Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Ahsan U. Shah is active.

Publication


Featured researches published by Ahsan U. Shah.


Gastroenterology | 2011

TLR9 and the NLRP3 Inflammasome Link Acinar Cell Death With Inflammation in Acute Pancreatitis

Rafaz Hoque; Muhammad A. Sohail; Ahsan F. Malik; Sherhayar Sarwar; Yuhuan Luo; Ahsan U. Shah; Franck J. Barrat; Richard A. Flavell; Fred S. Gorelick; Sohail Z. Husain; Wajahat Z. Mehal

BACKGROUND & AIMS Acute pancreatitis is characterized by early activation of intracellular proteases followed by acinar cell death and inflammation. Activation of damage-associated molecular pattern (DAMP) receptors and a cytosolic complex termed the inflammasome initiate forms of inflammation. In this study, we examined whether DAMP-receptors and the inflammasome provide the link between cell death and the initiation of inflammation in pancreatitis. METHODS Acute pancreatitis was induced by caerulein stimulation in wild-type mice and mice deficient in components of the inflammasome (apoptosis-associated speck-like protein containing a caspase recruitment domain [ASC], NLRP3, caspase-1), Toll-like receptor 9 (TLR9), or the purinergic receptor P2X(7). Resident and infiltrating immune cell populations and pro-interleukin-1β expression were characterized in control and caerulein-treated adult murine pancreas. TLR9 expression was quantified in pancreatic cell populations. Additionally, wild-type mice were pretreated with a TLR9 antagonist before induction of acute pancreatitis by caerulein or retrograde bile duct infusion of taurolithocholic acid 3-sulfate. RESULTS Caspase-1, ASC, and NLRP3 were required for inflammation in acute pancreatitis. Genetic deletion of Tlr9 reduced pancreatic edema, inflammation, and pro-IL-1β expression in pancreatitis. TLR9 was expressed in resident immune cells of the pancreas, which are predominantly macrophages. Pretreatment with the TLR9 antagonist IRS954 reduced pancreatic edema, inflammatory infiltrate, and apoptosis. Pretreatment with IRS954 reduced pancreatic necrosis and lung inflammation in taurolithocholic acid 3-sulfate-induced acute pancreatitis. CONCLUSIONS Components of the inflammasome, ASC, caspase-1, and NLRP3, are required for the development of inflammation in acute pancreatitis. TLR9 and P2X(7) are important DAMP receptors upstream of inflammasome activation, and their antagonism could provide a new therapeutic strategy for treating acute pancreatitis.


Journal of Pediatric Gastroenterology and Nutrition | 2009

Changing Referral Trends of Acute Pancreatitis in Children: A 12-year Single-center Analysis

Alexander Park; Sahibzada U. Latif; Ahsan U. Shah; Jianmin Tian; Steven L. Werlin; Allen Hsiao; Dinesh S. Pashankar; Vineet Bhandari; Anil B. Nagar; Sohail Z. Husain

Background:Acute pancreatitis is a painful inflammatory disorder known to occur in children. Recent reports, primarily on the basis of adult data, have suggested an increasing incidence. However, pediatric studies are limited. Objective:The study was performed to examine the frequency of acute pancreatitis in a pediatric population from 1994 to 2007 and to characterize etiologies by age subsets. Patients and Methods:In this retrospective study, cases of pancreatitis were identified by ICD-9 codes and subjected to inclusion criteria. Results:Two hundred and seventy-one cases of pancreatitis met inclusion criteria. Mean age of the subjects was 13.1 ± 5.6 years. The recurrence rate was 15.3%. Biliary disease was the most common etiology (32.6%). Acute pancreatitis cases evaluated at a single tertiary care center increased 53% between 1995 to 2000 and 2001 to 2006 (P < 0.02). However, when cases were normalized by all annual pediatric emergency department visits for all medical reasons, the increase was reduced to 22% and lost statistical significance (P = 0.16). The rise was not associated with a change in etiologies or body mass index (BMI). Conclusions:This is the first report demonstrating that an increase in pediatric pancreatitis may in part be due to growing referrals to tertiary care centers. The data on etiologies, particularly with regard to differing ages, may be helpful in managing children who present with acute pancreatitis.


Journal of Biological Chemistry | 2011

Ethanol Enhances Carbachol-induced Protease Activation and Accelerates Ca2+ Waves in Isolated Rat Pancreatic Acini

Abrahim I. Orabi; Ahsan U. Shah; Kamaldeen A. Muili; Yuhuan Luo; Asim Ahmad; Anamika Reed; Sohail Z. Husain

Alcohol abuse is a leading cause of pancreatitis, accounting for 30% of acute cases and 70–90% of chronic cases, yet the mechanisms leading to alcohol-associated pancreatic injury are unclear. An early and critical feature of pancreatitis is the aberrant signaling of Ca2+ within the pancreatic acinar cell. An important conductor of this Ca2+ is the basolaterally localized, intracellular Ca2+ channel ryanodine receptor (RYR). In this study, we examined the effect of ethanol on mediating both pathologic intra-acinar protease activation, a precursor to pancreatitis, as well as RYR Ca2+ signals. We hypothesized that ethanol sensitizes the acinar cell to protease activation by modulating RYR Ca2+. Acinar cells were freshly isolated from rat, pretreated with ethanol, and stimulated with the muscarinic agonist carbachol (1 μm). Ethanol caused a doubling in the carbachol-induced activation of the proteases trypsin and chymotrypsin (p < 0.02). The RYR inhibitor dantrolene abrogated the enhancement of trypsin and chymotrypsin activity by ethanol (p < 0.005 for both proteases). Further, ethanol accelerated the speed of the apical to basolateral Ca2+ wave from 9 to 18 μm/s (p < 0.0005; n = 18–22 cells/group); an increase in Ca2+ wave speed was also observed with a change from physiologic concentrations of carbachol (1 μm) to a supraphysiologic concentration (1 mm) that leads to protease activation. Dantrolene abrogated the ethanol-induced acceleration of wave speed (p < 0.05; n = 10–16 cells/group). Our results suggest that the enhancement of pathologic protease activation by ethanol is dependent on the RYR and that a novel mechanism for this enhancement may involve RYR-mediated acceleration of Ca2+ waves.


Journal of Biological Chemistry | 2011

Low Extracellular pH Induces Damage in the Pancreatic Acinar Cell by Enhancing Calcium Signaling

Anamika Reed; Sohail Z. Husain; Edwin C. Thrower; Martine Alexandre; Ahsan U. Shah; Fred S. Gorelick; Michael H. Nathanson

Low extracellular pH (pHe) occurs in a number of clinical conditions and sensitizes to the development of pancreatitis. The mechanisms responsible for this sensitization are unknown. Because abnormal Ca2+ signaling underlies many of the early steps in the pathogenesis of pancreatitis, we evaluated the effect of decreasing pHe from 7.4 to 7.0 on Ca2+ signals in the acinar cell. Low pHe significantly increased the amplitude of cerulein-induced Ca2+ signals. The enhancement in amplitude was localized to the basolateral region of the acinar cell and was reduced by pretreatment with ryanodine receptor (RYR) inhibitors. Because basolateral RYRs also have been implicated in the pathogenesis of pancreatitis, we evaluated the effects of RYR inhibitors on pancreatitis responses in acidic conditions. RYR inhibitors significantly reduced the sensitizing effects of low pHe on zymogen activation and cellular injury. These findings suggest that enhanced RYR-mediated Ca2+ signaling in the basolateral region of the acinar cell is responsible for the injurious effects of low pHe on the exocrine pancreas.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2010

Dantrolene mitigates caerulein-induced pancreatitis in vivo in mice.

Abrahim I. Orabi; Ahsan U. Shah; Mahwish U. Ahmad; Rayman Choo-Wing; Jerome Parness; Dhanpat Jain; Vineet Bhandari; Sohail Z. Husain

Acute pancreatitis is a painful, inflammatory disorder for which adequate treatments are lacking. An early, critical step in its development is the aberrant signaling of Ca(2+) within the pancreatic acinar cell. This Ca(2+) release is modulated by the intracellular Ca(2+) channel the ryanodine receptor (RYR). We have previously shown that RYR inhibition reduces pathological intra-acinar protease activation, an early marker of pancreatitis. In this study, we examined whether pretreatment with the RYR inhibitor dantrolene attenuates the severity of caerulein-induced pancreatitis in mice. Immunofluorescent labeling for RYR from mouse pancreatic sections showed localization to the basolateral region of the acinar cell. After 1 h of caerulein hyperstimulation in vivo, dantrolene 1) reduced pancreatic trypsin activity by 59% (P < 0.05) and 2) mitigated early ultrastructural derangements within the acinar cell. Eight hours after pancreatitis induction, dantrolene reduced pancreatic trypsin activity and serum amylase by 61 and 32%, respectively (P < 0.05). At this later time point, overall histological severity of pancreatitis was reduced by 63% with dantrolene pretreatment (P < 0.05). TUNEL-positive cells were reduced by 58% (P < 0.05). These data suggest that the RYR plays an important role in mediating early acinar cell events during in vivo pancreatitis and contributes to disease severity. Blockade of Ca(2+) signals and particularly RYR-Ca(2+) may be useful as prophylactic treatment for this disease in high-risk settings for pancreatitis.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2012

Pharmacological and genetic inhibition of calcineurin protects against carbachol-induced pathological zymogen activation and acinar cell injury

Kamaldeen A. Muili; Mahwish U. Ahmad; Abrahim I. Orabi; Ahsan U. Shah; Jeffery D. Molkentin; Sohail Z. Husain

Acute pancreatitis is a major health burden for which there are currently no targeted therapies. Premature activation of digestive proenzymes, or zymogens, within the pancreatic acinar cell is an early and critical event in this disease. A high-amplitude, sustained rise in acinar cell Ca(2+) is required for zymogen activation. We previously showed in a cholecystokinin-induced pancreatitis model that a potential target of this aberrant Ca(2+) signaling is the Ca(2+)-activated phosphatase calcineurin (Cn). However, in this study, we examined the role of Cn on both zymogen activation and injury, in the clinically relevant condition of neurogenic stimulation (by giving the acetylcholine analog carbachol) using three different Cn inhibitors or Cn-deficient acinar cells. In freshly isolated mouse acinar cells, pretreatment with FK506, calcineurin inhibitory peptide (CiP), or cyclosporine (CsA) blocked intra-acinar zymogen activation (n = 3; P < 0.05). The Cn inhibitors also reduced leakage of lactate dehydrogenase (LDH) by 79%, 62%, and 63%, respectively (n = 3; P < 0.05). Of the various Cn isoforms, the β-isoform of the catalytic A subunit (CnAβ) was strongly expressed in mouse acinar cells. For this reason, we obtained acinar cells from CnAβ-deficient mice (CnAβ-/-) and observed an 84% and 50% reduction in trypsin and chymotrypsin activation, respectively, compared with wild-type controls (n = 3; P < 0.05). LDH release in the CnAβ-deficient cells was reduced by 50% (n = 2; P < 0.05). The CnAβ-deficient cells were also protected against zymogen activation and cell injury induced by the cholecystokinin analog caerulein. Importantly, amylase secretion was generally not affected by either the Cn inhibitors or Cn deficiency. These data provide both pharmacological and genetic evidence that implicates Cn in intra-acinar zymogen activation and cell injury during pancreatitis.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2014

The ryanodine receptor is expressed in human pancreatic acinar cells and contributes to acinar cell injury

Christopher M. Lewarchik; Abrahim I. Orabi; Shunqian Jin; Dong Wang; Kamaldeen Muili; Ahsan U. Shah; John F. Eisses; Adeel Malik; Rita Bottino; Thottala Jayaraman; Sohail Z. Husain

Physiological calcium (Ca(2+)) signals within the pancreatic acinar cell regulate enzyme secretion, whereas aberrant Ca(2+) signals are associated with acinar cell injury. We have previously identified the ryanodine receptor (RyR), a Ca(2+) release channel on the endoplasmic reticulum, as a modulator of these pathological signals. In the present study, we establish that the RyR is expressed in human acinar cells and mediates acinar cell injury. We obtained pancreatic tissue from cadaveric donors and identified isoforms of RyR1 and RyR2 by qPCR. Immunofluorescence staining of the pancreas showed that the RyR is localized to the basal region of the acinar cell. Furthermore, the presence of RyR was confirmed from isolated human acinar cells by tritiated ryanodine binding. To determine whether the RyR is functionally active, mouse or human acinar cells were loaded with the high-affinity Ca(2+) dye (Fluo-4 AM) and stimulated with taurolithocholic acid 3-sulfate (TLCS) (500 μM) or carbachol (1 mM). Ryanodine (100 μM) pretreatment reduced the magnitude of the Ca(2+) signal and the area under the curve. To determine the effect of RyR blockade on injury, human acinar cells were stimulated with pathological stimuli, the bile acid TLCS (500 μM) or the muscarinic agonist carbachol (1 mM) in the presence or absence of the RyR inhibitor ryanodine. Ryanodine (100 μM) caused an 81% and 47% reduction in acinar cell injury, respectively, as measured by lactate dehydrogenase leakage (P < 0.05). Taken together, these data establish that the RyR is expressed in human acinar cells and that it modulates acinar Ca(2+) signals and cell injury.


Gastroenterology | 2010

1023 Alcohol Predisposes to Pathologic Calcium Signals in Pancreatic Acinar Cells by Hyper-Phosphorylating Ryanodine Receptors

Abrahim I. Orabi; Ahsan U. Shah; Zahir Mannan; Mahwish U. Ahmad; Xingtie Nie; Sohail Z. Husain

Alcohol abuse is a leading cause of pancreatitis, accounting for 30% of acute and up to 90% of chronic cases, yet the mechanisms leading to alcohol-induced injury remain elusive. An early and critical feature of this disease is the aberrant signaling of Ca2+ within the pancreatic acinar cell. An important conductor of this Ca2+ release is the basolaterally localized, intracellular Ca2+ channel the ryanodine receptor (RYR). In this study, we examined the role of the RYR in mediating Ca2+ signals during alcohol exposure. We hypothesized that alcohol triggers the release of Ca2+ from pathologically leaky, hyper-phosphorylated RYRs. Acinar cells were freshly isolated from rat, loaded with the Ca2+ dye fluo-4, and imaged by time lapse confocal microscopy. Spatial and temporal changes in Ca2+ release were examined after perfusion with the Ca2+-activating agonist carbachol (1 uM) with or without an ethanol concentration clinically achievable after heavy intoxication (100 mM). Ethanol accelerated the speed of the apical to basolateral Ca2+ wave from 9 to 18 um/sec (P<0.0005; n=18-22 cells/group). Ethanol also caused a similar doubling in intra-cellular cAMP levels from 1.5 to 3 fmoles. Acceleration of the acinar cell Ca2+ wave by alcohol was abrogated by the PKA inhibitor PKI (1 uM; P<0.05; n=10-16 cells/group). Using an epitope-specific phospho-antibody that recognizes the PKA site on the RYR, we found that ethanol increased RYR phosphorylation by nearly five-fold (P<0.05). Finally, the alcohol-induced acceleration of Ca2+ wave speed was completely reduced by the RYR inhibitor dantrolene (100 uM; P<0.05; n=10-16 cells/group). In summary, our results have for the first time implicated a pathological role for RYR-Ca2+ release in the predisposition to alcohol-induced pancreatitis via cAMP/PKA hyper-phosphorylation of the RYR.


Pancreas | 2006

CAERULEIN-INDUCED INTRACELLULAR PANCREATIC ZYMOGEN ACTIVATION IS DEPENDENT UPON CALCINEURIN

Wayne M. Grant; Ahsan U. Shah; Edwin C. Thrower; Sohail Z. Husain

Aberrant cytosolic Ca(2+) flux in pancreatic acinar cells is critical to the pathological pancreatic zymogen activation observed in acute pancreatitis, but the downstream effectors are not known. In this study, we examined the role of Ca(2+)-activated protein phosphatase 2B (or calcineurin) in zymogen activation. Isolated pancreatic acinar cells were stimulated with supraphysiological caerulein (100 nM) with or without the calcineurin inhibitors FK506 or cell-permeable calcineurin inhibitory peptide (CiP). Chymotrypsin activity was measured as a marker of zymogen activation, and the percent amylase secretion was used as a measure of enzyme secretion. Cytosolic Ca(2+) changes were recorded in acinar cells loaded with the intermediate Ca(2+)-affinity dye fluo-5F using a scanning confocal microscope. A 50% reduction in chymotrypsin activity was observed after pretreatment with 1 microM FK506 or 10 microM CiP. These pretreatments did not affect amylase secretion or the rise in cytosolic Ca(2+) after caerulein stimulation. These findings suggest that calcineurin mediates caerulein-induced intra-acinar zymogen activation but not enzyme secretion or the initial caerulein-induced cytosolic Ca(2+) signal.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2007

Caerulein-induced intracellular pancreatic zymogen activation is dependent on calcineurin

Sohail Z. Husain; Wayne M. Grant; Fred S. Gorelick; Michael H. Nathanson; Ahsan U. Shah

Collaboration


Dive into the Ahsan U. Shah's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge