Network


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

Hotspot


Dive into the research topics where Rajinder Dawra is active.

Publication


Featured researches published by Rajinder Dawra.


Cancer Research | 2007

Triptolide Induces Pancreatic Cancer Cell Death via Inhibition of Heat Shock Protein 70

Phoebe A. Phillips; Vikas Dudeja; Joshua A. McCarroll; Daniel Borja-Cacho; Rajinder Dawra; William E. Grizzle; Selwyn M. Vickers; Ashok K. Saluja

Pancreatic cancer is highly resistant to current chemotherapy agents. We therefore examined the effects of triptolide (a diterpenoid triepoxide) on pancreatic cancer growth and local-regional tumor spread using an orthotopic model of pancreatic cancer. We have recently shown that an increased level of HSP70 in pancreatic cancer cells confers resistance to apoptosis and that inhibiting HSP70 induces apoptosis in these cells. In addition, triptolide was recently identified as part of a small molecule screen, as a regulator of the human heat shock response. Therefore, our aims were to examine the effects of triptolide on (a) pancreatic cancer cells by assessing viability and apoptosis, (b) pancreatic cancer growth and local invasion in vivo, and (c) HSP70 levels in pancreatic cancer cells. Incubation of PANC-1 and MiaPaCa-2 cells with triptolide (50-200 nmol/L) significantly reduced cell viability, but had no effect on the viability of normal pancreatic ductal cells. Triptolide induced apoptosis (assessed by Annexin V, caspase-3, and terminal nucleotidyl transferase-mediated nick end labeling) and decreased HSP70 mRNA and protein levels in both cell lines. Triptolide (0.2 mg/kg/d for 60 days) administered in vivo decreased pancreatic cancer growth and significantly decreased local-regional tumor spread. The control group of mice had extensive local invasion into adjacent organs, including the spleen, liver, kidney, and small intestine. Triptolide causes pancreatic cancer cell death in vitro and in vivo by induction of apoptosis and its mechanism of action is mediated via the inhibition of HSP70. Triptolide is a potential therapeutic agent that can be used to prevent the progression and metastases of pancreatic cancer.


Cancer Research | 2007

Heat Shock Protein 70 Increases Tumorigenicity and Inhibits Apoptosis in Pancreatic Adenocarcinoma

Ali Aghdassi; Phoebe A. Phillips; Vikas Dudeja; D. Dhaulakhandi; R. Sharif; Rajinder Dawra; Markus M. Lerch; Ashok K. Saluja

Pancreatic carcinoma is a malignant disease that responds poorly to chemotherapy because of its resistance to apoptosis. Heat shock proteins (Hsp) are not only cytoprotective but also interfere with the apoptotic cascade. Here, we investigated the role of Hsp70 in regulating apoptosis in pancreatic cancer cells. Hsp70 expression was increased in pancreatic cancer cells compared with normal pancreatic ductal cells. This was confirmed by increased mRNA levels for Hsp70 in human pancreatic cancer tissue compared with neighboring normal tissue from the same patient. Depletion of Hsp70 by quercetin decreased cell viability and induced apoptosis in cancer cells but not in normal pancreatic ductal cells. To show that this is a specific effect of Hsp70 on apoptosis, levels of Hsp70 were knocked down by short interfering RNA treatment, which also induced apoptosis in cancer cells as indicated by Annexin V staining and caspase activation. Daily administration of quercetin to nude mice decreased tumor size as well as Hsp70 levels in tumor tissue. These findings indicate that Hsp70 plays an important role in apoptosis and that selective Hsp70 knockdown can be used to induce apoptosis in pancreatic cancer cells.


Gastroenterology | 1993

Pancreatic duct obstruction triggers acute necrotizing pancreatitis in the opossum

Markus M. Lerch; Ashok K. Saluja; Michael Rünzi; Rajinder Dawra; Manju Saluja; Michael L. Steer

BACKGROUND The common channel theory suggests that bile reflux, through a common biliopancreatic channel, triggers acute pancreatitis. In the present study, this controversial issue was evaluated using an experimental model of hemorrhagic necrotizing pancreatitis. METHODS American opossums underwent ligation of the pancreatic duct alone, bile and pancreatic duct separately, or common biliopancreatic duct; the severity of pancreatitis was evaluated at selected times after ligation. RESULTS Animals in all three experimental groups developed hemorrhagic necrotizing pancreatitis; the severity of pancreatitis was similar in each group, although only those subjected to common biliopancreatic duct ligation experienced bile reflux. CONCLUSIONS Bile reflux into the pancreatic duct, via a common biliopancreatic channel, is not necessary for the development of pancreatitis and does not worsen the severity of pancreatitis associated with pancreatic duct obstruction in this model.


Science Translational Medicine | 2012

A Preclinical Evaluation of Minnelide as a Therapeutic Agent Against Pancreatic Cancer

Rohit Chugh; Veena Sangwan; Satish Patil; Vikas Dudeja; Rajinder Dawra; Sulagna Banerjee; Robert J. Schumacher; Bruce R. Blazar; Gunda I. Georg; Selwyn M. Vickers; Ashok K. Saluja

Minnelide prevents tumor formation, causes tumor regression, and increases survival in multiple models of pancreatic cancer. Vegetation Is Good for You Your mom always told you to eat your vegetables, but what she probably didn’t tell you is that other plants can be good for you as well. Tripterygium wilfordii, sometimes known as the Thunder God vine, has various uses in traditional Chinese medicine. To better understand and improve upon the healing properties of this vine, the active ingredients have been isolated and characterized. One component of T. wilfordii, triptolide, has shown promising effects against pancreatic cancer cells. New therapies for pancreatic cancer—which is one of the most lethal human malignancies—are desperately needed, but triptolide is poorly soluble in water and thus has limited clinical use. Now, Chugh et al. synthesize a water-soluble form of triptolide, Minnelide, and demonstrate efficacy against pancreatic cancer in multiple animal models. The authors tested Minnelide both in vitro and in multiple preclinical models of pancreatic cancer. Each model has distinct advantages and limitations: Well-studied cancer cell lines and translationally relevant patient tumors were transplanted into mice that lack immune systems, whereas a spontaneous model in immunosufficient mice was, by necessity, a mouse tumor. By combining these approaches, the authors addressed many caveats that frequently plague preclinical studies. Indeed, Minnelide was highly effective in treating pancreatic cancer in all of these complementary models. The next step is to take Minnelide into early clinical trials to see if these results can be reproduced in human patients with pancreatic cancer. Pancreatic cancer is one of the most lethal human malignancies with an all-stage 5-year survival frequency of <5%, which highlights the urgent need for more effective therapeutic strategies. We have previously shown that triptolide, a diterpenoid, is effective against pancreatic cancer cells in vitro as well as in vivo. However, triptolide is poorly soluble in water, limiting its clinical use. We therefore synthesized a water-soluble analog of triptolide, named Minnelide. The efficacy of Minnelide was tested both in vitro and in multiple independent yet complementary in vivo models of pancreatic cancer: an orthotopic model of pancreatic cancer using human pancreatic cancer cell lines in athymic nude mice, a xenograft model where human pancreatic tumors were transplanted into severe combined immunodeficient mice, and a spontaneous pancreatic cancer mouse model (KRasG12D; Trp53R172H; Pdx-1Cre). In these multiple complementary models of pancreatic cancer, Minnelide was highly effective in reducing pancreatic tumor growth and spread, and improving survival. Together, our results suggest that Minnelide shows promise as a potent chemotherapeutic agent against pancreatic cancer, and support the evaluation of Minnelide in clinical trials against this deadly disease.


Gastroenterology | 1992

Acute necrotizing pancreatitis in the opossum : earliest morphological changes involve acinar cells

Markus M. Lerch; Ashok K. Saluja; Rajinder Dawra; P. Ramarao; Manju Saluja; Michael L. Steer

Acute pancreatitis was induced by ligating the opossum common biliopancreatic duct immediately proximal to its entry into the duodenum, and macroscopic as well as microscopic changes were evaluated during the subsequent 24 hours. Transient pancreatic edema and progressive hyperamylasemia were noted within 6 hours of pancreatic and bile duct ligation. Light microscopic evidence of pancreatic injury including acinar cell necrosis, hemorrhage, fat necrosis, and inflammatory cell infiltration was noted within 12 hours of duct obstruction. Electron microscopic changes included massive dilatation of the rough endoplasmic reticulum and disruption of the apical plasmalemma of acinar cells during the initial 3 hours. These observations indicate that pancreatic and bile duct ligation in the opossum results in the rapid (less than 24 hours) appearance of changes consistent with acute hemorrhagic and necrotizing pancreatitis and that the initial lesion in this model of experimental pancreatitis involves acinar cells.


Gastroenterology | 2011

Intra-acinar trypsinogen activation mediates early stages of pancreatic injury but not inflammation in mice with acute pancreatitis.

Rajinder Dawra; Raghuwansh P. Sah; Vikas Dudeja; Loveena Rishi; Rupjoyti Talukdar; Pramod Kumar Garg; Ashok K. Saluja

BACKGROUND & AIMS The role of trypsinogen activation in the pathogenesis of acute pancreatitis (AP) has not been clearly established. METHODS We generated and characterized mice lacking trypsinogen isoform 7 (T7) gene (T(-/-)). The effects of pathologic activation of trypsinogen were studied in these mice during induction of AP with cerulein. Acinar cell death, tissue damage, early intra-acinar activation of the transcription factor nuclear factor κB (NF-κB), and local and systemic inflammation were compared between T(-/-) and wild-type mice with AP. RESULTS Deletion of T7 reduced the total trypsinogen content by 60% but did not affect physiologic function. T(-/-) mice lacked pathologic activation of trypsinogen, which occurs within acinar cells during early stages of AP progression. Absence of trypsinogen activation in T(-/-) mice led to near complete inhibition of acinar cell death in vitro and a 50% reduction in acinar necrosis during AP progression. However, T(-/-) mice had similar degrees of local and systemic inflammation during AP progression and comparable levels of intra-acinar NF-κB activation, which was previously shown to occur concurrently with trypsinogen activation during early stages of pancreatitis. CONCLUSIONS T7 is activated during pathogenesis of AP in mice. Intra-acinar trypsinogen activation leads to acinar death during early stages of pancreatitis, which accounts for 50% of the pancreatic damage in AP. However, progression of local and systemic inflammation in AP does not require trypsinogen activation. NF-κB is activated early in acinar cells, independently of trypsinogen activation, and might be responsible for progression of AP.


Gastroenterology | 2010

Triptolide induces cell death in pancreatic cancer cells by apoptotic and autophagic pathways

N. Mujumdar; T.N. MacKenzie; Vikas Dudeja; Rohit Chugh; Mara B. Antonoff; Daniel Borja-Cacho; Veena Sangwan; Rajinder Dawra; Selwyn M. Vickers; Ashok K. Saluja

BACKGROUND & AIMS Pancreatic adenocarcinoma, among the most lethal human malignancies, is resistant to current chemotherapies. We previously showed that triptolide inhibits the growth of pancreatic cancer cells in vitro and prevents tumor growth in vivo. This study investigates the mechanism by which triptolide kills pancreatic cancer cells. METHODS Cells were treated with triptolide and viability and caspase-3 activity were measured using colorimetric assays. Annexin V, propidium iodide, and acridine orange staining were measured by flow cytometry. Immunofluorescence was used to monitor the localization of cytochrome c and Light Chain 3 (LC3) proteins. Caspase-3, Atg5, and Beclin1 levels were down-regulated by exposing cells to their respective short interfering RNA. RESULTS We show that triptolide induces apoptosis in MiaPaCa-2, Capan-1, and BxPC-3 cells and induces autophagy in S2-013, S2-VP10, and Hs766T cells. Triptolide-induced autophagy has a pro-death effect, requires autophagy-specific genes, atg5 or beclin1, and is associated with the inactivation of the Protein kinase B (Akt)/mammalian target of Rapamycin/p70S6K pathway and the up-regulation of the Extracellular Signal-Related Kinase (ERK)1/2 pathway. Inhibition of autophagy in S2-013 and S2-VP10 cells results in cell death via the apoptotic pathway whereas inhibition of both autophagy and apoptosis rescues cell death. CONCLUSIONS This study shows that triptolide kills pancreatic cancer cells by 2 different pathways. It induces caspase-dependent apoptotic death in MiaPaCa-2, Capan-1, and BxPC-3, and induces caspase-independent autophagic death in metastatic cell lines S2-013, S2-VP10, and Hs766T, thereby making it an attractive chemotherapeutic agent against a broad spectrum of pancreatic cancers.


Current Opinion in Gastroenterology | 2013

New Insights into the Pathogenesis of Pancreatitis

Raghuwansh P. Sah; Rajinder Dawra; Ashok K. Saluja

Purpose of review In this article, we review important advances in our understanding of the mechanisms of pancreatitis. Recent findings The relative contributions of intrapancreatic trypsinogen activation and nuclear factor kappa B (NF&kgr;B) activation, the two major early independent cellular events in pancreatitis, have been investigated using novel genetic models. Trypsinogen activation has traditionally held the spotlight for many decades as the central pathogenic event of pancreatitis. However, recent experimental evidence points to the role of trypsin activation in early acinar cell damage but not in the inflammatory response of acute pancreatitis, which was shown to be induced by NF&kgr;B activation. Further, chronic pancreatitis developed independently of trypsinogen activation in the caerulein model. Sustained NF&kgr;B activation, but not persistent intra-acinar expression of active trypsin, was shown to result in chronic pancreatitis. Calcineurin-NFAT (nuclear factor of activated T-cells) signaling was shown to mediate downstream effects of pathologic rise in intracellular calcium. Interleukin-6 was identified as a key cytokine mediating pancreatitis-associated lung injury. Summary Recent advances challenge the long-believed trypsin-centered understanding of pancreatitis. It is becoming increasingly clear that activation of intense inflammatory signaling mechanisms in acinar cells is crucial to the pathogenesis of pancreatitis, which may explain the strong systemic inflammatory response in pancreatitis.


Gut | 2009

Impact of Toll-like Receptor 4 on the Severity of Acute Pancreatitis and Pancreatitis-Associated Lung Injury in Mice

R. Sharif; Rajinder Dawra; Karen R. Wasiluk; Phoebe A. Phillips; Vikas Dudeja; Evelyn A. Kurt-Jones; Robert W. Finberg; Ashok K. Saluja

Background and Aims: Acute pancreatitis is an inflammatory disease involving acinar cell injury, and the rapid production and release of inflammatory cytokines, which play a dominant role in local pancreatic inflammation and systemic complications. Toll-like receptor 4 (TLR4) initiates a complex signalling pathway when it interacts with lipopolysaccharide (LPS), which ultimately results in a proinflammatory response. We hypothesised that TLR4 is important in the pathophysiology of acute pancreatitis, independently of LPS. Using two different models of acute pancreatitis, we investigated how genetic deletion of TLR4 or its co-receptor CD14 effects its progression and severity. Methods: We induced acute pancreatitis by administering either caerulein or l-arginine to wild-type, TLR4−/−, and CD14−/− mice. Control mice received normal saline injections. The severity of acute pancreatitis was determined by measuring serum amylase activity, quantifying myeloperoxidase (MPO) activity in the pancreatic tissue, and histologically assessing acinar cell injury. Results: It was found that administering caerulein and l-arginine to wild-type mice resulted in acute pancreatitis (as assessed by hyperamylasaemia, oedema, increased pancreatic MPO activity, and pancreatic necrosis) and associated lung injury. The same treatment to TLR4−/− or CD14−/− mice resulted in significantly less severe acute pancreatitis, and reduced lung injury. We found no evidence of either bacteria or LPS in the blood or in pancreatic tissue. Conclusions: The severity of acute pancreatitis is ameliorated in mice that lack either TLR4 or CD14 receptors. Furthermore, these results indicate that TLR4 plays a significant pro-inflammatory role independently of LPS in the progression of acute pancreatitis.


Gastroenterology | 2009

Heat Shock Protein 70 Inhibits Apoptosis in Cancer Cells Through Simultaneous and Independent Mechanisms

Vikas Dudeja; N. Mujumdar; Phoebe A. Phillips; Rohit Chugh; Daniel Borja–Cacho; Rajinder Dawra; Selwyn M. Vickers; Ashok K. Saluja

BACKGROUND & AIMS Heat shock proteins (HSPs) are highly conserved and serve a multitude of functions that mediate cell survival. HSP70, the only inducible form of the 70-kilodalton subfamily of HSPs, is overexpressed in pancreatic cancer cells and has been shown to inhibit caspase-dependent apoptosis. We aimed to elucidate the mechanism by which HSP70 inhibits apoptosis in cancer cells. METHODS HSP70 expression was down-regulated in cultured pancreatic cancer cells by exposure to quercetin, triptolide, or short interfering RNAs. Intracellular Ca2+, cytosolic cathepsin B activity, caspase-3 activity, cell viability, and lysosome integrity were measured using colorimetric assays. Immunofluorescence assays were used to localize cathepsin B and Lamp2. BAPTA-AM was used to chelate intracellular Ca2+. RESULTS Inhibition of HSP70 increased intracellular Ca2+ levels in pancreatic and colon cancer cell lines and led to loss of lysosome integrity in pancreatic cancer cells. The release of intracellular Ca2+ and lysosomal enzymes activated caspase-dependent apoptosis independently and simultaneously. CONCLUSIONS HSP70 inhibits apoptosis in cancer cells by 2 mechanisms: attenuation of cytosolic calcium and stabilization of lysosomes. HSP70-mediated cell survival might occur in other types of cancer cells.

Collaboration


Dive into the Rajinder Dawra's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Selwyn M. Vickers

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Rohit Chugh

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar

Phoebe A. Phillips

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

R. Sharif

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge