Network


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

Hotspot


Dive into the research topics where Anita Bhattacharyya is active.

Publication


Featured researches published by Anita Bhattacharyya.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Deficits in human trisomy 21 iPSCs and neurons

Jason P. Weick; Dustie L. Held; George F. Bonadurer; Matthew E. Doers; Yan Liu; Chelsie Maguire; Aaron D. Clark; Joshua Knackert; Katharine Molinarolo; Michael T. Musser; Lin Yao; Yingnan Yin; Jianfeng Lu; Xiaoqing Zhang; Su-Chun Zhang; Anita Bhattacharyya

Down syndrome (trisomy 21) is the most common genetic cause of intellectual disability, but the precise molecular mechanisms underlying impaired cognition remain unclear. Elucidation of these mechanisms has been hindered by the lack of a model system that contains full trisomy of chromosome 21 (Ts21) in a human genome that enables normal gene regulation. To overcome this limitation, we created Ts21-induced pluripotent stem cells (iPSCs) from two sets of Ts21 human fibroblasts. One of the fibroblast lines had low level mosaicism for Ts21 and yielded Ts21 iPSCs and an isogenic control that is disomic for human chromosome 21 (HSA21). Differentiation of all Ts21 iPSCs yielded similar numbers of neurons expressing markers characteristic of dorsal forebrain neurons that were functionally similar to controls. Expression profiling of Ts21 iPSCs and their neuronal derivatives revealed changes in HSA21 genes consistent with the presence of 50% more genetic material as well as changes in non-HSA21 genes that suggested compensatory responses to oxidative stress. Ts21 neurons displayed reduced synaptic activity, affecting excitatory and inhibitory synapses equally. Thus, Ts21 iPSCs and neurons display unique developmental defects that are consistent with cognitive deficits in individuals with Down syndrome and may enable discovery of the underlying causes of and treatments for this disorder.


Nature Reviews Neuroscience | 2001

Neurons from stem cells: preventing an identity crisis

Clive N. Svendsen; Anita Bhattacharyya; Yu-Tzu Tai

It is now possible to grow stem cells from a wide variety of tissues. Some of these cells have been shown to differentiate into presumptive neurons in vitro, or after transplantation into the developing or adult brain. When stem cells derived directly from the brain are induced to differentiate, there is a high probability that some of the resulting cells will be neurons. However, when stem cells from one tissue (for example, bone marrow or skin) take on the phenotype of another (for example, brain), rigorous criteria are required to define neurons. The aim of this review is to discuss the various techniques that are used to identify a cell as a neuron.


Stem Cells and Development | 2008

Normal Neurogenesis but Abnormal Gene Expression in Human Fragile X Cortical Progenitor Cells

Anita Bhattacharyya; Erin McMillan; Kyle Wallace; Thomas C. Tubon; Elizabeth E. Capowski; Clive N. Svendsen

Human stem and progenitor cells offer an innovative way to study early events in development. An exciting new opportunity for these cells is their application to study the underlying developmental consequences of genetic diseases. Because many diseases, ranging from leukemias to developmental disorders, are caused by single-gene defects, stem and progenitor cells that carry disease-causing genetic mutations are invaluable in understanding and treating disease. We have characterized human neural progenitor (hNPCs) cells that carry a single-gene defect that leads to the neurodevelopmental disorder Fragile X syndrome (FX). A loss-of-function mutation in the FMR1 gene leads to subtle changes in neural development and subsequent mental impairment characteristic of FX. hNPCs were isolated from fetal cortex carrying the FMR1 mutation to determine whether aberrations occur in their proliferation and differentiation. As expected, FX hNPCs have reduced expression of the FMR1 gene product Fragile X mental retardation protein (FMRP), and this decrease is maintained in culture and following differentiation. In contrast to a previously published report, the proliferation of FX hNPCs and their differentiation into neurons is not different from unaffected controls. Although the early development of FX hNPCs is essentially normal, microarray analysis reveals novel changes in the expression of signal transduction genes in FX hNPCs. Therefore, hNPCs have intrinsic characteristics that can be investigated to further our understanding and potential treatment of developmental disorders such as FX.


Developmental Neuroscience | 2009

A Critical Period in Cortical Interneuron Neurogenesis in Down Syndrome Revealed by Human Neural Progenitor Cells

Anita Bhattacharyya; Erin McMillan; Serene I. Chen; Kyle Wallace; Clive N. Svendsen

Down syndrome (DS) is a developmental disorder whose mental impairment is due to defective cortical development. Human neural progenitor cells (hNPCs) derived from fetal DS cortex initially produce normal numbers of neurons, but generate fewer neurons with time in culture, similar to the pattern of neurogenesis that occurs in DS in vivo. Microarray analysis of DS hNPCs at this critical time reveals gene changes indicative of defects in interneuron progenitor development. In addition, dysregulated expression of many genes involved in neural progenitor cell biology points to changes in the progenitor population and subsequent reduction in interneuron neurogenesis. Delineation of a critical period in interneuron development in DS provides a foundation for investigation of the basis of reduced neurogenesis in DS and defines a time when these progenitor cells may be amenable to therapeutic treatment.


PLOS ONE | 2007

The Cyclic AMP Cascade Is Altered in the Fragile X Nervous System

Daniel J. Kelley; Richard J. Davidson; Jamie L. Elliott; Garet P. Lahvis; Jerry C. P. Yin; Anita Bhattacharyya

Fragile X syndrome (FX), the most common heritable cause of mental retardation and autism, is a developmental disorder characterized by physical, cognitive, and behavioral deficits. FX results from a trinucleotide expansion mutation in the fmr1 gene that reduces levels of fragile X mental retardation protein (FMRP). Although research efforts have focused on FMRPs impact on mGluR signaling, how the loss of FMRP leads to the individual symptoms of FX is not known. Previous studies on human FX blood cells revealed alterations in the cyclic adenosine 3′, 5′-monophosphate (cAMP) cascade. We tested the hypothesis that cAMP signaling is altered in the FX nervous system using three different model systems. Induced levels of cAMP in platelets and in brains of fmr1 knockout mice are substantially reduced. Cyclic AMP induction is also significantly reduced in human FX neural cells. Furthermore, cAMP production is decreased in the heads of FX Drosophila and this defect can be rescued by reintroduction of the dfmr gene. Our results indicate that a robust defect in cAMP production in FX is conserved across species and suggest that cAMP metabolism may serve as a useful biomarker in the human disease population. Reduced cAMP induction has implications for the underlying causes of FX and autism spectrum disorders. Pharmacological agents known to modulate the cAMP cascade may be therapeutic in FX patients and can be tested in these models, thus supplementing current efforts centered on mGluR signaling.


Biochimica et Biophysica Acta | 2009

A systems biology approach to Down syndrome: Identification of Notch/Wnt dysregulation in a model of stem cells aging

Claire J. Cairney; Guido Sanguinetti; E. Ranghini; Andrew D. Chantry; M.C. Nostro; Anita Bhattacharyya; Clive N. Svendsen; W.N. Keith; Ilaria Bellantuono

Stem cells are central to the development and maintenance of many tissues. This is due to their capacity for extensive proliferation and differentiation into effector cells. More recently it has been shown that the proliferative and differentiative ability of stem cells decreases with age, suggesting that this may play a role in tissue aging. Down syndrome (DS), is associated with many of the signs of premature tissue aging including T-cell deficiency, increased incidence of early Alzheimer-type, Myelodysplastic-type disease and leukaemia. Previously we have shown that both hematopoietic (HSC) and neural stem cells (NSC) in patients affected by DS showed signs of accelerated aging. In this study we tested the hypothesis that changes in gene expression in HSC and NSC of patients affected by DS reflect changes occurring in stem cells with age. The profiles of genes expressed in HSC and NSC from DS patients highlight pathways associated with cellular aging including a downregulation of DNA repair genes and increases in proapoptotic genes, s-phase cell cycle genes, inflammation and angiogenesis genes. Interestingly, Notch signaling was identified as a potential hub, which when deregulated may drive stem cell aging. These data suggests that DS is a valuable model to study early events in stem cell aging.


Neuroscience & Biobehavioral Reviews | 2008

The cyclic AMP phenotype of fragile X and autism

Daniel J. Kelley; Anita Bhattacharyya; Garet P. Lahvis; Jerry C. P. Yin; Jim Malter; Richard J. Davidson

Cyclic AMP (cAMP) is a second messenger involved in many processes including mnemonic processing and anxiety. Memory deficits and anxiety are noted in the phenotype of fragile X (FX), the most common heritable cause of mental retardation and autism. Here we review reported observations of altered cAMP cascade function in FX and autism. Cyclic AMP is a potentially useful biochemical marker to distinguish autism comorbid with FX from autism per se and the cAMP cascade may be a viable therapeutic target for both FX and autism.


Science Translational Medicine | 2016

MDM2 inhibition rescues neurogenic and cognitive deficits in a mouse model of fragile X syndrome

Yue Li; Michael E. Stockton; Ismat Bhuiyan; Brian E. Eisinger; Yu Gao; Anita Bhattacharyya; Xinyu Zhao

The MDM2 inhibitor Nutlin-3 rescues neurogenic and cognitive deficits in mice with fragile X syndrome. MDM2 inhibitor rescues fragile X deficits Mutation of the FMRP protein in humans leads to fragile X syndrome, the most common inherited intellectual disability. Li et al. now show that FMRP controls the activities of neural stem cells in the adult mouse brain, which is critical for production of new neurons and learning and cognition. They discovered that FMRP regulates neural stem cells through controlling the expression of the E3 ubiquitin ligase MDM2. They found that treatment with an inhibitor of MDM2 called Nutlin-3 rebalanced neural stem cell activities and rescued cognitive deficits in a mouse model of fragile X syndrome. Fragile X syndrome, the most common form of inherited intellectual disability, is caused by loss of the fragile X mental retardation protein (FMRP). However, the mechanism remains unclear, and effective treatment is lacking. We show that loss of FMRP leads to activation of adult mouse neural stem cells (NSCs) and a subsequent reduction in the production of neurons. We identified the ubiquitin ligase mouse double minute 2 homolog (MDM2) as a target of FMRP. FMRP regulates Mdm2 mRNA stability, and loss of FMRP resulted in elevated MDM2 mRNA and protein. Further, we found that increased MDM2 expression led to reduced P53 expression in adult mouse NSCs, leading to alterations in NSC proliferation and differentiation. Treatment with Nutlin-3, a small molecule undergoing clinical trials for treating cancer, specifically inhibited the interaction of MDM2 with P53, and rescued neurogenic and cognitive deficits in FMRP-deficient mice. Our data reveal a potential regulatory role for FMRP in the balance between adult NSC activation and quiescence, and identify a potential new treatment for fragile X syndrome.


Stem Cells | 2017

Establishment of Reporter Lines for Detecting Fragile X Mental Retardation (FMR1) Gene Reactivation in Human Neural Cells

Meng Li; Huashan Zhao; Gene E. Ananiev; Michael T. Musser; Kathryn H. Ness; Dianne L. Maglaque; Krishanu Saha; Anita Bhattacharyya; Xinyu Zhao

Human patient‐derived induced pluripotent stem cells (hiPSCs) provide unique opportunities for disease modeling and drug development. However, adapting hiPSCs or their differentiated progenies to high throughput assays for phenotyping or drug screening has been challenging. Fragile X syndrome (FXS) is the most common inherited cause of intellectual disability and a major genetic cause of autism. FXS is caused by mutational trinucleotide expansion in the FMR1 gene leading to hypermethylation and gene silencing. One potential therapeutic strategy is to reactivate the silenced FMR1 gene, which has been attempted using both candidate chemicals and cell‐based screening. However, molecules that effectively reactivate the silenced FMR1 gene are yet to be identified; therefore, a high throughput unbiased screen is needed. Here we demonstrate the creation of a robust FMR1‐Nluc reporter hiPSC line by knocking in a Nano luciferase (Nluc) gene into the endogenous human FMR1 gene using the CRISPR/Cas9 genome editing method. We confirmed that luciferase activities faithfully report FMR1 gene expression levels and showed that neural progenitor cells derived from this line could be optimized for high throughput screening. The FMR1‐Nluc reporter line is a good resource for drug screening as well as for testing potential genetic reactivation strategies. In addition, our data provide valuable information for the generation of knockin human iPSC reporter lines for disease modeling, drug screening, and mechanistic studies. Stem Cells 2017;35:158–169


Scientific Reports | 2016

Efficient generation of region-specific forebrain neurons from human pluripotent stem cells under highly defined condition.

Fang Yuan; Kai-Heng Fang; Shi-Ying Cao; Zhuang-Yin Qu; Qi Li; Robert Krencik; Min Xu; Anita Bhattacharyya; Yuwen Su; Dong-Ya Zhu; Yan Liu

Human pluripotent stem cells (hPSCs) have potential to differentiate to unlimited number of neural cells, which provide powerful tools for neural regeneration. To date, most reported protocols were established with an animal feeder system. However, cells derived on this system are inappropriate for the translation to clinical applications because of the introduction of xenogenetic factors. In this study, we provided an optimized paradigm to generate region-specific forebrain neurons from hPSCs under a defined system. We assessed five conditions and found that a vitronectin-coated substrate was the most efficient method to differentiate hPSCs to neurons and astrocytes. More importantly, by applying different doses of purmorphamine, a small-molecule agonist of sonic hedgehog signaling, hPSCs were differentiated to different region-specific forebrain neuron subtypes, including glutamatergic neurons, striatal medium spiny neurons, and GABA interneurons. Our study offers a highly defined system without exogenetic factors to produce human neurons and astrocytes for translational medical studies, including cell therapy and stem cell-based drug discovery.

Collaboration


Dive into the Anita Bhattacharyya's collaboration.

Top Co-Authors

Avatar

Clive N. Svendsen

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Su-Chun Zhang

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Erin McMillan

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Xinyu Zhao

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Kyle Wallace

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Jason P. Weick

University of New Mexico

View shared research outputs
Top Co-Authors

Avatar

Krishanu Saha

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Matthew E. Doers

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Michael E. Stockton

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Michael T. Musser

University of Wisconsin-Madison

View shared research outputs
Researchain Logo
Decentralizing Knowledge