Network


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

Hotspot


Dive into the research topics where Kuldeepsinh Rana is active.

Publication


Featured researches published by Kuldeepsinh Rana.


Science Translational Medicine | 2011

Age-Related Intimal Stiffening Enhances Endothelial Permeability and Leukocyte Transmigration

John Huynh; Nozomi Nishimura; Kuldeepsinh Rana; John M. Peloquin; Joseph P. Califano; Christine R. Montague; Michael R. King; Chris B. Schaffer; Cynthia A. Reinhart-King

Inhibiting endothelial cell contractility reverses the deleterious effects of age-related matrix stiffening on normal cell function, which could help prevent the development of atherosclerosis. Rock Your Heart Out According to novelist Thomas Bailey Aldrich, “To keep the heart unwrinkled, to be hopeful, kindly, cheerful, reverent, is to triumph over old age” (from Ponkapoag Papers). Unfortunately, despite a positive attitude, aging is accompanied by several changes of heart, at least at the cellular level. One age-related “wrinkle” is stiffening of the extracellular matrix that lines the blood vessels, a change that has been linked to atherosclerosis; yet, the cellular and mechanical features that couple the two conditions have remained elusive. Now, using a clever combination of biomaterials, cells, aortas, and mice, Huynh and colleagues have demystified the correlation between aging and atherosclerosis, showing that cell contractility is at the heart of it all. The authors first developed an in vitro system that mimicked the basic structures of both young and old blood vessels. Synthetic hydrogel matrices of varying stiffnesses were seeded with bovine aortic endothelial cells. By administering a solution of fluorescently labeled molecules to the cell-gel system and watching how the dye moved across the cell layer, Huynh et al. determined that permeability increased as a function of matrix stiffness, suggesting that age alone was a disruptive factor. These results were confirmed ex vivo by performing atomic force microscopy with decellularized thoracic aortas from both young (~10 weeks) and old (~92 weeks) mice. In both of these systems, the enhanced vessel permeability resulted from an increase in the distance—or junction—between neighboring cells. This increase in the so-called gap junction width also permitted the passage of leukocytes through the endothelial cell monolayer; along with leaky vasculature, cellular transmigration is a hallmark of atherosclerosis progression. Because the Rho signaling pathway is linked to the cellular cytoskeleton and, in turn, contractility, Huynh et al. hypothesized that they could reverse the effects of age-related intimal stiffening by inhibiting Rho-associated kinase (ROCK). By administering a pharmacological ROCK inhibitor (Y-27632) to their in vitro setup and to old mice, the authors showed that gap junction widths and endothelial cellular forces decreased. In vitro, the inhibitor also prevented leukocyte transmigration. These observations suggest that directly interfering with Rho signaling is a viable treatment option for age-related atherosclerosis. And because inhibitors of Rho signaling, such as fasudil, are already available in the clinic, one might say that physicians and researchers are ready to rock. Age is the most significant risk factor for atherosclerosis; however, the link between age and atherosclerosis is poorly understood. During both aging and atherosclerosis progression, the blood vessel wall stiffens owing to alterations in the extracellular matrix. Using in vitro and ex vivo models of vessel wall stiffness and aging, we show that stiffening of extracellular matrix within the intima promotes endothelial cell permeability—a hallmark of atherogenesis. When cultured on hydrogels fabricated to match the elasticity of young and aging intima, endothelial monolayers exhibit increased permeability and disrupted cell-cell junctions on stiffer matrices. In parallel experiments, we showed a corresponding increase in cell-cell junction width with age in ex vivo aortas from young (10 weeks) and old (21 to 25 months) healthy mice. To investigate the mechanism by which matrix stiffening alters monolayer integrity, we found that cell contractility increases with increased matrix stiffness, mechanically destabilizing cell-cell junctions. This increase in endothelial permeability results in increased leukocyte extravasation, which is a critical step in atherosclerotic plaque formation. Mild inhibition of Rho-dependent cell contractility using Y-27632, an inhibitor of Rho-associated kinase, or small interfering RNA restored monolayer integrity in vitro and in vivo. Our results suggest that extracellular matrix stiffening alone, which occurs during aging, can lead to endothelial monolayer disruption and atherosclerosis pathogenesis. Because previous therapeutics designed to decrease vascular stiffness have been met with limited success, our findings could be the basis for the design of therapeutics that target the Rho-dependent cellular contractile response to matrix stiffening, rather than stiffness itself, to more effectively prevent atherosclerosis progression.


Proceedings of the National Academy of Sciences of the United States of America | 2014

TRAIL-coated leukocytes that kill cancer cells in the circulation

Michael J. Mitchell; Elizabeth C. Wayne; Kuldeepsinh Rana; Chris B. Schaffer; Michael R. King

Significance This paper describes a unique approach to target and kill cancer cells in the bloodstream, in which the extensive surface area of circulating leukocytes is used to display the cancer-specific TNF-related apoptosis inducing ligand (TRAIL) and E-selectin adhesion receptor to the surrounding fluid. The approach is inspired by the cytotoxic activity of natural killer cells and is quite effective at killing cancer cells both in vitro with human blood samples and in mouse blood circulation. The mechanism is surprising and unexpected in that this repurposing of leukocytes in flowing blood is more effective than directly targeting the cancer cells with liposomes or soluble protein. Metastasis through the bloodstream contributes to poor prognosis in many types of cancer. Mounting evidence implicates selectin-based adhesive interactions between cancer cells and the blood vessel wall as facilitating this process, in a manner similar to leukocyte trafficking during inflammation. Here, we describe a unique approach to target and kill colon and prostate cancer cells in the blood that causes circulating leukocytes to present the cancer-specific TNF-related apoptosis inducing ligand (TRAIL) on their surface along with E-selectin adhesion receptor. This approach, demonstrated in vitro with human blood and also in mice, mimics the cytotoxic activity of natural killer cells and increases the surface area available for delivery of the receptor-mediated signal. The resulting “unnatural killer cells” hold promise as an effective means to neutralize circulating tumor cells that enter blood with the potential to form new metastases.


Biotechnology and Bioengineering | 2009

Delivery of apoptotic signal to rolling cancer cells: A novel biomimetic technique using immobilized TRAIL and E-selectin

Kuldeepsinh Rana; Jane L. Liesveld; Michael R. King

The survival rate for patients with metastases versus localized cancer is dramatically reduced, with most deaths being associated with the formation of secondary tumors. Circulating cancer cells interact with the endothelial lining of the vasculature via a series of adhesive interactions that facilitate tethering and firm adhesion of cancer cells in the initial steps of metastasis. TNF‐related apoptosis‐inducing ligand (TRAIL) holds promise as a tumor‐specific cancer therapeutic, by inducing a death signal by apoptosis via the caspase pathway. In this study, we exploit this phenomenon to deliver a receptor‐mediated apoptosis signal to leukemic cells adhesively rolling along a TRAIL and selectin‐bearing surface. Results show that cancer cells exhibit selectin‐mediated rolling in capillary flow chambers, and that the rolling velocities can be controlled by varying the selectin and selectin surface density and the applied shear stress. It was determined that a 1 h rolling exposure to a functionalized TRAIL and E‐selectin surface was sufficient to kill 30% of captured cells compared to static conditions in which 4 h exposure was necessary to kill 30% of the cells. Thus, we conclude that rolling delivery is more effective than static exposure to a TRAIL immobilized surface. We have also verified that there is no significant effect of TRAIL on hematopoietic stem cells and other normal blood cells. This represents the first demonstration of a novel biomimetic method to capture metastatic cells from circulation and deliver an apoptotic signal. Biotechnol. Bioeng. 2009;102: 1692–1702.


Carbohydrate Research | 2010

Knockdown of fucosyltransferase III disrupts the adhesion of circulating cancer cells to E-selectin without affecting hematopoietic cell adhesion.

Xiaoyan Yin; Kuldeepsinh Rana; Varun Ponmudi; Michael R. King

Adhesive interactions between selectins and their ligands play an essential role during cancer extravasation. Fucosylation of these proteins by fucosyltransferases, or FUTs, is critical for their functions. Using quantitative RT-PCR, we demonstrated that FUT4 and FUT7 are the predominant FUTs expressed in hematopoietic cell line, while FUT3 is heavily expressed by multiple cancer cell lines including the prostate cancer cell line MDA PCa2b. Knockdown of FUT3 expression in MDA PCa2b cells by small interference RNA (siRNA) significantly reduced FUT3 expression. Cell-surface sialyl Lewis antigens were largely abolished. Cell adhesion and cell rolling on the blood vessel wall were simulated by perfusing cancer cells through microtubes coated with recombinant human E-selectin. At physiological levels of wall shear stress, the number of flowing cancer cells recruited to the microtube surface was dramatically reduced by FUT3 knockdown. Higher rolling velocity was also observed, which is consistent with reduced E-selectin binding activity. Interestingly, FUT3 siRNA treatment also significantly reduced the cell growth rate. Combined with the novel siRNA delivery platform recently developed in our laboratory, FUT3 siRNA could be a promising conjunctive therapy aiming at reducing the metastatic virulence of circulating epithelial cancer cells.


Molecular Pharmaceutics | 2012

Inducing Apoptosis in Rolling Cancer Cells: A Combined Therapy with Aspirin and Immobilized TRAIL and E-Selectin

Kuldeepsinh Rana; Cynthia A. Reinhart-King; Michael R. King

Though metastasis is considered an inefficient process, over 90% of cancer related deaths are attributed to the formation of secondary tumors. Thus, eliminating circulating cancer cells could lead to improved patient survival. This study was aimed at exploiting the interactions of cancer cells with selectins under flow to selectively kill captured colon cancer cells. Microtubes functionalized with E-selectin and TRAIL were perfused with colon cancer cell line Colo205 either treated with 1 mM aspirin or untreated for 1 or 2 h. Cells were collected from the microtube and analyzed by flow cytometry. Aspirin treatment alone killed only 3% cells in culture. A 95% difference in the number of cells killed between control and TRAIL + ES surfaces was seen when aspirin treated cells were perfused over the functionalized surface for 2 h. We have demonstrated a novel biomimetic method to capture and neutralize cancer cells in flow, thus reducing the chances for the formation of secondary tumors.


Journal of Bionic Engineering | 2009

Biomolecular Surfaces for the Capture and Reprogramming of Circulating Tumor Cells

Michael R. King; Laura T. Western; Kuldeepsinh Rana; Jane L. Liesveld

Circulating Tumor Cells (CTC) have the potential to be used clinically as a diagnostic tool and a treatment tool in the field of oncology. As a diagnostic tool, CTC may be used to indicate the presence of a tumor before it is large enough to cause noticeable symptoms. As a treatment tool, CTC isolated from patients may be used to test the efficacy of chemotherapy options to personalize patient treatment. One way for tumors to spread is through metastasis via the circulatory system. CTC are able to exploit the natural leukocyte recruitment process that is initially mediated by rolling on transient selectin bonds. Our capture devices take advantage of this naturally occurring recruitment step to isolate CTC from whole blood by flowing samples through selectin and antibody-coated microtubes. Whole blood was spiked with a known concentration of labeled cancer cells and then perfused through pre-coated microtubes. Microtubes were then rinsed to remove unbound cells and the number of labeled cells captured on the lumen was assessed. CTC were successfully captured from whole blood at a clinically relevant level on the order of 10 cells per mL. Combination tubes with selectin and antibody coated surface exhibited higher capture rate than tubes coated with selectin alone or antibody alone. Additionally, CTC capture was demonstrated with the KG1a hematopoietic cell line and the DU145 epithelial cell line. Thus, the in vivo process of selectin-mediated CTC recruitment to distant vessel walls can be used in vitro to target CTC to a tube lumen. The biomolecular coatings can also be used to capture CTC of hematopoietic and epithelial tumor origin and is demonstrated to sensitivities down to the order of 10 CTC per mL.In a related study aimed at reducing the blood borne metastatic cancer load, we have shown that cells captured to a surface can be neutralized by a receptor-mediated biochemical signal. In the proposed method we have shown that using a combined selectin and TRAIL (TNF Related Apoptosis Inducing Ligand or Apo 2L) functionalized surface we are able to kill about 30% of the captured cells in a short duration of one hour whereas it took about 4 hours to kill the same proportion of cells without flow on a similarly functionalized surface. Here we have taken the approach a step further by showing that with very small doses of chemotherapeutic agents like bortezomib, we can increase the kill rate of CTC, thus allowing the device to function in scenarios where the patient is undergoing treatment. We show here that, with leukemic cells treated with bortezomib we are able to kill about 41% of the captured cells.


northeast bioengineering conference | 2014

Unnatural killer cells: TRAIL-coated leukocytes that kill cancer cells in the circulation

Michael J. Mitchell; Elizabeth C. Wayne; Kuldeepsinh Rana; Chris B. Schaffer; Michael R. King

Metastasis through the bloodstream contributes to poor prognosis in many types of cancer. Mounting evidence implicates selectin-based adhesive interactions between cancer cells and the blood vessel wall as facilitating this process, in a manner similar to leukocyte trafficking during inflammation. Here, we describe a unique approach to target and kill colon and prostate cancer cells in the blood that causes circulating leukocytes to present the cancer-specific apoptosis ligand TRAIL on their surface along with E-selectin adhesion receptor. This approach, demonstrated in vitro with human blood and also in mice, mimics the cytotoxic activity of natural killer cells and increases the surface area available for delivery of the receptor-mediated signal. The resulting “unnatural killer cells” hold promise as an effective means to neutralize circulating tumor cells that enter blood with the potential to form new metastases.


ASME 2009 7th International Conference on Nanochannels, Microchannels, and Minichannels | 2009

Flow-Based Isolation and Neutralization of Circulating Tumor Cells

Laura T. Western; Kuldeepsinh Rana; Michael R. King

Circulating tumor cells (CTC) have the potential to be used clinically as a diagnostic tool and a treatment tool in the field of oncology. As a diagnostic tool, CTC may be used to indicate the presence of a tumor before the tumor is large enough to cause noticeable symptoms. As a treatment tool, CTC isolated from patients may be used to test the efficacy of chemotherapy options to personalize patient treatment. One way for tumors to spread is through metastasis via the circulatory system. CTC are able to exploit the natural leukocyte recruitment process that is initially mediated by rolling on transient selectin bonds. Our capture devices take advantage of this naturally occurring recruitment step to isolate CTC from whole blood by flowing samples through selectin and antibody-coated microtubes. Whole blood was spiked with a known concentration of labeled cancer cells and then perfused through pre-coated microtubes. Microtubes were then rinsed to remove unbound cells and the number of labeled cells captured on the lumen was assessed. CTC were successfully captured from whole blood at a clinically relevant level on the order of 10 cells per mL. Combination tubes with selectin and antibody coated surface exhibited higher capture rate than tubes coated with selectin alone or antibody alone. Additionally, CTC capture was demonstrated with the KG1a hematopoietic cell line and the Du145 epithelial cell line. Thus, the in vivo process of selectin-mediated CTC recruitment to distant vessel walls can be used in vitro to target CTC to a tube lumen. The microtube device can also be used to capture CTC of hematopoietic and epithelial tumor origin and is demonstrated sensitivity down to the order of 10 CTC per mL. In a related study aimed at reducing the blood borne metastatic cancer load, we have shown that cells captured to a surface can be neutralized by a receptor-mediated biochemical signal (Rana et al. 2008). In the proposed method we have shown that using a combined selecting and TRAIL (TNF Related Apoptosis Inducing Ligand or Apo 2L) functionalized surface we are able to kill about 30% of the captured cells in a short duration of 1 hour whereas it took about 4 hours to kill the same proportion of cells without flow on a similarly functionalized. Here we have taken the approach a step further by showing that with very small doses of chemotherapeutic agents like Bortezomib, we can increase the kill rate of CTCs., thus allowing the device to function in senarios where the patient is undergoing treatment. We show here with leukemic cells that are treated with Bortezomib that we are able kill about 41% of the captured cells.Copyright


Scientific Reports | 2013

A physical sciences network characterization of non-tumorigenic and metastatic cells

David B. Agus; Jenolyn F. Alexander; Wadih Arap; Shashanka Ashili; Joseph E. Aslan; Robert H. Austin; Vadim Backman; Kelly Bethel; Richard Bonneau; Wei Chiang Chen; Chira Chen-Tanyolac; Nathan C. Choi; Steven A. Curley; Matthew R. Dallas; Dhwanil Damania; Paul Davies; Paolo Decuzzi; Laura E. Dickinson; Luis Estévez-Salmerón; Veronica Estrella; Mauro Ferrari; Claudia Fischbach; Jasmine Foo; Stephanie I. Fraley; Christian Frantz; Alexander Fuhrmann; Philippe Gascard; Robert A. Gatenby; Yue Geng; Sharon Gerecht


Archive | 2008

Flow chamber device for neutralization of cancer cells

Michael R. King; Jane L. Liesveld; Kuldeepsinh Rana

Collaboration


Dive into the Kuldeepsinh Rana'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

John M. Peloquin

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge