when detecting relapse. Patients in the high AMH or PLA2G10 expression group had significantly worse relapse-free survival (RFS) compared to the respective low expression group. PLA2G10 was highly expressed in STLMS, but not in other sarcoma subtypes. PLA2G10 overexpression promoted SK-LMS-1 cell growth and G1/S transition, while PLA2G10 knockdown slowed the growth and resulted in G1 phase arrest. PLA2G10 overexpression markedly increased the expression of CDK2 and cyclin E1, but did not influence CDK4, CDK6, cyclin D1, CDK1 or cyclin A expression. PLA2G10 overexpression enhanced SK-LMS-1 cell-derived xenograft tumor growth in nude mice, while PLA2G10 inhibition slowed the growth. Mutation of two critical catalyzing amino acid residues (p.H88A and p.D89A) abrogated the capability of PLA2G10 to catalyze the production of arachidonic acid (AA), and also canceled the regulatory effects on cyclin E1 and CDK2 expression, as well as G1/S transition. In conclusion, PLA2G10 was a specific relapse-associated gene in STLMS. It facilitated the cell-cycle progression of STLMS cells at least by elevating the expression of cyclin E1 and CDK2. The hydrolytic activity was crucial for its oncogenic properties.


Assuntos
Ciclina E/genética , Quinase 2 Dependente de Ciclina/genética , Regulação Neoplásica da Expressão Gênica , Fosfolipases A2 do Grupo X/genética , Leiomiossarcoma/genética , Proteínas Oncogênicas/genética , Animais , Ciclo Celular , Linhagem Celular Tumoral , Feminino , Humanos , Leiomiossarcoma/patologia , Camundongos Endogâmicos BALB C , Camundongos Nus , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/patologia
4.
Proc Natl Acad Sci U S A ; 116(41): 20689-20699, 2019 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-31548400

RESUMO

Mutations in the iPLA2-VIA/PLA2G6 gene are responsible for PARK14-linked Parkinson's disease (PD) with α-synucleinopathy. However, it is unclear how iPLA2-VIA mutations lead to α-synuclein (α-Syn) aggregation and dopaminergic (DA) neurodegeneration. Here, we report that iPLA2-VIA-deficient Drosophila exhibits defects in neurotransmission during early developmental stages and progressive cell loss throughout the brain, including degeneration of the DA neurons. Lipid analysis of brain tissues reveals that the acyl-chain length of phospholipids is shortened by iPLA2-VIA loss, which causes endoplasmic reticulum (ER) stress through membrane lipid disequilibrium. The introduction of wild-type human iPLA2-VIA or the mitochondria-ER contact site-resident protein C19orf12 in iPLA2-VIA-deficient flies rescues the phenotypes associated with altered lipid composition, ER stress, and DA neurodegeneration, whereas the introduction of a disease-associated missense mutant, iPLA2-VIA A80T, fails to suppress these phenotypes. The acceleration of α-Syn aggregation by iPLA2-VIA loss is suppressed by the administration of linoleic acid, correcting the brain lipid composition. Our findings suggest that membrane remodeling by iPLA2-VIA is required for the survival of DA neurons and α-Syn stability.


Assuntos
Encéfalo/patologia , Membrana Celular/patologia , Neurônios Dopaminérgicos/patologia , Proteínas de Drosophila/metabolismo , Fosfolipases A2 do Grupo X/metabolismo , Degeneração Neural/patologia , Doença de Parkinson/patologia , alfa-Sinucleína/química , Animais , Animais Geneticamente Modificados , Encéfalo/metabolismo , Membrana Celular/metabolismo , Neurônios Dopaminérgicos/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster , Estresse do Retículo Endoplasmático , Feminino , Fosfolipases A2 do Grupo VI/genética , Fosfolipases A2 do Grupo VI/metabolismo , Fosfolipases A2 do Grupo X/genética , Humanos , Masculino , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Degeneração Neural/metabolismo , Doença de Parkinson/metabolismo , Fosfolipídeos/metabolismo , Transmissão Sináptica , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
5.
FASEB J ; 33(7): 7942-7952, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30922124

RESUMO

Hypersecretion of hepatic very LDL (VLDL)-associated triglyceride (TG) is the hallmark of hypertriglyceridemia. The estrogen-related receptor γ (ERRγ), an orphan nuclear receptor, plays crucial roles in the regulation of metabolic homeostasis, including TG formation in the liver. It remains unclear whether ERRγ regulates hepatic VLDL-TG secretion. We demonstrated that knockdown of ERRγ impairs hepatic VLDL-TG secretion in mice, whereas overexpression of ERRγ favors the secretion, indicating a novel role of ERRγ in hepatic TG metabolism. We found that ERRγ transcriptionally regulates the expression of PLA2G12B by binding to the promoter region of the Pla2g12b gene. In Pla2g12b-null mice, ERRγ fails to regulate hepatic VLDL-TG secretion. There is an apparent accumulation of large lipid droplets in the liver of Pla2g12b-null mice. These data suggest that ERRγ is a novel regulator of hepatic VLDL-TG secretion, which is mediated through the action on PLA2G12B.-Chen, L., Wu, M., Zhang, S., Tan, W., Guan, M., Feng, L., Chen, C., Tao, J., Chen, L., Qu, L. Estrogen-related receptor γ regulates hepatic triglyceride metabolism through phospholipase A2 G12B.


Assuntos
Fosfolipases A2 do Grupo X/fisiologia , Lipoproteínas VLDL/metabolismo , Fígado/metabolismo , Receptores de Estrogênio/fisiologia , Triglicerídeos/metabolismo , Animais , Linhagem Celular , Colesterol/sangue , Técnicas de Silenciamento de Genes , Fosfolipases A2 do Grupo X/deficiência , Fosfolipases A2 do Grupo X/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Regiões Promotoras Genéticas , Interferência de RNA , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Receptores de Estrogênio/genética , Proteínas Recombinantes/metabolismo , Transcrição Gênica , Triglicerídeos/sangue , Regulação para Cima
6.
Sci Rep ; 8(1): 2939, 2018 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-29440694

RESUMO

Infantile neuroaxonal dystrophy (INAD) is a fatal neurodegenerative disorder that typically begins within the first few years of life and leads to progressive impairment of movement and cognition. Several years ago, it was shown that >80% of patients with INAD have mutations in the phospholipase gene, PLA2G6. Interestingly, mutations in PLA2G6 are also causative in two other related neurodegenerative diseases, atypical neuroaxonal dystrophy and Dystonia-parkinsonism. While all three disorders give rise to similar defects in movement and cognition, some defects are unique to a specific disorder. At present, the cellular mechanisms underlying PLA2G6-associated neuropathology are poorly understood and there is no cure or treatment that can delay disease progression. Here, we show that loss of iPLA2-VIA, the Drosophila homolog of PLA2G6, gives rise to age-dependent defects in climbing and spontaneous locomotion. Moreover, using a newly developed assay, we show that iPLA2-VIA mutants also display impairments in fine-tune motor movements, motor coordination and psychomotor learning, which are distinct features of PLA2G6-associated disease in humans. Finally, we show that iPLA2-VIA mutants exhibit increased sensitivity to oxidative stress, progressive neurodegeneration and a severely reduced lifespan. Altogether, these data demonstrate that Drosophila iPLA2-VIA mutants provide a useful model to study human PLA2G6-associated neurodegeneration.


Assuntos
Proteínas de Drosophila/genética , Drosophila melanogaster , Fosfolipases A2 do Grupo X/genética , Atividade Motora/genética , Mutação , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/fisiopatologia , Homologia de Sequência do Ácido Nucleico , Envelhecimento/genética , Alelos , Animais , Cálcio/metabolismo , Proteínas de Drosophila/metabolismo , Feminino , Fosfolipases A2 do Grupo X/metabolismo , Humanos , Longevidade , Masculino , Doenças Neurodegenerativas/metabolismo , Estresse Oxidativo/genética
7.
JCI Insight ; 2(21)2017 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-29093264

RESUMO

Phospholipase A2 (PLA2) enzymes regulate the formation of eicosanoids and lysophospholipids that contribute to allergic airway inflammation. Secreted PLA2 group X (sPLA2-X) was recently found to be increased in the airways of asthmatics and is highly expressed in airway epithelial cells and macrophages. In the current study, we show that allergen exposure increases sPLA2-X in humans and in mice, and that global deletion of Pla2g10 results in a marked reduction in airway hyperresponsiveness (AHR), eosinophil and T cell trafficking to the airways, airway occlusion, generation of type-2 cytokines by antigen-stimulated leukocytes, and antigen-specific immunoglobulins. Further, we found that Pla2g10-/- mice had reduced IL-33 levels in BALF, fewer type-2 innate lymphoid cells (ILC2s) in the lung, less IL-33-induced IL-13 expression in mast cells, and a marked reduction in both the number of newly recruited macrophages and the M2 polarization of these macrophages in the lung. These results indicate that sPLA2-X serves as a central regulator of both innate and adaptive immune response to proteolytic allergen.


Assuntos
Imunidade Adaptativa/imunologia , Alérgenos/imunologia , Asma/imunologia , Fosfolipases A2 do Grupo X/imunologia , Imunidade Inata/imunologia , Fosfolipases A2/imunologia , Fosfolipases A2/metabolismo , Animais , Citocinas/imunologia , Modelos Animais de Doenças , Eicosanoides/análise , Feminino , Deleção de Genes , Fosfolipases A2 do Grupo X/genética , Fosfolipases A2 do Grupo X/metabolismo , Imunoglobulinas , Inflamação , Interleucina-13/metabolismo , Interleucina-33/metabolismo , Leucócitos/imunologia , Pulmão/imunologia , Pulmão/metabolismo , Macrófagos , Mastócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
8.
Mol Cell Endocrinol ; 436: 93-101, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27471003

RESUMO

As a novel mediator of hepatic very low-density lipoproteins (VLDL) secretion, phospholipase A2 G12B (PLA2G12B) is transcriptionally regulated by hepatocyte nuclear factor-4 alpha (HNF-4α). Farnesoid X receptor (FXR) plays a critical role in maintaining bile acids and triglycerides (TG) homeostasis. Here we report that FXR regulates serum TG level in part through PLA2G12B. Activation of FXR by chenodeoxycholic acid (CDCA) or GW4064 significantly decreased PLA2G12B expression in HepG2 cells. PLA2G12B expression was transcriptionally repressed due to an FXR-mediated up-regulation of small heterodimer partner (SHP) which functionally suppresses HNF-4α activity. We found that hepatic PLA2G12B expression was suppressed and serum TG level reduced in high fat diet mice treated with CDCA. Concurrently, CDCA treatment lowered hepatic VLDL-TG secretion. Our data demonstrate that activation of FXR promotes TG lowering, not only by decreasing de novo lipogenesis but also reducing hepatic secretion of TG-rich VLDL particles in part through suppressing PLA2G12B expression.


Assuntos
Fosfolipases A2 do Grupo X/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Triglicerídeos/metabolismo , Animais , Ácido Quenodesoxicólico/farmacologia , Dieta Hiperlipídica , Regulação da Expressão Gênica/efeitos dos fármacos , Fosfolipases A2 do Grupo X/metabolismo , Células Hep G2 , Humanos , Hiperlipidemias/patologia , Isoxazóis/farmacologia , Ligantes , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/genética , Lipoproteínas VLDL/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Regiões Promotoras Genéticas , Receptores Citoplasmáticos e Nucleares/agonistas
9.
J Biol Chem ; 291(13): 6895-911, 2016 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-26828067

RESUMO

Within the secreted phospholipase A2(sPLA2) family, group X sPLA2(sPLA2-X) has the highest capacity to hydrolyze cellular membranes and has long been thought to promote inflammation by releasing arachidonic acid, a precursor of pro-inflammatory eicosanoids. Unexpectedly, we found that transgenic mice globally overexpressing human sPLA2-X (PLA2G10-Tg) displayed striking immunosuppressive and lean phenotypes with lymphopenia and increased M2-like macrophages, accompanied by marked elevation of free ω3 polyunsaturated fatty acids (PUFAs) and their metabolites. Studies usingPla2g10-deficient mice revealed that endogenous sPLA2-X, which is highly expressed in the colon epithelium and spermatozoa, mobilized ω3 PUFAs or their metabolites to protect against dextran sulfate-induced colitis and to promote fertilization, respectively. In colitis, sPLA2-X deficiency increased colorectal expression of Th17 cytokines, and ω3 PUFAs attenuated their production by lamina propria cells partly through the fatty acid receptor GPR120. In comparison, cytosolic phospholipase A2(cPLA2α) protects from colitis by mobilizing ω6 arachidonic acid metabolites, including prostaglandin E2 Thus, our results underscore a previously unrecognized role of sPLA2-X as an ω3 PUFA mobilizerin vivo, segregated mobilization of ω3 and ω6 PUFA metabolites by sPLA2-X and cPLA2α, respectively, in protection against colitis, and the novel role of a particular sPLA2-X-driven PUFA in fertilization.


Assuntos
Colite/genética , Colo/enzimologia , Ácidos Graxos Ômega-3/biossíntese , Fertilidade/genética , Fosfolipases A2 do Grupo X/genética , Espermatozoides/enzimologia , Animais , Ácido Araquidônico/antagonistas & inibidores , Ácido Araquidônico/biossíntese , Colite/induzido quimicamente , Colite/enzimologia , Colite/terapia , Colo/patologia , Sulfato de Dextrana , Ácidos Graxos Ômega-3/metabolismo , Ácidos Graxos Ômega-6/biossíntese , Ácidos Graxos Ômega-6/metabolismo , Expressão Gênica , Perfilação da Expressão Gênica , Fosfolipases A2 do Grupo X/metabolismo , Humanos , Interleucina-17/biossíntese , Masculino , Camundongos , Camundongos Transgênicos , Fosfolipases A2/genética , Fosfolipases A2/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Contagem de Espermatozoides , Motilidade dos Espermatozoides , Espermatozoides/patologia , Células Th17/metabolismo , Células Th17/patologia , Transgenes
10.
J Biol Chem ; 291(6): 3076-89, 2016 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-26655718

RESUMO

Phospholipase A2 (PLA2) activity has been shown to be involved in the sperm acrosome reaction (AR), but the molecular identity of PLA2 isoforms has remained elusive. Here, we have tested the role of two intracellular (iPLA2ß and cytosolic PLA2α) and one secreted (group X) PLA2s in spontaneous and progesterone (P4)-induced AR by using a set of specific inhibitors and knock-out mice. iPLA2ß is critical for spontaneous AR, whereas both iPLA2ß and group X secreted PLA2 are involved in P4-induced AR. Cytosolic PLA2α is dispensable in both types of AR. P4-induced AR spreads over 30 min in the mouse, and kinetic analyses suggest the presence of different sperm subpopulations, using distinct PLA2 pathways to achieve AR. At low P4 concentration (2 µm), sperm undergoing early AR (0-5 min post-P4) rely on iPLA2ß, whereas sperm undergoing late AR (20-30 min post-P4) rely on group X secreted PLA2. Moreover, the role of PLA2s in AR depends on P4 concentration, with the PLA2s being key actors at low physiological P4 concentrations (≤2 µm) but not at higher P4 concentrations (~10 µm).


Assuntos
Reação Acrossômica/efeitos dos fármacos , Acrossomo/enzimologia , Exocitose/efeitos dos fármacos , Fosfolipases A2 do Grupo VI/metabolismo , Fosfolipases A2 do Grupo X/metabolismo , Progesterona/farmacologia , Animais , Fosfolipases A2 do Grupo VI/genética , Fosfolipases A2 do Grupo X/genética , Masculino , Camundongos , Camundongos Knockout , Progesterona/metabolismo
11.
Anticancer Res ; 35(5): 2983-90, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25964585

RESUMO

BACKGROUND: Although secretory phospholipase A2 (sPLA2) has been shown to be involved in various biological processes, its specific roles in sub-types of cancer development remain to be elucidated. MATERIALS AND METHODS: We examined the expression of sPLA2 group III (GIII) in 142 patients with colorectal cancer using immunohistochemistry, and its correlation with clinicopathological features and outcomes. In addition, we examined the co-expression of sPLA2GIII and sPLA2GX using serial tissue sections to clarify the roles of both proteins in colorectal carcinogenesis. RESULTS: In 66 cases, diffuse staining of sPLA2GIII was seen; this was defined as the group with high expression. High expression was associated with a significantly higher rate of lymph node metastasis (p=0.02) and poorer survival (p=0.03) compared with low expression. Patients with low sPLA2GIII and high sPLA2GX expression had a significantly higher survival rate than those with high sPLA2GIII and low sPLA2GX expression (p=0.038). CONCLUSION: sPLA2GIII expression may be used as a risk factor for lymph node metastasis and a prognostic marker in colorectal cancer. In addition, sPLA2GIII and sPLA2GX may play opposing roles in colorectal carcinogenesis.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias Colorretais/genética , Fosfolipases A2 do Grupo III/genética , Fosfolipases A2 do Grupo X/genética , Adulto , Idoso , Neoplasias Colorretais/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Fosfolipases A2 do Grupo III/biossíntese , Fosfolipases A2 do Grupo X/biossíntese , Humanos , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Prognóstico , Fatores de Risco , Taxa de Sobrevida
12.
Brain ; 138(Pt 7): 1801-16, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26001724

RESUMO

The PLA2G6 gene encodes a group VIA calcium-independent phospholipase A2 beta enzyme that selectively hydrolyses glycerophospholipids to release free fatty acids. Mutations in PLA2G6 have been associated with disorders such as infantile neuroaxonal dystrophy, neurodegeneration with brain iron accumulation type II and Karak syndrome. More recently, PLA2G6 was identified as the causative gene in a subgroup of patients with autosomal recessive early-onset dystonia-parkinsonism. Neuropathological examination revealed widespread Lewy body pathology and the accumulation of hyperphosphorylated tau, supporting a link between PLA2G6 mutations and parkinsonian disorders. Here we show that knockout of the Drosophila homologue of the PLA2G6 gene, iPLA2-VIA, results in reduced survival, locomotor deficits and organismal hypersensitivity to oxidative stress. Furthermore, we demonstrate that loss of iPLA2-VIA function leads to a number of mitochondrial abnormalities, including mitochondrial respiratory chain dysfunction, reduced ATP synthesis and abnormal mitochondrial morphology. Moreover, we show that loss of iPLA2-VIA is strongly associated with increased lipid peroxidation levels. We confirmed our findings using cultured fibroblasts taken from two patients with mutations in the PLA2G6 gene. Similar abnormalities were seen including elevated mitochondrial lipid peroxidation and mitochondrial membrane defects, as well as raised levels of cytoplasmic and mitochondrial reactive oxygen species. Finally, we demonstrated that deuterated polyunsaturated fatty acids, which inhibit lipid peroxidation, were able to partially rescue the locomotor abnormalities seen in aged flies lacking iPLA2-VIA gene function, and restore mitochondrial membrane potential in fibroblasts from patients with PLA2G6 mutations. Taken together, our findings demonstrate that loss of normal PLA2G6 gene activity leads to lipid peroxidation, mitochondrial dysfunction and subsequent mitochondrial membrane abnormalities. Furthermore we show that the iPLA2-VIA knockout fly model provides a useful platform for the further study of PLA2G6-associated neurodegeneration.


Assuntos
Proteínas de Drosophila/genética , Fosfolipases A2 do Grupo VI/genética , Fosfolipases A2 do Grupo X/genética , Peroxidação de Lipídeos/genética , Mitocôndrias/metabolismo , Estresse Oxidativo/genética , Animais , Linhagem Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Fibroblastos/metabolismo , Técnicas de Inativação de Genes , Fosfolipases A2 do Grupo VI/metabolismo , Fosfolipases A2 do Grupo X/metabolismo , Humanos , Espectrometria de Massas , Potencial da Membrana Mitocondrial/genética , Microscopia de Fluorescência , Mitocôndrias/patologia , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Circ Cardiovasc Genet ; 8(2): 356-62, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25583995

RESUMO

BACKGROUND: Observational studies report that secretory phospholipase A2 (sPLA2) activity is a marker for coronary heart disease (CHD) risk, and activity measures are thought to represent the composite activity of sPLA2-IIA, -V, and -X. The aim of this study was to use genetic variants of PLA2G10, encoding sPLA2-X, to investigate the contribution of sPLA2-X to the measure of sPLA2 activity and coronary heart disease (CHD) risk traits and outcome. METHODS AND RESULTS: Three PLA2G10 tagging single-nucleotide polymorphisms (rs72546339, rs72546340, and rs4003232) and a previously studied PLA2G10 coding single-nucleotide polymorphism rs4003228, R38C, were genotyped in a nested case: control cohort drawn from the prospective EPIC-Norfolk Study (2175 cases and 2175 controls). Meta-analysis of rs4003228 (R38C) and CHD was performed using data from the Northwick Park Heart Study II and 2 published cohorts AtheroGene and SIPLAC, providing in total an additional 1884 cases and 3119 controls. EPIC-Norfolk subjects in the highest tertile of sPLA2 activity were older and had higher inflammatory markers compared with those in the lowest tertile for sPLA2 activity. None of the PLA2G10 tagging single-nucleotide polymorphism nor R38C, a functional variant, were significantly associated with sPLA2 activity, intermediate CHD risk traits, or CHD risk. In meta-analysis, the summary odds ratio for R38C was odds ratio=0.97 (95% confidence interval, 0.77-1.22). CONCLUSIONS: PLA2G10 variants are not significantly associated with plasma sPLA2 activity or with CHD risk.


Assuntos
Doença das Coronárias , Fosfolipases A2 do Grupo X , Polimorfismo de Nucleotídeo Único , Característica Quantitativa Herdável , Idoso , Idoso de 80 Anos ou mais , Doença das Coronárias/sangue , Doença das Coronárias/enzimologia , Doença das Coronárias/genética , Feminino , Seguimentos , Fosfolipases A2 do Grupo X/sangue , Fosfolipases A2 do Grupo X/genética , Humanos , Masculino , Metanálise como Assunto , Pessoa de Meia-Idade , Estudos Prospectivos , Fatores de Risco
14.
J Biol Chem ; 289(40): 27410-7, 2014 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-25122761

RESUMO

Group X secretory phospholipase A2 (GX sPLA2) potently hydrolyzes membrane phospholipids to release arachidonic acid (AA). While AA is an activator of glucose-stimulated insulin secretion (GSIS), its metabolite prostaglandin E2 (PGE2) is a known inhibitor. In this study, we determined that GX sPLA2 is expressed in insulin-producing cells of mouse pancreatic islets and investigated its role in beta cell function. GSIS was measured in vivo in wild-type (WT) and GX sPLA2-deficient (GX KO) mice and ex vivo using pancreatic islets isolated from WT and GX KO mice. GSIS was also assessed in vitro using mouse MIN6 pancreatic beta cells with or without GX sPLA2 overexpression or exogenous addition. GSIS was significantly higher in islets isolated from GX KO mice compared with islets from WT mice. Conversely, GSIS was lower in MIN6 cells overexpressing GX sPLA2 (MIN6-GX) compared with control (MIN6-C) cells. PGE2 production was significantly higher in MIN6-GX cells compared with MIN6-C cells and this was associated with significantly reduced cellular cAMP. The effect of GX sPLA2 on GSIS was abolished when cells were treated with NS398 (a COX-2 inhibitor) or L-798,106 (a PGE2-EP3 receptor antagonist). Consistent with enhanced beta cell function, GX KO mice showed significantly increased plasma insulin levels following glucose challenge and were protected from age-related reductions in GSIS and glucose tolerance compared with WT mice. We conclude that GX sPLA2 plays a previously unrecognized role in negatively regulating pancreatic insulin secretion by augmenting COX-2-dependent PGE2 production.


Assuntos
Ciclo-Oxigenase 2/metabolismo , Fosfolipases A2 do Grupo X/metabolismo , Células Secretoras de Insulina/enzimologia , Insulina/metabolismo , Animais , Ciclo-Oxigenase 2/genética , Dinoprostona/metabolismo , Glucose/metabolismo , Fosfolipases A2 do Grupo X/genética , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/enzimologia , Ilhotas Pancreáticas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
15.
Virology ; 454-455: 78-92, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24725934

RESUMO

The role of Group X secreted phospholipase A2 (GX-sPLA2) during influenza infection has not been previously investigated. We examined the role of GX-sPLA2 during H1N1 pandemic influenza infection in a GX-sPLA2 gene targeted mouse (GX(-/-)) model and found that survival after infection was significantly greater in GX(-/-) mice than in GX(+/+) mice. Downstream products of GX-sPLA2 activity, PGD2, PGE2, LTB4, cysteinyl leukotrienes and Lipoxin A4 were significantly lower in GX(-/-) mice BAL fluid. Lung microarray analysis identified an earlier and more robust induction of T and B cell associated genes in GX(-/-) mice. Based on the central role of sPLA2 enzymes as key initiators of inflammatory processes, we propose that activation of GX-sPLA2 during H1N1pdm infection is an early step of pulmonary inflammation and its inhibition increases adaptive immunity and improves survival. Our findings suggest that GX-sPLA2 may be a potential therapeutic target during influenza.


Assuntos
Fosfolipases A2 do Grupo X/deficiência , Vírus da Influenza A Subtipo H1N1/imunologia , Infecções por Orthomyxoviridae/patologia , Infecções por Orthomyxoviridae/virologia , Animais , Linfócitos B/imunologia , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Fosfolipases A2 do Grupo X/genética , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise em Microsséries , Análise de Sobrevida , Linfócitos T/imunologia
16.
Biochem Biophys Res Commun ; 445(1): 230-5, 2014 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-24508801

RESUMO

Secreted phospholipases A2 (sPLA2s) have recently been associated with several cancers, but their role in breast cancer is unknown. Here we demonstrate that mRNA expression of group IIA, III and X sPLA2s differs both in vivo in tumour biopsies and in breast cancer cells in vitro. Their expression is differentially regulated by DNA methylation and histone acetylation and, significantly, all three genes are silenced in aggressive triple negative cells due to both mechanisms. The transcription start site promoter region and the upstream CpG islands, exclusive to the group X sPLA2 gene, have variable roles in the regulation of sPLA2 expression. Our results suggest that the differential expression of hGIIA, hGIII and hGX sPLA2s in breast cancer cells is a consequence of various degrees of epigenetic silencing due to DNA hypermethylation and histone deacetylation.


Assuntos
Neoplasias da Mama/genética , Epigênese Genética , Perfilação da Expressão Gênica , Fosfolipases A2 do Grupo II/genética , Fosfolipases A2 do Grupo III/genética , Fosfolipases A2 do Grupo X/genética , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular , Linhagem Celular Tumoral , Ilhas de CpG/genética , Metilação de DNA/efeitos dos fármacos , Decitabina , Feminino , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Fosfolipases A2 do Grupo II/metabolismo , Fosfolipases A2 do Grupo III/metabolismo , Fosfolipases A2 do Grupo X/metabolismo , Humanos , Ácidos Hidroxâmicos/farmacologia , Células MCF-7 , Regiões Promotoras Genéticas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
PLoS One ; 8(10): e76641, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24204651

RESUMO

BACKGROUND: Phospholipases A2 (PLA2) hydrolyzes phospholipids, initiating the production of inflammatory lipid mediators. We have previously shown that in rats, sPLA2 and cPLA2 play opposing roles in the pathophysiology of ovalbumin (OVA)-induced experimental allergic bronchitis (OVA-EAB), an asthma model: Upon disease induction sPLA2 expression and production of the broncho-constricting CysLTs are elevated, whereas cPLA2 expression and the broncho-dilating PGE2 production are suppressed. These were reversed upon disease amelioration by treatment with an sPLA2 inhibitor. However, studies in mice reported the involvement of both sPLA2 and cPLA2 in EAB induction. OBJECTIVES: To examine the relevance of mouse and rat models to understanding asthma pathophysiology. METHODS: OVA-EAB was induced in mice using the same methodology applied in rats. Disease and biochemical markers in mice were compared with those in rats. RESULTS: As in rats, EAB in mice was associated with increased mRNA of sPLA2, specifically sPLA2gX, in the lungs, and production of the broncho-constricting eicosanoids CysLTs, PGD2 and TBX2 in bronchoalveolar lavage (BAL). In contrast, EAB in mice was associated also with elevated cPLA2 mRNA and PGE2 production. Yet, treatment with an sPLA2 inhibitor ameliorated the EAB concomitantly with reverting the expression of both cPLA2 and sPLA2, and eicosanoid production. CONCLUSIONS: In both mice and rats sPLA2 is pivotal in OVA-induced EAB. Yet, amelioration of asthma markers in mouse models, and human tissues, was observed also upon cPLA2 inhibition. It is plausible that airway conditions, involving multiple cell types and organs, require the combined action of more than one, essential, PLA2s.


Assuntos
Asma/imunologia , Bronquite/imunologia , Fosfolipases A2 Citosólicas/imunologia , Fosfolipases A2 Secretórias/imunologia , Animais , Araquidonato 5-Lipoxigenase/imunologia , Araquidonato 5-Lipoxigenase/metabolismo , Arginase/genética , Arginase/imunologia , Arginase/metabolismo , Asma/genética , Asma/metabolismo , Western Blotting , Bronquite/genética , Bronquite/metabolismo , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/imunologia , Quitinases/genética , Quitinases/imunologia , Quitinases/metabolismo , Cisteína/imunologia , Cisteína/metabolismo , Dinoprostona/imunologia , Dinoprostona/metabolismo , Modelos Animais de Doenças , Feminino , Fosfolipases A2 do Grupo X/genética , Fosfolipases A2 do Grupo X/imunologia , Fosfolipases A2 do Grupo X/metabolismo , Humanos , Leucotrienos/imunologia , Leucotrienos/metabolismo , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Ovalbumina/imunologia , Fosfolipases A2 Citosólicas/genética , Fosfolipases A2 Citosólicas/metabolismo , Fosfolipases A2 Secretórias/genética , Fosfolipases A2 Secretórias/metabolismo , Prostaglandina D2/imunologia , Prostaglandina D2/metabolismo , Ratos , Receptores de Leucotrienos/imunologia , Receptores de Leucotrienos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas com Domínio T/imunologia , Proteínas com Domínio T/metabolismo
19.
Am J Respir Crit Care Med ; 188(1): 42-50, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23614662

RESUMO

RATIONALE: Indirect airway hyperresponsiveness (AHR) is a fundamental feature of asthma that is manifest as exercise-induced bronchoconstriction (EIB). Secreted phospholipase A2 group X (sPLA2-X) plays a key role in regulating eicosanoid formation and the development of inflammation and AHR in murine models. OBJECTIVES: We sought to examine sPLA2-X in the airway epithelium and airway wall of patients with asthma, the relationship to AHR in humans, and the regulation and function of sPLA2-X within the epithelium. METHODS: We precisely phenotyped 34 patients with asthma (19 with and 15 without EIB) and 10 normal control subjects to examine in vivo differences in epithelial gene expression, quantitative morphometry of endobronchial biopsies, and levels of secreted protein. The regulation of sPLA2-X gene (PLA2G10) expression was examined in primary airway epithelial cell cultures. The function of epithelial sPLA2-X in eicosanoid formation was examined using PLA2 inhibitors and murine tracheal epithelial cells with Pla2g10 deletion. MEASUREMENTS AND MAIN RESULTS: We found that sPLA2-X protein is increased in the airways of patients with asthma and that epithelial-derived sPLA2-X may be increased in association with indirect AHR. The expression of sPLA2-X increases during in vitro epithelial differentiation; is regulated by inflammatory signals including tumor necrosis factor, IL-13, and IL-17; and is both secreted from the epithelium and directly participates in the release of arachidonic acid by epithelial cells. CONCLUSIONS: These data reveal a relationship between epithelial-derived sPLA2-X and indirect AHR in asthma and that sPLA2-X serves as an epithelial regulator of inflammatory eicosanoid formation. Therapies targeting epithelial sPLA2-X may be useful in asthma.


Assuntos
Asma/genética , Asma/imunologia , Células Epiteliais/imunologia , Fosfolipases A2 do Grupo X/genética , Fosfolipases A2 do Grupo X/imunologia , Adolescente , Adulto , Animais , Asma Induzida por Exercício/genética , Asma Induzida por Exercício/imunologia , Hiper-Reatividade Brônquica/genética , Hiper-Reatividade Brônquica/imunologia , Ensaio de Imunoadsorção Enzimática/métodos , Feminino , Expressão Gênica/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase em Tempo Real/métodos , Adulto Jovem
20.
PLoS One ; 8(2): e56172, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23451035

RESUMO

BACKGROUND: Previous work has shown that disruption of the gene for group X secreted phospholipase A2 (sPLA2-X) markedly diminishes airway hyperresponsiveness and remodeling in a mouse asthma model. With the large number of additional sPLA2s in the mammalian genome, the involvement of other sPLA2s in the asthma model is possible - in particular, the group V sPLA2 (sPLA2-V) that like sPLA2-X is highly active at hydrolyzing membranes of mammalian cells. METHODOLOGY AND PRINCIPAL FINDINGS: The allergen-driven asthma phenotype was significantly reduced in sPLA2-V-deficient mice but to a lesser extent than observed previously in sPLA2-X-deficient mice. The most striking difference observed between the sPLA2-V and sPLA2-X knockouts was the significant impairment of the primary immune response to the allergen ovalbumin (OVA) in the sPLA2-V(-/-) mice. The impairment in eicosanoid generation and dendritic cell activation in sPLA2-V(-/-) mice diminishes Th2 cytokine responses in the airways. CONCLUSIONS: This paper illustrates the diverse roles of sPLA2s in the immunopathogenesis of the asthma phenotype and directs attention to developing specific inhibitors of sPLA2-V as a potential new therapy to treat asthma and other allergic disorders.


Assuntos
Asma/enzimologia , Asma/imunologia , Fosfolipases A2 do Grupo V/metabolismo , Fosfolipases A2 do Grupo X/metabolismo , Animais , Asma/genética , Linfócitos T CD4-Positivos/metabolismo , Modelos Animais de Doenças , Fosfolipases A2 do Grupo V/genética , Fosfolipases A2 do Grupo X/genética , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Ovalbumina/imunologia , Reação em Cadeia da Polimerase , Células Th2/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA