Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Am J Pathol ; 193(2): 161-181, 2023 02.
Article in English | MEDLINE | ID: mdl-36410420

ABSTRACT

The roof plate-specific spondin-leucine-rich repeat-containing G-protein coupled receptor 4/5 (LGR4/5)-zinc and ring finger 3 (ZNRF3)/ring finger protein 43 (RNF43) module is a master regulator of hepatic Wnt/ß-catenin signaling and metabolic zonation. However, its impact on nonalcoholic fatty liver disease (NAFLD) remains unclear. The current study investigated whether hepatic epithelial cell-specific loss of the Wnt/ß-catenin modulator Lgr4/5 promoted NAFLD. The 3- and 6-month-old mice with hepatic epithelial cell-specific deletion of both receptors Lgr4/5 (Lgr4/5dLKO) were compared with control mice fed with normal diet (ND) or high-fat diet (HFD). Six-month-old HFD-fed Lgr4/5dLKO mice developed hepatic steatosis and fibrosis but the control mice did not. Serum cholesterol-high-density lipoprotein and total cholesterol levels in 3- and 6-month-old HFD-fed Lgr4/5dLKO mice were decreased compared with those in control mice. An ex vivo primary hepatocyte culture assay and a comprehensive bile acid (BA) characterization in liver, plasma, bile, and feces demonstrated that ND-fed Lgr4/5dLKO mice had impaired BA secretion, predisposing them to develop cholestatic characteristics. Lipidome and RNA-sequencing analyses demonstrated severe alterations in several lipid species and pathways controlling lipid metabolism in the livers of Lgr4/5dLKO mice. In conclusion, loss of hepatic Wnt/ß-catenin activity by Lgr4/5 deletion led to loss of BA secretion, cholestatic features, altered lipid homeostasis, and deregulation of lipoprotein pathways. Both BA and intrinsic lipid alterations contributed to the onset of NAFLD.


Subject(s)
Non-alcoholic Fatty Liver Disease , Animals , Mice , Non-alcoholic Fatty Liver Disease/metabolism , beta Catenin/metabolism , Leucine/metabolism , Liver/metabolism , Cholesterol/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Mice, Inbred C57BL , Diet, High-Fat/adverse effects
2.
PLoS One ; 16(8): e0255687, 2021.
Article in English | MEDLINE | ID: mdl-34379673

ABSTRACT

Identifying the fundamental molecular factors that drive weight gain even in the absence of hypercaloric food intake, is crucial to enable development of novel treatments for the global pandemic of obesity. Here we investigated both adipose tissue-specific and systemic events that underlie the physiological weight gain occurring during early adulthood in mice fed a normocaloric diet. In addition, we used three different genetic models to identify molecular factors that promote physiological weight gain during normocaloric and hypercaloric diets. We demonstrated that normal physiological weight gain was accompanied by an increase in adipose tissue mass and the presence of cellular and metabolic signatures typically found during obesity, including adipocyte hypertrophy, macrophage recruitment into visceral fat and perturbed glucose metabolism. At the molecular level, this was associated with an increase in adipose tissue tryptophan hydroxylase 1 (Tph1) transcripts, the key enzyme responsible for the synthesis of peripheral serotonin. Genetic inactivation of Tph1 was sufficient to limit adipose tissue expansion and associated metabolic alterations. Mechanistically, we discovered that Tph1 inactivation resulted in down-regulation of cyclin-dependent kinase inhibitor p21Waf1/Cip1 expression. Single or double ablation of Tph1 and p21 were equally effective in preventing adipocyte expansion and systemic perturbation of glucose metabolism, upon both normocaloric and hypercaloric diets. Our results suggest that serotonin and p21 act as a central molecular determinant of weight gain and associated metabolic alterations, and highlights the potential of targeting these molecules as a pharmacologic approach to prevent the development of obesity.


Subject(s)
Adipose Tissue/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Diet, Healthy/methods , Gene Deletion , Obesity/metabolism , Serotonin/biosynthesis , Signal Transduction/genetics , Adipocytes/pathology , Animals , Cell Size , Cyclin-Dependent Kinase Inhibitor p21/genetics , Diet, High-Fat , Glucose/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Tryptophan Hydroxylase/genetics , Tryptophan Hydroxylase/metabolism , Weight Gain/genetics
3.
J Pathol ; 250(1): 42-54, 2020 01.
Article in English | MEDLINE | ID: mdl-31531867

ABSTRACT

Molecular signalling mediated by the phosphatidylinositol-3-kinase (PI3K)-Akt axis is a key regulator of cellular functions. Importantly, alteration of the PI3K-Akt signalling underlies the development of different human diseases, thus prompting the investigation of the pathway as a molecular target for pharmacologic intervention. In this regard, recent studies showed that small molecule inhibitors of PI3K, the upstream regulator of the pathway, reduced the development of inflammation during acute pancreatitis, a highly debilitating and potentially lethal disease. Here we investigated whether a specific reduction of Akt activity, by using either pharmacologic Akt inhibition, or genetic inactivation of the Akt1 isoform selectively in pancreatic acinar cells, is effective in ameliorating the onset and progression of the disease. We discovered that systemic reduction of Akt activity did not protect the pancreas from initial damage and only transiently delayed leukocyte recruitment. However, reduction of Akt activity decreased acinar proliferation and exacerbated acinar-to-ductal metaplasia (ADM) formation, two critical events in the progression of pancreatitis. These phenotypes were recapitulated upon conditional inactivation of Akt1 in acinar cells, which resulted in reduced expression of 4E-BP1, a multifunctional protein of key importance in cell proliferation and metaplasia formation. Collectively, our results highlight the critical role played by Akt1 during the development of acute pancreatitis in the control of acinar cell proliferation and ADM formation. In addition, these results harbour important translational implications as they raise the concern that inhibitors of PI3K-Akt signalling pathways may negatively affect the regeneration of the pancreas. Finally, this work provides the basis for further investigating the potential of Akt1 activators to boost pancreatic regeneration following inflammatory insults. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Acinar Cells/enzymology , Cell Proliferation , Pancreas, Exocrine/enzymology , Pancreatic Ducts/enzymology , Pancreatitis/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Acinar Cells/drug effects , Acinar Cells/pathology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Ceruletide , Disease Models, Animal , Male , Metaplasia , Mice, Inbred C57BL , Mice, Knockout , Pancreas, Exocrine/drug effects , Pancreas, Exocrine/pathology , Pancreatic Ducts/drug effects , Pancreatic Ducts/pathology , Pancreatitis/chemically induced , Pancreatitis/genetics , Pancreatitis/pathology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/analysis , Proto-Oncogene Proteins c-akt/deficiency , Proto-Oncogene Proteins c-akt/genetics , Rats , Signal Transduction
4.
J Pathol ; 248(2): 217-229, 2019 06.
Article in English | MEDLINE | ID: mdl-30714146

ABSTRACT

Proliferation of pancreatic acinar cells is a critical process in the pathophysiology of pancreatic diseases, because limited or defective proliferation is associated with organ dysfunction and patient morbidity. In this context, elucidating the signalling pathways that trigger and sustain acinar proliferation is pivotal to develop therapeutic interventions promoting the regenerative process of the organ. In this study we used genetic and pharmacological approaches to manipulate both local and systemic levels of thyroid hormones to elucidate their role in acinar proliferation following caerulein-mediated acute pancreatitis in mice. In addition, molecular mechanisms mediating the effects of thyroid hormones were identified by genetic and pharmacological inactivation of selected signalling pathways.In this study we demonstrated that levels of the thyroid hormone 3,3',5-triiodo-l-thyronine (T3) transiently increased in the pancreas during acute pancreatitis. Moreover, by using genetic and pharmacological approaches to manipulate both local and systemic levels of thyroid hormones, we showed that T3 was required to promote proliferation of pancreatic acinar cells, without affecting the extent of tissue damage or inflammatory infiltration.Finally, upon genetic and pharmacological inactivation of selected signalling pathways, we demonstrated that T3 exerted its mitogenic effect on acinar cells via a tightly controlled action on different molecular effectors, including histone deacetylase, AKT, and TGFß signalling.In conclusion, our data suggest that local availability of T3 in the pancreas is required to promote acinar cell proliferation and provide the rationale to exploit thyroid hormone signalling to enhance pancreatic regeneration. Copyright © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Acinar Cells/metabolism , Cell Proliferation , Hyperthyroidism/metabolism , Pancreas, Exocrine/metabolism , Pancreatitis/metabolism , Triiodothyronine/metabolism , Acinar Cells/pathology , Animals , Ceruletide , Disease Models, Animal , Histone Deacetylases/metabolism , Hyperthyroidism/genetics , Hyperthyroidism/pathology , Iodide Peroxidase/deficiency , Iodide Peroxidase/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Pancreas, Exocrine/pathology , Pancreatitis/chemically induced , Pancreatitis/genetics , Pancreatitis/pathology , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Transforming Growth Factor-beta Type II/deficiency , Receptor, Transforming Growth Factor-beta Type II/genetics , Signal Transduction , Thyroxine/metabolism , Transforming Growth Factor beta/metabolism , Up-Regulation
5.
J Pathol ; 246(3): 352-365, 2018 11.
Article in English | MEDLINE | ID: mdl-30058725

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC), which is the primary cause of pancreatic cancer mortality, is poorly responsive to currently available interventions. Identifying new targets that drive PDAC formation and progression is critical for developing alternative therapeutic strategies to treat this lethal malignancy. Using genetic and pharmacological approaches, we investigated in vivo and in vitro whether uptake of the monoamine serotonin [5-hydroxytryptamine (5-HT)] is required for PDAC development. We demonstrated that pancreatic acinar cells have the ability to readily take up 5-HT in a transport-mediated manner. 5-HT uptake promoted activation of the small GTPase Ras-related C3 botulinum toxin substrate 1 (Rac1), which is required for transdifferentiation of acinar cells into acinar-to-ductal metaplasia (ADM), a key determinant in PDAC development. Consistent with the central role played by Rac1 in ADM formation, inhibition of the 5-HT transporter Sert (Slc6a4) with fluoxetine reduced ADM formation both in vitro and in vivo in a cell-autonomous manner. In addition, fluoxetine treatment profoundly compromised the stromal reaction and affected the proliferation and lipid metabolism of malignant PDAC cells. We propose that Sert is a promising therapeutic target to counteract the early event of ADM, with the potential to stall the initiation and progression of pancreatic carcinogenesis. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Carcinoma, Pancreatic Ductal/enzymology , Cell Proliferation , Genes, ras , Neuropeptides/metabolism , Pancreas/enzymology , Pancreatic Neoplasms/enzymology , Serotonin/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/prevention & control , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Transdifferentiation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Chick Embryo , Disease Models, Animal , Enzyme Activation , Fluoxetine/pharmacology , Genetic Predisposition to Disease , Humans , Metaplasia , Mice, Inbred C57BL , Mice, Transgenic , Neovascularization, Pathologic , Pancreas/drug effects , Pancreas/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/prevention & control , Phenotype , Rats , Serotonin Plasma Membrane Transport Proteins/drug effects , Serotonin Plasma Membrane Transport Proteins/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Signal Transduction , Tryptophan Hydroxylase/genetics , Tryptophan Hydroxylase/metabolism
6.
Mol Pharmacol ; 94(2): 793-801, 2018 08.
Article in English | MEDLINE | ID: mdl-29880639

ABSTRACT

Pancreatic fibrosis is the hallmark of chronic pancreatitis, a highly debilitating disease for which there is currently no cure. The key event at the basis of pancreatic fibrosis is the deposition of extracellular matrix proteins by activated pancreatic stellate cells (PSCs). Transforming growth factor ß (TGFß) is a potent profibrotic factor in the pancreas as it promotes the activation of PSC; thus, pharmacologic interventions that effectively reduce TGFß expression harbor considerable therapeutic potential in the treatment of chronic pancreatitis. In this study, we investigated whether TGFß expression is reduced by pharmacologic inhibition of the epigenetic modifiers histone deacetylases (HDACs). To address this aim, chronic pancreatitis was induced in C57BL/6 mice with serial injections of cerulein, and the selective class 1 HDAC inhibitor MS-275 was administered in vivo in a preventive and therapeutic manner. Both MS-275 regimens potently reduced deposition of extracellular matrix and development of fibrosis in the pancreas after 4 weeks of chronic pancreatitis. Reduced pancreatic fibrosis was concomitant with lower expression of pancreatic TGFß and consequent reduced PSC activation. In search of the cell types targeted by the inhibitor, we found that MS-275 treatment abrogated the expression of TGFß in acinar cells stimulated by cerulein treatment. Our study demonstrates that MS-275 is an effective antifibrotic agent in the context of experimental chronic pancreatitis and thus may constitute a valid therapeutic intervention for this severe disease.


Subject(s)
Benzamides/administration & dosage , Histone Deacetylase Inhibitors/administration & dosage , Pancreas/drug effects , Pancreatitis, Chronic/chemically induced , Pancreatitis, Chronic/drug therapy , Pyridines/administration & dosage , Transforming Growth Factor beta/metabolism , Animals , Benzamides/pharmacology , Cell Line , Ceruletide/adverse effects , Disease Models, Animal , Fibrosis/prevention & control , Gene Expression Regulation , Histone Deacetylase Inhibitors/pharmacology , Mice , Mice, Inbred C57BL , Pancreas/pathology , Pancreatic Stellate Cells/cytology , Pancreatic Stellate Cells/drug effects , Pancreatic Stellate Cells/metabolism , Pancreatitis, Chronic/metabolism , Pyridines/pharmacology , Rats
7.
Br J Pharmacol ; 174(21): 3865-3880, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28832971

ABSTRACT

BACKGROUND AND PURPOSE: Pancreatitis is a common inflammation of the pancreas with rising incidence in many countries. Despite improvements in diagnostic techniques, the disease is associated with high risk of severe morbidity and mortality and there is an urgent need for new therapeutic interventions. In this study, we evaluated whether histone deacetylases (HDACs), key epigenetic regulators of gene transcription, are involved in the development of the disease. EXPERIMENTAL APPROACH: We analysed HDAC regulation during cerulein-induced acute, chronic and autoimmune pancreatitis using different transgenic mouse models. The functional relevance of class I HDACs was tested with the selective inhibitor MS-275 in vivo upon pancreatitis induction and in vitro in activated macrophages and primary acinar cell explants. KEY RESULTS: HDAC expression and activity were up-regulated in a time-dependent manner following induction of pancreatitis, with the highest abundance observed for class I HDACs. Class I HDAC inhibition did not prevent the initial acinar cell damage. However, it effectively reduced the infiltration of inflammatory cells, including macrophages and T cells, in both acute and chronic phases of the disease, and directly disrupted macrophage activation. In addition, MS-275 treatment reduced DNA damage in acinar cells and limited acinar de-differentiation into acinar-to-ductal metaplasia in a cell-autonomous manner by impeding the EGF receptor signalling axis. CONCLUSIONS AND IMPLICATIONS: These results demonstrate that class I HDACs are critically involved in the development of acute and chronic forms of pancreatitis and suggest that blockade of class I HDAC isoforms is a promising target to improve the outcome of the disease.


Subject(s)
Autoimmune Diseases/drug therapy , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/drug effects , Pancreatitis/drug therapy , Acinar Cells/metabolism , Acute Disease , Animals , Autoimmune Diseases/physiopathology , Benzamides/pharmacology , Disease Models, Animal , ErbB Receptors/metabolism , Histone Deacetylases/metabolism , Leukocytes/metabolism , Male , Metaplasia , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pancreatitis/physiopathology , Pancreatitis, Chronic/drug therapy , Pancreatitis, Chronic/physiopathology , Pyridines/pharmacology , Time Factors
8.
J Hepatol ; 66(3): 560-570, 2017 03.
Article in English | MEDLINE | ID: mdl-27771454

ABSTRACT

BACKGROUND & AIMS: ALPPS, a novel two-staged approach for the surgical removal of large/multiple liver tumors, combines portal vein ligation (PVL) with parenchymal transection. This causes acceleration of compensatory liver growth, enabling faster and more extensive tumor removal. We sought to identify the plasma factors thought to mediate the regenerative acceleration following ALPPS. METHODS: We compared a mouse model of ALPPS against PVL and additional control surgeries (n=6 per group). RNA deep sequencing was performed to identify candidate molecules unique to ALPPS liver (n=3 per group). Recombinant protein and a neutralizing antibody combined with appropriate surgeries were used to explore candidate functions in ALPPS (n=6 per group). Indian hedgehog (IHH/Ihh) levels were assessed in human ALPPS patient plasma (n=6). RESULTS: ALPPS in mouse confirmed significant acceleration of liver regeneration relative to PVL (p<0.001). Ihh mRNA, coding for a secreted ligand inducing hedgehog signaling, was uniquely upregulated in ALPPS liver (p<0.001). Ihh plasma levels rose 4h after surgery (p<0.01), along with hedgehog pathway activation and subsequent cyclin D1 induction in the liver. When combined with PVL, Ihh alone was sufficient to induce ALPPS-like acceleration of liver growth. Conversely, blocking Ihh markedly inhibited the accelerating effects of ALPPS. In the small cohort of ALPPS patients, IHH tended to be elevated early after surgery. CONCLUSIONS: Ihh and hedgehog pathway activation provide the first mechanistic insight into the acceleration of liver regeneration triggered by ALPPS surgery. The accelerating potency of recombinant Ihh, and its potential effect in human ALPPS may lead to a clinical role for this protein. LAY SUMMARY: ALPPS, a novel two-staged hepatectomy, accelerates liver regeneration, thereby helping to treat patients with otherwise unresectable liver tumors. The molecular mechanisms behind this accelerated regeneration are unknown. Here, we elucidate that Indian hedgehog, a secreted ligand important for fetal development, is a crucial mediator of the regenerative acceleration triggered by ALPPS surgery.


Subject(s)
Hedgehog Proteins/metabolism , Hepatectomy/methods , Liver Regeneration/physiology , Animals , Hedgehog Proteins/administration & dosage , Hedgehog Proteins/blood , Hedgehog Proteins/genetics , Humans , Ligation , Liver Neoplasms/blood , Liver Neoplasms/blood supply , Liver Neoplasms/surgery , Liver Regeneration/genetics , Male , Mice , Mice, Inbred C57BL , Models, Animal , Portal Vein/surgery , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/administration & dosage
9.
J Pathol ; 238(3): 434-45, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26510396

ABSTRACT

Determining signalling pathways that regulate pancreatic regeneration following pancreatitis is critical for implementing therapeutic interventions. In this study we elucidated the molecular mechanisms underlying the effects of transforming growth factor-ß (TGFß) in pancreatic epithelial cells during tissue regeneration. To this end, we conditionally inactivated TGFß receptor II (TGFß-RII) using a Cre-LoxP system under the control of pancreas transcription factor 1a (PTF1a) promoter, specific for the pancreatic epithelium, and evaluated the molecular and cellular changes in a mouse model of cerulein-induced pancreatitis. We show that TGFß-RII signalling does not mediate the initial acinar cell damage observed at the onset of pancreatitis. However, TGFß-RII signalling not only restricts acinar cell replication during the regenerative phase of the disease but also limits ADM formation in vivo and in vitro in a cell-autonomous manner. Analyses of molecular mechanisms underlying the observed phenotype revealed that TGFß-RII signalling stimulates the expression of cyclin-dependent kinase inhibitors and intersects with the EGFR signalling axis. Finally, TGFß-RII ablation in epithelial cells resulted in increased infiltration of inflammatory cells in the early phases of pancreatitis and increased activation of pancreatic stellate cells in the later stages of pancreatitis, thus highlighting a TGFß-based crosstalk between epithelial and stromal cells regulating the development of pancreatic inflammation and fibrosis. Collectively, our data not only contribute to clarifying the cellular processes governing pancreatic tissue regeneration, but also emphasize the conserved role of TGFß as a tumour suppressor, both in the regenerative process following pancreatitis and in the initial phases of pancreatic cancer.


Subject(s)
Acinar Cells/pathology , Pancreas/pathology , Pancreatitis/pathology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Amylases/metabolism , Animals , Carcinoma, Pancreatic Ductal/pathology , Cell Cycle Checkpoints/physiology , Cell Proliferation/physiology , Cell Transformation, Neoplastic/pathology , Cells, Cultured , Ceruletide/toxicity , Epithelial Cells/pathology , Fibrosis/pathology , Irritants/toxicity , Lipase/metabolism , Male , Metaplasia/pathology , Mice, Knockout , Mice, Transgenic , Pancreas/enzymology , Pancreatic Neoplasms/pathology , Pancreatitis/enzymology , Receptor, Transforming Growth Factor-beta Type II
10.
J Pathol ; 237(4): 495-507, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26235267

ABSTRACT

The exocrine pancreas exhibits a distinctive capacity for tissue regeneration and renewal following injury. This regenerative ability has important implications for a variety of disorders, including pancreatitis and pancreatic cancer, diseases associated with high morbidity and mortality. Thus, understanding its underlying mechanisms may help in developing therapeutic interventions. Serotonin has been recognized as a potent mitogen for a variety of cells and tissues. Here we investigated whether serotonin exerts a mitogenic effect in pancreatic acinar cells in three regenerative models, inflammatory tissue injury following pancreatitis, tissue loss following partial pancreatectomy, and thyroid hormone-stimulated acinar proliferation. Genetic and pharmacological techniques were used to modulate serotonin levels in vivo. Acinar dedifferentiation and cell cycle progression during the regenerative phase were investigated over the course of 2 weeks. By comparing acinar proliferation in the different murine models of regeneration, we found that serotonin did not affect the clonal regeneration of mature acinar cells. Serotonin was, however, required for acinar dedifferentiation following inflammation-mediated tissue injury. Specifically, lack of serotonin resulted in delayed up-regulation of progenitor genes and delayed the formation of acinar-to-ductal metaplasia and defective acinar cell proliferation. We identified serotonin-dependent acinar secretion as a key step in progenitor-based regeneration, as it promoted acinar cell dedifferentiation and the recruitment of type 2 macrophages. Finally, we identified a regulatory Hes1-Ptfa axis in the uninjured adult pancreas, activated by zymogen secretion. Our findings indicated that serotonin plays a critical role in the regeneration of the adult pancreas following pancreatitis by promoting the dedifferentiation of acinar cells.


Subject(s)
Acinar Cells/cytology , Cell Dedifferentiation/physiology , Pancreas, Exocrine/physiology , Serotonin/metabolism , Aging , Animals , Disease Models, Animal , Immunoblotting , Immunohistochemistry , Metaplasia , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Pancreatitis/pathology , Regeneration
11.
J Pathol ; 235(3): 502-14, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25212177

ABSTRACT

Trans-differentiation of pancreatic acinar cells into ductal-like lesions, a process defined as acinar-to-ductal metaplasia (ADM), is observed in the course of organ regeneration following pancreatitis. In addition, ADM is found in association with pre-malignant PanIN lesions and correlates with an increased risk of pancreatic adenocarcinoma (PDAC). Human PDAC samples show down-regulation of p21(WAF1) (/Cip1) , a key regulator of cell cycle and cell differentiation. Here we investigated whether p21 down-regulation is implicated in controlling the early events of acinar cell trans-differentiation and ADM formation. p21-mediated regulation of ADM formation and regression was analysed in vivo during the course of cerulein-induced pancreatitis, using wild-type (WT) and p21-deficient (p21(-/-) ) mice. Biochemical and immunohistochemical methods were used to evaluate disease progression over 2 weeks of the disease and during a recovery phase. We found that p21 was strongly up-regulated in WT acinar cells during pancreatitis, while it was absent in ADM areas, suggesting that p21 down-regulation is associated with ADM formation. In support of this hypothesis, p21(-/-) mice showed a significant increase in number and size of metaplasia. In addition, p21 over-expression in acinar cells reduced ADM formation in vitro, suggesting that the protein regulates the metaplastic transition in a cell-autonomous manner. p21(-/-) mice displayed increased expression and relocalization of ß-catenin both during pancreatitis and in the subsequent recovery phase. Finally, loss of p21 was accompanied by increased DNA damage and development of senescence. Our findings are consistent with a gate-keeper role of p21 in acinar cells to limit senescence activation and ADM formation during pancreatic regeneration.


Subject(s)
Cell Differentiation/physiology , Cellular Senescence/physiology , Cyclin-Dependent Kinase Inhibitor p21/physiology , Pancreas/pathology , Pancreas/physiopathology , Pancreatitis/pathology , Pancreatitis/physiopathology , Animals , Cell Cycle/physiology , Cell Proliferation/physiology , Ceruletide/adverse effects , Cyclin-Dependent Kinase Inhibitor p21/deficiency , Cyclin-Dependent Kinase Inhibitor p21/genetics , Disease Models, Animal , Down-Regulation/physiology , In Vitro Techniques , Metaplasia , Mice , Mice, Knockout , Pancreatitis/chemically induced , Regeneration/physiology , beta Catenin/physiology
12.
Diabetes ; 63(4): 1326-39, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24379346

ABSTRACT

Irreversible failure of pancreatic ß-cells is the main culprit in the pathophysiology of diabetes, a disease that is now a global epidemic. Recently, elevated plasma levels of deoxysphingolipids, including 1-deoxysphinganine, have been identified as a novel biomarker for the disease. In this study, we analyzed whether deoxysphingolipids directly compromise the functionality of insulin-producing Ins-1 cells and primary islets. Treatment with 1-deoxysphinganine induced dose-dependent cytotoxicity with senescent, necrotic, and apoptotic characteristics and compromised glucose-stimulated insulin secretion. In addition, 1-deoxysphinganine altered cytoskeleton dynamics, resulting in intracellular accumulation of filamentous actin and activation of the Rho family GTPase Rac1. Moreover, 1-deoxysphinganine selectively upregulated ceramide synthase 5 expression and was converted to 1-deoxy-dihydroceramides without altering normal ceramide levels. Inhibition of intracellular 1-deoxysphinganine trafficking and ceramide synthesis improved the viability of the cells, indicating that the intracellular metabolites of 1-deoxysphinganine contribute to its cytotoxicity. Analyses of signaling pathways identified Jun N-terminal kinase and p38 mitogen-activated protein kinase as antagonistic effectors of cellular senescence. The results revealed that 1-deoxysphinganine is a cytotoxic lipid for insulin-producing cells, suggesting that the increased levels of this sphingolipid observed in diabetic patients may contribute to the reduced functionality of pancreatic ß-cells. Thus, targeting deoxysphingolipid synthesis may complement the currently available therapies for diabetes.


Subject(s)
Insulin-Secreting Cells/drug effects , Sphingosine/analogs & derivatives , Animals , Apoptosis/drug effects , Biomarkers , Blood Glucose/metabolism , Cell Survival/drug effects , Cells, Cultured , Cellular Senescence/drug effects , Cytoskeleton/drug effects , Humans , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Lipids , Mice , Rats , Signal Transduction , Sphingosine/metabolism , Sphingosine/pharmacology , Sphingosine/toxicity
13.
Gut ; 62(6): 890-8, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22591619

ABSTRACT

OBJECTIVE: Serotonin (5-hydroxytryptamine, 5-HT) is a potent bioactive molecule involved in a variety of physiological processes. In this study, the authors analysed whether 5-HT regulates zymogen secretion in pancreatic acinar cells and the development of pancreatic inflammation, a potentially lethal disease whose pathophysiology is not completely understood. METHODS: 5-HT regulation of zymogen secretion was analysed in pancreatic acini isolated from wild-type or tryptophan hydoxylase-1 knock-out (TPH1(-/-)) mice, which lack peripheral 5-HT, and in amylase-secreting pancreatic cell lines. Pancreatitis was induced by cerulein stimulation and biochemical and immunohistochemical methods were used to evaluate disease progression over 2 weeks. RESULTS: Absence and reduced intracellular levels of 5-HT inhibited the secretion of zymogen granules both ex vivo and in vitro and altered cytoskeleton dynamics. In addition, absence of 5-HT resulted in attenuated pro-inflammatory response after induction of pancreatitis. TPH1(-/-) mice showed limited zymogen release, reduced expression of the pro-inflammatory chemokine MCP-1 and minimal leucocyte infiltration compared with wild-type animals. Restoration of 5-HT levels in TPH1(-/-) mice recovered the blunted inflammatory processes observed during acute pancreatitis. However, cellular damage, inflammatory and fibrotic processes accelerated in TPH1(-/-) mice during disease progression. CONCLUSIONS: These results identify a 5-HT-mediated regulation of zymogen secretion in pancreatic acinar cells. In addition, they demonstrate that 5-HT is required for the onset but not for the progression of pancreatic inflammation. These findings provide novel insights into the normal physiology of pancreatic acinar cells and into the pathophysiology of pancreatitis, with potential therapeutic implications.


Subject(s)
Acinar Cells/pathology , Amylases/metabolism , Enzyme Precursors/metabolism , Pancreatitis/enzymology , Serotonin/physiology , Acinar Cells/enzymology , Actin Cytoskeleton/pathology , Animals , Cell Line , Ceruletide/adverse effects , Chemotaxis, Leukocyte , Disease Progression , Fibrosis , Immunohistochemistry , Inflammation/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreatitis/chemically induced , Pancreatitis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...