Network


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

Hotspot


Dive into the research topics where Ramandeep Rattan is active.

Publication


Featured researches published by Ramandeep Rattan.


Journal of Immunology | 2004

The 15-Deoxy-δ12,14-Prostaglandin J 2 Inhibits the Inflammatory Response in Primary Rat Astrocytes via Down-Regulating Multiple Steps in Phosphatidylinositol 3-Kinase-Akt-NF-κB-p300 Pathway Independent of Peroxisome Proliferator-Activated Receptor γ

Shailendra Giri; Ramandeep Rattan; Avtar K. Singh; Inderjit Singh

Ligands for peroxisome proliferator-activated receptor γ (PPARγ), such as 15-deoxy-12,14-PGJ2 (15d-PGJ2), have been proposed as a new class of anti-inflammatory compounds because 15d-PGJ2 was able to inhibit the induction of inflammatory response genes such as inducible NO synthase (iNOS) and TNF (TNF-α) in a PPAR-dependent manner in various cell types. In primary astrocytes, the anti-inflammatory effects (inhibition of TNF-α, IL-1β, IL-6, and iNOS gene expression) of 15d-PGJ2 are observed to be independent of PPARγ. Overexpression (wild-type and dominant-negative forms) of PPARγ and its antagonist (GW9662) did not alter the 15d-PGJ2-induced inhibition of LPS/IFN-γ-mediated iNOS and NF-κB activation. The 15d-PGJ2 inhibited the inflammatory response by inhibiting IκB kinase activity, which leads to the inhibition of degradation of IκB and nuclear translocation of p65, thereby regulating the NF-κB pathway. Moreover, 15d-PGJ2 also inhibited the LPS/IFN-γ-induced PI3K-Akt pathway. The 15d-PGJ2 inhibited the recruitment of p300 by NF-κB (p65) and down-regulated the p300-mediated induction of iNOS and NF-κB luciferase reporter activity. Coexpression of constitutive active Akt and PI3K (p110) reversed the 15d-PGJ2-mediated inhibition of p300-induced iNOS and NF-κB luciferase activity. This study demonstrates that 15d-PGJ2 suppresses inflammatory response by inhibiting NF-κB signaling at multiple steps as well as by inhibiting the PI3K/Akt pathway independent of PPARγ in primary astrocytes.


PLOS ONE | 2013

Nanoceria: A Rare-Earth Nanoparticle as a Novel Anti-Angiogenic Therapeutic Agent in Ovarian Cancer

Shailendra Giri; Ajay S. Karakoti; Rondell P. Graham; Jacie Maguire; Christopher M. Reilly; Sudipta Seal; Ramandeep Rattan; Viji Shridhar

Ovarian cancer (OvCa) is the fifth most common cause of death from all cancers among women in United Sates and the leading cause of death from gynecological malignancies. While most OvCa patients initially respond to surgical debulking and chemotherapy, 75% of patients later succumb to the disease. Thus, there is an urgent need to test novel therapeutic agents to counteract the high mortality rate associated with OvCa. In this context, we have developed and engineered Nanoceria (NCe), nanoparticles of cerium oxide, possessing anti-oxidant properties, to be used as a therapeutic agent in OvCa. We show for the first time that NCe significantly inhibited production of reactive oxygen species (ROS) in A2780 cells, attenuated growth factor (SDF1, HB-EGF, VEGF165 and HGF) mediated cell migration and invasion of SKOV3 cells, without affecting the cell proliferation. NCe treatment also inhibited VEGF165 induced proliferation, capillary tube formation, activation of VEGFR2 and MMP2 in human umbilical vascular endothelial cells (HUVEC). NCe (0.1 mg/kg body weigh) treatment of A2780 ovarian cancer cells injected intra-peritoneally in nude mice showed significant reduction (p<0.002) in tumor growth accompanied by decreased tumor cell proliferation as evident from reduced tumor size and Ki67 staining. Accumulation of NCe was found in tumors isolated from treated group using transmission electron microscopy (TEM) and inductively coupled plasma mass spectroscopy (ICP-MS). Reduction of the tumor mass was accompanied by attenuation of angiogenesis, as observed by reduced CD31 staining and specific apoptosis of vascular endothelial cells. Collectively, these results indicate that cerium oxide based NCe is a novel nanoparticle that can potentially be used as an anti-angiogenic therapeutic agent in ovarian cancer.


Journal of Oncology | 2012

Metformin: An Emerging New Therapeutic Option for Targeting Cancer Stem Cells and Metastasis

Ramandeep Rattan; Rouba Ali Fehmi; Adnan R. Munkarah

Metastasis is an intricate process by which a small number of cancer cells from the primary tumor site undergo numerous alterations, which enables them to form secondary tumors at another and often multiple sites in the host. Transition of a cancer cell from epithelial to mesenchymal phenotype is thought to be the first step in the progression of metastasis. Recently, the recognition of cancer stem cells has added to the perplexity in understanding metastasis, as studies suggest cancer stem cells to be the originators of metastasis. All current and investigative drugs have been unable to prevent or reverse metastasis, as a result of which most metastatic cancers are incurable. A potential drug that can be considered is metformin, an oral hypoglycemic drug. In this review we discuss the potential of metformin in targeting both epithelial to mesenchymal transition and cancer stem cells in combating cancer metastases.


Journal of Medicinal Chemistry | 2016

Isatin Derived Spirocyclic Analogues with α-Methylene-γ-butyrolactone as Anticancer Agents: A Structure–Activity Relationship Study

Sandeep Rana; Elizabeth C. Blowers; Calvin Tebbe; Jacob I. Contreras; Prakash Radhakrishnan; Smitha Kizhake; Tian Zhou; Rajkumar N. Rajule; Jamie L. Arnst; Adnan R. Munkarah; Ramandeep Rattan; Amarnath Natarajan

Design, synthesis, and evaluation of α-methylene-γ-butyrolactone analogues and their evaluation as anticancer agents is described. SAR identified a spirocyclic analogue 19 that inhibited TNFα-induced NF-κB activity, cancer cell growth and tumor growth in an ovarian cancer model. A second iteration of synthesis and screening identified 29 which inhibited cancer cell growth with low-μM potency. Our data suggest that an isatin-derived spirocyclic α-methylene-γ-butyrolactone is a suitable core for optimization to identify novel anticancer agents.


Journal of Ovarian Research | 2015

A metabolomic approach to identifying platinum resistance in ovarian cancer

Laila M. Poisson; Adnan R. Munkarah; Hala Madi; Indrani Datta; Sharon Hensley-Alford; Calvin Tebbe; Thomas Buekers; Shailendra Giri; Ramandeep Rattan

BackgroundAcquisition of metabolic alterations has been shown to be essential for the unremitting growth of cancer, yet the relation of such alterations to chemosensitivity has not been investigated. In the present study our aim was to identify the metabolic alterations that are specifically associated with platinum resistance in ovarian cancer. A global metabolic analysis of the A2780 platinum-sensitive and its platinum-resistant derivative C200 ovarian cancer cell line was performed utilizing ultra-high performance liquid chromatography/mass spectroscopy and gas chromatography/mass spectroscopy. Per-metabolite comparisons were made between cell lines and an interpretive analysis was carried out using the Kyoto Encyclopedia of Genes and Genomes (KEGG) metabolic library and the Ingenuity exogenous molecule library.ResultsWe observed 288 identified metabolites, of which 179 were found to be significantly different (t-test p < 0.05) between A2780 and C200 cells. Of these, 70 had increased and 109 had decreased levels in platinum resistant C200 cells. The top altered KEGG pathways based on number or impact of alterations involved the cysteine and methionine metabolism. An Ingenuity Pathway Analysis also revealed that the methionine degradation super-pathway and cysteine biosynthesis are the top two canonical pathways affected. The highest scoring network of altered metabolites was related to carbohydrate metabolism, energy production, and small molecule biochemistry. Compilation of KEGG analysis and the common network molecules revealed methionine and associated pathways of glutathione synthesis and polyamine biosynthesis to be most significantly altered.ConclusionOur findings disclose that the chemoresistant C200 ovarian cancer cells have distinct metabolic alterations that may contribute to its platinum resistance. This distinct metabolic profile of platinum resistance is a first step towards biomarker development for the detection of chemoresistant disease and metabolism-based drug targets specific for chemoresistant tumors.


Journal of Immunology | 2016

AMP-Activated Protein Kinase Suppresses Autoimmune Central Nervous System Disease by Regulating M1-Type Macrophage-Th17 Axis.

Ashutosh Mangalam; Ramandeep Rattan; Hamid Suhail; Jaspreet Singh; Nasrul Hoda; Mandar Deshpande; Sadanand Fulzele; Alexander Denic; Viji Shridhar; Ashok Kumar; Benoit Viollet; Moses Rodriguez; Shailendra Giri

The AMP-activated protein kinase, AMPK, is an energy-sensing, metabolic switch implicated in various metabolic disorders; however, its role in inflammation is not well defined. We have previously shown that loss of AMPK exacerbates experimental autoimmune encephalomyelitis (EAE) disease severity. In this study, we investigated the mechanism through which AMPK modulates inflammatory disease like EAE. AMPKα1 knockout (α1KO) mice with EAE showed severe demyelination and inflammation in the brain and spinal cord compared with wild-type due to higher expression of proinflammatory Th17 cytokines, including IL-17, IL-23, and IL-1β, impaired blood–brain barrier integrity, and increased infiltration of inflammatory cells in the CNS. Infiltrated CD4 cells in the brains and spinal cords of α1KO with EAE were significantly higher compared with wild-type EAE and were characterized as IL-17 (IL-17 and GM-CSF double-positive) CD4 cells. Increased inflammatory response in α1KO mice was due to polarization of macrophages (Mϕ) to proinflammatory M1 type phenotype (IL-10lowIL-23/IL-1β/IL-6high), and these M1 Mϕ showed stronger capacity to induce allogenic as well as Ag-specific (myelin oligodendrocyte glycoprotein [MOG]35–55) T cell response. Mϕ from α1KO mice also enhanced the encephalitogenic property of MOG35–55–primed CD4 T cells in B6 mice. The increased encephalitogenic MOG-restricted CD4+ T cells were due to an autocrine effect of IL-1β/IL-23–mediated induction of IL-6 production in α1KO Mϕ, which in turn induce IL-17 and GM-CSF production in CD4 cells. Collectively, our data indicate that AMPK controls the inflammatory disease by regulating the M1 phenotype–Th17 axis in an animal model of multiple sclerosis.


Journal of Biological Chemistry | 2015

Untargeted plasma metabolomics identifies endogenous metabolite with drug-like properties in chronic animal model of multiple sclerosis

Laila M. Poisson; Hamid Suhail; Jaspreet Singh; Indrani Datta; Aleksandar Denic; Krzysztof Labuzek; Nasrul Hoda; Ashray Shankar; Ashok Kumar; Mirela Cerghet; Stanton B. Elias; Robert P. Mohney; Moses Rodriguez; Ramandeep Rattan; Ashutosh Mangalam; Shailendra Giri

We performed untargeted metabolomics in plasma of B6 mice with experimental autoimmune encephalitis (EAE) at the chronic phase of the disease in search of an altered metabolic pathway(s). Of 324 metabolites measured, 100 metabolites that mapped to various pathways (mainly lipids) linked to mitochondrial function, inflammation, and membrane stability were observed to be significantly altered between EAE and control (p < 0.05, false discovery rate <0.10). Bioinformatics analysis revealed six metabolic pathways being impacted and altered in EAE, including α-linolenic acid and linoleic acid metabolism (PUFA). The metabolites of PUFAs, including ω-3 and ω-6 fatty acids, are commonly decreased in mouse models of multiple sclerosis (MS) and in patients with MS. Daily oral administration of resolvin D1, a downstream metabolite of ω-3, decreased disease progression by suppressing autoreactive T cells and inducing an M2 phenotype of monocytes/macrophages and resident brain microglial cells. This study provides a proof of principle for the application of metabolomics to identify an endogenous metabolite(s) possessing drug-like properties, which is assessed for therapy in preclinical mouse models of MS.


Cancer | 2015

Protective effect of bisphosphonates on endometrial cancer incidence in data from the Prostate, Lung, Colorectal and Ovarian (PLCO) cancer screening trial.

Sharon Hensley Alford; Ramandeep Rattan; Thomas E. Buekers; Adnan R. Munkarah

Preclinical studies have demonstrated antitumor effects of bisphosphonates. The objective of the current study was to determine the effect of exposure to bisphosphonate on the incidence of endometrial cancer.


PLOS ONE | 2014

Preclinical Therapeutic Potential of a Nitrosylating Agent in the Treatment of Ovarian Cancer

Shailendra Giri; Ramandeep Rattan; Mandar Deshpande; Jacie Maguire; Zachary P. Johnson; Rondell P. Graham; Viji Shridhar

This study examines the role of s-nitrosylation in the growth of ovarian cancer using cell culture based and in vivo approaches. Using the nitrosylating agent, S-nitrosoglutathione (GSNO), a physiological nitric oxide molecule, we show that GSNO treatment inhibited proliferation of chemoresponsive and chemoresistant ovarian cancer cell lines (A2780, C200, SKVO3, ID8, OVCAR3, OVCAR4, OVCAR5, OVCAR7, OVCAR8, OVCAR10, PE01 and PE04) in a dose dependent manner. GSNO treatment abrogated growth factor (HB-EGF) induced signal transduction including phosphorylation of Akt, p42/44 and STAT3, which are known to play critical roles in ovarian cancer growth and progression. To examine the therapeutic potential of GSNO in vivo, nude mice bearing intra-peritoneal xenografts of human A2780 ovarian carcinoma cell line (2×106) were orally administered GSNO at the dose of 1 mg/kg body weight. Daily oral administration of GSNO significantly attenuated tumor mass (p<0.001) in the peritoneal cavity compared to vehicle (phosphate buffered saline) treated group at 4 weeks. GSNO also potentiated cisplatin mediated tumor toxicity in an A2780 ovarian carcinoma nude mouse model. GSNO’s nitrosylating ability was reflected in the induced nitrosylation of various known proteins including NFκB p65, Akt and EGFR. As a novel finding, we observed that GSNO also induced nitrosylation with inverse relationship at tyrosine 705 phosphorylation of STAT3, an established player in chemoresistance and cell proliferation in ovarian cancer and in cancer in general. Overall, our study underlines the significance of S-nitrosylation of key cancer promoting proteins in modulating ovarian cancer and proposes the therapeutic potential of nitrosylating agents (like GSNO) for the treatment of ovarian cancer alone or in combination with chemotherapeutic drugs.


Science immunology | 2017

Suppression of FIP200 and autophagy by tumor-derived lactate promotes naïve T cell apoptosis and affects tumor immunity

Houjun Xia; Wei Wang; Joel Crespo; Ilona Kryczek; Wei Li; Shuang Wei; Zhaoqun Bian; Tomasz Maj; Ming-Xiao He; Rebecca J. Liu; You-Wen He; Ramandeep Rattan; Adnan R. Munkarah; Jun-Lin Guan; Weiping Zou

Tumor-derived lactate induced naïve T cell apoptosis via suppressing FIP200 expression and autophagy formation, resulting in mitochondria activation and imbalance of Bcl-2 family members in T cells. Tumor immunity flounders without FIP200 The tumor microenvironment impairs the function of effector and memory T cells, but its effect on naïve T cells is not well understood. Xia et al. show that the autophagy component FAK family–interacting protein of 200 kDa (FIP200) is critical for promoting antitumor responses mediated by T cells. Ovarian cancer patients and mouse tumor models had significantly higher rates of apoptosis and impaired autophagy in naïve T cells, as well as the selective loss of FIP200, which disrupted the balance of pro- and antiapoptotic factors. Tumor-derived lactate was shown to suppress FIP200 expression, thus causing enhanced apoptosis and attenuated antitumor responses. Naïve T cells are poorly studied in cancer patients. We report that naïve T cells are prone to undergo apoptosis due to a selective loss of FAK family–interacting protein of 200 kDa (FIP200) in ovarian cancer patients and tumor-bearing mice. This results in poor antitumor immunity via autophagy deficiency, mitochondria overactivation, and high reactive oxygen species production in T cells. Mechanistically, loss of FIP200 disables the balance between proapoptotic and antiapoptotic Bcl-2 family members via enhanced argonaute 2 (Ago2) degradation, reduced Ago2 and microRNA1198-5p complex formation, less microRNA1198-5p maturation, and consequently abolished microRNA1198-5p–mediated repression on apoptotic gene Bak1. Bcl-2 overexpression and mitochondria complex I inhibition rescue T cell apoptosis and promoted tumor immunity. Tumor-derived lactate translationally inhibits FIP200 expression by down-regulating the nicotinamide adenine dinucleotide level while potentially up-regulating the inhibitory effect of adenylate-uridylate–rich elements within the 3′ untranslated region of Fip200 mRNA. Thus, tumors metabolically target naïve T cells to evade immunity.

Collaboration


Dive into the Ramandeep Rattan's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

J. Chhina

Henry Ford Health System

View shared research outputs
Top Co-Authors

Avatar

Calvin Tebbe

Henry Ford Health System

View shared research outputs
Top Co-Authors

Avatar

Ismail Mert

Wayne State University

View shared research outputs
Top Co-Authors

Avatar

S. Dar

Henry Ford Health System

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

R.T. Morris

Wayne State University

View shared research outputs
Top Co-Authors

Avatar

Z. Al-Wahab

Wayne State University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge