Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Int J Mol Sci ; 24(16)2023 Aug 21.
Article in English | MEDLINE | ID: mdl-37629186

ABSTRACT

Cancer cachexia is a multifactorial syndrome that interferes with treatment and reduces the quality of life and survival of patients. Currently, there is no effective treatment or biomarkers, and pathophysiology is not clear. Our group reported alterations on tryptophan metabolites in cachectic patients, so we aim to investigate the role of tryptophan using two cancer-associated cachexia syngeneic murine models, melanoma B16F10, and pancreatic adenocarcinoma that is KPC-based. Injected mice showed signs of cancer-associated cachexia as reduction in body weight and raised spleen weight, MCP1, and carbonilated proteins in plasma. CRP and Myostatin also increased in B16F10 mice. Skeletal muscle showed a decrease in quadriceps weight and cross-sectional area (especially in B16F10). Higher expression of atrophy genes, mainly Atrogin1, was also observed. Plasmatic tryptophan levels in B16F10 tumor-bearing mice decreased even at early steps of tumorigenesis. In KPC-injected mice, tryptophan fluctuated but were also reduced and in cachectic patients were significantly lower. Treatment with 1-methyl-tryptophan, an inhibitor of tryptophan degradation, in the murine models resulted in the restoration of plasmatic tryptophan levels and an improvement on splenomegaly and carbonilated proteins levels, while changes in plasmatic inflammatory markers were mild. After the treatment, CCR2 expression in monocytes diminished and lymphocytes, Tregs, and CD8+, were activated (seen by increased in CD127 and CD25 expression, respectively). These immune cell changes pointed to an improvement in systemic inflammation. While treatment with 1-MT did not show benefits in terms of muscle wasting and atrophy in our experimental setting, muscle functionality was not affected and central nuclei fibers appeared, being a feature of regeneration. Therefore, tryptophan metabolism pathway is a promising target for inflammation modulation in cancer-associated cachexia.


Subject(s)
Adenocarcinoma , Pancreatic Neoplasms , Animals , Mice , Cachexia/etiology , Quality of Life , Tryptophan , Muscular Atrophy/etiology , Inflammation
2.
Biomater Adv ; 150: 213426, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37104961

ABSTRACT

Acquired muscle diseases such as cancer cachexia are responsible for the poor prognosis of many patients suffering from cancer. In vitro models are needed to study the underlying mechanisms of those pathologies. Extrusion bioprinting is an emerging tool to emulate the aligned architecture of fibers while implementing additive manufacturing techniques in tissue engineering. However, designing bioinks that reconcile the rheological needs of bioprinting and the biological requirements of muscle tissue is a challenging matter. Here we formulate a biomaterial with dual crosslinking to modulate the physical properties of bioprinted models. We design 3D bioprinted muscle models that resemble the mechanical properties of native tissue and show improved proliferation and high maturation of differentiated myotubes suggesting that the GelMA-AlgMA-Fibrin biomaterial possesses myogenic properties. The electrical stimulation of the 3D model confirmed the contractile capability of the tissue and enhanced the formation of sarcomeres. Regarding the functionality of the models, they served as platforms to recapitulate skeletal muscle diseases such as muscle wasting produced by cancer cachexia. The genetic expression of 3D models demonstrated a better resemblance to the muscular biopsies of cachectic mouse models. Altogether, this biomaterial is aimed to fabricate manipulable skeletal muscle in vitro models in a non-costly, fast and feasible manner.


Subject(s)
Cachexia , Neoplasms , Mice , Animals , Cachexia/etiology , Cachexia/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy/etiology , Muscular Atrophy/metabolism , Neoplasms/complications , Neoplasms/metabolism , Biocompatible Materials
3.
J Pathol ; 253(2): 174-185, 2021 02.
Article in English | MEDLINE | ID: mdl-33079429

ABSTRACT

Cystic neoplasms of the pancreas are an increasingly important public health problem. The majority of these lesions are benign but some progress to invasive pancreatic ductal adenocarcinoma (PDAC). There is a dearth of mouse models of these conditions. The orphan nuclear receptor NR5A2 regulates development, differentiation, and inflammation. Germline Nr5a2 heterozygosity sensitizes mice to the oncogenic effects of mutant Kras in the pancreas. Here, we show that - unlike constitutive Nr5a2+/- mice - conditional Nr5a2 heterozygosity in pancreatic epithelial cells, combined with mutant Kras (KPN+/- ), leads to a dramatic replacement of the pancreatic parenchyma with cystic structures and an accelerated development of high-grade PanINs and PDAC. Timed histopathological analyses indicated that in KPN+/- mice PanINs precede the formation of cystic lesions and the latter precede PDAC. A single episode of acute caerulein pancreatitis is sufficient to accelerate the development of cystic lesions in KPN+/- mice. Epithelial cells of cystic lesions of KPN+/- mice express MUC1, MUC5AC, and MUC6, but lack expression of MUC2, CDX2, and acinar markers, indicative of a pancreato-biliary/gastric phenotype. In accordance with this, in human samples we found a non-significantly decreased expression of NR5A2 in mucinous tumours, compared with conventional PDAC. These results highlight that the effects of loss of one Nr5a2 allele are time- and cell context-dependent. KPN+/- mice represent a new model to study the formation of cystic pancreatic lesions and their relationship with PanINs and classical PDAC. Our findings suggest that pancreatitis could also contribute to acceleration of cystic tumour progression in patients. © 2020 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/pathology , Receptors, Cytoplasmic and Nuclear/metabolism , Adult , Aged , Aged, 80 and over , Animals , Carcinoma, Pancreatic Ductal/genetics , Disease Progression , Epithelial Cells/pathology , Female , Heterozygote , Humans , Male , Mice , Middle Aged , Pancreatic Cyst/pathology , Pancreatic Ducts/pathology , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Receptors, Cytoplasmic and Nuclear/genetics
4.
Nat Cell Biol ; 22(11): 1307-1318, 2020 11.
Article in English | MEDLINE | ID: mdl-33106654

ABSTRACT

Tissue regeneration declines with ageing but little is known about whether this arises from changes in stem-cell heterogeneity. Here, in homeostatic skeletal muscle, we identify two quiescent stem-cell states distinguished by relative CD34 expression: CD34High, with stemness properties (genuine state), and CD34Low, committed to myogenic differentiation (primed state). The genuine-quiescent state is unexpectedly preserved into later life, succumbing only in extreme old age due to the acquisition of primed-state traits. Niche-derived IGF1-dependent Akt activation debilitates the genuine stem-cell state by imposing primed-state features via FoxO inhibition. Interventions to neutralize Akt and promote FoxO activity drive a primed-to-genuine state conversion, whereas FoxO inactivation deteriorates the genuine state at a young age, causing regenerative failure of muscle, as occurs in geriatric mice. These findings reveal transcriptional determinants of stem-cell heterogeneity that resist ageing more than previously anticipated and are only lost in extreme old age, with implications for the repair of geriatric muscle.


Subject(s)
Antigens, CD34/metabolism , Cell Proliferation , Cell Self Renewal , Cellular Senescence , Forkhead Transcription Factors/metabolism , Muscle, Skeletal/metabolism , Regeneration , Satellite Cells, Skeletal Muscle/metabolism , Age Factors , Animals , Cardiotoxins/toxicity , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Self Renewal/drug effects , Cell Self Renewal/genetics , Cells, Cultured , Cellular Senescence/drug effects , Cellular Senescence/genetics , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O3/genetics , Forkhead Box Protein O3/metabolism , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Muscle, Skeletal/transplantation , Phenotype , Proto-Oncogene Proteins c-akt/metabolism , Regeneration/drug effects , Regeneration/genetics , Satellite Cells, Skeletal Muscle/drug effects , Satellite Cells, Skeletal Muscle/pathology , Satellite Cells, Skeletal Muscle/transplantation , Signal Transduction , Stem Cell Niche
5.
Nature ; 554(7693): 533-537, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29443959

ABSTRACT

Chronic inflammation increases the risk of developing one of several types of cancer. Inflammatory responses are currently thought to be controlled by mechanisms that rely on transcriptional networks that are distinct from those involved in cell differentiation. The orphan nuclear receptor NR5A2 participates in a wide variety of processes, including cholesterol and glucose metabolism in the liver, resolution of endoplasmic reticulum stress, intestinal glucocorticoid production, pancreatic development and acinar differentiation. In genome-wide association studies, single nucleotide polymorphisms in the vicinity of NR5A2 have previously been associated with the risk of pancreatic adenocarcinoma. In mice, Nr5a2 heterozygosity sensitizes the pancreas to damage, impairs regeneration and cooperates with mutant Kras in tumour progression. Here, using a global transcriptomic analysis, we describe an epithelial-cell-autonomous basal pre-inflammatory state in the pancreas of Nr5a2+/- mice that is reminiscent of the early stages of pancreatitis-induced inflammation and is conserved in histologically normal human pancreases with reduced expression of NR5A2 mRNA. In Nr5a2+/-mice, NR5A2 undergoes a marked transcriptional switch, relocating from differentiation-specific to inflammatory genes and thereby promoting gene transcription that is dependent on the AP-1 transcription factor. Pancreatic deletion of Jun rescues the pre-inflammatory phenotype, as well as binding of NR5A2 to inflammatory gene promoters and the defective regenerative response to damage. These findings support the notion that, in the pancreas, the transcriptional networks involved in differentiation-specific functions also suppress inflammatory programmes. Under conditions of genetic or environmental constraint, these networks can be subverted to foster inflammation.


Subject(s)
Cell Differentiation/genetics , Gene Expression Regulation , Inflammation/genetics , Pancreas/metabolism , Pancreas/pathology , Receptors, Cytoplasmic and Nuclear/metabolism , Transcriptome , Acinar Cells/metabolism , Acinar Cells/pathology , Animals , Chromatin/genetics , Chromatin/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Regulatory Networks/genetics , Genes, jun/genetics , Heterozygote , Humans , Mice , Organ Specificity/genetics , Pancreatitis/genetics , Promoter Regions, Genetic/genetics , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factor AP-1/metabolism
6.
Mediators Inflamm ; 2015: 942517, 2015.
Article in English | MEDLINE | ID: mdl-26441484

ABSTRACT

We show that galactomannan, a polysaccharide consisting of a mannose backbone with galactose side groups present on the cell wall of several fungi, induces a reprogramming of the inflammatory response in human macrophages through dectin-1 receptor. The nuclear factor kappa-light-chain-enhancer of activated B cells 2 (NFκB2)/p100 was overexpressed after galactomannan challenge. Knocking down NFκB2/p100 using small interfering RNA (siRNA) indicated that NFκB2/p100 expression is a crucial factor in the progression of the galactomannan-induced refractoriness. The data presented in this study could be used as a modulator of inflammatory response in clinical situations where refractory state is required.


Subject(s)
Inflammation/drug therapy , Macrophages/drug effects , Macrophages/immunology , Mannans/therapeutic use , NF-kappa B p52 Subunit/metabolism , Blotting, Western , Cell Proliferation/drug effects , Cells, Cultured , Flow Cytometry , Galactose/analogs & derivatives , Humans , Inflammation/chemically induced , Lipopolysaccharides/pharmacology , RNA, Small Interfering
7.
Carcinogenesis ; 35(12): 2670-8, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25233928

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is driven by the accumulation of somatic mutations, epigenetic modifications and changes in the micro-environment. New approaches to investigating disruptions of gene expression networks promise to uncover key regulators and pathways in carcinogenesis. We performed messenger RNA-sequencing in pancreatic normal (n = 10) and tumor (n = 8) derived tissue samples, as well as in pancreatic cancer cell lines (n = 9), to determine differential gene expression (DE) patterns. Sub-network enrichment analyses identified HNF1A as the regulator of the most significantly and consistently dysregulated expression sub-network in pancreatic tumor tissues and cells (median P = 7.56×10(-7), median rank = 1, range = 1-25). To explore the effects of HNF1A expression in pancreatic tumor-derived cells, we generated stable HNF1A-inducible clones in two pancreatic cancer cell lines (PANC-1 and MIA PaCa-2) and observed growth inhibition (5.3-fold, P = 4.5×10(-5) for MIA PaCa-2 clones; 7.2-fold, P = 2.2×10(-5) for PANC-1 clones), and a G0/G1 cell cycle arrest and apoptosis upon induction. These effects correlated with HNF1A-induced down-regulation of 51 of 84 cell cycle genes (e.g. E2F1, CDK2, CDK4, MCM2/3/4/5, SKP2 and CCND1), decreased expression of anti-apoptotic genes (e.g. BIRC2/5/6 and AKT) and increased expression of pro-apoptotic genes (e.g. CASP4/9/10 and APAF1). In light of the established role of HNF1A in the regulation of pancreatic development and homeostasis, our data suggest that it also functions as an important tumor suppressor in the pancreas.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Pancreatic Ductal/genetics , Gene Expression Profiling , Genes, Tumor Suppressor , Hepatocyte Nuclear Factor 1-alpha/genetics , Pancreatic Neoplasms/genetics , Apoptosis , Biomarkers, Tumor/metabolism , Blotting, Western , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Cycle , Cell Proliferation , Cells, Cultured , Flow Cytometry , Gene Regulatory Networks , Hepatocyte Nuclear Factor 1-alpha/metabolism , Humans , Immunoenzyme Techniques , Pancreas/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
8.
Gut ; 63(4): 647-55, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23598351

ABSTRACT

OBJECTIVES: Nr5a2 participates in biliary acid metabolism and is a major regulator of the pancreatic exocrine programme. Single nucleotide polymorphisms in the vicinity of NR5A2 are associated with the risk of pancreatic ductal adenocarcinoma (PDAC). AIMS: To determine the role of Nr5a2 in pancreatic homeostasis, damage-induced regeneration and mutant KRas-driven pancreatic tumourigenesis. DESIGN: Nr5a2+/- and KRas(G12V);Ptf1a-Cre;Nr5a2+/- mice were used to investigate whether a full dose of Nr5a2 is required for normal pancreas development, recovery from caerulein-induced pancreatitis, and protection from tumour development. RESULTS: Adult Nr5a2+/- mice did not display histological abnormalities in the pancreas but showed a more severe acute pancreatitis, increased acino-ductal metaplasia and impaired recovery from damage. This was accompanied by increased myeloid cell infiltration and proinflammatory cytokine gene expression, and hyperactivation of nuclear factor κb and signal transducer and activator of transcription 3 signalling pathways. Induction of multiple episodes of acute pancreatitis was associated with more severe damage and delayed regeneration. Inactivation of one Nr5a2 allele selectively in pancreatic epithelial cells was sufficient to cause impaired recovery from pancreatitis. In comparison with Nr5a2+/+ mice, KRas(G12V);Ptf1a(Cre/+);Nr5a2+/- mice showed a non-statistically significant increase in the area affected by preneoplastic lesions. However, a single episode of acute pancreatitis cooperated with loss of one Nr5a2 allele to accelerate KRas(G12V)-driven development of preneoplastic lesions. CONCLUSIONS: A full Nr5a2 dose is required to restore pancreatic homeostasis upon damage and to suppress the KRas(G12V)-driven mouse pancreatic intraepithelial neoplasia progression, indicating that Nr5a2 is a novel pancreatic tumour suppressor. Nr5a2 could contribute to PDAC through a role in the recovery from pancreatitis-induced damage.


Subject(s)
Pancreatic Neoplasms/genetics , Pancreatitis/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Animals , Blotting, Western , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/physiopathology , Ceruletide/pharmacology , Heterozygote , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , NF-kappa B/physiology , Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/physiopathology , Pancreatitis/chemically induced , Pancreatitis/physiopathology , Polymerase Chain Reaction , Proto-Oncogene Proteins p21(ras)/physiology , Receptors, Cytoplasmic and Nuclear/physiology , STAT3 Transcription Factor/physiology , Signal Transduction/physiology
9.
Eur Urol ; 65(2): 360-6, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24018021

ABSTRACT

BACKGROUND: Hotspot mutations in the promoter of the gene coding for telomerase reverse transcriptase (TERT) have been described and proposed to activate gene expression. OBJECTIVES: To investigate TERT mutation frequency, spectrum, association with expression and clinical outcome, and potential for detection of recurrences in urine in patients with urothelial bladder cancer (UBC). DESIGN, SETTING, AND PARTICIPANTS: A set of 111 UBCs of different stages was used to assess TERT promoter mutations by Sanger sequencing and TERT messenger RNA (mRNA) expression by reverse transcription-quantitative polymerase chain reaction. The two most frequent mutations were investigated, using a SNaPshot assay, in an independent set of 184 non-muscle-invasive and 173 muscle-invasive UBC (median follow-up: 53 mo and 21 mo, respectively). Voided urine from patients with suspicion of incident UBC (n=174), or under surveillance after diagnosis of non-muscle-invasive UBC (n=194), was tested using a SNaPshot assay. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: Association of mutation status with age, sex, tobacco, stage, grade, fibroblast growth factor receptor 3 (FGFR3) mutation, progression-free survival, disease-specific survival, and overall survival. RESULTS AND LIMITATIONS: In the two series, 78 of 111 (70%) and 283 of 357 (79%) tumors harbored TERT mutations, C228T being the most frequent substitution (83% for both series). TERT mutations were not associated with clinical or pathologic parameters, but were more frequent among FGFR3 mutant tumors (p=0.0002). There was no association between TERT mutations and mRNA expression (p=0.3). Mutations were not associated with clinical outcome. In urine, TERT mutations had 90% specificity in subjects with hematuria but no bladder tumor, and 73% in recurrence-free UBC patients. The sensitivity was 62% in incident and 42% in recurrent UBC. A limitation of the study is its retrospective nature. CONCLUSIONS: Somatic TERT promoter mutations are an early, highly prevalent genetic event in UBC and are not associated with TERT mRNA levels or disease outcomes. A SNaPshot assay in urine may help to detect UBC recurrences.


Subject(s)
Biomarkers, Tumor/genetics , Mutation , Promoter Regions, Genetic , Telomerase/genetics , Urinary Bladder Neoplasms/enzymology , Urinary Bladder Neoplasms/genetics , Aged , Biomarkers, Tumor/urine , Cell Line, Tumor , DNA Mutational Analysis , Disease Progression , Disease-Free Survival , Female , Genetic Predisposition to Disease , Genetic Testing/methods , Humans , Male , Neoplasm Grading , Neoplasm Recurrence, Local , Neoplasm Staging , Netherlands , Phenotype , Predictive Value of Tests , RNA, Messenger/urine , Retrospective Studies , Reverse Transcriptase Polymerase Chain Reaction , Risk Factors , Spain , Telomerase/urine , Time Factors , Urinary Bladder Neoplasms/mortality , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/therapy , Urinary Bladder Neoplasms/urine
10.
Gut ; 61(8): 1187-96, 2012 Aug.
Article in English | MEDLINE | ID: mdl-21948943

ABSTRACT

OBJECTIVES: During pancreatitis, specific transcriptional programmes govern functional regeneration after injury. The objective of this study was to analyse the dynamic regulation of pancreatic genes and the role of transcriptional regulators during recovery from pancreatitis. DESIGN: Wild-type and genetically modified mice (Hnf1α(-/-) and Ptf1a(+/-)) were used. After caerulein or L-arginine induced pancreatitis, blood or pancreata were processed for enzymatic assays, ELISA, histology, immunohistochemistry, western blotting and quantitative reverse transcriptase-PCR. Nr5a2 promoter reporter and chromatin immunoprecipitation assays for Hnf1α were also performed. RESULTS: After caerulein pancreatic injury, expression of acinar and endocrine genes rapidly decreased, but eventually recovered, depicting distinct cell-type-specific patterns. Pdx1 and Hnf1α mRNAs underwent marked downregulation, matching endocrine/exocrine gene expression profiles. Ptf1a, Pdx1 and Hnf1α protein levels were also reduced and recovered gradually. These changes were associated with transient impairment of exocrine and endocrine function, including abnormal glucose tolerance. On l-arginine pancreatitis, changes in Ptf1a, Pdx1 and Hnf1α gene and protein expression were recapitulated. Reduced Hnf1α and Ptf1a levels after pancreatitis coincided with increased acinar cell proliferation, both in Hnf1α(-/-) and Ptf1a(+/-) mice. Moreover, Hnf1α(-/-) mice had reduced Ptf1a protein as well as transcripts for Ptf1a and digestive enzymes. Dispersed acini from Hnf1α(-/-) mice showed suboptimal secretory responses to caerulein. Bioinformatics analysis did not support a role for Hnf1α as a direct regulator of digestive enzyme genes. Instead, it was found that Hnf1α binds to, and regulates, the promoter of Nr5a2, coding an orphan nuclear receptor that regulates acinar gene expression. CONCLUSIONS: Dynamic changes in gene expression occur on pancreatitis induction, determining altered exocrine and endocrine function. This analysis uncovers roles for Hnf1α in the regulation of acinar cell determination and function. This effect may be mediated, in part, through direct regulation of Nr5a2.


Subject(s)
Acinar Cells/metabolism , Gene Expression Regulation , Hepatocyte Nuclear Factor 1-alpha/genetics , Homeostasis/genetics , Pancreatitis/genetics , RNA, Messenger/genetics , Acinar Cells/pathology , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Hepatocyte Nuclear Factor 1-alpha/biosynthesis , Immunohistochemistry , Immunoprecipitation , Male , Mice , Mice, Inbred C57BL , Pancreatitis/metabolism , Pancreatitis/pathology , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transcription Factors/metabolism
11.
Mol Genet Genomics ; 281(3): 345-59, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19123063

ABSTRACT

The activity of protein phosphatases on mitogen-activated protein kinases (MAPKS) is essential in the modulation of the final outcome of MAPK-signalling pathways. The yeast dual-specificity phosphatase (DSP) Msg5, expressed as two isoforms of different length, dephosphorylates the MAPKs of mating and cell integrity pathways, Fus3 and Slt2, respectively, but its action on the MAPK Kss1 is unclear. Here we analyse the global impact of Msg5 on the yeast transcriptome. Both Fus3- and Slt2- but not Kss1-mediated gene expression is induced in cells lacking Msg5. However, although these cells show high Slt2 phosphorylation, the Rlm1-dependent Slt2-regulated transcriptional response is weak. Therefore, mechanisms concomitant with Slt2 phosphorylation are required for a strong Rlm1 activation. The limited Slt2 activity on Rlm1 is not a specific effect on this substrate but a consequence of its low kinase activity in msg5Delta cells. Lack of Msg5 does not increase Kss1 phosphorylation although both proteins physically interact. Both Msg5 isoforms interact similarly with Slt2, whereas the long form binds Fus3 with higher affinity and consequently down-regulates it more efficiently than the short one. We propose that specific binding of DSP isoforms to distinct MAPKs provides a novel mechanism for fine tuning different pathways by the same phosphatase.


Subject(s)
Dual-Specificity Phosphatases/metabolism , MAP Kinase Signaling System , Protein Tyrosine Phosphatases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Base Sequence , DNA Primers/genetics , DNA, Fungal/genetics , DNA-Binding Proteins/metabolism , Dual-Specificity Phosphatases/genetics , Gene Deletion , Gene Expression Profiling , Genes, Fungal , Isoenzymes/genetics , Isoenzymes/metabolism , Mitogen-Activated Protein Kinases/metabolism , Mutation , Oligonucleotide Array Sequence Analysis , Phosphorylation , Protein Tyrosine Phosphatases/genetics , Repressor Proteins , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
12.
J Proteomics ; 71(5): 530-46, 2008 Dec 02.
Article in English | MEDLINE | ID: mdl-18824147

ABSTRACT

Intestinal epithelial cells undergo progressive cell maturation as they migrate along the crypt-villus axis. To determine molecular signatures that define this process, proteins differentially expressed between the crypt and villus were identified by 2D-DIGE and MALDI-MS. Forty-six differentially expressed proteins were identified, several of which were validated by immunohistochemistry. Proteins upregulated in the villus were enriched for those involved in brush border assembly and lipid uptake, established features of differentiated intestinal epithelial cells. Multiple proteins involved in glycolysis were also upregulated in the villus, suggesting increased glycolysis is a feature of intestinal cell differentiation. Conversely, proteins involved in nucleotide metabolism, and protein processing and folding were increased in the crypt, consistent with functions associated with cell proliferation. Three novel paneth cell markers, AGR2, HSPA5 and RRBP1 were also identified. Notably, significant correlation was observed between overall proteomic changes and corresponding gene expression changes along the crypt-villus axis, indicating intestinal cell maturation is primarily regulated at the transcriptional level. This proteomic profiling analysis identified several novel proteins and functional processes differentially induced during intestinal cell maturation in vivo. Integration of proteomic, immunohistochemical, and parallel gene expression datasets demonstrate the coordinated manner in which intestinal cell maturation is regulated.


Subject(s)
Intestinal Mucosa/physiology , Intestine, Small/physiology , Proteomics , Animals , Coloring Agents , Electrophoresis, Gel, Two-Dimensional , Endoplasmic Reticulum Chaperone BiP , Enzymes/chemistry , Enzymes/genetics , Enzymes/isolation & purification , Gene Expression Regulation , Intestinal Mucosa/chemistry , Intestinal Mucosa/cytology , Intestine, Small/chemistry , Intestine, Small/cytology , Lipids/physiology , Mice , Proteins/chemistry , Proteins/genetics , Proteins/isolation & purification , Rats , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Steroids/metabolism
13.
Gastroenterology ; 135(3): 849-60, 860.e1-6, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18621050

ABSTRACT

BACKGROUND & AIMS: Notch downstream targets are fundamental to intestinal cell lineage commitment and are suggested as therapeutic targets for colon cancer cells. However, the role of endogenous Notch signaling through receptor-ligand interaction, and effects of its longer term down-regulation on intestinal homeostasis, are unclear. METHODS: To address these issues, the gene encoding protein O-fucosyltransferase 1, an enzyme required for Notch ligand binding and thus activation of all Notch receptors, was deleted in the mouse intestinal and colonic epithelium, through Villin-Cre-mediated recombination. RESULTS: Pofut1 deletion inactivated Notch signaling, giving rise to smaller but viable mice. These mice exhibited a large increase in all intestinal secretory cell lineages, which accumulated in the crypts, resulting in crypt hyperplasia. Although proliferating cells were largely reduced in the colon, the transit amplifying compartment was maintained in the upper crypts of the intestinal mucosa. By 9 months, these perturbations in cell maturation altered mucus-associated gut microbiota and caused chronic intestinal inflammation, with evidence of bacterial translocation to the mesenteric lymph nodes, macrophage, and T-lymphocyte infiltration, and Th1/Th17 immune response. Dysplastic foci were also observed in Pofut1-deficient small intestine with occasional progression to tumor formation. CONCLUSIONS: Mucus hypersecretion upon Pofut1 inactivation is accompanied by alteration of the mucus-associated flora, which likely contributes to the development of enterocolitis. Therefore, these data identify important potential complications in strategies to target Notch signaling in therapeutic approaches to colon cancer.


Subject(s)
Enterocolitis/metabolism , Fucosyltransferases/metabolism , Intestinal Mucosa/metabolism , Receptors, Notch/metabolism , Signal Transduction , Animals , Bacterial Translocation , Cell Fractionation , Enterocolitis/pathology , Fatty Acid-Binding Proteins/metabolism , Fucosyltransferases/genetics , Gene Silencing , High Mobility Group Proteins/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Ligands , Lymph Nodes/microbiology , Mice , Mice, Transgenic , SOX9 Transcription Factor , Tissue Culture Techniques , Transcription Factors/metabolism , Galactoside 2-alpha-L-fucosyltransferase
14.
Mol Microbiol ; 58(1): 6-16, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16164545

ABSTRACT

Because of their key role in cell signalling, a rigorous regulation of mitogen-activated protein kinases (MAPKs) is essential in eukaryotic physiology. Whereas the use of binding motifs and scaffold proteins guarantees the selective activation of a specific MAPK pathway, activating kinases and downregulating phosphatases control the appropriate intensity and timing of MAPK activation. Tyrosine, serine/threonine and dual-specificity phosphatases co-ordinately dephosphorylate and thereby inactivate MAPKs. In budding yeast, enzymes that belong to these three types of phosphatases have been shown to counteract the MAPKs that govern the cellular response to varied extracellular stimuli. Studies carried out with these yeast phosphatases have expanded our knowledge of essential key aspects of the biology of these negative regulators, such as their function, the mechanisms that operate in their modulation by MAPK pathways and their binding to MAPK substrates. Furthermore, yeast MAPK phosphatases have been shown to play additional and essential roles in MAPK-mediated signalling, controlling MAPK localization or cross-talk among pathways. This review stresses the importance of these negative regulators in eukaryotic signalling by discussing the recent developments and perspectives in the study of yeast MAPK phosphatases.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Phosphoprotein Phosphatases/metabolism , Saccharomyces cerevisiae/enzymology , Signal Transduction , Adaptation, Physiological , Saccharomyces cerevisiae/metabolism
15.
J Biol Chem ; 279(12): 11027-34, 2004 Mar 19.
Article in English | MEDLINE | ID: mdl-14703512

ABSTRACT

Dual-specificity protein phosphatases (DSPs) are involved in the negative regulation of mitogen-activated protein kinases (MAPKs) by dephosphorylating both threonine- and tyrosine-conserved residues located at the activation loop. Here we show that Msg5 DSP activity is essential for maintaining a low level of signaling through the cell integrity pathway in Saccharomyces cerevisiae. Consistent with a role of this phosphatase on cell wall physiology, cells lacking Msg5 displayed an increased sensitivity to the cell wall-interfering compound Congo Red. We have observed that the N-terminal non-catalytic region of this phosphatase was responsible for binding to the kinase domain of Slt2, the MAPK that operates in this pathway. In vivo and in vitro experiments revealed that both proteins act on each other. Msg5 bound and dephosphorylated activated Slt2. Reciprocally, Slt2 phosphorylated Msg5 as a consequence of the activation of the cell integrity pathway. In addition, alternative use of translation initiation sites at MSG5 resulted in two protein forms that are functional on Slt2 and became equally phosphorylated following activation of this MAPK. Under activating conditions, a decrease in the affinity between Msg5 and Slt2 was observed, leading us to suggest that the mechanism by which Slt2 controls the action of Msg5 was via the modulation of protein-protein interactions. Our results indicate the existence of posttranscriptional mechanisms of regulation of DSPs in yeast and provide new insights into the negative control of the cell integrity pathway.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Protein Tyrosine Phosphatases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Amino Acid Sequence , Base Sequence , DNA Primers , Phosphorylation , RNA Processing, Post-Transcriptional , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Signal Transduction , Substrate Specificity , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...