Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Transl Med ; 19(1): 466, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34781960

ABSTRACT

BACKGROUND: Abdominal aortic aneurysm (AAA), an irreversible cardiovascular disease prevalent in the artery, causes the increase of the aneurysm diameter over time, and is a fatal phenomenon inducing sidewall rupture. Long noncoding RNAs (lncRNAs) serve as promising biomarkers for AAA. In the present study, we sought to define the role of lncRNA growth-arrest-specific transcript 5 (GAS5) in growth of smooth muscle cells (SMC) and progression of AAA. METHODS: Initially, we established angiotensin II (Ang II)-induced AAA mouse models and Ang II-treated vascular SMC model. RT-qPCR and Western blot analysis were adopted to determine expression of GAS5 and zeste homolog 2 (EZH2). After ectopic expression and depletion experiments in Ang II-treated mice and vascular SMCs, cell apoptosis was detected in SMCs using flow cytometry and in mice using TUNEL staining. The binding of GAS5 and EZH2 was evaluated using RNA binding protein immunoprecipitation (RIP) and Co-IP assays. RESULTS: Increased GAS5 and RIG-I but decreased EZH2 were found in aortic tissues of AAA mice. EZH2 overexpression inhibited AAA formation and suppressed SMC apoptosis. Functionally, EZH2 blocked the RIG-I signaling pathway and consequently inhibited SMC apoptosis. GAS5 regulated EZH2 transcription in a negative manner in SMCs. Knockdown of GAS5 attenuated SMC apoptosis, which was reversed by EZH2 inhibition or RIG-I overexpression. CONCLUSIONS: The current study demonstrated that GAS5 induced SMC apoptosis and subsequent AAA onset by activating EZH2-mediated RIG-I signaling pathway, highlighting GAS5 as a novel biomarker for AAA.


Subject(s)
Aortic Aneurysm, Abdominal , RNA, Long Noncoding , Angiotensin II , Animals , Aortic Aneurysm, Abdominal/genetics , Apoptosis/genetics , Mice , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , RNA, Long Noncoding/genetics , Signal Transduction
2.
Cell Mol Gastroenterol Hepatol ; 3(1): 119-128, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28090570

ABSTRACT

BACKGROUND AND AIMS: There is a pressing need to develop effective preventative therapies for post-ERCP pancreatitis (PEP). We demonstrated that early PEP events are induced through the calcium-activated phosphatase calcineurin and that global calcineurin deletion abolishes PEP in mice. A crucial question is whether acinar cell calcineurin controls the initiation of PEP in vivo. METHODS: We used a mouse model of PEP and examined the effects of in vivo acinar cell-specific calcineurin deletion by either generating a conditional knockout line or infusing a novel AAV-Ela-iCre into the pancreatic duct of a calcineurin floxed line. RESULTS: We found that PEP is dependent on acinar cell calcineurin in vivo, and this led us to determine that calcineurin inhibitors, infused within the radiocontrast, can largely prevent PEP. CONCLUSIONS: These results provide impetus for launching clinical trials to test the efficacy of intraductal calcineurin inhibitors to prevent PEP.

3.
Vascular ; 25(4): 442-446, 2017 Aug.
Article in English | MEDLINE | ID: mdl-27928064

ABSTRACT

Objective To describe a novel approach, 3D printing guiding stent graft fenestration, for fenestration during endovascular aneurysm repair for juxtarenal abdominal aortic aneurysm. Methods A 69-year-old male with juxtarenal abdominal aortic aneurysm underwent endovascular aneurysm repair with "off the label" fenestrated stent graft. To precisely locate the fenestration position, we reconstructed a 3D digital abdominal aortic aneurysm model and created a skin template covering this abdominal aortic aneurysm model. Then the skin template was physically printed and the position of the visceral vessel was hollowed out, thereby helping in locating the fenestration on stent graft. Results and conclusions With the help of this 3D printed skin template, we fenestrated the stent graft accurately and rebuilt the bilateral renal artery successfully. This is the first clinical case that used 3D printing guiding stent graft fenestration, which is a novel approach for precise fenestration on stent graft on the table during endovascular aneurysm repair.


Subject(s)
Aortic Aneurysm, Abdominal/surgery , Blood Vessel Prosthesis Implantation/instrumentation , Blood Vessel Prosthesis , Endovascular Procedures/instrumentation , Models, Cardiovascular , Patient-Specific Modeling , Printing, Three-Dimensional , Stents , Aged , Anatomic Landmarks , Aortic Aneurysm, Abdominal/diagnostic imaging , Aortography/methods , Computed Tomography Angiography , Humans , Male , Prosthesis Design , Renal Artery/diagnostic imaging , Renal Artery/surgery , Treatment Outcome
4.
Gastroenterology ; 149(3): 753-64.e11, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25980752

ABSTRACT

BACKGROUND & AIMS: Radiocontrast agents are required for radiographic procedures, but these agents can injure tissues by unknown mechanisms. We investigated whether exposure of pancreatic tissues to radiocontrast agents during endoscopic retrograde cholangiopancreatography (ERCP) causes pancreatic inflammation, and studied the effects of these agents on human cell lines and in mice. METHODS: We exposed mouse and human acinar cells to the radiocontrast agent iohexol (Omnipaque; GE Healthcare, Princeton, NJ) and measured intracellular release of Ca(2+), calcineurin activation (using a luciferase reporter), activation of nuclear factor-κB (NF-κB, using a luciferase reporter), and cell necrosis (via propidium iodide uptake). We infused the radiocontrast agent into the pancreatic ducts of wild-type mice (C57BL/6) to create a mouse model of post-ERCP pancreatitis; some mice were given intraperitoneal injections of the calcineurin inhibitor FK506 before and after infusion of the radiocontrast agent. CnAß(-/-) mice also were used. This experiment also was performed in mice given infusions of adeno-associated virus 6-NF-κB-luciferase, to assess activation of this transcription factor in vivo. RESULTS: Incubation of mouse and human acinar cells, but not HEK293 or COS7 cells, with iohexol led to a peak and then plateau in Ca(2+) signaling, along with activation of the transcription factors NF-κB and nuclear factor of activated T cells. Suppressing Ca(2+) signaling or calcineurin with BAPTA, cyclosporine A, or FK506 prevented activation of NF-κB and acinar cell injury. Calcineurin Aß-deficient mice were protected against induction of pancreatic inflammation by iohexol. The calcineurin inhibitor FK506 prevented contrast-induced activation of NF-κB in pancreata of mice, this was observed by live imaging of mice given infusions of adeno-associated virus 6-NF-κB-luciferase. CONCLUSIONS: Radiocontrast agents cause pancreatic inflammation in mice, via activation of NF-κB, Ca(2+) signaling, and calcineurin. Calcineurin inhibitors might be developed to prevent post-ERCP pancreatitis in patients.


Subject(s)
Calcineurin/metabolism , Calcium Signaling , Contrast Media , Iohexol , NF-kappa B/metabolism , Pancreas, Exocrine/enzymology , Pancreatitis/enzymology , Animals , COS Cells , Calcineurin/deficiency , Calcineurin/genetics , Calcineurin Inhibitors/pharmacology , Calcium Signaling/drug effects , Chlorocebus aethiops , Disease Models, Animal , Gene Expression Regulation , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/genetics , Necrosis , Pancreas, Exocrine/drug effects , Pancreas, Exocrine/pathology , Pancreatitis/chemically induced , Pancreatitis/genetics , Pancreatitis/pathology , Pancreatitis/prevention & control , Tacrolimus/pharmacology , Time Factors
5.
Pancreas ; 44(3): 493-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25469547

ABSTRACT

OBJECTIVE: The most common etiology of acute pancreatitis results from the impaction of gallstones or sludge in the distal common bile duct (CBD). The result is pancreatic duct obstruction, diversion of bile into the pancreas, or cholestasis. In the current study, we examined whether combining both aspects, that is, infusion of the bile acid taurocholate (TC) followed by bile duct ligation (BDL), could yield a more severe form of pancreatitis that mimics biliary pancreatitis. METHODS: In mice, after laparotomy, the CBD was infused with either normal saline (NS) or TC. Subsequently, the CBD was ligated at the ampulla. RESULTS: Mice receiving TC infusion followed by BDL (TC + BDL) had higher mortality compared with animals receiving intraductal NS with BDL (NS + BDL). The TC + BDL arm developed more severe and diffuse pancreatic necrosis. In addition, serum amylase, IL-6, and bilirubin were significantly higher. However, pancreatic edema as well as lung and liver injury were unchanged between TC + BDL and NS + BDL. CONCLUSIONS: In summary, the combination of bile infusion into the pancreas followed by BDL causes a more severe, necrotizing pancreatitis. We believe that this novel model of pancreatitis is useful because it can be used in transgenic mice and recapitulates several aspects of biliary pancreatitis.


Subject(s)
Cholestasis/complications , Common Bile Duct/surgery , Gallstones/chemically induced , Pancreatitis, Acute Necrotizing/chemically induced , Taurocholic Acid , Amylases/blood , Animals , Bilirubin/blood , Biomarkers/blood , Disease Models, Animal , Interleukin-6/blood , Ligation , Male , Mice , Pancreas/enzymology , Pancreas/pathology , Pancreatitis, Acute Necrotizing/blood , Pancreatitis, Acute Necrotizing/pathology , Severity of Illness Index , Time Factors
6.
J Biol Chem ; 288(29): 21065-21073, 2013 Jul 19.
Article in English | MEDLINE | ID: mdl-23744075

ABSTRACT

Biliary pancreatitis is the most common etiology of acute pancreatitis, accounting for 30-60% of cases. A dominant theory for the development of biliary pancreatitis is the reflux of bile into the pancreatic duct and subsequent exposure to pancreatic acinar cells. Bile acids are known to induce aberrant Ca(2+) signals in acinar cells as well as nuclear translocation of NF-κB. In this study, we examined the role of the downstream Ca(2+) target calcineurin on NF-κB translocation. Freshly isolated mouse acinar cells were infected for 24 h with an adenovirus expressing an NF-κB luciferase reporter. The bile acid taurolithocholic acid-3-sulfate caused NF-κB activation at concentrations (500 µm) that were associated with cell injury. We show that the NF-κB inhibitor Bay 11-7082 (1 µm) blocked translocation and injury. Pretreatment with the Ca(2+) chelator 1,2-bis(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid, the calcineurin inhibitors FK506 and cyclosporine A, or use of acinar cells from calcineurin Aß-deficient mice each led to reduced NF-κB activation with taurolithocholic acid-3-sulfate. Importantly, these manipulations did not affect LPS-induced NF-κB activation. A critical upstream regulator of NF-κB activation is protein kinase C, which translocates to the membranes of various organelles in the active state. We demonstrate that pharmacologic and genetic inhibition of calcineurin blocks translocation of the PKC-δ isoform. In summary, bile-induced NF-κB activation and acinar cell injury are mediated by calcineurin, and a mechanism for this important early inflammatory response appears to be upstream at the level of PKC translocation.


Subject(s)
Acinar Cells/metabolism , Bile Acids and Salts/pharmacology , Calcineurin/metabolism , NF-kappa B/metabolism , Pancreas/pathology , Acinar Cells/drug effects , Acinar Cells/pathology , Animals , Humans , Lipopolysaccharides/pharmacology , Male , Mice , Models, Biological , Protein Kinase C-delta/metabolism , Protein Transport/drug effects , Rats , Taurolithocholic Acid/analogs & derivatives , Taurolithocholic Acid/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...