18 results on '"Jean J. Zhao"'
Search Results
2. Genomic Landscape and Immunological Profile of Glioblastoma in East Asians
- Author
-
Sheng Zhong, Bo Wu, Frank Dubois, Davy Deng, Tao Jiang, Rameen Beroukhim, Jean J. Zhao, and Yonggao Mou
- Subjects
History ,Polymers and Plastics ,Business and International Management ,Industrial and Manufacturing Engineering - Published
- 2022
3. Activation of Sarm1 Produces cADPR to Increase Intra-Axonal Calcium and Promote Axon Degeneration in CIPN
- Author
-
Daina Avizonis, Jian Zhu, Jeffrey Milbrandt, Yihang Li, Maria F. Pazyra-Murphy, sophia tang, Johann Bergholz, Mariana De Sa Tavares Russo, Kwang Woo Ko, Aaron DiAntonio, Tao Jiang, Rosalind A. Segal, and Jean J. Zhao
- Subjects
business.industry ,chemistry.chemical_element ,Degeneration (medical) ,Calcium ,Somatosensory system ,medicine.disease ,chemistry.chemical_compound ,Peripheral neuropathy ,Allodynia ,Paclitaxel ,chemistry ,Calcium flux ,medicine ,medicine.symptom ,business ,Neuroscience ,Intracellular - Abstract
Cancer patients frequently develop chemotherapy-induced peripheral neuropathy (CIPN), a painful and long-lasting disorder with profound somatosensory deficits. There are no effective therapies to prevent or treat this disorder. Pathologically, CIPN is characterized by a “dying-back” axonopathy that begins at intra-epidermal nerve terminals of sensory neurons and progresses in a retrograde fashion. Calcium dysregulation constitutes a critical event in CIPN, but it is not known how chemotherapies such as paclitaxel alter intra-axonal calcium and cause degeneration. Here, we demonstrate that paclitaxel triggers Sarm1-dependent cADPR production in distal axons, promoting intra-axonal calcium flux from both intracellular and extracellular calcium stores. Genetic or pharmacologic antagonists of cADPR signaling prevent paclitaxel-induced axon degeneration and allodynia symptoms, without mitigating the anti-neoplastic efficacy of paclitaxel. Our data demonstrate that cADPR is a calcium modulating factor that promotes paclitaxel-induced axon degeneration and suggest that targeting cADPR signaling provides a potential therapeutic approach for treating CIPN.
- Published
- 2021
4. Combined inhibition of PI3K and PARP is effective in the treatment of ovarian cancer cells with wild-type PIK3CA genes
- Author
-
Dong Wang, Pixu Liu, Hailing Cheng, Ting Li, Min Wang, Lin Xiang, Chengbo Li, Jean J. Zhao, Thomas M. Roberts, Yuan Zhang, and Nan Jiang
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,endocrine system diseases ,Class I Phosphatidylinositol 3-Kinases ,Morpholines ,Genes, BRCA2 ,Cell ,Genes, BRCA1 ,Aminopyridines ,Poly(ADP-ribose) Polymerase Inhibitors ,Poly (ADP-Ribose) Polymerase Inhibitor ,Piperazines ,Article ,Olaparib ,Phosphatidylinositol 3-Kinases ,03 medical and health sciences ,Ovarian tumor ,chemistry.chemical_compound ,Prostate cancer ,0302 clinical medicine ,Cell Line, Tumor ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Viability assay ,Protein Kinase Inhibitors ,Phosphoinositide-3 Kinase Inhibitors ,Ovarian Neoplasms ,business.industry ,Obstetrics and Gynecology ,Drug Synergism ,Middle Aged ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,030220 oncology & carcinogenesis ,PARP inhibitor ,Cancer research ,Phthalazines ,Female ,Phosphatidylinositol 3-Kinase ,Poly(ADP-ribose) Polymerases ,business ,Ovarian cancer - Abstract
Objective Combined inhibition of PI3K and PARP has been shown to be effective in the treatment of preclinical models of breast cancer and prostate cancer independent of BRCA or PIK3CA mutational status. However, the knowledge about this combination treatment in ovarian cancer is limited. The aim of this study was to evaluate the therapeutic effect of PI3K inhibitor BKM120 and PARP inhibitor Olaparib on ovarian cancer cell lines bearing wild-type PIK3CA genes. Methods We exposed three wild-type PIK3CA ovarian cancer cell lines to a PI3K inhibitor BKM120 and/or a PARP inhibitor Olaparib. The effect of BKM120 as a single-agent or in combination with Olaparib was evaluated by Cell Count Kit (CCK8) assay, immunoblotting, comet assay, flow cytometry and immunofluorescence staining assay. The combination indexes for synergistic effect on cell viability were calculated with the Chou-Talalay method. Ex vivo cultured ovarian cancer tissues from patients were analyzed by histological and immunohistochemical analyses. Results Combined inhibition of PI3K and PARP effectively synergized to block the growth of three wild-type PIK3CA ovarian cancer cell lines and explants of a primary ovarian tumor specimen. Mechanistically, dual blockade of PI3K and PARP in these ovarian cancer cell lines resulted in substantially attenuated PI3K/AKT/mTOR signaling, impaired DNA damage response and deficient homologous recombination repair, with remarkable BRCA downregulation. Conclusions The combined use of PI3K inhibitor BKM120 and PARP inhibitor Olaparib may be effective in ovarian cancers with a broader spectrum of cancer-associated genetic alterations but not limited to those with mutant PIK3CA or BRCA genes. BRCA downregulation may be a potential biomarker for the effective response to the proposed combination treatment.
- Published
- 2016
5. A cell-based quantitative high-throughput image screening identified novel autophagy modulators
- Author
-
Yan Zhou, Hua Zhong, Steven R. McGreal, Yuan Li, Menghang Xia, Jean J. Zhao, Wen-Xing Ding, and Ruili Huang
- Subjects
0301 basic medicine ,Chlorpromazine ,Recombinant Fusion Proteins ,Green Fluorescent Proteins ,Cell ,Cellular homeostasis ,Antineoplastic Agents ,Pharmacology ,Biology ,Transfection ,Article ,Receptors, Dopamine ,Methylamines ,Mice ,03 medical and health sciences ,Neoplasms ,Lysosome ,Drug Discovery ,Autophagy ,Fluphenazine ,medicine ,Animals ,Humans ,Protein Kinase Inhibitors ,PI3K/AKT/mTOR pathway ,Dose-Response Relationship, Drug ,TOR Serine-Threonine Kinases ,Fibroblasts ,HCT116 Cells ,Chlorpromazine Hydrochloride ,High-Throughput Screening Assays ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,A549 Cells ,Dopamine receptor ,Indans ,Dopamine Antagonists ,Biological Assay ,Signal transduction ,Microtubule-Associated Proteins ,Signal Transduction - Abstract
Macroautophagy is a major cellular degradation pathway for long-lived proteins and cellular organelles to maintain cellular homeostasis. Reduced autophagy has been implicated in neurodegenerative diseases, metabolic syndrome, and tumorigenesis. In contrast, increased autophagy has been shown to protect against tissue injury and aging. Here we employed a cell-based quantitative high-throughput image screening (qHTS) for autophagy modulators using mouse embryonic fibroblasts (MEFs) that are stably expressing GFP-LC3. The library of pharmacologically active compounds (LOPAC) was used to screen for the autophagy modulators in compounds alone or in combination with the lysosome inhibitor chloroquine (CQ). The GFP-LC3 puncta were then quantified to measure autophagic flux. The primary screening revealed 173 compounds with efficacy more than 40%. These compounds were cherry-picked and re-tested at multiple different concentrations using the same assay. A number of novel autophagy inducers, inhibitors, and modulators with dual-effects on autophagy were identified from the cherry-pick screening. Interestingly, we found a group of compounds that induce autophagy are related to dopamine receptors and are commonly used as clinical psychiatric drugs. Among them, indatraline hydrochloride (IND), a dopamine inhibitor, and chlorpromazine hydrochloride (CPZ) and fluphenazine dihydrochloride (FPZ), two dopamine receptor antagonists, were further evaluated. We found that FPZ-induced autophagy through mTOR inhibition but IND and CPZ induced autophagy in an mTOR-independent manner. Our data suggest that image-based autophagic flux qHTS can efficiently identify autophagy inducers and inhibitors.
- Published
- 2016
6. Overcoming Therapeutic Resistance in HER2-Positive Breast Cancers with CDK4/6 Inhibitors
- Author
-
April C. Watt, Sara M. Tolaney, X. Shirley Liu, Lyndsay Harris, Deborah A. Dillon, Ian E. Krop, Jean J. Zhao, Vinay Varadan, Wei Li, Adam C. Palmer, Susanne Ramm, Eric P. Winer, Qi Wang, Piotr Sicinski, Kwok-Kin Wong, David Tuck, Haluk Yuzugullu, and Shom Goel
- Subjects
0301 basic medicine ,Cancer Research ,endocrine system diseases ,Receptor, ErbB-2 ,medicine.medical_treatment ,mTORC1 ,Pharmacology ,environment and public health ,Targeted therapy ,Mice ,0302 clinical medicine ,Phosphorylation ,skin and connective tissue diseases ,integumentary system ,biology ,Kinase ,TOR Serine-Threonine Kinases ,3. Good health ,ErbB Receptors ,Oncology ,030220 oncology & carcinogenesis ,Female ,biological phenomena, cell phenomena, and immunity ,CDK4/6 Inhibition ,endocrine system ,Mice, Nude ,Breast Neoplasms ,Mice, Transgenic ,P70-S6 Kinase 1 ,Mechanistic Target of Rapamycin Complex 1 ,Article ,03 medical and health sciences ,Cyclin D1 ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Protein Kinase Inhibitors ,neoplasms ,Cyclin-dependent kinase 4 ,Tumor Suppressor Proteins ,Cyclin-Dependent Kinase 4 ,Cyclin-Dependent Kinase 6 ,Cell Biology ,Disease Models, Animal ,enzymes and coenzymes (carbohydrates) ,030104 developmental biology ,Drug Resistance, Neoplasm ,Multiprotein Complexes ,biology.protein ,Cyclin-dependent kinase 6 ,Neoplasm Recurrence, Local - Abstract
Using transgenic mouse models, cell line-based functional studies, and clinical specimens, we show that cyclin D1/CDK4 mediate resistance to targeted therapy for HER2-positive breast cancer. This is overcome using CDK4/6 inhibitors. Inhibition of CDK4/6 not only suppresses Rb phosphorylation, but also reduces TSC2 phosphorylation and thus partially attenuates mTORC1 activity. This relieves feedback inhibition of upstream EGFR family kinases, resensitizing tumors to EGFR/HER2 blockade. Consequently, dual inhibition of EGFR/HER2 and CDK4/6 invokes a more potent suppression of TSC2 phosphorylation and hence mTORC1/S6K/S6RP activity. The suppression of both Rb and S6RP enhances G1 arrest and a phenotype resembling cellular senescence. In vivo, CDK4/6 inhibitors sensitize patient-derived xenograft tumors to HER2-targeted therapies and delay tumor recurrence in a transgenic model of HER2-positive breast cancer.
- Published
- 2016
7. Allele-Specific Chromatin Recruitment and Therapeutic Vulnerabilities of ESR1 Activating Mutations
- Author
-
Nikhil Wagle, Weihan Liu, Ariel Feiglin, Myles Brown, Kayley Abell-Hart, Rinath Jeselsohn, René Houtman, Matthew Pun, MacIntosh Cornwell, Jean J. Zhao, Johann Bergholz, Wei Li, Tengfei Xiao, Eric P. Winer, Henry W. Long, Gilles Buchwalter, Tinghu Zhang, Nathanael S. Gray, Nicholas Kwiatkowski, X. Shirley Liu, Agostina Nardone, Prakash Rao, Teng Fei, and Ofir Cohen
- Subjects
Cistrome ,Cancer research ,Wild type ,Estrogen receptor ,FOXA1 ,Biology ,Estrogen receptor alpha ,Gene ,Phenotype ,Chromatin - Abstract
Estrogen receptor (ER) ligand-binding domain mutations are found in up to 40% of patients with endocrine therapy resistant metastatic ER-positive (ER ) breast cancer. We investigated the chromatin recruitment, transcriptional network and genetic vulnerabilities in breast cancer models harboring the most clinically relevant ER mutations. These ER mutations exhibit both ligand-independent functions that mimic estradiol bound wild type ER as well as allele-specific neomorphic properties that promote a pro-metastatic phenotype. The transcriptomes of a large set of ER metastatic biopsies validated the mutant allele-specific programs identified in the models. The mutant-selective ER cistrome is FOXA1 independent and mediates the allele-specific transcriptional program. Genome-wide CRISPR knockout screens identified genes that are essential for the ligand independent growth driven by the ER mutants including mutantselective synthetic vulnerabilities. These studies provide new insights into the mechanism of endocrine therapy resistance engendered by ER ligand binding domain mutations including clinically relevant allele-specific differences and potential new therapeutic targets.
- Published
- 2018
8. PARP Inhibition Elicits STING-Dependent Antitumor Immunity in Brca1-Deficient Ovarian Cancer
- Author
-
Jean J. Zhao, Tao Jiang, Elizabeth H. Stover, Suzan Lazo, Liya Ding, Thomas M. Roberts, Roderick T. Bronson, Joyce F. Liu, Hye-Jung Kim, Brooke E. Howitt, Ben Li, Ursula A. Matulonis, Gordon J. Freeman, Michael Kearns, Carolynn E. Ohlson, Panagiotis A. Konstantinopoulos, Shaozhen Xie, and Qiwei Wang
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Programmed Cell Death 1 Receptor ,Poly(ADP-ribose) Polymerase Inhibitors ,Article ,Piperazines ,General Biochemistry, Genetics and Molecular Biology ,Targeted therapy ,Olaparib ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Immune system ,Animals ,Humans ,Medicine ,lcsh:QH301-705.5 ,Ovarian Neoplasms ,Tumor microenvironment ,Innate immune system ,BRCA1 Protein ,business.industry ,Immunity ,Membrane Proteins ,Cancer ,Dendritic Cells ,Immunotherapy ,medicine.disease ,3. Good health ,Mice, Inbred C57BL ,HEK293 Cells ,Treatment Outcome ,030104 developmental biology ,lcsh:Biology (General) ,chemistry ,030220 oncology & carcinogenesis ,Cancer research ,Phthalazines ,Female ,business ,Ovarian cancer ,T-Lymphocytes, Cytotoxic - Abstract
SUMMARY PARP inhibitors have shown promising clinical activities for patients with BRCA mutations and are changing the landscape of ovarian cancer treatment. However, the therapeutic mechanisms of action for PARP inhibition in the interaction of tumors with the tumor microenvironment and the host immune system remain unclear. We find that PARP inhibition by olaparib triggers robust local and systemic antitumor immunity involving both adaptive and innate immune responses through a STING-dependent antitumor immune response in mice bearing Brca1-deficient ovarian tumors. This effect is further augmented when olaparib is combined with PD-1 blockade. Our findings thus provide a molecular mechanism underlying antitumor activity by PARP inhibition and lay a foundation to improve therapeutic outcome for cancer patients., In Brief Ding et al. show that PARP inhibition in Brca1-deficient tumors elicits strong antitumor immunity involving activation of both innate and adaptive immune responses, a process that is dependent on STING pathway activation. In addition, they show that addition of PD-1 blockade augments the therapeutic efficacy of PARP inhibitor treatment., Graphical Abstract
- Published
- 2018
9. PI3K Signaling in the Ventromedial Hypothalamic Nucleus Is Required for Normal Energy Homeostasis
- Author
-
Ki Woo Kim, Jeffrey M. Zigman, Jennifer W. Hill, Laurent Gautron, Danielle A. Lauzon, Joel K. Elmquist, Charlotte E. Lee, Michelle J. Choi, Jean J. Zhao, Bradford B. Lowell, Jong Woo Sohn, Makoto Fukuda, Yong Xu, and Harveen Dhillon
- Subjects
Leptin ,Male ,medicine.medical_specialty ,Physiology ,Mutant ,HUMDISEASE ,Biology ,MOLNEURO ,Energy homeostasis ,Article ,03 medical and health sciences ,Mice ,Phosphatidylinositol 3-Kinases ,0302 clinical medicine ,Internal medicine ,Appetite Depressants ,medicine ,Animals ,Homeostasis ,Obesity ,Molecular Biology ,PI3K/AKT/mTOR pathway ,030304 developmental biology ,Phosphoinositide-3 Kinase Inhibitors ,Mice, Knockout ,Neurons ,0303 health sciences ,Cell Biology ,Dietary Fats ,Endocrinology ,Hypothalamus ,Ventromedial Hypothalamic Nucleus ,Signal transduction ,Energy Metabolism ,030217 neurology & neurosurgery ,Intracellular ,Signal Transduction - Abstract
SummaryPhosphatidyl inositol 3-kinase (PI3K) signaling in the hypothalamus has been implicated in the regulation of energy homeostasis, but the critical brain sites where this intracellular signal integrates various metabolic cues to regulate food intake and energy expenditure are unknown. Here, we show that mice with reduced PI3K activity in the ventromedial hypothalamic nucleus (VMH) are more sensitive to high-fat diet-induced obesity due to reduced energy expenditure. In addition, inhibition of PI3K in the VMH impaired the ability to alter energy expenditure in response to acute high-fat diet feeding and food deprivation. Furthermore, the acute anorexigenic effects induced by exogenous leptin were blunted in the mutant mice. Collectively, our results indicate that PI3K activity in VMH neurons plays a physiologically relevant role in the regulation of energy expenditure.
- Published
- 2010
- Full Text
- View/download PDF
10. Specific Roles of the p110α Isoform of Phosphatidylinsositol 3-Kinase in Hepatic Insulin Signaling and Metabolic Regulation
- Author
-
Pixu Liu, C. Ronald Kahn, Mini P. Sajan, Jean J. Zhao, Ryo Suzuki, Thien T. Tran, Victoria Rotter Sopasakis, Jonathon N. Winnay, Graham Smyth, Tatsuya Kondo, Robert Vito Farese, and Tomoichiro Asano
- Subjects
Blood Glucose ,Leptin ,medicine.medical_specialty ,Class I Phosphatidylinositol 3-Kinases ,Physiology ,medicine.medical_treatment ,HUMDISEASE ,Down-Regulation ,Biology ,P110α ,Article ,Impaired glucose tolerance ,Mice ,Phosphatidylinositol 3-Kinases ,03 medical and health sciences ,0302 clinical medicine ,Phosphatidylinositol Phosphates ,Downregulation and upregulation ,Internal medicine ,Glucose Intolerance ,Diabetes Mellitus ,medicine ,Animals ,Insulin ,Molecular Biology ,Protein kinase B ,PI3K/AKT/mTOR pathway ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,Cell Biology ,medicine.disease ,Lipids ,Metformin ,Insulin receptor ,Endocrinology ,Liver ,SIGNALING ,030220 oncology & carcinogenesis ,biology.protein ,Signal transduction ,Energy Metabolism ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
The class I(A) phosphatidylinsositol 3-kinases (PI3Ks) form a critical node in the insulin metabolic pathway; however, the precise roles of the different isoforms of this enzyme remain elusive. Using tissue-specific gene inactivation, we demonstrate that p110alpha catalytic subunit of PI3K is a key mediator of insulin metabolic actions in the liver. Thus, deletion of p110alpha in liver results in markedly blunted insulin signaling with decreased generation of PIP(3) and loss of insulin activation of Akt, defects that could not be rescued by overexpression of p110beta. As a result, mice with hepatic knockout of p110alpha display reduced insulin sensitivity, impaired glucose tolerance, and increased gluconeogenesis, hypolipidemia, and hyperleptinemia. The diabetic syndrome induced by loss of p110alpha in liver did not respond to metformin treatment. Together, these data indicate that the p110alpha isoform of PI3K plays a fundamental role in insulin signaling and control of hepatic glucose and lipid metabolism.
- Published
- 2010
11. Allele-Specific Chromatin Recruitment and Therapeutic Vulnerabilities of ESR1 Activating Mutations
- Author
-
Ariel Feiglin, X. Shirley Liu, Tengfei Xiao, Xintao Qiu, MacIntosh Cornwell, Jean J. Zhao, Henry W. Long, Eric P. Winer, Nicholas Kwiatkowski, Prakash Rao, Teng Fei, Matthew Pun, Wei Li, Gilles Buchwalter, Agostina Nardone, Johann Bergholz, Tinghu Zhang, Rinath Jeselsohn, Weihan Liu, Kayley Abell-Hart, Nathanael S. Gray, Ofir Cohen, Nikhil Wagle, René Houtman, Myles Brown, and Diane Melchers
- Subjects
0301 basic medicine ,Cancer Research ,Antineoplastic Agents, Hormonal ,Estrogen receptor ,Breast Neoplasms ,Mice, Transgenic ,Biology ,medicine.disease_cause ,Article ,03 medical and health sciences ,Cell Line, Tumor ,Drug Discovery ,medicine ,Animals ,Humans ,Gene ,Alleles ,Mutation ,Estrogen Receptor alpha ,Cell Biology ,Phenotype ,Chromatin ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Oncology ,Cistrome ,Drug Resistance, Neoplasm ,Cancer research ,Estrogen receptor alpha ,Genetic screen - Abstract
Estrogen receptor α (ER) ligand-binding domain (LBD) mutations are found in a substantial number of endocrine treatment-resistant metastatic ER-positive (ER+) breast cancers. We investigated the chromatin recruitment, transcriptional network, and genetic vulnerabilities in breast cancer models harboring the clinically relevant ER mutations. These mutants exhibit both ligand-independent functions that mimic estradiol-bound wild-type ER as well as allele-specific neomorphic properties that promote a pro-metastatic phenotype. Analysis of the genome-wide ER binding sites identified mutant ER unique recruitment mediating the allele-specific transcriptional program. Genetic screens identified genes that are essential for the ligand-independent growth driven by the mutants. These studies provide insights into the mechanism of endocrine therapy resistance engendered by ER mutations and potential therapeutic targets.
- Published
- 2018
12. Should individual PI3 kinase isoforms be targeted in cancer?
- Author
-
Jean J. Zhao, Shidong Jia, and Thomas M. Roberts
- Subjects
Neovascularization, Physiologic ,Antineoplastic Agents ,P110α ,Biology ,medicine.disease_cause ,Article ,Receptor tyrosine kinase ,Phosphatidylinositol 3-Kinases ,Neoplasms ,medicine ,Animals ,Humans ,PTEN ,Protein Kinase Inhibitors ,PI3K/AKT/mTOR pathway ,G protein-coupled receptor ,Neovascularization, Pathologic ,Kinase ,PTEN Phosphohydrolase ,Receptor Protein-Tyrosine Kinases ,Cell Biology ,Cell biology ,Enzyme Activation ,Isoenzymes ,Mutation ,ras Proteins ,Cancer research ,biology.protein ,Intercellular Signaling Peptides and Proteins ,Signal transduction ,Carcinogenesis ,Signal Transduction - Abstract
Activation of the phosphoinositide-3-kinase (PI3K) signaling pathway is frequently found in common human cancers, brought about by oncogenic receptor tyrosine kinases (RTKs) acting upstream, PTEN loss, or activating mutations of PI3K itself. Recent studies have delineated distinct but overlapping functions in cell signaling and tumorigenesis for p110alpha and p110beta, the two major catalytic subunits of PI3K expressed in the tissues of origin for the common tumor types. In most cell types studied, p110alpha carries the majority of the PI3K signal in classic RTK signal transduction, while p110beta responds to GPCRs. Both p110alpha and p110beta function in cellular transformation induced by alterations in components of PI3K pathway. Specifically, p110alpha is essential for the signaling and growth of tumors driven by PIK3CA mutations and/or oncogenic RTKs/Ras, whereas p110beta is the major isoform in mediating PTEN-deficient tumorigenesis. While pan-PI3K inhibitors are currently being tested in the clinic, p110 isoform-specific inhibition holds promise as a therapeutic strategy.
- Published
- 2009
13. Integrative Genomic Approaches Identify IKBKE as a Breast Cancer Oncogene
- Author
-
Marc Vidal, Greg Hinkle, Andrea L. Richardson, Thomas M. Roberts, Lauren Ambrogio, Sarah K. Sjostrom, Matthew Meyerson, Jesse S. Boehm, Levi A. Garraway, Heidi Greulich, William C. Hahn, Laura Mulvey, Rhine R. Shen, Jun Yao, So Young Kim, Kornelia Polyak, David E. Hill, Jennifer K. Grenier, Eric S. Lander, Tomoko Hirozane-Kishikawa, Jean J. Zhao, Ian F. Dunn, Carly J. Stewart, David E. Root, Ron Firestein, and Stanislawa Weremowicz
- Subjects
MAPK/ERK pathway ,Breast Neoplasms ,Biology ,Models, Biological ,General Biochemistry, Genetics and Molecular Biology ,Malignant transformation ,Cell Line ,Phosphatidylinositol 3-Kinases ,Breast cancer ,medicine ,IKBKE ,Humans ,Extracellular Signal-Regulated MAP Kinases ,PI3K/AKT/mTOR pathway ,Alleles ,Gene Library ,Genome ,Oncogene ,Kinase ,Biochemistry, Genetics and Molecular Biology(all) ,NF-kappa B ,Genomics ,medicine.disease ,I-kappa B Kinase ,Gene Expression Regulation, Neoplastic ,Cell Transformation, Neoplastic ,Cancer research ,Signal transduction ,Signal Transduction - Abstract
The karyotypic chaos exhibited by human epithelial cancers complicates efforts to identify mutations critical for malignant transformation. Here we integrate complementary genomic approaches to identify human oncogenes. We show that activation of the ERK and phosphatidylinositol 3-kinase (PI3K) signaling pathways cooperate to transform human cells. Using a library of activated kinases, we identify several kinases that replace PI3K signaling and render cells tumorigenic. Whole genome structural analyses reveal that one of these kinases, IKBKE (IKKepsilon), is amplified and overexpressed in breast cancer cell lines and patient-derived tumors. Suppression of IKKepsilon expression in breast cancer cell lines that harbor IKBKE amplifications induces cell death. IKKepsilon activates the nuclear factor-kappaB (NF-kappaB) pathway in both cell lines and breast cancers. These observations suggest a mechanism for NF-kappaB activation in breast cancer, implicate the NF-kappaB pathway as a downstream mediator of PI3K, and provide a framework for integrated genomic approaches in oncogene discovery.
- Published
- 2007
- Full Text
- View/download PDF
14. A Genetic Screen for Candidate Tumor Suppressors Identifies REST
- Author
-
Thomas F. Westbrook, Anthony C. Liang, Eric S. Martin, Stephen J. Elledge, Jean J. Zhao, Thomas M. Roberts, Ronald A. DePinho, Gail Mandel, Yumei Leng, Gregory J. Hannon, Michael R. Schlabach, Bin Feng, and Lynda Chin
- Subjects
Candidate gene ,RE1-silencing transcription factor ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Malignant transformation ,Frameshift mutation ,Phosphatidylinositol 3-Kinases ,03 medical and health sciences ,0302 clinical medicine ,Transforming Growth Factor beta ,Cell Line, Tumor ,medicine ,Humans ,PTEN ,Genes, Tumor Suppressor ,Genetic Testing ,Epigenetics ,Cells, Cultured ,Phosphoinositide-3 Kinase Inhibitors ,030304 developmental biology ,Genetics ,0303 health sciences ,biology ,Biochemistry, Genetics and Molecular Biology(all) ,Epithelial Cells ,3. Good health ,Repressor Proteins ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,RNA Interference ,Carcinogenesis ,Signal Transduction ,Transcription Factors ,Genetic screen - Abstract
SummaryTumorigenesis is a multistep process characterized by a myriad of genetic and epigenetic alterations. Identifying the causal perturbations that confer malignant transformation is a central goal in cancer biology. Here we report an RNAi-based genetic screen for genes that suppress transformation of human mammary epithelial cells. We identified genes previously implicated in proliferative control and epithelial cell function including two established tumor suppressors, TGFBR2 and PTEN. In addition, we uncovered a previously unrecognized tumor suppressor role for REST/NRSF, a transcriptional repressor of neuronal gene expression. Array-CGH analysis identified REST as a frequent target of deletion in colorectal cancer. Furthermore, we detect a frameshift mutation of the REST gene in colorectal cancer cells that encodes a dominantly acting truncation capable of transforming epithelial cells. Cells lacking REST exhibit increased PI(3)K signaling and are dependent upon this pathway for their transformed phenotype. These results implicate REST as a human tumor suppressor and provide a novel approach to identifying candidate genes that suppress the development of human cancer.
- Published
- 2005
15. Functional genetics and experimental models of human cancer
- Author
-
Thomas M. Roberts, William C. Hahn, and Jean J. Zhao
- Subjects
Genetics ,Telomerase ,Cancer ,Biology ,Human cell ,medicine.disease ,Models, Biological ,Malignant transformation ,Cancer pathogenesis ,Cell Transformation, Neoplastic ,Cell culture ,Neoplasms ,Mutation ,medicine ,Animals ,Humans ,Molecular Medicine ,Molecular Biology ,Gene ,Human cancer - Abstract
Abundant evidence supports the hypothesis that cancer arises from normal cells through the stepwise accumulation of genetic mutations. The study of cells obtained from patients with cancer has identified numerous molecules and pathways that fundamentally contribute to malignant transformation; however, cancer cell lines are often difficult to isolate or maintain, and the cell lines that are available for experimentation represent only a small subset of late-stage human cancers. Recent work has elucidated the role of telomerase in regulating human cell lifespan and has enabled the development of new experimental systems to study human cancer. This review highlights the recent progress in combining genetic methods and primary human cells to understand the role of specific genes and pathways in cancer pathogenesis.
- Published
- 2004
16. A chemical genetic screen identifies inhibitors of regulated nuclear export of a Forkhead transcription factor in PTEN-deficient tumor cells
- Author
-
Tweeny R Kau, Jean J. Zhao, Shivapriya Ramaswamy, Pamela A. Silver, Cheryl L Wojciechowski, Frank C. Schroeder, William R. Sellers, Jon Clardy, and Thomas M. Roberts
- Subjects
Models, Molecular ,Cancer Research ,Active Transport, Cell Nucleus ,Receptors, Cytoplasmic and Nuclear ,FOXO1 ,Karyopherins ,03 medical and health sciences ,Phosphatidylinositol 3-Kinases ,0302 clinical medicine ,Calmodulin ,Tumor Cells, Cultured ,PTEN ,Animals ,Humans ,Benzothiazoles ,Enzyme Inhibitors ,Phosphorylation ,Nuclear export signal ,Protein kinase B ,Transcription factor ,PI3K/AKT/mTOR pathway ,030304 developmental biology ,Phosphoinositide-3 Kinase Inhibitors ,Cell Nucleus ,0303 health sciences ,biology ,Forkhead Box Protein O1 ,Forkhead Transcription Factors ,Cell Biology ,Phosphoric Monoester Hydrolases ,DNA-Binding Proteins ,Gene Expression Regulation, Neoplastic ,Oncology ,030220 oncology & carcinogenesis ,Drug Design ,FOXO4 ,biology.protein ,Cancer research ,Benzimidazoles ,Genetic screen ,Signal Transduction ,Transcription Factors - Abstract
The PI3K/PTEN/Akt signal transduction pathway plays a key role in many tumors. Downstream targets of this pathway include the Forkhead family of transcription factors (FOXO1a, FOXO3a, FOXO4). In PTEN null cells, FOXO1a is inactivated by PI3K-dependent phosphorylation and mislocalization to the cytoplasm, yet still undergoes nucleocytoplasmic shuttling. Since forcible localization of FOXO1a to the nucleus can reverse tumorigenicity of PTEN null cells, a high-content, chemical genetic screen for inhibitors of FOXO1a nuclear export was performed. The compounds detected in the primary screen were retested in secondary assays, and structure-function relationships were identified. Novel general export inhibitors were found that react with CRM1 as well as a number of compounds that inhibit PI3K/Akt signaling, among which are included multiple antagonists of calmodulin signaling.
- Published
- 2003
- Full Text
- View/download PDF
17. Human mammary epithelial cell transformation through the activation of phosphatidylinositol 3-kinase
- Author
-
Jean J. Zhao, Thomas M. Roberts, Yuan Cheng, Wen Chen, William C. Hahn, Romesh R. Subramanian, and Ole Gjoerup
- Subjects
rac1 GTP-Binding Protein ,Cancer Research ,DNA, Complementary ,Time Factors ,SV40 large T antigen ,Cell division ,Genetic Vectors ,Immunoblotting ,AKT1 ,RAC1 ,Protein Serine-Threonine Kinases ,Biology ,Transfection ,Proto-Oncogene Proteins c-myc ,Phosphatidylinositol 3-Kinases ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Proto-Oncogene Proteins ,Humans ,Phosphatidylinositol ,Phosphorylation ,Mammary Glands, Human ,PI3K/AKT/mTOR pathway ,030304 developmental biology ,0303 health sciences ,Dose-Response Relationship, Drug ,Kinase ,Cell Biology ,Fibroblasts ,Precipitin Tests ,Cell biology ,Enzyme Activation ,Agar ,Alternative Splicing ,Cell Transformation, Neoplastic ,Retroviridae ,Oncology ,chemistry ,Biochemistry ,030220 oncology & carcinogenesis ,Proto-Oncogene Proteins c-akt ,Cell Division - Abstract
Recent studies have demonstrated that introduction of hTERT in combination with SV40 large T antigen (LT), small t antigen (st), and H-rasV12 suffices to transform many primary human cells. In human mammary epithelial cells (HMECs) expressing elevated c-Myc, activated H-Ras is dispensable for anchorage-independent growth. Using this system, we show that st activates the PI3K pathway and that constitutive PI3K signaling substitutes for st in transformation. Moreover, using constitutively active versions of Akt1 and Rac1, we show that these downstream pathways of PI3K synergize to achieve anchorage-independent growth. At lower levels of c-myc expression, activated PI3K also replaces st to complement H-rasV12 and LT and confers both soft agar growth and tumorigenicity. However, elevated c-myc expression cannot replace H-rasV12 for tumorigenesis. These observations begin to define the pathways perturbed during the transformation of HMECs.
- Published
- 2003
18. Cell-Cycle-Targeting MicroRNAs as Therapeutic Tools against Refractory Cancers
- Author
-
Jan M. Suski, Roman L. Bogorad, Per Hydbring, Kimmie Ng, Daniel G. Anderson, Jean J. Zhao, Kevin J. Kauffman, Hao Yin, Yoon Jong Choi, Kornelia Polyak, Ewa Sicinska, Lars Anders, Haluk Yuzugullu, Anne Fassl, Katharine E. Sweeney, Davide Floris, Xiaoting Li, Richard Swanson, Veronica Matia, Piotr Sicinski, Junghoon Yang, Yinan Wang, Yingxiang Li, Shom Goel, Tobias Otto, Chong Jin, Cheng Li, Massachusetts Institute of Technology. Department of Chemical Engineering, Massachusetts Institute of Technology. Institute for Medical Engineering & Science, Harvard University--MIT Division of Health Sciences and Technology, and Koch Institute for Integrative Cancer Research at MIT
- Subjects
0301 basic medicine ,Cancer Research ,biology ,Kinase ,Cell cycle ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Cyclin-dependent kinase ,Cell culture ,Tumor progression ,030220 oncology & carcinogenesis ,microRNA ,Systemic administration ,biology.protein ,Cyclin - Abstract
Cyclins and cyclin-dependent kinases (CDKs) are hyperactivated in numerous human tumors. To identify means of interfering with cyclins/CDKs, we performed nine genome-wide screens for human microRNAs (miRNAs) directly regulating cell-cycle proteins. We uncovered a distinct class of miRNAs that target nearly all cyclins/CDKs, which are very effective in inhibiting cancer cell proliferation. By profiling the response of over 120 human cancer cell lines, we derived an expression-based algorithm that can predict the response of tumors to cell-cycle-targeting miRNAs. Using systemic administration of nanoparticle-formulated miRNAs, we inhibited tumor progression in seven mouse xenograft models, including three treatment-refractory patient-derived tumors, without affecting normal tissues. Our results highlight the utility of using cell-cycle-targeting miRNAs for treatment of refractory cancer types. Keywords: cell cycle; cyclins; cyclin-dependent kinases; microRNAs; cancers
- Published
- 2017
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.