Sagar R. Shah
Johns Hopkins University
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Sagar R. Shah.
Journal of Neurosurgery | 2011
Wesley Hsu; Ahmed Mohyeldin; Sagar R. Shah; Colette M. J. ap Rhys; Lakesha F. Johnson; Neda I. Sedora-Roman; Thomas A. Kosztowski; Ola Awad; Edward F. McCarthy; David M. Loeb; Jean Paul Wolinsky; Ziya L. Gokaslan; Alfredo Quinones-Hinojosa
OBJECT Chordoma is a malignant bone neoplasm hypothesized to arise from notochordal remnants along the length of the neuraxis. Recent genomic investigation of chordomas has identified T (Brachyury) gene duplication as a major susceptibility mutation in familial chordomas. Brachyury plays a vital role during embryonic development of the notochord and has recently been shown to regulate epithelial-to-mesenchymal transition in epithelial-derived cancers. However, current understanding of the role of this transcription factor in chordoma is limited due to the lack of availability of a fully characterized chordoma cell line expressing Brachyury. Thus, the objective of this study was to establish the first fully characterized primary chordoma cell line expressing gain of the T gene locus that readily recapitulates the original parental tumor phenotype in vitro and in vivo. METHODS Using an intraoperatively obtained tumor sample from a 61-year-old woman with primary sacral chordoma, a chordoma cell line (JHC7, or Johns Hopkins Chordoma Line 7) was established. Molecular characterization of the primary tumor and cell line was conducted using standard immunostaining and Western blotting. Chromosomal aberrations and genomic amplification of the T gene in this cell line were determined. Using this cell line, a xenograft model was established and the histopathological analysis of the tumor was performed. Silencing of Brachyury and changes in gene expression were assessed. RESULTS The authors report, for the first time, the successful establishment of a chordoma cell line (JHC7) from a patient with pathologically confirmed sacral chordoma. This cell line readily forms tumors in immunodeficient mice that recapitulate the parental tumor phenotype with conserved histological features consistent with the parental tumor. Furthermore, it is demonstrated for the first time that silencing of Brachyury using short hairpin RNA renders the morphology of chordoma cells to a more differentiated-like state and leads to complete growth arrest and senescence with an inability to be passaged serially in vitro. CONCLUSIONS This report represents the first xenograft model of a sacral chordoma line described in the literature and the first cell line established with stable Brachyury expression. The authors propose that Brachyury is an attractive therapeutic target in chordoma and that JHC7 will serve as a clinically relevant model for the study of this disease.
Stem Cells Translational Medicine | 2015
Christopher Smith; Kaisorn L. Chaichana; Young M. Lee; Benjamin Lin; Kevin M. Stanko; Thomas O’Donnell; Saksham Gupta; Sagar R. Shah; Joanne Wang; Olindi Wijesekera; Michael Delannoy; Andre Levchenko; Alfredo Quiñones-Hinojosa
Recent research advances have established mesenchymal stem cells (MSCs) as a promising vehicle for therapeutic delivery. Their intrinsic tropism for brain injury and brain tumors, their lack of immunogenicity, and their ability to breach the blood‐brain barrier make these cells an attractive potential treatment of brain disorders, including brain cancer. Despite these advantages, the efficiency of MSC homing to the brain has been limited in commonly used protocols, hindering the feasibility of such therapies. In the present study, we report a reproducible, comprehensive, cell culture‐based approach to enhance human adipose‐derived MSC (hAMSC) engraftment to brain tumors. We used micro‐ and nanotechnological tools to systematically model several steps in the putative homing process. By pre‐exposing hAMSCs to glioma‐conditioned media and the extracellular matrix proteins fibronectin and laminin, we achieved significant enhancements of the individual homing steps in vitro. This homing was confirmed in an in vivo rodent model of brain cancer. This comprehensive, cell‐conditioning approach provides a novel method to enhance stem cell homing to gliomas and, potentially, other neurological disorders.
Carcinogenesis | 2014
Yunping Hu; Akiva Mintz; Sagar R. Shah; Alfredo Quiñones-Hinojosa; Wesley Hsu
Recent evidence suggests that the expression of brachyury is necessary for chordoma growth. However, the mechanism associated with brachyury-regulated cell growth is poorly understood. Fibroblast growth factor (FGF), a regulator of brachyury expression in normal tissue, may also play an important role in chordoma pathophysiology. Using a panel of chordoma cell lines, we explored the role of FGF signaling and brachyury in cell growth and survival. Western blots showed that all chordoma cell lines expressed fibroblast growth factor receptor 2 (FGFR2), FGFR3, mitogen-activated protein kinase kinase (MEK) and extracellular signal-regulated kinase (ERK), whereas no cell lines expressed FGFR1 and FGFR4. Results of enzyme-linked immunosorbent assay indicated that chordoma cells produced FGF2. Neutralization of FGF2 inhibited MEK/ERK phosphorylation, decreased brachyury expression and induced apoptosis while reducing cell growth. Activation of the FGFR/MEK/ERK/brachyury pathway by FGF2-initiated phosphorylation of FGFR substrate 2 (FRS2)-α (Tyr196) prevented apoptosis while promoting cell growth and epithelial-mesenchymal transition (EMT). Immunofluorescence staining showed that FGF2 promoted the translocation of phosphorylated ERK to the nucleus and increased brachyury expression. The selective inhibition of FGFR, MEK and ERK phosphorylation by PD173074, PD0325901 and PD184352, respectively, decreased brachyury expression, induced apoptosis, and inhibited cell growth and EMT. Moreover, knockdown of brachyury by small hairpin RNA reduced FGF2 secretion, inhibited FGFR/MEK/ERK phosphorylation and blocked the effects of FGF2 on cell growth, apoptosis and EMT. Those findings highlight that FGFR/MEK/ERK/brachyury pathway coordinately regulates chordoma cell growth and survival and may represent a novel chemotherapeutic target for chordoma.
Journal of Neuro-oncology | 2015
Dimitrios Mathios; Jacob Ruzevick; Christopher Jackson; Haiying Xu; Sagar R. Shah; Janis M. Taube; Peter C. Burger; Edward F. McCarthy; Alfredo Quiñones-Hinojosa; Drew M. Pardoll; Michael Lim
Chordomas are rare malignant tumors that are postulated to arise from remnants of the notochord. Currently, the interaction between chordomas and the host immune system is poorly understood. The checkpoint protein, PD-1 is expressed by circulating lymphocytes and is a marker of activation and exhaustion. Its ligands, PD-L1 (B7-H1, CD274) and PD-L2 (B7-DC, CD273), are expressed on a variety of human cancers; however this pathway has not been previously reported in chordomas. We used flow cytometric and RT-PCR analysis in three established primary and recurrent chordoma cell lines (U-CH1, U-CH2, and JHC7) as well as immunohistochemical analysis of chordoma tissues from 10 patients to identify and localize expression of PD-1 pathway proteins. PD-1 ligands are not constitutively expressed by chordoma cells, but their expression is induced in the setting of pro-inflammatory cytokines in all cell lines examined. In paraffin embedded tissues, we found that tumor infiltrating lymphocytes expressed PD-1 in 3/6 cases. We also found that, although chordoma cells did not express significant levels of PD-L1, PD-L1 expression was observed on tumor-infiltrating macrophages and tumor infiltrating lymphocytes. Our study suggests that PD-1, PD-L1, and PD-L2 are present in the microenvironment of a subset of chordomas analyzed. Future studies are needed to evaluate the contribution of the PD-1 pathway to the immunosuppressive microenvironment of chordomas.
Neurosurgery | 2012
Wesley Hsu; Ahmed Mohyeldin; Sagar R. Shah; Ziya L. Gokaslan; Alfredo Quinones-Hinojosa
The management of spinal column tumors continues to be a challenge for clinicians. The mechanisms of tumor recurrence after surgical intervention as well as resistance to radiation and chemotherapy continue to be elucidated. Furthermore, the pathophysiology of metastatic spread remains an area of active investigation. There is a growing body of evidence pointing to the existence of a subset of tumor cells with high tumorigenic potential in many spine cancers that exhibit characteristics similar to those of stem cells. The ability to self-renew and differentiate into multiple lineages is the hallmark of stem cells, and tumor cells that exhibit these characteristics have been described as cancer stem cells (CSCs). The mechanisms that allow nonmalignant stem cells to promote normal developmental programming by way of enhanced proliferation, promotion of angiogenesis, and increased motility may be used by CSCs to fuel carcinogenesis. The purpose of this review is to discuss what is known about the role of CSCs in tumors of the osseous spine. First, this article reviews the fundamental concepts critical to understanding the role of CSCs with respect to chemoresistance, radioresistance, and metastatic disease. This discussion is followed by a review of what is known about the role of CSCs in the most common primary tumors of the osseous spine.
Journal of Neurosurgery | 2013
Jonathan Isaacs; Joseph Feher; Mary S. Shall; Scott Vota; Michael A. Fox; Satya Mallu; Ashkon Razavi; Ilvy Friebe; Sagar R. Shah; Nathalie Spita
OBJECT Suboptimal recovery following repair of major peripheral nerves has been partially attributed to denervation atrophy. Administration of anabolic steroids in conjunction with neurotization may improve functional recovery of chronically denervated muscle. The purpose of this study was to evaluate the effect of the administration of nandrolone on muscle recovery following prolonged denervation in a rat model. METHODS Eight groups of female Sprague-Dawley rats (15 rats per group, 120 in all) were divided into 3- or 6-month denervated hind limb and sham surgery groups and, then, nandrolone treatment groups and sham treatment groups. Evaluation of treatment effects included nerve conduction, force of contraction, comparative morphology, histology (of muscle fibers), protein electrophoresis (for muscle fiber grouping), and immunohistochemical evaluation. RESULTS Although a positive trend was noted, neither reinnervated nor normal muscle showed a statistically significant increase in peak muscle force following nandrolone treatment. Indirect measures, including muscle mass (weight and diameter), muscle cell size, muscle fiber type, and satellite cell counts, all failed to support significant anabolic effect. CONCLUSIONS There does not seem to be a functional benefit from nandrolone treatment following reinnervation of either mild or moderately atrophic muscle (related to prolonged denervation) in a rodent model.
Cell Reports | 2017
Sagar R. Shah; Justin M. David; Nathaniel D. Tippens; Ahmed Mohyeldin; Juan Carlos Martinez-Gutierrez; Sara Ganaha; Paula Schiapparelli; Duane H. Hamilton; Claudia Palena; Andre Levchenko; Alfredo Quinones-Hinojosa
Molecular factors that define stem cell identity have recently emerged as oncogenic drivers. For instance, brachyury, a key developmental transcriptional factor, is also implicated in carcinogenesis, most notably of chordoma, through mechanisms that remain elusive. Here, we show that brachyury is a crucial regulator of stemness in chordoma and in more common aggressive cancers. Furthermore, this effect of brachyury is mediated by control of synthesis and stability of Yes-associated protein (YAP), a key regulator of tissue growth and homeostasis, providing an unexpected mechanism of control of YAP expression. We further demonstrate that the brachyury-YAP regulatory pathway is associated with tumor aggressiveness. These results elucidate a mechanism of controlling both tumor stemness and aggressiveness through regulatory coupling of two developmental factors.
Neurosurgery | 2016
Sagar R. Shah; Nathaniel D. Tippens; Jin Seok Park; Ahmed Mohyeldin; Guillermo Vela; Juan Carlos Martinez-Gutierrez; Seth S. Margolis; Susanne Schmidt; Andre Levchenko; Alfredo Quinones-Hinojosa
INTRODUCTION Molecular pathways linking cell polarization and migration to extracellular cues regulate many pathological processes, including progression of aggressive and infiltrative cancers. Glioblastoma (GBM), the most common and lethal form of primary brain cancer, is characterized by its pronounced ability to disseminate into the intricate microenvironment of the human brain, confounding surgical excision and radiotherapy, leading to a median patient survival of 14 months. Progression results from defects in molecular pathways linking cell migration and invasion into surrounding tissue. However, the molecular engines are not known. METHODS Using patient-derived GBM tissues and cells, we profiled the expression of network moieties via Western blotting, quantitative real-time polymerase chain reaction and performed live cell time-lapse microscopy, bioinformatics analyses, in vivo intracranial GBM experiments using genetic and pharmacological inhibitors to delineate a prodispersal mechanism for management and treatment of GBMs. RESULTS Yes-associated protein (YAP), a transcriptional coactivator, is overexpressed and hyperactive in 78% of GBMs and 50% of metastatic tumors to the brain (P < .05). Our studies demonstrate that YAP activates a Rho-GTPase switch to potentiate migratory speed by interacting with canonical pathways through direct transcriptional control. In addition, YAP mediates a proinvasive genetic network by direct posttranslational regulation. By coupling the regulation of migration and invasion, YAP drives tumor cell dispersal in vitro and in vivo (P < .05). Hyperactivation of this YAP-driven network in GBM confers poor patient outcome in clinical biopsies and The Cancer Genome Atlas (P < .05), suggesting a new signature in clinical prognosis of this aggressive and infiltrative cancer. Targeting this network using a proprietary pharmacological inhibitor attenuates tumor dispersal and growth (P < .05). CONCLUSION YAP can critically control cellular locomotion through direct interaction with canonical molecular pathways controlling invasion and migration. Understanding the molecular underpinnings of this network is vital to the development of imperative prognostic and treatment approaches for cancer such as the new proprietary pharmacological inhibitor presented in this study.INTRODUCTION: Molecular pathways linking cell polarization and migration to extracellular cues regulate many pathological processes, including progression of aggressive and infiltrative cancers. Glioblastoma (GBM), the most common and lethal form of primary brain cancer, is characterized by its pronounced ability to disseminate into the intricate microenvironment of the human brain, confounding surgical excision and radiotherapy, leading to a median patient survival of 14 months. Progression results from defects in molecular pathways linking cell migration and invasion into surrounding tissue. However, the molecular engines are not known.
PLOS ONE | 2018
Christopher T. Saeui; Alison V. Nairn; Melina Galizzi; Christopher Douville; Prateek Gowda; Marian Park; Vrinda Dharmarha; Sagar R. Shah; Amelia Clarke; Melissa A. Austin; Kelley W. Moremen; Kevin J. Yarema
In this report we use ‘high-flux’ tributanoyl-modified N-acetylmannosamine (ManNAc) analogs with natural N-acetyl as well as non-natural azido- and alkyne N-acyl groups (specifically, 1,3,4-O-Bu3ManNAc, 1,3,4-O-Bu3ManNAz, and 1,3,4-O-Bu3ManNAl respectively) to probe intracellular sialic acid metabolism in the near-normal MCF10A human breast cell line in comparison with earlier stage T-47D and more advanced stage MDA-MB-231 breast cancer lines. An integrated view of sialic acid metabolism was gained by measuring intracellular sialic acid production in tandem with transcriptional profiling of genes linked to sialic acid metabolism. The transcriptional profiling showed several differences between the three lines in the absence of ManNAc analog supplementation that helps explain the different sialoglycan profiles naturally associated with cancer. Only minor changes in mRNA transcript levels occurred upon exposure to the compounds confirming that metabolic flux alone can be a key determinant of sialoglycoconjugate display in breast cancer cells; this result complements the well-established role of genetic control (e.g., the transcription of STs) of sialylation abnormalities ubiquitously associated with cancer. A notable result was that the different cell lines produced significantly different levels of sialic acid upon exogenous ManNAc supplementation, indicating that feedback inhibition of UDP-GlcNAc 2-epimerase/ManNAc kinase (GNE)—generally regarded as the ‘gatekeeper’ enzyme for titering flux into sialic acid biosynthesis—is not the only regulatory mechanism that limits production of this sugar. A notable aspect of our metabolic glycoengineering approach is its ability to discriminate cell subtype based on intracellular metabolism by illuminating otherwise hidden cell type-specific features. We believe that this strategy combined with multi-dimensional analysis of sialic acid metabolism will ultimately provide novel insights into breast cancer subtypes and provide a foundation for new methods of diagnosis.
Oncogene | 2018
Shuang Wei; Jie Wang; Olutobi Oyinlade; Ding Ma; Shuyan Wang; Lisa E. Kratz; Bachchu Lal; Qingfu Xu; Senquan Liu; Sagar R. Shah; Hao Zhang; Yunqing Li; Alfredo Quinones-Hinojosa; Heng Zhu; Zhi-yong Huang; Linzhao Cheng; Jiang Qian; Shuli Xia
Mutations in the isocitrate dehydrogenase 1 (IDH1) gene have been identified in a number of cancer types, including brain cancer. The Cancer Genome Atlas project has revealed that IDH1 mutations occur in 70–80% of grade II and grade III gliomas. Until recently, most of the functional studies of IDH1 mutations in cellular models have been conducted in overexpression systems with the IDH1 wild type background. In this study, we employed a modified CRISPR/Cas9 genome editing technique called “single base editing”, and efficiently introduced heterozygous IDH1 R132H mutation (IDH1R132H/WT) in human astroglial cells. Global DNA methylation profiling revealed hypermethylation as well as hypomethylation induced by IDH1R132H/WT. Global gene expression analysis identified molecular targets and pathways altered by IDH1R132H/WT, including cell proliferation, extracellular matrix (ECM), and cell migration. Our phenotype analysis indicated that compared with IDH1 wild type cells, IDH1R132H/WT promoted cell migration by upregulating integrin β4 (ITGB4); and significantly inhibited cell proliferation. Using our mutated IDH1 models generated by “single base editing”, we identified novel molecular targets of IDH1R132H/WT, namely Yes-associated protein (YAP) and its downstream signaling pathway Notch, to mediate the cell growth-inhibiting effect of IDH1R132H/WT. In summary, the “single base editing” strategy has successfully created heterozygous IDH1 R132H mutation that recapitulates the naturally occurring IDH1 mutation. Our isogenic cellular systems that differ in a single nucleotide in one allele of the IDH1 gene provide a valuable model for novel discoveries of IDH1R132H/WT-driven biological events.
Collaboration
Dive into the Sagar R. Shah's collaboration.
Juan Carlos Martinez-Gutierrez
Johns Hopkins University School of Medicine
View shared research outputs