Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38187663

RESUMO

Pancreatic stellate cells (PSCs) are primarily responsible for producing the stiff tumor tissue in pancreatic ductal adenocarcinoma (PDAC). Thereby, PSCs generate a stiffness gradient between the healthy pancreas and the tumor. This gradient induces durotaxis, a form of directional cell migration driven by differential stiffness. The molecular sensors behind durotaxis are still unclear. To investigate the role of mechanosensitive ion channels in PSC durotaxis, we established a two-dimensional stiffness gradient mimicking PDAC. Using pharmacological and genetic methods, we investigated the role of the ion channels Piezo1, TRPC1, and TRPV4 in PSC durotaxis. We found that PSC migration towards a stiffer substrate is diminished by altering Piezo1 activity. Moreover, disrupting TRPC1 along with TRPV4 abolishes PSC durotaxis even when Piezo1 is functional. Hence, PSC durotaxis is optimal with an intermediary level of mechanosensitive channel activity, which we simulated using a numerically discretized mathematical model. Our findings suggest that mechanosensitive ion channels, particularly Piezo1, detect the mechanical microenvironment to guide PSC migration.

2.
JCI Insight ; 7(8)2022 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-35451372

RESUMO

Pancreatic fibrosis is a complication of chronic pancreatitis and is a prominent feature of pancreatic cancer. Pancreatic fibrosis is commonly observed in patients with prolonged pancreatic duct obstruction, which elevates intrapancreatic pressure. We show here that increased pancreatic duct pressure causes fibrosis and describes the mechanism by which pressure increases deposition of extracellular matrix proteins and fibrosis. We found that pancreatic stellate cells (PSCs), the source of the extracellular matrix proteins in fibrosis, express the mechanically activated ion channel Piezo1. By increasing intracellular calcium, mechanical stress or the Piezo1 agonist Yoda1-activated PSCs manifest by loss of perinuclear fat droplets and increased TGF-ß1, fibronectin, and type I collagen expression. These effects were blocked by the Piezo1 inhibitor GsMTx4 and absent in PSCs from mice with conditional genetic deletion of Piezo1 in stellate cells, as was pancreatic duct ligation-induced fibrosis. Although TRPV4 has been proposed to have direct mechanosensing properties, we discovered that PSCs from Trpv4-KO mice were protected against Yoda1-triggered activation. Moreover, mice devoid of TRPV4 were protected from pancreatic duct ligation-induced fibrosis. Thus, high pressure within the pancreas stimulates Piezo1 channel opening, and subsequent activation of TRPV4 leads to stellate cell activation and pressure-induced chronic pancreatitis and fibrosis.


Assuntos
Canais Iônicos , Pancreatite Crônica , Canais de Cátion TRPV , Animais , Fibrose , Humanos , Canais Iônicos/genética , Canais Iônicos/metabolismo , Camundongos , Pâncreas/patologia , Células Estreladas do Pâncreas , Pancreatite Crônica/induzido quimicamente , Pancreatite Crônica/genética , Pancreatite Crônica/metabolismo , Canais de Cátion TRPV/genética
3.
Am J Physiol Gastrointest Liver Physiol ; 322(6): G561-G570, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35293263

RESUMO

Proper mitochondrial function and adequate cellular ATP are necessary for normal pancreatic protein synthesis and sorting, maintenance of intracellular organelles and enzyme secretion. Inorganic phosphate is required for generating ATP and its limited availability may lead to reduced ATP production causing impaired Ca2+ handling, defective autophagy, zymogen activation, and necrosis, which are all features of acute pancreatitis. We hypothesized that reduced dietary phosphate leads to hypophosphatemia and exacerbates pancreatitis severity of multiple causes. We observed that mice fed a low-phosphate diet before the induction of pancreatitis by either repeated caerulein administration or pancreatic duct injection as a model of pressure-induced pancreatitis developed hypophosphatemia and exhibited more severe pancreatitis than normophosphatemic mice. Pancreatitis severity was significantly reduced in mice treated with phosphate. In vitro modeling of secretagogue- and pressure-induced pancreatic injury was evaluated in isolated pancreatic acini using cholecystokinin and the mechanoreceptor Piezo1 agonist, Yoda1, under low and normal phosphate conditions. Isolated pancreatic acini were more sensitive to cholecystokinin- and Yoda1-induced acinar cell damage and mitochondrial dysfunction under low-phosphate conditions and improved following phosphate supplementation. Importantly, even mice on a normal phosphate diet exhibited less severe pancreatitis when treated with supplemental phosphate. Thus, hypophosphatemia sensitizes animals to pancreatitis and phosphate supplementation reduces pancreatitis severity. These appear to be direct effects of phosphate on acinar cells through restoration of mitochondrial function. We propose that phosphate administration may be useful in the treatment of acute pancreatitis.NEW & NOTEWORTHY Impaired ATP synthesis disrupts acinar cell homeostasis and is an early step in pancreatitis. We report that reduced phosphate availability impairs mitochondrial function and worsens pancreatic injury. Phosphate supplementation improves mitochondrial function and protects against experimental pancreatitis, raising the possibility that phosphate supplementation may be useful in treating pancreatitis.


Assuntos
Hipofosfatemia , Pancreatite , Doença Aguda , Trifosfato de Adenosina/metabolismo , Animais , Ceruletídeo/farmacologia , Colecistocinina/metabolismo , Hipofosfatemia/metabolismo , Canais Iônicos/metabolismo , Camundongos , Pâncreas/metabolismo , Pancreatite/induzido quimicamente , Pancreatite/tratamento farmacológico , Pancreatite/metabolismo , Fosfatos/metabolismo
4.
J Clin Invest ; 130(5): 2527-2541, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-31999644

RESUMO

Elevated pressure in the pancreatic gland is the central cause of pancreatitis following abdominal trauma, surgery, endoscopic retrograde cholangiopancreatography, and gallstones. In the pancreas, excessive intracellular calcium causes mitochondrial dysfunction, premature zymogen activation, and necrosis, ultimately leading to pancreatitis. Although stimulation of the mechanically activated, calcium-permeable ion channel Piezo1 in the pancreatic acinar cell is the initial step in pressure-induced pancreatitis, activation of Piezo1 produces only transient elevation in intracellular calcium that is insufficient to cause pancreatitis. Therefore, how pressure produces a prolonged calcium elevation necessary to induce pancreatitis is unknown. We demonstrate that Piezo1 activation in pancreatic acinar cells caused a prolonged elevation in intracellular calcium levels, mitochondrial depolarization, intracellular trypsin activation, and cell death. Notably, these effects were dependent on the degree and duration of force applied to the cell. Low or transient force was insufficient to activate these pathological changes, whereas higher and prolonged application of force triggered sustained elevation in intracellular calcium, leading to enzyme activation and cell death. All of these pathological events were rescued in acinar cells treated with a Piezo1 antagonist and in acinar cells from mice with genetic deletion of Piezo1. We discovered that Piezo1 stimulation triggered transient receptor potential vanilloid subfamily 4 (TRPV4) channel opening, which was responsible for the sustained elevation in intracellular calcium that caused intracellular organelle dysfunction. Moreover, TRPV4 gene-KO mice were protected from Piezo1 agonist- and pressure-induced pancreatitis. These studies unveil a calcium signaling pathway in which a Piezo1-induced TRPV4 channel opening causes pancreatitis.


Assuntos
Canais Iônicos/agonistas , Pancreatite/etiologia , Pancreatite/fisiopatologia , Canais de Cátion TRPV/fisiologia , Células Acinares/efeitos dos fármacos , Células Acinares/patologia , Células Acinares/fisiologia , Animais , Cálcio/metabolismo , Sinalização do Cálcio , Morte Celular , Modelos Animais de Doenças , Feminino , Canais Iônicos/genética , Canais Iônicos/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Pâncreas/efeitos dos fármacos , Pâncreas/patologia , Pâncreas/fisiopatologia , Pancreatite/patologia , Pressão , Pirazinas/farmacologia , Canais de Cátion TRPV/deficiência , Canais de Cátion TRPV/genética , Tiadiazóis/farmacologia
5.
Nat Commun ; 9(1): 1715, 2018 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-29712913

RESUMO

Merely touching the pancreas can lead to premature zymogen activation and pancreatitis but the mechanism is not completely understood. Here we demonstrate that pancreatic acinar cells express the mechanoreceptor Piezo1 and application of pressure within the gland produces pancreatitis. To determine if this effect is through Piezo1 activation, we induce pancreatitis by intrapancreatic duct instillation of the Piezo1 agonist Yoda1. Pancreatitis induced by pressure within the gland is prevented by a Piezo1 antagonist. In pancreatic acinar cells, Yoda1 stimulates calcium influx and induces calcium-dependent pancreatic injury. Finally, selective acinar cell-specific genetic deletion of Piezo1 protects mice against pressure-induced pancreatitis. Thus, activation of Piezo1 in pancreatic acinar cells is a mechanism for pancreatitis and may explain why pancreatitis develops following pressure on the gland as in abdominal trauma, pancreatic duct obstruction, pancreatography, or pancreatic surgery. Piezo1 blockade may prevent pancreatitis when manipulation of the gland is anticipated.


Assuntos
Células Acinares/efeitos dos fármacos , Cálcio/metabolismo , Canais Iônicos/genética , Mecanotransdução Celular/efeitos dos fármacos , Pâncreas/efeitos dos fármacos , Pancreatite/prevenção & controle , Células Acinares/metabolismo , Células Acinares/patologia , Animais , Modelos Animais de Doenças , Expressão Gênica , Humanos , Pressão Hidrostática/efeitos adversos , Peptídeos e Proteínas de Sinalização Intercelular , Canais Iônicos/agonistas , Canais Iônicos/antagonistas & inibidores , Canais Iônicos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pâncreas/metabolismo , Pâncreas/patologia , Pancreatite/etiologia , Pancreatite/genética , Pancreatite/patologia , Peptídeos/farmacologia , Cultura Primária de Células , Bibliotecas de Moléculas Pequenas/farmacologia , Venenos de Aranha/farmacologia
6.
Cell Mol Gastroenterol Hepatol ; 1(1): 75-86, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25729765

RESUMO

BACKGROUND & AIMS: In the pancreas, activation of primary sensory nerves through the transient receptor potential ion channel TRPV1 contributes to the early stages of development of pancreatitis. Little is known about the mechanism by which this occurs. We investigated whether leukotriene B4 (LTB4) is an endogenous agonist of TRPV1 and mediates pancreatitis. METHODS: Acute inflammation was induced in the pancreata of Trpv1-/- mice and their wild-type littermates by retrograde infusion of the main pancreatic duct with 2% sodium taurocholate (NaT) or intraperitoneal injections of caerulein. Mice were also given injections of resiniferatoxin (an excitotoxin that desensitizes TRPV1) or MK886 (a drug that inhibits LTB4 biosynthesis). Pancreatic tissues and plasma were collected and analyzed. RESULTS: Retrograde perfusion of the main pancreatic ducts of wild-type mice with NaT caused severe acute pancreatitis; severity was reduced by co-administration of resiniferatoxin. Trpv1-/- mice developed a less severe pancreatitis following NaT administration than controls. Administration of MK886 before perfusion with NaT also significantly reduced the severity of pancreatitis in wild-type mice. Pancreatic tissues from mice given NaT had a marked increase in the level of 5-lipoxygenase immunoreactivity specifically in acinar cells. Bile acid and caerulein induced secretion of LTB4 by cultured pancreatic acinar cells; MK886 inhibited this process. CONCLUSIONS: Administration of caerulein or intraductal bile acids in mice causes production of LTB4 by pancreatic acinar cells. This activates TRPV1 on primary sensory nerves to induce acute pancreatitis.

7.
Eur J Clin Invest ; 45(3): 237-46, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25641074

RESUMO

BACKGROUND: Regular gall bladder contraction reduces bile stasis and prevents gallstone formation. Intraduodenal administration of exogenous pancreatic secretory trypsin inhibitor-I (PSTI-I, also known as monitor peptide) causes cholecystokinin (CCK) secretion. DESIGN: We proposed that stimulation of CCK release by PSTI would produce gall bladder contraction and prevent gallstones in mice fed a lithogenic diet. Therefore, we tested the effect of overexpression of rat PSTI-I in pancreatic acinar cells on plasma CCK levels and gall bladder function in a transgenic mouse line (TgN[Psti1]; known hereafter as PSTI-I tg). RESULTS: Importantly, PSTI tg mice had elevated fasting and fed plasma CCK levels compared to wild-type (WT) mice. Only mice fed the lithogenic diet developed gallstones. Both fasting and stimulated plasma CCK levels were substantially reduced in both WT and PSTI-I tg mice on the lithogenic diet. Moreover, despite higher CCK levels PSTI-I tg animals developed more gallstones than WT animals. CONCLUSIONS: Together with the previously observed decrease in CCK-stimulated gall bladder emptying in mice fed a lithogenic diet, our findings suggest that a lithogenic diet causes gallstone formation by impaired CCK secretion in addition to reduced gall bladder sensitivity to CCK.


Assuntos
Colecistocinina/metabolismo , Cálculos Biliares/prevenção & controle , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Células Acinares/metabolismo , Animais , Colecistite/patologia , Dieta , Vesícula Biliar/patologia , Esvaziamento da Vesícula Biliar/fisiologia , Cálculos Biliares/fisiopatologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Metabolismo dos Lipídeos/fisiologia , Masculino , Camundongos Endogâmicos C57BL , Tamanho do Órgão/fisiologia , Pâncreas/citologia , Ratos , Inibidor da Tripsina Pancreática de Kazal
8.
Am J Physiol Gastrointest Liver Physiol ; 302(5): G535-41, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22173919

RESUMO

IL-1ß is believed to play a pathogenic role in the development of pancreatitis. Expression of human IL-1ß in pancreatic acinar cells produces chronic pancreatitis, characterized by extensive intrapancreatic inflammation, atrophy, and fibrosis. To determine if activation of trypsinogen is important in the pathogenesis of chronic pancreatitis in this model, we crossed IL-1ß transgenic [Tg(IL1ß)] mice with mice expressing a trypsin inhibitor that is normally produced in rat pancreatic acinar cells [pancreatic secretory trypsin inhibitor (PTSI) I]. We previously demonstrated that transgenic expression of PSTI-I [Tg(Psti1)] increased pancreatic trypsin inhibitor activity by 190%. Tg(IL1ß) mice were found to have marked pancreatic inflammation, characterized by histological changes, including acinar cell loss, inflammatory cell infiltration, and fibrosis, as well as elevated myeloperoxidase activity and elevated pancreatic trypsin activity, as early as 6 wk of age. In contrast to Tg(IL1ß) mice, pancreatitis was significantly less severe in dual-transgenic [Tg(IL1ß)-Tg(Psti1)] mice expressing IL-1ß and PSTI-I in pancreatic acinar cells. These findings indicate that overexpression of PSTI-I reduces the severity of pancreatitis and that pancreatic trypsin activity contributes to the pathogenesis of an inflammatory model of chronic pancreatitis.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Interleucina-1beta/biossíntese , Pâncreas/metabolismo , Pancreatite Crônica/prevenção & controle , Actinas/biossíntese , Amilases/metabolismo , Animais , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Masculino , Camundongos , Pâncreas/patologia , Pancreatite/patologia , Pancreatite Crônica/patologia , Ratos , Tripsina/metabolismo , Inibidor da Tripsina Pancreática de Kazal , Inibidores da Tripsina/biossíntese , Inibidores da Tripsina/fisiologia
9.
Am J Physiol Gastrointest Liver Physiol ; 298(4): G518-24, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20110462

RESUMO

Endogenous trypsin inhibitors are synthesized, stored, and secreted by pancreatic acinar cells. It is believed that they play a protective role in the pancreas by inhibiting trypsin within the cell should trypsinogen become prematurely activated. Rodent trypsin inhibitors are highly homologous to human serine protease inhibitor Kazal-type 1 (SPINK1). The mouse has one pancreatic trypsin inhibitor known as SPINK3, and the rat has two trypsin inhibitors commonly known as pancreatic secretory trypsin inhibitors I and II (PSTI-I and -II). Rat PSTI-I is a 61-amino acid protein that shares 65% sequence identity with mouse SPINK3. It was recently demonstrated that mice with genetic deletion of the Spink3 gene (Spink3(-/-)) do not survive beyond 15 days and lack normal pancreata because of pancreatic autophagy. We have shown that targeted transgenic expression of the rat Psti1 gene to acinar cells in mice [TgN(Psti1)] protects mice against caerulein-induced pancreatitis. To determine whether the autophagic phenotype and lethality in Spink3(-/-) mice were due to lack of pancreatic trypsin inhibitor, we conducted breeding studies with Spink3(+/-) heterozygous mice and TgN(Psti1) mice. We observed that, whereas Spink3(+/+), Spink3(+/-), and Spink3(-/-)/TgN(Psti1) mice had similar survival rates, no Spink3(-/-) mice survived longer than 1 wk. The level of expression of SPINK3 protein in acini was reduced in heterozygote mice compared with wild-type mice. Furthermore, endogenous trypsin inhibitor capacity was reduced in the pancreas of heterozygote mice compared with wild-type or knockout mice rescued with the rat Psti1 gene. Surprisingly, the lesser amount of SPINK3 present in the pancreata of heterozygote mice did not predispose animals to increased susceptibility to caerulein-induced acute pancreatitis. We propose that a threshold level of expression is sufficient to protect against pancreatitis.


Assuntos
Glicoproteínas/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Pâncreas/patologia , Pancreatite/genética , Proteínas Secretadas pela Próstata/genética , Transgenes/genética , Sequência de Aminoácidos , Amilases/sangue , Animais , Tamanho Corporal/genética , Ceruletídeo/farmacologia , Feminino , Glicoproteínas/metabolismo , Heterozigoto , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Dados de Sequência Molecular , Tamanho do Órgão/genética , Pâncreas/efeitos dos fármacos , Pâncreas/metabolismo , Pâncreas Exócrino/patologia , Pâncreas Exócrino/ultraestrutura , Pancreatite/induzido quimicamente , Pancreatite/patologia , Proteínas Secretadas pela Próstata/metabolismo , Ratos , Homologia de Sequência de Aminoácidos , Taxa de Sobrevida , Tripsina/metabolismo , Inibidor da Tripsina Pancreática de Kazal
10.
Pancreas ; 36(4): 394-401, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18437086

RESUMO

OBJECTIVES: Transient receptor potential subtype vanilloid 1 (TRPV1) is an ion channel that is primarily expressed by primary sensory neurons where it mediates pain and heat sensation and participates in neurogenic inflammation. In this study, we examined the role of TRPV1 during neurogenic activation of pancreatic inflammation using a secretagogue-induced model in mice. METHODS: A supramaximal dose of caerulein (50 microg/kg) was injected hourly for 12 hours. Mice lacking TRPV1 were compared to wild-type animals. RESULTS: All the parameters: serum amylase, pancreatic myeloperoxidase activity, histological scoring, pancreatic wet weight/body weight ratio, and quantification of neurokinin-1 receptor internalization indicated that null mice were not protected from acute pancreatitis. However, when primary sensory neurons were ablated by injection of the neurotoxin and TRPV1 agonist, resiniferatoxin, pancreatitis was ameliorated in wild-type mice but not in null mice, indicating that nerves bearing TRPV1 are part of the inflammatory pathway in acute pancreatitis because disappearance significantly reduced the inflammatory response. CONCLUSIONS: Nerves expressing TRPV1 participate in the neurogenic inflammation during acute pancreatitis. The lack of protection in TRPV1 null mice suggests that an alternate pathway to TRPV1 coexists in the same neurons.


Assuntos
Ceruletídeo/farmacologia , Deleção de Genes , Regulação da Expressão Gênica , Neurônios Aferentes/fisiologia , Pancreatite/genética , Canais de Cátion TRPV/genética , Doença Aguda , Animais , Cruzamentos Genéticos , Modelos Animais de Doenças , Endocitose , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios Aferentes/efeitos dos fármacos , Pancreatite/prevenção & controle , Canais de Cátion TRPV/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA