Network


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

Hotspot


Dive into the research topics where Sidhartha Tulachan is active.

Publication


Featured researches published by Sidhartha Tulachan.


Clinical Cancer Research | 2004

N-Cadherin Expression and Epithelial-Mesenchymal Transition in Pancreatic Carcinoma

Sanae Nakajima; Ryuichiro Doi; Eiji Toyoda; Shoichiro Tsuji; Michihiko Wada; Masayuki Koizumi; Sidhartha Tulachan; Daisuke Ito; Kazuhiro Kami; Tomohiko Mori; Yoshiya Kawaguchi; Koji Fujimoto; Ryo Hosotani; Masayuki Imamura

Purpose: Loss of intercellular adhesion and increased cell motility promote tumor cell invasion. In the present study, E- and N-cadherin, members of the classical cadherin family, are investigated as inducers of epithelial-to-mesenchymal transition (EMT) that is thought to play a fundamental role during the early steps of invasion and metastasis of carcinomas. Cell growth factors are known to regulate cell adhesion molecules. The purpose of the study presented here was to investigate whether a gain in N-cadherin in pancreatic cancer is involved in the process of metastasis via EMT and whether its expression is affected by growth factors. Experimental Design: We immunohistochemically examined the expression of N- and E-cadherins and vimentin, a mesenchymal marker, in pancreatic primary and metastatic tumors. Correlations among the expressions of N-cadherin, transforming growth factor (TGF)β, and fibroblast growth factor 2 was evaluated in both tumors, and the induction of cadherin and vimentin by growth factors was examined in cultured cell lines. Results: N-cadherin expression was observed in 13 of 30 primary tumors and in 8 of 15 metastatic tumors. N-cadherin expression correlated with neural invasion (P = 0.008), histological type (P = 0.043), fibroblast growth factor expression in primary tumors (P = 0.007), and TGF expression (P = 0.004) and vimentin (P = 0.01) in metastatic tumors. Vimentin, a mesenchymal marker, was observed in a few cancer cells of primary tumor but was substantially expressed in liver metastasis. TGF stimulated N-cadherin and vimentin protein expression and decreased E-cadherin expression of Panc-1 cells with morphological change. Conclusion: This study provided the morphological evidence of EMT in pancreatic carcinoma and revealed that overexpression of N-cadherin is involved in EMT and is affected by growth factors.


International Journal of Cancer | 2003

Endogenous decoy receptor 3 blocks the growth inhibition signals mediated by Fas ligand in human pancreatic adenocarcinoma

Shoichiro Tsuji; Ryo Hosotani; Shin Yonehara; Toshihiko Masui; Sidhartha Tulachan; Sanae Nakajima; Hiroyuki Kobayashi; Masayuki Koizumi; Eiji Toyoda; Daisuke Ito; Kazuhiro Kami; Tomohiko Mori; Koji Fujimoto; Ryuichiro Doi; Masayuki Imamura

Many cancers are resistant to Fas‐mediated apoptosis despite the expression of Fas. To investigate the mechanisms by which Fas signals are attenuated, we focused on decoy receptor 3 (DcR3). DcR3 is a soluble receptor against Fas ligand belonging to the tumor necrosis factor receptor superfamily and overexpresses in some forms of cancers. Exogenous DcR3 inhibits Fas‐mediated apoptosis in Fas‐sensitive Jurkat cells. In our study, we examined the expression and function of DcR3 in pancreatic cancers. TaqMan RT‐PCR showed that DcR3 mRNA was highly expressed in pancreatic cancer cell lines (71%) and tissues (67%). Its expression significantly correlated with cancer invasion to veins. Western blotting showed that the DcR3 protein was produced and secreted in 4 of 6 cell lines. The protein expressions were compatible with the mRNA expression. Five of 7 pancreatic cancer cell lines became sensitive to agonistic anti‐Fas antibody (CH‐11) to various extents, without Fas upregulation, when exposed to CH‐11 for 48 hr after pretreatment with IFNγ. Four of 7 pancreatic cancer cell lines were inhibited from growing, compared to control cells, when cocultured with membrane‐bounded Fas ligand (mFasL) transfected lymphomas for 48 hr after pretreatment with IFNγ. DcR3 reduced this growth inhibition when added exogenously. Regression analysis showed that the DcR3 expression significantly correlated with the sensitivity to mFasL, and not to CH‐11. These results suggest that DcR3 is highly expressed in many pancreatic cancers and endogenous DcR3 blocks the growth inhibition signals mediated by mFasL. DcR3 can be a candidate target molecule for the therapeutic intervention.


Journal of Virological Methods | 2012

Rapid and simplified purification of recombinant adeno-associated virus.

Ping Guo; Yousef El-Gohary; Krishna Prasadan; Chiyo Shiota; Xiangwei Xiao; John Wiersch; Jose Paredes; Sidhartha Tulachan; George K. Gittes

Preclinical gene therapy studies both in vitro and in vivo require high purity preparations of adeno-associated virus (AAV). Current methods for purification of AAV entail the use of centrifugation over either a CsCl or iodixanol gradient, or the use of chromatography. These methods can be cumbersome and expensive, necessitating ultrahigh speed gradient centrifugation or, for chromatography the use of other expensive equipment. In addition, these methods are time consuming, and the viral yield is not high. Currently no commercial purification kits are available for other than AAV serotype 2. A simplified method was used for the purification of AAV, with a viral yield that is able to be used effectively in adult and embryo mice. The method does not require ultrahigh speed gradient centrifugation nor chromatography. Instead, polyethylene glycol (PEG)/aqueous two-phase partitioning is used to remove soluble proteins from the PEG8000 precipitated virus-protein mixture. The procedure obtained rapidly up to 95% recovery of high quality purified AAV. The entire purification process, including HEK293 cell transfection, can be completed readily within one week, with purity seemingly higher than that obtained after one round of CsCl gradient purification.


Diabetes | 2014

A Smad Signaling Network Regulates Islet Cell Proliferation

Yousef El-Gohary; Sidhartha Tulachan; John Wiersch; P. Guo; Carey Welsh; K. Prasadan; Jose Paredes; C. Shiota; X. Xiao; Yoko Wada; Marilyn Diaz; George K. Gittes

Pancreatic β-cell loss and dysfunction are critical components of all types of diabetes. Human and rodent β-cells are able to proliferate, and this proliferation is an important defense against the evolution and progression of diabetes. Transforming growth factor-β (TGF-β) signaling has been shown to affect β-cell development, proliferation, and function, but β-cell proliferation is thought to be the only source of new β-cells in the adult. Recently, β-cell dedifferentiation has been shown to be an important contributory mechanism to β-cell failure. In this study, we tie together these two pathways by showing that a network of intracellular TGF-β regulators, smads 7, 2, and 3, control β-cell proliferation after β-cell loss, and specifically, smad7 is necessary for that β-cell proliferation. Importantly, this smad7-mediated proliferation appears to entail passing through a transient, nonpathologic dedifferentiation of β-cells to a pancreatic polypeptide–fold hormone-positive state. TGF-β receptor II appears to be a receptor important for controlling the status of the smad network in β-cells. These studies should help our understanding of properly regulated β-cell replication.


Developmental Biology | 2013

Smad signaling pathways regulate pancreatic endocrine development.

Yousef El-Gohary; Sidhartha Tulachan; P. Guo; Carey Welsh; John Wiersch; K. Prasadan; Jose Paredes; C. Shiota; X. Xiao; Yoko Wada; Marilyn Diaz; George K. Gittes

Expansion of the pancreatic endocrine cell population occurs during both embryonic development and during post-natal pancreatic growth and regeneration. Mechanisms of the expansion of endocrine cells during embryonic development are not completely understood, and no clear mechanistic link has been established between growth of the embryonic endocrine pancreas and the islet cell replication that occurs in an adult animal. We found that transforming growth factor-beta (TGF-β) superfamily signaling, which has been implicated in many developmental processes, plays a key role in regulating pancreatic endocrine maturation and development. Specifically, the intracellular mediators of TGF-β signaling, smad2 and smad3, along with their inhibitor smad7, appear to mediate this process. Smad2, smad3 and smad7 were all broadly expressed throughout the early embryonic pancreatic epithelium. However, during later stages of development, smad2 and smad3 became strongly localized to the nuclei of the endocrine positive cells, whereas the inhibitory smad7 became absent in the endocrine component. Genetic inactivation of smad2 and smad3 led to a significant expansion of the embryonic endocrine compartment, whereas genetic inactivation of smad7 led to a significant decrease in the endocrine compartment. In vitro antisense studies further corroborated these results and supported the possibility that interplay between the inhibitory smad7 and the intracellular mediators smad2/3 is a control point for pancreatic endocrine development. These results should provide a better understanding of the key control mechanisms for β-cell development.


Anatomical Record-advances in Integrative Anatomy and Evolutionary Biology | 2012

Three‐Dimensional Analysis of the Islet Vasculature

Yousef El-Gohary; Sunder Sims-Lucas; N. Lath; Sidhartha Tulachan; Ping Guo; Xiangwei Xiao; Carey Welsh; Jose Paredes; John Wiersch; Krishna Prasadan; Chiyo Shiota; George K. Gittes

The pancreatic islets of Langerhans are highly vascularized structures scattered throughout the pancreas that contain a capillary network 5–10 times denser than that of the exocrine pancreas. A simple method for three‐dimensional (3D) analysis of this intricate intraislet vasculature has been difficult because of the intrinsic opacity of the pancreas. We developed a whole‐mount imaging technique that allows relatively easy visualization of the islet vasculature. In combination with confocal microscopy and the use of 3D imaging software, we were able to readily reconstruct the 3D architecture of an islet, allowing delineation of the islet volume, length of the intraislet vessels, and the number of vessel branch‐points. This technique allows for straightforward 3D image analysis that may help toward understanding islet function. Anat Rec, 2012.


Anatomical Record-advances in Integrative Anatomy and Evolutionary Biology | 2012

Whole-mount imaging demonstrates hypervascularity of the pancreatic ducts and other pancreatic structures.

Yousef El-Gohary; Sidhartha Tulachan; Maria F. Branca; Sunder Sims-Lucas; Ping Guo; Krishna Prasadan; Chiyo Shiota; George K. Gittes

Confocal microscopy in combination with commercial software is frequently used to generate three‐dimensional images of tissue architecture. Here we report a novel, whole‐mount imaging protocol technique that allows detailed three‐dimensional imaging of adult pancreatic structures. This technique provides an improved appreciation of the anatomical detail of pancreatic structures and of the relationship between the pancreatic ducts and islets. In addition, imaging of the pancreatic ducts revealed a previously unappreciated high degree of hypervascularity. Anat Rec, 2012.


Mechanisms of Development | 2014

Barrier function of the coelomic epithelium in the developing pancreas.

Ping Guo; Barry Preuett; Prasadan Krishna; Xiangwei Xiao; Chiyo Shiota; John Wiersch; Iliana Gaffar; Sidhartha Tulachan; Yousef El-Gohary; Zewen Song; George K. Gittes

Tight spatial regulation of extracellular morphogen signaling within the close confines of a developing embryo is critical for proper organogenesis. Given the complexity of extracellular signaling in developing organs, together with the proximity of adjacent organs that use disparate signaling pathways, we postulated that a physical barrier to signaling may exist between organs in the embryo. Here we describe a previously unrecognized role for the embryonic coelomic epithelium in providing a physical barrier to contain morphogenic signaling in the developing mouse pancreas. This layer of cells appears to function both to contain key factors required for pancreatic epithelial differentiation, and to prevent fusion of adjacent organs during critical developmental windows. During early foregut development, this barrier appears to play a role in preventing splenic anlage-derived activin signaling from inducing intestinalization of the pancreas-specified epithelium.


Clinical Cancer Research | 2003

RECK Expression in Pancreatic Cancer Its Correlation with Lower Invasiveness and Better Prognosis

Toshihiko Masui; Ryuichiro Doi; Takatomo Koshiba; Koji Fujimoto; Shoichiro Tsuji; Sanae Nakajima; Masayuki Koizumi; Eiji Toyoda; Sidhartha Tulachan; Daisuke Ito; Kazuhiro Kami; Tomohiko Mori; Michihiko Wada; Makoto Noda; Masayuki Imamura


Clinical Cancer Research | 2001

Expression of METH-1 and METH-2 in Pancreatic Cancer

Toshihiko Masui; Ryo Hosotani; Shoichiro Tsuji; Yoshiharu Miyamoto; Seichi Yasuda; Jun Ida; Sanae Nakajima; Michiya Kawaguchi; Hiroyuki Kobayashi; Masayuki Koizumi; Eiji Toyoda; Sidhartha Tulachan; Shigeki Arii; Ryuichiro Doi; Masayuki Imamura

Collaboration


Dive into the Sidhartha Tulachan's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Masayuki Imamura

Tokyo Medical and Dental University

View shared research outputs
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