Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 101
Filtrer
1.
Funct Integr Genomics ; 24(5): 140, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-39160285

RÉSUMÉ

Acute pancreatitis (AP) is an inflammatory disease of the pancreas and the main cause of hospital admissions for gastrointestinal diseases. Here, the work studied the circular RNA DTNB/microRNA-485-5p/MCL1 axis in AP and hoped to unravel the related mechanism. Caerulein exposure replicated an AP model in AR42J cells, and caerulein-mediated expression of circDTNB, miR-485-5p, and MCL1 was recorded. After exposure, cells were intervened with transfection plasmids and tested for LDH release, apoptosis, and inflammation. To determine the interwork of circDTNB, miR-485-5p, and MCL1, prediction results and verification experiments were conducted. Caerulein exposure reduced circDTNB and MCL1, while elevated miR-485-5p levels in AR42J cells. Upregulating circDTNB protected AR42J cells from caerulein-induced LDH cytotoxicity, apoptosis, and inflammation, but circDTNB upregulation-induced protections could be muffled by inhibiting MCL1. On the contrary, downregulating circDTNB further damaged AR42J cells under caerulein exposure, however, this phenomenon could be partially rescued after silencing miR-485-5p. miR-485-5p was mechanistically verified to be a target of circDTNB to mediate MCL1. Overall, the circDTNB/miR-485-5p/MCL1 axis protects inflammatory response and apoptosis in caerulein-exposed AR42J cells, promisingly identifying circDTNB as a novel molecule for AP treatment.


Sujet(s)
Apoptose , Céruléine , Inflammation , microARN , Protéine Mcl-1 , ARN circulaire , microARN/génétique , microARN/métabolisme , Protéine Mcl-1/génétique , Protéine Mcl-1/métabolisme , ARN circulaire/génétique , ARN circulaire/métabolisme , Animaux , Rats , Inflammation/génétique , Pancréatite/génétique , Pancréatite/métabolisme , Pancréatite/induit chimiquement , Pancréatite/anatomopathologie , Lignée cellulaire
2.
Int J Mol Sci ; 25(15)2024 Jul 29.
Article de Anglais | MEDLINE | ID: mdl-39125854

RÉSUMÉ

Acute pancreatitis (AP) is a significant cause of morbidity, even in children, and is frequently associated with systemic manifestations. There are many cytokines involved in the inflammatory response characteristic of this disease. Interleukin 6 (IL-6) is one of the most important cytokines involved in AP, beginning from cellular injury and continuing to the systemic inflammatory response and distant organ involvement. IL-6 is a multifunctional cytokine that regulates acute-phase response and inflammation. It is produced by various cells and exerts its biological role on many cells through its high-affinity complex receptor. IL-6 has been investigated as a predicting maker for severe forms of AP. Many studies have validated the use of IL-6 serum levels in the first 48 h as a reliable marker for severe evolution and multisystemic involvement. Still, it has not been used in daily practice until now. This review discusses the main binding mechanisms by which IL-6 triggers cellular response and the AP pathogenetic mechanisms in which IL-6 is involved. We then emphasize the promising role of IL-6 as a prognostic marker, which could be added as a routine marker at admission in children with AP.


Sujet(s)
Marqueurs biologiques , Interleukine-6 , Pancréatite , Humains , Interleukine-6/sang , Interleukine-6/métabolisme , Pancréatite/métabolisme , Pancréatite/sang , Pancréatite/diagnostic , Marqueurs biologiques/sang , Pronostic , Maladie aigüe , Animaux
3.
J Biochem Mol Toxicol ; 38(8): e23802, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39132808

RÉSUMÉ

Hyperlipidemic pancreatitis (HP) is an inflammatory injury of the pancreas triggered by elevated serum triglyceride (TG) levels. The mechanistic target of rapamycin (mTOR) signaling pathway plays a crucial role in regulating lipid homeostasis and inflammation. This study aimed to investigate whether the activity of mTOR complex 2 (mTORC2) affects the progression of HP and its underlying mechanisms. In vivo, a high-fat diet and retrograde administration of sodium taurocholate were employed to establish the HP models in rats, with pancreatic tissue pathology evaluated. The expression of Rictor and peroxisome proliferator-activator receptor (PPAR) was examined. The serum levels of TG, fatty acid metabolites, inflammatory and lipid metabolism-related factors were determined. In vitro, pancreatic acinar cells (PACs) were exposed to palmitic acid and cholecystokinin-8. PAC apoptosis, pyroptosis, and ferroptosis were assessed. In the HP models, rats and PACs exhibited upregulated Rictor and downregulated PPARα, and Rictor knockdown promoted PPARα expression. In vivo, Rictor knockdown decreased the serum levels of TG, α-amylase, total cholesterol, low-density lipoprotein cholesterol, lactate dehydrogenase, and inflammatory factors, while increasing high-density lipoprotein cholesterol levels. Rictor knockdown increased ACOX1 and CPT1α and decreased SREBP-1, CD36, SCD1, ACLY, and ACACA. Rictor knockdown reduced damage to pancreatic tissue structure. In vitro, Rictor knockdown inhibited PAC apoptosis, pyroptosis, and ferroptosis. Treatment with the PPARα antagonist GW6471 abolished the beneficial effects of Rictor knockdown. Rictor/mTORC2 deficiency reduces serum TG levels, maintains lipid homeostasis, and suppresses inflammation by inhibiting PPARα expression. Weakening mTORC2 activity holds promise as a novel therapeutic strategy for HP.


Sujet(s)
Hyperlipidémies , Métabolisme lipidique , Complexe-2 cible mécanistique de la rapamycine , Récepteur PPAR alpha , Pancréatite , Rat Sprague-Dawley , Animaux , Récepteur PPAR alpha/métabolisme , Récepteur PPAR alpha/génétique , Rats , Pancréatite/métabolisme , Pancréatite/anatomopathologie , Pancréatite/induit chimiquement , Pancréatite/génétique , Hyperlipidémies/métabolisme , Hyperlipidémies/génétique , Métabolisme lipidique/effets des médicaments et des substances chimiques , Mâle , Complexe-2 cible mécanistique de la rapamycine/métabolisme , Techniques de knock-down de gènes
4.
BMC Gastroenterol ; 24(1): 245, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39090535

RÉSUMÉ

BACKGROUND: Ferroptosis is a newly recognized form of regulatory cell death characterized by severe lipid peroxidation triggered by iron overload and the production of reactive oxygen species (ROS). However, the role of ferroptosis in severe acute pancreatitis(SAP) has not been fully elucidated. METHODS: We established four severe acute pancreatitis models of rats including the sham control group, the SAP group, the Fer -1-treated SAP (SAP + Fer-1) group, the 3-MA-treated SAP (SAP + 3-MA) group. The SAP group was induced by retrograde injection of sodium taurocholate into the pancreatic duct. The other two groups were intraperitoneally injected with ferroptosis inhibitor (Fer-1) and autophagy inhibitor (3-MA), respectively. The model of severe acute pancreatitis with amylase crest-related inflammatory factors was successfully established. Then we detected ferroptosis (GPX4, SLC7A1 etc.) and autophagy-related factors (LC3II, p62 ect.) to further clarify the relationship between ferroptosis and autophagy. RESULTS: Our study found that ferroptosis occurs during the development of SAP, such as iron and lipid peroxidation in pancreatic tissues, decreased levels of reduced glutathione peroxidase 4 (GPX 4) and glutathione (GSH), and increased malondialdehyde(MDA) and significant mitochondrial damage. In addition, ferroptosis related proteins such as GPX4, solute carrier family 7 member 11(SLC7A11) and ferritin heavy chain 1(FTH1) were significantly decreased. Next, the pathogenesis of ferroptosis in SAP was studied. First, treatment with the ferroptosis inhibitor ferrostatin-1(Fer-1) significantly alleviated ferroptosis in SAP. Interestingly, autophagy occurs during the pathogenesis of SAP, and autophagy promotes the occurrence of ferroptosis in SAP. Moreover, 3-methyladenine (3-MA) inhibition of autophagy can significantly reduce iron overload and ferroptosis in SAP. CONCLUSIONS: Our results suggest that ferroptosis is a novel pathogenesis of SAP and is dependent on autophagy. This study provides a new theoretical basis for the study of SAP.


Sujet(s)
Autophagie , Modèles animaux de maladie humaine , Ferroptose , Peroxydation lipidique , Pancréatite , Rat Sprague-Dawley , Animaux , Pancréatite/métabolisme , Pancréatite/anatomopathologie , Rats , Mâle , Adénine/analogues et dérivés , Adénine/pharmacologie , Phospholipid hydroperoxide glutathione peroxidase/métabolisme , Acide taurocholique , Cyclohexylamines/pharmacologie , Pancréas/anatomopathologie , Pancréas/métabolisme , Phénylènediamines/pharmacologie , Malonaldéhyde/métabolisme , Espèces réactives de l'oxygène/métabolisme , Maladie aigüe , Glutathion/métabolisme , Fer/métabolisme
5.
World J Gastroenterol ; 30(23): 2927-2930, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38946872

RÉSUMÉ

In this editorial, we focus specifically on the mechanisms by which pancreatic inflammation affects pancreatic cancer. Cancer of the pancreas remains one of the deadliest cancer types. The highest incidence and mortality rates of pancreatic cancer are found in developed countries. Trends of pancreatic cancer incidence and mortality vary considerably worldwide. A better understanding of the etiology and identification of the risk factors is essential for the primary prevention of this disease. Pancreatic tumors are characterized by a complex microenvironment that orchestrates metabolic alterations and supports a milieu of interactions among various cell types within this niche. In this editorial, we highlight the foundational studies that have driven our understanding of these processes. In our experimental center, we have carefully studied the mechanisms of that link pancreatic inflammation and pancreatic cancer. We focused on the role of mast cells (MCs). MCs contain pro-angiogenic factors, including tryptase, that are associated with increased angiogenesis in various tumors. In this editorial, we address the role of MCs in angiogenesis in both pancreatic ductal adenocarcinoma tissue and adjacent normal tissue. The assessment includes the density of c-Kit receptor-positive MCs, the density of tryptase-positive MCs, the area of tryptase-positive MCs, and angiogenesis in terms of microvascularization density.


Sujet(s)
Mastocytes , Néovascularisation pathologique , Tumeurs du pancréas , Microenvironnement tumoral , Humains , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/immunologie , Mastocytes/métabolisme , Mastocytes/immunologie , Microenvironnement tumoral/immunologie , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/immunologie , Carcinome du canal pancréatique/métabolisme , Pancréas/anatomopathologie , Pancréas/immunologie , Pancréas/métabolisme , Animaux , Pancréatite/métabolisme , Pancréatite/anatomopathologie , Pancréatite/immunologie , Facteurs de risque , Médiateurs de l'inflammation/métabolisme , Tryptases/métabolisme , Inflammation/métabolisme
6.
Immun Inflamm Dis ; 12(7): e1301, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38967361

RÉSUMÉ

OBJECTIVE: Acute pancreatitis (AP) stands as a frequent cause for clinical emergency hospital admissions. The X-box binding protein 1 (XBP1) was found to be implicated in pancreatic acinar cell apoptosis. The objective is to unveil the potential mechanisms governed by XBP1 and SIRT6 in the context of AP. METHODS: Caerulein-treated human pancreatic duct epithelial (HPDE) cells to establish an in vitro research model. The levels and regulatory role of SIRT6 in the treated cells were evaluated, including its effects on inflammatory responses, oxidative stress, apoptosis, and endoplasmic reticulum stress. The relationship between XBP1 and SIRT6 was explored by luciferase and ChIP experiments. Furthermore, the effect of XBP1 overexpression on the regulatory function of SIRT6 on cells was evaluated. RESULTS: Caerulein promoted the decrease of SIRT6 and the increase of XBP1 in HPDE cells. Overexpression of SIRT6 slowed down the secretion of inflammatory factors, oxidative stress, apoptosis level, and endoplasmic reticulum stress in HPDE cells. However, XBP1 negatively regulated SIRT6, and XBP1 overexpression partially reversed the regulation of SIRT6 on the above aspects. CONCLUSION: Our study illuminates the role of XBP1 in downregulating SIRT6 in HPDE cells, thereby promoting cellular injury. Inhibiting XBP1 or augmenting SIRT6 levels holds promise in preserving cell function and represents a potential therapeutic avenue in the management of AP.


Sujet(s)
Apoptose , Régulation négative , Cellules épithéliales , Conduits pancréatiques , Pancréatite , Sirtuines , Protéine-1 liant la boite X , Humains , Sirtuines/métabolisme , Sirtuines/génétique , Cellules épithéliales/métabolisme , Protéine-1 liant la boite X/métabolisme , Protéine-1 liant la boite X/génétique , Pancréatite/métabolisme , Pancréatite/anatomopathologie , Conduits pancréatiques/métabolisme , Conduits pancréatiques/anatomopathologie , Stress du réticulum endoplasmique , Stress oxydatif , Lignée cellulaire , Céruléine/toxicité
7.
ACS Nano ; 18(29): 19283-19302, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-38990194

RÉSUMÉ

Developing strategies to target injured pancreatic acinar cells (PACs) in conjunction with primary pathophysiology-specific pharmacological therapy presents a challenge in the management of acute pancreatitis (AP). We designed and synthesized a trypsin-cleavable organosilica precursor bridged by arginine-based amide bonds, leveraging trypsin's ability to selectively identify guanidino groups on arginine via Asp189 at the active S1 pocket and cleave the carboxy-terminal (C-terminal) amide bond via catalytic triads. The precursors were incorporated into the framework of mesoporous silica nanoparticles (MSNs) for encapsulating the membrane-permeable Ca2+ chelator BAPTA-AM with a high loading content (∼43.9%). Mesenchymal stem cell membrane coating and surface modification with PAC-targeting ligands endow MSNs with inflammation recruitment and precise PAC-targeting abilities, resulting in the highest distribution at 3 h in the pancreas with 4.7-fold more accumulation than that of naked MSNs. The outcomes transpired as follows: After bioinspired MSNs' skeleton biodegradation by prematurely and massively activated trypsin, BAPTA-AM was on-demand released in injured PACs, thereby effectively eliminating intracellular calcium overload (reduced Ca2+ level by 81.3%), restoring cellular redox status, blocking inflammatory cascades, and inhibiting cell necrosis by impeding the IκBα/NF-κB/TNF-α/IL-6 and CaMK-II/p-RIP3/p-MLKL/caspase-8,9 signaling pathways. In AP mice, a single dose of the formulation significantly restored pancreatic function (lipase and amylase reduced more by 60%) and improved the survival rate from 50 to 91.6%. The formulation offers a potentially effective strategy for clinical translation in AP treatment.


Sujet(s)
Pancréatite , Trypsine , Animaux , Pancréatite/traitement médicamenteux , Pancréatite/anatomopathologie , Pancréatite/métabolisme , Trypsine/métabolisme , Trypsine/composition chimique , Souris , Porosité , Nanomédecine , Matériaux biomimétiques/composition chimique , Matériaux biomimétiques/pharmacologie , Nanoparticules/composition chimique , Silice/composition chimique , Composés organiques du silicium/composition chimique , Composés organiques du silicium/pharmacologie , Mâle , Humains , Cellules acineuses/effets des médicaments et des substances chimiques , Cellules acineuses/métabolisme , Cellules acineuses/anatomopathologie , Souris de lignée C57BL
8.
Am J Physiol Gastrointest Liver Physiol ; 327(3): G333-G344, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-38981616

RÉSUMÉ

The serine protease chymotrypsin protects the pancreas against pancreatitis by degrading trypsinogen, the precursor to the digestive protease trypsin. Taking advantage of previously generated mouse models with either the Ctrb1 gene (encoding chymotrypsin B1) or the Ctrl gene (encoding chymotrypsin-like protease) disrupted, here we generated the novel Ctrb1-del × Ctrl-KO strain in the C57BL/6N genetic background, which harbors a naturally inactivated Ctrc gene (encoding chymotrypsin C). The newly created mice are devoid of chymotrypsin, yet the animals develop normally, breed well, and show no spontaneous phenotype, indicating that chymotrypsin is dispensable under laboratory conditions. When given cerulein, the Ctrb1-del × Ctrl-KO strain exhibited markedly increased intrapancreatic trypsin activation and more severe acute pancreatitis, relative to wild-type C57BL/6N mice. After the acute episode, Ctrb1-del × Ctrl-KO mice spontaneously progressed to chronic pancreatitis, whereas C57BL/6N mice recovered rapidly. The cerulein-induced pancreas pathology in Ctrb1-del × Ctrl-KO mice was highly similar to that previously observed in Ctrb1-del mice; however, trypsin activation was more robust and pancreatitis severity was increased. Taken together, the results confirm and extend prior observations demonstrating that chymotrypsin safeguards the pancreas against pancreatitis by limiting pathologic trypsin activity. In mice, the CTRB1 isoform, which constitutes about 90% of the total chymotrypsin content, is responsible primarily for the anti-trypsin defenses and protection against pancreatitis; however, the minor isoform CTRL also contributes to an appreciable extent.NEW & NOTEWORTHY Chymotrypsins defend the pancreas against the inflammatory disorder pancreatitis by degrading harmful trypsinogen. This study demonstrates that mice devoid of pancreatic chymotrypsins are phenotypically normal but become sensitized to secretagogue hyperstimulation and exhibit increased intrapancreatic trypsin activation, more severe acute pancreatitis, and rapid progression to chronic pancreatitis. The observations confirm and extend the essential role of chymotrypsins in pancreas health.


Sujet(s)
Céruléine , Chymotrypsine , Souris de lignée C57BL , Souris knockout , Pancréatite , Trypsine , Animaux , Chymotrypsine/métabolisme , Chymotrypsine/génétique , Céruléine/toxicité , Pancréatite/induit chimiquement , Pancréatite/anatomopathologie , Pancréatite/métabolisme , Pancréatite/génétique , Souris , Trypsine/métabolisme , Sécrétagogues/métabolisme , Pancréas/métabolisme , Pancréas/anatomopathologie , Modèles animaux de maladie humaine , Mâle
9.
Neurosci Lett ; 837: 137902, 2024 Aug 10.
Article de Anglais | MEDLINE | ID: mdl-39029612

RÉSUMÉ

AIM: Tryptophan (TRP), an essential amino acid, undergoes catabolism through various pathways. Notably, the kynurenine pathway (KP), constituting one of these pathways, exhibits a unidirectional impact on immune response and energy metabolism. Nonetheless, its influence on pain sensation is characterized by biphasic dynamics. This study aims to scrutinize the influence of the KP pathway on pain sensation, particularly within the context of pancreatic inflammation. METHODS: Our prospective case-control study involved individuals diagnosed with acute pancreatitis and a control group matched for gender and age. The patient cohort was subsequently subdivided into severe and non-severe subgroups. To assess metabolites within KP, two blood samples were collected from the patient cohort, one at the time of diagnosis and another during the recovery phase. Furthermore, for pain quantification, daily pain scores utilizing the Visual Analog Scale (VAS) were extracted from the patients' medical records. RESULTS: The study incorporated 30 patients along with an equivalent number of controls. A noticeable distinction was evident between the patient and control groups, characterized by an increase in kynurenine levels and a decrease in the tryptophan/kynurenine ratio. Throughout the process of disease recovery, a uniform decrease was observed in all KP metabolites, excluding 3-Hydroxykynurenine. Elevated levels of Kynurenic acid (KYNA) were correlated with increased pain scores. Critically, no apparent distinctions in KP metabolites were discerned concerning pain severity in patients with comorbidities characterized by neural involvement. CONCLUSION: Based on our results, the kynurenine pathway (KP) is activated in instances of acute pancreatitis. Elevated levels of KYNA were found to be associated with heightened pain scores. The operative stages within the KP responsible for pain modulation are impaired in cases characterized by neuropathy-induced pain sensation.


Sujet(s)
Cynurénine , Perception de la douleur , Pancréatite , Tryptophane , Humains , Cynurénine/sang , Cynurénine/métabolisme , Pancréatite/sang , Pancréatite/métabolisme , Pancréatite/complications , Pancréatite/physiopathologie , Mâle , Femelle , Adulte d'âge moyen , Études cas-témoins , Tryptophane/sang , Tryptophane/métabolisme , Perception de la douleur/physiologie , Adulte , Études prospectives , Sujet âgé , Maladie aigüe
10.
Front Immunol ; 15: 1405622, 2024.
Article de Anglais | MEDLINE | ID: mdl-38827741

RÉSUMÉ

Background: Severe acute pancreatitis (SAP) is an inflammatory disorder affecting the gastrointestinal system. Intestinal injury plays an important role in the treatment of severe acute pancreatitis. In this study, we mainly investigated the role of S1PR2 in regulating macrophage pyroptosis in the intestinal injury of severe acute pancreatitis. Methods: The SAP model was constructed using cerulein and lipopolysaccharide, and the expression of S1PR2 was inhibited by JTE-013 to detect the degree of pancreatitis and intestinal tissue damage in mice. Meanwhile, the level of pyroptosis-related protein was detected by western blot, the level of related mRNA was detected by PCR, and the level of serum inflammatory factors was detected by ELISA. In vitro experiments, LPS+ATP was used to construct the pyroptosis model of THP-1. After knockdown and overexpression of S1PR2, the pyroptosis proteins level was detected by western blot, the related mRNA level was detected by PCR, and the level of cell supernatant inflammatory factors were detected by ELISA. A rescue experiment was used to verify the sufficient necessity of the RhoA/ROCK pathway in S1PR2-induced pyroptosis. Meanwhile, THP-1 and FHC were co-cultured to verify that cytokines released by THP-1 after damage could regulate FHC damage. Results: Our results demonstrated that JTE-013 effectively attenuated intestinal injury and inflammation in mice with SAP. Furthermore, we observed a significant reduction in the expression of pyroptosis-related proteins within the intestinal tissue of SAP mice upon treatment with JTE-013. We confirmed the involvement of S1PR2 in THP-1 cell pyroptosis in vitro. Specifically, activation of S1PR2 triggered pyroptosis in THP-1 cells through the RhoA/ROCK signaling pathway. Moreover, it was observed that inflammatory factors released during THP-1 cell pyroptosis exerted an impact on cohesin expression in FHC cells. Conclusion: The involvement of S1PR2 in SAP-induced intestinal mucosal injury may be attributed to its regulation of macrophage pyroptosis.


Sujet(s)
Modèles animaux de maladie humaine , Macrophages , Pancréatite , Pyroptose , Récepteurs de la sphingosine-1-phosphate , Animaux , Souris , Humains , Macrophages/métabolisme , Macrophages/immunologie , Pancréatite/métabolisme , Pancréatite/immunologie , Pancréatite/anatomopathologie , Pancréatite/induit chimiquement , Récepteurs de la sphingosine-1-phosphate/métabolisme , Récepteurs de la sphingosine-1-phosphate/génétique , Mâle , Transduction du signal , Souris de lignée C57BL , Protéine G RhoA/métabolisme , Cellules THP-1 , rho-Associated Kinases/métabolisme , rho-Associated Kinases/génétique , Muqueuse intestinale/métabolisme , Muqueuse intestinale/anatomopathologie , Intestins/anatomopathologie , Intestins/immunologie , Cytokines/métabolisme , Lipopolysaccharides , Pyrazoles , Pyridines
11.
J Clin Lab Anal ; 38(10): e25076, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38853390

RÉSUMÉ

BACKGROUND: Severe acute pancreatitis (SAP) is associated with tremendous systemic inflammation, T-helper 17 (Th17) cells, and regulatory T (Treg) cells play an essential role in the inflammatory responses. Meanwhile, soluble fibrinogen-like protein 2 (Sfgl2) is a critical immunosuppressive effector cytokine of Treg cells and modulates immune responses. However, the impact of SAP induction on Sfgl2 expression and the role of Sfgl2 in immunomodulation under SAP conditions are largely unknown. METHODS: A taurocholate-induced mouse SAP model was established. The ratios of CD4+CD25+Foxp3+ Treg cells or CD4+IL-17+ Th17 cells in blood and pancreatic tissues as well as surface expression of CD80, CD86, and major histocompatibility complex class II (MHC-II) were determined by flow cytometry. Gene mRNA expression was determined by qPCR. Serum amylase and soluble factors were quantitated by commercial kits. Bone marrow-derived dendritic cells (DCs) were generated, and NF-κB/p65 translocation was measured by immunofluorescence staining. RESULTS: SAP induction in mice decreased the Th17/Treg ratio in the pancreatic tissue and increased the Th17/Treg ratio in the peripheral blood. In addition, SAP was associated with a reduced level of Sfgl2 in the pancreatic tissue and blood: higher levels of serum IL-17, IL-2, IFN-α, and TNF-α, and lower levels of serum IL-4 and IL-10. Furthermore, the SAP-induced reduction in Sfgl2 expression was accompanied by dysregulated maturation of bone marrow-derived DCs. CONCLUSIONS: SAP causes reduced Sfgl2 expression and Th17/Treg imbalance, thus providing critical insights for the development of Sfgl2- and Th17/Treg balance-targeted immunotherapies for patients with SAP.


Sujet(s)
Modèles animaux de maladie humaine , Fibrinogène , Pancréatite , Lymphocytes T régulateurs , Acide taurocholique , Cellules Th17 , Animaux , Cellules Th17/immunologie , Lymphocytes T régulateurs/immunologie , Pancréatite/immunologie , Pancréatite/induit chimiquement , Pancréatite/métabolisme , Souris , Fibrinogène/métabolisme , Mâle , Souris de lignée C57BL , Régulation négative , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Maladie aigüe , Pancréas/immunologie , Pancréas/anatomopathologie , Pancréas/métabolisme
12.
Biomed Pharmacother ; 176: 116854, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38824834

RÉSUMÉ

BACKGROUND: Acute pancreatitis (APS) is a prevalent acute pancreatic inflammation, where oxidative stress, inflammatory signaling pathways, and apoptosis activation contribute to pancreatic injury. METHODS: Pinocembrin, the predominant flavonoid in propolis, was explored for its likely shielding effect against APS provoked by two intraperitoneal doses of L-arginine (250 mg / 100 g) in a rat model. RESULTS: Pinocembrin ameliorated the histological and immunohistochemical changes in pancreatic tissues and lowered the activities of pancreatic amylase and lipase that were markedly elevated with L-arginine administration. Moreover, pinocembrin reinstated the oxidant/antioxidant equilibrium, which was perturbed by L-arginine, and boosted the pancreatic levels of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1). Pinocembrin markedly reduced the elevation in serum C-reactive protein (CRP) level induced by L-arginine. Additionally, it decreased the expression of high motility group box protein 1 (HMGB1), toll-like receptor 4 (TLR4), nuclear factor kappa B (NF-κB), tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), and NOD-like receptor (NLR) Family Pyrin Domain Containing 3 (NLRP3) inflammasome in the pancreas. Furthermore, it also reduced myeloperoxidase (MPO) activity. Pinocembrin markedly downregulated miR-34a-5p expression and upregulated the protein levels of peroxisome proliferator-activated receptor alpha (PPAR-α) and Sirtuin 1 (SIRT1) and the gene expression level of the inhibitor protein of NF-κB (IκB-α), along with normalizing the Bax/Bcl-2 ratio. CONCLUSIONS: Pinocembrin notably improved L-arginine-induced APS by its antioxidant, anti-inflammatory, and anti-apoptotic activities. Pinocembrin exhibited a protective role in APS by suppressing inflammatory signaling via the TLR4/NF-κB/NLRP3 pathway and enhancing cytoprotective signaling via the miR-34a-5p/SIRT1/Nrf2/HO-1 pathway.


Sujet(s)
Modèles animaux de maladie humaine , Flavanones , Heme oxygenase (decyclizing) , microARN , Facteur-2 apparenté à NF-E2 , Facteur de transcription NF-kappa B , Protéine-3 de la famille des NLR contenant un domaine pyrine , Pancréatite , Rat Sprague-Dawley , Transduction du signal , Sirtuine-1 , Récepteur de type Toll-4 , Animaux , Pancréatite/induit chimiquement , Pancréatite/prévention et contrôle , Pancréatite/métabolisme , Pancréatite/anatomopathologie , Pancréatite/traitement médicamenteux , Sirtuine-1/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Récepteur de type Toll-4/métabolisme , Mâle , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , microARN/métabolisme , microARN/génétique , Flavanones/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Rats , Heme oxygenase (decyclizing)/métabolisme , Facteur-2 apparenté à NF-E2/métabolisme , Arginine/pharmacologie , Maladie aigüe , Pancréas/effets des médicaments et des substances chimiques , Pancréas/anatomopathologie , Pancréas/métabolisme , Antioxydants/pharmacologie , Stress oxydatif/effets des médicaments et des substances chimiques
13.
Int J Mol Sci ; 25(12)2024 Jun 16.
Article de Anglais | MEDLINE | ID: mdl-38928326

RÉSUMÉ

Diagnostic markers are desperately needed for the early detection of pancreatic ductal adenocarcinoma (PDA). We describe sets of markers expressed in temporal order in mouse models during pancreatitis, PDA initiation and progression. Cell type specificity and the differential expression of PDA markers were identified by screening single cell (sc) RNAseq from tumor samples of a mouse model for PDA (KIC) at early and late stages of PDA progression compared to that of a normal pancreas. Candidate genes were identified from three sources: (1) an unsupervised screening of the genes preferentially expressed in mouse PDA tumors; (2) signaling pathways that drive PDA, including the Ras pathway, calcium signaling, and known cancer genes, or genes encoding proteins that were identified by differential mass spectrometry (MS) of mouse tumors and conditioned media from human cancer cell lines; and (3) genes whose expression is associated with poor or better prognoses (PAAD, oncolnc.org). The developmental progression of PDA was detected in the temporal order of gene expression in the cancer cells of the KIC mice. The earliest diagnostic markers were expressed in epithelial cancer cells in early-stage, but not late-stage, PDA tumors. Other early markers were expressed in the epithelium of both early- and late-state PDA tumors. Markers that were expressed somewhat later were first elevated in the epithelial cancer cells of the late-stage tumors, then in both epithelial and mesenchymal cells, or only in mesenchymal cells. Stromal markers were differentially expressed in early- and/or late-stage PDA neoplasia in fibroblast and hematopoietic cells (lymphocytes and/or macrophages) or broadly expressed in cancer and many stromal cell types. Pancreatitis is a risk factor for PDA in humans. Mouse models of pancreatitis, including caerulein treatment and the acinar-specific homozygous deletion of differentiation transcription factors (dTFs), were screened for the early expression of all PDA markers identified in the KIC neoplasia. Prognostic markers associated with a more rapid decline were identified and showed differential and cell-type-specific expression in PDA, predominately in late-stage epithelial and/or mesenchymal cancer cells. Select markers were validated by immunohistochemistry in mouse and human samples of a normal pancreas and those with early- and late-stage PDA. In total, we present 2165 individual diagnostic and prognostic markers for disease progression to be tested in humans from pancreatitis to late-stage PDA.


Sujet(s)
Marqueurs biologiques tumoraux , Carcinome du canal pancréatique , Tumeurs du pancréas , Pancréatite , Animaux , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/métabolisme , Carcinome du canal pancréatique/diagnostic , Carcinome du canal pancréatique/anatomopathologie , Pancréatite/métabolisme , Pancréatite/génétique , Pancréatite/anatomopathologie , Pancréatite/diagnostic , Souris , Tumeurs du pancréas/génétique , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/diagnostic , Tumeurs du pancréas/anatomopathologie , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Humains , Pronostic , Régulation de l'expression des gènes tumoraux , Modèles animaux de maladie humaine , Lignée cellulaire tumorale , Évolution de la maladie
14.
Aging (Albany NY) ; 16(11): 10132-10141, 2024 06 11.
Article de Anglais | MEDLINE | ID: mdl-38862253

RÉSUMÉ

BACKGROUND: Acute pancreatitis (AP) is a prevalent acute abdominal condition, and AP induced colonic barrier dysfunction is commonly observed. Total flavonoids of Chrysanthemum indicum L (TFC) have exhibited noteworthy anti-inflammatory and anti-apoptotic properties. METHODS: We established AP models, both in animals and cell cultures, employing Cerulein. 16S rRNA gene sequencing was performed to investigate the gut microorganisms changes. RESULTS: In vivo, TFC demonstrated a remarkable capacity to ameliorate AP, as indicated by the inhibition of serum amylase, myeloperoxidase (MPO) levels, and the reduction in pancreatic tissue water content. Furthermore, TFC effectively curtailed the heightened inflammatory response. The dysfunction of colonic barrier induced by AP was suppressed by TFC. At the in vitro level, TFC treatment resulted in attenuation of increased cell apoptosis, and regulation of apoptosis related proteins expression in AR42J cells. The increase of Bacteroides sartorial, Lactobacillus reuteri, Muribaculum intestinale, and Parabacteroides merdae by AP, and decrease of of Helicobacter rodentium, Pasteurellaceae bacterium, Streptococcus hyointestinalis by AP were both reversed by TFC treatment. CONCLUSIONS: TFC can effectively suppress AP progression and AP induced colonic barrier dysfunction by mitigating elevated serum amylase, MPO levels, water content in pancreatic tissue, as well as curtailing inflammation, apoptosis. The findings presented herein shed light on the potential mechanisms by which TFC inhibit the development of AP progression and AP induced colonic barrier dysfunction.


Sujet(s)
Chrysanthemum , Flavonoïdes , Microbiome gastro-intestinal , Pancréatite , Animaux , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Chrysanthemum/composition chimique , Pancréatite/métabolisme , Pancréatite/microbiologie , Pancréatite/traitement médicamenteux , Flavonoïdes/pharmacologie , Mâle , Rats , Côlon/effets des médicaments et des substances chimiques , Côlon/métabolisme , Côlon/anatomopathologie , Apoptose/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Lignée cellulaire , Muqueuse intestinale/effets des médicaments et des substances chimiques , Muqueuse intestinale/métabolisme , Muqueuse intestinale/microbiologie , Muqueuse intestinale/anatomopathologie
15.
Biomolecules ; 14(6)2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38927047

RÉSUMÉ

Acute pancreatitis (AP) entails pancreatic inflammation, tissue damage and dysregulated enzyme secretion, including pancreatic lipase (PL). The role of irisin, an anti-inflammatory and anti-apoptotic cytokine, in AP and exocrine pancreatic stress is unclear. We have previously shown that irisin regulates PL through the PPARγ-PGC1α-FNDC5 pathway. In this study, we investigated irisin and irisin's pathway on AP in in vitro (AR42J-B13) and ex vivo (rat primary acinar) models using molecular, biochemical and immunohistochemistry methodology. Pancreatitis induction (cerulein (cer)) resulted in a significant up-regulation of the PPARγ-PGC1α-FNDC5 axis, PL expression and secretion and endoplasmic reticulum (ER) stress unfolded protein response (UPR) signal-transduction markers (CHOP, XBP-1 and ATF6). Irisin addition in the cer-pancreatitis state resulted in a significant down-regulation of the PPARγ-PGC1α-FNDC5 axis, PPARγ nucleus-translocation and inflammatory state (TNFα and IL-6) in parallel to diminished PL expression and secretion (in vitro and ex vivo models). Irisin addition up-regulated the expression of pro-survival UPR markers (ATF6 and XBP-1) and reduced UPR pro-apoptotic markers (CHOP) under cer-pancreatitis and induced ER stress (tunicamycin), consequently increasing cells viability. Irisin's pro-survival effect under cer-pancreatitis state was abolished under PPARγ inhibition. Our findings suggest irisin as a potential therapeutic option for AP via its ability to up-regulate pro-survival UPR signals and activate the PPARγ-PGC1α-FNDC5 pathway.


Sujet(s)
Cellules acineuses , Survie cellulaire , Fibronectines , Récepteur PPAR gamma , Pancréatite , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes , Transduction du signal , Réponse aux protéines mal repliées , Fibronectines/métabolisme , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/métabolisme , Animaux , Pancréatite/métabolisme , Pancréatite/anatomopathologie , Pancréatite/traitement médicamenteux , Récepteur PPAR gamma/métabolisme , Réponse aux protéines mal repliées/effets des médicaments et des substances chimiques , Rats , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules acineuses/métabolisme , Cellules acineuses/effets des médicaments et des substances chimiques , Cellules acineuses/anatomopathologie , Transduction du signal/effets des médicaments et des substances chimiques , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Céruléine , Mâle , Lignée cellulaire , Triacylglycerol lipase/métabolisme
16.
Biomolecules ; 14(6)2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38927046

RÉSUMÉ

Acute pancreatitis (AP) is a complex inflammatory condition that can lead to systemic inflammatory responses and multiple organ dysfunction. This study investigates the role of Galectin-3 (Gal-3), a ß-galactoside-binding lectin, in modulating acquired immune responses in AP. Acute pancreatitis was induced by ligation of the bile-pancreatic duct in wild-type and Galectin-3-deficient C57BL/6 mice. We determined the phenotypic and molecular features of inflammatory cells, serum concentrations of amylase, pancreatic trypsin activity, and pancreatic and lung pathology. Galectin-3 deficiency decreased the total number of CD3+CD49- T cells and CD4+ T helper cells, downregulated the production of inflammatory cytokine and IFN-γ, and increased the accumulation of IL-10-producing Foxp3+ T regulatory cells and regulatory CD4+ T cells in the pancreata of diseased animals. The deletion of Galectin-3 ameliorates acute pancreatitis characterized by lowering serum amylase concentration and pancreatic trypsin activity, and attenuating of the histopathology of the lung. These findings shed light on the role of Galectin-3 in acquired immune response in acute pancreatitis and identify Galectin-3 as an attractive target for investigation of the immunopathogenesis of disease and for consideration as a potential therapeutic target for patients with acute inflammatory disease of the pancreas.


Sujet(s)
Galectine -3 , Souris de lignée C57BL , Pancréatite , Lymphocytes T régulateurs , Animaux , Pancréatite/immunologie , Pancréatite/anatomopathologie , Pancréatite/métabolisme , Pancréatite/génétique , Galectine -3/métabolisme , Galectine -3/génétique , Souris , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme , Souris knockout , Maladie aigüe , Mâle , Amylases/sang
17.
Drug Des Devel Ther ; 18: 2043-2061, 2024.
Article de Anglais | MEDLINE | ID: mdl-38863767

RÉSUMÉ

Background: Despite its extensive utilization in Chinese hospitals for treating acute pancreatitis (AP) and related acute respiratory distress syndrome (ARDS), the active components and mechanisms underlying the action of Qingyi Granule (QYKL) remain elusive. Methods: This study consists of four parts. First, we used Mendelian randomization (MR) to investigate the causal relationship between AP, cytokine, and ARDS. Next, 321 patients were collected to evaluate the efficacy of QYKL combined with dexamethasone (DEX) in treating AP. In addition, we used UHPLC-QE-MS to determine the chemical constituents of QYKL extract and rat serum after the oral administration of QYKL. The weighted gene coexpression network analysis (WGCNA) method was used to find the main targets of AP-related ARDS using the GSE151572 dataset. At last, a AP model was established by retrograde injection of 5% sodium taurocholate. Results: MR showed that AP may have a causal relationship with ARDS by mediating cytokine storms. Retrospective study results showed early administration of QYKL was associated with a lower incidence of ARDS, mortality, admissions to the intensive care unit, and length of stay in AP patients compared to the Control group. Furthermore, we identified 23 QYKL prototype components absorbed into rat serum. WGCNA and differential expression analysis identified 1558 APALI-related genes. The prototype components exhibited strong binding activity with critical targets. QYKL has a significant protective effect on pancreatic and lung injury in AP rats, and the effect is more effective after combined treatment with DEX, which may be related to the regulation of the IL-6/STAT3 signaling pathway. Conclusion: By integrating MR, retrospective analysis, and systematic pharmacological methodologies, this study systematically elucidated the therapeutic efficacy of QYKL in treating AP-related ARDS, establishing a solid foundation for its medicinal use.


Sujet(s)
Médicaments issus de plantes chinoises , Pancréatite , , Médicaments issus de plantes chinoises/pharmacologie , Médicaments issus de plantes chinoises/composition chimique , /traitement médicamenteux , Pancréatite/traitement médicamenteux , Pancréatite/métabolisme , Animaux , Rats , Humains , Études rétrospectives , Mâle , Rat Sprague-Dawley , Dexaméthasone/pharmacologie , Dexaméthasone/administration et posologie , Maladie aigüe , Femelle , Adulte d'âge moyen
18.
J Gastroenterol ; 59(9): 869-879, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38864913

RÉSUMÉ

BACKGROUND: Accumulating evidence has shown that the NOD-like receptor protein 3 (NLRP3) inflammasome plays a crucial role in the inflammatory cascades involved in the development of acute pancreatitis (AP). However, the specific agonist responsible for activating the NLRP3 inflammasome in this process has not yet been identified. The purpose of this study is to clarify whether heparan sulfate (HS) works as an NLRP3 inflammasome activator to evoke inflammatory cascades in the progression of AP. METHODS: Two experimental mouse models of AP were utilized to investigate the pro-inflammatory activity of HS in the development of AP by measuring the secretion of inflammatory cytokines and the neutrophil infiltration in pancreatic tissue. The ability of HS to activate the NLRP3 inflammasome was evaluated both in vitro and in vivo. The nuclear factor kappa B (NF-κB)-mediated expression of NLRP3 inflammasome components in response to HS treatment was determined to decipher the role of HS in transcriptional priming of NLRP3 inflammasome. Furthermore, HS-triggered deubiquitination of NLRP3 was analyzed to reveal the promoting effect of HS on the NLRP3 inflammasome priming via a non-transcriptional pathway. RESULTS: High plasma level of HS was observed with a positive correlation to that of inflammatory cytokines in AP mice. Administration of HS to mice resulted in an exacerbated inflammatory profile, while reducing HS production by an inhibitor of heparanase significantly attenuated inflammatory response. Pharmacological inhibition or genetic deletion of NLRP3 substantially suppressed the HS-stimulated elevation of IL-1ß levels in AP mice. The in vitro data demonstrated that HS primarily serves as a priming signal for the activation of the NLRP3 inflammasome. HS possesses the ability to increase the transcriptional activity of NF-κB and TLR4/NF-κB-driven transcriptional pathway is employed for NLRP3 inflammasome priming. Moreover, HS-induced deubiquitination of NLRP3 is another pathway responsible for non-transcriptional priming of NLRP3 inflammasome. CONCLUSIONS: Our current work has unveiled HS as a new activator of the NLRP3 inflammasome responsible for the secondary inflammatory cascades during the development of AP, highlighting the HS-NLRP3 pathway as a potential target for future preventive and therapeutic approaches of AP.


Sujet(s)
Modèles animaux de maladie humaine , Héparitine sulfate , Inflammasomes , Facteur de transcription NF-kappa B , Protéine-3 de la famille des NLR contenant un domaine pyrine , Pancréatite , Animaux , Mâle , Souris , Maladie aigüe , Cytokines/métabolisme , Héparitine sulfate/métabolisme , Inflammasomes/métabolisme , Inflammation/métabolisme , Souris de lignée C57BL , Infiltration par les neutrophiles/effets des médicaments et des substances chimiques , Facteur de transcription NF-kappa B/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Pancréatite/métabolisme
19.
Adv Sci (Weinh) ; 11(31): e2401423, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38884169

RÉSUMÉ

Effectively neutralizing inflammatory cytokines is crucial for managing a variety of inflammatory disorders. Current techniques that target only a subset of cytokines often fall short due to the intricate nature of redundant and compensatory cytokine networks. A promising solution to this challenge is using cell membrane-coated nanoparticles (CNPs). These nanoparticles replicate the complex interactions between cells and cytokines observed in disease pathology, providing a potential avenue for multiplex cytokine scavenging. While the development of CNPs using experimental animal models has shown great promise, their effectiveness in scavenging multiple cytokines in human diseases has yet to be demonstrated. To bridge this gap, this study selected macrophage membrane-coated CNPs (MФ-CNPs) and assessed their ability to scavenge inflammatory cytokines in serum samples from patients with COVID-19, sepsis, acute pancreatitis, or type-1 diabetes, along with synovial fluid samples from patients with rheumatoid arthritis. The results show that MФ-CNPs effectively scavenge critical inflammatory cytokines, including interleukin (IL)-6, IL-8, interferon (IFN)-γ, and tumor necrosis factor (TNF)-α, in a dose-dependent manner. Overall, this study demonstrates MФ-CNPs as a multiplex cytokine scavenging formulation with promising applications in clinical settings to treat a range of inflammatory disorders.


Sujet(s)
COVID-19 , Cytokines , Macrophages , Nanoparticules , Humains , Cytokines/métabolisme , Nanoparticules/composition chimique , COVID-19/immunologie , Macrophages/métabolisme , Macrophages/immunologie , Inflammation/métabolisme , Diabète de type 1/métabolisme , Diabète de type 1/immunologie , Sepsie/métabolisme , Sepsie/immunologie , Pancréatite/immunologie , Pancréatite/métabolisme , Mâle , Polyarthrite rhumatoïde/immunologie , Polyarthrite rhumatoïde/métabolisme , Femelle , Adulte d'âge moyen , SARS-CoV-2/immunologie
20.
Biochim Biophys Acta Mol Basis Dis ; 1870(7): 167310, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-38901651

RÉSUMÉ

The pancreas is a glandular organ with both endocrine and exocrine functions. Researchers have investigated the roles of several Rab proteins, which are major regulators of membrane trafficking, in pancreatic exocytosis of zymogen granules in exocrine cells, also known as acinar cells. However, detailed molecular mechanisms mediated by Rab proteins are not fully understood. RASEF/Rab45 is an atypical Rab GTPase that contains N-terminal EF-hand and coiled-coil domains, as well as a C-terminal Rab-GTPase domain. In this study, we investigated the in vivo role of RASEF in pancreatic acinar cells using RASEF-knockout (KO) mice. Morphological analyses revealed that pancreatic acinar cells in RASEF-KO mice had an increased number of zymogen granules and abnormal formations of organelles, such as the endoplasmic reticulum (ER) and lysosomes. Biochemical analyses showed that ER proteins were decreased, but digestive enzymes were increased in the RASEF-KO pancreas. Moreover, trypsinogen was activated and co-localized with the endo-lysosomal marker LAMP1 in RASEF-KO pancreas. Upon cerulein administration to induce acute pancreatitis, impaired enzyme release from the pancreas was observed in the serum of RASEF-KO mice. These findings suggest that RASEF likely regulates the formation and sorting of zymogen granules and secretion of digestive enzymes by pancreatic acinar cells.


Sujet(s)
Cellules acineuses , Vésicules de sécrétion , Facteur ras d'échange de nucléotides guanyliques , Animaux , Mâle , Souris , Cellules acineuses/métabolisme , Réticulum endoplasmique/métabolisme , Exocytose , Lysosomes/métabolisme , Souris knockout , Pancréas/métabolisme , Pancréas exocrine/métabolisme , Pancréatite/métabolisme , Pancréatite/anatomopathologie , Pancréatite/induit chimiquement , Transport des protéines , Protéines G rab/métabolisme , Protéines G rab/génétique , Vésicules de sécrétion/métabolisme , Trypsinogène/métabolisme , Facteur ras d'échange de nucléotides guanyliques/génétique , Facteur ras d'échange de nucléotides guanyliques/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE