Network


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

Hotspot


Dive into the research topics where Kumaran Sundaram is active.

Publication


Featured researches published by Kumaran Sundaram.


Cancer Immunology, Immunotherapy | 2008

Gamma-IFN-inducible-lysosomal thiol reductase modulates acidic proteases and HLA class II antigen processing in melanoma

Oliver Goldstein; Laela M. Hajiaghamohseni; Shereen Amria; Kumaran Sundaram; Sakamuri V. Reddy; Azizul Haque

HLA class II-restricted antigen (Ag) processing and presentation are important for the activation of CD4+ T cells, which are the central orchestrating cells of immune responses. The majority of melanoma cells either expresses, or can be induced to express, HLA class II proteins. Thus, they are prime targets for immune mediated elimination by class II-restricted CD4+ T cells. We have previously shown that human melanoma cells lack an important enzyme, gamma interferon-inducible lysosomal thiol-reductase (GILT), capable of perturbing immune recognition of these tumors. Here, we show that GILT expression in human melanoma cells enhances Ag processing and presentation via HLA class II molecules. We also show that GILT expression influences the generation of active forms of cysteinyl proteases, cathepsins B, L and S, as well as an aspartyl protease cathepsin D in melanoma cells. Mechanistic studies revealed that GILT does not regulate acidic cathepsins at the transcriptional level; rather it colocalizes with the cathepsins and influences HLA class II Ag processing. GILT expression in melanoma cells also elevated HLA-DM molecules, which favor epitope loading onto class II in the endolysosomal compartments, enhancing CD4+ T cell recognition. These data suggest that GILT-expressing melanoma cells could prove to be very promising for direct antigen presentation and CD4+ T cell recognition, and may have direct implications for the design of cancer vaccines.


Endocrinology | 2011

Mutant p62P392L Stimulation of Osteoclast Differentiation in Paget's Disease of Bone

Kumaran Sundaram; Srinivasan Shanmugarajan; D. Sudhaker Rao; Sakamuri V. Reddy

Pagets disease of the bone (PDB) is an autosomal dominant trait with genetic heterogeneity, characterized by abnormal osteoclastogenesis. Sequestosome 1 (p62) is a scaffold protein that plays an important role in receptor activator of nuclear factor κB (RANK) signaling essential for osteoclast (OCL) differentiation. p62P392L mutation in the ubiquitin-associated (UBA) domain is widely associated with PDB; however, the mechanisms by which p62P392L stimulate OCL differentiation in PDB are not completely understood. Deubiquitinating enzyme cylindromatosis (CYLD) has been shown to negatively regulate RANK ligand-RANK signaling essential for OCL differentiation. Here, we report that CYLD binds with the p62 wild-type (p62WT), non-UBA mutant (p62A381V) but not with the UBA mutant (p62P392L) in OCL progenitor cells. Also, p62P392L induces expression of c-Fos (2.8-fold) and nuclear factor of activated T cells c1 (6.0-fold) transcription factors critical for OCL differentiation. Furthermore, p62P392L expression results in accumulation of polyubiquitinated TNF receptor-associated factor (TRAF)6 and elevated levels of phospho-IκB during OCL differentiation. Retroviral transduction of p62P392L/CYLD short hairpin RNA significantly increased TRAP positive multinucleated OCL formation/bone resorption activity in mouse bone marrow cultures. Thus, the p62P392L mutation abolished CYLD interaction and enhanced OCL development/bone resorption activity in PDB.


Oncogene | 2013

CXCL13 activation of c-Myc induces RANK ligand expression in stromal/preosteoblast cells in the oral squamous cell carcinoma tumor–bone microenvironment

Yuvaraj Sambandam; Kumaran Sundaram; Angen Liu; Keith L. Kirkwood; William L. Ries; Sakamuri V. Reddy

CXC chemokine ligand-13 (CXCL13) has been implicated in oral squamous cell carcinoma (OSCC) tumor progression and osteolysis. The tumor necrosis factor family member RANKL (receptor activator of NF-κB ligand), a critical bone resorbing osteoclastogenic factor, has an important role in cancer invasion of bone/osteolysis. Here, we show high-level expression of CXCL13 in primary human OSCC tumor specimens; however, human bone marrow-derived stromal (SAKA-T) and murine preosteoblast (MC3T3-E1) cells produce at very low level. Recombinant CXCL13 (0–15 ng/ml) dose dependently induced CXCR5 expression in SAKA-T and MC3T3-E1 cells. Conditioned media obtained from OSCC cell lines increased the RANKL expression and an antibody against the CXCL13 specific receptor, CXCR5 markedly decreased RANKL expression in these cells. Furthermore, CXCL13 increased hRANKL-Luc promoter activity. Superarray screening identified c-Myc and NFATc3 transcription factors upregulated in CXCL13-stimulated SAKA-T cells. Immunohistochemical analysis of OSCC tumors that developed in athymic mice demonstrated RANKL and NFATc3 expression in tumor and osteoblast cells, however, showed p-c-Myc expression specific to osteoblastic cells at the tumor–bone interface. We further identified NFATc3 expression, but not c-Myc activation in primary human OSCC tumor specimens compared with adjacent normal tissue. Also, CXCL13 significantly increased p-ERK1/2 in SAKA-T and MC3T3-E1 cells. siRNA suppression of c-Myc expression markedly decreased CXCL13-induced RANKL and NFATc3 expression in preosteoblast cells. Chromatin-immuno precipitation assay confirmed p-c-Myc binding to the hRANKL promoter region. In summary, c-Myc activation through CXCL13–CXCR5 signaling axis stimulates RANKL expression in stromal/preosteoblast cells. Thus, our results implicate CXCL13 as a potential therapeutic target to prevent OSCC invasion of bone/osteolysis.


Molecular Cancer Research | 2009

A Novel Function of CXCL13 to Stimulate RANK Ligand Expression in Oral Squamous Cell Carcinoma Cells

Sambandam Yuvaraj; Alfred C. Griffin; Kumaran Sundaram; Keith L. Kirkwood; James S. Norris; Sakamuri V. Reddy

Oral squamous cell carcinomas (OSCC) are malignant tumors with a potent activity of local bone invasion/osteolysis. The chemokine ligand, CXCL13, has been identified as a prognostic marker for OSCC development and progression. Here in, we show that recombinant hCXCL13 treatment of OSCC cells stimulates (5-fold) RANK ligand (RANKL), a critical bone resorbing osteoclastogenic factor expression. Anti-CXCR5 chemokine receptor antibody abrogates CXCL13-induced RANKL expression in these cells. Also, CXCL13 stimulated (3.0-fold) hRANKL gene promoter activity in SCC14a cells. SuperArray screening for transcription factors by real-time RT-PCR identified significant increase in the levels of c-Jun and NFATc3 mRNA expression in CXCL13-stimulated OSCC cells. CXCL13 treatment significantly increased (3.5-fold) phospho-c-Jun levels in these cells and a c-Jun-NH2-kinase inhibitor abolished CXCL13-stimulated RANKL expression. Furthermore, we show that CXCL13 stimulation induced nuclear translocation of NFATc3 in OSCC cells. Chromatin-immune precipitation assay confirmed NFATc3 binding to the RANKL promoter region. We also show that overexpression of NFATc3 stimulates RANKL expression/promoter activity and that siRNA suppression of NFATc3 abolished CXCL13-stimulated RANKL expression. Thus, our results suggest that NFATc3 is a downstream target of the CXCL13/CXCR5 axis to stimulate RANKL expression in OSCC cells and implicates CXCL13 as a potential therapeutic target to prevent OSCC bone invasion/osteolysis. (Mol Cancer Res 2009;7(8):1399–407)


International Journal of Cancer | 2009

Role of CXC chemokine ligand 13 in oral squamous cell carcinoma associated osteolysis in athymic mice

Subramanya N.M. Pandruvada; Sambandam Yuvaraj; Xiang Liu; Kumaran Sundaram; Srinivasan Shanmugarajan; William L. Ries; James S. Norris; Steven D. London; Sakamuri V. Reddy

Oral squamous cell carcinomas (OSCC) are malignant tumors with a potent activity of local bone invasion; however, the molecular mechanisms of tumor osteolysis are unclear. In this study, we identified high level expression of chemokine ligand, CXCL13 and RANK ligand (RANKL) in OSCC cells (SCC1, SCC12 and SCC14a). OSCC cell‐conditioned media (20%) induced osteoclast differentiation which was inhibited by OPG in peripheral blood monocyte cultures indicating that OSCC cells produce soluble RANKL. Recombinant hCXCL13 (10 ng/ml) significantly enhanced RANKL‐stimulated osteoclast differentiation in these cultures. Trans‐well migration assay identified that CXCL13 induces chemotaxis of peripheral blood monocytes in vitro which was inhibited by addition of anti‐CXCR5 receptor antibody. Zymogram analysis of conditioned media from OSCC cells revealed matrix metalloproteinase‐9 (MMP‐9) activity. Interestingly, CXCL13 treatment to OSCC cells induced CXCR5 and MMP‐9 expression suggesting an autocrine regulatory function in OSCC cells. To examine the OSCC tumor cell bone invasion/osteolysis, we established an in vivo model for OSCC by subcutaneous injection of OSCC cells onto the surface of calvaria in NCr‐nu/nu athymic mice, which developed tumors in 4–5 weeks. μCT analysis revealed numerous osteolytic lesions in calvaria from OSCC tumor‐bearing mice. Histochemical staining of calvarial sections from these mice revealed a significant increase in the numbers of TRAP‐positive osteoclasts at the tumor‐bone interface. Immunohistochemical analysis confirmed CXCL13 and MMP‐9 expression in tumor cells. Thus, our data implicate a functional role for CXCL13 in bone invasion and may be a potential therapeutic target to prevent osteolysis associated with OSCC tumors in vivo.


Cellular Immunology | 2011

Enhancement of HLA class II-restricted CD4+ T cell recognition of human melanoma cells following treatment with bryostatin-1.

Dan Zhao; Shereen Amria; Azim Hossain; Kumaran Sundaram; Peter Komlosi; Mitzi Nagarkatti; Azizul Haque

The majority of melanoma cells express detectable levels of HLA class II proteins, and an increased threshold of cell surface class II is crucial for the stimulation of CD4+ T cells. Bryostatin-1, a protein kinase C (PKC) activator, has been considered as a potent chemotherapeutic agent in a variety of in vitro tumor models. Little is known about the role of bryostatin-1 in HLA class II Ag presentation and immune activation in malignant tumors, especially in melanoma. In this study, we show that bryostatin-1 treatment enhances CD4+ T cell recognition of melanoma cells in the context of HLA class II molecules. We also show that bryostatin-1 treatment of melanoma cells increases class II protein levels by upregulating the class II transactivator (CIITA) gene. Flow cytometry and confocal microscopic analyses revealed that bryostatin-1 treatment upregulated the expression of costimulatory molecules (CD80 and CD86) in melanoma cells, which could prolong the interaction of immune cells and tumors. Bryostatin-1 also induced cellular differentiation in melanoma cells, and reduced tumorigenic factors such as pro-cathepsins and matrix-metalloproteinase-9. These data suggest that bryostatin-1 could be used as a chemo-immunotherapeutic agent for reducing tumorigenic potential of melanoma cells while enhancing CD4+ T cell recognition to prevent tumor recurrence.


Laboratory Investigation | 2013

CXCL5 stimulation of RANK ligand expression in Paget’s disease of bone

Kumaran Sundaram; D. Sudhaker Rao; William L. Ries; Sakamuri V. Reddy

Paget’s disease of bone (PDB) is a chronic focal skeletal disorder that affects 2–3% of the population over 55 years of age. PDB is marked by highly localized areas of bone turnover with increased osteoclast activity. Evidence suggests a functional role for measles virus nucleocapsid protein (MVNP) in the pathogenesis of PDB. In the present study, we identified elevated levels (∼180-fold) of CXCL5 mRNA expression in bone marrow cells from patients with PDB compared with that in normal subjects. In addition, CXCL5 levels are increased (five-fold) in serum samples from patients with PDB. Furthermore, MVNP transduction in human bone marrow monocytes significantly increased CXCL5 mRNA expression. Real-time PCR analysis showed that CXCL5 stimulation increased (6.8-fold) RANKL mRNA expression in normal human bone marrow-derived stromal (SAKA-T) cells. Moreover, CXCL5 increased (5.2-fold) CXCR1 receptor expression in these cells. We further showed that CXCL5 treatment elevated the expression levels of phospho-ERK1/2 and phospho-p38. CXCL5 also significantly increased phosphorylation of CREB (cAMP response element-binding) in bone marrow stromal/preosteoblast cells. Chromatin immuneprecipitation (ChIP) assay confirmed phospho-CREB binding to RANKL gene promoter region. Further, the suppression of p-CREB expression by the inhibitors of ERK1/2, p38 and PKA significantly decreased CXCL5 stimulation of hRANKL gene promoter activity. Thus, our results suggest that CREB is a downstream effector of CXCL5 signaling and that increased levels of CXCL5 contribute to enhanced levels of RANKL expression in PDB.


Journal of Cellular Biochemistry | 2008

DACH1 negatively regulates the human RANK ligand gene expression in stromal/preosteoblast cells.

Kumaran Sundaram; Santhosh K. Mani; Kazuyuki Kitatani; Kongming Wu; Richard G. Pestell; Sakamuri V. Reddy

Receptor activator of NF‐κB ligand (RANKL) is a critical osteoclastogenic factor that is expressed on bone marrow stromal/preosteoblast cells. Most bone resorption stimuli induce osteoclast formation by modulating RANKL expression in these cells. However, little is known about the mechanisms regulating RANKL gene expression. We recently reported that heat shock factor‐2 (HSF‐2) is a downstream target for FGF‐2 signaling to enhance RANKL gene transcription in marrow stromal/preosteoblast cells. In this study, we show that DACH1 (human homologue of Drosophila dachshund gene) negatively regulates RANKL gene expression and suppresses FGF‐2‐enhanced RANKL gene expression in these cells. DACH1 contains a conserved dachshund domain (DS) in the N‐terminal region, which interacts with the nuclear co‐repressor (NCoR) to repress gene expression. Co‐expression of DACH1 with hRANKL promoter‐luciferase reporter plasmid in normal human bone marrow‐derived stromal cells significantly decreased (3.3‐fold) FGF‐2‐stimulated hRANKL gene promoter activity. Deletion of DS domain abolished DACH1 inhibition of FGF‐2‐enhanced RANKL gene promoter activity. Western blot analysis confirmed that DACH1 suppressed FGF‐2‐stimulated RANKL expression in marrow stromal/preosteoblast cells. We show HSF‐2 co‐immune precipitated with DACH1 and that FGF‐2 stimulation significantly increased (2.7‐fold) HSF‐2 binding to DACH1. Confocal microscopy analysis further demonstrated that FGF‐2 promotes HSF‐2 nuclear transport and co‐localization with DACH1 in marrow stromal cells. Co‐expression of NCoR with DACH1 significantly decreased (5.3‐fold) and siRNA suppression of NCoR in DACH1 co‐transfected cells increased (3.6‐fold) RANKL promoter activity. Furthermore, DACH1 co‐expression with NCoR significantly decreased (7.5‐fold) RANKL mRNA expression in marrow stromal cells. Collectively, these studies indicate that NCoR participates in DACH1 repression of RANKL gene expression in marrow stromal/preosteoblast cells. Thus, DACH1 plays an important role in negative regulation of RANKL gene expression in marrow stromal/preosteoblast cells in the bone microenvironment. J. Cell. Biochem. 103: 1747–1759, 2008.


Bone | 2015

STAT-6 mediates TRAIL induced RANK ligand expression in stromal/preosteoblast cells

Kumaran Sundaram; Yuvaraj Sambandam; Sundaravadivel Balasubramanian; Balakrishnan Pillai; Christina Voelkel-Johnson; William L. Ries; Sakamuri V. Reddy

Receptor activator of nuclear factor kappa-B ligand (RANKL) is a critical osteoclastogenic factor expressed in bone marrow stromal/osteoblast lineage cells. Tumor necrosis factor (TNF) related apoptosis-inducing ligand (TRAIL) levels are elevated in pathologic conditions such as multiple myeloma and inflammatory arthritis, and have been positively correlated with osteolytic markers. Osteoprotegerin (OPG) which inhibits osteoclastogenesis is a decoy receptor for RANKL and also known to interact with TRAIL. Herein, we show that TRAIL increases DR5 and DcR1 receptors but no change in the levels of DR4 and DcR2 expression in human bone marrow derived stromal/preosteoblast (SAKA-T) cell line. We further demonstrated that TRAIL treatment significantly decreased OPG mRNA expression. Interestingly, TRAIL treatment induced RANKL mRNA expression in these cells. In addition, TRAIL significantly increased NF-kB and c-Jun N-terminal kinase (JNK) activity. Human transcription factor array screening by real-time RT-PCR identified TRAIL up-regulation of the signal transducers and activators of the transcription (STAT)-6 expression in SAKA-T cells. TRAIL stimulation induced p-STAT-6 expression in human bone marrow derived primary stromal/preosteoblast cells. Confocal microscopy analysis further revealed p-STAT-6 nuclear localization in SAKA-T cells. Chromatin immunoprecipitation (ChIP) assay confirmed p-STAT-6 binding to the hRANKL gene distal promoter region. In addition, siRNA suppression of STAT-6 expression inhibits TRAIL increased hRANKL gene promoter activity. Thus, our results suggest that TRAIL induces RANKL expression through a STAT-6 dependent transcriptional regulatory mechanism in bone marrow stromal/preosteoblast cells.


Journal of Cellular Biochemistry | 2013

SOCS‐1/3 participation in FGF‐2 signaling to modulate RANK ligand expression in paget's disease of bone

Kumaran Sundaram; Joseph Senn; Sakamuri V. Reddy

Pagets disease of bone (PDB) is a chronic focal skeletal disorder characterized by excessive bone resorption followed by disorganized new bone formation. Measles virus nucleocapsid (MVNP) is implicated in pathogenesis of PDB. RANK ligand (RANKL), a critical osteoclastogenic factor expressed on bone marrow stromal/preosteoblast cells is upregulated in PDB. We recently demonstrated that fibroblast growth factor‐2 (FGF‐2) which induces RANKL expression is elevated in PDB. In this study, we hypothesized that FGF‐2 modulates suppressors of cytokine signaling (SOCS) to induce RANKL expression in PDB. We identified increased levels of SOCS‐1/3 mRNA expression in bone marrow mononuclear cells derived from patients with PDB compared to normal subjects. Interestingly, conditioned media obtained from MVNP transduced osteoclast progenitor cells significantly increased SOCS‐1/3 mRNA expression in stromal/preosteoblast cells. We next examined if SOCS participates in FGF‐2 signaling to modulate RANKL gene expression. We showed that FGF‐2 stimulation significantly increased SOCS‐1/3 expression in human bone marrow stromal/preosteoblast cells. In addition, co‐expression of SOCS‐1/3 with hRANKL gene promoter‐luciferase reporter plasmid in marrow stromal cells demonstrated a significant increase in promoter activity without FGF‐2 stimulation. Furthermore, siRNA inhibition of STAT‐1 suppresses FGF‐2 increased SOCS‐1/3 expression in these cells. Thus, our results suggest that SOCS participates in FGF‐2 modulation of RANKL expression in PDB. J. Cell. Biochem. 114: 2032–2038, 2013.

Collaboration


Dive into the Kumaran Sundaram's collaboration.

Top Co-Authors

Avatar

Sakamuri V. Reddy

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Yuvaraj Sambandam

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

William L. Ries

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Joseph Senn

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Sambandam Yuvaraj

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Srinivasan Shanmugarajan

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Azizul Haque

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

James S. Norris

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Keith L. Kirkwood

Medical University of South Carolina

View shared research outputs
Researchain Logo
Decentralizing Knowledge