651
|
Bayne LJ, Beatty GL, Jhala N, Clark CE, Rhim AD, Stanger BZ, Vonderheide RH. Tumor-derived granulocyte-macrophage colony-stimulating factor regulates myeloid inflammation and T cell immunity in pancreatic cancer. Cancer Cell 2012; 21:822-35. [PMID: 22698406 PMCID: PMC3575028 DOI: 10.1016/j.ccr.2012.04.025] [Citation(s) in RCA: 736] [Impact Index Per Article: 56.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Revised: 02/04/2012] [Accepted: 04/09/2012] [Indexed: 02/07/2023]
Abstract
Cancer-associated inflammation is thought to be a barrier to immune surveillance, particularly in pancreatic ductal adenocarcinoma (PDA). Gr-1(+) CD11b(+) cells are a key feature of cancer inflammation in PDA, but remain poorly understood. Using a genetically engineered mouse model of PDA, we show that tumor-derived granulocyte-macrophage colony-stimulating factor (GM-CSF) is necessary and sufficient to drive the development of Gr-1(+) CD11b(+) cells that suppressed antigen-specific T cells. In vivo, abrogation of tumor-derived GM-CSF inhibited the recruitment of Gr-1(+) CD11b(+) cells to the tumor microenvironment and blocked tumor development-a finding that was dependent on CD8(+) T cells. In humans, PDA tumor cells prominently expressed GM-CSF in vivo. Thus, tumor-derived GM-CSF is an important regulator of inflammation and immune suppression within the tumor microenvironment.
Collapse
Affiliation(s)
- Lauren J. Bayne
- Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Gregory L. Beatty
- Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
- Division of Hematology-Oncology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Nirag Jhala
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Carolyn E. Clark
- Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Andrew D. Rhim
- Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
- Division of Gastroenterology, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Ben Z. Stanger
- Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
- Abramson Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
- Division of Gastroenterology, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Robert H. Vonderheide
- Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
- Abramson Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
- Division of Hematology-Oncology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| |
Collapse
|
652
|
Ihara S, Kida H, Arase H, Tripathi LP, Chen YA, Kimura T, Yoshida M, Kashiwa Y, Hirata H, Fukamizu R, Inoue R, Hasegawa K, Goya S, Takahashi R, Minami T, Tsujino K, Suzuki M, Kohmo S, Inoue K, Nagatomo I, Takeda Y, Kijima T, Mizuguchi K, Tachibana I, Kumanogoh A. Inhibitory Roles of Signal Transducer and Activator of Transcription 3 in Antitumor Immunity during Carcinogen-Induced Lung Tumorigenesis. Cancer Res 2012; 72:2990-9. [DOI: 10.1158/0008-5472.can-11-4062] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
653
|
Deng J, Liu Y, Lee H, Herrmann A, Zhang W, Zhang C, Shen S, Priceman SJ, Kujawski M, Pal SK, Raubitschek A, Hoon DSB, Forman S, Figlin RA, Liu J, Jove R, Yu H. S1PR1-STAT3 signaling is crucial for myeloid cell colonization at future metastatic sites. Cancer Cell 2012; 21:642-654. [PMID: 22624714 PMCID: PMC3360884 DOI: 10.1016/j.ccr.2012.03.039] [Citation(s) in RCA: 206] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Revised: 11/28/2011] [Accepted: 03/05/2012] [Indexed: 12/20/2022]
Abstract
Recent studies underscore the importance of myeloid cells in rendering distant organs hospitable for disseminating tumor cells to colonize. However, what enables myeloid cells to have an apparently superior capacity to colonize distant organs is unclear. Here, we show that S1PR1-STAT3 upregulation in tumor cells induces factors that activate S1PR1-STAT3 in various cells in premetastatic sites, leading to premetastatic niche formation. Targeting either S1PR1 or STAT3 in myeloid cells disrupts existing premetastatic niches. S1PR1-STAT3 pathway enables myeloid cells to intravasate, prime the distant organ microenvironment and mediate sustained proliferation and survival of their own and other stromal cells at future metastatic sites. Analyzing tumor-free lymph nodes from cancer patients shows elevated myeloid infiltrates, STAT3 activity, and increased survival signal.
Collapse
Affiliation(s)
- Jiehui Deng
- Department of Cancer Immunotherapeutics and Tumor Immunology, Beckman Research Institute and City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Yong Liu
- Department of Cancer Immunotherapeutics and Tumor Immunology, Beckman Research Institute and City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Heehyoung Lee
- Department of Cancer Immunotherapeutics and Tumor Immunology, Beckman Research Institute and City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Andreas Herrmann
- Department of Cancer Immunotherapeutics and Tumor Immunology, Beckman Research Institute and City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Wang Zhang
- Department of Cancer Immunotherapeutics and Tumor Immunology, Beckman Research Institute and City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Chunyan Zhang
- Department of Cancer Immunotherapeutics and Tumor Immunology, Beckman Research Institute and City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Shudan Shen
- Department of Cancer Immunotherapeutics and Tumor Immunology, Beckman Research Institute and City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Saul J Priceman
- Department of Cancer Immunotherapeutics and Tumor Immunology, Beckman Research Institute and City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Maciej Kujawski
- Department of Cancer Immunotherapeutics and Tumor Immunology, Beckman Research Institute and City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Sumanta K Pal
- Department of Medical Oncology, Beckman Research Institute and City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Andrew Raubitschek
- Department of Cancer Immunotherapeutics and Tumor Immunology, Beckman Research Institute and City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Dave S B Hoon
- Department of Molecular Oncology, John Wayne Cancer Institute, Santa Monica, CA 90404, USA
| | - Stephen Forman
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute and City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Robert A Figlin
- Department of Hematology-Oncology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jie Liu
- Department of Digestive Diseases of Huashan Hospital, Department of Immunology of Shanghai Medical School, Fudan University, Shanghai, 200040, China; Center for Translational Medicine, Zhangjiang High-Tech Park, Shanghai, 201203, China
| | - Richard Jove
- Department of Molecular Medicine, Beckman Research Institute and City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Hua Yu
- Department of Cancer Immunotherapeutics and Tumor Immunology, Beckman Research Institute and City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; Center for Translational Medicine, Zhangjiang High-Tech Park, Shanghai, 201203, China.
| |
Collapse
|
654
|
Tudor CS, Dawson CW, Eckhardt J, Niedobitek G, Büttner AC, Seliger B, Hartmann A, Buettner M. c-Myc and EBV-LMP1: two opposing regulators of the HLA class I antigen presentation machinery in epithelial cells. Br J Cancer 2012; 106:1980-8. [PMID: 22588558 PMCID: PMC3388564 DOI: 10.1038/bjc.2012.197] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background: Epstein–Barr virus (EBV)-encoded latent membrane protein 1 (LMP1) up-regulates the human leukocyte antigen (HLA) class I antigen presentation machinery (APM). This appears counterintuitive with immune evasion in EBV-associated tumours like nasopharyngeal carcinoma (NPC). Methods: Latent membrane protein 1-transfected epithelial cell lines were used as a model system to study the impact of LMP1 and c-Myc on HLA class I components. The expression of components of the HLA class I APM, c-Myc and Ki-67 was analysed in LMP1+ and LMP1− NPC by immunohistochemistry. Results: In epithelial cells, LMP1 up-regulated HLA class I APM. This effect could be counteracted by c-Myc, which itself was up-regulated by LMP1 apparently through IL6 induction and Jak3/STAT3 activation. Studies of NPC biopsies revealed down-regulation of HLA class I APM expression. No difference was observed between LMP1+ and LMP1− NPC. However, expression of Ki-67 and c-Myc were up-regulated in LMP1+ tumours. Conclusion: These findings raise the possibility that c-Myc activation in NPC might antagonise the effect of LMP1 on HLA class I expression thus contributing to immune escape of tumour cells.
Collapse
Affiliation(s)
- C S Tudor
- Institute of Pathology, Department of Nephropathology, Friedrich-Alexander-University, Erlangen-Nuremberg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
655
|
Pérez-Mancera PA, Guerra C, Barbacid M, Tuveson DA. What we have learned about pancreatic cancer from mouse models. Gastroenterology 2012; 142:1079-92. [PMID: 22406637 DOI: 10.1053/j.gastro.2012.03.002] [Citation(s) in RCA: 132] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Revised: 02/29/2012] [Accepted: 03/06/2012] [Indexed: 12/15/2022]
Affiliation(s)
- Pedro A Pérez-Mancera
- Li Ka Shing Centre, Cambridge Research Institute, and Department of Oncology, Cancer Research UK, Cambridge, England
| | | | | | | |
Collapse
|
656
|
The expression of the receptor for advanced glycation endproducts (RAGE) is permissive for early pancreatic neoplasia. Proc Natl Acad Sci U S A 2012; 109:7031-6. [PMID: 22509024 DOI: 10.1073/pnas.1113865109] [Citation(s) in RCA: 120] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Pancreatic cancer is an almost uniformly lethal disease, characterized by late diagnosis, early metastasis, resistance to chemotherapy, and early mutation of the Kras oncogene. Here we show that the receptor for advanced glycation endproducts (RAGE) is required for the activation of interleukin 6 (IL-6)-mediated mitochondrial signal transducers and activators of transcription 3 (STAT3) signaling in pancreatic carcinogenesis. RAGE expression correlates with elevated levels of autophagy in pancreatic cancer in vivo and in vitro, and this heightened state of autophagy is required for IL-6-induced STAT3 activation. To further explore the intersection of RAGE, autophagy, and pancreatic carcinogenesis, we created a transgenic murine model, backcrossing RAGE-null mice to a spontaneous mouse model of pancreatic cancer, Pdx1-Cre:Kras(G12D/+) (KC). Targeted ablation of Rage in KC mice delayed neoplasia development, decreased levels of autophagy, and inhibited mitochondrial STAT3 activity and subsequent ATP production. Our results suggest a critical role for RAGE expression in the earliest stages of pancreatic carcinogenesis, potentially acting as the "autophagic switch," regulating mitochondrial STAT3 signaling.
Collapse
|
657
|
Abstract
Myeloid cells are the most abundant nucleated haematopoietic cells in the human body and are a collection of distinct cell populations with many diverse functions. The three groups of terminally differentiated myeloid cells - macrophages, dendritic cells and granulocytes - are essential for the normal function of both the innate and adaptive immune systems. Mounting evidence indicates that the tumour microenvironment alters myeloid cells and can convert them into potent immunosuppressive cells. Here, we consider myeloid cells as an intricately connected, complex, single system and we focus on how tumours manipulate the myeloid system to evade the host immune response.
Collapse
|
658
|
Pini M, Rhodes DH, Castellanos KJ, Hall AR, Cabay RJ, Chennuri R, Grady EF, Fantuzzi G. Role of IL-6 in the resolution of pancreatitis in obese mice. J Leukoc Biol 2012; 91:957-66. [PMID: 22427681 DOI: 10.1189/jlb.1211627] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Obesity increases severity of acute pancreatitis and risk of pancreatic cancer. Pancreatitis and obesity are associated with elevated IL-6, a cytokine involved in inflammation and tumorigenesis. We studied the role of IL-6 in the response of lean and obese mice to pancreatitis induced by IL-12 + IL-18. Lean and diet-induced obese (DIO) WT and IL-6 KO mice and ob/ob mice pretreated with anti-IL-6 antibodies were evaluated at Days 1, 7, and 15 after induction of pancreatitis. Prolonged elevation of IL-6 in serum and visceral adipose tissue was observed in DIO versus lean WT mice, whereas circulating sIL-6R declined in DIO but not lean mice with pancreatitis. The severe inflammation and lethality of DIO mice were also observed in IL-6 KO mice. However, the delayed resolution of neutrophil infiltration; sustained production of CXCL1, CXCL2, and CCL2; prolonged activation of STAT-3; and induction of MMP-7 in the pancreas, as well as heightened induction of serum amylase A of DIO mice, were blunted significantly in DIO IL-6 KO mice. In DIO mice, production of OPN and TIMP-1 was increased for a prolonged period, and this was mediated by IL-6 in the liver but not the pancreas. Results obtained in IL-6 KO mice were confirmed in ob/ob mice pretreated with anti-IL-6 antibodies. In conclusion, IL-6 does not contribute to the increased severity of pancreatitis of obese mice but participates in delayed recovery from acute inflammation and may favor development of a protumorigenic environment through prolonged activation of STAT-3, induction of MMP-7, and sustained production of chemokines.
Collapse
Affiliation(s)
- Maria Pini
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL, USA
| | | | | | | | | | | | | | | |
Collapse
|
659
|
Allavena P, Mantovani A. Immunology in the clinic review series; focus on cancer: tumour-associated macrophages: undisputed stars of the inflammatory tumour microenvironment. Clin Exp Immunol 2012; 167:195-205. [PMID: 22235995 DOI: 10.1111/j.1365-2249.2011.04515.x] [Citation(s) in RCA: 285] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mononuclear phagocytes are cells of the innate immunity that defend the host against harmful pathogens and heal tissues after injury. Contrary to expectations, in malignancies, tumour-associated macrophages (TAM) promote disease progression by supporting cancer cell survival, proliferation and invasion. TAM and related myeloid cells [Tie2(+) monocytes and myeloid-derived suppressor cells (MDSC)] also promote tumour angiogenesis and suppress adaptive immune responses. These divergent biological activities are mediated by macrophages/myeloid cells with distinct functional polarization, which are ultimately dictated by microenvironmental cues. Clinical and experimental evidence has shown that cancer tissues with high infiltration of TAM are associated with poor patient prognosis and resistance to therapies. Targeting of macrophages in tumours is considered a promising therapeutic strategy: depletion of TAM or their 're-education' as anti-tumour effectors is under clinical investigation and will hopefully contribute to the success of conventional anti-cancer treatments.
Collapse
Affiliation(s)
- P Allavena
- Department of Immunology and Inflammation, IRCCS Humanitas Clinical Institute Department of Translational Medicine, University of Milan, Milan, Rozzano, Italy.
| | | |
Collapse
|
660
|
Sansone P, Bromberg J. Targeting the interleukin-6/Jak/stat pathway in human malignancies. J Clin Oncol 2012; 30:1005-14. [PMID: 22355058 DOI: 10.1200/jco.2010.31.8907] [Citation(s) in RCA: 407] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The Janus kinase/signal transducer and activator of transcription (Jak/Stat) pathway was discovered 20 years ago as a mediator of cytokine signaling. Since this time, more than 2,500 articles have been published demonstrating the importance of this pathway in virtually all malignancies. Although there are dozens of cytokines and cytokine receptors, four Jaks, and seven Stats, it seems that interleukin-6-mediated activation of Stat3 is a principal pathway implicated in promoting tumorigenesis. This transcription factor regulates the expression of numerous critical mediators of tumor formation and metastatic progression. This review will examine the relative importance and function of this pathway in nonmalignant conditions as well as malignancies (including tumor intrinsic and extrinsic), the influence of other Stats, the development of inhibitors to this pathway, and the potential role of inhibitors in controlling or eradicating cancers.
Collapse
Affiliation(s)
- Pasquale Sansone
- Memorial Sloan-Kettering Cancer Center, Weill Cornell Medical College, New York, NY 10021, USA
| | | |
Collapse
|
661
|
Quante M, Bhagat G, Abrams J, Marache F, Good P, Lee MD, Lee Y, Friedman R, Asfaha S, Dubeykovskaya Z, Mahmood U, Figueiredo JL, Kitajewski J, Shawber C, Lightdale C, Rustgi AK, Wang TC. Bile acid and inflammation activate gastric cardia stem cells in a mouse model of Barrett-like metaplasia. Cancer Cell 2012; 21:36-51. [PMID: 22264787 PMCID: PMC3266546 DOI: 10.1016/j.ccr.2011.12.004] [Citation(s) in RCA: 369] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Revised: 06/02/2011] [Accepted: 12/01/2011] [Indexed: 02/06/2023]
Abstract
Esophageal adenocarcinoma (EAC) arises from Barrett esophagus (BE), intestinal-like columnar metaplasia linked to reflux esophagitis. In a transgenic mouse model of BE, esophageal overexpression of interleukin-1β phenocopies human pathology with evolution of esophagitis, Barrett-like metaplasia and EAC. Histopathology and gene signatures closely resembled human BE, with upregulation of TFF2, Bmp4, Cdx2, Notch1, and IL-6. The development of BE and EAC was accelerated by exposure to bile acids and/or nitrosamines, and inhibited by IL-6 deficiency. Lgr5(+) gastric cardia stem cells present in BE were able to lineage trace the early BE lesion. Our data suggest that BE and EAC arise from gastric progenitors due to a tumor-promoting IL-1β-IL-6 signaling cascade and Dll1-dependent Notch signaling.
Collapse
Affiliation(s)
- Michael Quante
- Division of Digestive and Liver Diseases, Irving Cancer Research Center, Department of Medicine, Columbia University Medical Center, New York, NY
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675 München
- Corresponding authors: Timothy C. Wang, M.D., Division of Digestive and Liver Diseases, Columbia University Medical Center, 1130 St. Nicholas Avenue, Room 925, 9th Floor; New York, NY 10032, Phone: (212) 851-4581; Fax: (212) 851-4590; . Michael Quante, M.D., II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675 München, Phone: +49 89 4140 6795; Fax: +49 89 4140 6796;
| | - Govind Bhagat
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY
| | - Julian Abrams
- Division of Digestive and Liver Diseases, Irving Cancer Research Center, Department of Medicine, Columbia University Medical Center, New York, NY
| | - Frederic Marache
- Division of Digestive and Liver Diseases, Irving Cancer Research Center, Department of Medicine, Columbia University Medical Center, New York, NY
| | - Pamela Good
- Division of Digestive and Liver Diseases, Irving Cancer Research Center, Department of Medicine, Columbia University Medical Center, New York, NY
| | - Michele D. Lee
- Division of Digestive and Liver Diseases, Irving Cancer Research Center, Department of Medicine, Columbia University Medical Center, New York, NY
| | - Yoomi Lee
- Division of Digestive and Liver Diseases, Irving Cancer Research Center, Department of Medicine, Columbia University Medical Center, New York, NY
| | - Richard Friedman
- Department of Biomedical Informatics, Columbia University Medical Center, New York, NY
| | - Samuel Asfaha
- Division of Digestive and Liver Diseases, Irving Cancer Research Center, Department of Medicine, Columbia University Medical Center, New York, NY
| | - Zinaida Dubeykovskaya
- Division of Digestive and Liver Diseases, Irving Cancer Research Center, Department of Medicine, Columbia University Medical Center, New York, NY
| | - Umar Mahmood
- Nuclear Medicine & Molecular Imaging, Harvard Medical School and Massachusetts General Hospital, Boston, MA
| | - Jose-Luiz Figueiredo
- Center for Systems Biology, Harvard Medical School and Massachusetts General Hospital, Boston, MA
| | - Jan Kitajewski
- Pathology, Obstetrics and Gynecology, and Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Carrie Shawber
- Pathology, Obstetrics and Gynecology, and Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Charles Lightdale
- Division of Digestive and Liver Diseases, Irving Cancer Research Center, Department of Medicine, Columbia University Medical Center, New York, NY
| | - Anil K. Rustgi
- Division of Gastroenterology, Department of Medicine and Genetics, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Timothy C. Wang
- Division of Digestive and Liver Diseases, Irving Cancer Research Center, Department of Medicine, Columbia University Medical Center, New York, NY
- Corresponding authors: Timothy C. Wang, M.D., Division of Digestive and Liver Diseases, Columbia University Medical Center, 1130 St. Nicholas Avenue, Room 925, 9th Floor; New York, NY 10032, Phone: (212) 851-4581; Fax: (212) 851-4590; . Michael Quante, M.D., II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675 München, Phone: +49 89 4140 6795; Fax: +49 89 4140 6796;
| |
Collapse
|
662
|
Yoneyama T, Angata K, Bao X, Courtneidge S, Chanda SK, Fukuda M. Fer kinase regulates cell migration through α-dystroglycan glycosylation. Mol Biol Cell 2012; 23:771-80. [PMID: 22238358 PMCID: PMC3290637 DOI: 10.1091/mbc.e11-06-0517] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
This is the first report on the role of Fer kinase in down-regulating the expression of laminin-binding glycans that suppress cell migration. The data show a novel biochemical interaction between glycan-based adhesion and cell migration, mediated by a tyrosine kinase. Glycans of α-dystroglycan (α-DG), which is expressed at the epithelial cell–basement membrane (BM) interface, play an essential role in epithelium development and tissue organization. Laminin-binding glycans on α-DG expressed on cancer cells suppress tumor progression by attenuating tumor cell migration from the BM. However, mechanisms controlling laminin-binding glycan expression are not known. Here, we used small interfering RNA (siRNA) library screening and identified Fer kinase, a non–receptor-type tyrosine kinase, as a key regulator of laminin-binding glycan expression. Fer overexpression decreased laminin-binding glycan expression, whereas siRNA-mediated down-regulation of Fer kinase increased glycan expression on breast and prostate cancer cell lines. Loss of Fer kinase function via siRNA or mutagenesis increased transcription levels of glycosyltransferases, including protein O-mannosyltransferase 1, β3-N-acetylglucosaminyltransferase 1, and like-acetylglucosaminyltransferase that are required to synthesize laminin-binding glycans. Consistently, inhibition of Fer expression decreased cell migration in the presence of laminin fragment. Fer kinase regulated STAT3 phosphorylation and consequent activation, whereas knockdown of STAT3 increased laminin-binding glycan expression on cancer cells. These results indicate that the Fer pathway negatively controls expression of genes required to synthesize laminin-binding glycans, thus impairing BM attachment and increasing tumor cell migration.
Collapse
Affiliation(s)
- Tohru Yoneyama
- Glycobiology Unit, Tumor Microenvironment Program, Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, USA
| | | | | | | | | | | |
Collapse
|
663
|
Collins MA, Bednar F, Zhang Y, Brisset JC, Galbán S, Galbán CJ, Rakshit S, Flannagan KS, Adsay NV, Pasca di Magliano M. Oncogenic Kras is required for both the initiation and maintenance of pancreatic cancer in mice. J Clin Invest 2012; 122:639-53. [PMID: 22232209 DOI: 10.1172/jci59227] [Citation(s) in RCA: 585] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Accepted: 11/16/2011] [Indexed: 01/03/2023] Open
Abstract
Pancreatic cancer is almost invariably associated with mutations in the KRAS gene, most commonly KRASG12D, that result in a dominant-active form of the KRAS GTPase. However, how KRAS mutations promote pancreatic carcinogenesis is not fully understood, and whether oncogenic KRAS is required for the maintenance of pancreatic cancer has not been established. To address these questions, we generated two mouse models of pancreatic tumorigenesis: mice transgenic for inducible KrasG12D, which allows for inducible, pancreas-specific, and reversible expression of the oncogenic KrasG12D, with or without inactivation of one allele of the tumor suppressor gene p53. Here, we report that, early in tumorigenesis, induction of oncogenic KrasG12D reversibly altered normal epithelial differentiation following tissue damage, leading to precancerous lesions. Inactivation of KrasG12D in established precursor lesions and during progression to cancer led to regression of the lesions, indicating that KrasG12D was required for tumor cell survival. Strikingly, during all stages of carcinogenesis, KrasG12D upregulated Hedgehog signaling, inflammatory pathways, and several pathways known to mediate paracrine interactions between epithelial cells and their surrounding microenvironment, thus promoting formation and maintenance of the fibroinflammatory stroma that plays a pivotal role in pancreatic cancer. Our data establish that epithelial KrasG12D influences multiple cell types to drive pancreatic tumorigenesis and is essential for tumor maintenance. They also strongly support the notion that inhibiting KrasG12D, or its downstream effectors, could provide a new approach for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Meredith A Collins
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
664
|
Meyer C, Eydeler K, Magbanua E, Zivkovic T, Piganeau N, Lorenzen I, Grötzinger J, Mayer G, Rose-John S, Hahn U. Interleukin-6 receptor specific RNA aptamers for cargo delivery into target cells. RNA Biol 2012; 9:67-80. [PMID: 22258147 PMCID: PMC3342945 DOI: 10.4161/rna.9.1.18062] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Aptamers represent an emerging strategy to deliver cargo molecules, including dyes, drugs, proteins or even genes, into specific target cells. Upon binding to specific cell surface receptors aptamers can be internalized, for example by macropinocytosis or receptor mediated endocytosis. Here we report the in vitro selection and characterization of RNA aptamers with high affinity (Kd = 20 nM) and specificity for the human IL-6 receptor (IL-6R). Importantly, these aptamers trigger uptake without compromising the interaction of IL-6R with its natural ligands the cytokine IL-6 and glycoprotein 130 (gp130). We further optimized the aptamers to obtain a shortened, only 19-nt RNA oligonucleotide retaining all necessary characteristics for high affinity and selective recognition of IL-6R on cell surfaces. Upon incubation with IL-6R presenting cells this aptamer was rapidly internalized. Importantly, we could use our aptamer, to deliver bulky cargos, exemplified by fluorescently labeled streptavidin, into IL-6R presenting cells, thereby setting the stage for an aptamer-mediated escort of drug molecules to diseased cell populations or tissues.
Collapse
Affiliation(s)
- Cindy Meyer
- Institute for Biochemistry and Molecular Biology; Chemistry Department; MIN-Faculty; Hamburg University; Hamburg, Germany
| | - Katja Eydeler
- Institute for Biochemistry and Molecular Biology; Chemistry Department; MIN-Faculty; Hamburg University; Hamburg, Germany
| | - Eileen Magbanua
- Institute for Biochemistry and Molecular Biology; Chemistry Department; MIN-Faculty; Hamburg University; Hamburg, Germany
| | - Tijana Zivkovic
- Institute for Biochemistry and Molecular Biology; Chemistry Department; MIN-Faculty; Hamburg University; Hamburg, Germany
| | - Nicolas Piganeau
- Institute for Biochemistry and Molecular Biology; Chemistry Department; MIN-Faculty; Hamburg University; Hamburg, Germany
| | - Inken Lorenzen
- Institute of Biochemistry; Medical Faculty; Christian-Albrechts-University; Kiel, Germany
| | - Joachim Grötzinger
- Institute of Biochemistry; Medical Faculty; Christian-Albrechts-University; Kiel, Germany
| | - Günter Mayer
- Life and Medical Sciences Institute; University of Bonn; Bonn, Germany
| | - Stefan Rose-John
- Institute of Biochemistry; Medical Faculty; Christian-Albrechts-University; Kiel, Germany
| | - Ulrich Hahn
- Institute for Biochemistry and Molecular Biology; Chemistry Department; MIN-Faculty; Hamburg University; Hamburg, Germany
| |
Collapse
|
665
|
Tumor promotion via injury- and death-induced inflammation. Immunity 2011; 35:467-77. [PMID: 22035839 DOI: 10.1016/j.immuni.2011.09.006] [Citation(s) in RCA: 208] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Revised: 09/03/2011] [Accepted: 09/06/2011] [Indexed: 12/20/2022]
Abstract
Inhibition of programmed cell death is considered to be a major aspect of tumorigenesis. Indeed, several key oncogenic transcription factors, such as NF-κB and STAT3, exert their tumor-promoting activity at least in part through upregulation of survival genes. However, many cancers develop in response to chronic tissue injury, in which the resulting cell death increases the tumorigenic potential of the neighboring cells. In this review, we discuss a resolution to this paradox based on cell death-mediated induction of tumor promoting inflammatory cytokines, which enhance cell survival and trigger compensatory proliferation in response to tissue injury.
Collapse
|
666
|
Tai WT, Cheng AL, Shiau CW, Liu CY, Ko CH, Lin MW, Chen PJ, Chen KF. Dovitinib induces apoptosis and overcomes sorafenib resistance in hepatocellular carcinoma through SHP-1-mediated inhibition of STAT3. Mol Cancer Ther 2011; 11:452-63. [PMID: 22180308 DOI: 10.1158/1535-7163.mct-11-0412] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The multiple kinase inhibitor dovitinib is currently under clinical investigation for hepatocellular carcinoma (HCC). Here, we investigated the mechanistic basis for the effects of dovitinib in HCCs. Dovitinib showed significant antitumor activity in HCC cell lines PLC5, Hep3B, Sk-Hep1, and Huh-7. Dovitinib downregulated phospho-STAT3 (p-STAT3) at tyrosine 705 and subsequently reduced the levels of expression of STAT3-related proteins Mcl-1, survivin, and cyclin D1 in a time-dependent manner. Ectopic expression of STAT3 abolished the apoptotic effect of dovitinib, indicating that STAT3 is indispensable in mediating the effect of dovitinib in HCC. SHP-1 inhibitor reversed downregulation of p-STAT3 and apoptosis induced by dovitinib, and silencing of SHP-1 by RNA interference abolished the effects of dovitinib on p-STAT3, indicating that SHP-1, a protein tyrosine phosphatase, mediates the effects of dovitinib. Notably, dovitinib increased SHP-1 activity in HCC cells. Incubation of dovitinib with pure SHP-1 protein enhanced its phosphatase activity, indicating that dovitinib upregulates the activity of SHP-1 via direct interactions. In addition, dovitinib induced apoptosis in two sorafenib-resistant cell lines through inhibition of STAT3, and sorafenib-resistant cells showed significant activation of STAT3, suggesting that targeting STAT3 may be a useful approach to overcome drug resistance in HCC. Finally, in vivo, dovitinib significantly suppressed growth of both Huh-7 and PLC5 xenograft tumors and downregulated p-STAT3 by increasing SHP-1 activity. In conclusion, dovitinib induces significant apoptosis in HCC cells and sorafenib-resistant cells via SHP-1-mediated inhibition of STAT3.
Collapse
Affiliation(s)
- Wei-Tien Tai
- Department of Medical Research, National Taiwan University Hospital, Chung-Shan S Rd, Taipei 100, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
667
|
Bednar F, Simeone DM. Pancreatic cancer stem cell biology and its therapeutic implications. J Gastroenterol 2011; 46:1345-52. [PMID: 22048257 DOI: 10.1007/s00535-011-0494-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Accepted: 10/08/2011] [Indexed: 02/04/2023]
Abstract
Pancreatic cancer remains one of the most difficult malignancies to treat. Significant developments in our understanding of pancreatic cancer biology have occurred over the past decade. One of the key advances has been the formulation of the cancer stem cell model of tumor growth and subsequent experimental proof of pancreatic cancer stem cell existence. Cancer stem cells contribute to pancreatic tumor growth and progression and are at least partially responsible for the relative resistance of the tumor to systemic chemotherapy and radiation. Significant questions remain about how the mutational profile of the tumor, the tumor microenvironment, and normal pancreatic developmental pathways contribute to pancreatic cancer stem cell biology. Answers to these questions will likely yield new therapeutic approaches for this deadly disease.
Collapse
Affiliation(s)
- Filip Bednar
- Department of Surgery, Box 5343, University of Michigan Medical Center, 1500 E. Medical Center Dr., Ann Arbor, MI 48109, USA
| | | |
Collapse
|
668
|
Reichert M, Rustgi AK. Pancreatic ductal cells in development, regeneration, and neoplasia. J Clin Invest 2011. [PMID: 22133881 DOI: 10.1172/jci57131.4572] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The pancreas is a complex organ comprised of three critical cell lineages: islet (endocrine), acinar, and ductal. This review will focus upon recent insights and advances in the biology of pancreatic ductal cells. In particular, emphasis will be placed upon the regulation of ductal cells by specific transcriptional factors during development as well as the underpinnings of acinar-ductal metaplasia as an important adaptive response during injury and regeneration. We also address the potential contributions of ductal cells to neoplastic transformation, specifically in pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Maximilian Reichert
- Division of Gastroenterology, Department of Medicine, Abramson Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
669
|
Reichert M, Rustgi AK. Pancreatic ductal cells in development, regeneration, and neoplasia. J Clin Invest 2011; 121:4572-8. [PMID: 22133881 DOI: 10.1172/jci57131] [Citation(s) in RCA: 164] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The pancreas is a complex organ comprised of three critical cell lineages: islet (endocrine), acinar, and ductal. This review will focus upon recent insights and advances in the biology of pancreatic ductal cells. In particular, emphasis will be placed upon the regulation of ductal cells by specific transcriptional factors during development as well as the underpinnings of acinar-ductal metaplasia as an important adaptive response during injury and regeneration. We also address the potential contributions of ductal cells to neoplastic transformation, specifically in pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Maximilian Reichert
- Division of Gastroenterology, Department of Medicine, Abramson Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
670
|
Acute pancreatitis accelerates initiation and progression to pancreatic cancer in mice expressing oncogenic Kras in the nestin cell lineage. PLoS One 2011; 6:e27725. [PMID: 22140463 PMCID: PMC3225359 DOI: 10.1371/journal.pone.0027725] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Accepted: 10/23/2011] [Indexed: 12/27/2022] Open
Abstract
Targeting of oncogenic Kras to the pancreatic Nestin-expressing embryonic progenitor cells and subsequently to the adult acinar compartment and Nestin-expressing cells is sufficient for the development of low grade pancreatic intraepithelial neoplasia (PanIN) between 2 and 4 months. The mice die around 6 month-old of unrelated causes, and it is therefore not possible to assess whether the lesions will progress to carcinoma. We now report that two brief episodes of caerulein-induced acute pancreatitis in 2 month-old mice causes rapid PanIN progression and pancreatic ductal adenocarcinoma (PDAC) development by 4 months of age. These events occur with similar frequency as observed in animals where the oncogene is targeted during embryogenesis to all pancreatic cell types. Thus, these data show that oncogenic Kras-driven PanIN originating in a non-ductal compartment can rapidly progress to PDAC when subjected to a brief inflammatory insult.
Collapse
|
671
|
Zhang WN, Wang L, Wang Q, Luo X, Fang DF, Chen Y, Pan X, Man JH, Xia Q, Jin BF, Li WH, Li T, Liang B, Chen L, Gong WL, Yu M, Li AL, Zhou T, Li HY. CUEDC2 (CUE domain-containing 2) and SOCS3 (suppressors of cytokine signaling 3) cooperate to negatively regulate Janus kinase 1/signal transducers and activators of transcription 3 signaling. J Biol Chem 2011; 287:382-392. [PMID: 22084247 DOI: 10.1074/jbc.m111.276832] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Janus kinase 1/signal transducers and activators of transcription 3 (JAK1/STAT3) pathway is one of the recognized oncogenic signaling pathways that frequently overactivated in a variety of human tumors. Despite rapid progress in elucidating the molecular mechanisms of activation of JAK/STAT pathway, the processes that regulate JAK/STAT deactivation need to be further clarified. Here we demonstrate that CUE domain-containing 2 (CUEDC2) inhibits cytokine-induced phosphorylation of JAK1 and STAT3 and the subsequent STAT3 transcriptional activity. Further analysis by a yeast two-hybrid assay showed that CUEDC2 could engage in a specific interaction with a key JAK/STAT inhibitor, SOCS3 (suppressors of cytokine signaling 3). The interaction between CUEDC2 and SOCS3 is required for the inhibitory effect of CUEDC2 on JAK1 and STAT3 activity. Additionally, we found CUEDC2 functions collaboratively with SOCS3 to inhibit JAK1/STAT3 signaling by increasing SOCS3 stability via enhancing its association with Elongin C. Therefore, our findings revealed a new biological activity for CUEDC2 as the regulator of JAK1/STAT3 signaling and paved the way to a better understanding of the mechanisms by which SOCS3 has been linked to suppression of the JAK/STAT pathway.
Collapse
Affiliation(s)
- Wei-Na Zhang
- Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing 100850, China
| | - Li Wang
- Beijing Institute of Biotechnology, Beijing 100071, China
| | - Qiong Wang
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Xue Luo
- Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing 100850, China
| | - Di-Feng Fang
- Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing 100850, China
| | - Yuan Chen
- Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing 100850, China
| | - Xin Pan
- Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing 100850, China
| | - Jiang-Hong Man
- Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing 100850, China
| | - Qing Xia
- Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing 100850, China
| | - Bao-Feng Jin
- Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing 100850, China
| | - Wei-Hua Li
- Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing 100850, China
| | - Tao Li
- Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing 100850, China
| | - Bing Liang
- Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing 100850, China
| | - Liang Chen
- Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing 100850, China
| | - Wei-Li Gong
- Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing 100850, China
| | - Ming Yu
- Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing 100850, China
| | - Ai-Ling Li
- Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing 100850, China
| | - Tao Zhou
- Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing 100850, China.
| | - Hui-Yan Li
- Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing 100850, China.
| |
Collapse
|
672
|
An intermediate-risk multiple myeloma subgroup is defined by sIL-6r: levels synergistically increase with incidence of SNP rs2228145 and 1q21 amplification. Blood 2011; 119:503-12. [PMID: 22072558 DOI: 10.1182/blood-2011-07-367052] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
IL-6 signaling can be enhanced through transsignaling by the soluble IL-6 receptor (sIL-6r), allowing for the pleiotropic cytokine to affect cells it would not ordinarily have an effect on. Serum levels of sIL-6r can be used as an independent prognostic indicator and further stratify the GEP 70-gene low-risk group to identify an intermediate-risk group in multiple myeloma (MM). By analyzing more than 600 MM patients with ELISA, genotyping, and gene expression profiling tools, we show how the combination of 2 independent molecular genetic events is related to synergistic increases in sIL-6r levels. We also show that the rs2228145 minor allele is related to increased expression levels of an IL-6r splice variant that purportedly codes exclusively for a sIL-6r isoform. Together, the SNP rs2228145 minor allele C and amplification of chromosome 1q21 are significantly correlated to an increase in sIL-6r levels, which are associated with lower overall survival in 70-gene low-risk disease, and aid in identification of the intermediate-risk MM group.
Collapse
|
673
|
Gierut J, Zheng Y, Bie W, Carroll RE, Ball-Kell S, Haegebarth A, Tyner AL. Disruption of the mouse protein tyrosine kinase 6 gene prevents STAT3 activation and confers resistance to azoxymethane. Gastroenterology 2011; 141:1371-80, 1380.e1-2. [PMID: 21741923 PMCID: PMC3448944 DOI: 10.1053/j.gastro.2011.06.071] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Revised: 06/18/2011] [Accepted: 06/24/2011] [Indexed: 01/05/2023]
Abstract
BACKGROUND & AIMS Protein tyrosine kinase 6 (PTK6) is expressed throughout the gastrointestinal tract and is a negative regulator of proliferation that promotes differentiation and DNA-damage-induced apoptosis in the small intestine. PTK6 is not expressed in normal mammary gland, but is induced in most human breast tumors. Signal transducer and activator of transcription 3 (STAT3) mediates pathogenesis of colon cancer and is a substrate of PTK6. We investigated the role of PTK6 in colon tumorigenesis. METHODS Ptk6+/+ and Ptk6-/- mice were injected with azoxymethane alone or in combination with dextran sodium sulfate; formation of aberrant crypt foci and colon tumors was examined. Effects of disruption of Ptk6 on proliferation, apoptosis, and STAT3 activation were examined by immunoblot and immunohistochemical analyses. Regulation of STAT3 activation was examined in the HCT116 colon cancer cell line and young adult mouse colon cells. RESULTS Ptk6-/- mice developed fewer azoxymethane-induced aberrant crypt foci and tumors. Induction of PTK6 increased apoptosis, proliferation, and STAT3 activation in Ptk6+/+ mice injected with azoxymethane. Disruption of Ptk6 impaired STAT3 activation following azoxymethane injection, and reduced active STAT3 levels in Ptk6-/- tumors. Stable knockdown of PTK6 reduced basal levels of active STAT3, as well as activation of STAT3 by epidermal growth factor in HCT116 cells. Disruption of Ptk6 reduced activity of STAT3 in young adult mouse colon cells. CONCLUSIONS PTK6 promotes STAT3 activation in the colon following injection of the carcinogen azoxymethane and regulates STAT3 activity in mouse colon tumors and in the HCT116 and young adult mouse colon cell lines. Disruption of Ptk6 decreases azoxymethane-induced colon tumorigenesis in mice.
Collapse
Affiliation(s)
- Jessica Gierut
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607
| | - Yu Zheng
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607
| | - Wenjun Bie
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607
| | - Robert E. Carroll
- Department of Medicine, University of Illinois at Chicago, Chicago, IL 60607
| | - Susan Ball-Kell
- University of Illinois College of Veterinary Medicine Veterinary Diagnostic Laboratory Urbana, IL 61802
| | - Andrea Haegebarth
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607
| | - Angela L. Tyner
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607,Department of Medicine, University of Illinois at Chicago, Chicago, IL 60607,Corresponding author: University of Illinois College of Medicine, Department of Biochemistry and Molecular Genetics, M/C 669, 900 South Ashland Avenue, Chicago, Illinois 60607, Phone: 312-996-7964, Fax. 312-413-4892,
| |
Collapse
|
674
|
Rose-John S. PL2-2. Molecular Switch between Pro- and Anti-Inflammatory Cytokine Activities. Cytokine 2011. [DOI: 10.1016/j.cyto.2011.07.296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
675
|
Fisher DT, Chen Q, Skitzki JJ, Muhitch JB, Zhou L, Appenheimer MM, Vardam TD, Weis EL, Passanese J, Wang WC, Gollnick SO, Dewhirst MW, Rose-John S, Repasky EA, Baumann H, Evans SS. IL-6 trans-signaling licenses mouse and human tumor microvascular gateways for trafficking of cytotoxic T cells. J Clin Invest 2011; 121:3846-59. [PMID: 21926464 DOI: 10.1172/jci44952] [Citation(s) in RCA: 192] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2010] [Accepted: 08/03/2011] [Indexed: 12/26/2022] Open
Abstract
Immune cells are key regulators of neoplastic progression, which is often mediated through their release of cytokines. Inflammatory cytokines such as IL-6 exert tumor-promoting activities by driving growth and survival of neoplastic cells. However, whether these cytokines also have a role in recruiting mediators of adaptive anticancer immunity has not been investigated. Here, we report that homeostatic trafficking of tumor-reactive CD8+ T cells across microvascular checkpoints is limited in tumors despite the presence of inflammatory cytokines. Intravital imaging in tumor-bearing mice revealed that systemic thermal therapy (core temperature elevated to 39.5°C ± 0.5°C for 6 hours) activated an IL-6 trans-signaling program in the tumor blood vessels that modified the vasculature such that it could support enhanced trafficking of CD8+ effector/memory T cells (Tems) into tumors. A concomitant decrease in tumor infiltration by Tregs during systemic thermal therapy resulted in substantial enhancement of Tem/Treg ratios. Mechanistically, IL-6 produced by nonhematopoietic stromal cells acted cooperatively with soluble IL-6 receptor-α and thermally induced gp130 to promote E/P-selectin- and ICAM-1-dependent extravasation of cytotoxic T cells in tumors. Parallel increases in vascular adhesion were induced by IL-6/soluble IL-6 receptor-α fusion protein in mouse tumors and patient tumor explants. Finally, a causal link was established between IL-6-dependent licensing of tumor vessels for Tem trafficking and apoptosis of tumor targets. These findings suggest that the unique IL-6-rich tumor microenvironment can be exploited to create a therapeutic window to boost T cell-mediated antitumor immunity and immunotherapy.
Collapse
Affiliation(s)
- Daniel T Fisher
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
676
|
Jones SA, Scheller J, Rose-John S. Therapeutic strategies for the clinical blockade of IL-6/gp130 signaling. J Clin Invest 2011; 121:3375-83. [PMID: 21881215 PMCID: PMC3163962 DOI: 10.1172/jci57158] [Citation(s) in RCA: 544] [Impact Index Per Article: 38.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The successful treatment of certain autoimmune conditions with the humanized anti-IL-6 receptor (IL-6R) antibody tocilizumab has emphasized the clinical importance of cytokines that signal through the β-receptor subunit glycoprotein 130 (gp130). In this Review, we explore how gp130 signaling controls disease progression and examine why IL-6 has a special role among these cytokines as an inflammatory regulator. Attention will be given to the role of the soluble IL-6R, and we will provide a perspective into the clinical blockade of IL-6 activity in autoimmunity, inflammation, and cancer.
Collapse
Affiliation(s)
- Simon A. Jones
- Department of Infection, Immunity and Biochemistry, The School of Medicine, Cardiff University, Cardiff, United Kingdom.
Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany.
Institute of Biochemistry, Christian-Albrechts-University, Kiel, Germany
| | - Jürgen Scheller
- Department of Infection, Immunity and Biochemistry, The School of Medicine, Cardiff University, Cardiff, United Kingdom.
Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany.
Institute of Biochemistry, Christian-Albrechts-University, Kiel, Germany
| | - Stefan Rose-John
- Department of Infection, Immunity and Biochemistry, The School of Medicine, Cardiff University, Cardiff, United Kingdom.
Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany.
Institute of Biochemistry, Christian-Albrechts-University, Kiel, Germany
| |
Collapse
|
677
|
Rai TS, Adams PD. Lessons from senescence: Chromatin maintenance in non-proliferating cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2011; 1819:322-31. [PMID: 21839870 DOI: 10.1016/j.bbagrm.2011.07.014] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 07/25/2011] [Accepted: 07/27/2011] [Indexed: 02/08/2023]
Abstract
Cellular senescence is an irreversible proliferation arrest, thought to contribute to tumor suppression, proper wound healing and, perhaps, tissue and organismal aging. Two classical tumor suppressors, p53 and pRB, control cell cycle arrest associated with senescence. Profound molecular changes occur in cells undergoing senescence. At the level of chromatin, for example, senescence associated heterochromatic foci (SAHF) form in some cell types. Chromatin is inherently dynamic and likely needs to be actively maintained to achieve a stable cell phenotype. In proliferating cells chromatin is maintained in conjunction with DNA replication, but how non-proliferating cells maintain chromatin structure is poorly understood. Some histone variants, such as H3.3 and macroH2A increase as cells undergo senescence, suggesting histone variants and their associated chaperones could be important in chromatin structure maintenance in senescent cells. Here, we discuss options available for senescent cells to maintain chromatin structure and the relative contribution of histone variants and chaperones in this process. This article is part of a Special Issue entitled: Histone chaperones and chromatin assembly.
Collapse
|
678
|
Li N, Grivennikov SI, Karin M. The unholy trinity: inflammation, cytokines, and STAT3 shape the cancer microenvironment. Cancer Cell 2011; 19:429-31. [PMID: 21481782 PMCID: PMC3111086 DOI: 10.1016/j.ccr.2011.03.018] [Citation(s) in RCA: 199] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Tumor-associated inflammation is a consequence and a driver of tumorigenesis. Three papers in this issue of Cancer Cell demonstrate the importance of tumor-elicited inflammation in the development and progression of pancreatic ductal adenocarcinoma and esophageal squamous carcinoma. Disruption of tissue homeostasis culminates in activation of STAT3, generating a pro-tumorigenic inflammatory microenvironment.
Collapse
Affiliation(s)
- Ning Li
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | | | | |
Collapse
|
679
|
|