176 results on '"Vijaya Ramesh"'
Search Results
2. Translatome analysis of tuberous sclerosis complex 1 patient-derived neural progenitor cells reveals rapamycin-dependent and independent alterations
- Author
-
Inci S. Aksoylu, Pauline Martin, Francis Robert, Krzysztof J. Szkop, Nicholas E. Redmond, Srirupa Bhattacharyya, Jennifer Wang, Shan Chen, Roberta L. Beauchamp, Irene Nobeli, Jerry Pelletier, Ola Larsson, and Vijaya Ramesh
- Subjects
Tuberous sclerosis complex ,TSC1 ,Neural progenitor cells ,Translatome ,Polysome profiling ,Autism spectrum disorder ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background Tuberous sclerosis complex (TSC) is an inherited neurocutaneous disorder caused by mutations in the TSC1 or TSC2 genes, with patients often exhibiting neurodevelopmental (ND) manifestations termed TSC-associated neuropsychiatric disorders (TAND) including autism spectrum disorder (ASD) and intellectual disability. Hamartin (TSC1) and tuberin (TSC2) proteins form a complex inhibiting mechanistic target of rapamycin complex 1 (mTORC1) signaling. Loss of TSC1 or TSC2 activates mTORC1 that, among several targets, controls protein synthesis by inhibiting translational repressor eIF4E-binding proteins. Using TSC1 patient-derived neural progenitor cells (NPCs), we recently reported early ND phenotypic changes, including increased cell proliferation and altered neurite outgrowth in TSC1-null NPCs, which were unaffected by the mTORC1 inhibitor rapamycin. Methods Here, we used polysome profiling, which quantifies changes in mRNA abundance and translational efficiencies at a transcriptome-wide level, to compare CRISPR-edited TSC1-null with CRISPR-corrected TSC1-WT NPCs generated from one TSC donor (one clone/genotype). To assess the relevance of identified gene expression alterations, we performed polysome profiling in postmortem brains from ASD donors and age-matched controls. We further compared effects on translation of a subset of transcripts and rescue of early ND phenotypes in NPCs following inhibition of mTORC1 using the allosteric inhibitor rapamycin versus a third-generation bi-steric, mTORC1-selective inhibitor RMC-6272. Results Polysome profiling of NPCs revealed numerous TSC1-associated alterations in mRNA translation that were largely recapitulated in human ASD brains. Moreover, although rapamycin treatment partially reversed the TSC1-associated alterations in mRNA translation, most genes related to neural activity/synaptic regulation or ASD were rapamycin-insensitive. In contrast, treatment with RMC-6272 inhibited rapamycin-insensitive translation and reversed TSC1-associated early ND phenotypes including proliferation and neurite outgrowth that were unaffected by rapamycin. Conclusions Our work reveals ample mRNA translation alterations in TSC1 patient-derived NPCs that recapitulate mRNA translation in ASD brain samples. Further, suppression of TSC1-associated but rapamycin-insensitive translation and ND phenotypes by RMC-6272 unveils potential implications for more efficient targeting of mTORC1 as a superior treatment strategy for TAND.
- Published
- 2023
- Full Text
- View/download PDF
3. Gene replacement therapy in a schwannoma mouse model of neurofibromatosis type 2
- Author
-
Shilpa Prabhakar, Roberta L. Beauchamp, Pike See Cheah, Akiko Yoshinaga, Edwina Abou Haidar, Sevda Lule, Gayathri Mani, Katia Maalouf, Anat Stemmer-Rachamimov, David H. Jung, D. Bradley Welling, Marco Giovannini, Scott R. Plotkin, Casey A. Maguire, Vijaya Ramesh, and Xandra O. Breakefield
- Subjects
neurofibromatosis type 2 ,gene therapy ,adeno-associated viral vector ,schwannoma ,Schwann cells ,Genetics ,QH426-470 ,Cytology ,QH573-671 - Abstract
Loss of function of the neurofibromatosis type 2 (NF2) tumor suppressor gene leads to the formation of schwannomas, meningiomas, and ependymomas, comprising ∼50% of all sporadic cases of primary nervous system tumors. NF2 syndrome is an autosomal dominant condition, with bi-allelic inactivation of germline and somatic alleles resulting in loss of function of the encoded protein merlin and activation of mammalian target of rapamycin (mTOR) pathway signaling in NF2-deficient cells. Here we describe a gene replacement approach through direct intratumoral injection of an adeno-associated virus vector expressing merlin in a novel human schwannoma model in nude mice. In culture, the introduction of an AAV1 vector encoding merlin into CRISPR-modified human NF2-null arachnoidal cells (ACs) or Schwann cells (SCs) was associated with decreased size and mTORC1 pathway activation consistent with restored merlin activity. In vivo, a single injection of AAV1-merlin directly into human NF2-null SC-derived tumors growing in the sciatic nerve of nude mice led to regression of tumors over a 10-week period, associated with a decrease in dividing cells and an increase in apoptosis, in comparison with vehicle. These studies establish that merlin re-expression via gene replacement in NF2-null schwannomas is sufficient to cause tumor regression, thereby potentially providing an effective treatment for NF2.
- Published
- 2022
- Full Text
- View/download PDF
4. High-content image-based analysis and proteomic profiling identifies Tau phosphorylation inhibitors in a human iPSC-derived glutamatergic neuronal model of tauopathy
- Author
-
Chialin Cheng, Surya A. Reis, Emily T. Adams, Daniel M. Fass, Steven P. Angus, Timothy J. Stuhlmiller, Jared Richardson, Hailey Olafson, Eric T. Wang, Debasis Patnaik, Roberta L. Beauchamp, Danielle A. Feldman, M. Catarina Silva, Mriganka Sur, Gary L. Johnson, Vijaya Ramesh, Bruce L. Miller, Sally Temple, Kenneth S. Kosik, Bradford C. Dickerson, and Stephen J. Haggarty
- Subjects
Medicine ,Science - Abstract
Abstract Mutations in MAPT (microtubule-associated protein tau) cause frontotemporal dementia (FTD). MAPT mutations are associated with abnormal tau phosphorylation levels and accumulation of misfolded tau protein that can propagate between neurons ultimately leading to cell death (tauopathy). Recently, a p.A152T tau variant was identified as a risk factor for FTD, Alzheimer's disease, and synucleinopathies. Here we used induced pluripotent stem cells (iPSC) from a patient carrying this p.A152T variant to create a robust, functional cellular assay system for probing pathophysiological tau accumulation and phosphorylation. Using stably transduced iPSC-derived neural progenitor cells engineered to enable inducible expression of the pro-neural transcription factor Neurogenin 2 (Ngn2), we generated disease-relevant, cortical-like glutamatergic neurons in a scalable, high-throughput screening compatible format. Utilizing automated confocal microscopy, and an advanced image-processing pipeline optimized for analysis of morphologically complex human neuronal cultures, we report quantitative, subcellular localization-specific effects of multiple kinase inhibitors on tau, including ones under clinical investigation not previously reported to affect tau phosphorylation. These results demonstrate the potential for using patient iPSC-derived ex vivo models of tauopathy as genetically accurate, disease-relevant systems to probe tau biochemistry and support the discovery of novel therapeutics for tauopathies.
- Published
- 2021
- Full Text
- View/download PDF
5. TSC patient-derived isogenic neural progenitor cells reveal altered early neurodevelopmental phenotypes and rapamycin-induced MNK-eIF4E signaling
- Author
-
Pauline Martin, Vilas Wagh, Surya A. Reis, Serkan Erdin, Roberta L. Beauchamp, Ghalib Shaikh, Michael Talkowski, Elizabeth Thiele, Steven D. Sheridan, Stephen J. Haggarty, and Vijaya Ramesh
- Subjects
Tuberous sclerosis complex ,TSC1 ,mTORC1 ,Induced pluripotent stem cells ,Neural progenitor cells ,Early neurodevelopment ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background Tuberous sclerosis complex (TSC) is a neurodevelopmental disorder with frequent occurrence of epilepsy, autism spectrum disorder (ASD), intellectual disability (ID), and tumors in multiple organs. The aberrant activation of mTORC1 in TSC has led to treatment with mTORC1 inhibitor rapamycin as a lifelong therapy for tumors, but TSC-associated neurocognitive manifestations remain unaffected by rapamycin. Methods Here, we generated patient-specific, induced pluripotent stem cells (iPSCs) from a TSC patient with a heterozygous, germline, nonsense mutation in exon 15 of TSC1 and established an isogenic set of heterozygous (Het), null and corrected wildtype (Corr-WT) iPSCs using CRISPR/Cas9-mediated gene editing. We differentiated these iPSCs into neural progenitor cells (NPCs) and examined neurodevelopmental phenotypes, signaling and changes in gene expression by RNA-seq. Results Differentiated NPCs revealed enlarged cell size in TSC1-Het and Null NPCs, consistent with mTORC1 activation. TSC1-Het and Null NPCs also revealed enhanced proliferation and altered neurite outgrowth in a genotype-dependent manner, which was not reversed by rapamycin. Transcriptome analyses of TSC1-NPCs revealed differentially expressed genes that display a genotype-dependent linear response, i.e., genes upregulated/downregulated in Het were further increased/decreased in Null. In particular, genes linked to ASD, epilepsy, and ID were significantly upregulated or downregulated warranting further investigation. In TSC1-Het and Null NPCs, we also observed basal activation of ERK1/2, which was further activated upon rapamycin treatment. Rapamycin also increased MNK1/2-eIF4E signaling in TSC1-deficient NPCs. Conclusion MEK-ERK and MNK-eIF4E pathways regulate protein translation, and our results suggest that aberrant translation distinct in TSC1/2-deficient NPCs could play a role in neurodevelopmental defects. Our data showing upregulation of these signaling pathways by rapamycin support a strategy to combine a MEK or a MNK inhibitor with rapamycin that may be superior for TSC-associated CNS defects. Importantly, our generation of isogenic sets of NPCs from TSC patients provides a valuable platform for translatome and large-scale drug screening studies. Overall, our studies further support the notion that early developmental events such as NPC proliferation and initial process formation, such as neurite number and length that occur prior to neuronal differentiation, represent primary events in neurogenesis critical to disease pathogenesis of neurodevelopmental disorders such as ASD.
- Published
- 2020
- Full Text
- View/download PDF
6. Brigatinib causes tumor shrinkage in both NF2-deficient meningioma and schwannoma through inhibition of multiple tyrosine kinases but not ALK.
- Author
-
Long-Sheng Chang, Janet L Oblinger, Abbi E Smith, Marc Ferrer, Steven P Angus, Eric Hawley, Alejandra M Petrilli, Roberta L Beauchamp, Lars Björn Riecken, Serkan Erdin, Ming Poi, Jie Huang, Waylan K Bessler, Xiaohu Zhang, Rajarshi Guha, Craig Thomas, Sarah S Burns, Thomas S K Gilbert, Li Jiang, Xiaohong Li, Qingbo Lu, Jin Yuan, Yongzheng He, Shelley A H Dixon, Andrea Masters, David R Jones, Charles W Yates, Stephen J Haggarty, Salvatore La Rosa, D Bradley Welling, Anat O Stemmer-Rachamimov, Scott R Plotkin, James F Gusella, Justin Guinney, Helen Morrison, Vijaya Ramesh, Cristina Fernandez-Valle, Gary L Johnson, Jaishri O Blakeley, D Wade Clapp, and Synodos for NF2 Consortium
- Subjects
Medicine ,Science - Abstract
Neurofibromatosis Type 2 (NF2) is an autosomal dominant genetic syndrome caused by mutations in the NF2 tumor suppressor gene resulting in multiple schwannomas and meningiomas. There are no FDA approved therapies for these tumors and their relentless progression results in high rates of morbidity and mortality. Through a combination of high throughput screens, preclinical in vivo modeling, and evaluation of the kinome en masse, we identified actionable drug targets and efficacious experimental therapeutics for the treatment of NF2 related schwannomas and meningiomas. These efforts identified brigatinib (ALUNBRIG®), an FDA-approved inhibitor of multiple tyrosine kinases including ALK, to be a potent inhibitor of tumor growth in established NF2 deficient xenograft meningiomas and a genetically engineered murine model of spontaneous NF2 schwannomas. Surprisingly, neither meningioma nor schwannoma cells express ALK. Instead, we demonstrate that brigatinib inhibited multiple tyrosine kinases, including EphA2, Fer and focal adhesion kinase 1 (FAK1). These data demonstrate the power of the de novo unbiased approach for drug discovery and represents a major step forward in the advancement of therapeutics for the treatment of NF2 related malignancies.
- Published
- 2021
- Full Text
- View/download PDF
7. Traditional and systems biology based drug discovery for the rare tumor syndrome neurofibromatosis type 2.
- Author
-
Synodos for NF2 Consortium, Robert Allaway, Steve P Angus, Roberta L Beauchamp, Jaishri O Blakeley, Marga Bott, Sarah S Burns, Annemarie Carlstedt, Long-Sheng Chang, Xin Chen, D Wade Clapp, Patrick A Desouza, Serkan Erdin, Cristina Fernandez-Valle, Justin Guinney, James F Gusella, Stephen J Haggarty, Gary L Johnson, Salvatore La Rosa, Helen Morrison, Alejandra M Petrilli, Scott R Plotkin, Abhishek Pratap, Vijaya Ramesh, Noah Sciaky, Anat Stemmer-Rachamimov, Tim J Stuhlmiller, Michael E Talkowski, D Bradley Welling, Charles W Yates, Jon S Zawistowski, and Wen-Ning Zhao
- Subjects
Medicine ,Science - Abstract
Neurofibromatosis 2 (NF2) is a rare tumor suppressor syndrome that manifests with multiple schwannomas and meningiomas. There are no effective drug therapies for these benign tumors and conventional therapies have limited efficacy. Various model systems have been created and several drug targets have been implicated in NF2-driven tumorigenesis based on known effects of the absence of merlin, the product of the NF2 gene. We tested priority compounds based on known biology with traditional dose-concentration studies in meningioma and schwann cell systems. Concurrently, we studied functional kinome and gene expression in these cells pre- and post-treatment to determine merlin deficient molecular phenotypes. Cell viability results showed that three agents (GSK2126458, Panobinostat, CUDC-907) had the greatest activity across schwannoma and meningioma cell systems, but merlin status did not significantly influence response. In vivo, drug effect was tumor specific with meningioma, but not schwannoma, showing response to GSK2126458 and Panobinostat. In culture, changes in both the transcriptome and kinome in response to treatment clustered predominantly based on tumor type. However, there were differences in both gene expression and functional kinome at baseline between meningioma and schwannoma cell systems that may form the basis for future selective therapies. This work has created an openly accessible resource (www.synapse.org/SynodosNF2) of fully characterized isogenic schwannoma and meningioma cell systems as well as a rich data source of kinome and transcriptome data from these assay systems before and after treatment that enables single and combination drug discovery based on molecular phenotype.
- Published
- 2018
- Full Text
- View/download PDF
8. Survival benefit and phenotypic improvement by hamartin gene therapy in a tuberous sclerosis mouse brain model
- Author
-
Shilpa Prabhakar, Xuan Zhang, June Goto, Sangyeul Han, Charles Lai, Roderick Bronson, Miguel Sena-Esteves, Vijaya Ramesh, Anat Stemmer-Rachamimov, David J. Kwiatkowski, and Xandra O. Breakefield
- Subjects
Tuberous sclerosis complex ,TSC ,TSC1 ,TSC2 ,Gene therapy ,AAV ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
We examined the potential benefit of gene therapy in a mouse model of tuberous sclerosis complex (TSC) in which there is embryonic loss of Tsc1 (hamartin) in brain neurons. An adeno-associated virus (AAV) vector (serotype rh8) expressing a tagged form of hamartin was injected into the cerebral ventricles of newborn pups with the genotype Tsc1cc (homozygous for a conditional floxed Tsc1 allele) SynI-cre+, in which Tsc1 is lost selectively in neurons starting at embryonic day 12. Vector-treated Tsc1ccSynIcre+ mice showed a marked improvement in survival from a mean of 22 days in non-injected mice to 52 days in AAV hamartin vector-injected mice, with improved weight gain and motor behavior in the latter. Pathologic studies showed normalization of neuron size and a decrease in markers of mTOR activation in treated as compared to untreated mutant littermates. Hence, we show that gene replacement in the brain is an effective therapeutic approach in this mouse model of TSC1. Our strategy for gene therapy has the advantages that therapy can be achieved from a single application, as compared to repeated treatment with drugs, and that AAV vectors have been found to have minimal to no toxicity in clinical trials for other neurologic conditions. Although there are many additional issues to be addressed, our studies support gene therapy as a useful approach in TSC patients.
- Published
- 2015
- Full Text
- View/download PDF
9. Modeling NF2 with human arachnoidal and meningioma cell culture systems: NF2 silencing reflects the benign character of tumor growth
- Author
-
Marianne F. James, Johanna M. Lelke, Mia MacCollin, Scott R. Plotkin, Anat O. Stemmer-Rachamimov, Vijaya Ramesh, and James F. Gusella
- Subjects
Merlin ,NF2 ,Neurofibromatosis 2 ,Meningioma ,Arachnoidal ,Senescence ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Meningiomas, common tumors arising from arachnoidal cells of the meninges, may occur sporadically, or in association with the inherited disorder, neurofibromatosis 2 (NF2). Most sporadic meningiomas result from NF2 inactivation, resulting in loss of tumor suppressor merlin, implicated in regulating membrane–cytoskeletal organization. To investigate merlin function in an authentic target cell type for NF2 tumor formation, we established primary cultures from genetically-matched meningioma and normal arachnoidal tissues. Our studies revealed novel and distinct cell biological and biochemical properties unique to merlin-deficient meningioma cells compared to merlin-expressing arachnoidal and meningioma cells, and other NF2-deficient cell types. Merlin-deficient meningioma cells displayed cytoskeletal and cell contact defects, altered cell morphology and growth properties, most notably cell senescence, implicating the activation of senescence pathways in limiting benign meningioma growth. Merlin suppression by RNAi in arachnoidal cells replicated merlin-deficient meningioma features, thus establishing these cell systems as disease-relevant models for studying NF2 tumorigenesis.
- Published
- 2008
- Full Text
- View/download PDF
10. Mediator Subunit Med28 Is Essential for Mouse Peri-Implantation Development and Pluripotency.
- Author
-
Lin Li, Ryan M Walsh, Vilas Wagh, Marianne F James, Roberta L Beauchamp, Yuh-Shin Chang, James F Gusella, Konrad Hochedlinger, and Vijaya Ramesh
- Subjects
Medicine ,Science - Abstract
The multi-subunit mammalian Mediator complex acts as an integrator of transcriptional regulation by RNA Polymerase II, and has emerged as a master coordinator of development and cell fate determination. We previously identified the Mediator subunit, MED28, as a cytosolic binding partner of merlin, the Neurofibromatosis 2 (NF2) tumor suppressor, and thus MED28 is distinct in having a cytosolic role as an NF2 interacting protein as well as a nuclear role as a Mediator complex subunit. Although limited in vitro studies have been performed on MED28, its in vivo function remains unknown. Employing a knockout mouse model, we describe for the first time the requirement for Med28 in the developing mouse embryo. Med28-deficiency causes peri-implantation lethality resulting from the loss of pluripotency of the inner cell mass accompanied by reduced expression of key pluripotency transcription factors Oct4 and Nanog. Further, overexpression of Med28 in mouse embryonic fibroblasts enhances the efficiency of their reprogramming to pluripotency. Cre-mediated inactivation of Med28 in induced pluripotent stem cells shows that Med28 is required for their survival. Intriguingly, heterozygous loss of Med28 results in differentiation of induced pluripotent stem cells into extraembryonic trophectoderm and primitive endoderm lineages. Our findings document the essential role of Med28 in the developing embryo as well as in acquisition and maintenance of pluripotency during reprogramming.
- Published
- 2015
- Full Text
- View/download PDF
11. TorsinA protein and neuropathology in early onset generalized dystonia with GAG deletion
- Author
-
Kevin Rostasy, Sarah J Augood, Jeffrey W Hewett, Joanne Chung-on Leung, Hikaru Sasaki, Laurie J Ozelius, Vijaya Ramesh, David G Standaert, Xandra O Breakefield, and John C Hedreen
- Subjects
Torsin ,Dystonia ,Human brain ,Substantia nigra ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Familial, early onset, generalized torsion dystonia is the most common and severe primary dystonia. Most cases are caused by a 3-bp deletion (GAG) in the coding region of the TOR1A (DYT1) gene, which is widely expressed in human brain and encodes the protein torsinA. This study compares neuropathology and torsinA expression in the normal human brain with that in dystonia cases with and without the GAG deletion. TorsinA-like protein was expressed in neuronal cytoplasm throughout the human brain, including cerebellum, substantia nigra, hippocampus, and neostriatum, with higher levels in specific neurons. This immunostaining pattern was not discernibly different in dystonia and normal brains in midbrain and neostriatal regions. However, nigral dopaminergic neurons appeared to be larger in both GAG-deletion and non-GAG-deletion dystonia brains compared to normal, and may be more closely spaced in GAG-deletion brains. Beyond these apparent changes in neuronal size and spacing in dystonia brains, there was no indication of neuron loss, inflammation, DNA strand breaks, or altered distribution of torsin-like immunoreactivity, supporting a functional rather than degenerative etiology of early onset torsion dystonia.
- Published
- 2003
- Full Text
- View/download PDF
12. Comparison of Therapeutic Efficacy of Gene Therapy for Tuberous Sclerosis Type 2 with Standard of Care Everolimus in Preclinical Model (S14.002)
- Author
-
Edwina Abou Haidar, Shilpa Prabhakar, Pike See Cheah, Sevda Lule, Roberta L. Beauchamp, Akiko Yoshinaga, Alexandra L. Geffrey, Anat Stemmer-Rachamimov, Vijaya Ramesh, Casey A. Maguire, and Xandra O. Breakefield
- Published
- 2023
13. Supplementary Figure 1 from Regulation of mTOR Complex 2 Signaling in Neurofibromatosis 2–Deficient Target Cell Types
- Author
-
Vijaya Ramesh, Anat O. Stemmer-Rachamimov, James F. Gusella, Margaret R. Wallace, Hua Li, Sangyeul Han, Roberta Beauchamp, Elizabeth Stivison, and Marianne F. James
- Abstract
PDF file - 105K, Receptor phosphorylation in merlin-deficient Schwann cells in response to serum deprivation, IGF-1 and PDGF stimulation
- Published
- 2023
14. Translatome analysis of Tuberous Sclerosis Complex-1 patient-derived neural progenitor cells reveal rapamycin-dependent and independent alterations
- Author
-
Inci Aksoylu, Pauline Martin, Francis Robert, Krzysztof Szkop, Nicholas Redmond, Shan Chen, Roberta Beauchamp, Irene Nobeli, Jerry Pelletier, Ola Larsson, and Vijaya Ramesh
- Abstract
Tuberous sclerosis complex (TSC) is an inherited neurocutaneous disorder caused by mutations in TSC1 or TSC2 genes, with patients often exhibiting neurodevelopmental (ND) manifestations termed TSC-associated neuropsychiatric disorders (TAND) including autism spectrum disorder (ASD). The hamartin-tuberin (TSC1-TSC2) protein complex inactivates mechanistic target of rapamycin complex 1 (mTORC1) signaling, leading to increased protein synthesis via inactivation of translational repressor eIF4E-binding proteins (4E-BPs). In TSC1-null neural progenitor cells (NPCs), we previously reported early ND phenotypic changes, including increased proliferation/altered neurite outgrowth, which were unaffected by mTORC1-inhibitor rapamycin. Here, using polysome-profiling to quantify translational efficiencies at a transcriptome-wide level, we observed numerous TSC1-dependent alterations in NPCs, largely recapitulated in post-mortem brains from ASD donors. Although rapamycin partially reversed TSC1-associated alterations, most neural activity/synaptic- or ASD-related genes remained insensitive but were inhibited by third-generation bi-steric, mTORC1-selective inhibitor RMC-6272, which also reversed altered ND phenotypes. Together these data reveal potential implications for treatment of TAND.
- Published
- 2023
15. Proteasomal pathway inhibition as a potential therapy for NF2-associated meningioma and schwannoma
- Author
-
Srirupa Bhattacharyya, Janet L Oblinger, Roberta L Beauchamp, Zhenzhen Yin, Serkan Erdin, Priya Koundinya, Anna D Ware, Marc Ferrer, Justin T Jordan, Scott R Plotkin, Lei Xu, Long-Sheng Chang, and Vijaya Ramesh
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
BackgroundNeurofibromatosis 2 (NF2) is an inherited disorder caused by bi-allelic inactivation of the NF2 tumor suppressor gene. NF2-associated tumors, including schwannoma and meningioma, are resistant to chemotherapy, often recurring despite surgery and/or radiation, and have generally shown cytostatic response to signal transduction pathway inhibitors, highlighting the need for improved cytotoxic therapies.MethodsLeveraging data from our previous high-throughput drug screening in NF2 preclinical models, we identified a class of compounds targeting the ubiquitin–proteasome pathway (UPP), and undertook studies using candidate UPP inhibitors, ixazomib/MLN9708, pevonedistat/MLN4924, and TAK-243/MLN7243. Employing human primary and immortalized meningioma (MN) cell lines, CRISPR-modified Schwann cells (SCs), and mouse Nf2 −/− SCs, we performed dose response testing, flow cytometry-based Annexin V and cell cycle analyses, and RNA-sequencing to identify potential underlying mechanisms of apoptosis. In vivo efficacy was also assessed in orthotopic NF2-deficient meningioma and schwannoma tumor models.ResultsTesting of three UPP inhibitors demonstrated potent reduction in cell viability and induction of apoptosis for ixazomib or TAK-243, but not pevonedistat. In vitro analyses revealed that ixazomib or TAK-243 downregulates expression of c-KIT and PDGFRα, as well as the E3 ubiquitin ligase SKP2 while upregulating genes associated with endoplasmic reticulum stress-mediated activation of the unfolded protein response (UPR). In vivo treatment of mouse models revealed delayed tumor growth, suggesting a therapeutic potential.ConclusionsThis study demonstrates the efficacy of proteasomal pathway inhibitors in meningioma and schwannoma preclinical models and lays the groundwork for use of these drugs as a promising novel treatment strategy for NF2 patients.
- Published
- 2023
16. Prospective phase II trial of the dual mTORC1/2 inhibitor vistusertib for progressive or symptomatic meningiomas in persons with neurofibromatosis 2
- Author
-
Justin T Jordan, Christina C Orr, Raquel D Thalheimer, Josephine V Cambillo, Roberta L Beauchamp, Ghalib Shaikh, Alona Muzikansky, Anat Stemmer-Rachamimov, Marco Giovannini, Michel Kalamarides, Fred G Barker, Vijaya Ramesh, and Scott R Plotkin
- Subjects
Oncology ,Surgery ,Neurology (clinical) - Abstract
Background Meningiomas occur in 80% of persons with neurofibromatosis 2 (NF2) and cause significant mortality and morbidity, yet there are no effective medical treatments. NF2-deficient tumors have constitutive activation of mammalian/mechanistic target of rapamycin (mTOR), and treatment with mTORC1 inhibitors results in growth arrest in a minority of tumors, with paradoxical activation of the mTORC2/AKT pathway. We studied the effect of vistusertib, a dual mTORC1/mTORC2 inhibitor, in NF2 patients with progressive or symptomatic meningiomas. Methods Vistusertib was administered orally at 125 mg twice daily for 2 consecutive days each week. The primary endpoint was the imaging response in the target meningioma, defined as a volume decrease of 20% compared with the baseline. Secondary endpoints included toxicity, imaging response of nontarget tumors, quality of life, and genetic biomarkers. Results Eighteen participants (13 female), median age of 41 (range, 18–61) years, were enrolled. In target meningiomas, the best response was partial response (PR) in 1/18 tumors (6%) and stable disease (SD) in 17/18 tumors (94%). For all measured intracranial meningiomas and vestibular schwannomas, the best imaging response was PR in 6/59 tumors (10%) and SD in 53 (90%). Treatment-related grade 3/4 adverse events occurred in 14 (78%) participants, and 9 participants discontinued treatment due to side effects. Conclusions Although the study did not meet the primary endpoint, vistusertib treatment was associated with high rates of SD in progressive NF2-related tumors. However, this dosing regimen for vistusertib was poorly tolerated. Future studies of dual mTORC inhibitors for NF2 should focus on optimizing tolerability and evaluating the relevance of tumor stability in participants.
- Published
- 2023
17. Rapid Multiplex Loop-Mediated Isothermal Amplification (m-LAMP) Assay for Differential Diagnosis of Leprosy and Post–Kala-Azar Dermal Leishmaniasis
- Author
-
Ruchi Singh, Shweta Joshi, Keerti Kaumudee Dixit, Poonam Salotra, Vijaya Ramesh, and Vanila Sharma
- Subjects
Adult ,Male ,Adolescent ,Leishmania donovani ,Loop-mediated isothermal amplification ,Leishmaniasis, Cutaneous ,Antigens, Protozoan ,Diagnosis, Differential ,Young Adult ,Leprosy ,Virology ,parasitic diseases ,medicine ,Humans ,Multiplex ,Mycobacterium leprae ,Aged ,Post-kala-azar dermal leishmaniasis ,biology ,business.industry ,Leishmaniasis ,Articles ,Middle Aged ,medicine.disease ,biology.organism_classification ,Infectious Diseases ,Molecular Diagnostic Techniques ,Female ,Parasitology ,Differential diagnosis ,business ,Multiplex Polymerase Chain Reaction ,Nucleic Acid Amplification Techniques - Abstract
Leprosy and post–kala-azar dermal leishmaniasis (PKDL) are co-endemic neglected tropical diseases often misdiagnosed because of close resemblance in their clinical manifestations. The test that aids in differential diagnosis of leprosy and PKDL would be useful in endemic areas. Here, we report development of a multiplex loop-mediated isothermal amplification (m-LAMP) assay for differential detection of Mycobacterium leprae and Leishmania donovani using a real-time fluorometer. The m-LAMP assay was rapid with a mean amplification time of 15 minutes, and analytical sensitivity of 1 fg for L. donovani and 100 fg for M. leprae. The distinct mean Tm values for M. leprae and L. donovani allowed differentiation of the two organisms in the m-LAMP assay. Diagnostic sensitivity of the assay was evaluated by using confirmed cases of leprosy (n = 40) and PKDL (n = 40) (tissue and slit aspirate samples). All the leprosy and PKDL samples used in this study were positive by organism-specific QPCR and loop-mediated isothermal amplification assays. The diagnostic sensitivity of the m-LAMP assay was 100% (95% CI: 91.2–100.0%) for detecting PKDL and 95% for leprosy (95% CI: 83.1–99.4%). Our m-LAMP assay was successfully used to detect both M. leprae and L. donovani in a patient coinfected with leprosy and macular PKDL. The m-LAMP assay is rapid, accurate, and applicable for differential diagnosis of leprosy versus PKDL, especially in endemic areas.
- Published
- 2021
18. TSC patient-derived isogenic neural progenitor cells reveal altered early neurodevelopmental phenotypes and rapamycin-induced MNK-eIF4E signaling
- Author
-
Vilas Wagh, Pauline Martin, Ghalib Shaikh, Roberta L. Beauchamp, Steven D. Sheridan, Michael E. Talkowski, Stephen J. Haggarty, Elizabeth A. Thiele, Surya A. Reis, Vijaya Ramesh, and Serkan Erdin
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,Neurite ,mTORC1 ,Biology ,Early neurodevelopment ,lcsh:RC346-429 ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Neurodevelopmental disorder ,Developmental Neuroscience ,medicine ,Neural progenitor cells ,Induced pluripotent stem cell ,Molecular Biology ,lcsh:Neurology. Diseases of the nervous system ,030304 developmental biology ,0303 health sciences ,Neurogenesis ,medicine.disease ,Neural stem cell ,Cell biology ,TSC1 ,Psychiatry and Mental health ,Induced pluripotent stem cells ,medicine.anatomical_structure ,Tuberous sclerosis complex ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Background Tuberous sclerosis complex (TSC) is a neurodevelopmental disorder with frequent occurrence of epilepsy, autism spectrum disorder (ASD), intellectual disability (ID), and tumors in multiple organs. The aberrant activation of mTORC1 in TSC has led to treatment with mTORC1 inhibitor rapamycin as a lifelong therapy for tumors, but TSC-associated neurocognitive manifestations remain unaffected by rapamycin. Methods Here, we generated patient-specific, induced pluripotent stem cells (iPSCs) from a TSC patient with a heterozygous, germline, nonsense mutation in exon 15 of TSC1 and established an isogenic set of heterozygous (Het), null and corrected wildtype (Corr-WT) iPSCs using CRISPR/Cas9-mediated gene editing. We differentiated these iPSCs into neural progenitor cells (NPCs) and examined neurodevelopmental phenotypes, signaling and changes in gene expression by RNA-seq. Results Differentiated NPCs revealed enlarged cell size in TSC1-Het and Null NPCs, consistent with mTORC1 activation. TSC1-Het and Null NPCs also revealed enhanced proliferation and altered neurite outgrowth in a genotype-dependent manner, which was not reversed by rapamycin. Transcriptome analyses of TSC1-NPCs revealed differentially expressed genes that display a genotype-dependent linear response, i.e., genes upregulated/downregulated in Het were further increased/decreased in Null. In particular, genes linked to ASD, epilepsy, and ID were significantly upregulated or downregulated warranting further investigation. In TSC1-Het and Null NPCs, we also observed basal activation of ERK1/2, which was further activated upon rapamycin treatment. Rapamycin also increased MNK1/2-eIF4E signaling in TSC1-deficient NPCs. Conclusion MEK-ERK and MNK-eIF4E pathways regulate protein translation, and our results suggest that aberrant translation distinct in TSC1/2-deficient NPCs could play a role in neurodevelopmental defects. Our data showing upregulation of these signaling pathways by rapamycin support a strategy to combine a MEK or a MNK inhibitor with rapamycin that may be superior for TSC-associated CNS defects. Importantly, our generation of isogenic sets of NPCs from TSC patients provides a valuable platform for translatome and large-scale drug screening studies. Overall, our studies further support the notion that early developmental events such as NPC proliferation and initial process formation, such as neurite number and length that occur prior to neuronal differentiation, represent primary events in neurogenesis critical to disease pathogenesis of neurodevelopmental disorders such as ASD.
- Published
- 2020
19. Utility of Blood as the Clinical Specimen for the Molecular Diagnosis of Post-Kala-Azar Dermal Leishmaniasis
- Author
-
Poonam Salotra, Abhishek Kumar Singh, Ruchi Singh, Vijaya Ramesh, Shyam Sundar, Keerti Kaumudee Dixit, S. N. Upadhyay, and Om Prakash Singh
- Subjects
Microbiology (medical) ,medicine.medical_specialty ,India ,Leishmaniasis, Cutaneous ,Real-Time Polymerase Chain Reaction ,Parasite load ,parasitic diseases ,medicine ,Humans ,Inverse correlation ,Post-kala-azar dermal leishmaniasis ,medicine.diagnostic_test ,business.industry ,Leishmaniasis ,medicine.disease ,Dermatology ,Highly sensitive ,Indian subcontinent ,Visceral leishmaniasis ,Molecular Diagnostic Techniques ,Skin biopsy ,Leishmaniasis, Visceral ,Parasitology ,business ,Nucleic Acid Amplification Techniques ,Leishmania donovani - Abstract
The countries in the Indian subcontinent have reported a dramatic decline in visceral leishmaniasis (VL) cases. However, the presence of the parasite reservoir in the form of post-kala-azar dermal leishmaniasis (PKDL), a dermal sequel of VL, is a hurdle in attaining VL elimination. Presently employed clinical specimens for the diagnosis of PKDL include skin biopsy specimens and slit skin smears. In this study, the use of blood as a clinical specimen was investigated in different manifestations of PKDL in India. This is a bicentric study (National Institute of Pathology, Indian Council of Medical Research [ICMR], New Delhi, and Institute of Medical Sciences [IMS], Banaras Hindu University, Varanasi), with 215 participants (120 PKDL patients and 95 controls). Highly sensitive quantitative real-time PCR (Q-PCR) and field-deployable loop-mediated isothermal amplification (LAMP) were employed using blood samples for diagnosis. Promising sensitivities of 77.50% (95% confidence interval [CI], 69.24 to 84.05%) for Q-PCR and 70.83% (95% CI, 62.16 to 78.22%) for LAMP were obtained for the diagnosis of PKDL. Further, enhanced sensitivities of 83.33% (95% CI, 71.28 to 90.98%) and 77.78% (95% CI, 65.06 to 86.80%) for Q-PCR and LAMP, respectively, were recorded for the detection of macular cases. The study revealed an inverse correlation between the parasite load estimated in slit and blood samples, thereby favoring the use of blood for the diagnosis of the macular variant, which may be missed due to scant parasite loads in the slit. This study is the first to propose the promising potential of blood as a clinical specimen for accurate diagnosis of PKDL, which would aid in fast-tracking VL elimination.
- Published
- 2021
20. High-content image-based analysis and proteomic profiling identifies Tau phosphorylation inhibitors in a human iPSC-derived glutamatergic neuronal model of tauopathy
- Author
-
Daniel M. Fass, Roberta L. Beauchamp, Steven P. Angus, Chialin Cheng, Kenneth S. Kosik, Timothy J. Stuhlmiller, Bradford C. Dickerson, Bruce L. Miller, Debasis Patnaik, Danielle A. Feldman, Jared I Richardson, Stephen J. Haggarty, Hailey Olafson, Surya A. Reis, Mriganka Sur, Gary L. Johnson, Vijaya Ramesh, Eric T. Wang, Sally Temple, Emily T Adams, and M. Catarina Silva
- Subjects
Proteomics ,Aging ,Pyridines ,Image Processing ,Stem cells ,Neurodegenerative ,Alzheimer's Disease ,Computer-Assisted ,Glutamates ,Models ,Stem Cell Research - Nonembryonic - Human ,Basic Helix-Loop-Helix Transcription Factors ,Image Processing, Computer-Assisted ,2.1 Biological and endogenous factors ,Neurogenin-2 ,Aetiology ,Phosphorylation ,Induced pluripotent stem cell ,Alzheimer's Disease Related Dementias (ADRD) ,Neurons ,Multidisciplinary ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Molecular medicine ,Drug discovery ,Chemical biology ,Neural stem cell ,Cell biology ,Frontotemporal Dementia (FTD) ,Tauopathies ,5.1 Pharmaceuticals ,Neurological ,Medicine ,Tauopathy ,Development of treatments and therapeutic interventions ,Frontotemporal dementia ,Science ,Tau protein ,Induced Pluripotent Stem Cells ,Nerve Tissue Proteins ,tau Proteins ,Biology ,Models, Biological ,Article ,Cell Line ,Small Molecule Libraries ,Medical research ,mental disorders ,medicine ,Acquired Cognitive Impairment ,Humans ,Synucleinopathies ,Stem Cell Research - Induced Pluripotent Stem Cell ,Neurosciences ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,medicine.disease ,Biological ,Stem Cell Research ,Brain Disorders ,Pyrimidines ,biology.protein ,Dementia ,Protein Kinases ,Biomarkers ,Neuroscience - Abstract
Mutations in MAPT (microtubule-associated protein tau) cause frontotemporal dementia (FTD). MAPT mutations are associated with abnormal tau phosphorylation levels and accumulation of misfolded tau protein that can propagate between neurons ultimately leading to cell death (tauopathy). Recently, a p.A152T tau variant was identified as a risk factor for FTD, Alzheimer's disease, and synucleinopathies. Here we used induced pluripotent stem cells (iPSC) from a patient carrying this p.A152T variant to create a robust, functional cellular assay system for probing pathophysiological tau accumulation and phosphorylation. Using stably transduced iPSC-derived neural progenitor cells engineered to enable inducible expression of the pro-neural transcription factor Neurogenin 2 (Ngn2), we generated disease-relevant, cortical-like glutamatergic neurons in a scalable, high-throughput screening compatible format. Utilizing automated confocal microscopy, and an advanced image-processing pipeline optimized for analysis of morphologically complex human neuronal cultures, we report quantitative, subcellular localization-specific effects of multiple kinase inhibitors on tau, including ones under clinical investigation not previously reported to affect tau phosphorylation. These results demonstrate the potential for using patient iPSC-derived ex vivo models of tauopathy as genetically accurate, disease-relevant systems to probe tau biochemistry and support the discovery of novel therapeutics for tauopathies.
- Published
- 2021
21. Brigatinib causes tumor shrinkage in both NF2-deficient meningioma and schwannoma through inhibition of multiple tyrosine kinases but not ALK
- Author
-
Salvatore La Rosa, Helen Morrison, Jie Huang, Rajarshi Guha, Li Jiang, Yongzheng He, Gary L. Johnson, Steven P. Angus, Eric T. Hawley, Vijaya Ramesh, Justin Guinney, Sarah S. Burns, Jin Yuan, Roberta L. Beauchamp, Lars Björn Riecken, Cristina Fernandez-Valle, Marc Ferrer, Charles W. Yates, Serkan Erdin, Abbi Smith, Waylan K. Bessler, D. Bradley Welling, D. Wade Clapp, Shelley Dixon, Scott R. Plotkin, Ming Poi, Thomas S. K. Gilbert, Anat Stemmer-Rachamimov, Alejandra M. Petrilli, Craig J. Thomas, Xiaohong Li, Qingbo Lu, David R. Jones, James F. Gusella, Long-Sheng Chang, Andrea R. Masters, Xiaohu Zhang, Janet L. Oblinger, Jaishri O. Blakeley, and Stephen J. Haggarty
- Subjects
0301 basic medicine ,Macroglial Cells ,Cancer Treatment ,Schwannoma ,Biochemistry ,Tyrosine Kinases ,0302 clinical medicine ,Medical Conditions ,Animal Cells ,Medicine and Health Sciences ,Meningeal Neoplasms ,Kinome ,Neurofibromatosis type 2 ,Neurological Tumors ,Neurofibromin 2 ,Multidisciplinary ,Pharmaceutics ,Animal Models ,Protein-Tyrosine Kinases ,EPH receptor A2 ,Enzymes ,Oncology ,Neurology ,Experimental Organism Systems ,Genetic Diseases ,030220 oncology & carcinogenesis ,Medicine ,Cellular Types ,Meningioma ,Tyrosine kinase ,Neurilemmoma ,Research Article ,Brigatinib ,Tumor suppressor gene ,Science ,Mouse Models ,Glial Cells ,Research and Analysis Methods ,03 medical and health sciences ,Model Organisms ,Organophosphorus Compounds ,Drug Therapy ,medicine ,otorhinolaryngologic diseases ,Humans ,neoplasms ,Cell Proliferation ,Clinical Genetics ,business.industry ,Autosomal Dominant Diseases ,Cancers and Neoplasms ,Biology and Life Sciences ,Proteins ,Cell Biology ,medicine.disease ,nervous system diseases ,030104 developmental biology ,Pyrimidines ,Neurofibromatosis Type 2 ,Mutation ,Cancer research ,Animal Studies ,Enzymology ,Schwann Cells ,business ,Protein Kinases - Abstract
Neurofibromatosis Type 2 (NF2) is an autosomal dominant genetic syndrome caused by mutations in the NF2 tumor suppressor gene resulting in multiple schwannomas and meningiomas. There are no FDA approved therapies for these tumors and their relentless progression results in high rates of morbidity and mortality. Through a combination of high throughput screens, preclinical in vivo modeling, and evaluation of the kinome en masse, we identified actionable drug targets and efficacious experimental therapeutics for the treatment of NF2 related schwannomas and meningiomas. These efforts identified brigatinib (ALUNBRIG®), an FDA-approved inhibitor of multiple tyrosine kinases including ALK, to be a potent inhibitor of tumor growth in established NF2 deficient xenograft meningiomas and a genetically engineered murine model of spontaneous NF2 schwannomas. Surprisingly, neither meningioma nor schwannoma cells express ALK. Instead, we demonstrate that brigatinib inhibited multiple tyrosine kinases, including EphA2, Fer and focal adhesion kinase 1 (FAK1). These data demonstrate the power of the de novo unbiased approach for drug discovery and represents a major step forward in the advancement of therapeutics for the treatment of NF2 related malignancies.
- Published
- 2021
22. SMARCE1 deficiency generates a targetable mSWI/SNF dependency in clear cell meningioma
- Author
-
Roodolph St. Pierre, Clayton K. Collings, Daniel D. Samé Guerra, Christian J. Widmer, Olubusayo Bolonduro, Nazar Mashtalir, Akshay Sankar, Yu Liang, Wenya Linda Bi, Erica H. Gerkes, Vijaya Ramesh, Jun Qi, Miriam J. Smith, David M. Meredith, and Cigall Kadoch
- Subjects
Mammals ,EXPRESSION ,AKT1 ,COMPLEX ,MUTATIONS ,Chromatin Assembly and Disassembly ,GENE ,CANCER ,ARID1A ,Chromatin ,TRANSCRIPTION FACTORS ,FUSION ,GERMLINE ,Meningeal Neoplasms ,Genetics ,Animals ,Meningioma - Abstract
SMARCE1 loss destabilizes the canonical BAF complex and increases the formation of BRD9-containing non-canonical (ncBAF) complexes. SMARCE1-deficient cells, which are a model for clear cell meningioma, are sensitive to ncBAF complex inhibition.Mammalian SWI/SNF (mSWI/SNF) ATP-dependent chromatin remodeling complexes establish and maintain chromatin accessibility and gene expression, and are frequently perturbed in cancer. Clear cell meningioma (CCM), an aggressive tumor of the central nervous system, is uniformly driven by loss of SMARCE1, an integral subunit of the mSWI/SNF core. Here, we identify a structural role for SMARCE1 in selectively stabilizing the canonical BAF (cBAF) complex core-ATPase module interaction. In CCM, cBAF complexes fail to stabilize on chromatin, reducing enhancer accessibility, and residual core module components increase the formation of BRD9-containing non-canonical BAF (ncBAF) complexes. Combined attenuation of cBAF function and increased ncBAF complex activity generates the CCM-specific gene expression signature, which is distinct from that of NF2-mutated meningiomas. Importantly, SMARCE1-deficient cells exhibit heightened sensitivity to small-molecule inhibition of ncBAF complexes. These data inform the function of a previously elusive SWI/SNF subunit and suggest potential therapeutic approaches for intractable SMARCE1-deficient CCM tumors.
- Published
- 2022
23. Gene therapy for tuberous sclerosis complex type 2 in a mouse model by delivery of AAV9 encoding a condensed form of tuberin
- Author
-
Xandra O. Breakefield, Shingo Kasamatsu, King Hwa Ling, Vijaya Ramesh, Pike See Cheah, Casey A. Maguire, Bakhos A. Tannous, Masao Kaneki, Roberta L. Beauchamp, Roderick T. Bronson, Xuan Zhang, David J. Kwiatkowski, Shilpa Prabhakar, Anat Stemmer-Rachamimov, Bence György, Elizabeth A. Thiele, and David Yellen
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,Tumor suppressor gene ,Genetic enhancement ,Diseases and Disorders ,Biology ,Virus ,03 medical and health sciences ,Tuberous sclerosis ,0302 clinical medicine ,medicine ,Recombinase ,Research Articles ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,Cell growth ,fungi ,food and beverages ,SciAdv r-articles ,Human Genetics ,medicine.disease ,nervous system diseases ,medicine.anatomical_structure ,Cancer research ,TSC1 ,TSC2 ,030217 neurology & neurosurgery ,Research Article - Abstract
Gene therapy for tuberous sclerosis type 2 proved beneficial in a mouse model of the disease, extending life span., Tuberous sclerosis complex (TSC) results from loss of a tumor suppressor gene - TSC1 or TSC2, encoding hamartin and tuberin, respectively. These proteins formed a complex to inhibit mTORC1-mediated cell growth and proliferation. Loss of either protein leads to overgrowth lesions in many vital organs. Gene therapy was evaluated in a mouse model of TSC2 using an adeno-associated virus (AAV) vector carrying the complementary for a “condensed” form of human tuberin (cTuberin). Functionality of cTuberin was verified in culture. A mouse model of TSC2 was generated by AAV-Cre recombinase disruption of Tsc2-floxed alleles at birth, leading to a shortened lifespan (mean 58 days) and brain pathology consistent with TSC. When these mice were injected intravenously on day 21 with AAV9-cTuberin, the mean survival was extended to 462 days with reduction in brain pathology. This demonstrates the potential of treating life-threatening TSC2 lesions with a single intravenous injection of AAV9-cTuberin.
- Published
- 2021
24. mTOR kinase inhibition disrupts neuregulin 1-ERBB3 autocrine signaling and sensitizes NF2-deficient meningioma cellular models to IGF1R inhibition
- Author
-
Roberta L. Beauchamp, Vijaya Ramesh, Justin T. Jordan, Serkan Erdin, Scott R. Plotkin, James F. Gusella, and Luke Witt
- Subjects
0301 basic medicine ,Receptor, ErbB-3 ,insulin-like growth factor (IGF) receptor 1 ,DRC, dose–response curve ,mTORC1 ,SGK1, serum-/glucocorticoid-responsive kinase-1 ,Biochemistry ,meningioma ,Receptor, IGF Type 1 ,mTORC1/mTORC2, mTOR complex 1/mTOR complex 2 ,Cell Movement ,PI3K, Phosphoinositide 3-kinase ,Meningeal Neoplasms ,Akt PKB ,FOXO, Forkhead box protein O ,ERBB3 ,tumor suppressor gene ,CM, concentrated medium ,Benzoxazoles ,Neurofibromin 2 ,biology ,Chemistry ,Triazines ,Receptor, EphA2 ,TOR Serine-Threonine Kinases ,mammalian target of rapamycin (mTOR) ,APJ, apelin receptor ,Autocrine Communication ,MR, maximum response ,NF2, Neurofibromatosis 2 ,ERBB, V-ERB-B avian erythroblastic leukemia viral oncogene homolog ,Benzamides ,IRS, insulin receptor substrate ,PDK1, phosphoinositide-dependent kinase-1 ,RTK, receptor tyrosine kinase ,signaling ,brain tumor ,Research Article ,IC50, inhibitory concentration 50% ,Signal Transduction ,NRG1, neuregulin-1/heregulin ,Morpholines ,Neuregulin-1 ,mTOR, mechanistic/mammalian target of rapamycin ,Antibodies, Monoclonal, Humanized ,WHO, World Health Organization ,03 medical and health sciences ,Growth factor receptor ,TGFA, transforming growth factor-alpha ,Cell Line, Tumor ,otorhinolaryngologic diseases ,Humans ,Autocrine signalling ,Molecular Biology ,Mechanistic target of rapamycin ,PI3K/AKT/mTOR pathway ,dual mTORC1/mTORC2 inhibition ,Insulin-like growth factor 1 receptor ,Cell Proliferation ,Sirolimus ,Phosphoinositide 3-kinase ,030102 biochemistry & molecular biology ,qPCR, quantitative RT-PCR ,Dose-Response Relationship, Drug ,EPH receptor, erythropoietin-producing hepatocellular receptor ,Lapatinib ,Cell Biology ,EGFR, epidermal growth factor receptor ,AC, arachnoid cell ,030104 developmental biology ,Pyrimidines ,Gene Expression Regulation ,NF2 ,Cancer research ,biology.protein ,Pyrazoles ,NRG1-ERBB3 ,MN, meningioma ,Transcriptome ,IGF1R/IR, insulin-like growth factor receptor 1/insulin receptor ,Proto-Oncogene Proteins c-akt - Abstract
Meningiomas (MNs), arising from the arachnoid/meningeal layer, are nonresponsive to chemotherapies, with ∼50% showing loss of the Neurofibromatosis 2 (NF2) tumor suppressor gene. Previously, we established NF2 loss activates mechanistic target of rapamycin complex 1 (mTORC1) and mechanistic target of rapamycin complex 2 (mTORC2) signaling, leading to clinical trials for NF2 and MN. Recently our omics studies identified activated ephrin (EPH) receptor and Src family kinases upon NF2 loss. Here, we report increased expression of several ligands in NF2-null human arachnoidal cells (ACs) and the MN cell line Ben-Men-1, particularly neuregulin-1/heregulin (NRG1), and confirm increased NRG1 secretion and activation of V-ERB-B avian erythroblastic leukemia viral oncogene homolog 3 (ERBB3) receptor kinase. Conditioned-medium from NF2-null ACs or exogenous NRG1 stimulated ERBB3, EPHA2, and mTORC1/2 signaling, suggesting pathway crosstalk. NF2-null cells treated with an ERBB3-neutralizing antibody partially downregulated mTOR pathway activation but showed no effect on viability. mTORC1/2 inhibitor treatment decreased NRG1 expression and downregulated ERBB3 while re-activating pAkt T308, suggesting a mechanism independent of NRG1–ERBB3 but likely involving activation of another upstream receptor kinase. Transcriptomics after mTORC1/2 inhibition confirmed decreased ERBB3/ERBB4 while revealing increased expression of insulin-like growth factor receptor 1 (IGF1R). Drug treatment co-targeting mTORC1/2 and IGF1R/insulin receptor attenuated pAkt T308 and showed synergistic effects on viability. Our findings indicate potential autocrine signaling where NF2 loss leads to secretion/activation of NRG1-ERBB3 signaling. mTORC1/2 inhibition downregulates NRG1-ERBB3, while upregulating pAkt T308 through an adaptive response involving IGF1R/insulin receptor and co-targeting these pathways may prove effective for treatment of NF2-deficient MN.
- Published
- 2020
25. Elevated IL-6R on CD4+ T cells promotes IL-6 driven Th17 cell responses in patients with T1R leprosy reactions
- Author
-
Alpana Sharma, Santosh Kurra, Neena Khanna, Vijaya Ramesh, Mohd Tarique, Chaman Saini, and Rupesh K. Srivastava
- Subjects
Adult ,CD4-Positive T-Lymphocytes ,Male ,0301 basic medicine ,Adolescent ,Cellular differentiation ,Immunology ,030231 tropical medicine ,Cell ,lcsh:Medicine ,Diseases ,Microbiology ,T-Lymphocytes, Regulatory ,Article ,Pathogenesis ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Transforming Growth Factor beta ,Leprosy ,medicine ,Humans ,Secretion ,lcsh:Science ,Receptor ,Interleukin 6 ,Mycobacterium leprae ,Multidisciplinary ,biology ,Interleukin-6 ,Chemistry ,lcsh:R ,Interleukin-17 ,biology.organism_classification ,Receptors, Interleukin-6 ,030104 developmental biology ,medicine.anatomical_structure ,biology.protein ,Th17 Cells ,lcsh:Q ,Female ,Signal transduction ,Signal Transduction - Abstract
Th17 cells play vital role during pathogenesis of leprosy reactions. Previously, we have reported that IL-23 is involved in Th17 cells differentiation. Subsequently, our group also showed that IL-6 induces Th17 cell differentiation along with TGF-β in leprosy reactions. Here, we next asked the question that whether IL-6 or IL-23 induced Th17 cells are different in nature? In this study, Type 1 Reactions (T1R) showed significantly (p +IL6R+ T cells as compared to non-reaction (NR) patients. Furthermore, recombinant IL-6, IL-23 and TGF-β promoted IL-17A secretion by CD4+IL6R+ T cells. Subsequently, IL-6R and IL-23R blocking experiments showed significantly (p Mycobacterium leprae and its reactions.
- Published
- 2020
26. γδ T cells are associated with inflammation and immunopathogenesis of leprosy reactions
- Author
-
Chaman Saini, Alpana Sharma, Mohd Tarique, Vijaya Ramesh, and Neena Khanna
- Subjects
STAT3 Transcription Factor ,0301 basic medicine ,medicine.medical_treatment ,CD3 ,Immunology ,Gene Expression ,Inflammation ,T-Lymphocytes, Regulatory ,Peripheral blood mononuclear cell ,Immunophenotyping ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,T-Lymphocyte Subsets ,Leprosy ,STAT5 Transcription Factor ,medicine ,Humans ,Immunology and Allergy ,Phosphorylation ,biology ,business.industry ,FOXP3 ,Receptors, Antigen, T-Cell, gamma-delta ,medicine.disease ,030104 developmental biology ,Cytokine ,Antigens, Surface ,biology.protein ,Cytokines ,Interleukin 17 ,medicine.symptom ,business ,Biomarkers ,030215 immunology - Abstract
Background Leprosy reactions appear episodically in leprosy patients, which lead to high inflammation, morbidity and peripheral nerve damage. The role of Th17 cell has been well studied in leprosy reactions but the role of γδ or unconventional T cells which is an other major source of IL-17 in many diseases, not studied in leprosy reactional episodes. Objective The aim of the present study to elucidate the role of γδ T cells in leprosy reactions. Methodology A total of 40 untreated non-reaction and reactions patients were recruited. PBMCs were isolated and stimulated with M. leprae sonicated antigen (MLSA) for 48 h and immuno-phenotyping was done using flow cytometry. Moreover, γδ T cells were isolated by Magnetic beads technology and mRNA expression of IL-17, IFN-γ, TGF-β and FOXP3 were analyzed by real-time PCR (qPCR) and cytokine was estimated in the culture supernatant by ELISA. Results γδ T cells were significantly increased in both Reversal reaction (RR) and Erythema nodosum leprosum (ENL) reaction patients. These cells produced significant amount of IL-17 and IFN-γ. Furthermore, CD3+TCRγδ+ T cells expressed transient FOXP3 with a low amount of TGF-β in both reactions as compared to stable patients. Moreover, low TGF-β producing TCR-γδ cells were associated with low phosphorylation of STAT5A. Conclusion This study will add to our understanding of the immunological features that mediate and regulate the pathogenesis of leprosy and may helpful to reduce the immuno-pathogenesis of leprosy reaction by targeting these cells.
- Published
- 2018
27. EPH receptor signaling as a novel therapeutic target in NF2-deficient meningioma
- Author
-
Patrick A. DeSouza, Timothy J. Stuhlmiller, Thomas S. K. Gilbert, Steven P. Angus, Xin Chen, James F. Gusella, Gary L. Johnson, Roberta L. Beauchamp, Vijaya Ramesh, Janet L. Oblinger, Anat Stemmer-Rachamimov, Scott R. Plotkin, Luke Witt, Stephen J. Haggarty, Justin T. Jordan, and Long-Sheng Chang
- Subjects
0301 basic medicine ,Cancer Research ,Tumor suppressor gene ,Antineoplastic Agents ,Apoptosis ,Mice, SCID ,mTORC1 ,Receptor tyrosine kinase ,Mice ,03 medical and health sciences ,Mice, Inbred NOD ,Biomarkers, Tumor ,Meningeal Neoplasms ,Tumor Cells, Cultured ,otorhinolaryngologic diseases ,Animals ,Humans ,Medicine ,Kinome ,Cell Proliferation ,Receptors, Eph Family ,Neurofibromin 2 ,biology ,business.industry ,Erythropoietin-producing hepatocellular (Eph) receptor ,Xenograft Model Antitumor Assays ,Gene Expression Regulation, Neoplastic ,Dasatinib ,030104 developmental biology ,Oncology ,Tumor progression ,Basic and Translational Investigations ,biology.protein ,Cancer research ,Neurology (clinical) ,Meningioma ,business ,Proto-oncogene tyrosine-protein kinase Src ,medicine.drug - Abstract
Background Meningiomas are the most common primary brain tumor in adults, and somatic loss of the neurofibromatosis 2 (NF2) tumor suppressor gene is a frequent genetic event. There is no effective treatment for tumors that recur or continue to grow despite surgery and/or radiation. Therefore, targeted therapies that either delay tumor progression or cause tumor shrinkage are much needed. Our earlier work established mammalian target of rapamycin complex mTORC1/mTORC2 activation in NF2-deficient meningiomas. Methods High-throughput kinome analyses were performed in NF2-null human arachnoidal and meningioma cell lines to identify functional kinome changes upon NF2 loss. Immunoblotting confirmed the activation of kinases and demonstrated effectiveness of drugs to block the activation. Drugs, singly and in combination, were screened in cells for their growth inhibitory activity. Antitumor drug efficacy was tested in an orthotopic meningioma model. Results Erythropoietin-producing hepatocellular receptor tyrosine kinases (EPH RTKs), c-KIT, and Src family kinase (SFK) members, which are biological targets of dasatinib, were among the top candidates activated in NF2-null cells. Dasatinib significantly inhibited phospho-EPH receptor A2 (pEPHA2), pEPHB1, c-KIT, and Src/SFK in NF2-null cells, showing no cross-talk with mTORC1/2 signaling. Posttreatment kinome analyses showed minimal adaptive changes. While dasatinib treatment showed some activity, dual mTORC1/2 inhibitor and its combination with dasatinib elicited stronger growth inhibition in meningiomas. Conclusion Co-targeting mTORC1/2 and EPH RTK/SFK pathways could be a novel effective treatment strategy for NF2-deficient meningiomas.
- Published
- 2018
28. CTNI-54. A SINGLE ARM PHASE II STUDY OF THE DUAL MTORC1/MTORC2 INHIBITOR VISTUSERTIB PROVIDED FOR SPORADIC PATIENTS WITH GRADE II-III MENINGIOMAS THAT RECUR OR PROGRESS AFTER SURGERY AND RADIATION
- Author
-
Patrick Y. Wen, Fred G. Barker, Vijaya Ramesh, Anat Stemmer-Rachamimov, Alona Muzikansky, Miriam J. Smith, Justin T. Jordan, Roberta L. Beauchamp, Priya Kumthekar, Elizabeth R. Gerstner, and Scott R. Plotkin
- Subjects
Cancer Research ,medicine.medical_specialty ,business.industry ,Surrogate endpoint ,Nausea ,Phases of clinical research ,medicine.disease ,Meningioma ,Oncology ,Troponin I ,Vomiting ,Medicine ,Neurology (clinical) ,Radiology ,Progression-free survival ,medicine.symptom ,business ,Adverse effect - Abstract
Grade II/III meningiomas have increased rates of recurrence with no approved medical therapies. The historical progression-free survival at 6 months (PFS-6) is 25% with rates >35% declared of interest for drug development. NF2 gene inactivation occurs in about half of meningiomas. Based on our studies showing mTORC1 and mTORC2/SGK1 pathway activation in NF2-deficient meningiomas and the paradoxical activation of the mTORC2/AKT pathway, we hypothesized that mTORC1/mTORC2 inhibitors would be active in meningiomas. We studied the effect of vistusertib in patients with progressive/recurrent grade II/III meningiomas (NCT03071874). Vistusertib was administered orally at 125mg twice daily on two consecutive days each week. MRIs were obtained every 56 days. Tumor size was defined as the largest cross-sectional area. Progression was defined as ≥ 25% increase in the sum of products of all measurable lesions over smallest sum observed. The primary endpoint was PFS-6. Secondary endpoints included toxicity, radiographic response, and correlative studies including immunohistochemistry for mTORC1/2 pathway activation and genetic biomarkers. Twenty-eight patients (13 female, median age 58 years, median KPS 80%) were enrolled. Median tumor size was 4.4cm; 71% were grade II and 50% harbored pathogenic NF2 variants. Four patients discontinued treatment voluntarily and 1 each withdrew for intercurrent illness and non-compliance. PFS-6 is 47% (CI, 26%-65%) and OS-12 is 72% (95%CI, 48%-86%). PFS but not OS was shorter for patients with grade 3 meningiomas; there was no difference in PFS/OS between genetic groups. Adverse events at least possibly related to vistusertib with frequency >10% include nausea, fatigue, hypophosphatemia, diarrhea, anorexia, dry mouth, hypertriglyceridemia, hypertension, vomiting, increased ALT, constipation, and weight loss. Vistusertib treatment was associated with a PFS-6 rate exceeding the target of 35% for recurrent high-grade meningioma. Adverse events were tolerable in this patient population. These data support the continued development of mTORC1/2 inhibitors in this setting.
- Published
- 2021
29. Immunomodulatory and Mast Cell Stabilizing Potential of Ethanol Extract of Eleusinecoracana L.
- Author
-
Padmavathy, Kesavaram, primary and Vijaya Ramesh, K., additional
- Published
- 2019
- Full Text
- View/download PDF
30. Combination therapy with mTOR kinase inhibitor and dasatinib as a novel therapeutic strategy for vestibular schwannoma
- Author
-
Konstantina M. Stankovic, Yanling Zhang, Wenjianlong Zhou, Limeng Wu, Sasa Vasilijic, Patrick A. DeSouza, Lei Xu, Roberta L. Beauchamp, Jessica E. Sagers, Vijaya Ramesh, and Richard Seist
- Subjects
Male ,Dasatinib ,lcsh:Medicine ,mTORC1 ,Tyrosine-kinase inhibitor ,Receptor tyrosine kinase ,Mice ,0302 clinical medicine ,lcsh:Science ,Tumour-suppressor proteins ,Mice, Knockout ,0303 health sciences ,Neurofibromin 2 ,Multidisciplinary ,biology ,Receptor, EphA1 ,TOR Serine-Threonine Kinases ,Neuroma, Acoustic ,3. Good health ,030220 oncology & carcinogenesis ,Benzamides ,Drug Therapy, Combination ,Female ,biological phenomena, cell phenomena, and immunity ,medicine.drug ,Combination therapy ,medicine.drug_class ,Morpholines ,Article ,03 medical and health sciences ,In vivo ,medicine ,otorhinolaryngologic diseases ,Animals ,Humans ,Protein Kinase Inhibitors ,PI3K/AKT/mTOR pathway ,030304 developmental biology ,Pharmacology ,business.industry ,lcsh:R ,Erythropoietin-producing hepatocellular (Eph) receptor ,Mice, Inbred C57BL ,Disease Models, Animal ,Pyrimidines ,Cancer research ,biology.protein ,lcsh:Q ,business - Abstract
Neurofibromatosis type 2 (NF2) is an inherited disorder characterized by bilateral vestibular schwannomas (VS) that arise from neoplastic Schwann cells (SCs). NF2-associated VSs are often accompanied by meningioma (MN), and the majority of NF2 patients show loss of the NF2 tumor suppressor. mTORC1 and mTORC2-specific serum/glucocorticoid-regulated kinase 1 (SGK1) are constitutively activated in MN with loss of NF2. In a recent high-throughput kinome screen in NF2-null human arachnoidal and meningioma cells, we showed activation of EPH RTKs, c-KIT, and SFK members independent of mTORC1/2 activation. Subsequently, we demonstrated in vitro and in vivo efficacy of combination therapy with the dual mTORC1/2 inhibitor AZD2014 and the multi-kinase inhibitor dasatinib. For these reasons, we investigated activated mTORC1/2 and EPH receptor-mediated signaling in sporadic and NF2-associated VS. Using primary human VS cells and a mouse allograft model of schwannoma, we evaluated the dual mTORC1/2 inhibitor AZD2014 and the tyrosine kinase inhibitor dasatinib as monotherapies and in combination. Escalating dose-response experiments on primary VS cells grown from 15 human tumors show that combination therapy with AZD2014 and dasatinib is more effective at reducing metabolic activity than either drug alone and exhibits a therapeutic effect at a physiologically reasonable concentration (~0.1 µM). In vivo, while AZD2014 and dasatinib each inhibit tumor growth alone, the effect of combination therapy exceeds that of either drug. Co-targeting the mTOR and EPH receptor pathways with these or similar compounds may constitute a novel therapeutic strategy for VS, a condition for which there is no FDA-approved pharmacotherapy.
- Published
- 2019
31. First international conference on RASopathies and neurofibromatoses in Asia : identification and advances of new therapeutics
- Author
-
Vijaya Ramesh, D. Gareth Evans, Susan M Huson, Jaishri O. Blakeley, Joshi George, Ludwine Messiaen, Ype Elgersma, Rick van Minkelen, Pierre Wolkenstein, Eric Legius, Meena Upadhyaya, Shay Ben-Shachar, Anup Raji, Bruce R. Korf, Ratna Dua Puri, Katherine A. Rauen, Luis F. Parada, Miikka Vikkula, Brigitte C. Widemann, Shubha R. Phadke, Sheetal Sharda, Sheela Nampoothiri, Michael Fisher, Isabel Cordeiro, Abeer Al-Saegh, Ashok Pillai, Ian M. Frayling, Suma P. Shankar, Nancy Ratner, Scott R. Plotkin, Yemima Berman, Anant Tambe, Uday Khire, Bronwyn Kerr, Joanne Ngeow, Lee Kong Chian School of Medicine (LKCMedicine), First International Conference on RASopathies and Neurofibromatoses in Asia, and Neurosciences
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Biomedical ,Neurofibromatoses ,Intellectual and Developmental Disabilities (IDD) ,Clinical Sciences ,030105 genetics & heredity ,RASopathy ,Article ,Neurofibromatosis ,03 medical and health sciences ,Congenital ,Rare Diseases ,Costello syndrome ,Translational Research ,medicine ,Genetics ,Humans ,Genetic Predisposition to Disease ,Medicine [Science] ,Molecular Targeted Therapy ,Schwannomatosis ,Genetics (clinical) ,Genetic Association Studies ,Legius syndrome ,Pediatric ,therapy ,business.industry ,Neurosciences ,Disease Management ,signal transduction pathway ,medicine.disease ,Dermatology ,Clinical Trial ,Brain Disorders ,030104 developmental biology ,Molecular Diagnostic Techniques ,ras Proteins ,Noonan syndrome ,Mitogen-Activated Protein Kinases ,business ,Noonan Syndrome with Multiple Lentigines ,Biomarkers ,Signal Transduction - Abstract
The neurofibromatoses, which include neurofibromatosis type I (NF1), neurofibromatosis type II (NF2), and schwannomatosis, are a group of syndromes characterized by tumor growth in the nervous system. The RASopathies are a group of syndromes caused by germline mutations in genes that encode components of the RAS/mitogen-activated protein kinase (MAPK) pathway. The RASopathies include NF1, Noonan syndrome, Noonan syndrome with multiple lentigines, Costello syndrome, cardio-facio-cutaneous syndrome, Legius syndrome, capillary malformation arterio-venous malformation syndrome, and SYNGAP1 autism. Due to their common underlying pathogenetic etiology, all these syndromes have significant phenotypic overlap of which one common feature include a predisposition to tumors, which may be benign or malignant. Together as a group, they represent one of the most common multiple congenital anomaly syndromes estimating to affect approximately one in 1000 individuals worldwide. The subcontinent of India represents one of the largest populations in the world, yet remains underserved from an aspect of clinical genetics services. In an effort to bridge this gap, the First International Conference on RASopathies and Neurofibromatoses in Asia: Identification and Advances of New Therapeutics was held in Kochi, Kerala, India. These proceedings chronicle this timely and topical international symposium directed at discussing the best practices and therapies for individuals with neurofibromatoses and RASopathies. Children's Tumor Foundation; Fonds De La Recherche Scientifique ‐ FNRS, Grant/Award Numbers: T002614, T024719F; Manchester NIHR Biomedical Research Centre, Grant/Award Number: IS‐BRC‐1215‐20007; NIH/NIAMS, Grant/Award Number: R01AR062165; Axis Health Biomedicals and the Doctor's Academy; Schiller India; MedGenome; Wales Gene Park; University of Wales Trinity Saint David; Cardiff University; AstraZeneca
- Published
- 2019
32. Multi-center, single arm phase II study of the dual mTORC1/mTORC2 inhibitor vistusertib for patients with recurrent or progressive grade II-III meningiomas
- Author
-
Justin T. Jordan, Vijaya Ramesh, Scott R. Plotkin, Fred G. Barker, Alona Muzikansky, Anat Stemmer-Rachamimov, Patrick Y. Wen, Priya Kumthekar, and Roberta L. Beauchamp
- Subjects
Cancer Research ,medicine.medical_specialty ,Oncology ,business.industry ,VISTUSERTIB ,Medicine ,Phases of clinical research ,Center (algebra and category theory) ,Radiology ,business - Abstract
2024 Background: Grade II/III meningiomas represent about 20% of tumors and have increased rates of recurrence with no approved medical therapies. Historically, the progression-free survival at 6 months (PFS-6) for these tumors is 25%. The Response Assessment in Neuro-Oncology (RANO) group identified a PFS-6 rate of > 35% to be of interest for trials of grade II/III meningioma. Methods : NF2 gene inactivation occurs in the majority of meningiomas and is associated with mTORC1 activation. Human studies of everolimus for neurofibromatosis 2 patients documented growth arrest in only a minority of tumors. Based on our studies showing mTORC2/SGK1 pathway activation in NF2-deficient meningiomas and the known paradoxical activation of the mTORC2/AKT pathway in meningiomas, we hypothesized that dual inhibition of mTORC1/2 would be superior in meningiomas. Treatment of primary meningioma cells with vistusertib led to decreased cell proliferation and showed greater efficacy than rapamycin, regardless of NF2 expression. We studied the effect of vistusertib in patients with progressive or recurrent grade II/III meningiomas (NCT03071874). Vistusertib was administered orally at 125mg twice daily on two consecutive days each week. MRIs were obtained every 2 cycles (1 cycle = 28 days). Tumor size was defined as the largest cross-sectional area. Progression was defined as ≥25% increase in the sum of products of all measurable lesions over smallest sum observed. The primary endpoint was PFS-6. Secondary endpoints included toxicity, radiographic response, and correlative studies including immunohistochemistry for mTORC1/2 pathway activation and genetic biomarkers. Results: Twenty-eight patients (13 female), with a median age of 58 years (range, 32 to 77 years), were enrolled in this multicenter study. The median Karnofsky performance status was 80. Twenty-five patients have been followed to six months or to tumor progression. The median duration of treatment was 6.5 month (range, 1-18 months). Four patients chose to discontinue treatment, 1 withdrew to intercurrent illness, and 1 was withdrawn due to non-compliance. PFS-6 is 51.5% (CI, 29.3% - 70.0%). Adverse events at least possibly related to vistusertib with frequency > 10% include nausea (54%); fatigue (36%); hypophosphatemia (29%); diarrhea, anorexia, dry mouth, and hypertriglyceridemia (all 14%); hypertension, vomiting, increased ALT, constipation, and weight loss (all 11%). Conclusions: Vistusertib treatment was associated with a PFS-6 rate that exceeds the RANO target of 35% for recurrent high-grade meningioma. The follow-up data continue to mature. Adverse events were tolerable in this patient population. Correlative studies to identify biological factors that correlate with response are under way. These data support the initiation of larger randomized studies of vistusertib in this setting. Clinical trial information: NCT03071874.
- Published
- 2021
33. Evaluation of cellular immunological responses in mono- and polymorphic clinical forms of post-kala-azar dermal leishmaniasis in India
- Author
-
Kumar Avishek, Elodie Petitdidier, S. Kumar, Gérard-Marie Papierok, Poonam Salotra, D. Kumar Deep, Rachel Bras-Gonçalves, Vijaya Ramesh, Jean-Loup Lemesre, and Himanshu Kaushal
- Subjects
CD4-Positive T-Lymphocytes ,Cytotoxicity, Immunologic ,Male ,0301 basic medicine ,Cellular immunity ,Gene Expression ,CD8-Positive T-Lymphocytes ,Lymphocyte Activation ,Granzymes ,Parasite Load ,0302 clinical medicine ,granzyme B ,Immunology and Allergy ,Cytotoxic T cell ,Skin ,biology ,post-kala-azar dermal ,Flow Cytometry ,Killer Cells, Natural ,medicine.anatomical_structure ,polymorphic ,Cytokines ,Leishmaniasis, Visceral ,Female ,Adult ,Adolescent ,T cell ,Primary Cell Culture ,030231 tropical medicine ,Immunology ,India ,Leishmaniasis, Cutaneous ,Antigens, Protozoan ,Immunophenotyping ,03 medical and health sciences ,leishmania ,monomorphic ,Immune system ,Antigen ,parasitic diseases ,medicine ,Humans ,leishmaniasis ,Cell Proliferation ,Post-kala-azar dermal leishmaniasis ,Original Articles ,medicine.disease ,Granzyme B ,030104 developmental biology ,Granzyme ,Leukocytes, Mononuclear ,biology.protein ,Leishmania donovani - Abstract
Summary Post-kala-azar dermal leishmaniasis (PKDL) is a chronic dermal complication that occurs usually after recovery from visceral leishmaniasis (VL). The disease manifests into macular, papular and/or nodular clinical types with mono- or polymorphic presentations. Here, we investigated differences in immunological response between these two distinct clinical forms in Indian PKDL patients. Peripheral blood mononuclear cells of PKDL and naive individuals were exposed in vitro to total soluble Leishmania antigen (TSLA). The proliferation index was evaluated using an enzyme-linked immunosorbent assay (ELISA)-based lymphoproliferative assay. Cytokines and granzyme B levels were determined by cytometric bead array. Parasite load in tissue biopsy samples of PKDL was quantified by quantitative polymerase chain reaction (qPCR). The proportion of different lymphoid subsets in peripheral blood and the activated T cell population were estimated using flow cytometry. The study demonstrated heightened cellular immune responses in the polymorphic PKDL group compared to the naive group. The polymorphic group showed significantly higher lymphoproliferation, increased cytokines and granzyme B levels upon TSLA stimulation, and a raised proportion of circulating natural killer (NK) T cells against naive controls. Furthermore, the polymorphic group showed a significantly elevated proportion of activated CD4+ and CD8+ T cells upon in-vitro TSLA stimulation. Thus, the polymorphic variants showed pronounced cellular immunity while the monomorphic form demonstrated a comparatively lower cellular response. Additionally, the elevated level of both activated CD4+ and CD8+ T cells, coupled with high granzyme B secretion upon in-vitro TSLA stimulation, indicated the role of cytotoxic cells in resistance to L. donovani infection in polymorphic PKDL.
- Published
- 2016
34. CSIG-42. HIGH THROUGHPUT KINOME AND TRANSCRIPTOME ANALYSES REVEAL NOVEL THERAPEUTIC TARGETS IN NF2-DEFICIENT MENINGIOMA
- Author
-
Stephen J. Haggarty, James F. Gusella, Gary L. Johnson, Janet L. Oblinger, Steven P. Angus, Vijaya Ramesh, Roberta L. Beauchamp, Justin T. Jordan, Long-Sheng Chang, Timothy J. Stuhlmiller, Serkan Erdin, and Scott R. Plotkin
- Subjects
Transcriptome ,Cancer Research ,Abstracts ,Text mining ,Oncology ,business.industry ,otorhinolaryngologic diseases ,Kinome ,Neurology (clinical) ,Computational biology ,Biology ,business ,Throughput (business) - Abstract
Meningiomas (MN), the most common adult primary intracranial tumor, arise from the arachnoid/meninges and are non-responsive to chemotherapies with a high recurrence rate despite surgery, necessitating effective non-invasive therapies. Our previous work showed that NF2 loss activates mechanistic target of rapamycin complex 1 (mTORC1) and mTORC2 signaling, which led to past NF2 clinical trials using rapalogs (RAD001/everolimus), and current meningioma clinical trials with dual mTORC1/mTORC2 inhibitor (mTORi) AZD2014. To understand additional dysregulated, potentially druggable pathways, we undertook an ‘omics approach of large-scale kinomics and RNA-sequencing employing CRISPR-modified human arachnoidal cells (ACs), NF2-expressing vs NF2-null. In NF2-null ACs, several kinases were elevated including erythropoietin-producing hepatocellular (EPH)-receptor tyrosine kinase (RTK) family members, Src family kinase (SFK) members, and c-KIT, all targets of dasatinib. In vitro treatment of MN cells using mTORi (AZD2014 or INK128) and dasatinib enhanced growth inhibition upon combination mTORi+dasatinib. In vivo treatment of an orthotopic mouse MN model showed moderate response to dasatinib with stronger response using INK128 or INK128+dasatinib (e-published in Neuro-Oncology). Our transcriptomic data also revealed increased expression of several ligands/growth factors, particularly NRG1/neuregulin. Expanding these results, we have confirmed increased expression of NRG1 in human NF2-null ACs. We also find NF2-null ACs secrete NRG1, and in conditioned-media experiments we observe stimulation of ErbB3, EPHA2 and mTOR pathways, suggesting an autocrine signaling mechanism. NF2-null AC or MN cells, when stimulated with exogenous NRG1, show enhanced activation of mTOR and EPH pathways besides ErbB3 signaling. Further, lapatinib (multi-ErbB inhibitor) but not erlotinib (EGFR inhibitor) attenuates the NRG1-stimulated activation of ErbB3, EPHA2 and mTOR, suggesting that NRG1-induced activation is EGFR-independent. Taken together, our results support a mechanistic link where NF2 loss increases NRG1/ErbB signaling to EPH/SFK and mTOR pathways, which may be a critical driver of tumorigenesis, thus providing a therapeutic opportunity to co-target these pathways in NF2-deficient meningiomas.
- Published
- 2018
35. Immunomodulatory and Mast Cell Stabilizing Potential of Ethanol Extract of Eleusinecoracana L
- Author
-
Kesavaram Padmavathy and K. Vijaya Ramesh
- Subjects
Ethanol ,biology ,Bran ,Phagocytosis ,Public Health, Environmental and Occupational Health ,Degranulation ,Dietary fibre ,Pharmacology ,biology.organism_classification ,Mast cell ,In vitro ,chemistry.chemical_compound ,medicine.anatomical_structure ,chemistry ,medicine ,Candida albicans - Abstract
Eleusinecoracanal (seed)(finger millet bran)is rich in dietary fibre (free of glutan) and micronutrients and has been extensively studied for its anti-diabetic, anti-tumorogenic, anti-diarrheal, anti-ulcerative, anti-oxidant and anti-microbial potential. However, the immuno-modulatory potential and mast cell membrane stabilizing ability of E. coracana L (Ragi seeds) needs to be evaluated. Hence, this study was designed to evaluate the ability of ethanol extract of E. coracana L. to potentiate immunomodulation and inhibit mast cell degranulation of human macrophages and mice peritoneal mast cells respectively. Ethanol extraction of E. coracana L (Ragi seeds) was performed by cold percolation method. Assessment of In vitro immunomodulatory activity was carried out using human PMN layer, Candida albicans and morphological criteria was adopted to calculate the phagocytic index. In vitroevaluation of inhibition of mast cell degranulation was determined using isolated mice peritoneal mast cells. In vitro evaluation of phagocytosis revealed that ethanol extract of E. corocana L. (seed) induced vacuolation of human macrophages. Nevertheless, E. corocana L (seed, ethanol extract) (25 mg/mL) was found to inhibit polysorbate 80 induced mast cell degranulation.
- Published
- 2019
36. Pain correlates with germline mutation in schwannomatosis
- Author
-
Michael E. Talkowski, James F. Gusella, Vanessa L. Merker, Justin T. Jordan, Gordon J. Harris, Vijaya Ramesh, Serkan Erdin, Alessandra Suuberg, Miriam A. Bredella, Miriam J. Smith, Scott R. Plotkin, James A. Walker, Marlon Seijo, and Wenli Cai
- Subjects
Male ,0301 basic medicine ,Skin Neoplasms ,SMARCB1 ,Gastroenterology ,Cohort Studies ,0302 clinical medicine ,Quality of life ,Whole Body Imaging ,pain ,Schwannomatosis ,Pain Measurement ,Medicine(all) ,medicine.diagnostic_test ,Visual Analog Pain Scale ,Cancer Pain ,SMARCB1 Protein ,General Medicine ,Middle Aged ,Magnetic Resonance Imaging ,Tumor Burden ,Peripheral ,Female ,Neurilemmoma ,Research Article ,Cohort study ,Adult ,medicine.medical_specialty ,Neurofibromatoses ,Observational Study ,03 medical and health sciences ,Germline mutation ,Internal medicine ,medicine ,Humans ,Genetic Association Studies ,Germ-Line Mutation ,schwannomatosis ,business.industry ,whole-body MRI ,Magnetic resonance imaging ,medicine.disease ,030104 developmental biology ,Quality of Life ,LZTR1 ,business ,030217 neurology & neurosurgery ,Transcription Factors - Abstract
Schwannomatosis has been linked to germline mutations in the SMARCB1 and LZTR1 genes, and is frequently associated with pain. In a cohort study, we assessed the mutation status of 37 patients with clinically diagnosed schwannomatosis and compared to clinical data, whole body MRI (WBMRI), visual analog pain scale, and Short Form 36 (SF-36) bodily pain subscale. We identified a germline mutation in LZTR1 in 5 patients (13.5%) and SMARCB1 in 15 patients (40.5%), but found no germline mutation in 17 patients (45.9%). Peripheral schwannomas were detected in 3 LZTR1-mutant (60%) and 10 SMARCB1-mutant subjects (66.7%). Among those with peripheral tumors, the median tumor number was 4 in the LZTR1 group (median total body tumor volume 30 cc) and 10 in the SMARCB1 group (median volume 85cc), (P=.2915 for tumor number and P = .2289 for volume). mutation was associated with an increased prevalence of spinal schwannomas (100% vs 41%, P = .0197). The median pain score was 3.9/10 in the LZTR1 group and 0.5/10 in the SMARCB1 group (P = .0414), and SF-36 pain-associated quality of life was significantly worse in the LZTR1 group (P = .0106). Pain scores correlated with total body tumor volume (rho = 0.32471, P = .0499), but not with number of tumors (rho = 0.23065, P = .1696). We found no significant difference in quantitative tumor burden between mutational groups, but spinal schwannomas were more common in LZTR1-mutant patients. Pain was significantly higher in LZTR1-mutant than in SMARCB1-mutant patients, though spinal tumor location did not significantly correlate with pain. This suggests a possible genetic association with schwannomatosis-associated pain.
- Published
- 2018
37. Isolation and functional characterization Of bacterial strain b1 from erode region,Tamilnadu, india
- Author
-
VIJAYA NANDINI, GOVIN DAN, and VIJAYA RAMESH
- Subjects
Cell Biology ,Biology ,Isolation (microbiology) ,Molecular Biology ,Biochemistry ,Bacterial strain ,Biotechnology ,Microbiology - Published
- 2018
38. Survival benefit and phenotypic improvement by hamartin gene therapy in a tuberous sclerosis mouse brain model
- Author
-
Charles P. Lai, June Goto, Xuan Zhang, Roderick T. Bronson, Miguel Sena-Esteves, Xandra O. Breakefield, David J. Kwiatkowski, Sangyeul Han, Vijaya Ramesh, Shilpa Prabhakar, and Anat Stemmer-Rachamimov
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,Pathology ,medicine.medical_specialty ,Genetic enhancement ,Mutant ,Biology ,Article ,Tuberous Sclerosis Complex 1 Protein ,lcsh:RC321-571 ,Mice ,Tuberous sclerosis ,Gene therapy ,Tuberous Sclerosis ,medicine ,Animals ,Allele ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,PI3K/AKT/mTOR pathway ,TSC ,Neurons ,Tumor Suppressor Proteins ,Brain ,AAV ,Genetic Therapy ,medicine.disease ,TSC2 ,TSC1 ,Disease Models, Animal ,Phenotype ,Treatment Outcome ,medicine.anatomical_structure ,Neurology ,Tuberous sclerosis complex ,Mutation ,Cancer research ,Neuron - Abstract
We examined the potential benefit of gene therapy in a mouse model of tuberous sclerosis complex (TSC) in which there is embryonic loss of Tsc1 (hamartin) in brain neurons. An adeno-associated virus (AAV) vector (serotype rh8) expressing a tagged form of hamartin was injected into the cerebral ventricles of newborn pups with the genotype Tsc1cc (homozygous for a conditional floxed Tsc1 allele) SynI-cre+, in which Tsc1 is lost selectively in neurons starting at embryonic day 12. Vector-treated Tsc1ccSynIcre+ mice showed a marked improvement in survival from a mean of 22 days in non-injected mice to 52 days in AAV hamartin vector-injected mice, with improved weight gain and motor behavior in the latter. Pathologic studies showed normalization of neuron size and a decrease in markers of mTOR activation in treated as compared to untreated mutant littermates. Hence, we show that gene replacement in the brain is an effective therapeutic approach in this mouse model of TSC1. Our strategy for gene therapy has the advantages that therapy can be achieved from a single application, as compared to repeated treatment with drugs, and that AAV vectors have been found to have minimal to no toxicity in clinical trials for other neurologic conditions. Although there are many additional issues to be addressed, our studies support gene therapy as a useful approach in TSC patients.
- Published
- 2015
39. A high-throughput kinome screen reveals serum/glucocorticoid-regulated kinase 1 as a therapeutic target for NF2-deficient meningiomas
- Author
-
Vilas Wagh, Patrick A. DeSouza, Anat Stemmer-Rachamimov, Justin T. Jordan, Scott R. Plotkin, Wen-Ning Zhao, Stephen J. Haggarty, James F. Gusella, Roberta L. Beauchamp, Marianne James, and Vijaya Ramesh
- Subjects
Neurofibromatosis 2 ,Morpholines ,Immunoblotting ,Antineoplastic Agents ,mTORC1 ,Biology ,Protein Serine-Threonine Kinases ,mTORC2 ,meningioma ,Polymerase Chain Reaction ,Immediate early protein ,Immediate-Early Proteins ,Meningioma ,PAK1 ,AZD2014 ,Cell Line, Tumor ,medicine ,otorhinolaryngologic diseases ,Meningeal Neoplasms ,Humans ,Kinome ,SGK1 ,neoplasms ,Kinase ,High-Throughput Nucleotide Sequencing ,medicine.disease ,Molecular biology ,nervous system diseases ,Pyrimidines ,Oncology ,NF2 ,Gene Knockdown Techniques ,Benzamides ,Cancer research ,mTOR signaling ,Signal transduction ,Priority Research Paper ,Signal Transduction - Abstract
Meningiomas are the most common primary intracranial adult tumor. All Neurofibromatosis 2 (NF2)-associated meningiomas and ~60% of sporadic meningiomas show loss of NF2 tumor suppressor protein. There are no effective medical therapies for progressive and recurrent meningiomas. Our previous work demonstrated aberrant activation of mTORC1 signaling that led to ongoing clinical trials with rapamycin analogs for NF2 and sporadic meningioma patients. Here we performed a high-throughput kinome screen to identify kinases responsible for mTORC1 pathway activation in NF2-deficient meningioma cells. Among the emerging top candidates were the mTORC2-specific target serum/glucocorticoid-regulated kinase 1 (SGK1) and p21-activated kinase 1 (PAK1). In NF2-deficient meningioma cells, inhibition of SGK1 rescues mTORC1 activation, and SGK1 activation is sensitive to dual mTORC1/2 inhibitor AZD2014, but not to rapamycin. PAK1 inhibition also leads to attenuated mTORC1 but not mTORC2 signaling, suggesting that mTORC2/SGK1 and Rac1/PAK1 pathways are independently responsible for mTORC1 activation in NF2-deficient meningiomas. Using CRISPR-Cas9 genome editing, we generated isogenic human arachnoidal cell lines (ACs), the origin cell type for meningiomas, expressing or lacking NF2. NF2-null CRISPR ACs recapitulate the signaling of NF2-deficient meningioma cells. Interestingly, we observe increased SGK1 transcription and protein expression in NF2-CRISPR ACs and in primary NF2-negative meningioma lines. Moreover, we demonstrate that the dual mTORC1/mTORC2 inhibitor, AZD2014 is superior to rapamycin and PAK inhibitor FRAX597 in blocking proliferation of meningioma cells. Importantly, AZD2014 is currently in use in several clinical trials of cancer. Therefore, we believe that AZD2014 may provide therapeutic advantage over rapalogs for recurrent and progressive meningiomas.
- Published
- 2015
40. The path forward: 2015 International Children's Tumor Foundation conference on neurofibromatosis type 1, type 2, and schwannomatosis
- Author
-
Eric Legius, Vincent M. Riccardi, Anat Stemmer-Rachamimov, Meena Upadhyaya, Alison C. Lloyd, Vijaya Ramesh, Matthias A. Karajannis, Annette Bakker, Wade Clapp, Rosalie E. Ferner, Joseph L. Kissil, Roger J. Packer, David H. Gutmann, Michael Fisher, Anne Barker, Jaishri O. Blakeley, David A. Stevenson, Marco Giovannini, and Nicole J. Ullrich
- Subjects
0301 basic medicine ,Neurofibromatosis 2 ,Neurofibromatosis 1 ,Skin Neoplasms ,neurofibromatosis type 2 ,Neurofibromatoses ,Clinical Sciences ,rare disease ,Diagnostic tools ,Pediatrics ,neurofibromatosis type 1 ,03 medical and health sciences ,0302 clinical medicine ,therapeutic discovery ,medicine ,Genetics ,Humans ,Neurofibromatosis type 2 ,Neurofibromatosis ,Schwannomatosis ,Child ,Genetics (clinical) ,Discovery science ,schwannomatosis ,Foundation (evidence) ,medicine.disease ,pediatric tumors ,Rare tumor ,030104 developmental biology ,Engineering ethics ,Psychology ,030217 neurology & neurosurgery ,Neurilemmoma - Abstract
The Annual Children's Tumor Foundation International Neurofibromatosis Meeting is the premier venue for connecting discovery, translational and clinical scientists who are focused on neurofibromatosis types 1 and 2 (NF1 and NF2) and schwannomatosis (SWN). The meeting also features rare tumors such as glioma, meningioma, sarcoma, and neuroblastoma that occur both within these syndromes and spontaneously; associated with somatic mutations in NF1, NF2, and SWN. The meeting addresses both state of the field for current clinical care as well as emerging preclinical models fueling discovery of new therapeutic targets and discovery science initiatives investigating mechanisms of tumorigenesis. Importantly, this conference is a forum for presenting work in progress and bringing together all stakeholders in the scientific community. A highlight of the conference was the involvement of scientists from the pharmaceutical industry who presented growing efforts for rare disease therapeutic development in general and specifically, in pediatric patients with rare tumor syndromes. Another highlight was the focus on new investigators who presented new data about biomarker discovery, tumor pathogenesis, and diagnostic tools for NF1, NF2, and SWN. This report summarizes the themes of the meeting and a synthesis of the scientific discoveries presented at the conference in order to make the larger research community aware of progress in the neurofibromatoses.
- Published
- 2017
41. Post-kala-azar dermal leishmaniasis in HIV-coinfected individuals: problems in diagnosis and treatment
- Author
-
Kumar Avishek, Vijaya Ramesh, and Poonam Salotra
- Subjects
Male ,Anti-HIV Agents ,Antiprotozoal Agents ,Human immunodeficiency virus (HIV) ,Leishmania donovani ,Leishmaniasis, Cutaneous ,HIV Infections ,Dermatology ,medicine.disease_cause ,Immune reconstitution inflammatory syndrome ,Immune Reconstitution Inflammatory Syndrome ,medicine ,Humans ,Post-kala-azar dermal leishmaniasis ,biology ,Coinfection ,business.industry ,Middle Aged ,medicine.disease ,biology.organism_classification ,Immunology ,Facial Dermatosis ,Leishmaniasis, Visceral ,business ,Facial Dermatoses - Published
- 2014
42. Gene deleted live attenuated Leishmania vaccine candidates against visceral leishmaniasis elicit pro-inflammatory cytokines response in human PBMCs
- Author
-
Kumar Avishek, Sreenivas Gannavaram, Angamuthu Selvapandiyan, Vijaya Ramesh, Himanshu Kaushal, Poonam Salotra, Ranadhir Dey, Narender Singh Negi, Uma S. Dubey, and Hira L. Nakhasi
- Subjects
0301 basic medicine ,Male ,030231 tropical medicine ,Genes, Protozoan ,Leishmania donovani ,Cell Cycle Proteins ,Vaccines, Attenuated ,Article ,Proinflammatory cytokine ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,parasitic diseases ,medicine ,Humans ,Leishmaniasis Vaccines ,Multidisciplinary ,Attenuated vaccine ,biology ,Monokines ,Leishmaniasis ,Leishmania ,biology.organism_classification ,medicine.disease ,Virology ,030104 developmental biology ,Visceral leishmaniasis ,Gene Knockdown Techniques ,Immunology ,Leukocytes, Mononuclear ,Leishmaniasis, Visceral ,Female ,CD8 - Abstract
Currently no effective vaccine is available for human visceral leishmaniasis(VL) caused by Leishmania donovani. Previously, we showed that centrin1 and p27gene deleted live attenuated Leishmania parasites (LdCen1−/− and Ldp27−/−) are safe, immunogenic and protective in animal models. Here, to assess the correlates of protection, we evaluated immune responses induced by LdCen1−/− and Ldp27−/− in human blood samples obtained from healthy, healed VL (HVL), post kala-azar dermal leishmaniasis(PKDL) and VL subjects. Both parasites infected human macrophages, as effectively as the wild type parasites. Further, LdCen1−/− and Ldp27−/− strongly stimulated production of pro-inflammatory cytokines including, IL-12, IFN-γ, TNF-α, IL-2, IL-6 and IL-17 in the PBMCs obtained from individuals with a prior exposure to Leishmania (HVL and PKDL). There was no significant stimulation of anti-inflammatory cytokines (IL-4 and IL-10). Induction of Th1 biased immune responses was supported by a remarkable increase in IFN-γ secreting CD4+ and CD8+ T cells and IL-17 secreting CD4+ cells in PBMCs from HVL cases with no increase in IL-10 secreting T cells. Hence, LdCen1−/− and Ldp27−/− are promising as live vaccine candidates against VL since they elicit strong protective immune response in human PBMCs from HVL, similar to the wild type parasite infection, mimicking a naturally acquired protection following cure.
- Published
- 2016
43. Lichen scrofulosorum: importance of early recognition
- Author
-
Vijaya Ramesh and A. Molpariya
- Subjects
medicine.medical_specialty ,Tuberculosis ,integumentary system ,business.industry ,fungi ,Dermatology ,medicine.disease ,Asymptomatic ,030207 dermatology & venereal diseases ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,medicine ,medicine.symptom ,Young adult ,business ,Lichen scrofulosorum ,POSITIVE TUBERCULIN TEST - Abstract
Lichen scrofulosorum (LS), a tuberculid affecting children and young adults, usually presents with asymptomatic skin-coloured to erythematous follicular and perifollicular papules over the trunk and extremities. An underlying focus of tuberculosis can be detected, but occasionally other mycobacterial infections may be responsible. The eruption is associated with a positive tuberculin test or positive interferon-gamma release assay. Perifollicular granulomas and absence of bacilli are the histopathological characteristics. LS can go unsuspected because of its waxing and waning course and close resemblance to other dermatoses. This article reviews the different clinical presentations and various associations of LS. We reiterate that the focus of internal infection may be subtle and go undetected, emphasizing the need for correct diagnosis and early treatment.
- Published
- 2016
44. Decreased miltefosine susceptibility in clinical isolates of Leishmania donovani derived from visceral leishmaniasis and post kala-azar dermal leishmaniasis: Apparent mechanisms and clinical implications
- Author
-
Vijaya Ramesh, P. salotra, V. bhandari, D.K. Deep, Ruchi Singh, Narendra Singh Negi, and Vinod Sharma
- Subjects
Post-kala-azar dermal leishmaniasis ,Microbiology (medical) ,Miltefosine ,biology ,business.industry ,Leishmania donovani ,General Medicine ,medicine.disease ,biology.organism_classification ,Virology ,Visceral leishmaniasis ,Infectious Diseases ,Medicine ,business ,medicine.drug - Published
- 2016
- Full Text
- View/download PDF
45. Air-dried 3D-collagen-chitosan biocomposite scaffold for tissue engineering application
- Author
-
D. Vijaya Ramesh, B. Santhosh Kumar, and Sahaja Aigal
- Subjects
Scaffold ,Materials science ,Polymers and Plastics ,Composite number ,Substrate (chemistry) ,General Chemistry ,Chitosan ,chemistry.chemical_compound ,chemistry ,Tissue engineering ,Ultimate tensile strength ,Emulsion ,Materials Chemistry ,Ceramics and Composites ,Biocomposite ,Composite material - Abstract
Collagen–chitosan scaffolds of different compositions were developed using emulsion air-drying method. The scaffolds prepared adding 10–30 wt% of chitosan to collagen improved the mechanical properties of the composite scaffold, and 7:3 ratio (collagen :chitosan) was found to be a better composite having a tensile strength of 13.57 MPa with 9% elongation at break. The water-uptake characteristics were performed at different pH and found to be ameliorated for the composite scaffolds compared to pure collagen and chitosan scaffold, respectively. The pores ranging from 100 to 300 μm were well interconnected, and their distribution was fairly homogeneous in the scaffold as observed through scanning electron microscopy. Furthermore, the scaffold decreased the bacterial counts and supported fibroblasts attachment and proliferation, thus demonstrating this composite to be a good substrate for biomedical application.POLYM. COMPOS., 33:2029–2035, 2012. © 2012 Society of Plastics Engineers
- Published
- 2012
46. Preparation and comparative characterization of keratin–chitosan and keratin–gelatin composite scaffolds for tissue engineering applications
- Author
-
P. Neela Kanta Reddy, Ramadhar Kumar, P.K. Sehgal, Prachi Kakkar, D. Vijaya Ramesh, R. Sripriya, and S. Balaji
- Subjects
Scaffold ,Materials science ,food.ingredient ,Biocompatibility ,technology, industry, and agriculture ,Bioengineering ,Gelatin ,Artificial skin ,Biomaterials ,Chitosan ,chemistry.chemical_compound ,food ,Tissue engineering ,chemistry ,Chemical engineering ,Mechanics of Materials ,Ultimate tensile strength ,medicine ,Swelling ,medicine.symptom ,Composite material - Abstract
We report fabrication of three dimensional scaffolds with well interconnected matrix of high porosity using keratin, chitosan and gelatin for tissue engineering and other biomedical applications. Scaffolds were fabricated using porous Keratin–Gelatin (KG), Keratin–Chitosan (KC) composites. The morphology of both KG and KC was investigated using SEM. The scaffolds showed high porosity with interconnected pores in the range of 20–100 μm. They were further tested by FTIR, DSC, CD, tensile strength measurement, water uptake and swelling behavior. In vitro cell adhesion and cell proliferation tests were carried out to study the biocompatibility behavior and their application as an artificial skin substitute. Both KG and KC composite scaffolds showed similar properties and patterns for cell proliferation. Due to rapid degradation of gelatin in KG, we found that it has limited application as compared to KC scaffold. We conclude that KC scaffold owing to its slow degradation and antibacterial properties would be a better substrate for tissue engineering and other biomedical application.
- Published
- 2012
47. Back to the future: Proceedings from the 2010 NF Conference
- Author
-
Allan J. Belzberg, Maria T. Acosta, Filippo G. Giancotti, David Viskochil, Karlyne M. Reilly, Elizabeth K. Schorry, Kathryn N. North, Roger J. Packer, John W. Risner, Kim Hunter-Schaedle, Susan M Huson, Vijaya Ramesh, D. Gareth Evans, Juha Peltonen, Rosalie E. Ferner, Andre Bernards, Luis F. Parada, Robert Listernick, Meena Upadhyaya, Bruce R. Korf, Karen Cichowski, Jonathan Chernoff, Marco Giovannini, and Yuan Zhu
- Subjects
0303 health sciences ,medicine.medical_specialty ,Molecular signaling ,Blindness ,Extramural ,business.industry ,medicine.disease ,Disfigurement ,Article ,3. Good health ,Clinical trial ,03 medical and health sciences ,0302 clinical medicine ,Immunology ,Genetics ,medicine ,Severe morbidity ,Schwannomatosis ,Intensive care medicine ,business ,030217 neurology & neurosurgery ,Genetics (clinical) ,030304 developmental biology ,Neurofibromatoses - Abstract
The neurofibromatoses (NF) encompass the rare diseases NF1, NF2, and schwannomatosis. The NFs affect 100,000 Americans; over 2 million persons worldwide; and are caused by mutation of tumor suppressor genes. Individuals with NF1 in particular may develop tumors anywhere in the nervous system; additional manifestations can include learning disabilities, bone dysplasia, cardiovascular defects, unmanageable pain, and physical disfigurement. Ultimately, the NFs can cause blindness, deafness, severe morbidity, and increased mortality and NF1 includes a risk of malignant cancer. Today there is no treatment for the NFs (other than symptomatic); however, research efforts to understand these genetic conditions have made tremendous strides in the past few years. Progress is being made on all fronts, from discovery studies-understanding the molecular signaling deficits that cause the manifestations of NF-to the growth of preclinical drug screening initiatives and the emergence of a number of clinical trials. An important element in fuelling this progress is the sharing of knowledge, and to this end, for over 20 years the Children's Tumor Foundation has convened an annual NF Conference, bringing together NF professionals to share ideas and build collaborations. The 2010 NF Conference held in Baltimore, MD June 5-8, 2010 hosted over 300 NF researchers and clinicians. This paper provides a synthesis of the highlights presented at the Conference and as such, is a "state-of-the-field" for NF research in 2010.
- Published
- 2010
48. Traditional and systems biology based drug discovery for the rare tumor syndrome neurofibromatosis type 2
- Author
-
Jaishri O. Blakeley, Anat Stemmer-Rachamimov, Wen-Ning Zhao, Noah Sciaky, Sarah S. Burns, Cristina Fernandez-Valle, Marga Bott, Long-Sheng Chang, Gary L. Johnson, Vijaya Ramesh, Justin Guinney, Michael E. Talkowski, Steve Angus, Serkan Erdin, Annemarie Carlstedt, Alejandra M. Petrilli, Roberta L. Beauchamp, Charles W. Yates, Xin Chen, Stephen J. Haggarty, Patrick A. DeSouza, Tim J. Stuhlmiller, Abhishek Pratap, D. Bradley Welling, Jon S. Zawistowski, D. Wade Clapp, Scott R. Plotkin, Salvatore La Rosa, James F. Gusella, Helen Morrison, and Robert J. Allaway
- Subjects
0301 basic medicine ,Neurofibromatosis 2 ,Carcinogenesis ,Cell Survival ,Morpholines ,lcsh:Medicine ,Schwannoma ,Biology ,medicine.disease_cause ,Meningioma ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Cell Line, Tumor ,Panobinostat ,Meningeal Neoplasms ,otorhinolaryngologic diseases ,medicine ,Animals ,Humans ,Kinome ,Neurofibromatosis ,Neurofibromatosis type 2 ,lcsh:Science ,neoplasms ,Neurofibromin 2 ,Sulfonamides ,Multidisciplinary ,Systems Biology ,lcsh:R ,medicine.disease ,nervous system diseases ,Gene Expression Regulation, Neoplastic ,Pyridazines ,Merlin (protein) ,Pyrimidines ,030104 developmental biology ,chemistry ,Quinolines ,Cancer research ,lcsh:Q ,Transcriptome ,Neurilemmoma - Abstract
Neurofibromatosis 2 (NF2) is a rare tumor suppressor syndrome that manifests with multiple schwannomas and meningiomas. There are no effective drug therapies for these benign tumors and conventional therapies have limited efficacy. Various model systems have been created and several drug targets have been implicated in NF2-driven tumorigenesis based on known effects of the absence of merlin, the product of the NF2 gene. We tested priority compounds based on known biology with traditional dose-concentration studies in meningioma and schwann cell systems. Concurrently, we studied functional kinome and gene expression in these cells pre- and post-treatment to determine merlin deficient molecular phenotypes. Cell viability results showed that three agents (GSK2126458, Panobinostat, CUDC-907) had the greatest activity across schwannoma and meningioma cell systems, but merlin status did not significantly influence response. In vivo, drug effect was tumor specific with meningioma, but not schwannoma, showing response to GSK2126458 and Panobinostat. In culture, changes in both the transcriptome and kinome in response to treatment clustered predominantly based on tumor type. However, there were differences in both gene expression and functional kinome at baseline between meningioma and schwannoma cell systems that may form the basis for future selective therapies. This work has created an openly accessible resource (www.synapse.org/SynodosNF2) of fully characterized isogenic schwannoma and meningioma cell systems as well as a rich data source of kinome and transcriptome data from these assay systems before and after treatment that enables single and combination drug discovery based on molecular phenotype.
- Published
- 2018
49. What's new in neurofibromatosis? Proceedings from the 2009 NF Conference:New Frontiers
- Author
-
Vijaya Ramesh, Susan M Huson, Frank McCormick, Helen Morrison, Kim Hunter-Schaedle, Joseph L. Kissil, Rosalie E. Ferner, Nancy Ratner, Michel Kalamarides, Victor F. Mautner, Jaishri O. Blakeley, Roger J. Packer, David A. Stevenson, Kathryn N. North, and Katherine A. Rauen
- Subjects
medicine.medical_specialty ,business.industry ,Extramural ,Attendance ,Translational research ,medicine.disease ,Article ,Clinical trial ,Basic research ,Family medicine ,Genetics ,Medicine ,Neurofibromatosis type 2 ,Neurofibromatosis ,business ,Schwannomatosis ,Genetics (clinical) - Abstract
The NF Conference is the largest annual gathering of researchers and clinicians focused on neurofibromatosis and has been convened by the Children’s Tumor Foundation for over 20 years. The 2009 NF Conference was held in Portland, Oregon from June 13th – June 16th, 2009 and co-chaired by Kathryn North from the University of Sydney and The Children’s Hospital at Westmead, Sydney, Australia; and Joseph Kissil from the Wistar Institute, Philadelphia. The Conference included 80 platform presentations in 9 sessions over 4 days; over 100 abstracts presented as posters; and three Keynote presentations. To date, there have been tremendous advances in basic research in the pathogenesis of neurofibromatosis, and more recently in progress toward identifying effective drug therapies and the commencement of neurofibromatosis clinical trials. The NF Conference attendees have significantly increased (doubling from 140 in 2005 to 280 attending in 2009) with a significant increase in attendance of physicians and clinical researchers. Correspondingly the NF Conference scope has expanded to include translational research, clinical trials and clinical management issues while retaining a core of basic research. These themes are reflected in the highlights from the 2009 NF Conference presented here.
- Published
- 2010
50. NF2/Merlin Is a Novel Negative Regulator of mTOR Complex 1, and Activation of mTORC1 Is Associated with Meningioma and Schwannoma Growth
- Author
-
Vijaya Ramesh, Brendan D. Manning, Carolyn Polizzano, Anat Stemmer-Rachamimov, James F. Gusella, Sangyeul Han, Marianne James, and Scott R. Plotkin
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,Immunoblotting ,mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,Biology ,Transfection ,Cell Line ,Mice ,Phosphatidylinositol 3-Kinases ,Tumor Cells, Cultured ,otorhinolaryngologic diseases ,Animals ,Humans ,RNA, Small Interfering ,Protein kinase A ,Molecular Biology ,Transcription factor ,Cells, Cultured ,Cell Proliferation ,Mice, Knockout ,Sirolimus ,Neurofibromin 2 ,Antibiotics, Antineoplastic ,Cell growth ,TOR Serine-Threonine Kinases ,Cell Cycle ,Proteins ,Articles ,Cell Biology ,Fibroblasts ,Cell cycle ,Flow Cytometry ,Molecular biology ,Merlin (protein) ,Multiprotein Complexes ,Cancer research ,Arachnoid ,TSC2 ,Signal transduction ,Meningioma ,Proto-Oncogene Proteins c-akt ,Neurilemmoma ,Signal Transduction ,Transcription Factors - Abstract
Inactivating mutations of the neurofibromatosis 2 (NF2) gene, NF2, result predominantly in benign neurological tumors, schwannomas and meningiomas, in humans; however, mutations in murine Nf2 lead to a broad spectrum of cancerous tumors. The tumor-suppressive function of the NF2 protein, merlin, a membrane-cytoskeleton linker, remains unclear. Here, we identify the mammalian target of rapamycin complex 1 (mTORC1) as a novel mediator of merlin's tumor suppressor activity. Merlin-deficient human meningioma cells and merlin knockdown arachnoidal cells, the nonneoplastic cell counterparts of meningiomas, exhibit rapamycin-sensitive constitutive mTORC1 activation and increased growth. NF2 patient tumors and Nf2-deficient mouse embryonic fibroblasts demonstrate elevated mTORC1 signaling. Conversely, the exogenous expression of wild-type merlin isoforms, but not a patient-derived L64P mutant, suppresses mTORC1 signaling. Merlin does not regulate mTORC1 via the established mechanism of phosphoinositide 3-kinase-Akt or mitogen-activated protein kinase/extracellular signal-regulated kinase-mediated TSC2 inactivation and may instead regulate TSC/mTOR signaling in a novel fashion. In conclusion, the deregulation of mTORC1 activation underlies the aberrant growth and proliferation of NF2-associated tumors and may restrain the growth of these lesions through negative feedback mechanisms, suggesting that rapamycin in combination with phosphoinositide 3-kinase inhibitors may be therapeutic for NF2.
- Published
- 2009
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.