Network


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

Hotspot


Dive into the research topics where Ganesh Rao is active.

Publication


Featured researches published by Ganesh Rao.


Cell | 2016

Molecular Profiling Reveals Biologically Discrete Subsets and Pathways of Progression in Diffuse Glioma

Michele Ceccarelli; Floris P. Barthel; Tathiane Maistro Malta; Thais S. Sabedot; Sofie R. Salama; Bradley A. Murray; Olena Morozova; Yulia Newton; Amie Radenbaugh; Stefano Maria Pagnotta; Samreen Anjum; Jiguang Wang; Ganiraju C. Manyam; Pietro Zoppoli; Shiyun Ling; Arjun A. Rao; Mia Grifford; Andrew D. Cherniack; Hailei Zhang; Laila M. Poisson; Carlos Gilberto Carlotti; Daniela Tirapelli; Arvind Rao; Tom Mikkelsen; Ching C. Lau; W. K. Alfred Yung; Raul Rabadan; Jason T. Huse; Daniel J. Brat; Norman L. Lehman

Therapy development for adult diffuse glioma is hindered by incomplete knowledge of somatic glioma driving alterations and suboptimal disease classification. We defined the complete set of genes associated with 1,122 diffuse grade II-III-IV gliomas from The Cancer Genome Atlas and used molecular profiles to improve disease classification, identify molecular correlations, and provide insights into the progression from low- to high-grade disease. Whole-genome sequencing data analysis determined that ATRX but not TERT promoter mutations are associated with increased telomere length. Recent advances in glioma classification based on IDH mutation and 1p/19q co-deletion status were recapitulated through analysis of DNA methylation profiles, which identified clinically relevant molecular subsets. A subtype of IDH mutant glioma was associated with DNA demethylation and poor outcome; a group of IDH-wild-type diffuse glioma showed molecular similarity to pilocytic astrocytoma and relatively favorable survival. Understanding of cohesive disease groups may aid improved clinical outcomes.


Oncogene | 2004

Sonic hedgehog and insulin-like growth factor signaling synergize to induce medulloblastoma formation from nestin-expressing neural progenitors in mice

Ganesh Rao; Carolyn A. Pedone; Luis Del Valle; Krzysztof Reiss; Eric C. Holland; Daniel W. Fults

Medulloblastoma (MB) is a malignant brain tumor that arises in the cerebellum of children. Activation of the Sonic hedgehog/Patched (Shh/Ptc) signaling pathway in neural progenitor cells of the cerebellum induces MBs in mice. The incomplete penetrance of tumor formation in mice, coupled with the low frequency of mutations in Shh/Ptc pathway genes in human tumors, suggests that other signaling molecules cooperate with Shh to enhance MB formation. We modeled the ability of insulin-like growth factor (IGF) signaling to induce MB using the RCAS/tv-a system, which allows postnatal gene transfer and expression in a cell-type-specific manner. We used RCAS retroviral vectors to target expression of Shh, IGF2, and activated Akt to nestin-expressing neural progenitors in the cerebella of newborn mice. The incidence of Shh-induced tumor formation (15%) was enhanced by coexpression with IGF2 (39%) and Akt (48%). Neither IGF2 nor Akt caused tumors when expressed independently. The induced tumors showed upregulated expression of insulin receptor substrate 1 and phosphorylated forms of IGF1 receptor and Akt, mimicking activated IGF signaling found in human MBs. These results indicate that combined activation of the Shh/Ptc and IGF signaling pathways is an important mechanism in MB pathogenesis.


Genes & Development | 2011

The transcriptional coactivator TAZ regulates mesenchymal differentiation in malignant glioma

Krishna P.L. Bhat; Katrina Salazar; Veerakumar Balasubramaniyan; Khalida Wani; Lindsey Heathcock; Faith Hollingsworth; Johanna D. James; Joy Gumin; Kristin Diefes; Se Hoon Kim; Alice Turski; Yasaman Azodi; Yuhui Yang; Tiffany Doucette; Howard Colman; Erik P. Sulman; Frederick F. Lang; Ganesh Rao; Sjef Copray; Brian Vaillant; Kenneth D. Aldape

Recent molecular classification of glioblastoma (GBM) has shown that patients with a mesenchymal (MES) gene expression signature exhibit poor overall survival and treatment resistance. Using regulatory network analysis of available expression microarray data sets of GBM, including The Cancer Genome Atlas (TCGA), we identified the transcriptional coactivator with PDZ-binding motif (TAZ), to be highly associated with the MES network. TAZ expression was lower in proneural (PN) GBMs and lower-grade gliomas, which correlated with CpG island hypermethylation of the TAZ promoter compared with MES GBMs. Silencing of TAZ in MES glioma stem cells (GSCs) decreased expression of MES markers, invasion, self-renewal, and tumor formation. Conversely, overexpression of TAZ in PN GSCs as well as murine neural stem cells (NSCs) induced MES marker expression and aberrant osteoblastic and chondrocytic differentiation in a TEAD-dependent fashion. Using chromatin immunoprecipitation (ChIP), we show that TAZ is directly recruited to a majority of MES gene promoters in a complex with TEAD2. The coexpression of TAZ, but not a mutated form of TAZ that lacks TEAD binding, with platelet-derived growth factor-B (PDGF-B) resulted in high-grade tumors with MES features in a murine model of glioma. Our studies uncover a direct role for TAZ and TEAD in driving the MES differentiation of malignant glioma.


Neuro-oncology | 2014

IDH1 mutant malignant astrocytomas are more amenable to surgical resection and have a survival benefit associated with maximal surgical resection

Jason Beiko; Dima Suki; Kenneth R. Hess; Benjamin D. Fox; Vincent Cheung; Matthew Cabral; N. A. Shonka; Mark R. Gilbert; Raymond Sawaya; Sujit S. Prabhu; Jeffrey S. Weinberg; Frederick F. Lang; Kenneth D. Aldape; Erik P. Sulman; Ganesh Rao; Ian E. McCutcheon; Daniel P. Cahill

BACKGROUND IDH1 gene mutations identify gliomas with a distinct molecular evolutionary origin. We sought to determine the impact of surgical resection on survival after controlling for IDH1 status in malignant astrocytomas-World Health Organization grade III anaplastic astrocytomas and grade IV glioblastoma. METHODS Clinical parameters including volumetric assessment of preoperative and postoperative MRI were recorded prospectively on 335 malignant astrocytoma patients: n = 128 anaplastic astrocytomas and n = 207 glioblastoma. IDH1 status was assessed by sequencing and immunohistochemistry. RESULTS IDH1 mutation was independently associated with complete resection of enhancing disease (93% complete resections among mutants vs 67% among wild-type, P < .001), indicating IDH1 mutant gliomas were more amenable to resection. The impact of residual tumor on survival differed between IDH1 wild-type and mutant tumors. Complete resection of enhancing disease among IDH1 wild-type tumors was associated with a median survival of 19.6 months versus 10.7 months for incomplete resection; however, no survival benefit was observed in association with further resection of nonenhancing disease (minimization of total tumor volume). In contrast, IDH1 mutants displayed an additional survival benefit associated with maximal resection of total tumor volume (median survival 9.75 y for >5 cc residual vs not reached for <5 cc, P = .025). CONCLUSIONS The survival benefit associated with surgical resection differs based on IDH1 genotype in malignant astrocytic gliomas. Therapeutic benefit from maximal surgical resection, including both enhancing and nonenhancing tumor, may contribute to the better prognosis observed in the IDH1 mutant subgroup. Thus, individualized surgical strategies for malignant astrocytoma may be considered based on IDH1 status.


Neurosurgery | 2009

Impact of intraoperative high-field magnetic resonance imaging guidance on glioma surgery: a prospective volumetric analysis.

Mustafa Aziz Hatiboglu; Jeffrey S. Weinberg; Dima Suki; Ganesh Rao; Sujit S. Prabhu; Komal Shah; Edward F. Jackson; Raymond Sawaya

OBJECTIVETo determine the impact of intraoperative magnetic resonance imaging (iMRI) on the decision to proceed with additional glioma resection during surgery and to maximize extent of resection (EOR). METHODSPatients who underwent craniotomy for glioma resection with high-field iMRI guidance were prospectively evaluated between September 2006 and August 2007. Volumetric analysis and EOR were assessed with iMRI, using postcontrast T1-weighted images for tumors showing contrast enhancement and T2-weighted images for nonenhancing tumors. RESULTSForty-six patients underwent resection using iMRI guidance, with iMRI being used to evaluate the EOR in 44 patients and for reregistration in 2 patients. Surgery was terminated after iMRI in 23 patients (52%) because gross total resection was achieved or because of residual tumor infiltration in an eloquent brain region. Twenty-one patients (47%) underwent additional resection of residual tumor after iMRI. For enhancing gliomas, the median EOR increased significantly from 84% (range, 59%–97%) to 99% (range, 85%–100%) with additional tumor removal after iMRI (P < 0.001). For nonenhancing gliomas, the median EOR increased (from 63% to 80%) with additional tumor removal after iMRI, but not significantly, owing to the small sample size (7 patients). Overall, the EOR increased from 76% (range, 35%–97%) to 96% (range, 48%–100%) (P < 0.001). Gross total resection was achieved after additional tumor removal after iMRI in 15 of 21 patients (71%). Overall, 29 patients (65%) experienced gross total resection, and in 15 (52%), this was achieved with the contribution of iMRI. CONCLUSIONHigh-field iMRI is a safe and reliable technique, and its use optimizes the extent of glioma resection.


Neoplasia | 2003

c-Myc Enhances Sonic Hedgehog-Induced Medulloblastoma Formation from Nestin-Expressing Neural Progenitors in Mice

Ganesh Rao; Carolyn A. Pedone; Cheryl M. Coffin; Eric C. Holland; Daniel W. Fults

Medulloblastomas are malignant brain tumors that arise in the cerebella of children. The presumed cells-of-origin are undifferentiated precursors of granule neurons that occupy the external granule layer (EGL) of the developing cerebellum. The overexpression of proteins that normally stimulate proliferation of neural progenitor cells may initiate medulloblastoma formation. Two known mitogens for neural progenitors are the c-Myc oncoprotein and Sonic hedgehog (Shh), a crucial determinant of embryonic pattern formation in the central nervous system. We modeled the ability of c-Myc and Shh to induce medulloblastoma in mice using the RCAS/tv-a system, which allows postnatal gene transfer and expression in a cell type-specific manner. We targeted the expression of Shh and c-Myc to nestin-expressing neural progenitor cells by injecting replication-competent ALV splice acceptor (RCAS) vectors into the cerebella of newborn mice. Following injection with RCAS-Shh alone, 3/32 (9%) mice developed medulloblastomas and 5/32 showed multifocal hyperproliferation of the EGL, possibly a precursor stage of medulloblastoma. Following injection with RCAS-Shh plus RCAS-Myc, 9/39 (23%) mice developed medulloblastomas. We conclude that nestin-expressing neural progenitors, present in the cerebellum at birth, can act as the cells-of-origin for medulloblastoma, and that c-Myc cooperates with Shh to enhance tumorigenicity.


Cancer Research | 2013

MiR-124 inhibits STAT3 signaling to enhance T cell-mediated immune clearance of glioma

Jun Wei; Fei Wang; Ling Yuan Kong; Shuo Xu; Tiffany Doucette; Sherise D. Ferguson; Yuhui Yang; Kayla McEnery; Krishan Jethwa; Olsi Gjyshi; Wei Qiao; Nicholas B. Levine; Frederick F. Lang; Ganesh Rao; Gregory N. Fuller; George A. Calin; Amy B. Heimberger

miRNAs (miR) have been shown to modulate critical gene transcripts involved in tumorigenesis, but their role in tumor-mediated immunosuppression is largely unknown. On the basis of miRNA gene expression in gliomas using tissue microarrays, in situ hybridization, and molecular modeling, miR-124 was identified as a lead candidate for modulating STAT3 signaling, a key pathway mediating immunosuppression in the tumor microenvironment. miR-124 is absent in all grades and pathologic types of gliomas. Upon upregulating miR-124 in glioma cancer stem cells (gCSC), the STAT3 pathway was inhibited, and miR-124 reversed gCSC-mediated immunosuppression of T-cell proliferation and induction of forkhead box P3 (Foxp3)(+) regulatory T cells (Treg). Treatment of T cells from immunosuppressed glioblastoma patients with miR-124 induced marked effector response including upregulation of interleukin (IL)-2, IFN-γ, and TNF-α. Both systemic administration of miR-124 or adoptive miR-124-transfected T-cell transfers exerted potent anti-glioma therapeutic effects in clonotypic and genetically engineered murine models of glioblastoma and enhanced effector responses in the local tumor microenvironment. These therapeutic effects were ablated in both CD4(+)- and CD8(+)-depleted mice and nude mouse systems, indicating that the therapeutic effect of miR-124 depends on the presence of a T-cell-mediated antitumor immune response. Our findings highlight the potential application of miR-124 as a novel immunotherapeutic agent for neoplasms and serve as a model for identifying miRNAs that can be exploited as immunotherapeutics.


Clinical Cancer Research | 2011

Glioma-Associated Cytomegalovirus Mediates Subversion of the Monocyte Lineage to a Tumor Propagating Phenotype

Kristine Dziurzynski; Jun Wei; Wei Qiao; Mustafa Aziz Hatiboglu; Ling Yuan Kong; Adam Wu; Yongtao Wang; Daniel P. Cahill; Nicholas B. Levine; Sujit S. Prabhu; Ganesh Rao; Raymond Sawaya; Amy B. Heimberger

Purpose: Cytomegalovirus (CMV) has been ubiquitously detected within high-grade gliomas, but its role in gliomagenesis has not been fully elicited. Experimental Design: Glioblastoma multiforme (GBM) tumors were analyzed by flow cytometry to determine CMV antigen expression within various glioma-associated immune populations. The glioma cancer stem cell (gCSC) CMV interleukin (IL)-10 production was determined by ELISA. Human monocytes were stimulated with recombinant CMV IL-10 and levels of expression of p-STAT3, VEGF (vascular endothelial growth factor), TGF-β, viral IE1, and pp65 were determined by flow cytometry. The influence of CMV IL-10–treated monocytes on gCSC biology was ascertained by functional assays. Results: CMV showed a tropism for macrophages (MΦ)/microglia and CD133+ gCSCs within GBMs. The gCSCs produce CMV IL-10, which induces human monocytes (the precursor to the central nervous system MΦs/microglia) to assume an M2 immunosuppressive phenotype (as manifested by downmodulation of the major histocompatibility complex and costimulatory molecules) while upregulating immunoinhibitory B7-H1. CMV IL-10 also induces expression of viral IE1, a modulator of viral replication and transcription in the monocytes. Finally, the CMV IL-10–treated monocytes produced angiogenic VEGF, immunosuppressive TGF-β, and enhanced migration of gCSCs. Conclusions: CMV triggers a feedforward mechanism of gliomagenesis by inducing tumor-supportive monocytes. Clin Cancer Res; 17(14); 4642–9. ©2011 AACR.


Neurosurgery Clinics of North America | 2011

Epidemiology of Metastatic Brain Tumors

Benjamin D. Fox; Vincent Cheung; Akash J. Patel; Dima Suki; Ganesh Rao

Metastatic tumors are the most common brain tumors in adults, and their incidence is increasing. An accurate understanding of the epidemiology of metastatic brain tumors is useful for health care professionals to allocate appropriate clinical, diagnostic, therapeutic, and research resources. Reported incidences in the literature are derived from epidemiologic population-based studies; clinical studies from surgical, radiological, and autopsy series; and reviews of hospital and clinical medical records. Despite these various sources of information, an accurate incidence of metastatic brain tumors is difficult, and current figures are estimates at best. Here, we review the available data regarding the epidemiology of metastatic brain tumors.


Journal of Neurosurgery | 2008

Os odontoideum: presentation, diagnosis, and treatment in a series of 78 patients

Paul Klimo; Peter Kan; Ganesh Rao; Ronald I. Apfelbaum; Douglas L. Brockmeyer

OBJECT The most contentious issue in the management of os odontoideum surrounds the decision to attempt atlantoaxial fusion in patients with asymptomatic lesions. The authors examined the clinical presentation and outcome in patients with os odontoideum who underwent surgical stabilization, with an emphasis on 3 patients who initially received conservative treatment and suffered delayed neurological injury. METHODS Seventy-eight patients (mean age 20.5 years; median 15 years) were identified in a 17-year retrospective review. The median follow-up period was 14 months (range 1-115 months). Neck pain was the most common symptom (64%), and 56% of patients presented after traumatic injury. Eighteen patients had neurological signs or symptoms at presentation, and an additional 15 had a history of intermittent or prior neurological symptoms. Fifteen patients had undergone > or = 1 attempt at atlantoaxial fusion elsewhere. RESULTS Seventy-seven patients underwent posterior fusion and rigid screw fixation combined with a graft/wire construct: 75 had C1-2 fusion and 2 had occipitocervical fusion. One patient had an odontoid screw placed. Fusion was achieved in all patients at a median of 4.8 months (range 2-17 months). Approximately 90% of patients had resolution or improvement of their neck pain or neurological symptoms. CONCLUSIONS The authors believe that patients with os odontoideum are at risk for future spinal cord compromise. Forty-four percent of our patients had myelopathic symptoms at referral, and 3 had significant neurological deterioration when a known os odontoideum was left untreated. This risk of late neurological deterioration should be considered when counseling patients. Stabilization using internal screw fixation techniques resulted in 100% fusion, whereas 15% of patients had previously undergone unsuccessful wire and external bracing attempts.

Collaboration


Dive into the Ganesh Rao's collaboration.

Top Co-Authors

Avatar

Sujit S. Prabhu

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jeffrey S. Weinberg

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Dima Suki

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Amy B. Heimberger

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Laurence D. Rhines

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Raymond Sawaya

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Frederick F. Lang

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Arvind Rao

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Gregory N. Fuller

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ian E. McCutcheon

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge