32 results on '"Spring DJ"'
Search Results
2. Inhibition of Toll-like receptor 4 with eritoran attenuates myocardial ischemia-reperfusion injury.
- Author
-
Shimamoto A, Chong AJ, Yada M, Shomura S, Takayama H, Fleisig AJ, Agnew ML, Hampton CR, Rothnie CL, Spring DJ, Pohlman TH, Shimpo H, and Verrier ED
- Published
- 2006
3. Telomere dysfunction alters intestinal stem cell dynamics to promote cancer.
- Author
-
LaBella KA, Hsu WH, Li J, Qi Y, Liu Y, Liu J, Wu CC, Liu Y, Song Z, Lin Y, Blecher JM, Jiang S, Shang X, Han J, Spring DJ, Zhang J, Xia Y, and DePinho RA
- Subjects
- Animals, Mice, Enhancer of Zeste Homolog 2 Protein metabolism, Enhancer of Zeste Homolog 2 Protein genetics, Adenoma pathology, Adenoma genetics, Adenoma metabolism, Intestines pathology, Cell Differentiation, Humans, Glycogen Synthase Kinase 3 beta metabolism, Glycogen Synthase Kinase 3 beta genetics, DNA Damage, Mice, Inbred C57BL, Wnt Signaling Pathway, Telomere metabolism, Adenomatous Polyposis Coli Protein genetics, Adenomatous Polyposis Coli Protein metabolism, Stem Cells metabolism, Stem Cells pathology
- Abstract
Telomere dynamics are linked to aging hallmarks, and age-associated telomere loss fuels the development of epithelial cancers. In Apc-mutant mice, the onset of DNA damage associated with telomere dysfunction has been shown to accelerate adenoma initiation via unknown mechanisms. Here, we observed that Apc-mutant mice engineered to experience telomere dysfunction show accelerated adenoma formation resulting from augmented cell competition and clonal expansion. Mechanistically, telomere dysfunction induces the repression of EZH2, resulting in the derepression of Wnt antagonists, which causes the differentiation of adjacent stem cells and a relative growth advantage to Apc-deficient telomere dysfunctional cells. Correspondingly, in this mouse model, GSK3β inhibition countered the actions of Wnt antagonists on intestinal stem cells, resulting in impaired adenoma formation of telomere dysfunctional Apc-mutant cells. Thus, telomere dysfunction contributes to cancer initiation through altered stem cell dynamics, identifying an interception strategy for human APC-mutant cancers with shortened telomeres., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
4. Oncogenic KRAS Drives Lipofibrogenesis to Promote Angiogenesis and Colon Cancer Progression.
- Author
-
Hsu WH, LaBella KA, Lin Y, Xu P, Lee R, Hsieh CE, Yang L, Zhou A, Blecher JM, Wu CJ, Lin K, Shang X, Jiang S, Spring DJ, Xia Y, Chen P, Shen JP, Kopetz S, and DePinho RA
- Subjects
- Animals, Humans, Mice, Angiogenesis, Colorectal Neoplasms metabolism, DNA-Binding Proteins metabolism, Fibroblasts metabolism, Lipids, Transcription Factors metabolism, Tumor Microenvironment genetics, Cancer-Associated Fibroblasts metabolism, Colonic Neoplasms genetics, Colonic Neoplasms pathology, Proto-Oncogene Proteins p21(ras) genetics, Proto-Oncogene Proteins p21(ras) metabolism
- Abstract
Oncogenic KRAS (KRAS*) contributes to many cancer hallmarks. In colorectal cancer, KRAS* suppresses antitumor immunity to promote tumor invasion and metastasis. Here, we uncovered that KRAS* transforms the phenotype of carcinoma-associated fibroblasts (CAF) into lipid-laden CAFs, promoting angiogenesis and tumor progression. Mechanistically, KRAS* activates the transcription factor CP2 (TFCP2) that upregulates the expression of the proadipogenic factors BMP4 and WNT5B, triggering the transformation of CAFs into lipid-rich CAFs. These lipid-rich CAFs, in turn, produce VEGFA to spur angiogenesis. In KRAS*-driven colorectal cancer mouse models, genetic or pharmacologic neutralization of TFCP2 reduced lipid-rich CAFs, lessened tumor angiogenesis, and improved overall survival. Correspondingly, in human colorectal cancer, lipid-rich CAF and TFCP2 signatures correlate with worse prognosis. This work unveils a new role for KRAS* in transforming CAFs, driving tumor angiogenesis and disease progression, providing an actionable therapeutic intervention for KRAS*-driven colorectal cancer., Significance: This study identified a molecular mechanism contributing to KRAS*-driven colorectal cancer progression via fibroblast transformation in the tumor microenvironment to produce VEGFA driving tumor angiogenesis. In preclinical models, targeting the KRAS*-TFCP2-VEGFA axis impaired tumor progression, revealing a potential novel therapeutic option for patients with KRAS*-driven colorectal cancer. This article is featured in Selected Articles from This Issue, p. 2489., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
5. Stromal-derived NRG1 enables oncogenic KRAS bypass in pancreas cancer.
- Author
-
Han J, Xu J, Liu Y, Liang S, LaBella KA, Chakravarti D, Spring DJ, Xia Y, and DePinho RA
- Subjects
- Humans, Animals, Mice, Proto-Oncogene Proteins p21(ras) genetics, Proto-Oncogene Proteins p21(ras) metabolism, Cell Proliferation, Neuregulin-1 genetics, Neuregulin-1 metabolism, Pancreatic Neoplasms metabolism, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal pathology
- Abstract
Activating KRAS mutations (KRAS*) in pancreatic ductal adenocarcinoma (PDAC) drive anabolic metabolism and support tumor maintenance. KRAS* inhibitors show initial antitumor activity followed by recurrence due to cancer cell-intrinsic and immune-mediated paracrine mechanisms. Here, we explored the potential role of cancer-associated fibroblasts (CAFs) in enabling KRAS* bypass and identified CAF-derived NRG1 activation of cancer cell ERBB2 and ERBB3 receptor tyrosine kinases as a mechanism by which KRAS*-independent growth is supported. Genetic extinction or pharmacological inhibition of KRAS* resulted in up-regulation of ERBB2 and ERBB3 expression in human and murine models, which prompted cancer cell utilization of CAF-derived NRG1 as a survival factor. Genetic depletion or pharmacological inhibition of ERBB2/3 or NRG1 abolished KRAS* bypass and synergized with KRAS
G12D inhibitors in combination treatments in mouse and human PDAC models. Thus, we found that CAFs can contribute to KRAS* inhibitor therapy resistance via paracrine mechanisms, providing an actionable therapeutic strategy to improve the effectiveness of KRAS* inhibitors in PDAC patients., (© 2023 Han et al.; Published by Cold Spring Harbor Laboratory Press.)- Published
- 2023
- Full Text
- View/download PDF
6. Histone demethylase KDM5D upregulation drives sex differences in colon cancer.
- Author
-
Li J, Lan Z, Liao W, Horner JW, Xu X, Liu J, Yoshihama Y, Jiang S, Shim HS, Slotnik M, LaBella KA, Wu CJ, Dunner K Jr, Hsu WH, Lee R, Khanduri I, Terranova C, Akdemir K, Chakravarti D, Shang X, Spring DJ, Wang YA, and DePinho RA
- Subjects
- Animals, Female, Humans, Male, Mice, CD8-Positive T-Lymphocytes immunology, Disease Models, Animal, Mice, Transgenic, Up-Regulation, Colorectal Neoplasms genetics, Colorectal Neoplasms metabolism, Colorectal Neoplasms pathology, Histone Demethylases genetics, Histone Demethylases metabolism, Minor Histocompatibility Antigens genetics, Minor Histocompatibility Antigens metabolism, Sex Characteristics
- Abstract
Sex exerts a profound impact on cancer incidence, spectrum and outcomes, yet the molecular and genetic bases of such sex differences are ill-defined and presumptively ascribed to X-chromosome genes and sex hormones
1 . Such sex differences are particularly prominent in colorectal cancer (CRC) in which men experience higher metastases and mortality. A murine CRC model, engineered with an inducible transgene encoding oncogenic mutant KRASG12D and conditional null alleles of Apc and Trp53 tumour suppressors (designated iKAP)2 , revealed higher metastases and worse outcomes specifically in males with oncogenic mutant KRAS (KRAS*) CRC. Integrated cross-species molecular and transcriptomic analyses identified Y-chromosome gene histone demethylase KDM5D as a transcriptionally upregulated gene driven by KRAS*-mediated activation of the STAT4 transcription factor. KDM5D-dependent chromatin mark and transcriptome changes showed repression of regulators of the epithelial cell tight junction and major histocompatibility complex class I complex components. Deletion of Kdm5d in iKAP cancer cells increased tight junction integrity, decreased cell invasiveness and enhanced cancer cell killing by CD8+ T cells. Conversely, iAP mice engineered with a Kdm5d transgene to provide constitutive Kdm5d expression specifically in iAP cancer cells showed an increased propensity for more invasive tumours in vivo. Thus, KRAS*-STAT4-mediated upregulation of Y chromosome KDM5D contributes substantially to the sex differences in KRAS* CRC by means of its disruption of cancer cell adhesion properties and tumour immunity, providing an actionable therapeutic strategy for metastasis risk reduction for men afflicted with KRAS* CRC., (© 2023. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2023
- Full Text
- View/download PDF
7. Targeting T cell checkpoints 41BB and LAG3 and myeloid cell CXCR1/CXCR2 results in antitumor immunity and durable response in pancreatic cancer.
- Author
-
Gulhati P, Schalck A, Jiang S, Shang X, Wu CJ, Hou P, Ruiz SH, Soto LS, Parra E, Ying H, Han J, Dey P, Li J, Deng P, Sei E, Maeda DY, Zebala JA, Spring DJ, Kim M, Wang H, Maitra A, Moore D, Clise-Dwyer K, Wang YA, Navin NE, and DePinho RA
- Subjects
- Humans, Myeloid Cells pathology, T-Lymphocyte Subsets metabolism, T-Lymphocyte Subsets pathology, Receptors, Interleukin-8A immunology, Pancreatic Neoplasms, Carcinoma, Pancreatic Ductal drug therapy, Pancreatic Neoplasms drug therapy
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) is considered non-immunogenic, with trials showing its recalcitrance to PD1 and CTLA4 immune checkpoint therapies (ICTs). Here, we sought to systematically characterize the mechanisms underlying de novo ICT resistance and to identify effective therapeutic options for PDAC. We report that agonist 41BB and antagonist LAG3 ICT alone and in combination, increased survival and antitumor immunity, characterized by modulating T cell subsets with antitumor activity, increased T cell clonality and diversification, decreased immunosuppressive myeloid cells and increased antigen presentation/decreased immunosuppressive capability of myeloid cells. Translational analyses confirmed the expression of 41BB and LAG3 in human PDAC. Since single and dual ICTs were not curative, T cell-activating ICTs were combined with a CXCR1/2 inhibitor targeting immunosuppressive myeloid cells. Triple therapy resulted in durable complete responses. Given similar profiles in human PDAC and the availability of these agents for clinical testing, our findings provide a testable hypothesis for this lethal disease., (© 2022. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2023
- Full Text
- View/download PDF
8. Synthetic Essentiality of Tryptophan 2,3-Dioxygenase 2 in APC-Mutated Colorectal Cancer.
- Author
-
Lee R, Li J, Li J, Wu CJ, Jiang S, Hsu WH, Chakravarti D, Chen P, LaBella KA, Li J, Spring DJ, Zhao D, Wang YA, and DePinho RA
- Subjects
- Humans, Tryptophan, Tryptophan Oxygenase metabolism, Tumor Microenvironment, Wnt Signaling Pathway genetics, beta Catenin genetics, beta Catenin metabolism, Adenomatous Polyposis Coli genetics, Adenomatous Polyposis Coli metabolism, Adenomatous Polyposis Coli pathology, Colorectal Neoplasms metabolism, Dioxygenases metabolism
- Abstract
Inactivation of adenomatous polyposis coli (APC) is common across many cancer types and serves as a critical initiating event in most sporadic colorectal cancers. APC deficiency activates WNT signaling, which remains an elusive target for cancer therapy, prompting us to apply the synthetic essentiality framework to identify druggable vulnerabilities for APC-deficient cancers. Tryptophan 2,3-dioxygenase 2 (TDO2) was identified as a synthetic essential effector of APC-deficient colorectal cancer. Mechanistically, APC deficiency results in the TCF4/β-catenin-mediated upregulation of TDO2 gene transcription. TDO2 in turn activates the Kyn-AhR pathway, which increases glycolysis to drive anabolic cancer cell growth and CXCL5 secretion to recruit macrophages into the tumor microenvironment. Therapeutically, APC-deficient colorectal cancer models were susceptible to TDO2 depletion or pharmacologic inhibition, which impaired cancer cell proliferation and enhanced antitumor immune profiles. Thus, APC deficiency activates a TCF4-TDO2-AhR-CXCL5 circuit that affects multiple cancer hallmarks via autonomous and nonautonomous mechanisms and illuminates a genotype-specific vulnerability in colorectal cancer., Significance: This study identifies critical effectors in the maintenance of APC-deficient colorectal cancer and demonstrates the relationship between APC/WNT pathway and kynurenine pathway signaling. It further determines the tumor-associated macrophage biology in APC-deficient colorectal cancer, informing genotype-specific therapeutic targets and the use of TDO2 inhibitors. This article is highlighted in the In This Issue feature, p. 1599., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
9. AR-negative prostate cancer is vulnerable to loss of JMJD1C demethylase.
- Author
-
Yoshihama Y, LaBella KA, Kim E, Bertolet L, Colic M, Li J, Shang X, Wu CJ, Spring DJ, Wang YA, Hart T, and DePinho RA
- Subjects
- Apoptosis drug effects, Cell Line, Tumor, Databases, Genetic, Histone Demethylases metabolism, Humans, Intracellular Signaling Peptides and Proteins genetics, Intracellular Signaling Peptides and Proteins metabolism, Jumonji Domain-Containing Histone Demethylases metabolism, Male, Oxidoreductases, N-Demethylating metabolism, Promoter Regions, Genetic drug effects, Prostate pathology, Protein Serine-Threonine Kinases genetics, Receptors, Androgen genetics, Signal Transduction drug effects, Transcriptional Activation drug effects, Tumor Necrosis Factor-alpha metabolism, Jumonji Domain-Containing Histone Demethylases genetics, Oxidoreductases, N-Demethylating genetics, Prostatic Neoplasms genetics, Receptors, Androgen metabolism
- Abstract
Prostate cancer is a leading cause of cancer-related mortality in men. The widespread use of androgen receptor (AR) inhibitors has generated an increased incidence of AR-negative prostate cancer, triggering the need for effective therapies for such patients. Here, analysis of public genome-wide CRISPR screens in human prostate cancer cell lines identified histone demethylase JMJD1C (KDM3C) as an AR-negative context-specific vulnerability. Secondary validation studies in multiple cell lines and organoids, including isogenic models, confirmed that small hairpin RNA (shRNA)-mediated depletion of JMJD1C potently inhibited growth specifically in AR-negative prostate cancer cells. To explore the cooperative interactions of AR and JMJD1C, we performed comparative transcriptomics of 1) isogenic AR-positive versus AR-negative prostate cancer cells, 2) AR-positive versus AR-negative prostate cancer tumors, and 3) isogenic JMJD1C-expressing versus JMJD1C-depleted AR-negative prostate cancer cells. Loss of AR or JMJD1C generates a modest tumor necrosis factor alpha (TNFα) signature, whereas combined loss of AR and JMJD1C strongly up-regulates the TNFα signature in human prostate cancer, suggesting TNFα signaling as a point of convergence for the combined actions of AR and JMJD1C. Correspondingly, AR-negative prostate cancer cells showed exquisite sensitivity to TNFα treatment and, conversely, TNFα pathway inhibition via inhibition of its downstream effector MAP4K4 partially reversed the growth defect of JMJD1C-depleted AR-negative prostate cancer cells. Given the deleterious systemic side effects of TNFα therapy in humans and the viability of JMJD1C-knockout mice, the identification of JMJD1C inhibition as a specific vulnerability in AR-negative prostate cancer may provide an alternative drug target for prostate cancer patients progressing on AR inhibitor therapy., Competing Interests: Competing interest statement: T.H. is a consultant for Repare Therapeutics. R.A.D. is a cofounder, advisor, and/or director of Tvardi Therapeutics, Asylia Therapeutics, Stellanova Therapeutics, Nirogy Therapeutics, and Sporos Bioventures. The work of this manuscript was not supported by, nor is related to, these companies.
- Published
- 2021
- Full Text
- View/download PDF
10. Telomere dysfunction activates YAP1 to drive tissue inflammation.
- Author
-
Chakravarti D, Hu B, Mao X, Rashid A, Li J, Li J, Liao WT, Whitley EM, Dey P, Hou P, LaBella KA, Chang A, Wang G, Spring DJ, Deng P, Zhao D, Liang X, Lan Z, Lin Y, Sarkar S, Terranova C, Deribe YL, Blutt SE, Okhuysen P, Zhang J, Vilar E, Nielsen OH, Dupont A, Younes M, Patel KR, Shroyer NF, Rai K, Estes MK, Wang YA, Bertuch AA, and DePinho RA
- Subjects
- Adaptor Proteins, Signal Transducing antagonists & inhibitors, Adaptor Proteins, Signal Transducing genetics, Animals, Anti-Bacterial Agents therapeutic use, Ataxia Telangiectasia Mutated Proteins antagonists & inhibitors, Ataxia Telangiectasia Mutated Proteins metabolism, Caspase 1 metabolism, Cell Cycle Proteins antagonists & inhibitors, Cell Cycle Proteins genetics, Child, Colon metabolism, Colon microbiology, Colon pathology, Gastrointestinal Diseases pathology, Gastrointestinal Microbiome drug effects, Gastrointestinal Microbiome physiology, Humans, Inflammation drug therapy, Inflammation metabolism, Inflammation microbiology, Interleukin-18 genetics, Interleukin-18 metabolism, Intestinal Mucosa metabolism, Intestinal Mucosa pathology, Mice, Mice, Mutant Strains, Phosphorylation, Protein Precursors genetics, Protein Precursors metabolism, Signal Transduction, Telomerase genetics, Telomerase metabolism, YAP-Signaling Proteins, Adaptor Proteins, Signal Transducing metabolism, Cell Cycle Proteins metabolism, Inflammation pathology, Telomere pathology
- Abstract
Germline telomere maintenance defects are associated with an increased incidence of inflammatory diseases in humans, yet whether and how telomere dysfunction causes inflammation are not known. Here, we show that telomere dysfunction drives pATM/c-ABL-mediated activation of the YAP1 transcription factor, up-regulating the major pro-inflammatory factor, pro-IL-18. The colonic microbiome stimulates cytosolic receptors activating caspase-1 which cleaves pro-IL-18 into mature IL-18, leading to recruitment of interferon (IFN)-γ-secreting T cells and intestinal inflammation. Correspondingly, patients with germline telomere maintenance defects exhibit DNA damage (γH2AX) signaling together with elevated YAP1 and IL-18 expression. In mice with telomere dysfunction, telomerase reactivation in the intestinal epithelium or pharmacological inhibition of ATM, YAP1, or caspase-1 as well as antibiotic treatment, dramatically reduces IL-18 and intestinal inflammation. Thus, telomere dysfunction-induced activation of the ATM-YAP1-pro-IL-18 pathway in epithelium is a key instigator of tissue inflammation.
- Published
- 2020
- Full Text
- View/download PDF
11. Chromatin Regulator CHD1 Remodels the Immunosuppressive Tumor Microenvironment in PTEN-Deficient Prostate Cancer.
- Author
-
Zhao D, Cai L, Lu X, Liang X, Li J, Chen P, Ittmann M, Shang X, Jiang S, Li H, Meng C, Flores I, Song JH, Horner JW, Lan Z, Wu CJ, Li J, Chang Q, Chen KC, Wang G, Deng P, Spring DJ, Wang YA, and DePinho RA
- Subjects
- Animals, Cell Line, Tumor, Disease Models, Animal, Gene Expression Regulation, Neoplastic immunology, Humans, Male, Mice, Transgenic, Prostatic Neoplasms immunology, Prostatic Neoplasms pathology, Smad4 Protein genetics, Tumor Microenvironment genetics, DNA-Binding Proteins metabolism, PTEN Phosphohydrolase genetics, Prostatic Neoplasms genetics, Tumor Escape genetics, Tumor Microenvironment immunology
- Abstract
Genetic inactivation of PTEN is common in prostate cancer and correlates with poorer prognosis. We previously identified CHD1 as an essential gene in PTEN-deficient cancer cells. Here, we sought definitive in vivo genetic evidence for, and mechanistic understanding of, the essential role of CHD1 in PTEN-deficient prostate cancer. In Pten and Pten / Smad4 genetically engineered mouse models, prostate-specific deletion of Chd1 resulted in markedly delayed tumor progression and prolonged survival. Chd1 deletion was associated with profound tumor microenvironment (TME) remodeling characterized by reduced myeloid-derived suppressor cells (MDSC) and increased CD8
+ T cells. Further analysis identified IL6 as a key transcriptional target of CHD1, which plays a major role in recruitment of immunosuppressive MDSCs. Given the prominent role of MDSCs in suppressing responsiveness to immune checkpoint inhibitors (ICI), our genetic and tumor biological findings support combined testing of anti-IL6 and ICI therapies, specifically in PTEN-deficient prostate cancer. SIGNIFICANCE: We demonstrate a critical role of CHD1 in MDSC recruitment and discover CHD1/IL6 as a major regulator of the immunosuppressive TME of PTEN-deficient prostate cancer. Pharmacologic inhibition of IL6 in combination with immune checkpoint blockade elicits robust antitumor responses in prostate cancer. This article is highlighted in the In This Issue feature, p. 1241 ., (©2020 American Association for Cancer Research.)- Published
- 2020
- Full Text
- View/download PDF
12. Tumor Microenvironment Remodeling Enables Bypass of Oncogenic KRAS Dependency in Pancreatic Cancer.
- Author
-
Hou P, Kapoor A, Zhang Q, Li J, Wu CJ, Li J, Lan Z, Tang M, Ma X, Ackroyd JJ, Kalluri R, Zhang J, Jiang S, Spring DJ, Wang YA, and DePinho RA
- Subjects
- Humans, Pancreatic Neoplasms pathology, Tumor Microenvironment, Pancreatic Neoplasms, Oncogenes physiology, Pancreatic Neoplasms genetics, Proto-Oncogene Proteins p21(ras) metabolism
- Abstract
Oncogenic KRAS (KRAS*) is a key tumor maintenance gene in pancreatic ductal adenocarcinoma (PDAC), motivating pharmacologic targeting of KRAS* and its effectors. Here, we explored mechanisms involving the tumor microenvironment (TME) as a potential basis for resistance to targeting KRAS*. Using the inducible Kras
G12D ; Trp53-/- PDAC mouse model, gain-of-function screens of epigenetic regulators identified HDAC5 as the top hit enabling KRAS* independent tumor growth. HDAC5 -driven escaper tumors showed a prominent neutrophil-to-macrophage switch relative to KRAS*-driven tumors. Mechanistically, HDAC5 represses Socs3 , a negative regulator of chemokine CCL2, resulting in increased CCL2, which recruits CCR2+ macrophages. Correspondingly, enforced Ccl2 promotes macrophage recruitment into the TME and enables tumor recurrence following KRAS* extinction. These tumor-associated macrophages in turn provide cancer cells with trophic support including TGFβ to enable KRAS* bypass in a SMAD4-dependent manner. Our work uncovers a KRAS* resistance mechanism involving immune cell remodeling of the PDAC TME. SIGNIFICANCE: Although KRAS* is required for PDAC tumor maintenance, tumors can recur following KRAS* extinction. The capacity of PDAC cancer cells to alter the TME myeloid cell composition to support KRAS*-independent tumor growth illuminates novel therapeutic targets that may enhance the effectiveness of therapies targeting KRAS* and its pathway components. See related commentary by Carr and Fernandez-Zapico, p. 910 . This article is highlighted in the In This Issue feature, p. 890 ., (©2020 American Association for Cancer Research.)- Published
- 2020
- Full Text
- View/download PDF
13. Oncogenic KRAS-Driven Metabolic Reprogramming in Pancreatic Cancer Cells Utilizes Cytokines from the Tumor Microenvironment.
- Author
-
Dey P, Li J, Zhang J, Chaurasiya S, Strom A, Wang H, Liao WT, Cavallaro F, Denz P, Bernard V, Yen EY, Genovese G, Gulhati P, Liu J, Chakravarti D, Deng P, Zhang T, Carbone F, Chang Q, Ying H, Shang X, Spring DJ, Ghosh B, Putluri N, Maitra A, Wang YA, and DePinho RA
- Subjects
- Animals, Cellular Reprogramming genetics, Humans, Mice, Oncogenes, Pancreatic Neoplasms metabolism, Pancreatic Neoplasms pathology, Proto-Oncogene Proteins p21(ras) metabolism, Transfection, Tumor Microenvironment, Cytokines metabolism, Pancreatic Neoplasms genetics, Proto-Oncogene Proteins p21(ras) genetics
- Abstract
A hallmark of pancreatic ductal adenocarcinoma (PDAC) is an exuberant stroma comprised of diverse cell types that enable or suppress tumor progression. Here, we explored the role of oncogenic KRAS in protumorigenic signaling interactions between cancer cells and host cells. We show that KRAS mutation (KRAS*) drives cell-autonomous expression of type I cytokine receptor complexes (IL2rγ-IL4rα and IL2rγ-IL13rα1) in cancer cells that in turn are capable of receiving cytokine growth signals (IL4 or IL13) provided by invading Th2 cells in the microenvironment. Early neoplastic lesions show close proximity of cancer cells harboring KRAS* and Th2 cells producing IL4 and IL13. Activated IL2rγ-IL4rα and IL2rγ-IL13rα1 receptors signal primarily via JAK1-STAT6. Integrated transcriptomic, chromatin occupancy, and metabolomic studies identified MYC as a direct target of activated STAT6 and that MYC drives glycolysis. Thus, paracrine signaling in the tumor microenvironment plays a key role in the KRAS*-driven metabolic reprogramming of PDAC. SIGNIFICANCE: Type II cytokines, secreted by Th2 cells in the tumor microenvironment, can stimulate cancer cell-intrinsic MYC transcriptional upregulation to drive glycolysis. This KRAS*-driven heterotypic signaling circuit in the early and advanced tumor microenvironment enables cooperative protumorigenic interactions, providing candidate therapeutic targets in the KRAS* pathway for this intractable disease., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
14. Circadian Regulator CLOCK Recruits Immune-Suppressive Microglia into the GBM Tumor Microenvironment.
- Author
-
Chen P, Hsu WH, Chang A, Tan Z, Lan Z, Zhou A, Spring DJ, Lang FF, Wang YA, and DePinho RA
- Subjects
- Animals, Cell Line, Tumor, Cell Self Renewal genetics, Cell Self Renewal immunology, Gene Expression Regulation, Neoplastic genetics, Glioblastoma immunology, Glioblastoma pathology, Glioblastoma therapy, Heterografts, Humans, Immunity, Cellular immunology, Mice, Microglia immunology, Microglia metabolism, Neoplastic Stem Cells immunology, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells pathology, Tumor Microenvironment genetics, Tumor Microenvironment immunology, ARNTL Transcription Factors genetics, CLOCK Proteins genetics, Glioblastoma genetics, Glycoproteins genetics
- Abstract
Glioblastoma (GBM) is a lethal brain tumor containing a subpopulation of glioma stem cells (GSC). Pan-cancer analyses have revealed that stemness of cancer cells correlates positively with immunosuppressive pathways in many solid tumors, including GBM, prompting us to conduct a gain-of-function screen of epigenetic regulators that may influence GSC self-renewal and tumor immunity. The circadian regulator CLOCK emerged as a top hit in enhancing stem-cell self-renewal, which was amplified in about 5% of human GBM cases. CLOCK and its heterodimeric partner BMAL1 enhanced GSC self-renewal and triggered protumor immunity via transcriptional upregulation of OLFML3, a novel chemokine recruiting immune-suppressive microglia into the tumor microenvironment. In GBM models, CLOCK or OLFML3 depletion reduced intratumoral microglia density and extended overall survival. We conclude that the CLOCK-BMAL1 complex contributes to key GBM hallmarks of GSC maintenance and immunosuppression and, together with its downstream target OLFML3, represents new therapeutic targets for this disease. SIGNIFICANCE: Circadian regulator CLOCK drives GSC self-renewal and metabolism and promotes microglia infiltration through direct regulation of a novel microglia-attracting chemokine, OLFML3. CLOCK and/or OLFML3 may represent novel therapeutic targets for GBM. This article is highlighted in the In This Issue feature, p. 327 ., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
15. USP21 deubiquitinase promotes pancreas cancer cell stemness via Wnt pathway activation.
- Author
-
Hou P, Ma X, Zhang Q, Wu CJ, Liao W, Li J, Wang H, Zhao J, Zhou X, Guan C, Ackroyd J, Jiang S, Zhang J, Spring DJ, Wang YA, and DePinho RA
- Subjects
- Animals, Cell Line, Tumor, Humans, Mice, Neoplastic Stem Cells pathology, Pancreatic Neoplasms genetics, Pancreatic Neoplasms physiopathology, T Cell Transcription Factor 1, Ubiquitination, Pancreatic Neoplasms, Gene Expression Regulation, Neoplastic genetics, Pancreatic Neoplasms enzymology, Ubiquitin Thiolesterase genetics, Ubiquitin Thiolesterase metabolism, Wnt Signaling Pathway genetics
- Abstract
The ubiquitin-specific protease (USP) family is the largest group of cysteine proteases. Cancer genomic analysis identified frequent amplification of USP21 (22%) in human pancreatic ductal adenocarcinoma (PDAC). USP21 overexpression correlates with human PDAC progression, and enforced expression of USP21 accelerates murine PDAC tumor growth and drives PanIN to PDAC progression in immortalized human pancreatic ductal cells. Conversely, depletion of USP21 impairs PDAC tumor growth. Mechanistically, USP21 deubiquitinates and stabilizes the TCF/LEF transcription factor TCF7, which promotes cancer cell stemness. Our work identifies and validates USP21 as a PDAC oncogene, providing a potential druggable target for this intractable disease., (© 2019 Hou et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2019
- Full Text
- View/download PDF
16. Symbiotic Macrophage-Glioma Cell Interactions Reveal Synthetic Lethality in PTEN-Null Glioma.
- Author
-
Chen P, Zhao D, Li J, Liang X, Li J, Chang A, Henry VK, Lan Z, Spring DJ, Rao G, Wang YA, and DePinho RA
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing metabolism, Animals, Antineoplastic Agents pharmacology, Biomarkers, Tumor deficiency, Brain Neoplasms drug therapy, Brain Neoplasms enzymology, Brain Neoplasms pathology, Cell Movement, Cell Proliferation, Enzyme Inhibitors pharmacology, Female, Focal Adhesion Kinase 2 genetics, Focal Adhesion Kinase 2 metabolism, Gene Expression Regulation, Neoplastic, Glioma drug therapy, Glioma enzymology, Glioma pathology, HEK293 Cells, Humans, Integrin beta1 genetics, Integrin beta1 metabolism, Macrophages drug effects, Macrophages pathology, Mice, Mice, Inbred C57BL, Mice, Inbred ICR, Mice, SCID, Osteopontin genetics, Osteopontin metabolism, PTEN Phosphohydrolase deficiency, Protein-Lysine 6-Oxidase antagonists & inhibitors, Protein-Lysine 6-Oxidase genetics, RAW 264.7 Cells, Signal Transduction, THP-1 Cells, Transcription Factors genetics, Transcription Factors metabolism, Tumor Burden, Xenograft Model Antitumor Assays, YAP-Signaling Proteins, Biomarkers, Tumor genetics, Brain Neoplasms genetics, Glioma genetics, Macrophages enzymology, PTEN Phosphohydrolase genetics, Paracrine Communication drug effects, Protein-Lysine 6-Oxidase metabolism, Synthetic Lethal Mutations
- Abstract
Heterotypic interactions across diverse cell types can enable tumor progression and hold the potential to expand therapeutic interventions. Here, combined profiling and functional studies of glioma cells in glioblastoma multiforme (GBM) models establish that PTEN deficiency activates YAP1, which directly upregulates lysyl oxidase (LOX) expression. Mechanistically, secreted LOX functions as a potent macrophage chemoattractant via activation of the β1 integrin-PYK2 pathway in macrophages. These infiltrating macrophages secrete SPP1, which sustains glioma cell survival and stimulates angiogenesis. In PTEN-null GBM models, LOX inhibition markedly suppresses macrophage infiltration and tumor progression. Correspondingly, YAP1-LOX and β1 integrin-SPP1 signaling correlates positively with higher macrophage density and lower overall survival in GBM patients. This symbiotic glioma-macrophage interplay provides therapeutic targets specifically for PTEN-deficient GBM., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
17. KRAS-IRF2 Axis Drives Immune Suppression and Immune Therapy Resistance in Colorectal Cancer.
- Author
-
Liao W, Overman MJ, Boutin AT, Shang X, Zhao D, Dey P, Li J, Wang G, Lan Z, Li J, Tang M, Jiang S, Ma X, Chen P, Katkhuda R, Korphaisarn K, Chakravarti D, Chang A, Spring DJ, Chang Q, Zhang J, Maru DM, Maeda DY, Zebala JA, Kopetz S, Wang YA, and DePinho RA
- Subjects
- Adenomatous Polyposis Coli Protein genetics, Adenomatous Polyposis Coli Protein metabolism, Adult, Aged, Animals, Cell Line, Tumor, Cell Movement, Chemokines, CXC metabolism, Colorectal Neoplasms genetics, Colorectal Neoplasms immunology, Colorectal Neoplasms metabolism, Female, Gene Expression Regulation, Neoplastic, Humans, Interferon Regulatory Factor-2 genetics, Male, Mice, Inbred C57BL, Mice, Inbred NOD, Mice, SCID, Mice, Transgenic, Middle Aged, Myeloid-Derived Suppressor Cells drug effects, Myeloid-Derived Suppressor Cells immunology, Myeloid-Derived Suppressor Cells metabolism, Programmed Cell Death 1 Receptor immunology, Programmed Cell Death 1 Receptor metabolism, Proto-Oncogene Proteins p21(ras) genetics, Receptors, Interleukin-8B metabolism, Signal Transduction, Tumor Microenvironment, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Young Adult, Antineoplastic Agents, Immunological pharmacology, Colorectal Neoplasms drug therapy, Drug Resistance, Neoplasm genetics, Interferon Regulatory Factor-2 metabolism, Programmed Cell Death 1 Receptor antagonists & inhibitors, Proto-Oncogene Proteins p21(ras) metabolism, Tumor Escape
- Abstract
The biological functions and mechanisms of oncogenic KRAS
G12D (KRAS∗ ) in resistance to immune checkpoint blockade (ICB) therapy are not fully understood. We demonstrate that KRAS∗ represses the expression of interferon regulatory factor 2 (IRF2), which in turn directly represses CXCL3 expression. KRAS∗ -mediated repression of IRF2 results in high expression of CXCL3, which binds to CXCR2 on myeloid-derived suppressor cells and promotes their migration to the tumor microenvironment. Anti-PD-1 resistance of KRAS∗ -expressing tumors can be overcome by enforced IRF2 expression or by inhibition of CXCR2. Colorectal cancer (CRC) showing higher IRF2 expression exhibited increased responsiveness to anti-PD-1 therapy. The KRAS∗ -IRF2-CXCL3-CXCR2 axis provides a framework for patient selection and combination therapies to enhance the effectiveness of ICB therapy in CRC., (Copyright © 2019 Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
18. Genetics and biology of prostate cancer.
- Author
-
Wang G, Zhao D, Spring DJ, and DePinho RA
- Subjects
- Animals, Drug Resistance, Neoplasm, Gene Fusion, Humans, Male, Mice, Neoplasm Metastasis, Prognosis, Prostate anatomy & histology, Prostatic Neoplasms diagnosis, Prostatic Neoplasms pathology, Tumor Microenvironment, Prostatic Neoplasms genetics, Prostatic Neoplasms therapy
- Abstract
Despite the high long-term survival in localized prostate cancer, metastatic prostate cancer remains largely incurable even after intensive multimodal therapy. The lethality of advanced disease is driven by the lack of therapeutic regimens capable of generating durable responses in the setting of extreme tumor heterogeneity on the genetic and cell biological levels. Here, we review available prostate cancer model systems, the prostate cancer genome atlas, cellular and functional heterogeneity in the tumor microenvironment, tumor-intrinsic and tumor-extrinsic mechanisms underlying therapeutic resistance, and technological advances focused on disease detection and management. These advances, along with an improved understanding of the adaptive responses to conventional cancer therapies, anti-androgen therapy, and immunotherapy, are catalyzing development of more effective therapeutic strategies for advanced disease. In particular, knowledge of the heterotypic interactions between and coevolution of cancer and host cells in the tumor microenvironment has illuminated novel therapeutic combinations with a strong potential for more durable therapeutic responses and eventual cures for advanced disease. Improved disease management will also benefit from artificial intelligence-based expert decision support systems for proper standard of care, prognostic determinant biomarkers to minimize overtreatment of localized disease, and new standards of care accelerated by next-generation adaptive clinical trials., (© 2018 Wang et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2018
- Full Text
- View/download PDF
19. PAF promotes stemness and radioresistance of glioma stem cells.
- Author
-
Ong DST, Hu B, Ho YW, Sauvé CG, Bristow CA, Wang Q, Multani AS, Chen P, Nezi L, Jiang S, Gorman CE, Monasterio MM, Koul D, Marchesini M, Colla S, Jin EJ, Sulman EP, Spring DJ, Yung WA, Verhaak RGW, Chin L, Wang YA, and DePinho RA
- Subjects
- Animals, Brain Neoplasms genetics, Brain Neoplasms mortality, Brain Neoplasms pathology, Carrier Proteins metabolism, DNA Damage genetics, DNA Damage radiation effects, DNA Repair genetics, DNA Repair radiation effects, DNA Replication drug effects, DNA-Binding Proteins, Female, Gene Expression Regulation, Neoplastic radiation effects, Glioblastoma genetics, Glioblastoma mortality, Glioblastoma pathology, Green Fluorescent Proteins genetics, Humans, Mice, SCID, Neoplastic Stem Cells drug effects, Neoplastic Stem Cells pathology, Pyrimidines biosynthesis, Radiation Tolerance, Xenograft Model Antitumor Assays, Brain Neoplasms radiotherapy, Carrier Proteins genetics, Glioblastoma radiotherapy, Neoplastic Stem Cells radiation effects
- Abstract
An integrated genomic and functional analysis to elucidate DNA damage signaling factors promoting self-renewal of glioma stem cells (GSCs) identified proliferating cell nuclear antigen (PCNA)-associated factor ( PAF ) up-regulation in glioblastoma. PAF is preferentially overexpressed in GSCs. Its depletion impairs maintenance of self-renewal without promoting differentiation and reduces tumor-initiating cell frequency. Combined transcriptomic and metabolomic analyses revealed that PAF supports GSC maintenance, in part, by influencing DNA replication and pyrimidine metabolism pathways. PAF interacts with PCNA and regulates PCNA-associated DNA translesion synthesis (TLS); consequently, PAF depletion in combination with radiation generated fewer tumorspheres compared with radiation alone. Correspondingly, pharmacological impairment of DNA replication and TLS phenocopied the effect of PAF depletion in compromising GSC self-renewal and radioresistance, providing preclinical proof of principle that combined TLS inhibition and radiation therapy may be a viable therapeutic option in the treatment of glioblastoma multiforme (GBM)., Competing Interests: The authors declare no conflict of interest., (Published under the PNAS license.)
- Published
- 2017
- Full Text
- View/download PDF
20. Erratum: Effective combinatorial immunotherapy for castration-resistant prostate cancer.
- Author
-
Lu X, Horner JW, Paul E, Shang X, Troncoso P, Deng P, Jiang S, Chang Q, Spring DJ, Sharma P, Zebala JA, Maeda DY, Wang YA, and DePinho RA
- Published
- 2017
- Full Text
- View/download PDF
21. Effective combinatorial immunotherapy for castration-resistant prostate cancer.
- Author
-
Lu X, Horner JW, Paul E, Shang X, Troncoso P, Deng P, Jiang S, Chang Q, Spring DJ, Sharma P, Zebala JA, Maeda DY, Wang YA, and DePinho RA
- Subjects
- Anilides pharmacology, Anilides therapeutic use, Animals, CD8-Positive T-Lymphocytes drug effects, CD8-Positive T-Lymphocytes immunology, Chimera, Cytokines immunology, Cytokines metabolism, Disease Models, Animal, Drug Synergism, Female, Humans, Imidazoles pharmacology, Imidazoles therapeutic use, Lymphocytes, Tumor-Infiltrating drug effects, Lymphocytes, Tumor-Infiltrating immunology, Male, Mice, Molecular Targeted Therapy, Myeloid-Derived Suppressor Cells cytology, Myeloid-Derived Suppressor Cells drug effects, Myeloid-Derived Suppressor Cells immunology, Phosphoinositide-3 Kinase Inhibitors, Prostatic Neoplasms, Castration-Resistant pathology, Pyridines pharmacology, Pyridines therapeutic use, Quinolines pharmacology, Quinolines therapeutic use, Signal Transduction drug effects, Tumor Microenvironment drug effects, Tumor Microenvironment immunology, Immunotherapy methods, Prostatic Neoplasms, Castration-Resistant immunology, Prostatic Neoplasms, Castration-Resistant therapy
- Abstract
A significant fraction of patients with advanced prostate cancer treated with androgen deprivation therapy experience relapse with relentless progression to lethal metastatic castration-resistant prostate cancer (mCRPC). Immune checkpoint blockade using antibodies against cytotoxic-T-lymphocyte-associated protein 4 (CTLA4) or programmed cell death 1/programmed cell death 1 ligand 1 (PD1/PD-L1) generates durable therapeutic responses in a significant subset of patients across a variety of cancer types. However, mCRPC showed overwhelming de novo resistance to immune checkpoint blockade, motivating a search for targeted therapies that overcome this resistance. Myeloid-derived suppressor cells (MDSCs) are known to play important roles in tumour immune evasion. The abundance of circulating MDSCs correlates with prostate-specific antigen levels and metastasis in patients with prostate cancer. Mouse models of prostate cancer show that MDSCs (CD11b
+ Gr1+ ) promote tumour initiation and progression. These observations prompted us to hypothesize that robust immunotherapy responses in mCRPC may be elicited by the combined actions of immune checkpoint blockade agents together with targeted agents that neutralize MDSCs yet preserve T-cell function. Here we develop a novel chimaeric mouse model of mCRPC to efficiently test combination therapies in an autochthonous setting. Combination of anti-CTLA4 and anti-PD1 engendered only modest efficacy. Targeted therapy against mCRPC-infiltrating MDSCs, using multikinase inhibitors such as cabozantinib and BEZ235, also showed minimal anti-tumour activities. Strikingly, primary and metastatic CRPC showed robust synergistic responses when immune checkpoint blockade was combined with MDSC-targeted therapy. Mechanistically, combination therapy efficacy stemmed from the upregulation of interleukin-1 receptor antagonist and suppression of MDSC-promoting cytokines secreted by prostate cancer cells. These observations illuminate a clinical path hypothesis for combining immune checkpoint blockade with MDSC-targeted therapies in the treatment of mCRPC.- Published
- 2017
- Full Text
- View/download PDF
22. Synthetic essentiality of chromatin remodelling factor CHD1 in PTEN-deficient cancer.
- Author
-
Zhao D, Lu X, Wang G, Lan Z, Liao W, Li J, Liang X, Chen JR, Shah S, Shang X, Tang M, Deng P, Dey P, Chakravarti D, Chen P, Spring DJ, Navone NM, Troncoso P, Zhang J, Wang YA, and DePinho RA
- Subjects
- Breast Neoplasms genetics, Breast Neoplasms metabolism, Breast Neoplasms pathology, Cell Line, Tumor, DNA Helicases chemistry, DNA Helicases deficiency, DNA Helicases genetics, DNA-Binding Proteins chemistry, DNA-Binding Proteins deficiency, DNA-Binding Proteins genetics, Female, Gene Expression Regulation, Neoplastic, Glycogen Synthase Kinase 3 beta metabolism, Histones metabolism, Humans, Lysine metabolism, Male, Methylation, Molecular Targeted Therapy, NF-kappa B metabolism, Neoplasms genetics, PTEN Phosphohydrolase genetics, PTEN Phosphohydrolase metabolism, Phosphorylation, Prostatic Neoplasms genetics, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology, Proteasome Endopeptidase Complex metabolism, Protein Stability, Proteolysis, Tumor Necrosis Factor-alpha metabolism, Ubiquitination, beta-Transducin Repeat-Containing Proteins metabolism, Chromatin Assembly and Disassembly genetics, DNA Helicases metabolism, DNA-Binding Proteins metabolism, Genes, Essential genetics, Neoplasms metabolism, Neoplasms pathology, PTEN Phosphohydrolase deficiency
- Abstract
Synthetic lethality and collateral lethality are two well-validated conceptual strategies for identifying therapeutic targets in cancers with tumour-suppressor gene deletions. Here, we explore an approach to identify potential synthetic-lethal interactions by screening mutually exclusive deletion patterns in cancer genomes. We sought to identify 'synthetic-essential' genes: those that are occasionally deleted in some cancers but are almost always retained in the context of a specific tumour-suppressor deficiency. We also posited that such synthetic-essential genes would be therapeutic targets in cancers that harbour specific tumour-suppressor deficiencies. In addition to known synthetic-lethal interactions, this approach uncovered the chromatin helicase DNA-binding factor CHD1 as a putative synthetic-essential gene in PTEN-deficient cancers. In PTEN-deficient prostate and breast cancers, CHD1 depletion profoundly and specifically suppressed cell proliferation, cell survival and tumorigenic potential. Mechanistically, functional PTEN stimulates the GSK3β-mediated phosphorylation of CHD1 degron domains, which promotes CHD1 degradation via the β-TrCP-mediated ubiquitination-proteasome pathway. Conversely, PTEN deficiency results in stabilization of CHD1, which in turn engages the trimethyl lysine-4 histone H3 modification to activate transcription of the pro-tumorigenic TNF-NF-κB gene network. This study identifies a novel PTEN pathway in cancer and provides a framework for the discovery of 'trackable' targets in cancers that harbour specific tumour-suppressor deficiencies.
- Published
- 2017
- Full Text
- View/download PDF
23. Epigenetic Activation of WNT5A Drives Glioblastoma Stem Cell Differentiation and Invasive Growth.
- Author
-
Hu B, Wang Q, Wang YA, Hua S, Sauvé CG, Ong D, Lan ZD, Chang Q, Ho YW, Monasterio MM, Lu X, Zhong Y, Zhang J, Deng P, Tan Z, Wang G, Liao WT, Corley LJ, Yan H, Zhang J, You Y, Liu N, Cai L, Finocchiaro G, Phillips JJ, Berger MS, Spring DJ, Hu J, Sulman EP, Fuller GN, Chin L, Verhaak RGW, and DePinho RA
- Subjects
- Endothelial Cells cytology, Endothelial Cells metabolism, Epigenomics, Gene Expression Regulation, Neoplastic, Homeodomain Proteins metabolism, Humans, Neural Stem Cells metabolism, PAX6 Transcription Factor metabolism, Proto-Oncogene Proteins c-akt metabolism, Transcription Factors metabolism, Glioblastoma genetics, Glioblastoma pathology, Neoplasm Invasiveness genetics, Wnt-5a Protein genetics
- Abstract
Glioblastoma stem cells (GSCs) are implicated in tumor neovascularization, invasiveness, and therapeutic resistance. To illuminate mechanisms governing these hallmark features, we developed a de novo glioblastoma multiforme (GBM) model derived from immortalized human neural stem/progenitor cells (hNSCs) to enable precise system-level comparisons of pre-malignant and oncogene-induced malignant states of NSCs. Integrated transcriptomic and epigenomic analyses uncovered a PAX6/DLX5 transcriptional program driving WNT5A-mediated GSC differentiation into endothelial-like cells (GdECs). GdECs recruit existing endothelial cells to promote peritumoral satellite lesions, which serve as a niche supporting the growth of invasive glioma cells away from the primary tumor. Clinical data reveal higher WNT5A and GdECs expression in peritumoral and recurrent GBMs relative to matched intratumoral and primary GBMs, respectively, supporting WNT5A-mediated GSC differentiation and invasive growth in disease recurrence. Thus, the PAX6/DLX5-WNT5A axis governs the diffuse spread of glioma cells throughout the brain parenchyma, contributing to the lethality of GBM., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
24. Cancer Genomics in Clinical Context.
- Author
-
Chin L, Wargo JA, Spring DJ, Kantarjian H, and Futreal PA
- Abstract
Precision medicine requires appropriate application of genomics in clinical practice. In cancer, we have witnessed practice-changing examples of how genomic knowledge is translated into more tailored and effective therapies. The next opportunity is to embed cancer genomics in clinical context so that patient-centric longitudinal clinical, genomic, and molecular phenotypes can be compiled for adaptive learning between precision medicine research and clinical care with the goal of accelerating clinically-actionable discoveries. We describe here an adaptive learning platform, APOLLO™ (adaptive patient-oriented longitudinal learning and optimization) designed to integrate genomic research in the context of, but not in the path of, routine and investigational clinical care for purposes of enabling data-driven discovery across disciplines such that every patient can contribute to and potentially benefit from research discoveries., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
25. Specific inhibition of p38 mitogen-activated protein kinase with FR167653 attenuates vascular proliferation in monocrotaline-induced pulmonary hypertension in rats.
- Author
-
Lu J, Shimpo H, Shimamoto A, Chong AJ, Hampton CR, Spring DJ, Yada M, Takao M, Onoda K, Yada I, Pohlman TH, and Verrier ED
- Subjects
- Animals, Cytokines metabolism, Hypertension, Pulmonary chemically induced, Hypertrophy, Right Ventricular physiopathology, Immunohistochemistry, Lung cytology, Male, Monocrotaline, Rats, Rats, Sprague-Dawley, Reverse Transcriptase Polymerase Chain Reaction, p38 Mitogen-Activated Protein Kinases drug effects, p38 Mitogen-Activated Protein Kinases metabolism, Anti-Inflammatory Agents, Non-Steroidal pharmacology, Hypertension, Pulmonary physiopathology, Pyrazoles pharmacology, Pyridines pharmacology, p38 Mitogen-Activated Protein Kinases antagonists & inhibitors
- Abstract
Objectives: p38 mitogen-activated protein kinase is associated with many clinical entities characterized by inflammation. We postulated that inhibition of p38 mitogen-activated protein kinase with FR167653 attenuates inflammation and the development of pulmonary hypertension in monocrotaline-treated rats., Methods: Rats were divided into 4 groups: (1) the control group (daily 0.9% saline), (2) the FR group (daily FR167653, 2 mg . kg(-1) . d(-1)), (3) the MCT group (daily 0.9% saline the day after a single monocrotaline dose, 60 mg/kg), and (4) the MCT+FR group (daily FR167653, 2 mg . kg(-1) . d(-1), the day after a single MCT dose). Body weight, pulmonary artery pressure, and morphometric changes of the pulmonary artery with the histopathologic method were observed weekly for 4 weeks. Also, p38 mitogen-activated protein kinase activity and inflammatory cytokine expression in the lung were measured., Results: Four weeks after monocrotaline administration, mean pulmonary artery pressure in the MCT+FR group was lower than in the MCT group (MCT+FR vs MCT: 24.7 +/- 1.9 vs 36.5 +/- 2.1 mm Hg; P < .05). In morphometric analysis the percentage of medial wall thickness and the percentage of muscularization in the MCT+FR group were reduced compared with those in the MCT group after 4 weeks (P < .05); however, the number of macrophages was not significantly different. p38 mitogen-activated protein kinase activity was significantly attenuated in the MCT+FR group compared with in the MCT group (7.2 +/- 0.52 vs 2.1 +/- 0.23 fold-increase, P < .05, at 1 week). Although mRNA levels of tumor necrosis factor alpha and interleukin 1beta were reduced in the MCT+FR group compared with in the MCT group (tumor necrosis factor alpha: 1.18 +/- 0.36 vs 3.05 +/- 1.12 fold-increase, P < .05, at 2 weeks; interleukin 1beta: 2.2 +/- 0.34 vs 4.4 +/- 1.09 fold-increase, P < .05, at 1 week), FR167653 did not suppress increased monocyte chemotactic protein 1 mRNA expression induced by monocrotaline (3.2 +/- 0.62 vs 3.1 +/- 0.42 fold-increase, at 1 week)., Conclusion: FR167653 significantly attenuates the expression of inflammatory cytokines, ultimately preventing the progression of pulmonary hypertension. These results suggest that p38 mitogen-activated protein kinase might play a central role in the molecular events that underlie the development and progression of pulmonary hypertension.
- Published
- 2004
- Full Text
- View/download PDF
26. FR167653 diminishes infarct size in a murine model of myocardial ischemia-reperfusion injury.
- Author
-
Yada M, Shimamoto A, Hampton CR, Chong AJ, Takayama H, Rothnie CL, Spring DJ, Shimpo H, Yada I, Pohlman TH, and Verrier ED
- Subjects
- Animals, Blotting, Western, Male, Mice, NF-kappa B antagonists & inhibitors, NF-kappa B metabolism, Phosphorylation, Premedication, p38 Mitogen-Activated Protein Kinases antagonists & inhibitors, Enzyme Inhibitors therapeutic use, Myocardial Reperfusion Injury prevention & control, Pyrazoles therapeutic use, Pyridines therapeutic use
- Abstract
Objective: During myocardial ischemia-reperfusion injury, p38 mitogen-activated protein kinase is activated. We examined the effect of a highly specific inhibitor of p38 mitogen-activated protein kinase, FR167653, in an experimental model of regional myocardial ischemia-reperfusion., Methods: CD-1 mice received FR167653 intraperitoneally 24 hours before 30 minutes of transient occlusion of the left anterior descending artery, followed by 120 minutes of reperfusion. The p38 mitogen-activated protein kinase activation and kinase activity were determined by Western blotting with monoclonal antibodies for the phosphorylated from of p38 mitogen-activated protein kinase or its substrate, activating transcription factor 2. Nuclear factor kappaB activity was measured by detecting translocation of nuclear factor kappaB to the nucleus. The expression of inflammatory cytokines was measured by ribonuclease protection assay., Results: Pretreatment of mice with FR167653 before myocardial ischemia-reperfusion resulted in a reduction in p38 mitogen-activated protein kinase phosphorylation (P =.018), an inhibition of p38 mitogen-activated protein kinase activity (P =.047), a smaller amount of nuclear factor kappaB (P =.001), and a decrease in the expression of inflammatory cytokines (tumor necrosis factor alpha: P =.023, interleukin 1beta: P =.038, monocyte chemotactic protein 1: P =.0001) in the heart and the development of a significantly smaller infarct (P =.0069) relative to hearts from mice treated with vehicle alone. Activation of c-Jun N-terminal kinase and extracellular signal-regulated kinase were observed after myocardial ischemia-reperfusion without inhibition by FR167653., Conclusion: We conclude that FR167653 selectively inhibits p38 mitogen-activated protein kinase activation and activity during regional myocardial ischemia-reperfusion injury and efficaciously reduces infarct size (by 73.6%). Thus p38 mitogen-activated protein kinase inhibition may have a role in the treatment of myocardial ischemia-reperfusion.
- Published
- 2004
- Full Text
- View/download PDF
27. Toll-like receptor 4 mediates ischemia/reperfusion injury of the heart.
- Author
-
Chong AJ, Shimamoto A, Hampton CR, Takayama H, Spring DJ, Rothnie CL, Yada M, Pohlman TH, and Verrier ED
- Subjects
- Animals, Inflammation Mediators physiology, Mice, Mice, Inbred C3H, Mitogen-Activated Protein Kinases physiology, Myocardial Infarction etiology, Toll-Like Receptors, Membrane Glycoproteins physiology, Myocardial Reperfusion Injury etiology, Receptors, Cell Surface physiology
- Abstract
Background: Restoration of blood flow to the ischemic heart may paradoxically exacerbate tissue injury (ischemia/reperfusion injury). Toll-like receptor 4, expressed on several cell types, including cardiomyocytes, is a mediator of the host inflammatory response to infection. Because ischemia/reperfusion injury is characterized by an acute inflammatory reaction, we investigated toll-like receptor 4 activation in a murine model of regional myocardial ischemia/reperfusion injury. We used C3H/HeJ mice, which express a nonfunctional toll-like receptor 4, to assess the pertinence of this receptor to tissue injury after reperfusion of ischemic myocardium., Methods: Wild-type mice (C3H/HeN) or toll-like receptor 4 mutant mice (C3H/HeJ) were subjected to 60 minutes of regional myocardial ischemia followed by 2 hours of reperfusion. At the end of reperfusion, the area at risk and the myocardial infarct size were measured as the end point of myocardial ischemia/reperfusion injury. Myocardial mitogen-activated protein kinase activation was measured by Western blotting, and nuclear translocation of nuclear factor-kappaB and activator protein-1 was determined by electrophoretic mobility shift assay. Ischemia/reperfusion-injured myocardium was also assessed by ribonuclease protection assay for expression of inflammatory mediators (tumor necrosis factor-alpha, interleukin-1beta, monocyte chemotactic factor-1, and interleukin-6)., Results: The area at risk was similar for all groups after myocardial ischemia/reperfusion injury. There was a 40% reduction in infarct size (as a percentage of the area at risk) in C3H/HeJ mice compared with C3H/HeN mice (P =.001). Within the myocardium, significant activation of c-Jun N-terminal kinase, p38, and extracellular signal-regulated kinase was observed in both strains after ischemia and during reperfusion as compared with an absence of mitogen-activated protein kinase activation during sham operations; however, c-Jun N-terminal kinase activity, but not p38 or extracellular signal-regulated kinase activity, was significantly reduced in C3H/HeJ mice (P <.05). In both groups, nuclear factor-kappaB and activator protein-1 nuclear translocation occurred in the myocardium during myocardial ischemia/reperfusion injury, but, by densitometric analysis, nuclear translocation of nuclear factor-kappaB and activator protein-1 was significantly decreased in C3H/HeJ mice compared with C3H/HeN mice. Interleukin-1beta, monocyte chemotactic factor-1, and interleukin-6 were detectable in reperfused ischemic myocardium but were not detected in sham-operated myocardium; the expression of each of these mediators was significantly decreased in the myocardial tissue of C3H/HeJ mice when compared with expression in the control C3H/HeN mouse strain., Conclusions: Our data suggest that toll-like receptor 4 may mediate, at least in part, myocardial ischemia/reperfusion injury. Inhibition of toll-like receptor 4 activation may be a potential therapeutic target to attenuate ischemia/reperfusion-induced tissue damage in the clinical setting.
- Published
- 2004
- Full Text
- View/download PDF
28. p90(RSK) is a serum-stimulated Na+/H+ exchanger isoform-1 kinase. Regulatory phosphorylation of serine 703 of Na+/H+ exchanger isoform-1.
- Author
-
Takahashi E, Abe J, Gallis B, Aebersold R, Spring DJ, Krebs EG, and Berk BC
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Blood, Cells, Cultured, DNA Primers, DNA, Complementary, Enzyme Activation, Enzyme Inhibitors pharmacology, Flavonoids pharmacology, MAP Kinase Kinase 1, Male, Mutagenesis, Site-Directed, Peptide Mapping, Phosphorylation, Precipitin Tests, Protein Isoforms chemistry, Protein Isoforms genetics, Protein Serine-Threonine Kinases antagonists & inhibitors, Protein-Tyrosine Kinases antagonists & inhibitors, Rats, Rats, Sprague-Dawley, Ribosomal Protein S6 Kinases genetics, Sodium-Hydrogen Exchangers chemistry, Sodium-Hydrogen Exchangers genetics, Thrombin pharmacology, Transfection, Mitogen-Activated Protein Kinase Kinases, Protein Isoforms metabolism, Ribosomal Protein S6 Kinases metabolism, Serine metabolism, Sodium-Hydrogen Exchangers metabolism
- Abstract
The Na+/H+ exchanger isoform-1 (NHE-1) is the key member of a family of exchangers that regulates intracellular pH and cell volume. Activation of NHE-1 by growth factors is rapid, correlates with increased NHE-1 phosphorylation and cell alkalinization, and plays a role in cell cycle progression. By two-dimensional tryptic peptide mapping of immunoprecipitated NHE-1, we identify serine 703 as the major serum-stimulated amino acid. Mutation of serine 703 to alanine had no effect on acid-stimulated Na+/H+ exchange but completely prevented the growth factor-mediated increase in NHE-1 affinity for H+. In addition, we show that p90 ribosomal S6 kinase (p90(RSK)) is a key NHE-1 kinase since p90(RSK) phosphorylates NHE-1 serine 703 stoichiometrically in vitro, and transfection with kinase-inactive p90(RSK) inhibits serum-induced phosphorylation of NHE-1 serine 703 in transfected 293 cells. These findings establish p90(RSK) as a serum-stimulated NHE-1 kinase and a mediator of increased Na+/H+ exchange in vivo.
- Published
- 1999
- Full Text
- View/download PDF
29. Deletion of 11 amino acids in p90(rsk-mo-1) abolishes kinase activity.
- Author
-
Spring DJ and Krebs EG
- Subjects
- Amino Acid Sequence, Animals, COS Cells, Humans, Mice, Molecular Sequence Data, Mutagenesis, Rats, Sequence Homology, Amino Acid, Structure-Activity Relationship, Ribosomal Protein S6 Kinases genetics, Ribosomal Protein S6 Kinases metabolism
- Abstract
p90(rsk) is a distal member of the mitogen-activated protein kinase signaling pathway. It has been cloned from a variety of species including Xenopus laevis, mouse, chicken, rat, and human. The clone p90(rsk-mo-1), isolated by others from a mouse library, contains a unique 33-nucleotide deletion not found in the p90(rsk) clones from any other species that have been examined. When p90(rsk-mo-1) was expressed in Cos-7 cells that were subsequently stimulated with epidermal growth factor, the immunoprecipitated p90(rsk-mo-1) protein showed no measurable kinase activity toward the ribosomal protein S6 peptide. By comparison, expression of rat p90(rsk-1) resulted in significant kinase activity. Deletion of the 33-nucleotide region missing in the p90(rsk-mo-1) clone from the p90(rsk-rat-1) cDNA abolished kinase activity in the resulting protein. When these 33 nucleotides were introduced into the p90(rsk-mo-1) cDNA, the expressed protein showed significant kinase activity. Reverse transcription-PCR and direct sequencing of mRNA isolated from several mouse tissues indicated the presence of the full-length form of p90(rsk-1) in the mouse and showed no conclusive evidence for a deletion-containing form. This study indicates the presence of a full-length p90(rsk-1) mRNA in mouse tissues that is homologous to that identified in other species and suggests that the deletion in p90(rsk-mo-1) may be a cloning artifact. The findings provide additional support for the conclusion that the first catalytic domain of p90(rsk) is responsible for its enzymatic activity toward ribosomal protein S6.
- Published
- 1999
- Full Text
- View/download PDF
30. The G protein gamma subunit. Requirements for dimerization with beta subunits.
- Author
-
Mende U, Schmidt CJ, Yi F, Spring DJ, and Neer EJ
- Subjects
- Amino Acid Sequence, Animals, Epitopes, GTP-Binding Proteins biosynthesis, GTP-Binding Proteins immunology, Molecular Sequence Data, Precipitin Tests, Rabbits, Reticulocytes metabolism, GTP-Binding Proteins chemistry
- Abstract
Guanine nucleotide-binding protein beta and gamma subunits form a tightly bound complex that can only be separated by denaturation. Assembly of beta and gamma subunits is a complicated process. The beta 1 and gamma 2 subunits can be synthesized in vitro in rabbit reticulocyte lysate and then assembled into dimers, but beta 1 cannot form beta gamma dimers when synthesized in a wheat germ extract. In contrast, gamma 2 translated in either system can dimerize with beta 1, suggesting that dimerization-competent gamma 2 can be synthesized without the aid of specific chaperonins or other cofactors. Dimerization-competent gamma 2 in solution forms an asymmetric particle with a Stokes radius of about 21 +/- 0.4 A (n = 4), s20,w of 0.9 S (range 0.8-1.0 S, n = 2), and frictional ratio of 1.57 (assuming no hydration). To define the part of gamma 2 that is needed for native beta gamma dimer formation, a series of N- and C-terminal truncations were generated, synthesized in vitro, and incubated with beta 1. Dimerization was assessed by stabilization of beta 1 to tryptic proteolysis. Truncation of up to 13 amino acids at the C terminus did not affect dimerization with beta 1, whereas removal of 27 amino acids prevented it. Therefore, a region between residues 45 and 59 of gamma 2 is important for dimerization. Truncation of 15 amino acids from the N terminus greatly diminished the formation of beta gamma dimers, while removal of 25 amino acids entirely blocked it. Thus, another region important for forming native beta gamma is near the N terminus. Extension of the N terminus by 12 amino acids that include the influenza virus hemagglutinin epitope did not prevent beta gamma dimerization. Furthermore, in intact 35S-labeled COS cells, epitope-tagged gamma 2 coimmunoprecipitates with beta and alpha subunits. The N-terminal epitope tag must lie at the surface of the heterotrimer since it prevents neither heterotrimer formation nor access of the antibody.
- Published
- 1995
- Full Text
- View/download PDF
31. A 14-amino acid region of the G protein gamma subunit is sufficient to confer selectivity of gamma binding to the beta subunit.
- Author
-
Spring DJ and Neer EJ
- Subjects
- Amino Acid Sequence, Animals, Binding Sites, Brain metabolism, Cattle, GTP-Binding Proteins biosynthesis, GTP-Binding Proteins chemistry, Macromolecular Substances, Molecular Sequence Data, Peptide Fragments chemistry, Polymerase Chain Reaction, Recombinant Fusion Proteins biosynthesis, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins metabolism, Restriction Mapping, Signal Transduction, Transducin biosynthesis, Transducin chemistry, Trypsin, GTP-Binding Proteins metabolism, Peptide Fragments metabolism, Transducin metabolism
- Abstract
Heterotrimeric guanine nucleotide-binding proteins are important signaling molecules composed of an alpha, beta, and gamma subunit. The beta subunits must form dimers with gamma subunits to function. Several subtypes of beta and gamma have been identified, but not all combinations of beta and gamma subtypes can form dimers. For example, the gamma 2 subunit can form dimers with beta 1 and beta 2, but gamma 1 forms dimers only with beta 1, not with beta 2. Selective dimerization may play a role in the regulation of beta gamma dimer-mediated signal transduction. In order to identify the region of gamma responsible for selective dimer formation, a series of gamma 1/gamma 2 chimeras was constructed, transcribed, and translated in vitro. The ability of these gamma chimeras to form dimers with beta 1 and beta 2 was assayed by trypsin protection and chemical cross-linking. When amino acids 36-49 of gamma 1 were substituted for 33-46 of gamma 2, the chimera behaved like gamma 1 and dimerized only with beta 1; the reciprocal chimera, in which 14 residues from gamma 2 were substituted for the corresponding amino acids of gamma 1, behaved like gamma 2 and interacted with both beta 1 and beta 2. This 14-amino acid region was sufficient for gamma 1 to discriminate between the beta subunits. All gamma chimeras were functional because they were able to interact with beta 1, which is capable of forming dimers with both gamma 1 and gamma 2. All dimers of chimeric gamma subunits plus beta 1 were able to interact with purified alpha o subunit, indicating that beta gamma dimers containing chimeric gamma molecules were capable of interacting with an appropriate third molecule. This lays the foundation for using these gamma chimeras to study selective dimer interactions with various effectors and receptors.
- Published
- 1994
32. Lipoprotein assembly. Apolipoprotein B size determines lipoprotein core circumference.
- Author
-
Spring DJ, Chen-Liu LW, Chatterton JE, Elovson J, and Schumaker VN
- Subjects
- Apolipoproteins B genetics, Carcinoma, Hepatocellular metabolism, Electrophoresis, Polyacrylamide Gel, Humans, Lipase metabolism, Lipoproteins ultrastructure, Liver enzymology, Liver Neoplasms metabolism, Microscopy, Electron, Molecular Weight, Plasmids, Puromycin pharmacology, Transfection, Tumor Cells, Cultured, Apolipoproteins B chemistry, Lipoproteins chemistry
- Abstract
Apolipoprotein B (apoB) is an essential structural protein for the two triglyceride-rich lipoproteins synthesized by humans: chylomicrons and very low density lipoproteins. Although much is known about the role of apoB in clearance of lipoproteins from the circulation, relatively little is known about its role in the assembly of nascent lipoproteins. Therefore, we have investigated the relationship between the length of various N-terminal apoB fragments and the characteristics of the lipoproteins with which these fragments were associated. After the addition of puromycin, HepG2 cells secreted a discrete series of C-terminally truncated apoB fragments on lipoprotein particles including apoB25, apoB29, apoB31, apoB33, apoB36, apoB38, apoB42, apoB45, apoB49, apoB51, apoB55, apoB70, and apoB80. Also, using plasmids encoding apoB26, apoB33, apoB37, apoB42, and apoB48, C-terminally truncated apoB fragments were expressed and secreted after transient transfection of HepG2 cells. Lipoproteins bearing the metabolically labeled apoB fragments were isolated from the cell culture media and characterized in terms of size, density, flotation coefficient, and composition. Lipoprotein radii, calculated from their flotation coefficients and buoyant densities, were used to derive the circumference of the non-polar core of each lipoprotein species. When plotted as a function of apoB size, core circumference defined a straight line of near-zero intercept. The slope of this line was approximately 1 A of core circumference/1 kDa of apoB molecular mass. A model for the mechanism of lipoprotein assembly in HepG2 cells, consistent with the concept that apoB size determines lipoprotein core circumference, is proposed.
- Published
- 1992
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.