Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
PLoS Biol ; 19(1): e3001062, 2021 01.
Article in English | MEDLINE | ID: mdl-33395408

ABSTRACT

Lyme carditis is an extracutaneous manifestation of Lyme disease characterized by episodes of atrioventricular block of varying degrees and additional, less reported cardiomyopathies. The molecular changes associated with the response to Borrelia burgdorferi over the course of infection are poorly understood. Here, we identify broad transcriptomic and proteomic changes in the heart during infection that reveal a profound down-regulation of mitochondrial components. We also describe the long-term functional modulation of macrophages exposed to live bacteria, characterized by an augmented glycolytic output, increased spirochetal binding and internalization, and reduced inflammatory responses. In vitro, glycolysis inhibition reduces the production of tumor necrosis factor (TNF) by memory macrophages, whereas in vivo, it produces the reversion of the memory phenotype, the recovery of tissue mitochondrial components, and decreased inflammation and spirochetal burdens. These results show that B. burgdorferi induces long-term, memory-like responses in macrophages with tissue-wide consequences that are amenable to be manipulated in vivo.


Subject(s)
Borrelia burgdorferi/immunology , Cardiomyopathies/etiology , Immunologic Memory , Lyme Disease/immunology , Macrophages/physiology , Animals , Cardiomyopathies/immunology , Cardiomyopathies/microbiology , Cardiomyopathies/pathology , Cells, Cultured , Endocarditis, Bacterial/complications , Endocarditis, Bacterial/immunology , Endocarditis, Bacterial/microbiology , Endocarditis, Bacterial/pathology , Female , HEK293 Cells , Heart/microbiology , Humans , Lyme Disease/pathology , Macrophage Activation/physiology , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/immunology , Myocytes, Cardiac/microbiology , Myocytes, Cardiac/pathology , RAW 264.7 Cells
2.
PLoS Genet ; 10(10): e1004721, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25329316

ABSTRACT

Exposure to ultraviolet (UV) radiation from sunlight accounts for 90% of the symptoms of premature skin aging and skin cancer. The tumor suppressor serine-threonine kinase LKB1 is mutated in Peutz-Jeghers syndrome and in a spectrum of epithelial cancers whose etiology suggests a cooperation with environmental insults. Here we analyzed the role of LKB1 in a UV-dependent mouse skin cancer model and show that LKB1 haploinsufficiency is enough to impede UVB-induced DNA damage repair, contributing to tumor development driven by aberrant growth factor signaling. We demonstrate that LKB1 and its downstream kinase NUAK1 bind to CDKN1A. In response to UVB irradiation, LKB1 together with NUAK1 phosphorylates CDKN1A regulating the DNA damage response. Upon UVB treatment, LKB1 or NUAK1 deficiency results in CDKN1A accumulation, impaired DNA repair and resistance to apoptosis. Importantly, analysis of human tumor samples suggests that LKB1 mutational status could be a prognostic risk factor for UV-induced skin cancer. Altogether, our results identify LKB1 as a DNA damage sensor protein regulating skin UV-induced DNA damage response.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage/radiation effects , Protein Serine-Threonine Kinases/metabolism , Ultraviolet Rays/adverse effects , AMP-Activated Protein Kinases , Animals , Animals, Newborn , Apoptosis/genetics , Apoptosis/radiation effects , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/genetics , Disease Models, Animal , Hepatocyte Growth Factor/genetics , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , Keratinocytes/radiation effects , Mice, Transgenic , Neoplasms, Squamous Cell/etiology , Neoplasms, Squamous Cell/pathology , Phosphorylation , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Repressor Proteins/metabolism , Skin Neoplasms/etiology , Skin Neoplasms/genetics , Skin Neoplasms/pathology
3.
Prostaglandins Other Lipid Mediat ; 106: 106-15, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23579063

ABSTRACT

Cyclooxygenase-2 (COX-2) has been associated with cell growth regulation, tissue remodeling and carcinogenesis. Overexpression of COX-2 in hepatocytes constitutes an ideal condition to evaluate the role of prostaglandins (PGs) in liver pathogenesis. The effect of COX-2-dependent PGs in genetic hepatocarcinogenesis has been investigated in triple c-myc/transforming growth factor α (TGF-α) transgenic mice that express human COX-2 in hepatocytes on a B6CBAxCD1xB6DBA2 background. Analysis of the contribution of COX-2-dependent PGs to the development of hepatocarcinogenesis, evaluated in this model, suggested a minor role of COX-2-dependent prostaglandins to liver oncogenesis as indicated by liver histopathology, morphometric analysis and specific markers of tumor progression. This allows concluding that COX-2 is insufficient for modifying the hepatocarcinogenesis course mediated by c-myc/TGF-α.


Subject(s)
Carcinogenesis/genetics , Carcinoma, Hepatocellular/pathology , Cyclooxygenase 2/genetics , Liver Neoplasms/pathology , Proto-Oncogene Proteins c-myc/genetics , Transforming Growth Factor alpha/genetics , Animals , Carcinoma, Hepatocellular/genetics , Disease Progression , Female , Gene Expression , Humans , Liver/metabolism , Liver/pathology , Liver Neoplasms/genetics , Male , Mice , Mice, Transgenic , Signal Transduction/genetics
4.
Am J Pathol ; 178(3): 1361-73, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21356386

ABSTRACT

Cyclooxygenase-2 (COX-2) has been associated with cell growth regulation, tissue remodeling, and carcinogenesis. Ectopic expression of COX-2 in hepatocytes constitutes a nonphysiological condition ideal for evaluating the role of prostaglandins (PGs) in liver pathogenesis. The effect of COX-2-dependent PGs in chronic liver disease, hepatitis, fibrosis, and chemical hepatocarcinogenesis, has been investigated in transgenic (Tg) mice that express human COX-2 in hepatocytes and in Tg hepatic human cell lines. We have used three different complementary approaches: i) diethylnitrosamine (DEN)-induced chemical hepatocarcinogenesis in COX-2 Tg mice, ii) DEN/phenobarbital treatment of human COX-2 Tg hepatocyte-like cells, and iii) COX-2 Tg hepatocyte-like cells implants in nude mice. The data suggest that PGs produced by COX-2 in hepatocytes promoted mild hepatitis in 60-week-old mice, as assessed by histological examination, but failed to contribute to the development of liver fibrogenesis after methionine- and choline-deficient diet treatment. Moreover, liver injury, collagen content, and hepatic stellate cell activation were equally severe in wild-type and COX-2 Tg mice. The contribution of COX-2-dependent PGs to the development of DEN-induced hepatocarcinogenesis was evaluated in Tg mice, Tg hepatocyte-like cells, and nude mice and the analysis revealed that COX-2 expression favors the development of preneoplastic foci without affecting malignant transformation. Endogenous COX-2 expression in wild-type mice is a late event in the development of hepatocellular carcinoma.


Subject(s)
Cyclooxygenase 2/metabolism , Hepatocytes/enzymology , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Precancerous Conditions/enzymology , Precancerous Conditions/pathology , Aging/pathology , Animals , Body Weight , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cyclin E/metabolism , Diethylnitrosamine , Gene Expression Regulation, Neoplastic , Hepatitis/complications , Hepatitis/pathology , Hepatocytes/pathology , Humans , Liver/metabolism , Liver/pathology , Liver Cirrhosis/complications , Liver Cirrhosis/enzymology , Liver Cirrhosis/pathology , Liver Neoplasms/genetics , Mice , Mice, Nude , Mice, Transgenic , Organ Size , Precancerous Conditions/chemically induced , Proto-Oncogene Proteins c-myc/metabolism , Transgenes/genetics , Xenograft Model Antitumor Assays
5.
Nat Commun ; 13(1): 5656, 2022 10 06.
Article in English | MEDLINE | ID: mdl-36202783

ABSTRACT

TRF1 is an essential component of the telomeric protective complex or shelterin. We previously showed that dysfunctional telomeres in alveolar type II (ATII) cells lead to interstitial lung fibrosis. Here, we study the lung pathologies upon telomere dysfunction in fibroblasts, club and basal cells. TRF1 deficiency in lung fibroblasts, club and basal cells induced telomeric damage, proliferative defects, cell cycle arrest and apoptosis. While Trf1 deletion in fibroblasts does not spontaneously lead to lung pathologies, upon bleomycin challenge exacerbates lung fibrosis. Unlike in females, Trf1 deletion in club and basal cells from male mice resulted in lung inflammation and airway remodeling. Here, we show that depletion of TRF1 in fibroblasts, Club and basal cells does not lead to interstitial lung fibrosis, underscoring ATII cells as the relevant cell type for the origin of interstitial fibrosis. Our findings contribute to a better understanding of proper telomere protection in lung tissue homeostasis.


Subject(s)
Pulmonary Fibrosis , Telomeric Repeat Binding Protein 1 , Animals , Bleomycin/toxicity , Female , Fibroblasts/metabolism , Male , Mice , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , Telomere/metabolism , Telomeric Repeat Binding Protein 1/genetics
6.
Cell Metab ; 3(3): 211-22, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16517408

ABSTRACT

Signaling cascades that control adipogenesis are essential in the regulation of body weight and obesity. The adaptor p62 controls pathways that modulate cell differentiation. We report here that p62(-/-) mice develop mature-onset obesity, leptin resistance, as well as impaired glucose and insulin intolerance. The metabolic rate was significantly reduced in p62(-/-) nonobese mice, which displayed increased mRNA levels of PPAR-gamma and reduced levels of UCP-1 in adipose tissue. Basal activity of ERK was enhanced in fat from nonobese mutant mice. Embryo fibroblasts from p62(-/-) mice differentiated better than the wild-type controls into adipocytes, which was abrogated by pharmacological inhibition of the ERK pathway. p62 is induced during adipocyte differentiation and inhibits ERK activation by direct interaction. We propose that p62 normally antagonizes basal ERK activity and adipocyte differentiation and that its loss leads to the hyperactivation of ERK that favors adipogenesis and obesity.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Insulin Resistance , Signal Transduction , Transcription Factors/deficiency , Transcription Factors/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Adipogenesis , Adipose Tissue/cytology , Animals , Embryo, Mammalian/metabolism , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Fibroblasts/metabolism , HeLa Cells , Humans , Mice , Mice, Knockout , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factor TFIIH , Transcription Factors/genetics
7.
Sci Rep ; 10(1): 572, 2020 01 17.
Article in English | MEDLINE | ID: mdl-31953445

ABSTRACT

Recent evidences indicate that mitochondrial genes and function are decreased in active ulcerative colitis (UC) patients, in particular, the activity of Complex I of the electron transport chain is heavily compromised. MCJ is a mitochondrial inner membrane protein identified as a natural inhibitor of respiratory chain Complex I. The induction of experimental colitis in MCJ-deficient mice leads to the upregulation of Timp3 expression resulting in the inhibition of TACE activity that likely inhibits Tnf and Tnfr1 shedding from the cell membrane in the colon. MCJ-deficient mice also show higher expression of Myd88 and Tlr9, proinflammatory genes and disease severity. Interestingly, the absence of MCJ resulted in distinct microbiota metabolism and composition, including a member of the gut community in UC patients, Ruminococcus gnavus. These changes provoked an effect on IgA levels. Gene expression analyses in UC patients showed decreased levels of MCJ and higher expression of TIMP3, suggesting a relevant role of mitochondrial genes and function among active UC. The MCJ deficiency disturbs the regulatory relationship between the host mitochondria and microbiota affecting disease severity. Our results indicate that mitochondria function may be an important factor in the pathogenesis. All together support the importance of MCJ regulation during UC.


Subject(s)
Bacteria/classification , Colitis, Ulcerative/genetics , Dysbiosis/genetics , HSP40 Heat-Shock Proteins/genetics , Mitochondrial Proteins/genetics , Molecular Chaperones/genetics , ADAM17 Protein/genetics , Animals , Bacteria/genetics , Bacteria/isolation & purification , Colitis, Ulcerative/microbiology , Disease Models, Animal , Gene Deletion , Gene Expression Regulation , Humans , Mice , Microbiota , Phylogeny , Receptors, Tumor Necrosis Factor, Type I/genetics , Severity of Illness Index , Tissue Inhibitor of Metalloproteinase-3/genetics , Tumor Necrosis Factor-alpha/genetics
8.
Biochem J ; 416(3): 337-46, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18671671

ABSTRACT

The effect of COX (cyclo-oxygenase)-2-dependent PGs (prostaglandins) in acute liver injury has been investigated in transgenic mice that express human COX-2 in hepatocytes. We have used three well-established models of liver injury: in LPS (lipopolysaccharide) injury in D-GalN (D-galactosamine)-preconditioned mice; in the hepatitis induced by ConA (concanavalin A); and in the proliferation of hepatocytes in regenerating liver after PH (partial hepatectomy). The results from the present study demonstrate that PG synthesis in hepatocytes decreases the susceptibility to LPS/D-GalN or ConA-induced liver injury as deduced by significantly lower levels of the pro-inflammatory profile and plasmatic aminotransferases in transgenic mice, an effect suppressed by COX-2-selective inhibitors. These Tg (transgenic) animals express higher levels of anti-apoptotic proteins and exhibit activation of proteins implicated in cell survival, such as Akt and AMP kinase after injury. The resistance to LPS/D-GalN-induced liver apoptosis involves an impairment of procaspase 3 and 8 activation. Protection against ConA-induced injury implies a significant reduction in necrosis. Moreover, hepatocyte commitment to start replication is anticipated in Tg mice after PH, due to the expression of PCNA (proliferating cell nuclear antigen), cyclin D1 and E. These results show, in a genetic model, that tissue-specific COX-2-dependent PGs exert an efficient protection against acute liver injury by an antiapoptotic/antinecrotic effect and by accelerated early hepatocyte proliferation.


Subject(s)
Cyclooxygenase 2/metabolism , Hepatocytes/enzymology , Liver/enzymology , Liver/pathology , Transgenes , Animals , Apoptosis/physiology , Biomarkers/blood , Caspases/metabolism , Cell Line , Cell Proliferation , Concanavalin A/pharmacology , Cyclooxygenase 2/genetics , Cytokines/metabolism , Galactosamine/pharmacology , Gene Expression , Gene Expression Profiling , Hepatectomy , Hepatocytes/cytology , Humans , Lipopolysaccharides/pharmacology , Liver/cytology , Liver/drug effects , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Prostaglandins/metabolism
9.
Elife ; 72018 01 30.
Article in English | MEDLINE | ID: mdl-29378675

ABSTRACT

Pulmonary fibrosis is a fatal lung disease characterized by fibrotic foci and inflammatory infiltrates. Short telomeres can impair tissue regeneration and are found both in hereditary and sporadic cases. We show here that telomerase expression using AAV9 vectors shows therapeutic effects in a mouse model of pulmonary fibrosis owing to a low-dose bleomycin insult and short telomeres. AAV9 preferentially targets regenerative alveolar type II cells (ATII). AAV9-Tert-treated mice show improved lung function and lower inflammation and fibrosis at 1-3 weeks after viral treatment, and improvement or disappearance of the fibrosis at 8 weeks after treatment. AAV9-Tert treatment leads to longer telomeres and increased proliferation of ATII cells, as well as lower DNA damage, apoptosis, and senescence. Transcriptome analysis of ATII cells confirms downregulation of fibrosis and inflammation pathways. We provide a proof-of-principle that telomerase activation may represent an effective treatment for pulmonary fibrosis provoked or associated with short telomeres.


Subject(s)
Genetic Therapy/methods , Pulmonary Fibrosis/therapy , Telomerase/pharmacology , Telomere/metabolism , Alveolar Epithelial Cells/physiology , Animals , Disease Models, Animal , Gene Expression Profiling , Lung/pathology , Lung/physiology , Mice , Pulmonary Fibrosis/pathology , Respiratory Function Tests , Telomerase/genetics , Therapeutic Uses
10.
J Exp Med ; 215(11): 2901-2918, 2018 11 05.
Article in English | MEDLINE | ID: mdl-30327417

ABSTRACT

MHCII in antigen-presenting cells (APCs) is a key regulator of adaptive immune responses. Expression of MHCII genes is controlled by the transcription coactivator CIITA, itself regulated through cell type-specific promoters. Here we show that the transcription factor NFAT5 is needed for expression of Ciita and MHCII in macrophages, but not in dendritic cells and other APCs. NFAT5-deficient macrophages showed defective activation of MHCII-dependent responses in CD4+ T lymphocytes and attenuated capacity to elicit graft rejection in vivo. Ultrasequencing analysis of NFAT5-immunoprecipitated chromatin uncovered an NFAT5-regulated region distally upstream of Ciita This region was required for CIITA and hence MHCII expression, exhibited NFAT5-dependent characteristics of active enhancers such as H3K27 acetylation marks, and required NFAT5 to interact with Ciita myeloid promoter I. Our results uncover an NFAT5-regulated mechanism that maintains CIITA and MHCII expression in macrophages and thus modulates their T lymphocyte priming capacity.


Subject(s)
Enhancer Elements, Genetic/immunology , Gene Expression Regulation/immunology , Histocompatibility Antigens Class II/immunology , Macrophages/immunology , Nuclear Proteins/immunology , Trans-Activators/immunology , Transcription Factors/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Gene Rearrangement/immunology , Histocompatibility Antigens Class II/genetics , Macrophages/cytology , Mice , Mice, Knockout , Nuclear Proteins/genetics , Trans-Activators/genetics , Transcription Factors/genetics
11.
Sci Rep ; 7(1): 10740, 2017 09 06.
Article in English | MEDLINE | ID: mdl-28878331

ABSTRACT

Salp15, a salivary protein of Ixodes ticks, inhibits the activation of naïve CD4 T cells. Treatment with Salp15 results in the inhibition of early signaling events and the production of the autocrine growth factor, interleukin-2. The fate of the CD4 T cells activated in the presence of Salp15 or its long-term effects are, however, unknown. We now show that Salp15 binding to CD4 is persistent and induces a long-lasting immunomodulatory effect. The activity of Salp15 results in sustained diminished cross-antigenic antibody production even after interruption of the treatment with the protein. Transcriptionally, the salivary protein provokes an acute effect that includes known activation markers, such as Il2 or Cd44, and that fades over time. The long-term effects exerted by Salp15 do not involve the induction of either anergy traits nor increased populations of regulatory T cells. Similarly, the treatment with Salp15 does not result in B cell anergy or the generation of myeloid suppressor cells. However, Salp15 induces the increased expression of the ectoenzyme, CD73, in regulatory T cells and increased production of adenosine. Our study provides a profound characterization of the immunomodulatory activity of Salp15 and suggests that its long-term effects are due to the specific regulation of CD73.


Subject(s)
Immune Tolerance/drug effects , Immunomodulation/drug effects , Immunosuppressive Agents/pharmacology , Salivary Proteins and Peptides/pharmacology , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Graft vs Host Disease/drug therapy , Graft vs Host Disease/etiology , Hematopoiesis/drug effects , Hematopoiesis/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Immunoglobulin G/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transcription, Genetic
12.
J Med Chem ; 58(5): 2256-64, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-25671648

ABSTRACT

Triple-negative breast cancer (TNBC) represents a subtype of breast cancer characterized by high aggressiveness. There is no current targeted therapy for these patients whose prognosis, as a group, is very poor. Here, we report the synthesis and evaluation of a potent antitumor agent in vivo for this type of breast cancer designed as a combination of quinone/cannabinoid pharmacophores. This new compound (10) has been selected from a series of chromenopyrazolediones with full selectivity for the nonpsychotropic CB2 cannabinoid receptor and with efficacy in inducing death of human TNBC cell lines. The dual concept quinone/cannabinoid was supported by the fact that compound 10 exerts antitumor effect by inducing cell apoptosis through activation of CB2 receptors and through oxidative stress. Notably, it did not show either cytotoxicity on noncancerous human mammary epithelial cells nor toxic effects in vivo, suggesting that it may be a new therapeutic tool for the management of TNBC.


Subject(s)
Antineoplastic Agents/pharmacology , Benzopyrans/pharmacology , Benzoquinones/chemistry , Breast/drug effects , Cannabinoids/chemistry , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB2/metabolism , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Benzopyrans/chemistry , Blotting, Western , Breast/cytology , Cell Proliferation/drug effects , Cells, Cultured , Female , Humans , Mice , Mice, Nude , Pyrazoles/chemistry , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptor, Cannabinoid, CB2/genetics , Reverse Transcriptase Polymerase Chain Reaction , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
13.
PLoS One ; 8(3): e58751, 2013.
Article in English | MEDLINE | ID: mdl-23516551

ABSTRACT

In humans and most mammals, differentiation of the embryonic gonad into ovaries or testes is controlled by the Y-linked gene SRY. Here we show a role for the Gadd45g protein in this primary sex differentiation. We characterized mice deficient in Gadd45a, Gadd45b and Gadd45g, as well as double-knockout mice for Gadd45ab, Gadd45ag and Gadd45bg, and found a specific role for Gadd45g in male fertility and testis development. Gadd45g-deficient XY mice on a mixed 129/C57BL/6 background showed varying degrees of disorders of sexual development (DSD), ranging from male infertility to an intersex phenotype or complete gonadal dysgenesis (CGD). On a pure C57BL/6 (B6) background, all Gadd45g(-/-) XY mice were born as completely sex-reversed XY-females, whereas lack of Gadd45a and/or Gadd45b did not affect primary sex determination or testis development. Gadd45g expression was similar in female and male embryonic gonads, and peaked around the time of sex differentiation at 11.5 days post-coitum (dpc). The molecular cause of the sex reversal was the failure of Gadd45g(-/-) XY gonads to achieve the SRY expression threshold necessary for testes differentiation, resulting in ovary and Müllerian duct development. These results identify Gadd45g as a candidate gene for male infertility and 46,XY sex reversal in humans.


Subject(s)
Fertility , Intracellular Signaling Peptides and Proteins/metabolism , Sex Determination Processes , Testis/growth & development , Animals , Cell Differentiation , Female , Gene Expression Regulation, Developmental , Gonadal Dysgenesis, 46,XY/genetics , Humans , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Phenotype , Pregnancy , Sertoli Cells/cytology , Sex-Determining Region Y Protein/genetics , Testis/cytology , Testis/metabolism , GADD45 Proteins
14.
PLoS One ; 8(6): e66933, 2013.
Article in English | MEDLINE | ID: mdl-23825589

ABSTRACT

Aberrant activation of MAP kinase signaling pathway and loss of tumor suppressor LKB1 have been implicated in lung cancer development and progression. Although oncogenic KRAS mutations are frequent, BRAF mutations (BRAF(V600E)) are found in 3% of human non-small cell lung cancers. Contrary to KRAS mutant tumors, BRAF(V600E)-induced tumors are benign adenomas that fail to progess. Interestingly, loss of tumor supressor LKB1 coexists with KRAS oncogenic mutations and synergizes in tumor formation and progression, however, its cooperation with BRAF(V600E) oncogene is unknown. Our results describe a lung cell population in neonates mice where expression of BRAF(V600E) leads to lung adenoma development. Importantly, expression of BRAF(V600E) concomitant with the loss of only a single-copy of Lkb1, overcomes senencence-like features of BRAF(V600E)-mutant adenomas leading malignization to carcinomas. These results posit LKB1 haploinsufficiency as a risk factor for tumor progression of BRAF(V600E) mutated lung adenomas in human cancer patients.


Subject(s)
Adenoma/pathology , Disease Progression , Gene Deletion , Lung Neoplasms/pathology , Mutation , Protein Serine-Threonine Kinases/deficiency , Proto-Oncogene Proteins B-raf/genetics , AMP-Activated Protein Kinases , Adenoma/enzymology , Adenoma/genetics , Animals , Animals, Newborn , Cadherins/metabolism , Carcinogenesis/drug effects , Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Mice , Mice, Transgenic , Oncogenes/genetics , Protein Serine-Threonine Kinases/genetics , Pulmonary Surfactant-Associated Protein C/metabolism , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Tumor Suppressor Protein p53/metabolism
15.
Fertil Steril ; 93(4): 1308-15, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-19446805

ABSTRACT

OBJECTIVE: To determine, in a sheep model, the effect of a short-term progestative treatment on growth dynamics and functionality of induced corpora lutea. DESIGN: Observational, model study. SETTING: Public university. PATIENT(S): Sixty adult female sheep. INTERVENTION(S): Synchronization and induction of ovulation with progestogens and prostaglandin analogues; ovarian ultrasonography, blood sampling, and ovariectomy. MAIN OUTCOME MEASURE(S): Determination of pituitary function and morphologic characteristics, expression of luteinizing hormone (LH) receptors, and progesterone secretion of corpora lutea. RESULT(S): The use of progestative pretreatments for assisted conception affect the growth patterns, the expression of LH receptors, and the progesterone secretion of induced corpora lutea. CONCLUSION(S): The current study indicates, in a sheep model, the existence of deleterious effects from progestogens on functionality of induced corpora lutea.


Subject(s)
Corpus Luteum/growth & development , Corpus Luteum/metabolism , Flurogestone Acetate/administration & dosage , Menstrual Cycle/physiology , Ovulation Induction , Progesterone/metabolism , Progestins/administration & dosage , Receptors, LH/biosynthesis , Animals , Corpus Luteum/drug effects , Female , Menstrual Cycle/drug effects , Models, Animal , Ovulation/drug effects , Ovulation/physiology , Ovulation Induction/methods , Sheep
18.
Blood ; 107(3): 1174-7, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16239434

ABSTRACT

Gene silencing by CpG island promoter hypermethylation has awakened the interest for DNA demethylating agents as chemotherapy drugs. Zebularine (1-[beta-D-ribofuranosil]-1,2-dihydropyrimidin-2-1) has been recently described as a new DNA methylation inhibitor. Here we have studied its effects in a mouse model of radiation-induced lymphomagenesis using nuclear magnetic resonance (NMR) and positron emission tomography (PET). All control animals presented large thymic T lymphomas and died between 4 and 5.5 months. In contrast, 40% (12 of 30) of zebularine-treated animals were still alive after 1 year (Kaplan-Meier P < .001). NMR and PET imaging showed that surviving animals presented a thymus structure/volume similar to normal mice of the same age. Most important, zebularine demonstrated a complete lack of toxicity in nonirradiated control mice. DNA hypomethylation induced by zebularine occurred in association with depletion in extractable DNA methyltransferase 1 protein. Thus, our data support the role of zebularine as a DNA demethylating agent with antitumor activity and little toxicity.


Subject(s)
Antineoplastic Agents/administration & dosage , Cytidine/analogs & derivatives , DNA Methylation/drug effects , Gene Silencing/drug effects , Lymphoma, T-Cell/drug therapy , Animals , Antineoplastic Agents/adverse effects , Cell Transformation, Neoplastic/drug effects , Cytidine/administration & dosage , Cytidine/adverse effects , Drug Evaluation, Preclinical , Injections, Intraperitoneal , Lymphoma, T-Cell/pathology , Mice , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology
19.
EMBO J ; 23(23): 4595-605, 2004 Nov 24.
Article in English | MEDLINE | ID: mdl-15526032

ABSTRACT

PKCzeta is required for nuclear factor kappa-B (NF-kappaB) activation in several cell systems. NF-kappaB is a suppressor of liver apoptosis during development and in concanavalin A (ConA)-induced T-cell-mediated hepatitis. Here we show that PKCzeta-/- mice display inhibited ConA-induced NF-kappaB activation and reduced damage in liver. As the IL-4/Stat6 pathway is necessary for ConA-induced hepatitis, we addressed here the potential role of PKCzeta in this cascade. Interestingly, the loss of PKCzeta severely attenuated serum IL-5 and liver eotaxin-1 levels, two critical mediators of liver damage. Stat6 tyrosine phosphorylation and Jak1 activation were ablated in the liver of ConA-injected PKCzeta-/- mice and in IL-4-stimulated PKCzeta-/- fibroblasts. PKCzeta interacts with and phosphorylates Jak1 and PKCzeta activity is required for Jak1 function. In contrast, Par-4-/- mice have increased sensitivity to ConA-induced liver damage and IL-4 signaling. This unveils a novel and critical involvement of PKCzeta in the IL-4/Stat6 signaling pathway in vitro and in vivo.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Interleukin-4/physiology , Liver/metabolism , Protein Kinase C/physiology , T-Lymphocytes/immunology , Trans-Activators/physiology , Animals , Cells, Cultured , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/immunology , Chemokine CCL11 , Chemokines, CC/metabolism , Concanavalin A , Enzyme Activation , Fibroblasts/metabolism , Interleukin-5/blood , Janus Kinase 1 , Liver/drug effects , Liver/pathology , Mice , Mice, Knockout , NF-kappa B/metabolism , Phosphorylation , Protein Kinase C/genetics , Protein-Tyrosine Kinases/metabolism , Receptors, Thrombin/genetics , Receptors, Thrombin/metabolism , STAT6 Transcription Factor , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL