Maria Castro

Maria Castro, Ph.D.

R.C. Schneider Collegiate Professor of Neurosurgery
Professor of Cell and Developmental Biology

Biography

Maria G. Castro, PhD, is the R.C. Schneider Professor of Neurosurgery, and Professor of Cell and Developmental Biology, at the University of Michigan Medical School. My research program focuses on epigenetic regulation of cancer progression, uncovering the role of oncometabolites in the brain tumor microenvironment (TME), and the development of new therapies for adult and pediatric gliomas, including DIPG. We are investigating the role of the tumor immune-microenvironment in tumor progression and response to therapeutics, crosstalk between cancer cells and hematopoietic stem/progenitor cells, and mechanisms affecting the migration of immune cells from peripheral lymphoid organs to the tumor microenvironment. The study of these basic immunological mechanisms will lead to clinical implementation. The goal of my program is to develop novel treatments for brain tumors based on immunotherapeutics, a new and exciting approach that targets inhibition of tumor growth and recurrence.

Research Interests

Pediatric and adult brain cancer biology and therapeutics. Epigenetics, cancer metabolism, and signaling networks that mediate tumor progression. Uncovering the role of secreted cancer ligands in hematopoietic stem cell development and anti-tumor immunity. Nanotechnology to develop novel anti-cancer therapies. DNA damage, DNA damage response, and development of novel combination therapies for both adult and pediatric brain tumors, including novel immunotherapeutic approaches.

Mutant IDH1 Glioma Project

Glioma genetic models are needed to uncover mechanisms that mediate tumor progression, the interplay with the tumor microenvironment (TME) and response to therapeutics. We have generated the first genetically engineered mutant IDH1 mouse glioma model and isolated primary neurospheres (NS) from the tumors, which exhibit cancer stem cell-like properties. This has enabled us to develop a transplantable mIDH1 glioma model amenable to testing novel therapies. NS are derived from fully immune-competent (C57BL/6) mice, thus allowing examination of the TME and the impact of tumor mutations on the immune response. Our goals are to assess the effect of mIDH1 on transcription (using mRNA-seq) and on global DNA and histone methylation. The mIDH1 glioma model will also be used to identify promoter/enhancer region-specific changes in histone methylation marks (using chromatin immunoprecipitation followed by deep sequencing, or ChIP-seq). We are collaborating with Dr. Mats Ljungman, who pioneered bromouridine sequencing (Bru-seq), to identify and quantify nascent mRNA and gene transcription rates. Uncovering epigenetic patterning of histone 3 hypermethylation and cytosine modifications using next generation sequencing (NGS) technologies will contribute to the identification of novel pathways and gene regulatory networks which will provide novel insights into disease biology and uncover novel therapeutic targets.

DIPG Project

Diffuse intrinsic pontine glioma (DIPG) is a brainstem tumor that affects mainly children and for which there is no effective treatment. The most frequent DIPG mutations affect the N-terminal tail of histone variant H3.3 (encoded by H3F3A) and histone variant H3.1 (encoded by HIST1H3B) and result in the change of a lysine to methionine at residue 27, precluding methylation or acetylation of this key regulatory post-translational modification. In addition, six recurrent somatic activating mutations in ACVR1, which encodes for a bone morphogenetic protein (BMP) type-I receptor, have also been identified in DIPG tumors. BMP has very context-specific roles in the brain during development but its role in pediatric cancer remains unknown. We are using the Sleeping Beauty Transposase System to generate spontaneous in vivo tumor models that will allow us to analyze the contributions of these genes to DIPG pathogenesis. This work will elucidate how ACVR1 and H3K27M mutations contribute to DIPG progression and evaluate their impact on tumor response to DNA damaging agents. These studies will uncover novel therapeutic targets to improve prognosis for patients who suffer from DIPG.

Immunotherapy Development

The adult and pediatric brain tumor models we have established in our lab are ideal for developing and testing novel immunotherapies as they are implemented in mice with a fully competent immune system. We are using both immune mediated gene therapy strategies and nanoparticle-based vaccination approaches to develop new treatment strategies for these devastating brain cancers.

Clinical Trials

Our innovative work has led to an FDA-approved gene therapy Phase 1 clinical trial for malignant brain cancer which is currently enrolling patients at the University of Michigan.

Research Opportunities for Rotating Students

Immune-suppressive Myeloid Cells in the Glioma Microenvironment: Signaling Mechanisms and Novel Therapeutic Strategies; Interactions between the tumor cells and the neuro-immune microenvironment in mutant IDH1 gliomas; Immune-mediated therapies in a genetically engineered murine model of diffuse intrinsic pontine glioma

Publications

Haase S, Garcia-Fabiani MB, Carney S, Altshuler D, Nunez FJ, Mendez FM, Nunez F, Lowenstein PR, Castro MG. Mutant ATRX: uncovering a new therapeutic target for glioma. Expert Opinion on Therapeutic Targets, 2018 Jul;22(7):599-613. PMCID: PMC6044414.

Nunez F, Mendez FM, Kadiyala P, Alghamri MS, Savelieff MG, Garcia-Fabiani MB, Haase S, Koschmann C, Calinescu AA, Kamran N, Patel R, Carney S, Guo MZ, Edwards M, Ljungman M, Qin T, Sartor MA, Tagett R, Venneti S, Brosnan-Cashman J, Meeker A, Gorbunova V, Zhao L, Kremer DM, Zhang L, Lyssiotis CA, Jones L, Herting CJ, Ross JL, Hambardzumyan D, Hervey-Jumper S, Figueroa ME, Lowenstein PR*, Castro MG*. IDH1R132H acts as a tumor suppressor in glioma via epigenetic upregulation of the DNA damage response. Science Translational Medicine 2019 Feb 13;11(479). PMCID: PMC6400220. (*Corresponding authors) Accessible online: https://www.biorxiv.org/content/10.1101/389817v1

Kadiyala P, Li D, Nunez FM, Altshuler D, Doherty R, Kuai R, Yu M, Kamran N, Edwards M, Moon JJ, Lowenstein PR, Castro MG*, Schwendeman A*. High density lipoprotein-mimicking nanodiscs for chemo-immunotherapy against glioblastoma multiforme. American Chemical Society Nano 2019 Feb 26;13(2):1365-1384. PMCID: PMC6484828. *Co-corresponding, co-lead authors.

Calinescu AA, Yadav VN, Carballo E, Kadiyala P, Tran D, Zamler D, Doherty R, Srikanth M, Lowenstein PR, Castro MG. (2017) Survival and proliferation of neural progenitor derived glioblastomas under hypoxic stress is controlled by a CXCL12/CXCR4 autocrine positive feedback mechanism. Clinical Cancer Research, 2017 Mar 1;23(5):1250-1262. Epub 2016 Aug 19. PMCID: PMC5316506

Koschmann C, Nunez FJ, Mendez F, Brosnan-Cashman JA, Meeker Ak, Lowenstein PR, Castro MG. (2017) Mutated Chromatin Regulatory Factors as Tumor Drivers in Cancer. Cancer Research. 2017 Jan 15;77(2):227-233. Epub 2017 Jan 6. PMCID: PMC5243833.

Kamran N, Kadiyala P, Saxena M, Candolfi M, Li Y, Moreno-Ayala MA, Raja N, Shah D, Lowenstein PR, Castro MG. (2017) Immunosuppressive myeloid cells’ blockade in the glioma microenvironment enhances the efficacy of immune stimulatory gene therapy. Molecular Therapy. 2017 Jan 4;25(1):232-238. PMCID: PMC5363306

Koschmann C, Calinescu AA, Nunez FJ, Mackay A, Fazal-Salom J, Thomas D, Mulpuri L, Kamran N, Mendez F, Dzaman M, Krasinkiewicz J, Doherty R, Lemons R, Li Y, Roh S, Zhao L, Appleman H, Ferguson D, Gorbunova V, Meeker A, Jones C, Lowenstein PR, Castro MG. (2016) ATRX Loss Promotes Tumor Growth and Impairs Non-Homologous End Joining DNA Repair in Glioma. Science Translational Medicine, 2016 March 2;8(328):328ra28. PMCID: PMC5381643.

Candolfi M, Yagiz K, Wibowo M, Ahlzadeh G, Puntel M, Kamran N, Paran C, Ghiasi H, Lowenstein PR, Castro MG. (2014) Temozolomide does not impair gene therapy-mediated antitumor immunity in syngenic brain tumor models. Clinical Cancer Research. March 15, 2014;20(6):1555-65. Epub February 5, 2014. Cover article. PMCID: PMC3959570.

Sanderson NS, Puntel M, Kroeger KM, Bondale NS, Swerdlow M, Iranmanesh N, Yagita H, Ibrahim A, Castro MG, Lowenstein PR. (2012) Cytotoxic immunological synapses do not restrict the action of interferon-γ to antigenic target cells. Proceedings of the National Academy of Sciences, USA, 2012 May 15: 109(20):7835-40. Epub April 30, 2012. PMCID: PMC3356634.

Mineharu Y, King G, Muhammad AKM, Bannykh S, Kroeger K, Liu C, Lowenstein PR, Castro MG. (2011) Engineering the brain tumor microenvironment enhances the efficacy of dendritic cells' vaccination: implications for clinical trials design. Clinical Cancer Research, 17(14):4705-4718. Epub June 1, 2011. PMCID: PMC3208508.

Candolfi M, Xiong W, Yagiz K, Liu C, Muhammad AK, Puntel M, Foulad D, Zadmehr A, Ahlzadeh GE, Kroeger KM, Tesarfreund M, Lee S, Debinski W, Sareen D, Svendsen CN, Rodriguez R, Lowenstein PR, Castro MG. (2010) Gene therapy-mediated delivery of targeted cytotoxins for glioma therapeutics. Proceedings of the National Academy of Sciences, USA, 107(46):20021-20026. Epub Oct. 28, 2010. PMCID: PMC2993419.

Larocque D, Sanderson NRS, Bergeron J, Curtin J, Girton J, Wibowo M, Bondale N, Kroeger KM, Yang J, Lacayo LM, Reyes KC, Farrokh C, Pechnick RN, Castro MG, Lowenstein PR. (2010) Exogenous fms-like tyrosine kinase 3 ligand overrides brain immune privilege and facilitates recognition of a neo-antigen without causing autoimmune neuropathology. Proceedings of the National Academy of Sciences, USA, 107(32):14443-14448. Epub July 26, 2010. PMCID: PMC2922551.

Curtin JF, Liu N, Candolfi M, Xiong W, Assi H, Yagiz Y, Edwards MR, Michelsen KS, Kroeger KM, Liu C, Muhammad AKM, Clark MC, Arditi M, Comin-Anduix B, Ribas A, Lowenstein PR, Castro MG. (2009) HMGB1 mediates endogenous TLR2 activation and brain tumor regression. PLoS Medicine. 2009 January 13;6(1):e10. PMCID: PMC2621261.

http://www.ncbi.nlm.nih.gov/sites/myncbi/maria.castro.1/bibliography/404...