Network


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

Hotspot


Dive into the research topics where Neha Garg is active.

Publication


Featured researches published by Neha Garg.


Leukemia | 2012

Differential regulation of miR-21 and miR-146a by Epstein–Barr virus-encoded EBNA2

Paola Rosato; Eleni Anastasiadou; Neha Garg; Dido Lenze; Francesco Boccellato; Sara Vincenti; Martina Severa; Eliana M. Coccia; Rachele Bigi; Mara Cirone; Elisabetta Ferretti; Antonio Francesco Campese; Michael Hummel; Luigi Frati; Carlo Presutti; Alberto Faggioni; Pankaj Trivedi

The discovery of microRNA (miR) represents a novel paradigm in RNA-based regulation of gene expression and their dysregulation has become a hallmark of many a tumor. In virally associated cancers, the host–pathogen interaction could involve alteration in miR expression. Epstein–Barr virus (EBV)-encoded EBNA2 is indispensable for the capacity of the virus to transform B cells in vitro. Here, we studied how it affects cellular miRs. Extensive miR profiling of the virus-infected and EBNA2-transfected B lymphoma cells revealed that oncomiR miR-21 is positively regulated by this viral protein. Conversely, Burkitt’s lymphoma (BL) cell lines infected with EBNA2 lacking P3HR1 strain did not show any increase in miR-21. EBNA2 increased phosphorylation of AKT and this was directly correlated with increased miR-21. In contrast, miR-146a was downregulated by EBNA2 in B lymphoma cells. Low miR-146a expression correlates with an elevated level of IRAK1 and type I interferon in EBNA2 transfectants. Taken together, the present data suggest that EBNA2 might contribute to EBV-induced B-cell transformation by altering miR expression and in particular by increasing oncomiR-like miR-21 and by affecting the antiviral responses of the innate immune system through downregulation of its key regulator miR-146a.


The EMBO Journal | 2013

microRNA‐17‐92 cluster is a direct Nanog target and controls neural stem cell through Trp53inp1

Neha Garg; Agnese Po; Evelina Miele; Antonio Francesco Campese; Federica Begalli; Marianna Silvano; Paola Infante; Carlo Capalbo; Enrico De Smaele; Gianluca Canettieri; Lucia Di Marcotullio; Isabella Screpanti; Elisabetta Ferretti; Alberto Gulino

The transcription factor Nanog plays a critical role in the self‐renewal of embryonic stem cells as well as in neural stem cells (NSCs). microRNAs (miRNAs) are also involved in stemness regulation. However, the miRNA network downstream of Nanog is still poorly understood. High‐throughput screening of miRNA expression profiles in response to modulated levels of Nanog in postnatal NSCs identifies miR‐17‐92 cluster as a direct target of Nanog. Nanog controls miR‐17‐92 cluster by binding to the upstream regulatory region and maintaining high levels of transcription in NSCs, whereas Nanog/promoter association and cluster miRNAs expression are lost alongside differentiation. The two miR‐17 family members of miR‐17‐92 cluster, namely miR‐17 and miR‐20a, target Trp53inp1, a downstream component of p53 pathway. To support a functional role, the presence of miR‐17/20a or the loss of Trp53inp1 is required for the Nanog‐induced enhancement of self‐renewal of NSCs. We unveil an arm of the Nanog/p53 pathway, which regulates stemness in postnatal NSCs, wherein Nanog counteracts p53 signals through miR‐17/20a‐mediated repression of Trp53inp1.


International Journal of Cancer | 2015

Epstein‐Barr virus infection induces miR‐21 in terminally differentiated malignant B cells

Eleni Anastasiadou; Neha Garg; Rachele Bigi; Shivangi Yadav; Antonio Francesco Campese; Caterina Lapenta; Massimo Spada; Laura Cuomo; Annalisa Botta; Filippo Belardelli; Luigi Frati; Elisabetta Ferretti; Alberto Faggioni; Pankaj Trivedi

The association of Epstein‐Barr virus (EBV) with plasmacytoid malignancies is now well established but how the virus influences microRNA expression in such cells is not known. We have used multiple myeloma (MM) cell lines to address this issue and find that an oncomiR, miR‐21 is induced after in vitro EBV infection. The PU.1 binding site in miR‐21 promoter was essential for its activation by the virus. In accordance with its noted oncogenic functions, miR‐21 induction in EBV infected MM cells caused downregulation of p21 and an increase in cyclin D3 expression. EBV infected MM cells were highly tumorigenic in SCID mice. Given the importance of miR‐21 in plasmacytoid malignancies, our findings that EBV could further exacerbate the disease by inducing miR‐21 has interesting implications both in terms of diagnosis and future miR based therapeutical approaches for the virus associated plasmacytoid tumors.


Clinical Cancer Research | 2015

Pyrvinium Targets CD133 in Human Glioblastoma Brain Tumor–Initiating Cells

Chitra Venugopal; Robin M. Hallett; Parvez Vora; Branavan Manoranjan; Sujeivan Mahendram; Maleeha Qazi; Nicole McFarlane; Minomi Subapanditha; Sara Nolte; Mohini Singh; David Bakhshinyan; Neha Garg; Thusyanth Vijayakumar; Boleslaw Lach; John Provias; Kesava Reddy; Naresh Murty; Bradley W. Doble; Mickie Bhatia; John A. Hassell; Sheila K. Singh

Purpose: Clonal evolution of cancer may be regulated by determinants of stemness, specifically self-renewal, and current therapies have not considered how genetic perturbations or properties of stemness affect such functional processes. Glioblastoma-initiating cells (GICs), identified by expression of the cell surface marker CD133, are shown to be chemoradioresistant. In the current study, we sought to elucidate the functional role of CD133 in self-renewal and identify compounds that can specifically target this CD133+ treatment-refractory population. Experimental Design: Using gain/loss-of-function studies for CD133 we assessed the in vitro self-renewal and in vivo tumor formation capabilities of patient-derived glioblastoma cells. We generated a CD133 signature combined with an in silico screen to find compounds that target GICs. Self-renewal and proliferation assays on CD133-sorted samples were performed to identify the preferential action of hit compounds. In vivo efficacy of the lead compound pyrvinium was assessed in intracranial GIC xenografts and survival studies. Lastly, microarray analysis was performed on pyrvinium-treated GICs to discover core signaling events involved. Results: We discovered pyrvinium, a small-molecule inhibitor of GIC self-renewal in vitro and in vivo, in part through inhibition of Wnt/β-catenin signaling and other essential stem cell regulatory pathways. We provide a therapeutically tractable strategy to target self-renewing, chemoradioresistant, and functionally important CD133+ stem cells that drive glioblastoma relapse and mortality. Conclusions: Our study provides an integrated approach for the eradication of clonal populations responsible for cancer progression, and may apply to other aggressive and heterogeneous cancers. Clin Cancer Res; 21(23); 5324–37. ©2015 AACR.


Neuro-oncology | 2014

High-throughput microRNA profiling of pediatric high-grade gliomas

Evelina Miele; Francesca R. Buttarelli; Antonella Arcella; Federica Begalli; Neha Garg; Marianna Silvano; Agnese Po; Caterina Baldi; Giuseppe Carissimo; Manila Antonelli; Gian Paolo Spinelli; Carlo Capalbo; Vittoria Donofrio; Isabella Morra; Paolo Nozza; Alberto Gulino; Felice Giangaspero; Elisabetta Ferretti

Background High-grade gliomas (HGGs) account for 15% of all pediatric brain tumors and are a leading cause of cancer-related mortality and morbidity. Pediatric HGGs (pHGGs) are histologically indistinguishable from their counterpart in adulthood. However, recent investigations indicate that differences occur at the molecular level, thus suggesting that the molecular path to gliomagenesis in childhood is distinct from that of adults. MicroRNAs (miRNAs) have been identified as key molecules in gene expression regulation, both in development and in cancer. miRNAs have been investigated in adult high-grade gliomas (aHGGs), but scant information is available for pHGGs. Methods We explored the differences in microRNAs between pHGG and aHGG, in both fresh-frozen and paraffin-embedded tissue, by high-throughput miRNA profiling. We also evaluated the biological effects of miR-17-92 cluster silencing on a pHGG cell line. Results Comparison of miRNA expression patterns in formalin versus frozen specimens resulted in high correlation between both types of samples. The analysis of miRNA profiling revealed a specific microRNA pattern in pHGG with an overexpression and a proliferative role of the miR-17-92 cluster. Moreover, we highlighted a possible quenching function of miR-17-92 cluster on its target gene PTEN, together with an activation of tumorigenic signaling such as sonic hedgehog in pHGG. Conclusions Our results suggest that microRNA profiling represents a tool to distinguishing pediatric from adult HGG and that miR-17-92 cluster sustains pHGG.


Oncotarget | 2015

STAT3 pathway regulates lung-derived brain metastasis initiating cell capacity through miR-21 activation.

Mohini Singh; Neha Garg; Chitra Venugopal; Robin M. Hallett; Tomas Tokar; Nicole McFarlane; Sujeivan Mahendram; David Bakhshinyan; Branavan Manoranjan; Parvez Vora; Maleeha Qazi; Carolynn C. Arpin; Brent D. G. Page; Sina Haftchenary; David A. Rosa; Ping-Shan Lai; Rodolfo F. Gómez-Biagi; Ahmed M. Ali; Andrew M. Lewis; Mulu Geletu; Naresh Murty; John A. Hassell; Igor Jurisica; Patrick T. Gunning; Sheila K. Singh

Brain metastases (BM) represent the most common tumor to affect the adult central nervous system. Despite the increasing incidence of BM, likely due to consistently improving treatment of primary cancers, BM remain severely understudied. In this study, we utilized patient-derived stem cell lines from lung-to-brain metastases to examine the regulatory role of STAT3 in brain metastasis initiating cells (BMICs). Annotation of our previously described BMIC regulatory genes with protein-protein interaction network mapping identified STAT3 as a novel protein interactor. STAT3 knockdown showed a reduction in BMIC self-renewal and migration, and decreased tumor size in vivo. Screening of BMIC lines with a library of STAT3 inhibitors identified one inhibitor to significantly reduce tumor formation. Meta-analysis identified the oncomir microRNA-21 (miR-21) as a target of STAT3 activity. Inhibition of miR-21 displayed similar reductions in BMIC self-renewal and migration as STAT3 knockdown. Knockdown of STAT3 also reduced expression of known downstream targets of miR-21. Our studies have thus identified STAT3 and miR-21 as cooperative regulators of stemness, migration and tumor initiation in lung-derived BM. Therefore, STAT3 represents a potential therapeutic target in the treatment of lung-to-brain metastases.


Journal of Parasitology Research | 2011

Inhibition of the Growth of Plasmodium falciparum in Culture by Stearylamine-Phosphatidylcholine Liposomes

Gulam Mustafa Hasan; Neha Garg; Enna Dogra; Ranu Surolia; Prahlad C. Ghosh

We have examined the effect of stearylamine (SA) in liposomes on the viability of Plasmodium falciparum in culture by studying the inhibition of incorporation of [3H]-hypoxanthine in the nucleic acid of parasites. Stearylamine in liposomes significantly inhibits the growth of the parasites depending on the phospholipids composition. The maximum inhibition was observed when SA was delivered through Soya phosphatidylcholine (SPC) liposomes. The chain length of alkyl group and density of SA in liposomes play a significant role in inhibiting the growth of the parasites. Incorporation of either cholesterol or Distearylphosphatidylethanolamine−Methoxy-Polyethylene glycol-2000 (DSPE-mPEG-2000) in Soya phosphatidylcholine-stearylamine (SPC-SA) liposomes improves the efficacy. Intraerythrocytic entry of intact SPC-SA liposomes into infected erythrocytes was visualized using fluorescent microscopy. No hemolysis was observed in uninfected erythrocytes, and slight hemolysis was noted in infected erythrocytes at high concentrations of SPC-SA liposomes. Overall, our data suggested SA in SPC-liposomes might have potential application in malaria chemotherapy.


Stem Cells International | 2015

MicroRNA Regulation of Brain Tumour Initiating Cells in Central Nervous System Tumours

Neha Garg; Thusyanth Vijayakumar; David Bakhshinyan; Chitra Venugopal; Sheila K. Singh

CNS tumours occur in both pediatric and adult patients and many of these tumours are associated with poor clinical outcome. Due to a paradigm shift in thinking for the last several years, these tumours are now considered to originate from a small population of stem-like cells within the bulk tumour tissue. These cells, termed as brain tumour initiating cells (BTICs), are perceived to be regulated by microRNAs at the posttranscriptional/translational levels. Proliferation, stemness, differentiation, invasion, angiogenesis, metastasis, apoptosis, and cell cycle constitute some of the significant processes modulated by microRNAs in cancer initiation and progression. Characterization and functional studies on oncogenic or tumour suppressive microRNAs are made possible because of developments in sequencing and microarray techniques. In the current review, we bring recent knowledge of the role of microRNAs in BTIC formation and therapy. Special attention is paid to two highly aggressive and well-characterized brain tumours: gliomas and medulloblastoma. As microRNA seems to be altered in the pathogenesis of many human diseases, “microRNA therapy” may now have potential to improve outcomes for brain tumour patients. In this rapidly evolving field, further understanding of miRNA biology and its contribution towards cancer can be mined for new therapeutic tools.


Stem Cells International | 2016

MicroRNAs-Proteomic Networks Characterizing Human Medulloblastoma-SLCs.

Giuseppina Catanzaro; Zein Mersini Besharat; Neha Garg; Maurizio Ronci; Luisa Pieroni; Evelina Miele; Angela Mastronuzzi; Andrea Carai; Vincenzo Alfano; Agnese Po; Isabella Screpanti; Franco Locatelli; Andrea Urbani; Elisabetta Ferretti

Medulloblastoma (MB) is the most common malignant brain tumor of pediatric age and is characterized by cells expressing stem, astroglial, and neuronal markers. Among them, stem-like cells (hMB-SLCs) represent a fraction of the tumor cell population with the potential of self-renewal and proliferation and have been associated with tumor poor prognosis. In this context, microRNAs have been described as playing a pivotal role in stem cells differentiation. In our paper, we analyze microRNAs profile and genes expression of hMB-SLCs before and after Retinoic Acid- (RA-) induced differentiation. We aimed to identify pivotal players of specific pathways sustaining stemness and/or tumor development and progression and integrate the results of our recent proteomic study. Our results uncovered 22 differentially expressed microRNAs that were used as input together with deregulated genes and proteins in the Genomatix Pathway System (GePS) analysis revealing 3 subnetworks that could be interestingly involved in the maintenance of hMB-SLCs proliferation. Taken together, our findings highlight microRNAs, genes, and proteins that are significantly modulated in hMB-SLCs with respect to their RA-differentiated counterparts and could open new perspectives for prognostic and therapeutic intervention on MB.


Oncogene | 2018

BMI1 is a therapeutic target in recurrent medulloblastoma

David Bakhshinyan; Chitra Venugopal; Ashley Adile; Neha Garg; Branavan Manoranjan; Robin M. Hallett; Xin Wang; Sujeivan Mahendram; Parvez Vora; Thusyanth Vijayakumar; Minomi Subapanditha; Mohini Singh; Michelle Kameda-Smith; Maleeha Qazi; Nicole McFarlane; Aneet Mann; Olufemi Ajani; Blake Yarascavitch; Vijay Ramaswamy; Hamza Farooq; Sorana Morrissy; Liangxian Cao; Nadiya Sydorenko; Ramil Baiazitov; Wu Du; Josephine Sheedy; Marla Weetall; Young-Choon Moon; Chang-Sun Lee; Jacek M. Kwiecien

Medulloblastoma (MB) is the most frequent malignant pediatric brain tumor, representing 20% of newly diagnosed childhood central nervous system malignancies. Although advances in multimodal therapy yielded a 5-year survivorship of 80%, MB still accounts for the leading cause of childhood cancer mortality. In this work, we describe the epigenetic regulator BMI1 as a novel therapeutic target for the treatment of recurrent human Group 3 MB, a childhood brain tumor for which there is virtually no treatment option beyond palliation. Current clinical trials for recurrent MB patients based on genomic profiles of primary, treatment-naive tumors will provide limited clinical benefit since recurrent metastatic MBs are highly genetically divergent from their primary tumor. Using a small molecule inhibitor against BMI1, PTC-028, we were able to demonstrate complete ablation of self-renewal of MB stem cells in vitro. When administered to mice xenografted with patient tumors, we observed significant reduction in tumor burden in both local and metastatic compartments and subsequent increased survival, without neurotoxicity. Strikingly, serial in vivo re-transplantation assays demonstrated a marked reduction in tumor initiation ability of recurrent MB cells upon re-transplantation of PTC-028-treated cells into secondary recipient mouse brains. As Group 3 MB is often metastatic and uniformly fatal at recurrence, with no current or planned trials of targeted therapy, an efficacious targeted agent would be rapidly transitioned to clinical trials.

Collaboration


Dive into the Neha Garg'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
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge