Network


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

Hotspot


Dive into the research topics where Michelle Monje is active.

Publication


Featured researches published by Michelle Monje.


Nature Medicine | 2002

Irradiation induces neural precursor-cell dysfunction

Michelle Monje; Shinichiro Mizumatsu; John R. Fike; Theo D. Palmer

In both pediatric and adult patients, cranial radiation therapy causes a debilitating cognitive decline that is poorly understood and currently untreatable. This decline is characterized by hippocampal dysfunction, and seems to involve a radiation-induced decrease in postnatal hippocampal neurogenesis. Here we show that the deficit in neurogenesis reflects alterations in the microenvironment that regulates progenitor-cell fate, as well as a defect in the proliferative capacity of the neural progenitor-cell population. Not only is hippocampal neurogenesis ablated, but the remaining neural precursors adopt glial fates and transplants of non-irradiated neural precursor cells fail to differentiate into neurons in the irradiated hippocampus. The inhibition of neurogenesis is accompanied by marked alterations in the neurogenic microenvironment, including disruption of the microvascular angiogenesis associated with adult neurogenesis and a marked increase in the number and activation status of microglia within the neurogenic zone. These findings provide clear targets for future therapeutic interventions.


Science | 2014

Neuronal Activity Promotes Oligodendrogenesis and Adaptive Myelination in the Mammalian Brain

Erin M. Gibson; David Purger; Christopher Mount; Andrea K. Goldstein; Grant Lin; Lauren Wood; Ingrid Inema; Sarah Miller; Gregor Bieri; J. Bradley Zuchero; Ben A. Barres; Pamelyn Woo; Hannes Vogel; Michelle Monje

Introduction Myelin is formed by mature oligodendrocytes to facilitate fast propagation of action potentials in axons. Small changes in myelin thickness can confer substantial changes in conduction speed and may thus alter neural circuit function. The idea that active neurons may modulate myelination is supported by in vitro studies and correlations between experience and myelin microstructure, but direct in vivo evidence demonstrating that neuronal activity regulates oligodendrocyte precursor cell (OPC) proliferation, differentiation, or changes in myelin microstructure has been lacking. We use in vivo optogenetic techniques in awake, behaving mice to provide direct evidence that neuronal activity regulates changes in myelin-forming cells within an active circuit. Neuronal activity promotes OPC proliferation, oligodendrogenesis, and myelin remodeling. Optogenetic stimulation of unilateral premotor cortex layer V projection neurons in awake, behaving Thy1::ChR2 mice promotes OPC proliferation (light green cells, red EdU+ nuclei), oligodendrogenesis (newly generated, EdU-marked oligodendrocyte; dark green cell), and an increase in myelin sheath thickness (viewed in cross section, gray). Together with these adaptive myelin changes, motor performance of the correlate limb is improved during normal gait 4 weeks after premotor cortex stimulation. Rationale Demonstrating the direct effects of behaviorally relevant neuronal activity on oligodendroglial lineage cells in vivo has been a challenge because traditional methods of directly promoting neuronal activity involve placement of an electrode, and the resultant tissue injury and subsequent inflammation affects OPC dynamics. Optogenetic technology allows for in vivo control of neuronal firing with millisecond precision using light delivered at a distance from the target and, thus, avoids extensive electrode-related tissue damage. We used an optogenetic (Thy1::ChR2) mouse model in which 470-nm light delivered near the brain surface stimulates the excitatory opsin channelrhodopsin expressed by cortical layer V projection neurons. Wild-type littermate controls lacking channelrhodopsin were identically manipulated to control for effects of surgery, optical fiber placement, and light exposure. In Thy1::ChR2 mice, light stimulation delivered unilaterally to the premotor cortex elicits complex motor behavior (unidirectional ambulation). The thymidine analog 5-ethynyl-2′-deoxyuridine (EdU) was administered at the time of optogenetic stimulation to mark actively dividing cells. Animals were evaluated at various time points to examine the effects of neuronal activity on myelin-forming cells and myelin microstructure, as well as the functional consequences of neuronal activity–regulated myelin changes. Results Optogenetic stimulation of cortical layer V projection neurons resulted in robust proliferation of OPCs within the premotor circuit, from the deep layers of the premotor cortex to the subcortical projections through the corpus callosum. Four weeks later, an increase in newly generated oligodendrocytes and increased myelin sheath thickness were found within the stimulated premotor circuit. Behavioral testing revealed increased swing speed of the correlate forelimb. Pharmacological blockade of OPC differentiation prevented activity-regulated oligodendrogenesis and myelin changes, as well as the associated behavioral change. Conclusion Neuronal activity regulates OPC proliferation, differentiation, and myelin remodeling in the murine brain with accompanying changes in behavioral function. Taken together, these findings suggest that adaptive changes in myelin-forming cells represent a type of behaviorally relevant neural plasticity, raising numerous conceptual and mechanistic questions. Mechanisms regulating myelin plasticity may be important for adaptive neural function and could be leveraged for interventions in diseases of myelin. Conversely, dysregulated myelin plasticity could conceivably contribute to disease. On-Demand Activity Oligodendroglia ensheath axons in the brain with myelin, which provides the insulation that speeds up transmission of neuronal electrical impulses. The process of myelination in the human brain goes on for decades, concurrent with all manner of brain development and cognitive activity. Gibson et al. (p. 10.1126/science.1252304, published online 10 April; see the Perspective by Bechler and ffrench-Constant) used optogenetics to study myelination in response to neural activity. Electrical activity in the motor cortex of the brain of awake mice led to proliferation and differentiation of oligodendrocytes and consequently increased myelination and alterations in motor response. Optogenetic stimulation of the mouse motor cortex incites proliferation of myelin-producing cells and axonal myelination. [Also see Perspective by Bechler and ffrench-Constant] Myelination of the central nervous system requires the generation of functionally mature oligodendrocytes from oligodendrocyte precursor cells (OPCs). Electrically active neurons may influence OPC function and selectively instruct myelination of an active neural circuit. In this work, we use optogenetic stimulation of the premotor cortex in awake, behaving mice to demonstrate that neuronal activity elicits a mitogenic response of neural progenitor cells and OPCs, promotes oligodendrogenesis, and increases myelination within the deep layers of the premotor cortex and subcortical white matter. We further show that this neuronal activity–regulated oligodendrogenesis and myelination is associated with improved motor function of the corresponding limb. Oligodendrogenesis and myelination appear necessary for the observed functional improvement, as epigenetic blockade of oligodendrocyte differentiation and myelin changes prevents the activity-regulated behavioral improvement.


Cancer Cell | 2013

Reduced H3K27me3 and DNA Hypomethylation Are Major Drivers of Gene Expression in K27M Mutant Pediatric High-Grade Gliomas

Sebastian Bender; Yujie Tang; Anders M. Lindroth; Volker Hovestadt; David T. W. Jones; Marcel Kool; Marc Zapatka; Paul A. Northcott; Dominik Sturm; Wei Wang; Bernhard Radlwimmer; Jonas W. Højfeldt; Nathalene Truffaux; David Castel; Simone Schubert; Marina Ryzhova; Huriye Şeker-Cin; Jan Gronych; Pascal-David Johann; Sebastian Stark; Jochen Meyer; Till Milde; Martin U. Schuhmann; Martin Ebinger; Camelia Maria Monoranu; Anitha Ponnuswami; Spenser Chen; Chris Jones; Olaf Witt; V. Peter Collins

Two recurrent mutations, K27M and G34R/V, within histone variant H3.3 were recently identified in ∼50% of pHGGs. Both mutations define clinically and biologically distinct subgroups of pHGGs. Here, we provide further insight about the dominant-negative effect of K27M mutant H3.3, leading to a global reduction of the repressive histone mark H3K27me3. We demonstrate that this is caused by aberrant recruitment of the PRC2 complex to K27M mutant H3.3 and enzymatic inhibition of the H3K27me3-establishing methyltransferase EZH2. By performing chromatin immunoprecipitation followed by next-generation sequencing and whole-genome bisulfite sequencing in primary pHGGs, we show that reduced H3K27me3 levels and DNA hypomethylation act in concert to activate gene expression in K27M mutant pHGGs.


Oncologist | 2008

Clinical Patterns and Biological Correlates of Cognitive Dysfunction Associated with Cancer Therapy

Jorg Dietrich; Michelle Monje; Jeffrey S. Wefel; Christina A. Meyers

Standard oncological therapies, such as chemotherapy and cranial radiotherapy, frequently result in a spectrum of neurocognitive deficits that includes impaired learning, memory, attention, and speed of information processing. In addition to classical mechanisms of neurotoxicity associated with chemo- and radiotherapy, such as radiation necrosis and leukoencephalopathy, damage to dynamic progenitor cell populations in the brain is emerging as an important etiologic factor. Radiation- and chemotherapy-induced damage to progenitor populations responsible for maintenance of white matter integrity and adult hippocampal neurogenesis is now believed to play a major role in the neurocognitive impairment many cancer survivors experience.


Nature Genetics | 2014

Recurrent activating ACVR1 mutations in diffuse intrinsic pontine glioma.

Kathryn R. Taylor; Alan Mackay; Nathalene Truffaux; Yaron S N Butterfield; Olena Morozova; Cathy Philippe; David Castel; Catherine S. Grasso; Maria Vinci; Diana Carvalho; Angel M. Carcaboso; Carmen Torres; Ofelia Cruz; Jaume Mora; Natacha Entz-Werle; Wendy J. Ingram; Michelle Monje; Darren Hargrave; Alex N. Bullock; Stéphanie Puget; Stephen Yip; Chris Jones; Jacques Grill

Diffuse intrinsic pontine gliomas (DIPGs) are highly infiltrative malignant glial neoplasms of the ventral pons that, due to their location within the brain, are unsuitable for surgical resection and consequently have a universally dismal clinical outcome. The median survival time is 9–12 months, with neither chemotherapeutic nor targeted agents showing substantial survival benefit in clinical trials in children with these tumors. We report the identification of recurrent activating mutations in the ACVR1 gene, which encodes a type I activin receptor serine/threonine kinase, in 21% of DIPG samples. Strikingly, these somatic mutations (encoding p.Arg206His, p.Arg258Gly, p.Gly328Glu, p.Gly328Val, p.Gly328Trp and p.Gly356Asp substitutions) have not been reported previously in cancer but are identical to mutations found in the germ line of individuals with the congenital childhood developmental disorder fibrodysplasia ossificans progressiva (FOP) and have been shown to constitutively activate the BMP–TGF-β signaling pathway. These mutations represent new targets for therapeutic intervention in this otherwise incurable disease.


Annals of Neurology | 2007

Impaired human hippocampal neurogenesis after treatment for central nervous system malignancies

Michelle Monje; Hannes Vogel; B A Marilyn Masek; Keith L. Ligon; Paul G. Fisher; Theo D. Palmer

The effects of cancer treatments such as cranial radiation and chemotherapy on human hippocampal neurogenesis remain unknown. In this study, we examine neuropathological markers of neurogenesis and inflammation in the human hippocampus after treatment for acute myelogenous leukemia or medulloblastoma. We demonstrate a persistent radiation‐induced microglial inflammation that is accompanied by nearly complete inhibition of neurogenesis after cancer treatment. These findings are consistent with preclinical animal studies and suggest potential therapeutic strategies. Ann Neurol 2007


Nature Medicine | 2015

Functionally defined therapeutic targets in diffuse intrinsic pontine glioma

Catherine S. Grasso; Yujie Tang; Nathalene Truffaux; Noah Berlow; Lining Liu; Marie Anne Debily; Michael J. Quist; Lara E. Davis; Elaine C. Huang; Pamelyn Woo; Anitha Ponnuswami; Spenser Chen; Tessa Johung; Wenchao Sun; Mari Kogiso; Yuchen Du; Lin Qi; Yulun Huang; Marianne Hütt-Cabezas; Katherine E. Warren; Ludivine Le Dret; Paul S. Meltzer; Hua Mao; Martha Quezado; Dannis G. van Vuurden; Jinu Abraham; Maryam Fouladi; Matthew N. Svalina; Nicholas Wang; Cynthia Hawkins

Diffuse intrinsic pontine glioma (DIPG) is a fatal childhood cancer. We performed a chemical screen in patient-derived DIPG cultures along with RNA-seq analyses and integrated computational modeling to identify potentially effective therapeutic strategies. The multi–histone deacetylase inhibitor panobinostat demonstrated therapeutic efficacy both in vitro and in DIPG orthotopic xenograft models. Combination testing of panobinostat and the histone demethylase inhibitor GSK-J4 revealed that the two had synergistic effects. Together, these data suggest a promising therapeutic strategy for DIPG.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Hedgehog-responsive candidate cell of origin for diffuse intrinsic pontine glioma

Michelle Monje; Siddhartha Mitra; Morgan Freret; Tal Raveh; James Kim; Marilyn Masek; Joanne L. Attema; Gordon Li; Terri Haddix; Michael S. B. Edwards; Paul G. Fisher; Irving L. Weissman; David H. Rowitch; Hannes Vogel; Albert J. Wong; Philip A. Beachy

Diffuse intrinsic pontine gliomas (DIPGs) are highly aggressive tumors of childhood that are almost universally fatal. Our understanding of this devastating cancer is limited by a dearth of available tissue for study and by the lack of a faithful animal model. Intriguingly, DIPGs are restricted to the ventral pons and occur during a narrow window of middle childhood, suggesting dysregulation of a postnatal neurodevelopmental process. Here, we report the identification of a previously undescribed population of immunophenotypic neural precursor cells in the human and murine brainstem whose temporal and spatial distributions correlate closely with the incidence of DIPG and highlight a candidate cell of origin. Using early postmortem DIPG tumor tissue, we have established in vitro and xenograft models and find that the Hedgehog (Hh) signaling pathway implicated in many developmental and oncogenic processes is active in DIPG tumor cells. Modulation of Hh pathway activity has functional consequences for DIPG self-renewal capacity in neurosphere culture. The Hh pathway also appears to be active in normal ventral pontine precursor-like cells of the mouse, and unregulated pathway activity results in hypertrophy of the ventral pons. Together, these findings provide a foundation for understanding the cellular and molecular origins of DIPG, and suggest that the Hh pathway represents a potential therapeutic target in this devastating pediatric tumor.


Nature | 2016

Single-cell RNA-seq supports a developmental hierarchy in human oligodendroglioma

Itay Tirosh; Andrew S. Venteicher; Christine Hebert; Leah E. Escalante; Anoop P. Patel; Keren Yizhak; Jonathan M. Fisher; Christopher Rodman; Christopher Mount; Mariella G. Filbin; Cyril Neftel; Niyati Desai; Jackson Nyman; Benjamin Izar; Christina C. Luo; Joshua M. Francis; Aanand A. Patel; Maristela L. Onozato; Nicolo Riggi; Kenneth J. Livak; Dave Gennert; Rahul Satija; Brian V. Nahed; William T. Curry; Robert L. Martuza; Ravindra Mylvaganam; A. John Iafrate; Matthew P. Frosch; Todd R. Golub; Miguel Rivera

Although human tumours are shaped by the genetic evolution of cancer cells, evidence also suggests that they display hierarchies related to developmental pathways and epigenetic programs in which cancer stem cells (CSCs) can drive tumour growth and give rise to differentiated progeny. Yet, unbiased evidence for CSCs in solid human malignancies remains elusive. Here we profile 4,347 single cells from six IDH1 or IDH2 mutant human oligodendrogliomas by RNA sequencing (RNA-seq) and reconstruct their developmental programs from genome-wide expression signatures. We infer that most cancer cells are differentiated along two specialized glial programs, whereas a rare subpopulation of cells is undifferentiated and associated with a neural stem cell expression program. Cells with expression signatures for proliferation are highly enriched in this rare subpopulation, consistent with a model in which CSCs are primarily responsible for fuelling the growth of oligodendroglioma in humans. Analysis of copy number variation (CNV) shows that distinct CNV sub-clones within tumours display similar cellular hierarchies, suggesting that the architecture of oligodendroglioma is primarily dictated by developmental programs. Subclonal point mutation analysis supports a similar model, although a full phylogenetic tree would be required to definitively determine the effect of genetic evolution on the inferred hierarchies. Our single-cell analyses provide insight into the cellular architecture of oligodendrogliomas at single-cell resolution and support the cancer stem cell model, with substantial implications for disease management.


Nature Medicine | 2014

Epigenetic targeting of Hedgehog pathway transcriptional output through BET bromodomain inhibition

Yujie Tang; Sharareh Gholamin; Simone Schubert; Minde Willardson; Alex G. Lee; Pratiti Bandopadhayay; Guillame Bergthold; Sabran Masoud; Brian Nguyen; Nujsaubnusi Vue; Brianna Balansay; Furong Yu; Sekyung Oh; Pamelyn Woo; Spenser Chen; Anitha Ponnuswami; Michelle Monje; Scott X. Atwood; Ramon J. Whitson; Siddhartha Mitra; Samuel H. Cheshier; Jun Qi; Rameen Beroukhim; Jean Y. Tang; Rob Wechsler-Reya; Anthony E. Oro; Brian A. Link; James E. Bradner; Yoon-Jae Cho

Hedgehog signaling drives oncogenesis in several cancers, and strategies targeting this pathway have been developed, most notably through inhibition of Smoothened (SMO). However, resistance to Smoothened inhibitors occurs by genetic changes of Smoothened or other downstream Hedgehog components. Here we overcome these resistance mechanisms by modulating GLI transcription through inhibition of bromo and extra C-terminal (BET) bromodomain proteins. We show that BRD4 and other BET bromodomain proteins regulate GLI transcription downstream of SMO and suppressor of fused (SUFU), and chromatin immunoprecipitation studies reveal that BRD4 directly occupies GLI1 and GLI2 promoters, with a substantial decrease in engagement of these sites after treatment with JQ1, a small-molecule inhibitor targeting BRD4. Globally, genes associated with medulloblastoma-specific GLI1 binding sites are downregulated in response to JQ1 treatment, supporting direct regulation of GLI activity by BRD4. Notably, patient- and GEMM (genetically engineered mouse model)-derived Hedgehog-driven tumors (basal cell carcinoma, medulloblastoma and atypical teratoid rhabdoid tumor) respond to JQ1 even when harboring genetic lesions rendering them resistant to Smoothened antagonists. Altogether, our results reveal BET proteins as critical regulators of Hedgehog pathway transcriptional output and nominate BET bromodomain inhibitors as a strategy for treating Hedgehog-driven tumors with emerged or a priori resistance to Smoothened antagonists.

Collaboration


Dive into the Michelle Monje's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yujie Tang

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chris Jones

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Kathryn R. Taylor

Institute of Cancer Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge