14 results on '"Tony E. Chavarria"'
Search Results
2. An EMT–primary cilium–GLIS2 signaling axis regulates mammogenesis and claudin-low breast tumorigenesis
- Author
-
Vincent J Guen, Jane E. Visvader, Qiong Ding, Philippe Juin, Matthieu Le Gallo, Anton M. Jetten, Tony E. Chavarria, Claude Prigent, Sophie G. Martin, Hong Soon Kang, Massimo Attanasio, Amandine Salamagnon, Patrick Tas, Arjun Bhutkar, Florence Godey, Roselyne Viel, Molly M. Wilson, Jacqueline A. Lees, Céline Callens, Svetlana Mironov, Robert A. Weinberg, Abena D. Peasah, Jonchère, Laurent, Massachusetts Institute of Technology (MIT), Institut de Génétique et Développement de Rennes (IGDR), Université de Rennes (UR)-Centre National de la Recherche Scientifique (CNRS)-Structure Fédérative de Recherche en Biologie et Santé de Rennes ( Biosit : Biologie - Santé - Innovation Technologique ), Chemistry, Oncogenesis, Stress and Signaling (COSS), Université de Rennes (UR)-CRLCC Eugène Marquis (CRLCC)-Institut National de la Santé et de la Recherche Médicale (INSERM), Carver College of Medicine, University of Iowa, H2P2 - Histo Pathologie Hight Precision (H2P2), Université de Rennes (UR)-Structure Fédérative de Recherche en Biologie et Santé de Rennes ( Biosit : Biologie - Santé - Innovation Technologique ), CRLCC Eugène Marquis (CRLCC), National Institute of Environmental Health Sciences [Durham] (NIEHS-NIH), National Institutes of Health [Bethesda] (NIH), Centre de Recherche en Cancérologie et Immunologie Nantes-Angers (CRCINA), Université d'Angers (UA)-Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Centre hospitalier universitaire de Nantes (CHU Nantes), University of Melbourne, Centre de recherche en Biologie cellulaire de Montpellier (CRBM), Université de Montpellier (UM)-Centre National de la Recherche Scientifique (CNRS), This work was supported by the MIT Stem Cell Initiative through Fondation MIT, Koch Institute Support (core) grant P30-CA14051 from the National Cancer Institute, Fondation ARC, Cancéropôle Grand Ouest, Université de Rennes 1, SIRIC ILIAD, and Fondation de France. M.M.W. was supported by the David H. Koch Graduate Fellowship. A.M.J. research was supported by the Intramural Research Program of the NIEHS, NIH Z01-ES-101585. V.J.G. was supported by Postdoctoral Fellowships from the Koch Institute and Fondation ARC., Structure Fédérative de Recherche en Biologie et Santé de Rennes ( Biosit : Biologie - Santé - Innovation Technologique )-Centre National de la Recherche Scientifique (CNRS)-Université de Rennes 1 (UR1), Université de Rennes (UNIV-RENNES)-Université de Rennes (UNIV-RENNES), Université de Rennes 1 (UR1), Université de Rennes (UNIV-RENNES)-Université de Rennes (UNIV-RENNES)-CRLCC Eugène Marquis (CRLCC)-Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Rennes (UNIV-RENNES)-Université de Rennes (UNIV-RENNES)-Structure Fédérative de Recherche en Biologie et Santé de Rennes ( Biosit : Biologie - Santé - Innovation Technologique ), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN)-Centre hospitalier universitaire de Nantes (CHU Nantes)-Centre National de la Recherche Scientifique (CNRS)-Université d'Angers (UA), Centre de recherche en Biologie Cellulaire (CRBM), Université Montpellier 2 - Sciences et Techniques (UM2)-Centre National de la Recherche Scientifique (CNRS)-Université de Montpellier (UM)-Université Montpellier 1 (UM1), Université de Rennes (UNIV-RENNES)-Université de Rennes (UNIV-RENNES)-Centre National de la Recherche Scientifique (CNRS)-Structure Fédérative de Recherche en Biologie et Santé de Rennes ( Biosit : Biologie - Santé - Innovation Technologique ), Institut National de la Santé et de la Recherche Médicale (INSERM)-CRLCC Eugène Marquis (CRLCC)-Université de Rennes 1 (UR1), and Université Montpellier 1 (UM1)-Université Montpellier 2 - Sciences et Techniques (UM2)-Université de Montpellier (UM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
[SDV.CAN]Life Sciences [q-bio]/Cancer ,[SDV.GEN] Life Sciences [q-bio]/Genetics ,Biology ,medicine.disease_cause ,03 medical and health sciences ,0302 clinical medicine ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,[SDV.BBM.GTP]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,Ciliogenesis ,medicine ,Transcription factor ,ComputingMilieux_MISCELLANEOUS ,Cancer ,030304 developmental biology ,[SDV.GEN]Life Sciences [q-bio]/Genetics ,0303 health sciences ,Multidisciplinary ,Fibroblast growth factor receptor 1 ,Cilium ,SciAdv r-articles ,Cell Biology ,Claudin-Low ,Cell biology ,030220 oncology & carcinogenesis ,ComputingMethodologies_DOCUMENTANDTEXTPROCESSING ,Biomedicine and Life Sciences ,Stem cell ,Carcinogenesis ,Ciliary base ,Research Article - Abstract
Description, EMT induces cilium assembly and signaling to promote mammogenesis and tumorigenesis., The epithelial-mesenchymal transition (EMT) and primary ciliogenesis induce stem cell properties in basal mammary stem cells (MaSCs) to promote mammogenesis, but the underlying mechanisms remain incompletely understood. Here, we show that EMT transcription factors promote ciliogenesis upon entry into intermediate EMT states by activating ciliogenesis inducers, including FGFR1. The resulting primary cilia promote ubiquitination and inactivation of a transcriptional repressor, GLIS2, which localizes to the ciliary base. We show that GLIS2 inactivation promotes MaSC stemness, and GLIS2 is required for normal mammary gland development. Moreover, GLIS2 inactivation is required to induce the proliferative and tumorigenic capacities of the mammary tumor–initiating cells (MaTICs) of claudin-low breast cancers. Claudin-low breast tumors can be segregated from other breast tumor subtypes based on a GLIS2-dependent gene expression signature. Collectively, our findings establish molecular mechanisms by which EMT programs induce ciliogenesis to control MaSC and MaTIC stemness, mammary gland development, and claudin-low breast cancer formation.
- Published
- 2021
3. Maternal Microbiota Modulate a Fragile X-like Syndrome in Offspring Mice
- Author
-
Bernard J. Varian, Katherine T. Weber, Lily J. Kim, Tony E. Chavarria, Sebastian E. Carrasco, Sureshkumar Muthupalani, Theofilos Poutahidis, Marwa Zafarullah, Reem R. Al Olaby, Mariana Barboza, Kemal Solakyildirim, Carlito Lebrilla, Flora Tassone, Fuqing Wu, Eric J. Alm, and Susan E. Erdman
- Subjects
Limosilactobacillus reuteri ,Intellectual and Developmental Disabilities (IDD) ,Lactobacillus reuteri ,microbiome ,Reproductive health and childbirth ,Mice ,Rare Diseases ,Pregnancy ,Complementary and Integrative Health ,Genetics ,2.1 Biological and endogenous factors ,Animals ,Humans ,Aetiology ,Genetics (clinical) ,Nutrition ,Pediatric ,Microbiota ,FXS ,FMRP ,probiotic ,Perinatal Period - Conditions Originating in Perinatal Period ,Brain Disorders ,Gastrointestinal Microbiome ,Good Health and Well Being ,Fragile X Syndrome ,Cytokines ,Dysbiosis ,Female ,Biotechnology - Abstract
Maternal microbial dysbiosis has been implicated in adverse postnatal health conditions in offspring, such as obesity, cancer, and neurological disorders. We observed that the progeny of mice fed a Westernized diet (WD) with low fiber and extra fat exhibited higher frequencies of stereotypy, hyperactivity, cranial features and lower FMRP protein expression, similar to what is typically observed in Fragile X Syndrome (FXS) in humans. We hypothesized that gut dysbiosis and inflammation during pregnancy influenced the prenatal uterine environment, leading to abnormal phenotypes in offspring. We found that oral in utero supplementation with a beneficial anti-inflammatory probiotic microbe, Lactobacillus reuteri, was sufficient to inhibit FXS-like phenotypes in offspring mice. Cytokine profiles in the pregnant WD females showed that their circulating levels of pro-inflammatory cytokine interleukin (Il)-17 were increased relative to matched gravid mice and to those given supplementary L. reuteri probiotic. To test our hypothesis of prenatal contributions to this neurodevelopmental phenotype, we performed Caesarian (C-section) births using dissimilar foster mothers to eliminate effects of maternal microbiota transferred during vaginal delivery or nursing after birth. We found that foster-reared offspring still displayed a high frequency of these FXS-like features, indicating significant in utero contributions. In contrast, matched foster-reared progeny of L. reuteri-treated mothers did not exhibit the FXS-like typical features, supporting a key role for microbiota during pregnancy. Our findings suggest that diet-induced dysbiosis in the prenatal uterine environment is strongly associated with the incidence of this neurological phenotype in progeny but can be alleviated by addressing gut dysbiosis through probiotic supplementation.
- Published
- 2022
4. An EMT-primary cilium-GLIS2 signaling axis regulates mammogenesis and claudin-low breast tumorigenesis
- Author
-
Sabrina Martin, M. M. Wilson, Jane E. Visvader, Céline Callens, Qiong Ding, Tony E. Chavarria, Jacqueline A. Lees, Claude Prigent, A. Peasah, Robert A. Weinberg, P. Tas, Massimo Attanasio, Florence Godey, Arjun Bhutkar, Svetlana Mironov, Anton M. Jetten, M. Le Gallo, Vincent J Guen, and A. Salamagnon
- Subjects
GLIS2 ,Ciliogenesis ,Cilium ,Fibroblast growth factor receptor 1 ,medicine ,Stem cell ,Biology ,Carcinogenesis ,medicine.disease_cause ,Claudin-Low ,Transcription factor ,Cell biology - Abstract
The Epithelial–Mesenchymal Transition (EMT) and primary ciliogenesis induce stem cell properties in basal Mammary Stem Cells (MaSCs) to promote mammogenesis, but the underlying mechanisms remain incompletely understood. Here, we show that EMT transcription factors promote ciliogenesis at intermediate EMT transition states by activating ciliogenesis inducers, including FGFR1. The resulting primary cilia promote BBS11-dependent ubiquitination and inactivation of a central signaling node, GLIS2. We show that GLIS2 inactivation promotes MaSC stemness, and GLIS2 is required for normal mammary gland development. Moreover, GLIS2 inactivation is required to induce the proliferative and tumorigenic capacities of the Mammary-Tumor-initiating cells (MaTICs) of claudin-low breast cancers. Claudin-low breast tumors can be segregated from other breast tumor subtypes based on a GLIS2-dependent gene expression signature. Collectively, our findings establish molecular mechanisms by which EMT programs induce ciliogenesis to control MaSC and MaTIC biology, mammary gland development, and claudin-low breast cancer formation.
- Published
- 2020
5. Phosphocholine accumulation and PHOSPHO1 depletion promote adipose tissue thermogenesis
- Author
-
Tony E. Chavarria, Bingbing Yuan, Mengxi Jiang, Harvey F. Lodish, and Nai-Jia Huang
- Subjects
Phosphorylcholine ,Adipose tissue ,chemistry.chemical_compound ,Mice ,Insulin resistance ,Adipose Tissue, Brown ,Commentaries ,Brown adipose tissue ,Gene expression ,medicine ,Choline ,Animals ,Uncoupling Protein 1 ,Phosphocholine ,Mice, Knockout ,Multidisciplinary ,Thermogenesis ,Biological Sciences ,medicine.disease ,Phosphoric Monoester Hydrolases ,Cell biology ,Cold Temperature ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Adipocytes, Brown ,chemistry ,Adipose Tissue ,Knockout mouse - Abstract
Phosphocholine phosphatase-1 (PHOSPHO1) is a phosphocholine phosphatase that catalyzes the hydrolysis of phosphocholine (PC) to choline. Here we demonstrate that the PHOSPHO1 transcript is highly enriched in mature brown adipose tissue (BAT) and is further induced by cold and isoproterenol treatments of BAT and primary brown adipocytes. In defining the functional relevance of PHOPSPHO1 in BAT thermogenesis and energy metabolism, we show that PHOSPHO1 knockout mice are cold-tolerant, with higher expression of thermogenic genes in BAT, and are protected from high-fat diet-induced obesity and development of insulin resistance. Treatment of mice with the PHOSPHO1 substrate phosphocholine is sufficient to induce cold tolerance, thermogenic gene expression, and allied metabolic benefits. Our results reveal a role of PHOSPHO1 as a negative regulator of BAT thermogenesis, and inhibition of PHOSPHO1 or enhancement of phosphocholine represent innovative approaches to manage the metabolic syndrome.
- Published
- 2020
6. SYK kinase mediates brown fat differentiation and activation
- Author
-
Anirudh Natarajan, Prathapan Thiru, Jacob B. Hansen, Marko Knoll, Dudley W. Lamming, Harvey F. Lodish, Mengxi Jiang, Tony E. Chavarria, Sally Winther, Aliakbar Shahsafaei, Lei Sun, and Huan Yang
- Subjects
Male ,0301 basic medicine ,Science ,General Physics and Astronomy ,Syk ,Stimulation ,chemical and pharmacologic phenomena ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Mediator ,Adipose Tissue, Brown ,Downregulation and upregulation ,Brown adipose tissue ,medicine ,Animals ,Humans ,Syk Kinase ,Glucose homeostasis ,Receptor ,lcsh:Science ,Cells, Cultured ,Uncoupling Protein 1 ,Cell Proliferation ,Mice, Knockout ,Multidisciplinary ,Chemistry ,Cell Differentiation ,hemic and immune systems ,General Chemistry ,Cell biology ,Mice, Inbred C57BL ,Adipocytes, Brown ,030104 developmental biology ,medicine.anatomical_structure ,lcsh:Q ,Thermogenesis - Abstract
Brown adipose tissue (BAT) metabolism influences glucose homeostasis and metabolic health in mice and humans. Sympathetic stimulation of β-adrenergic receptors in response to cold induces proliferation, differentiation, and UCP1 expression in pre-adipocytes and mature brown adipocytes. Here we show that spleen tyrosine kinase (SYK) is upregulated during brown adipocyte differentiation and activated by β-adrenergic stimulation. Deletion or inhibition of SYK, a kinase known for its essential roles in the immune system, blocks brown and white pre-adipocyte proliferation and differentiation in vitro, and results in diminished expression of Ucp1 and other genes regulating brown adipocyte function in response to β-adrenergic stimulation. Adipocyte-specific SYK deletion in mice reduces BAT mass and BAT that developed consisted of SYK-expressing brown adipocytes that had escaped homozygous Syk deletion. SYK inhibition in vivo represses β-agonist-induced thermogenesis and oxygen consumption. These results establish SYK as an essential mediator of brown fat formation and function., Spleen protein tyrosine kinase (Syk) has so far been mainly studied in haematopoietic and immune cells. Here, the authors show that Syk also has a role in brown adipose tissue, where it regulates the formation of brown adipocytes and their thermogenic activation in response to β-adrenergic stimulation.
- Published
- 2017
7. Abstract B61: The genotype of serous carcinomas shapes the tumor microenvironment and modulates responses to targeted and immune checkpoint therapies
- Author
-
Ferenc Reinhardt, Tony E. Chavarria, David Pépin, Tian Xia, Kevin M. Elias, Benjamin G. Neel, Esmee Hoefsmit, Shuang Zhang, Shailja Pathania, Paula T. Hammond, Yunlan Zhou, Sonia Iyer, Vera Poort, Anniina Färkkilä, Sean W. Smith, Robert A. Weinberg, and Raghav Mohan
- Subjects
Cancer Research ,Serous fluid ,Tumor microenvironment ,Oncology ,Genotype ,Cancer research ,Biology ,Immune checkpoint - Abstract
Immunotherapy in ovarian cancer has been disappointing, with only ~10% of patients responding to checkpoint blockade. The determinants of this low response rate remain poorly understood and there is a pressing need for immune-competent preclinical models to elucidate the biology of immune evasion in ovarian cancer. One critical area of interest is the role of homologous recombination (HR) DNA repair in immune evasion. The types and abundance of potential antigens present on cancer cells may depend on the genotype of the tumor, its mutational burden, and the cellular state. Unfortunately, the preclinical tools required to explore the relationship between the types of DNA damage repair deficiencies and immune evasion have been lacking. To this end, we have engineered novel syngeneic mouse models from murine fallopian tube epithelium using CRISPR/Cas9 technology. These tumors capture the most common combinations of co-occurring mutations observed in homologous recombination-deficient and -proficient patient samples. These models can identify the contribution of common driver mutations, which are TP53, BRCA1, PTEN, MYC, Cyclin E1 (CCNE1), AKT2, and Kras, to the heterotypic interactions between cancer and stromal/immune compartments and examine how DNA repair proficiency contributes to immunogenicity. To validate the DNA repair proficiency of the transformed cells, we measured Rad51 nuclear focus formation after ionizing radiation (IR) and PARP inhibitor and DNA-damaging agent sensitivity. The HR-deficient cell lines had significantly fewer Rad51 nuclear foci and were more sensitive to PARP inhibition in comparison to HR-proficient cells. Initial immune/stromal analysis using flow cytometry, single-cell RNASeq (scRNASeq) transcriptomic, and immunofluorescence imaging analysis revealed substantial differences in the myeloid and T-cell regulatory compartments between HR-proficient and -deficient primary and metastatic tumors and within the ascitic fluid. Preliminary results also suggest that inhibition of the DNA damage response (DDR), checkpoint kinase 1 (Chk1) in combination with immune checkpoint inhibitors, potentiates antitumor effects and augments cytotoxic T-cell infiltration. In conclusion, these results reveal how common mutational drivers, and particularly those associated with HR status, determine the microenvironment of the tumor and its response to treatment. Understanding the genetic basis of these complex cellular interactions will be critical to better tailor combinations of existing targeted treatments and immunotherapies in ovarian cancer to fight this devastating disease. Citation Format: Sonia Iyer, Shuang Zhang, Anniina Farkkila, Sean Smith, David Pepin, Raghav Mohan, Tian Xia, Ferenc Reinhardt, Tony Chavarria, Esmee Hoefsmit, Vera Poort, Shailja Pathania, Yunlan Zhou, Kevin M. Elias, Paula T. Hammond, Benjamin G. Neel, Robert A. Weinberg. The genotype of serous carcinomas shapes the tumor microenvironment and modulates responses to targeted and immune checkpoint therapies [abstract]. In: Proceedings of the AACR Special Conference on Advances in Ovarian Cancer Research; 2019 Sep 13-16, 2019; Atlanta, GA. Philadelphia (PA): AACR; Clin Cancer Res 2020;26(13_Suppl):Abstract nr B61.
- Published
- 2020
8. Abstract A29: The genomic architecture of serous carcinomas shapes the tumor microenvironment and modulates responses to targeted and immunotherapies
- Author
-
Sonia Iyer, Shuang Zhang, Anniina Farkkila, Sean G. Smith, David Pepin, Raghav Mohan, Esmee Hoefsmit, Tian Xia, Ferenc Reinhardt, Tony E. Chavarria, Shailja Pathania, Yunlan Zhou, Kevin M. Elias, Benjamin G. Neel, and Robert A. Weinberg
- Subjects
Cancer Research ,Immunology - Abstract
Immunotherapy in ovarian cancer has been disappointing, with only ~10% of patients responding to checkpoint blockade. The determinants of this low response rate remain poorly understood, and there is a pressing need for immune-competent preclinical models to elucidate the biology of immune evasion in ovarian cancer. One critical area of interest is the role of homologous recombination (HR) repair in immune evasion. The types and abundance of potential antigens present on cancer cells may depend on the genotype of the tumor, its mutational burden, and the cellular state. Unfortunately, the preclinical tools required to explore the relationship between the types of DNA damage repair deficiencies and immune evasion have been lacking. To this end, we have engineered novel syngeneic mouse models from murine fallopian tube epithelium using CRISPR/Cas9 technology. These tumors capture the most common combinations of co-occurring mutations observed in homologous recombination-deficient and -proficient patient samples. These models can identify the contribution of common driver mutations, which are TP53, BRCA1, PTEN, Myc, Cyclin E1 (CCNE1), Akt2, and Kras, to the heterotypic interactions between cancer and stromal/immune compartments and examine how DNA repair proficiency contributes to immunogenicity. To validate the DNA repair proficiency of the transformed cells, we measured Rad51 nuclear focus formation after ionizing radiation (IR) and PARP inhibitor and DNA-damaging agent sensitivity. The HR-deficient cell lines had significantly fewer Rad51 nuclear foci and were more sensitive to PARP inhibition in comparison to HR-proficient cells. Initial immune/stromal analysis using flow cytometry, single-cell RNA sequencing, and transcriptomic and immunofluorescence analysis revealed substantial differences in the myeloid and regulatory compartments between HR-proficient and -deficient tumors within the primary and metastatic tumors themselves and within the ascitic fluid. Preliminary results also suggest that inhibition of the DNA damage response (DDR), checkpoint kinase 1 (Chk1) in combination with immune checkpoint inhibitors, potentiates antitumor effects and augments cytotoxic T-cell infiltration. In conclusion, these results reveal how common mutational drivers, and particularly those associated with HR status, determine the microenvironment of the tumor and its response to treatment. Understanding the genetic basis of these complex cellular interactions will be critical to better tailor combinations of existing targeted treatments and immunotherapies in ovarian cancer to fight this devastating disease. Citation Format: Sonia Iyer, Shuang Zhang, Anniina Farkkila, Sean G. Smith, David Pepin, Raghav Mohan, Esmee Hoefsmit, Tian Xia, Ferenc Reinhardt, Tony E. Chavarria, Shailja Pathania, Yunlan Zhou, Kevin M. Elias, Benjamin G. Neel, Robert A. Weinberg. The genomic architecture of serous carcinomas shapes the tumor microenvironment and modulates responses to targeted and immunotherapies [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2019 Nov 17-20; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2020;8(3 Suppl):Abstract nr A29.
- Published
- 2020
9. Abstract PR04: The genomic architecture of serous carcinomas shapes the tumor microenvironment and modulates responses to targeted and immunotherapies
- Author
-
Tony E. Chavarria, Robert A. Weinberg, Kevin M. Elias, Yunlan Zhou, Sonia Iyer, Esmee Hoefsmit, Benjamin G. Neel, Ferenc Reinhardt, Anniina Färkkilä, Shuang Zhang, Tian Xia, David Pépin, Raghav Mohan, Sean W. Smith, and Shailja Pathania
- Subjects
Cancer Research ,Tumor microenvironment ,DNA repair ,medicine.medical_treatment ,Immunotherapy ,Biology ,medicine.disease ,Immune system ,Oncology ,PARP inhibitor ,Cancer cell ,medicine ,Cancer research ,CHEK1 ,Ovarian cancer - Abstract
High-grade serous ovarian cancer (HGSOC) is the most frequent and most aggressive histologic subtype of ovarian cancer. The cornerstone of the existing treatment of HGSOC is DNA-damaging chemotherapy; however, practically all patients eventually develop a progressive disease and the 5-year survival is only 40%. Immunotherapy would seem to be an attractive alternative treatment to chemotherapy, yet existing immunotherapies perform poorly in ovarian cancer, with only ~10% of patients responding to checkpoint blockade. Why this is the case remains poorly understood and there is a pressing need to understand the underlying biology of immune evasion in ovarian cancer. One critical area of interest is the role of homology-dependent DNA repair (HR) in immune evasion. The types and abundance of potential antigens present on cancer cells may depend on the genotype of the tumor, its mutational burden, and the cellular state. Unfortunately, the preclinical tools required to explore the relationship between the types of DNA damage repair deficiencies and immune evasion have been lacking. Hence, we have modeled the biology of ovarian cancer using patient-relevant mutational landscapes in an immune-proficient, syngeneic mouse model in order to help us identify the contribution of common driver mutations to the immune repertoire in the tumor microenvironment, and thus to responses of HGSOC tumors to immunotherapy. We hypothesize that the immune composition and gene expression signatures of the resulting tumors will vary based on the combination of genetic alterations and the DNA repair proficiency of the transformed cells. To this end, we have engineered novel syngeneic mouse models from murine fallopian tube epithelium using CRISPR/Cas9 technology. These tumors capture the most common combinations of co-occurring mutations observed in HR-deficient and -proficient patient samples. These models can identify the contribution of common driver mutations which are TP53, BRCA1, PTEN, MYC, Cyclin E1 (CCNE1), AKT2, and Kras to the heterotypic interactions between cancer and stromal/immune compartments and examine how DNA repair proficiency contributes to immunogenicity. To validate the DNA repair proficiency of the transformed cells, we measured Rad51 nuclear focus formation after ionizing radiation (IR) and PARP inhibitor and DNA-damaging agent sensitivity. The HR-deficient cell lines had significantly fewer Rad51 nuclear foci and were more sensitive to PARP inhibition in comparison to HR-proficient cells. Initial immune /stromal analysis using flow cytometry, scRNAseq transcriptomic and immunofluorescence analysis revealed substantial differences in the myeloid and T-cell regulatory compartments between HR-proficient and -deficient primary and metastatic tumors and within the ascitic fluid. Preliminary results also suggest that inhibition of the DNA damage response (DDR), checkpoint kinase 1 (Chk1) in combination with immune checkpoint inhibitors, potentiates antitumor effects and augments cytotoxic T-cell infiltration. In conclusion, these results reveal how common mutational drivers, and particularly those associated with HR status, determine the microenvironment of the tumor and its response to treatment. Understanding the genetic basis of these complex cellular interactions will be critical to better tailor combinations of existing targeted treatments and immunotherapies in ovarian cancer to fight this devastating disease. Citation Format: Sonia Iyer, Shuang Zhang, Anniina Farkkila, Sean Smith, David Pepin, Raghav Mohan, Ferenc Reinhardt, Tian Xia, Tony E. Chavarria, Esmee Hoefsmit, Shailja Pathania, Yunlan Zhou, Kevin M. Elias, Benjamin G. Neel, Robert A. Weinberg. The genomic architecture of serous carcinomas shapes the tumor microenvironment and modulates responses to targeted and immunotherapies [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference on Molecular Targets and Cancer Therapeutics; 2019 Oct 26-30; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2019;18(12 Suppl):Abstract nr PR04. doi:10.1158/1535-7163.TARG-19-PR04
- Published
- 2019
10. Excess cholesterol induces mouse egg activation and may cause female infertility
- Author
-
Ayce Yesilaltay, Nava Dekel, Eliza Vasile, Monty Krieger, Tony E. Chavarria, Dalia Walzer, Dalia Galiani, Linda Quilaqueo, David F. Albertini, Juan A. Orellana, David C. Page, Li Wang, Attilio Rigotti, Ruth Shalgi, Dolores Busso, and Gregoriy A Dokshin
- Subjects
Infertility ,medicine.medical_specialty ,Cell Survival ,MAP Kinase Signaling System ,Maturation promoting factor ,Polar Bodies ,Polar body ,chemistry.chemical_compound ,Mice ,Internal medicine ,medicine ,Animals ,Scavenger receptor ,Egtazic Acid ,Mice, Knockout ,Multidisciplinary ,Germinal vesicle ,biology ,Cholesterol ,urogenital system ,Female infertility ,Cholesterol, HDL ,beta-Cyclodextrins ,nutritional and metabolic diseases ,Oocyte activation ,Scavenger Receptors, Class B ,medicine.disease ,Mice, Inbred C57BL ,Meiosis ,Endocrinology ,chemistry ,PNAS Plus ,Strontium ,embryonic structures ,biology.protein ,Oocytes ,lipids (amino acids, peptides, and proteins) ,Female ,Infertility, Female - Abstract
The HDL receptor scavenger receptor, class B type I (SR-BI) controls the structure and fate of plasma HDL. Female SR-BI KO mice are infertile, apparently because of their abnormal cholesterol-enriched HDL particles. We examined the growth and meiotic progression of SR-BI KO oocytes and found that they underwent normal germinal vesicle breakdown; however, SR-BI KO eggs, which had accumulated excess cholesterol in vivo, spontaneously activated, and they escaped metaphase II (MII) arrest and progressed to pronuclear, MIII, and anaphase/telophase III stages. Eggs from fertile WT mice were activated when loaded in vitro with excess cholesterol by a cholesterol/methyl-β-cyclodextrin complex, phenocopying SR-BI KO oocytes. In vitro cholesterol loading of eggs induced reduction in maturation promoting factor and MAPK activities, elevation of intracellular calcium, extrusion of a second polar body, and progression to meiotic stages beyond MII. These results suggest that the infertility of SR-BI KO females is caused, at least in part, by excess cholesterol in eggs inducing premature activation and that cholesterol can activate WT mouse eggs to escape from MII arrest. Analysis of SR-BI KO female infertility raises the possibility that abnormalities in cholesterol metabolism might underlie some cases of human female infertility of unknown etiology.
- Published
- 2014
11. Essential function of Wnt-4 in mammary gland development downstream of progesterone signaling
- Author
-
Sudhansu K. Dey, Anna Heineman, Andrew P. McMahon, Tony E. Chavarria, Jill A. McMahon, Brian Elenbaas, Jian Tan, Robert A. Weinberg, and Cathrin Brisken
- Subjects
medicine.medical_specialty ,Morphogenesis ,Wnt signaling pathway ,Biology ,Epithelium ,Cell biology ,Transplantation ,medicine.anatomical_structure ,Endocrinology ,Mammary Epithelium ,Internal medicine ,Progesterone receptor ,Genetics ,medicine ,Ectopic expression ,Mammary gland morphogenesis ,Developmental Biology - Abstract
Female reproductive hormones control mammary gland morphogenesis. In the absence of the progesterone receptor (PR) from the mammary epithelium, ductal side-branching fails to occur. We can overcome this defect by ectopic expression of the protooncogene Wnt-1. Transplantation of mammary epithelia fromWnt-4−/− mice shows that Wnt-4 has an essential role in side-branching early in pregnancy. PR andWnt-4 mRNAs colocalize to the luminal compartment of the ductal epithelium. Progesterone induces Wnt-4 in mammary epithelial cells and is required for increased Wnt-4 expression during pregnancy. Thus, Wnt signaling is essential in mediating progesterone function during mammary gland morphogenesis.
- Published
- 2000
12. Laser-Assisted In Vitro Fertilization Facilitates Fertilization of Vitrified-Warmed C57BL/6 Mouse Oocytes with Fresh and Frozen-Thawed Spermatozoa, Producing Live Pups
- Author
-
John M. Mkandawire, Elizabeth K. Rosalia, Peimin Qi, Alexis García, James G. Fox, Tony E. Chavarria, Allan R. Discua, Stephanie E. Woods, Massachusetts Institute of Technology. Department of Biology, Massachusetts Institute of Technology. Division of Comparative Medicine, Koch Institute for Integrative Cancer Research at MIT, Woods, Stephanie E., Qi, Peimin, Rosalia, Elizabeth K., Chavarria, Tony E., Discua, Allan R., Mkandawire, John M., Fox, James G., and Garcia, Alexis
- Subjects
Male ,Anatomy and Physiology ,Pregnancy Rate ,Mouse ,medicine.medical_treatment ,lcsh:Medicine ,Veterinary Anatomy and Physiology ,Reproductive technology ,Cryopreservation ,Mice ,Sperm-Egg Interactions ,0302 clinical medicine ,Human fertilization ,Cell Movement ,Pregnancy ,Reproductive Physiology ,Animal Breeding ,Zona pellucida ,lcsh:Science ,reproductive and urinary physiology ,Animal Management ,0303 health sciences ,030219 obstetrics & reproductive medicine ,Multidisciplinary ,Obstetrics and Gynecology ,Embryo ,Animal Models ,Spermatozoa ,Embryo transfer ,3. Good health ,medicine.anatomical_structure ,Medicine ,Female ,Research Article ,Cell Survival ,Animal Types ,Mice, Transgenic ,Fertilization in Vitro ,Biology ,Andrology ,03 medical and health sciences ,Cryobiology ,Model Organisms ,Animal Production ,medicine ,Animal Genital Anatomy ,Animals ,Laboratory Animals ,030304 developmental biology ,In vitro fertilisation ,urogenital system ,Lasers ,lcsh:R ,Reproductive System ,Embryo, Mammalian ,Female Subfertility ,Sperm ,Mice, Inbred C57BL ,Fertilization ,Oocytes ,Veterinary Science ,lcsh:Q ,Developmental Biology - Abstract
The utility of cryopreserved mouse gametes for reproduction of transgenic mice depends on development of assisted reproductive technologies, including vitrification of unfertilized mouse oocytes. Due to hardening of the zona pellucida, spermatozoa are often unable to penetrate vitrified-warmed (V-W) oocytes. Laser-assisted in vitro fertilization (LAIVF) facilitates fertilization by allowing easier penetration of spermatozoa through a perforation in the zona. We investigated the efficiency of V-W C57BL/6NTac oocytes drilled by the XYClone laser, compared to fresh oocytes. By using DAP213 for cryoprotection, 83% (1,470/1,762) of vitrified oocytes were recovered after warming and 78% were viable. Four groups were evaluated for two-cell embryo and live offspring efficiency: 1) LAIVF using V-W oocytes, 2) LAIVF using fresh oocytes, 3) conventional IVF using V-W oocytes and 4) conventional IVF using fresh oocytes. First, the groups were tested using fresh C57BL/6NTac spermatozoa (74% motile, 15 million/ml). LAIVF markedly improved the two-cell embryo efficiency using both V-W (76%, 229/298) and fresh oocytes (69%, 135/197), compared to conventional IVF (7%, 12/182; 6%, 14/235, respectively). Then, frozen-thawed C57BL/6NTac spermatozoa (35% motile, 15 million/ml) were used and LAIVF was again found to enhance fertilization efficiency, with two-cell embryo rates of 87% (298/343) using V-W oocytes (P, National Institutes of Health (U.S.) (NIH National Research Service Award (T32-RR070036)), National Institutes of Health (U.S.) (NIH National Cancer Institute Program Project (P01CA10451))
- Published
- 2013
13. Reconstruction of functionally normal and malignant human breast tissues in mice
- Author
-
Min Wu, Greg Magrane, Charlotte Kuperwasser, Loucinda Carey, Joe W. Gray, Andrea L. Richardson, Tony E. Chavarria, and Robert A. Weinberg
- Subjects
Stromal cell ,Mammary gland ,Morphogenesis ,In situ hybridization ,Mice, SCID ,Biology ,medicine.disease_cause ,Transforming Growth Factor beta1 ,Mice ,Mammary Glands, Animal ,Transforming Growth Factor beta ,In vivo ,Mice, Inbred NOD ,medicine ,Animals ,Humans ,Breast ,In Situ Hybridization, Fluorescence ,Multidisciplinary ,Cancer ,Epithelial Cells ,Biological Sciences ,medicine.disease ,Immunohistochemistry ,Rats ,medicine.anatomical_structure ,Immunology ,Cancer research ,Commentary ,Stromal Cells ,Carcinogenesis ,Cell Division - Abstract
The study of normal breast epithelial morphogenesis and carcinogenesis in vivo has largely used rodent models. Efforts at studying mammary morphogenesis and cancer with xenotransplanted human epithelial cells have failed to recapitulate the full extent of development seen in the human breast. We have developed an orthotopic xenograft model in which both the stromal and epithelial components of the reconstructed mammary gland are of human origin. Genetic modification of human stromal cells before the implantation of ostensibly normal human mammary epithelial cells resulted in the outgrowth of benign and malignant lesions. This experimental model allows for studies of human epithelial morphogenesis and differentiation in vivo and underscores the critical role of heterotypic interactions in human breast development and carcinogenesis.
- Published
- 2004
14. Prolactin Controls Mammary Gland Development via Direct and Indirect Mechanisms
- Author
-
Sarabjeet Kaur, Paul A. Kelly, Robert L. Sutherland, Tony E. Chavarria, Robert A. Weinberg, Cathrin Brisken, Christopher J. Ormandy, and Nadine Binart
- Subjects
Heterozygote ,medicine.medical_specialty ,prolactin ,mammary gland ,prolactin receptor ,Receptors, Prolactin ,Mammary gland ,Biology ,Prolactin cell ,Mice ,Mammary Glands, Animal ,Pregnancy ,tissue recombination ,Internal medicine ,medicine ,Animals ,Endocrine system ,Molecular Biology ,development ,Gene knockout ,Mice, Knockout ,Histocytochemistry ,Prolactin receptor ,Gene Expression Regulation, Developmental ,Cell Biology ,Prolactin ,medicine.anatomical_structure ,Endocrinology ,Mammary Epithelium ,Tissue Transplantation ,Female ,Signal Transduction ,Developmental Biology ,Endocrine gland - Abstract
The inactivation of the prolactin receptor gene by homologous recombination has made it possible to investigate the role of prolactin signaling in mammary gland development without resort to ablative surgery of the endocrine glands. In knockout mice lacking the prolactin receptor, mammary development is normal up to puberty. Subsequently, the ducts branch less frequently than those of wild-type animals. While terminal end buds differentiate to alveolar buds in wild-type females by the end of puberty, in knockout females terminal end bud-like structures persist at the ductal ends. To distinguish between the developmental defects that are intrinsic to the epithelium and those that result from systemic endocrine alterations in prolactin receptor knockout mice, mammary epithelium from prolactin receptor knockouts was transplanted into mammary fat pads of wild-type mice. In virgin mice, the knockout epithelial transplants developed normally at puberty, indicating an indirect effect of prolactin on ductal development. Prolactin receptor knockout females are infertile due to multiple reproductive defects, but epithelial transplants allowed us to assess the extent to which the absence of prolactin receptor is limiting, under systemic conditions that allow full mammary gland development. During pregnancy, the prolactin receptor knockout transplants showed normal side branching and the formation of alveolar buds, but no lobuloalveolar development. Thus, prolactin affects mammary morphogenesis in two different ways: it controls ductal side branching and terminal end bud regression in virgin animals via indirect mechanisms, but acts directly on the mammary epithelium to produce lobuloalveolar development during pregnancy.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.