Network


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

Hotspot


Dive into the research topics where Tilat A. Rizvi is active.

Publication


Featured researches published by Tilat A. Rizvi.


Journal of Clinical Investigation | 2013

MEK inhibition exhibits efficacy in human and mouse neurofibromatosis tumors

Walter J. Jessen; Shyra J. Miller; Edwin Jousma; Jianqiang Wu; Tilat A. Rizvi; Meghan E. Brundage; David Eaves; Brigitte C. Widemann; Mi-Ok Kim; Eva Dombi; Jessica Sabo; Atira Hardiman Dudley; Michiko Niwa-Kawakita; Grier P. Page; Marco Giovannini; Bruce J. Aronow; Timothy P. Cripe; Nancy Ratner

Neurofibromatosis type 1 (NF1) patients develop benign neurofibromas and malignant peripheral nerve sheath tumors (MPNST). These incurable peripheral nerve tumors result from loss of NF1 tumor suppressor gene function, causing hyperactive Ras signaling. Activated Ras controls numerous downstream effectors, but specific pathways mediating the effects of hyperactive Ras in NF1 tumors are unknown. We performed cross-species transcriptome analyses of mouse and human neurofibromas and MPNSTs and identified global negative feedback of genes that regulate Ras/Raf/MEK/ERK signaling in both species. Nonetheless, ERK activation was sustained in mouse and human neurofibromas and MPNST. We used a highly selective pharmacological inhibitor of MEK, PD0325901, to test whether sustained Ras/Raf/MEK/ERK signaling contributes to neurofibroma growth in a neurofibromatosis mouse model (Nf1(fl/fl);Dhh-Cre) or in NF1 patient MPNST cell xenografts. PD0325901 treatment reduced aberrantly proliferating cells in neurofibroma and MPNST, prolonged survival of mice implanted with human MPNST cells, and shrank neurofibromas in more than 80% of mice tested. Our data demonstrate that deregulated Ras/ERK signaling is critical for the growth of NF1 peripheral nerve tumors and provide a strong rationale for testing MEK inhibitors in NF1 clinical trials.


Cancer Cell | 2008

Plexiform and Dermal Neurofibromas and Pigmentation Are Caused by Nf1 Loss in Desert Hedgehog-Expressing Cells

Jianqiang Wu; Jon P. Williams; Tilat A. Rizvi; Jennifer J. Kordich; David P. Witte; Dies Meijer; Anat Stemmer-Rachamimov; Jose A. Cancelas; Nancy Ratner

Neurofibromatosis type 1 (Nf1) mutation predisposes to benign peripheral nerve (glial) tumors called neurofibromas. The point(s) in development when Nf1 loss promotes neurofibroma formation are unknown. We show that inactivation of Nf1 in the glial lineage in vitro at embryonic day 12.5 + 1, but not earlier (neural crest) or later (mature Schwann cell), results in colony-forming cells capable of multilineage differentiation. In vivo, inactivation of Nf1 using a DhhCre driver beginning at E12.5 elicits plexiform neurofibromas, dermal neurofibromas, and pigmentation. Tumor Schwann cells uniquely show biallelic Nf1 inactivation. Peripheral nerve and tumors contain transiently proliferating Schwann cells that lose axonal contact, providing insight into early neurofibroma formation. We suggest that timing of Nf1 mutation is critical for neurofibroma formation.


Oncogene | 1998

Ruffling membrane, stress fiber, cell spreading and proliferation abnormalities in human Schwannoma cells

Patricia D. Pelton; Larry S. Sherman; Tilat A. Rizvi; Mark A. Marchionni; Patrick M. Wood; Rick A. Friedman; Nancy Ratner

Schwannomas are peripheral nerve tumors that typically have mutations in the NF2 tumor suppressor gene. We compared cultured schwannoma cells with Schwann cells from normal human peripheral nerves (NHSC). Both cell types expressed specific antigenic markers, interacted with neurons, and proliferated in response to glial growth factor, confirming their identity as Schwann cells. Schwannoma cells frequently had elevated basal proliferation compared to NHSC. Schwannoma cells also showed spread areas 5–7-fold greater than NHSC, aberrant membrane ruffling and numerous, frequently disorganized stress fibers. Dominant negative Rac inhibited schwannoma cell ruffling but had no apparent effect on NHSC. Schwannoma cell stress fibers were inhibited by C3 transferase, tyrphostin A25, or dominant negative RhoA. These data suggest that the Rho and Rac pathways are abnormally activated in schwannoma cells. Levels of ezrin and moesin, proteins related to the NF2 gene product, merlin, were unchanged in schwannoma cells compared to NHSC. Our findings demonstrate for the first time that cell proliferation and actin organization are aberrant in schwannoma cells. Because NF2 is mutant in most or all human schwannomas, we postulate that loss of NF2 contributes to the cell growth and cytoskeletal dysfunction reported here.


Neuroscience | 1999

The organization of preoptic-medullary circuits in the male rat: evidence for interconnectivity of neural structures involved in reproductive behavior, antinociception and cardiovascular regulation.

Anne Z. Murphy; Tilat A. Rizvi; Matthew Ennis; Michael T. Shipley

The present studies used anatomical tract-tracing techniques to delineate the organization of pathways linking the medial preoptic area and the ventral medulla, two key regions involved in neuroendocrine, autonomic and sensory regulation. Wheatgerm agglutinin-horseradish peroxidase injections into the ventromedial medulla retrogradely labeled a large number of neurons in the medial preoptic area, including both the median and medial preoptic nuclei. The termination pattern of preoptic projections to the medulla was mapped using the anterograde tracers Phaseolus vulgaris leucoagglutinin and biotinylated dextran amine. Tracer injections into the preoptic area produced a dense plexus of labeled fibers and terminals in the ventromedial and ventrolateral pons and medulla. Within the caudal pons/rostral medulla, medial preoptic projections terminated heavily in the nucleus raphe magnus; strong anterograde labeling was also present in the pontine reticular field. At mid-medullary levels, labeled fibers focally targeted the nucleus paragigantocellularis, in addition to the heavy fiber labeling present in the midline raphe nuclei. By contrast, very little labeling was observed in the caudal third of the medulla. Experiments were also conducted to map the distribution of ventral pontine and medullary neurons that project to the medial preoptic area. Wheatgerm agglutinin-horseradish peroxidase injections in the preoptic area retrogradely labeled a significant population of neurons in the ventromedial and ventrolateral medulla. Ascending projections from the medulla to the preoptic area were organized along rostral-caudal, medial-lateral gradients. In the caudal pons/rostral medulla, retrogradely labeled cells were aggregated along the midline raphe nuclei; no retrograde labeling was present laterally at this level. By contrast, in the caudal half of the medulla, cells retrogradely labeled from the medial preoptic area were concentrated as a discrete zone dorsal to the lateral reticular nucleus; labeled cells were not present in the ventromedial medulla at this level. The present findings suggest that the medial preoptic area and ventral midline raphe nuclei share reciprocal connections that are organized in a highly symmetrical fashion. By contrast, preoptic-lateral medullary pathways are not reciprocal. These preoptic-brainstem circuits may participate in antinociceptive, autonomic and reproductive behaviors.


The Journal of Neuroscience | 2005

Overexpression of the Epidermal Growth Factor Receptor Confers Migratory Properties to Nonmigratory Postnatal Neural Progenitors

Adan Aguirre; Tilat A. Rizvi; Nancy Ratner; Vittorio Gallo

Approaches to successful cell transplantation therapies for the injured brain involve selecting the appropriate neural progenitor type and optimizing the efficiency of the cell engraftment. Here we show that epidermal growth factor receptor (EGFR) expression enhances postnatal neural progenitor migration in vitro and in vivo. Migratory NG2-expressing (NG2+) progenitor cells of the postnatal subventricular zone (SVZ) express higher EGFR levels than nonmigratory, cortical NG2+ cells. The higher endogenous EGFR expression in SVZ NG2+ cells is causally related with their migratory potential in vitro as well as in vivo after cell engraftment. EGFR overexpression in cortical NG2+ cells by transient transfection converted these cells to a migratory phenotype in vitro and in vivo. Finally, cortical NG2+ cells purified from a transgenic mouse in which the EGFR is overexpressed under the CNP promoter exhibited enhanced migratory capability. These findings reveal a new role for EGFR in the postnatal brain and open new avenues to optimize cell engraftment for brain repair.


Archive | 1991

Topographical Specificity of Forebrain Inputs to the Midbrain Periaqueductal Gray: Evidence for Discrete Longitudinally Organized Input Columns

Michael T. Shipley; Matthew Ennis; Tilat A. Rizvi; Michael M. Behbehani

Over two decades ago it was discovered that electrical stimulation of the periaqueductal gray (PAG) caused profound analgesia (Reynolds, 1969). It was subsequently found that “PAG-analgesia” is, at least in part, mediated by opiate and neurotensin systems acting via PAG projections to the rostral medulla (Basbaum and Fields, 1984; Behbehani, 1981; Behbehani and Fields, 1979; Behbehani and Pert, 1984; Behbehani et al., 1987; Lakos and Basbaum, 1988; Reichling et al., 1988; Shipley et al., 1987). As a result of the observation that a discrete CNS structure exerted such a potent regulation of pain, much subsequent research has focused on the role of PAG in antinociception. At the same time there has been growing evidence that PAG plays a key role in the “defense reaction” (Bandler and Carrive, 1988; Bandler and Depaulis, this volume; Bandler et al., 1985a, 1991; Depaulis and Vergnes, 1986; Depaulis et al., 1989; Zhang et al., 1990), vocalization (Jurgens, 1976; Jurgens and Richter, 1986; Larson, 1985; Larson and Kistler, 1984; 1986), and in certain sexual behaviors (Ogawa et al., this volume; Sakuma and Pfaff; 1979a,b).


The New England Journal of Medicine | 2016

Activity of Selumetinib in Neurofibromatosis Type 1–Related Plexiform Neurofibromas

Eva Dombi; Andrea Baldwin; Leigh Marcus; Michael J. Fisher; Brian Weiss; AeRang Kim; Patricia Whitcomb; Staci Martin; Lindsey Aschbacher-Smith; Tilat A. Rizvi; Jianqiang Wu; Rachel Ershler; Pamela L. Wolters; Janet Therrien; John Glod; Jean B. Belasco; Elizabeth K. Schorry; Alessandra Brofferio; Amy J. Starosta; Andrea Gillespie; Austin L. Doyle; Nancy Ratner; Brigitte C. Widemann

BACKGROUND Effective medical therapies are lacking for the treatment of neurofibromatosis type 1-related plexiform neurofibromas, which are characterized by elevated RAS-mitogen-activated protein kinase (MAPK) signaling. METHODS We conducted a phase 1 trial of selumetinib (AZD6244 or ARRY-142886), an oral selective inhibitor of MAPK kinase (MEK) 1 and 2, in children who had neurofibromatosis type 1 and inoperable plexiform neurofibromas to determine the maximum tolerated dose and to evaluate plasma pharmacokinetics. Selumetinib was administered twice daily at a dose of 20 to 30 mg per square meter of body-surface area on a continuous dosing schedule (in 28-day cycles). We also tested selumetinib using a mouse model of neurofibromatosis type 1-related neurofibroma. Response to treatment (i.e., an increase or decrease from baseline in the volume of plexiform neurofibromas) was monitored by using volumetric magnetic resonance imaging analysis to measure the change in size of the plexiform neurofibroma. RESULTS A total of 24 children (median age, 10.9 years; range, 3.0 to 18.5) with a median tumor volume of 1205 ml (range, 29 to 8744) received selumetinib. Patients were able to receive selumetinib on a long-term basis; the median number of cycles was 30 (range, 6 to 56). The maximum tolerated dose was 25 mg per square meter (approximately 60% of the recommended adult dose). The most common toxic effects associated with selumetinib included acneiform rash, gastrointestinal effects, and asymptomatic creatine kinase elevation. The results of pharmacokinetic evaluations of selumetinib among the children in this trial were similar to those published for adults. Treatment with selumetinib resulted in confirmed partial responses (tumor volume decreases from baseline of ≥20%) in 17 of the 24 children (71%) and decreases from baseline in neurofibroma volume in 12 of 18 mice (67%). Disease progression (tumor volume increase from baseline of ≥20%) has not been observed to date. Anecdotal evidence of decreases in tumor-related pain, disfigurement, and functional impairment was observed. CONCLUSIONS Our early-phase data suggested that children with neurofibromatosis type 1 and inoperable plexiform neurofibromas benefited from long-term dose-adjusted treatment with selumetinib without having excess toxic effects. (Funded by the National Institutes of Health and others; ClinicalTrials.gov number, NCT01362803 .).


Journal of Neuropathology and Experimental Neurology | 1995

Neurofibromin Expression and Astrogliosis in Neurofibromatosis (Type 1) Brains

Michael L. Nordlund; Tilat A. Rizvi; Camilynn I. Brannan; Nancy Ratner

Abstract . Patients with type 1 neurofibromatosis (NF1) have mutations in the gene encoding the protein neurofibromin. Immunocytochemistry on sections of cortex and cerebellum of unaffected and NF1 individuals and wild-type and NF1-deficient mice showed that the distribution of neurofibromin was similar to that reported for rat. However, dystrophic neurofibromin-expressing neurons were found in human but not rodent brain. Intensity of anti-neurofibromin reactivity was reduced in NF1-deficient mice but not in human brains. GFAP was upregulated in three NF1 brains studied by immunocytochemistry; a 4–18-fold increase in GFAP levels was documented by Western blot analysis in three brains. GFAP content/ cell and the number of GFAP-immunoreactive astrocytes was increased in NF1 brains as compared to the controls. These results suggest that mutations in the NF1 gene do not grossly alter the pattern of neurofibromin expression, but activation of astrocytes may be common in NF1. Presence of degenerative debris in one of two brains using the cupric silver method suggests that degeneration is not always detectable in NF1 brains.


Cancer Research | 2012

PTEN and NF1 inactivation in Schwann cells produces a severe phenotype in the peripheral nervous system that promotes the development and malignant progression of peripheral nerve sheath tumors.

Vincent W. Keng; Eric P. Rahrmann; Adrienne L. Watson; Barbara R. Tschida; Christopher L. Moertel; Walter J. Jessen; Tilat A. Rizvi; Margaret H. Collins; Nancy Ratner; David A. Largaespada

The genetic evolution from a benign neurofibroma to a malignant sarcoma in patients with neurofibromatosis type 1 (NF1) syndrome remains unclear. Schwann cells and/or their precursor cells are believed to be the primary pathogenic cell in neurofibromas because they harbor biallelic neurofibromin 1 (NF1) gene mutations. However, the phosphatase and tensin homolog (Pten) and neurofibromatosis 1 (Nf1) genes recently were found to be comutated in high-grade peripheral nerve sheath tumors (PNST) in mice. In this study, we created transgenic mice that lack both Pten and Nf1 in Schwann cells and Schwann cell precursor cells to validate the role of these two genes in PNST formation in vivo. Haploinsufficiency or complete loss of Pten dramatically accelerated neurofibroma development and led to the development of higher grade PNSTs in the context of Nf1 loss. Pten dosage, together with Nf1 loss, was sufficient for the progression from low-grade to high-grade PNSTs. Genetic analysis of human malignant PNSTs (MPNST) also revealed downregulation of PTEN expression, suggesting that Pten-regulated pathways are major tumor-suppressive barriers to neurofibroma progression. Together, our findings establish a novel mouse model that can rapidly recapitulate the onset of human neurofibroma tumorigenesis and the progression to MPNSTs.


Cancer Research | 2011

Perinatal or adult Nf1 inactivation using tamoxifen-inducible PlpCre each cause neurofibroma formation

Debra A. Mayes; Tilat A. Rizvi; Jose A. Cancelas; Nathan T. Kolasinski; Georgianne Ciraolo; Anat Stemmer-Rachamimov; Nancy Ratner

Plexiform neurofibromas are peripheral nerve sheath tumors initiated by biallelic mutation of the NF1 tumor suppressor gene in the Schwann cell lineage. To understand whether neurofibroma formation is possible after birth, we induced Nf1 loss of function with an inducible proteolipid protein Cre allele. Perinatal loss of Nf1 resulted in the development of small plexiform neurofibromas late in life, whereas loss in adulthood caused large plexiform neurofibromas and morbidity beginning 4 months after onset of Nf1 loss. A conditional EGFP reporter allele identified cells showing recombination, including peripheral ganglia satellite cells, peripheral nerve S100β+ myelinating Schwann cells, and peripheral nerve p75+ cells. Neurofibromas contained cells with Remak bundle disruption but no recombination within GFAP+ nonmyelinating Schwann cells. Extramedullary lympho-hematopoietic expansion was also observed in PlpCre;Nf1fl/fl mice. These tumors contained EGFP+/Sca-1+ stromal cells among EGFP-negative lympho-hematopoietic cells indicating a noncell autonomous effect and unveiling a role of Nf1-deleted microenvironment on lympho-hematopoietic proliferation in vivo. Together these findings define a tumor suppressor role for Nf1 in the adult and narrow the range of potential neurofibroma-initiating cell populations.

Collaboration


Dive into the Tilat A. Rizvi's collaboration.

Top Co-Authors

Avatar

Nancy Ratner

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jianqiang Wu

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Timothy P. Cripe

Nationwide Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Punam Malik

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Shyra J. Miller

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eva Dombi

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Jose A. Cancelas

Cincinnati Children's Hospital Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge