1
|
Fnu G, Weber GF. Targeting the core program of metastasis with a novel drug combination. Cancer Med 2024; 13:e7291. [PMID: 38826119 PMCID: PMC11145026 DOI: 10.1002/cam4.7291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 04/29/2024] [Accepted: 05/04/2024] [Indexed: 06/04/2024] Open
Abstract
BACKGROUND We previously reported that metastases are generally characterized by a core program of gene expression that activates tissue remodeling/vascularization, alters ion homeostasis, induces the oxidative metabolism, and silences extracellular matrix interactions. This core program distinguishes metastases from their originating primary tumors as well as from their destination host tissues. Therefore, the gene products involved are potential targets for anti-metastasis drug treatment. METHODS Because the silencing of extracellular matrix interactions predisposes to anoiks in the absence of active survival mechanisms, we tested inhibitors against the other three components. RESULTS Individually, the low-specificity VEGFR blocker pazopanib (in vivo combined with marimastat), the antioxidant dimethyl sulfoxide (or the substitute atovaquone, which is approved for internal administration), and the ionic modulators bumetanide and tetrathiomolybdate inhibited soft agar colony formation by breast and pancreatic cancer cell lines. The individual candidate agents have a record of use in humans (with limited efficacy when administered individually) and are available for repurposing. In combination, the effects of these drugs were additive or synergistic. In two mouse models of cancer (utilizing 4T1 cells or B16-F10 cells), the combination treatment with these medications, applied immediately (to prevent metastasis formation) or after a delay (to suppress established metastases), dramatically reduced the occurrence of disseminated foci. CONCLUSIONS The combination of tissue remodeling inhibitors, suppressors of the oxidative metabolism, and ion homeostasis modulators has very strong promise for the treatment of metastases by multiple cancers.
Collapse
Affiliation(s)
- Gulimirerouzi Fnu
- James L. Winkle College of PharmacyUniversity of Cincinnati Academic Health CenterCincinnatiOhioUSA
| | - Georg F. Weber
- James L. Winkle College of PharmacyUniversity of Cincinnati Academic Health CenterCincinnatiOhioUSA
| |
Collapse
|
2
|
Fnu G, Weber GF. Osteopontin induces mitochondrial biogenesis in deadherent cancer cells. Oncotarget 2023; 14:957-969. [PMID: 38039408 PMCID: PMC10691814 DOI: 10.18632/oncotarget.28540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/16/2023] [Indexed: 12/03/2023] Open
Abstract
Metastasizing cells display a unique metabolism, which is very different from the Warburg effect that arises in primary tumors. Over short time frames, oxidative phosphorylation and ATP generation are prominent. Over longer time frames, mitochondrial biogenesis becomes a pronounced feature and aids metastatic success. It has not been known whether or how these two phenomena are connected. We hypothesized that Osteopontin splice variants, which synergize to increase ATP levels in deadherent cells, also increase the mitochondrial mass via the same signaling mechanisms. Here, we report that autocrine Osteopontin does indeed stimulate an increase in mitochondrial size, with the splice variant -c being more effective than the full-length form -a. Osteopontin-c achieves this via its receptor CD44v, jointly with the upregulation and co-ligation of the chloride-dependent cystine-glutamate transporter SLC7A11. The signaling proceeds through activation of the known mitochondrial biogenesis inducer PGC-1 (which acts as a transcription coactivator). Peroxide is an important intermediate in this cascade, but surprisingly acts upstream of PGC-1 and is likely produced as a consequence of SLC7A11 recruitment and activation. In vivo, suppression of the biogenesis-inducing mechanisms leads to a reduction in disseminated tumor mass. This study confirms a functional connection between the short-term oxidative metabolism and the longer-term mitochondrial biogenesis in cancer metastasis - both are induced by Osteopontin-c. The results imply possible mechanisms and targets for treating cancer metastasis.
Collapse
Affiliation(s)
- Gulimirerouzi Fnu
- University of Cincinnati Academic Health Center, James L. Winkle College of Pharmacy, Cincinnati, OH 45229, USA
| | - Georg F. Weber
- University of Cincinnati Academic Health Center, James L. Winkle College of Pharmacy, Cincinnati, OH 45229, USA
| |
Collapse
|
3
|
Oh A, Pardo M, Rodriguez A, Yu C, Nguyen L, Liang O, Chorzalska A, Dubielecka PM. NF-κB signaling in neoplastic transition from epithelial to mesenchymal phenotype. Cell Commun Signal 2023; 21:291. [PMID: 37853467 PMCID: PMC10585759 DOI: 10.1186/s12964-023-01207-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 06/25/2023] [Indexed: 10/20/2023] Open
Abstract
NF-κB transcription factors are critical regulators of innate and adaptive immunity and major mediators of inflammatory signaling. The NF-κB signaling is dysregulated in a significant number of cancers and drives malignant transformation through maintenance of constitutive pro-survival signaling and downregulation of apoptosis. Overactive NF-κB signaling results in overexpression of pro-inflammatory cytokines, chemokines and/or growth factors leading to accumulation of proliferative signals together with activation of innate and select adaptive immune cells. This state of chronic inflammation is now thought to be linked to induction of malignant transformation, angiogenesis, metastasis, subversion of adaptive immunity, and therapy resistance. Moreover, accumulating evidence indicates the involvement of NF-κB signaling in induction and maintenance of invasive phenotypes linked to epithelial to mesenchymal transition (EMT) and metastasis. In this review we summarize reported links of NF-κB signaling to sequential steps of transition from epithelial to mesenchymal phenotypes. Understanding the involvement of NF-κB in EMT regulation may contribute to formulating optimized therapeutic strategies in cancer. Video Abstract.
Collapse
Affiliation(s)
- Amy Oh
- Division of Hematology/Oncology, Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, One Hoppin St., Coro West, Suite 5.01, RI, 02903, Providence, USA
| | - Makayla Pardo
- Division of Hematology/Oncology, Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, One Hoppin St., Coro West, Suite 5.01, RI, 02903, Providence, USA
| | - Anaelena Rodriguez
- Division of Hematology/Oncology, Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, One Hoppin St., Coro West, Suite 5.01, RI, 02903, Providence, USA
| | - Connie Yu
- Division of Hematology/Oncology, Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, One Hoppin St., Coro West, Suite 5.01, RI, 02903, Providence, USA
| | - Lisa Nguyen
- Division of Hematology/Oncology, Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, One Hoppin St., Coro West, Suite 5.01, RI, 02903, Providence, USA
| | - Olin Liang
- Division of Hematology/Oncology, Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, One Hoppin St., Coro West, Suite 5.01, RI, 02903, Providence, USA
| | - Anna Chorzalska
- Division of Hematology/Oncology, Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, One Hoppin St., Coro West, Suite 5.01, RI, 02903, Providence, USA
| | - Patrycja M Dubielecka
- Division of Hematology/Oncology, Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, One Hoppin St., Coro West, Suite 5.01, RI, 02903, Providence, USA.
| |
Collapse
|
4
|
Fnu G, Weber GF. Alterations of Ion Homeostasis in Cancer Metastasis: Implications for Treatment. Front Oncol 2022; 11:765329. [PMID: 34988012 PMCID: PMC8721045 DOI: 10.3389/fonc.2021.765329] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 11/23/2021] [Indexed: 12/20/2022] Open
Abstract
We have previously reported that metastases from all malignancies are characterized by a core program of gene expression that suppresses extracellular matrix interactions, induces vascularization/tissue remodeling, activates the oxidative metabolism, and alters ion homeostasis. Among these features, the least elucidated component is ion homeostasis. Here we review the literature with the goal to infer a better mechanistic understanding of the progression-associated ionic alterations and identify the most promising drugs for treatment. Cancer metastasis is accompanied by skewing in calcium, zinc, copper, potassium, sodium and chloride homeostasis. Membrane potential changes and water uptake through Aquaporins may also play roles. Drug candidates to reverse these alterations are at various stages of testing, with some having entered clinical trials. Challenges to their utilization comprise differences among tumor types and the involvement of multiple ions in each case. Further, adverse effects may become a concern, as channel blockers, chelators, or supplemented ions will affect healthy and transformed cells alike.
Collapse
Affiliation(s)
- Gulimirerouzi Fnu
- College of Pharmacy, University of Cincinnati Academic Health Center, Cincinnati, OH, United States
| | - Georg F Weber
- College of Pharmacy, University of Cincinnati Academic Health Center, Cincinnati, OH, United States
| |
Collapse
|
5
|
Hinz N, Baranowsky A, Horn M, Kriegs M, Sibbertsen F, Smit DJ, Clezardin P, Lange T, Schinke T, Jücker M. Knockdown of AKT3 Activates HER2 and DDR Kinases in Bone-Seeking Breast Cancer Cells, Promotes Metastasis In Vivo and Attenuates the TGFβ/CTGF Axis. Cells 2021; 10:cells10020430. [PMID: 33670586 PMCID: PMC7922044 DOI: 10.3390/cells10020430] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/12/2021] [Accepted: 02/16/2021] [Indexed: 12/12/2022] Open
Abstract
Bone metastases frequently occur in breast cancer patients and lack appropriate treatment options. Hence, understanding the molecular mechanisms involved in the multistep process of breast cancer bone metastasis and tumor-induced osteolysis is of paramount interest. The serine/threonine kinase AKT plays a crucial role in breast cancer bone metastasis but the effect of individual AKT isoforms remains unclear. Therefore, AKT isoform-specific knockdowns were generated on the bone-seeking MDA-MB-231 BO subline and the effect on proliferation, migration, invasion, and chemotaxis was analyzed by live-cell imaging. Kinome profiling and Western blot analysis of the TGFβ/CTGF axis were conducted and metastasis was evaluated by intracardiac inoculation of tumor cells into NOD scid gamma (NSG) mice. MDA-MB-231 BO cells exhibited an elevated AKT3 kinase activity in vitro and responded to combined treatment with AKT- and mTOR-inhibitors. Knockdown of AKT3 significantly increased migration, invasion, and chemotaxis in vitro and metastasis to bone but did not significantly enhance osteolysis. Furthermore, knockdown of AKT3 increased the activity and phosphorylation of pro-metastatic HER2 and DDR1/2 but lowered protein levels of CTGF after TGFβ-stimulation, an axis involved in tumor-induced osteolysis. We demonstrated that AKT3 plays a crucial role in bone-seeking breast cancer cells by promoting metastatic potential without facilitating tumor-induced osteolysis.
Collapse
Affiliation(s)
- Nico Hinz
- Center for Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.H.); (F.S.); (D.J.S.)
| | - Anke Baranowsky
- Center for Experimental Medicine, Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (A.B.); (T.S.)
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Michael Horn
- University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
- Mildred Scheel Cancer Career Center Hamburg, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Malte Kriegs
- Department of Radiotherapy & Radiation Oncology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
- UCCH Kinomics Core Facility, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Freya Sibbertsen
- Center for Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.H.); (F.S.); (D.J.S.)
| | - Daniel J. Smit
- Center for Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.H.); (F.S.); (D.J.S.)
| | - Philippe Clezardin
- INSERM, Research Unit UMR S1033, LyOS, Faculty of Medicine Lyon-Est, University of Lyon 1, 69372 Lyon, France;
| | - Tobias Lange
- Center for Experimental Medicine, Department of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
| | - Thorsten Schinke
- Center for Experimental Medicine, Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (A.B.); (T.S.)
| | - Manfred Jücker
- Center for Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.H.); (F.S.); (D.J.S.)
- Correspondence: ; Tel.: +49-(0)-40-7410-56339
| |
Collapse
|
6
|
Hartung F, Patil A, Meshram RJ, Weber GF. Gene expression signatures of site-specificity in cancer metastases. Clin Exp Metastasis 2019; 37:159-171. [PMID: 31555944 DOI: 10.1007/s10585-019-09995-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 09/19/2019] [Indexed: 11/26/2022]
Abstract
We have previously shown that metastases are generally characterized by a core program of gene expression that induces the oxidative energy metabolism, activates vascularization/tissue remodeling, silences extracellular matrix interactions, and alters ion homeostasis. This core program distinguishes metastases from their originating primary tumors as well as from their target host tissues. We hypothesized that organ preference is reflected in additional, site-selective components within the metastatic gene expression programs. Expanding our prior analysis of 653 human gene expression profiles plus data from a murine model, we find that the release from the primary tumor is associated with a suppression of functions that are important for the identity of the organ of origin, such as a down-regulation of steroid hormone responsiveness in the disseminated foci derived from prostate cancer. Metastases adjust to their target microenvironment by up-regulating-even overexpressing-genes and genetic programs that are characteristic of that organ. Finally, alterations in RNA and protein processing as well as immune deviation are common. In the clinic, metastases are mostly treated with the chemotherapy protocols devised for their primary tumors. Adjustments that account for the gene expression differences between primary and metastatic cancers have the potential to improve the currently dismal success rates.
Collapse
Affiliation(s)
- Franz Hartung
- University of Cincinnati Academic Health Center, Cincinnati, OH, USA
| | - Aditya Patil
- Bioinformatics Centre, Savitribai Phule Pune University, Pune, Maharashtra, India
| | - Rohan J Meshram
- Bioinformatics Centre, Savitribai Phule Pune University, Pune, Maharashtra, India
| | - Georg F Weber
- University of Cincinnati Academic Health Center, Cincinnati, OH, USA.
- James L. Winkle College of Pharmacy, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, 45267-0514, USA.
| |
Collapse
|
7
|
Lamort AS, Giopanou I, Psallidas I, Stathopoulos GT. Osteopontin as a Link between Inflammation and Cancer: The Thorax in the Spotlight. Cells 2019; 8:cells8080815. [PMID: 31382483 PMCID: PMC6721491 DOI: 10.3390/cells8080815] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/25/2019] [Accepted: 07/31/2019] [Indexed: 12/23/2022] Open
Abstract
The glycoprotein osteopontin (OPN) possesses multiple functions in health and disease. To this end, osteopontin has beneficial roles in wound healing, bone homeostasis, and extracellular matrix (ECM) function. On the contrary, osteopontin can be deleterious for the human body during disease. Indeed, osteopontin is a cardinal mediator of tumor-associated inflammation and facilitates metastasis. The purpose of this review is to highlight the importance of osteopontin in malignant processes, focusing on lung and pleural tumors as examples.
Collapse
Affiliation(s)
- Anne-Sophie Lamort
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, University Hospital, Ludwig-Maximilians University of Munich and Helmholtz Center Munich, Member of the German Center for Lung Research, Max-Lebsche-Platz 31, 81377 Munich, Bavaria, Germany.
| | - Ioanna Giopanou
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Biomedical Sciences Research Center, 1 Asklepiou Str., University Campus, 26504 Rio, Achaia, Greece
| | - Ioannis Psallidas
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E6BT, UK
| | - Georgios T Stathopoulos
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, University Hospital, Ludwig-Maximilians University of Munich and Helmholtz Center Munich, Member of the German Center for Lung Research, Max-Lebsche-Platz 31, 81377 Munich, Bavaria, Germany.
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Biomedical Sciences Research Center, 1 Asklepiou Str., University Campus, 26504 Rio, Achaia, Greece.
| |
Collapse
|
8
|
Xue L, Maihle NJ, Yu X, Tang SC, Liu HY. Synergistic Targeting HER2 and EGFR with Bivalent Aptamer-siRNA Chimera Efficiently Inhibits HER2-Positive Tumor Growth. Mol Pharm 2018; 15:4801-4813. [PMID: 30222359 PMCID: PMC6220360 DOI: 10.1021/acs.molpharmaceut.8b00388] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
![]()
HER2 overexpression
is identified on 20–30% breast cancer
and other cancers at different levels. Although HER2 targeted monoclonal
antibody combined with chemical drugs has shown improved outcomes
in HER2 expressing patients, drug resistance and toxicity have limited
their efficacy. To overcome drug resistance, cotargeting multiple
HER receptors was proven to be effective. EGFR/HER2 dimerization can
active PI3K/AKT pathway, and resistance to HER2-targeted drugs is
associated with upregulation of EGFR. Here, we developed a novel HER2/EGFR
targeted nucleic acid therapeutic to address current drug limits.
The new therapeutic is constructed by fusing HER2 aptamer-EGFR siRNA
sense strand with HER2 aptamer-EGFR siRNA antisense strand into one
molecule: a bivalent HER2 aptamer-EGFR siRNA aptamer chimera (HEH).
In breast cancer cell lines, HEH can be selectively taken up into
HER2 expressing cells and successfully silence EGFR gene and down
regulate HER2 expression. In breast cancer xenograft models, HEH is
capable of triggering cell apoptosis, decreasing HER2 and EGFR expression,
and suppressing tumor growth. The therapeutic efficacy of HEH is superior
to HER2 aptamer only, which suggests that HEH has synergistic effect
by targeting HER2 and EGFR. This study demonstrated that HEH has great
potential as a new HER2 targeted drug to address toxicity and resistance
of current drugs and may provide a cure for many HER2 positive cancers.
Collapse
Affiliation(s)
- Lu Xue
- Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Medical College of Georgia , Augusta University , Augusta , Georgia 30912 , United States.,Department of Pediatrics Hematology , The First Hospital of Jilin University , Changchun 130021 , China
| | - Nita J Maihle
- Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Medical College of Georgia , Augusta University , Augusta , Georgia 30912 , United States
| | - Xiaolin Yu
- Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Medical College of Georgia , Augusta University , Augusta , Georgia 30912 , United States
| | - Shou-Ching Tang
- University of Mississippi Medical Center Cancer Institute , Jackson , Mississippi 39216 , United States
| | - Hong Yan Liu
- Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Medical College of Georgia , Augusta University , Augusta , Georgia 30912 , United States
| |
Collapse
|
9
|
Wynn ML, Egbert M, Consul N, Chang J, Wu ZF, Meravjer SD, Schnell S. Inferring Intracellular Signal Transduction Circuitry from Molecular Perturbation Experiments. Bull Math Biol 2017; 80:1310-1344. [PMID: 28455685 DOI: 10.1007/s11538-017-0270-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 03/15/2017] [Indexed: 12/28/2022]
Abstract
The development of network inference methodologies that accurately predict connectivity in dysregulated pathways may enable the rational selection of patient therapies. Accurately inferring an intracellular network from data remains a very challenging problem in molecular systems biology. Living cells integrate extremely robust circuits that exhibit significant heterogeneity, but still respond to external stimuli in predictable ways. This phenomenon allows us to introduce a network inference methodology that integrates measurements of protein activation from perturbation experiments. The methodology relies on logic-based networks to provide a predictive approximation of the transfer of signals in a network. The approach presented was validated in silico with a set of test networks and applied to investigate the epidermal growth factor receptor signaling of a breast epithelial cell line, MFC10A. In our analysis, we predict the potential signaling circuitry most likely responsible for the experimental readouts of several proteins in the mitogen-activated protein kinase and phosphatidylinositol-3 kinase pathways. The approach can also be used to identify additional necessary perturbation experiments to distinguish between a set of possible candidate networks.
Collapse
Affiliation(s)
- Michelle L Wynn
- Division of Hematology & Oncology and Comprehensive Cancer Center, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Computational Medicine & Bioinformatics, and Brehm Center for Diabetes Research, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Megan Egbert
- Division of Hematology & Oncology and Comprehensive Cancer Center, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Nikita Consul
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
- Columbia University College of Physicians & Surgeons, New York, NY, USA
| | - Jungsoo Chang
- Division of Hematology & Oncology and Comprehensive Cancer Center, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Zhi-Fen Wu
- Division of Hematology & Oncology and Comprehensive Cancer Center, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Sofia D Meravjer
- Division of Hematology & Oncology and Comprehensive Cancer Center, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Santiago Schnell
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Department of Computational Medicine & Bioinformatics, and Brehm Center for Diabetes Research, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
10
|
Ramchandani D, Unruh D, Lewis CS, Bogdanov VY, Weber GF. Activation of carbonic anhydrase IX by alternatively spliced tissue factor under late-stage tumor conditions. J Transl Med 2016; 96:1234-1245. [PMID: 27721473 PMCID: PMC5121009 DOI: 10.1038/labinvest.2016.103] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 08/12/2016] [Accepted: 09/06/2016] [Indexed: 02/07/2023] Open
Abstract
Molecules of the coagulation pathway predispose patients to cancer-associated thrombosis and also trigger intracellular signaling pathways that promote cancer progression. The primary transcript of tissue factor, the main physiologic trigger of blood clotting, can undergo alternative splicing yielding a secreted variant, termed asTF (alternatively spliced tissue factor). asTF is not required for normal hemostasis, but its expression levels positively correlate with advanced tumor stages in several cancers, including pancreatic adenocarcinoma. The asTF-overexpressing pancreatic ductal adenocarcinoma cell line Pt45.P1/asTF+ and its parent cell line Pt45.P1 were tested for growth and mobility under normoxic conditions that model early-stage tumors, and in the hypoxic environment of late-stage cancers. asTF overexpression in Pt45.P1 cells conveys increased proliferative ability. According to cell cycle analysis, the major fraction of Pt45.P1/asTF+ cells reside in the dividing G2/M phase of the cell cycle, whereas the parental Pt45.P1 cells are mostly confined to the quiescent G0/G1 phase. asTF overexpression is also associated with significantly higher mobility in cells plated under either normoxia or hypoxia. A hypoxic environment leads to upregulation of carbonic anhydrase IX (CAIX), which is more pronounced in Pt45.P1/asTF+ cells. Inhibition of CAIX by the compound U-104 significantly decreases cell growth and mobility of Pt45.P1/asTF+ cells in hypoxia, but not in normoxia. U-104 also reduces the growth of Pt45.P1/asTF+ orthotopic tumors in nude mice. CAIX is a novel downstream mediator of asTF in pancreatic cancer, particularly under hypoxic conditions that model late-stage tumor microenvironment.
Collapse
Affiliation(s)
| | | | | | - Vladimir Y. Bogdanov
- College of Medicine, University of Cincinnati,address correspondence to either: Georg F. Weber, College of Pharmacy, University of Cincinnati, 3225 Eden Avenue, Cincinnati, OH 45267-0004. , phone 513-558-0947 or : Vladimir Y. Bogdanov, College of Medicine, University of Cincinnati, OH 45267, USA.
| | - Georg F. Weber
- James L. Winkle College of Pharmacy, University of Cincinnati,address correspondence to either: Georg F. Weber, College of Pharmacy, University of Cincinnati, 3225 Eden Avenue, Cincinnati, OH 45267-0004. , phone 513-558-0947 or : Vladimir Y. Bogdanov, College of Medicine, University of Cincinnati, OH 45267, USA.
| |
Collapse
|
11
|
Kaempferol inhibits the production of ROS to modulate OPN–αvβ3 integrin pathway in HUVECs. J Physiol Biochem 2016; 72:303-13. [DOI: 10.1007/s13105-016-0479-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 03/08/2016] [Indexed: 12/22/2022]
|
12
|
Kuwabara Y, Katayama A, Tomiyama R, Piao H, Kurihara S, Ono S, Mine K, Akira S, Orimo H, Takeshita T. Gonadotropin regulation and role of ovarian osteopontin in the periovulatory period. J Endocrinol 2015; 224:49-59. [PMID: 25352284 DOI: 10.1530/joe-14-0203] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Osteopontin (OPN), a secreted glycoprotein, has multiple physiological functions. This study investigated the regulation and roles of OPN in the mouse ovary during the periovulatory stages. Immature female mice were treated with pregnant mare serum gonadotropin (PMSG) and human chorionic gonadotropin (hCG) to simulate follicle maturation and ovulation. In situ hybridization and real-time RT-PCR were performed to assess expression of Opn in the periovulatory ovary. Granulosa cells (GCs) from PMSG-primed immature mice were cultured with or without hCG in the presence or absence of OPN, and effects on expression of Opn, progesterone synthesis, and vascular endothelial growth factor (VEGF) signaling were assessed by real-time RT-PCR, ELISA, and western blotting analysis. Opn transcripts were significantly upregulated 3 h after hCG treatment, followed by a peak at 16 h, and the transcripts localized to GCs. Incubation with hCG significantly increased quantities of Opn transcripts in GCs and OPN levels in the culture medium at 12 and 24 h. Furthermore, OPN treatment caused a significant increase in the levels of Star protein, P 450 cholesterol side-chain cleavage enzyme (p450scc), 3-beta-hydroxysteroid dehydrogenase (Hsd3b), and progesterone in the culture medium. OPN treatment promoted Vegf expression in GCs, which was significantly suppressed by a phosphoinositide 3-kinase (PI3K) inhibitor. In addition, OPN treatment stimulated phosphorylation of AKT, a downstream PI3K signaling molecule. In conclusion, expression of Opn was upregulated in mouse ovarian GCs in response to a gonadotropin surge through epidermal growth factor receptor signaling, which enhances progesterone synthesis and Vegf expression during the early-luteal phase.
Collapse
Affiliation(s)
- Yoshimitsu Kuwabara
- Departments of Obstetrics and GynecologyBiochemistry and Molecular BiologyNippon Medical School, 1-1-5, Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Akira Katayama
- Departments of Obstetrics and GynecologyBiochemistry and Molecular BiologyNippon Medical School, 1-1-5, Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Ryoko Tomiyama
- Departments of Obstetrics and GynecologyBiochemistry and Molecular BiologyNippon Medical School, 1-1-5, Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Hu Piao
- Departments of Obstetrics and GynecologyBiochemistry and Molecular BiologyNippon Medical School, 1-1-5, Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Sachiko Kurihara
- Departments of Obstetrics and GynecologyBiochemistry and Molecular BiologyNippon Medical School, 1-1-5, Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan Departments of Obstetrics and GynecologyBiochemistry and Molecular BiologyNippon Medical School, 1-1-5, Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Shuichi Ono
- Departments of Obstetrics and GynecologyBiochemistry and Molecular BiologyNippon Medical School, 1-1-5, Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Katsuya Mine
- Departments of Obstetrics and GynecologyBiochemistry and Molecular BiologyNippon Medical School, 1-1-5, Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Shigeo Akira
- Departments of Obstetrics and GynecologyBiochemistry and Molecular BiologyNippon Medical School, 1-1-5, Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Hideo Orimo
- Departments of Obstetrics and GynecologyBiochemistry and Molecular BiologyNippon Medical School, 1-1-5, Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Toshiyuki Takeshita
- Departments of Obstetrics and GynecologyBiochemistry and Molecular BiologyNippon Medical School, 1-1-5, Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| |
Collapse
|
13
|
Shevde LA, Samant RS. Role of osteopontin in the pathophysiology of cancer. Matrix Biol 2014; 37:131-41. [PMID: 24657887 PMCID: PMC5916777 DOI: 10.1016/j.matbio.2014.03.001] [Citation(s) in RCA: 198] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 03/10/2014] [Accepted: 03/10/2014] [Indexed: 12/12/2022]
Abstract
Osteopontin (OPN) is a multifunctional cytokine that impacts cell proliferation, survival, drug resistance, invasion, and stem like behavior. Due to its critical involvement in regulating cellular functions, its aberrant expression and/or splicing is functionally responsible for undesirable alterations in disease pathologies, specifically cancer. It is implicated in promoting invasive and metastatic progression of many carcinomas. Due to its autocrine and paracrine activities OPN has been shown to be a crucial mediator of cellular cross talk and an influential factor in the tumor microenvironment. OPN has been implicated as a prognostic and diagnostic marker for several cancer types. It has also been explored as a possible target for treatment. In this article we hope to provide a broad perspective on the importance of OPN in the pathophysiology of cancer.
Collapse
Affiliation(s)
- Lalita A Shevde
- Department of Pathology and Comprehensive Cancer Center, The University of Alabama at Birmingham, United States.
| | - Rajeev S Samant
- Department of Pathology and Comprehensive Cancer Center, The University of Alabama at Birmingham, United States.
| |
Collapse
|
14
|
Yang G, Zhang S, Zhang Y, Zhou Q, Peng S, Zhang T, Yang C, Zhu Z, Zhang F. The inhibitory effects of extracellular ATP on the growth of nasopharyngeal carcinoma cells via P2Y2 receptor and osteopontin. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2014; 33:53. [PMID: 24961145 PMCID: PMC4078358 DOI: 10.1186/1756-9966-33-53] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 06/17/2014] [Indexed: 12/15/2022]
Abstract
Background Nasopharyngeal carcinoma (NPC) is a common malignant tumor observed in the populations of southern China and Southeast Asia. However, little is known about the effects of purinergic signal on the behavior of NPC cells. This study analyzed the effects of ATP on the growth and migration of NPC cells, and further investigated the potential mechanisms during the effects. Methods Cell viability was estimated by MTT assay. Transwell assay was utilized to assess the motility of NPC cells. Cell cycle and apoptosis were detected by flow cytometry analysis. Changes in OPN, P2Y2 and p65 expression were assessed by western blotting analysis or immunofluorescence. The effects of ATP and P2Y2 on promoter activity of OPN were analyzed by luciferase activity assay. The binding of p65 to the promoter region of OPN was examined by ChIP assay. Results An MTT assay indicated that ATP inhibited the proliferation of NPC cells in time- and dose-dependent manners, and a Transwell assay showed that extracellular ATP inhibited the motility of NPC cells. We further investigated the potential mechanisms involved in the inhibitory effect of extracellular ATP on the growth of NPC cells and found that extracellular ATP could reduce Bcl-2 and p-AKT levels while elevating Bax and cleaved caspase-3 levels in NPC cells. Decreased levels of p65 and osteopontin were also detected in the ATP-treated NPC cells. We demonstrated that extracellular ATP inhibited the growth of NPC cells via p65 and osteopontin and verified that P2Y2 overexpression elevated the inhibitory effect of extracellular ATP on the proliferation of NPC cells. Moreover, a dual luciferase reporter assay showed that the level of osteopontin transcription was inhibited by extracellular ATP and P2Y2. ATP decreased the binding of p65 to potential sites in the OPN promoter region in NPC cells. Conclusion This study indicated that extracellular ATP inhibited the growth of NPC cells via P2Y2, p65 and OPN. ATP could be a promising agent serving as an adjuvant in the treatment of NPC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Fujun Zhang
- State Key Laboratory of Oncology in South China, Department of Imaging and Interventional Radiology, Cancer Center, Sun Yat-sen University, Guangzhou Guangdong 510060, China.
| |
Collapse
|
15
|
Ramchandani D, Weber GF. An osteopontin promoter polymorphism is associated with aggressiveness in breast cancer. Oncol Rep 2013; 30:1860-8. [PMID: 23900428 DOI: 10.3892/or.2013.2632] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 07/01/2013] [Indexed: 11/06/2022] Open
Abstract
Metastasis-related genes are deregulated in cancer by aberrant expression or splicing. Here, we analyzed polymorphic sites in the osteopontin promoter as potential contributors to aberrant expression in breast cancers. This study comprised 241 breast cancer specimens, for which DNA from normal surrounding tissue was available for 111, and 65 healthy breast samples. The polymorphic site in position -443 of the promoter was associated with tumor grade. As expected, there was no association between promoter single nucleotide polymorphisms (SNPs) and tumor stage or in situ carcinoma versus cancer, as stage and early transformation are determined by the sampling time more than by tumor genetics. In a subset of samples, osteopontin RNA expression levels had previously been obtained. The allelic distribution in positions -443 and -1748 was distinct between high and low expressors, confirming the importance of promoter SNPs. These two sites also form a haplotype. Osteopontin expression has been associated with breast cancer progression, regardless of the histological subtype of the cancer. Remarkably, the polymorphic site at -443, but not -1748 or -1776, showed differences between ER-positive and ER-negative breast cancers and between PR-positive and PR-negative breast cancers, but there was no association with HER2 status. In five cases, the genotype of the tumor was different from the genotype of the host, implying the possibility of somatic mutations in the osteopontin promoter that may affect expression. Our results corroborate that the osteopontin promoter SNPs -443 (rs11730582) and -1748 (rs2728127) are important for gene expression and breast cancer aggressiveness.
Collapse
Affiliation(s)
- Divya Ramchandani
- University of Cincinnati Academic Health Center, College of Pharmacy, Cincinnati, OH 45267-0004, USA
| | | |
Collapse
|
16
|
Bayer CL, Joshi PP, Emelianov SY. Photoacoustic imaging: a potential tool to detect early indicators of metastasis. Expert Rev Med Devices 2013; 10:125-34. [PMID: 23278229 DOI: 10.1586/erd.12.62] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The metastasis of cancer is a multistage process involving complex biological interactions and difficult to predict outcomes. Accurate assessment of the extent of metastasis is critical for clinical practice; unfortunately, medical imaging methods capable of identifying the early stages of invasion and metastasis are lacking. Photoacoustic imaging is capable of providing noninvasive, real-time imaging of significant anatomical and physiological changes. indicating the progression of cancer invasion and metastasis. Preclinically, photoacoustic methods have been used to image lymphatic anatomy, including the sentinel lymph nodes, to identify circulating tumor cells within vasculature and to detect micrometastases. Progress has begun toward the development of clinically applicable photoacoustic imaging systems to assist with the determination of cancer stage and likelihood of metastatic invasion.
Collapse
Affiliation(s)
- Carolyn L Bayer
- Department of Biomedical Engineering, The University of Texas at Austin, 1 University Station, Austin, TX 78712, USA.
| | | | | |
Collapse
|
17
|
Shen H, Weber GF. The osteopontin-c splice junction is important for anchorage-independent growth. Mol Carcinog 2013; 53:480-7. [PMID: 23359517 DOI: 10.1002/mc.21997] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 11/29/2012] [Accepted: 12/07/2012] [Indexed: 01/17/2023]
Abstract
The secreted molecule osteopontin (OPN) is important for metastasis by various cancers. While the full-length form, OPN-a, is also present in healthy tissues, human breast tumors generate a shorter splice form, OPN-c, which has only been found in cancers. OPN-c is more potent than OPN-a in supporting anchorage-independence and may be critical for metastasis. Here we analyze the structural requirements for OPN-c function. An antibody specific to the splice junction neutralizes the OPN-c effects. A short peptide covering the sequence around the splice junction inhibits, whereas a long peptide mimics the effects of OPN-c. Site-directed mutagenesis identifies the distal, but not the proximal phosphorylation sites upstream and downstream of the splice junction as important for function. The interposed amino acids are also important as their mutation to alanines leads to a loss of OPN-c effects. We find that small peptides and specific antibodies can neutralize the anchorage-independence enhancing functions of OPN-c. This implies potential for future therapeutic approaches.
Collapse
Affiliation(s)
- Hui Shen
- University of Cincinnati Academic Health Center, College of Pharmacy, Cincinnati, Ohio
| | | |
Collapse
|
18
|
Ma H, Xu H, Qin J. Biomimetic tumor microenvironment on a microfluidic platform. BIOMICROFLUIDICS 2013; 7:11501. [PMID: 24396521 PMCID: PMC3556015 DOI: 10.1063/1.4774070] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Accepted: 12/17/2012] [Indexed: 05/23/2023]
Abstract
Tumor microenvironment is a highly complex system consisting of non-cancerous cells, soluble factors, signaling molecules, extracellular matrix, and mechanical cues, which provides tumor cells with integrated biochemical and biophysical cues. It has been recognized as a significant regulator in cancer initiation, progression, metastasis, and drug resistance, which is becoming a crucial component of cancer biology. Modeling microenvironmental conditions of such complexity in vitro are particularly difficult and technically challenging. Significant advances in microfluidic technologies have offered an unprecedented opportunity to closely mimic the physiological microenvironment that is normally encountered by cancer cells in vivo. This review highlights the recent advances of microfluidic platform in recapitulating many aspects of tumor microenvironment from biochemical and biophysical regulations. The major events relevant in tumorigenesis, angiogenesis, and spread of cancer cells dependent on specific combinations of cell types and soluble factors present in microenvironmental niche are summarized. The questions and challenges that lie ahead if this field is expected to transform the future cancer research are addressed as well.
Collapse
Affiliation(s)
- Huipeng Ma
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Hui Xu
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Jianhua Qin
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| |
Collapse
|
19
|
Jiang S, Chen X, Li C, Zhang X, Zhang T, Yue Y, Yang G. Suramin inhibits the growth of nasopharyngeal carcinoma cells via the downregulation of osteopontin. Mol Med Rep 2012; 6:1351-4. [PMID: 22965338 DOI: 10.3892/mmr.2012.1074] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Accepted: 08/28/2012] [Indexed: 11/06/2022] Open
Abstract
Radiotherapy is the principal therapy for nasopharyngeal carcinoma (NPC) at early stages. A number of chemotherapeutic methods have been used to inhibit the progression of NPC at elevated stages. Suramin has been reported to inhibit the growth of certain tumor cells via various pathways. In the present study, we aimed to analyze the effects of suramin on the proliferation of NPC cells (CNE-2). Suramin was proved to demonstrate NPC cell growth-inhibiting effects both in a dose- and time-dependent manner. To determine the potential mechanisms of these effects, western blotting and flow cytometric analysis were performed. Suramin was found to have the potential to induce cell cycle arrest in S-phase CNE-2 cells. Additionally, we found that the OPN level may decrease in suramin-treated CNE-2 cells. The changes of certain apoptosis- and p-AKT-associated proteins possibly regulating the OPN expression were measured by western blotting. In conclusion, suramin may function as a potential agent for the adjunctive therapy of NPC.
Collapse
Affiliation(s)
- Shan Jiang
- Department of Oncology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, P.R. China
| | | | | | | | | | | | | |
Collapse
|
20
|
Liu XP, Chen SY, Gao H, Pan QC. Clinical significance of OPN and VEGF-C expression in gastric cancer. Shijie Huaren Xiaohua Zazhi 2012; 20:2243-2247. [DOI: 10.11569/wcjd.v20.i24.2243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To evaluate the expression of osteopontin (OPN) and vascular endothelial growth factor C (VEGF-C) in gastric cancer and to analyze their correlation with clinical pathological characteristics of gastric cancer.
METHODS: Surgical specimens of gastric cancer and related clinical data were collected from the Affiliated Zhongshan Hospital of Fudan University. The expression of OPN and VEGF-C proteins in 93 gastric cancer specimens was examined by immunohistochemistry.
RESULTS: The positive rates of OPN and VEGF-C expression in 93 gastric cancer specimens were 64.5% (60/93) and 69.9% (65/93), respectively. The expression of OPN and VEGF-C was significantly correlated with serosal invasion, tumor TNM stage, and lymph node metastasis (all P < 0.05). Moreover, the expression of OPN was positively correlated with that of VEGF-C in gastric cancer (r = 0.493, P < 0.01).
CONCLUSION: OPN and VEGF-C protein expression might be associated with the development, invasion, and metastasis of gastric cancer. OPN may play a role in the lymph node metastasis of gastric cancer by up-regulating the expression of VEGF-C.
Collapse
|
21
|
Matušan-Ilijaš K, Damante G, Fabbro D, Dorđević G, Hadžisejdić I, Grahovac M, Avirović M, Grahovac B, Jonjić N, Lučin K. EGFR expression is linked to osteopontin and Nf-κB signaling in clear cell renal cell carcinoma. Clin Transl Oncol 2012; 15:65-71. [PMID: 22855173 DOI: 10.1007/s12094-012-0889-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Accepted: 03/19/2012] [Indexed: 10/28/2022]
Abstract
AIM AND BACKGROUND Epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase involved in many important aspects of cell biology that are related to tumorigenesis. There are opposite evidences of the role of EGFR in renal cancer and the outcome of EGFR-targeted therapies, suggesting the complexity of EGFR signaling pathways. In vitro, osteopontin (OPN) and nuclear factor kappa B (NF-κB) are thought to be involved in specific ligand-independent EGFR activation that could have a role in resistance to EGFR mAb therapy. Aim of this study was to analyze the relationship between EGFR and OPN at the protein and mRNA level, as well as their relation to NF-κB in clear cell renal cell carcinoma (CCRCC). MATERIALS AND METHODS Expression of EGFR, OPN, and p65 NF-κB protein was analyzed using immunohistochemistry and compared mutually in 88 CCRCC samples. Expression of EGFR and OPN mRNAs was analyzed using quantitative Real-time PCR in 22 CCRCC samples and compared mutually and with NF-κB protein expression. RESULTS Epidermal growth factor receptor mRNA level was higher in CCRCC samples in comparison with normal renal tissue (p = 0.012) and was associated with high OPN mRNA level, and with NF-κB activation (p < 0.001 and p = 0.045, respectively). Immunohistochemical staining showed the inverse association; high EGFR protein expression was related with low OPN and NF-κB protein expression (p < 0.001 and p = 0.047, respectively). CONCLUSION Epidermal growth factor receptor gene is upregulated in CRCC and associated with OPN gene expression and NF-kB signaling. The inverse relation between OPN and EGFR at the protein level could probably reflect dynamic changes that EGFR undergoes following activation.
Collapse
Affiliation(s)
- K Matušan-Ilijaš
- Department of Pathology, Rijeka University School of Medicine, Braće Branchetta 20, Rijeka, Croatia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Leng H, Zang WQ, Wang T, Wang YY, Ma J, Zhao GQ. Overexpression of miR-451 inhibits cell proliferation and invasion and promotes apoptosis in human esophageal carcinoma cell line EC9706. Shijie Huaren Xiaohua Zazhi 2012; 20:1323-1327. [DOI: 10.11569/wcjd.v20.i15.1323] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To explore the effect of overexpression of miR-451 on cell proliferation, apoptosis and invasion in human esophageal carcinoma cell line EC9706.
METHODS: MiR-451 mimics were constructed and transfected into EC9706 cells using LipofectamineTM2000. EC9706 cells transfected with Scramble-miR, empty liposomes or negative miR-451 were used as controls. Forty-eight hours after transfection, the expression of miR-451 was detected by RT-PCR, and the expression of Bcl-2, AKT and phosphorylated AKT proteins were detected by Western blot. Cell apoptosis, invasion and proliferation were assessed by flow cytometry, transwell assay and MTT assay, respectively.
RESULTS: Compared to control cells, the expression of miR-451 was significantly up-regulated (P < 0.01, F = 69.26); the expression of Bcl-2, AKT and phosphorylated AKT proteins was significantly down-regulated (P < 0.05, F = 5.83); cell apoptosis rate significantly increased (P < 0.01, F = 26.72); the average number of cells penetrating Matrigel membrane significantly decreased (P < 0.01, F = 34.55); and cell proliferation was significantly suppressed in a time-dependent manner (P < 0.05, F = 5.95) in EC9706 cells transfected with miR-451 mimics.
CONCLUSION: Over-expression of miR-451 induces apoptosis and suppresses cell proliferation and invasion in human esophageal carcinoma cell line EC9706.
Collapse
|
23
|
Morrow KA, Das S, Metge BJ, Ye K, Mulekar MS, Tucker JA, Samant RS, Shevde LA. Loss of tumor suppressor Merlin in advanced breast cancer is due to post-translational regulation. J Biol Chem 2011; 286:40376-85. [PMID: 21965655 DOI: 10.1074/jbc.m111.250035] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Unlike malignancies of the nervous system, there have been no mutations identified in Merlin in breast cancer. As such, the role of the tumor suppressor, Merlin, has not been investigated in breast cancer. We assessed Merlin expression in breast cancer tissues by immunohistochemistry and by real-time PCR. The expression of Merlin protein (assessed immunohistochemically) was significantly decreased in breast cancer tissues (although the transcript levels were comparable) simultaneous with increased expression of the tumor-promoting protein, osteopontin (OPN). We further demonstrate that the loss of Merlin in breast cancer is brought about, in part, due to OPN-initiated Akt-mediated phosphorylation of Merlin leading to its proteasomal degradation. Restoring expression of Merlin resulted in reduced malignant attributes of breast cancer, characterized by reduced invasion, migration, motility, and impeded tumor (xenograft) growth in immunocompromised mice. The possibility of developing a model using the relationship between OPN and Merlin was tested with a logistic regression model applied to immunohistochemistry data. This identified consistent loss of immunohistochemical expression of Merlin in breast tumor tissues. Thus, we demonstrate for the first time a role for Merlin in impeding breast malignancy, identify a novel mechanism for the loss of Merlin protein in breast cancer, and have developed a discriminatory model using Merlin and OPN expression in breast tumor tissues.
Collapse
Affiliation(s)
- K Adam Morrow
- Department of Oncologic Sciences, Mitchell Cancer Institute, Mobile, Alabama 36604, USA
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Flajollet S, Tian TV, Flourens A, Tomavo N, Villers A, Bonnelye E, Aubert S, Leroy X, Duterque-Coquillaud M. Abnormal expression of the ERG transcription factor in prostate cancer cells activates osteopontin. Mol Cancer Res 2011; 9:914-24. [PMID: 21669963 DOI: 10.1158/1541-7786.mcr-10-0537] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Osteopontin (OPN) is an extracellular matrix glycophosphoprotein that plays a key role in the metastasis of a wide variety of cancers. The high level of OPN expression in prostate cells is associated with malignancy and reduced survival of the patient. Recent studies on prostate cancer (PCa) tissue have revealed recurrent genomic rearrangements involving the fusion of the 5' untranslated region of a prostate-specific androgen-responsive gene with a gene coding for transcription factors from the ETS family. The most frequently identified fusion gene is TMPRSS2:ERG, which causes ERG protein overexpression in PCa cells. ERG is a transcription factor linked to skeletogenesis. This study was designed to test whether ERG and the product of the TMPRSS2:ERG fusion gene modulate OPN gene expression in PCa cells. To characterize ERG and TMPRSS2:ERG transcriptional activity of OPN, we focused on ETS binding sites (EBS) localized in conserved regions of the promoter. Using in vitro and in vivo molecular assays, we showed that ERG increases OPN expression and binds to an EBS (nt -115 to -118) in the OPN promoter. Moreover, stable transfection of prostate tumor cell lines by TMPRSS2:ERG upregulates endogenous OPN expression. Finally, in human prostate tumor samples, detection of the TMPRSS2:ERG fusion gene was significantly associated with OPN overexpression. Taken together, these data suggest that OPN is an ERG-target gene in PCa where the abnormal expression of the transcription factor ERG, due to the TMPRSS2:ERG fusion, disturbs the expression of genes that play an important role in PCa cells and associated metastases.
Collapse
|
25
|
De Laurentiis A, Pardo OE, Palamidessi A, Jackson SP, Schoenwaelder SM, Reichmann E, Scita G, Arcaro A. The catalytic class I(A) PI3K isoforms play divergent roles in breast cancer cell migration. Cell Signal 2010; 23:529-41. [PMID: 21056654 DOI: 10.1016/j.cellsig.2010.10.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Accepted: 10/23/2010] [Indexed: 02/05/2023]
Abstract
Transforming growth factor-β (TGFβ) plays an important role in breast cancer metastasis. Here phosphoinositide 3-kinase (PI3K) signalling was found to play an essential role in the enhanced migration capability of fibroblastoid cells (FibRas) derived from normal mammary epithelial cells (EpH4) by transduction of oncogenic Ras (EpRas) and TGFβ1. While expression of the PI3K isoform p110δ was down-regulated in FibRas cells, there was an increase in the expression of p110α and p110β in the fibroblastoid cells. The PI3K isoform p110β was found to specifically contribute to cell migration in FibRas cells, while p110α contributed to the response in EpH4, EpRas and FibRas cells. Akt, a downstream targets of PI3K signalling, had an inhibitory role in the migration of transformed breast cancer cells, while Rac, Cdc42 and the ribosomal protein S6 kinase (S6K) were necessary for the response. Together our data reveal a novel specific function of the PI3K isoform p110β in the migration of cells transformed by oncogenic H-Ras and TGF-β1.
Collapse
Affiliation(s)
- Angela De Laurentiis
- Division of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, 8032 Zurich, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Song G, Ouyang G, Mao Y, Ming Y, Bao S, Hu T. Osteopontin promotes gastric cancer metastasis by augmenting cell survival and invasion through Akt-mediated HIF-1alpha up-regulation and MMP9 activation. J Cell Mol Med 2010; 13:1706-1718. [PMID: 19602039 DOI: 10.1111/j.1582-4934.2008.00540.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Osteopontin (OPN) is a secreted, integrin-binding matrix phosphorylated glycoprotein. OPN has been shown to facilitate the progression and metastasis of malignancies and has prognostic value in several types of cancer, including gastric cancer. However, the functional mechanism of OPN mediated metastatic growth in gastric cancer remains unclear. Here, using multiple in vitro and in vivo models, we report that OPN strongly promoted the progression and metastasis of gastric cancer. Immunohistochemical staining revealed that OPN, matrix metalloproteinase (MMP)9 and hypoxia-inducible factor (HIF)-1alpha have statistically significant different expression patterns between well- and poorly differentiated tissue samples (P < 0.05). Correlations existed between OPN and MMP9, and between OPN and HIF-1 (r(1) = 0.872, p(1) < 0.01 and r(2) = 0.878, p(2) < 0.01). Furthermore, OPN dramatically increased colony formation and invasion of gastric cancer cells in vitro and promoted tumour growth and metastasis in vivo. In addition, OPN potently protected gastric cancer cells from serum depletion-induced apoptosis. Further study shows that OPN activated phosphoinositide 3-kinase/Akt survival pathway and up-regulated HIF-1alpha via binding to v3 integrins in gastric cancer cells. Moreover, we found that OPN could activate MMP9 and upregulate MMP2. Taken together, our results suggest that the survival-promoting function is crucial for OPN to promote the development of gastric cancer, and HIF-1 and MMP9 may play key roles during this process. Thus, targeting OPN and its related signalling network may develop an effective therapeutic approach for the management of gastric cancer.
Collapse
Affiliation(s)
- Gang Song
- Cancer Research Center, Xiamen University Medical College, Xiamen, China.,The Key Laboratory of Ministry of Education for Cell Biology and Tumor Cell Engineering, School of Life Sciences, Xiamen University, Xiamen, China
| | - Gaoliang Ouyang
- The Key Laboratory of Ministry of Education for Cell Biology and Tumor Cell Engineering, School of Life Sciences, Xiamen University, Xiamen, China
| | - Yubin Mao
- Cancer Research Center, Xiamen University Medical College, Xiamen, China
| | - Yanlin Ming
- The Key Laboratory of Ministry of Education for Cell Biology and Tumor Cell Engineering, School of Life Sciences, Xiamen University, Xiamen, China
| | - Shideng Bao
- The Key Laboratory of Ministry of Education for Cell Biology and Tumor Cell Engineering, School of Life Sciences, Xiamen University, Xiamen, China
| | - Tianhui Hu
- Cancer Research Center, Xiamen University Medical College, Xiamen, China
| |
Collapse
|
27
|
Shevde LA, Das S, Clark DW, Samant RS. Osteopontin: an effector and an effect of tumor metastasis. Curr Mol Med 2010; 10:71-81. [PMID: 20205680 DOI: 10.2174/156652410791065381] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2008] [Accepted: 05/11/2008] [Indexed: 12/12/2022]
Abstract
Osteopontin (OPN) is a matricellular protein that is produced by multiple tissues in our body and is most abundant in bone. It is also produced by cancer cells and plays a determinative role in the growth, progression and metastasis of cancer. Clinically, OPN has been reported to be upregulated in tumor cells per se; this is also reflected by increased levels of OPN in the circulation. Thus, increased OPN levels the plasma are an effect of tumor growth and progression. Functionally, high OPN levels are determinative of higher incidence of bone metastases in mouse models and are clinically correlated with metastatic bone disease and bone resorption in advanced breast cancer patients. Several research efforts have been made to therapeutically target and inhibit the activities of OPN. In this article we have reviewed OPN in its role as an effector of critical steps in tumor progression and metastasis, with a particular emphasis on its role in facilitating bone metastasis of breast cancer. We have also addressed the role of the host-derived OPN in influencing the malignant behavior of the tumor cells.
Collapse
Affiliation(s)
- L A Shevde
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA.
| | | | | | | |
Collapse
|
28
|
Xu Z, Zhang Y, Jiang J, Yang Y, Shi R, Hao B, Zhang Z, Huang Z, Kim JW, Zhang G. Epidermal growth factor induces HCCR expression via PI3K/Akt/mTOR signaling in PANC-1 pancreatic cancer cells. BMC Cancer 2010; 10:161. [PMID: 20423485 PMCID: PMC2880295 DOI: 10.1186/1471-2407-10-161] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2009] [Accepted: 04/27/2010] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Human cervical cancer oncoprotein 1 (HCCR-1), reported as a negative regulator of p53, is over-expressed in a variety of human cancers. However, it is yet unknown whether HCCR-1 plays any role in pancreatic cancer development. The aim of this study was to investigate the effect of epidermal growth factor on the expression of HCCR in pancreatic cancer cells, and to explore if PI3K/Akt/mTOR signaling pathway mediated this expression. METHODS A polyclonal antibody against HCCR protein was raised by immunizing Balb/c mice with the purified recombinant protein pMBPc-HCCR. Tissue samples were constructed on a tissue chip, and the expression of HCCR was investigated by immunohistochemistry assay and Western blotting. Pancreatic cell line, PANC-1 cells were stably transfected with plasmids containing sense-HCCR-1 fragment and HCCR siRNA fragment. MTT and transwell assay were used to investigate the proliferation and invasion of stable tansfectants. The specific inhibitor of PI3K and mTOR was used to see if PI3K/mTOR signal transduction was involved in the induction of HCCR gene expression. A Luciferase assay was used to see if Akt can enhance the HCCR promoter activity. RESULTS HCCR was up-regulated in pancreatic tumor tissues (mean Allred score 4.51+/-1.549 vs. 2.87+/-2.193, P<0.01), especially with high expression in poorly differentiated pancreatic cancer. The growth of cells decreased in HCCR-1 siRNA transfected cells compared with vector transfectants. The number of invasion cells was significantly lower in HCCR-1 siRNA transfected cells (24.4+/-9.9) than that in vector transfectants (49.1+/-15.4). Treatment of PANC-1 cells with epidermal growth factor increased HCCR protein level in a dose- and time-dependent manner. However, application of LY294002 and rapamycin caused a dramatic reduction of epidermal growth factor-induced HCCR expression. Over-expression of exogenous constitutively active Akt increased the HCCR promoter activity; in contrast, dominant negative Akt decreased the promoter activity. CONCLUSIONS EGF-induced HCCR-1 over-expression is mediated by PI3K/AKT/mTOR signaling which plays a pivotal role in pancreatic tumor progression, suggesting that HCCR-1 could be a potential target for cancer therapeutics.
Collapse
Affiliation(s)
- Zekuan Xu
- Department of Gastroenterology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Seow HF, Yip WK, Loh HW, Ithnin H, Por P, Rohaizak M. Immunohistochemical detection of phospho-Akt, phospho-BAD, HER2 and oestrogen receptors alpha and beta in Malaysian breast cancer patients. Pathol Oncol Res 2009; 16:239-48. [PMID: 19882362 DOI: 10.1007/s12253-009-9216-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2009] [Accepted: 10/07/2009] [Indexed: 01/12/2023]
Abstract
Activation of Akt signaling pathway has been documented in various human malignancies, including breast carcinoma. The objective of this study is to determine the incidence of Akt phosphorylation in breast tumours and its relationship with expression of ER-alpha, ER-beta, HER2, Ki-67 and phosphorylated Bcl-2 associated death domain (p-BAD). Immunohistochemical staining was performed to detect these molecules on 43 paraffin-embedded breast tumour tissues with commercially available antibodies. Eighteen (41.9%), 3 (7.0%), 23 (53.5%), 35 (81.4%), 21 (48.8%), 29 (67.4%), and 34 (81.0%) of breast tumours were positive for nuclear ER-alpha, nuclear ER-beta, membranous HER2, cytonuclear p-Akt (Thr308), p-Akt (Ser473), p-BAD and Ki-67, respectively. ER-alpha expression was inversely correlated with HER2 and Ki-67 (P = 0.041 and P = 0.040, respectively). The p-Akt (Ser473) was correlated with increased level of p-BAD (Ser136) (P = 0.012). No relationship of Akt phosphorylation with HER2, ER-alpha or ER-beta was found. The p-Akt (Ser473) immunoreactivity was significantly higher in stage IV than in stage I or II (P = 0.036 or P = 0.009). The higher Ki-67 and lower ER-alpha expression showed an association with patient age of <50 years (P = 0.004) and with positive nodal status (P = 0.033), respectively. Our data suggest that the Akt phosphorylation and inactivation of its downstream target, BAD may play a role in survival of breast cancer cell. This study does not support the simple model of linear HER2/PI3K/Akt pathway in breast cancer.
Collapse
Affiliation(s)
- Heng Fong Seow
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia.
| | | | | | | | | | | |
Collapse
|
30
|
Zhang J, Yamada O, Matsushita Y, Chagan-Yasutan H, Hattori T. Transactivation of human osteopontin promoter by human T-cell leukemia virus type 1-encoded Tax protein. Leuk Res 2009; 34:763-8. [PMID: 19767100 DOI: 10.1016/j.leukres.2009.08.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Revised: 08/25/2009] [Accepted: 08/26/2009] [Indexed: 01/01/2023]
Abstract
Osteopontin (OPN) is a cytokine that contributes substantially to the growth and metastasis in a wide spectrum of malignancies. We report here that OPN gene is transactivated by Tax protein of human T-cell leukemia virus type 1 (HTLV-1). Northern blot showed enhanced OPN gene expression in cells stably expressing Tax. Co-expression of Tax increased the reporter gene expression directed by OPN promoter. Tax-induced OPN activation was abrogated by treatment with LY294002 (PI3K inhibitor) or co-transfection with AKT siRNA, suggesting PI3K/AKT pathway is involved in Tax-mediated transactivation. Reporter assay with deletion mutants showed that the 5'-partial sequence between -765 and -660 of the OPN promoter is the region responsive to Tax, and further, disrupting the AP-1 site within this region abolished the OPN induction by Tax, indicating that Tax activation of OPN promoter is likely mediated by AP-1 site. This study suggests that OPN is one of the downstream mediators of aberrantly activated PI3K/AKT signaling by Tax, which may partially contribute to HTLV-1-associated leukemogenesis.
Collapse
Affiliation(s)
- Jing Zhang
- Research and Development Center, FUSO Pharmaceutical Industries, Ltd, Joto-ku, Osaka 536-8523, Japan.
| | | | | | | | | |
Collapse
|
31
|
Macrì A, Versaci A, Lupo G, Trimarchi G, Tomasello C, Loddo S, Sfuncia G, Caminiti R, Teti D, Famulari C. Role of osteopontin in breast cancer patients. TUMORI JOURNAL 2009; 95:48-52. [PMID: 19366056 DOI: 10.1177/030089160909500109] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2023]
Abstract
AIM AND BACKGROUND In breast cancer, as in almost all neoplastic diseases, the prognosis is strictly related to the invasive capacity, local and distant, that characterizes the growth of all tumors. Since the mechanisms that regulate replication of the neoplastic cells, with consequent capacity to metastasize, are not completely known, identification of new markers represents the gold standard of research in the stratification of patients with such a pathology. Osteopontin, a specific phosphoglycoprotein isolated from extracellular bone matrix and actively involved in mechanisms of bone reabsorption, appears to play a key role in osteoclastogenesis at the level of the skeleton in some pathologic situations. It has been found that patients with metastatic bone lesions from breast or prostate cancer present, with respect to subjects without repetitive bone lesions, elevated serum levels of the protein, indicating that osteopontin could play an important role in the development and progression of the neoplastic disease at the bone level. METHODS AND STUDY DESIGN The authors studied 26 patients with breast cancer, evaluating as a marker also serum osteopontin levels. RESULTS AND CONCLUSIONS The results, although obtained on a small number of patients, showed that osteopontin evaluation in breast cancer patients can be a particularly interesting method of research in staging of the disease as well as in the prognosis, thereby attributing a role of a biotumoral marker also in the follow-up of the therapy.
Collapse
Affiliation(s)
- Antonio Macrì
- Emergency Surgery Unit, Department of Human Pathology, University of Messina, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Song G, Ming Y, Mao Y, Bao S, Ouyang G. Osteopontin prevents curcumin-induced apoptosis and promotes survival through Akt activation via alpha v beta 3 integrins in human gastric cancer cells. Exp Biol Med (Maywood) 2008; 233:1537-45. [PMID: 18849546 DOI: 10.3181/0805-rm-164] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Osteopontin (OPN) is a secreted, integrin-binding matrix phosphorylated glycoprotein that is overexpressed in many advanced cancers. However, the functional mechanisms by which OPN contributes to gastric cancer development are poorly understood. Here, we report that curcumin inhibited the growth of SGC7901 cell and induced apoptosis in a concentration- and time-dependent manner, while the acquired expression of OPN in SGC7901 cells dramatically promoted cell survival under serum depletion and prevented curcumin-induced apoptosis. Furthermore, PI3-K inhibitor LY294002 attenuated OPN-mediated Akt activation. Moreover, inhibiting the binding of OPN to alpha(v)beta(3) integrins reduced activation of Akt. Taken together, these results demonstrate that the pro-survival and anti-apoptosis activities of OPN in gastric cancer cells are mediated in part through PI3-K/Akt pathway via alpha(v)beta(3) integrins.
Collapse
Affiliation(s)
- Gang Song
- Key Laboratory of the Ministry of Education for Cell Biology and Tumor Cell Engineering, School of Life Sciences, Xiamen University, Xiamen 361005, China
| | | | | | | | | |
Collapse
|
33
|
Song G, Cai QF, Mao YB, Ming YL, Bao SD, Ouyang GL. Osteopontin promotes ovarian cancer progression and cell survival and increases HIF-1alpha expression through the PI3-K/Akt pathway. Cancer Sci 2008; 99:1901-7. [PMID: 19016748 PMCID: PMC11158665 DOI: 10.1111/j.1349-7006.2008.00911.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2008] [Revised: 06/13/2008] [Accepted: 06/17/2008] [Indexed: 12/27/2022] Open
Abstract
Osteopontin (OPN) is a secreted, integrin-binding matrix phosphorylated glycoprotein that is overexpressed in many advanced cancers. However, the functional mechanisms by which OPN contributes to the development of ovarian cancer are poorly understood. Here, we reveal that acquired expression of OPN by HO-8910 ovarian cancer cells greatly promoted the progression of ovarian cancer. OPN expression dramatically increased the colony formation of ovarian cancer cells in vitro and tumor growth in vivo. Under the stress induced by serum depletion or curcumin treatment, OPN expression promoted the survival of ovarian cells through preventing stress-induced apoptosis. At the molecular level, both endogenous and exogenous OPN expression activated the PI3-K/Akt survival pathway and dramatically decreased p53 expression under serum depletion. In addition, HIF-1alpha was induced in OPN-producing cells under normoxia. Furthermore, we also found that inhibition of the PI3-K/Akt pathway attenuated OPN-mediated HIF-1alpha up-regulation in ovarian cancer cells. Taken together, these results indicate that OPN can increase the survival of ovarian cancer cells under stress conditions in vitro and promote the late progression of ovarian cancer in vivo, and the survival-promoting functions of OPN are mediated through Akt activation and the induction of HIF-1alpha expression.
Collapse
Affiliation(s)
- Gang Song
- The Key Laboratory of Ministry of Education for Cell Biology and Tumor Cell Engineering, School of Life Sciences, Xiamen University, Xiamen, China
| | | | | | | | | | | |
Collapse
|
34
|
Syed M, Fenoglio-Preiser C, Skau KA, Weber GF. Acetylcholinesterase supports anchorage independence in colon cancer. Clin Exp Metastasis 2008; 25:787-98. [PMID: 18612832 DOI: 10.1007/s10585-008-9192-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2007] [Accepted: 06/19/2008] [Indexed: 11/24/2022]
Abstract
Various roles have been attributed to Acetylcholinesterase (AChE) in cancer. Evidence exists for a pro-apoptotic function, consistent with a protective role of AChE. Because other reports suggested that upregulated AChE in some tumors may control cell adhesion, we tested the effects of AChE on anchorage independence (an essential component of metastasis) of colon tumor cells. Several AChE inhibitors dose-dependently suppressed colony formation of HTB-38 cells in soft agar. This effect of AChE was confirmed with HTB-38 cells stably overexpressing AChE. In contrast, cell proliferation was not altered by the effective doses of these chemical inhibitors or by transfected AChE. Protection from cell cycle arrest consecutive to cancer cell detachment may be conveyed by changes in cell-matrix interactions. Reflective of such changes, the AChE overexpressing cells adhered more strongly to Fibronectin than did the vector controls. The AChE-dependent adhesion was RGD-dependent and accompanied by increased c-Myb DNA-binding, suggesting that AChE upregulates an Integrin receptor via c-Myb. In support of these observations, we find AChE message and protein to be expressed in a large fraction of colon cancers and in all colon tumor cell lines analyzed, but only rarely in normal colon specimens. Our results imply a dual role for AChE in colon cancer. While the anti-apoptotic effects of AChE may be protective against early stages of tumorigenesis, this gene product may support the later stages of transformation by enhancing anchorage independent growth. The induction of Integrins could render the cells independent of microenvironmental cues and override cell cycle arrest after deadhesion.
Collapse
Affiliation(s)
- Moyeenuddin Syed
- College of Pharmacy, University of Cincinnati Academic Health Center, 3225 Eden Avenue, Cincinnati, OH 45267-0004, USA
| | | | | | | |
Collapse
|
35
|
Weber GF. Molecular mechanisms of metastasis. Cancer Lett 2008; 270:181-90. [PMID: 18522865 DOI: 10.1016/j.canlet.2008.04.030] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2008] [Revised: 04/11/2008] [Accepted: 04/14/2008] [Indexed: 11/26/2022]
Abstract
Metastasis formation is an essential aspect of cancer, for which the molecular underpinning has long been subject to debate. Although the organ preference for dissemination is governed by tumor-host interactions on the epigenetic level there is a genetic basis to the ability of cancer cells to disseminate. Metastasis genes encode homing receptors, their ligands, and extracellular matrix-degrading proteinases, which jointly cause invasion and anchorage-independence. They are developmentally non-essential stress response genes that physiologically mediate the homing of immune system cells. Metastatic potential is conferred to cancer cells by aberrant expression or splicing of these genes. Oncogenes act upstream of metastasis genes. In cancer cells, oncogenic signaling activates distinct genetic programs leading to cell cycle progression and invasiveness, respectively. The expression of metastasis genes is regulated by multi-subunit transcription factor complexes. The identification of genes that direct cancer metastasis implicates them as candidate drug targets.
Collapse
Affiliation(s)
- Georg F Weber
- University of Cincinnati Academic Health Center, College of Pharmacy, 3225 Eden Avenue, Cincinnati, OH 45267-0004, USA.
| |
Collapse
|
36
|
Polyomavirus middle T antigen induces the transcription of osteopontin, a gene important for the migration of transformed cells. J Virol 2008; 82:4946-54. [PMID: 18337582 DOI: 10.1128/jvi.02650-07] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Middle T antigen (MT) is the principal oncoprotein of murine polyomavirus. Experiments on the acute immediate effects of MT expression on cellular RNA levels showed that expression of osteopontin (OPN) was strongly induced by MT expression. Osteopontin is a protein known to be associated with cancer. It has a role in tumor progression and invasion. Protein analysis confirmed that MT induced the secretion of OPN into the extracellular medium. Expression of antisense OPN RNA had no effect on the growth of MT-transformed cells. However, it had a strong effect on the ability of MT transformants to migrate or to fill a wound. Analysis of MT mutants implicated both the SHC and phosphatidylinositol 3-kinase pathways in OPN induction. Reporter assays showed that MT regulated the OPN promoter through two of its PEA3 (polyoma enhancer activator 3) sites. As critical PEA3 sites are also part of the polyomavirus enhancer, the same signaling important for viral replication also contributes to virally induced metastatic potential.
Collapse
|
37
|
Mirza M, Shaughnessy E, Hurley JK, Vanpatten KA, Pestano GA, He B, Weber GF. Osteopontin-c is a selective marker of breast cancer. Int J Cancer 2008; 122:889-97. [PMID: 17960616 DOI: 10.1002/ijc.23204] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
While the acquisition of invasiveness is a critical step in early stage breast carcinomas (DCIS), no established molecular markers reliably identify tumor progression. The metastasis gene osteopontin is subject to alternative splicing, which yields 3 messages, osteopontin-a, osteopontin-b and osteopontin-c. Osteopontin-c is selectively expressed in invasive, but not in noninvasive, breast tumor cell lines, and it effectively supports anchorage independence. We evaluated osteopontin-c as a biomarker. The RNA message for osteopontin-c was present in 16 of 20 breast cancers (80%), but was undetectable in 22 normal specimens obtained from reduction mammoplasty. In contrast, osteopontin-a RNA was expressed at various levels in all 20 breast cancers, 11 tumor-surrounding tissues and 21 normal samples. The splice variant osteopontin-b was present at barely detectable levels in 18 of 20 cancers and in 6 of 22 normal breasts. By immunohistochemistry, 66 of 69 normal breasts were negative, while 3 showed low level staining. Among the breast cancers, 43 of 56 cores (77%) stained positive for osteopontin-c. When correlated with tumor grade, the staining for osteopontin-c increased from grade 1 to grade 3. In a total of 178 breast specimens analyzed, osteopontin-c was present in 78% of cancers, 36% of surrounding tissues and 0% of normal tissues. Furthermore, osteopontin-c detects a higher fraction of breast cancers than estrogen receptor (ER), progesterone receptor or HER2. In conjunction, osteopontin-c, ER and HER2 reliably predict grade 2-3 breast cancer. Hence, osteopontin-c is a diagnostic and prognostic marker that may have value in a diagnostic panel together with conventional breast cancer markers.
Collapse
Affiliation(s)
- Mana Mirza
- College of Pharmacy, University of Cincinnati Medical Center, Cincinnati, OH 45267, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
YB-1 is a Transcription/Translation Factor that Orchestrates the Oncogenome by Hardwiring Signal Transduction to Gene Expression. TRANSLATIONAL ONCOGENOMICS 2007; 2:49-65. [PMID: 23641145 PMCID: PMC3634714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The Y-box Binding Protein-1 (YB-1) is a highly conserved oncogenic transcription/translation factor that is expressed in cancers affecting adults and children. It is now believed that YB-1 plays a causal role in the development of cancer given recent work showing that its expression drives the tumorigenesis in the mammary gland. In human breast cancers, YB-1 is associated with rapidly proliferating tumors that are highly aggressive. Moreover, expression of YB-1 promotes the growth of breast cancer cell lines both in monolayer and anchorage independent conditions. The involvement of YB-1 in breast cancer pathogenesis has made it a putative therapeutic target; however, the mechanism(s) that regulate YB-1 are poorly understood. This review first describes the oncogenic properties of YB-1 in cancer. It also highlights the importance of YB-1 in hardwiring signal transduction pathways to the regulation of genes involved in the development of cancer.
Collapse
|
39
|
Abstract
Metastatic spread of cancer cells is the main cause of death of breast cancer patients, and elucidation of the molecular mechanisms underlying this process is a major focus in cancer research. The identification of appropriate therapeutic targets and proof-of-concept experimentation involves an increasing number of experimental mouse models, including spontaneous and chemically induced carcinogenesis, tumor transplantation, and transgenic and/or knockout mice. Here we give a progress report on how mouse models have contributed to our understanding of the molecular processes underlying breast cancer metastasis and on how such experimentation can open new avenues to the development of innovative cancer therapy.
Collapse
Affiliation(s)
- Anna Fantozzi
- Institute of Biochemistry and Genetics, Department of Clinical-Biological Sciences (DKBW), Center of Biomedicine, University of Basel, Mattenstrasse 28, CH-4058 Basel, Switzerland
| | - Gerhard Christofori
- Institute of Biochemistry and Genetics, Department of Clinical-Biological Sciences (DKBW), Center of Biomedicine, University of Basel, Mattenstrasse 28, CH-4058 Basel, Switzerland
| |
Collapse
|
40
|
Vasko VV, Saji M. Molecular mechanisms involved in differentiated thyroid cancer invasion and metastasis. Curr Opin Oncol 2007; 19:11-7. [PMID: 17133106 DOI: 10.1097/cco.0b013e328011ab86] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
PURPOSE OF REVIEW The majority of patients with thyroid cancer have an excellent prognosis, however patients with extensive local invasion and distant metastasis frequently do not respond to standard treatments and have worsened prognosis. Understanding the specific mechanisms involved in thyroid cancer invasion and metastasis is critical in order to develop new treatments specifically targeted for these patients. RECENT FINDINGS The genetic basis for thyroid cancer initiation and development is well characterized, with the majority of studies implicating activation of the RAS-RAF-ERK and PI3K/PDK1/Akt signaling pathways. Over the last several years, data from a concerted effort to define the pathways involved in invasion and metastasis suggest that reactivation of embryonic pathways involved in cell movement, to include epithelial to mesenchymal transition and collective cell migration, may be involved in cancer cell migration and invasion. The previously identified thyroid oncogenes, BRAF, RET/PTC and Ras, appear to be important regulators of this process. SUMMARY The molecular mechanisms that control cell migration during embryological development, such as epithelial to mesenchymal transition, appear to be reactivated in invading thyroid cancer cells. Elucidation of the signal-transduction networks and molecules that are involved in thyroid cancer invasion may lead to novel therapeutic targets.
Collapse
Affiliation(s)
- Vasyl V Vasko
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA.
| | | |
Collapse
|
41
|
Matoba R, Niwa H, Masui S, Ohtsuka S, Carter MG, Sharov AA, Ko MS. Dissecting Oct3/4-regulated gene networks in embryonic stem cells by expression profiling. PLoS One 2006; 1:e26. [PMID: 17183653 PMCID: PMC1762406 DOI: 10.1371/journal.pone.0000026] [Citation(s) in RCA: 142] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2006] [Accepted: 10/03/2006] [Indexed: 12/11/2022] Open
Abstract
POU transcription factor Pou5f1 (Oct3/4) is required to maintain ES cells in an undifferentiated state. Here we show that global expression profiling of Oct3/4-manipulated ES cells delineates the downstream target genes of Oct3/4. Combined with data from genome-wide chromatin-immunoprecipitation (ChIP) assays, this analysis identifies not only primary downstream targets of Oct3/4, but also secondary or tertiary targets. Furthermore, the analysis also reveals that downstream target genes are regulated either positively or negatively by Oct3/4. Identification of a group of genes that show both activation and repression depending on Oct3/4 expression levels provides a possible mechanism for the requirement of appropriate Oct3/4 expression to maintain undifferentiated ES cells. As a proof-of-principle study, one of the downstream genes, Tcl1, has been analyzed in detail. We show that Oct3/4 binds to the promoter region of Tcl1 and activates its transcription. We also show that Tcl1 is involved in the regulation of proliferation, but not differentiation, in ES cells. These findings suggest that the global expression profiling of gene-manipulated ES cells can help to delineate the structure and dynamics of gene regulatory networks.
Collapse
Affiliation(s)
- Ryo Matoba
- Developmental Genomics and Aging Section, Laboratory of Genetics, National Institute on Aging, National Institutes of HealthBaltimore, Maryland, United States of America
| | - Hitoshi Niwa
- Laboratory of Pluripotent Cell Studies, RIKEN Center for Developmental BiologyKobe, Japan
| | - Shinji Masui
- Laboratory of Pluripotent Cell Studies, RIKEN Center for Developmental BiologyKobe, Japan
| | - Satoshi Ohtsuka
- Laboratory of Pluripotent Cell Studies, RIKEN Center for Developmental BiologyKobe, Japan
| | - Mark G. Carter
- Developmental Genomics and Aging Section, Laboratory of Genetics, National Institute on Aging, National Institutes of HealthBaltimore, Maryland, United States of America
| | - Alexei A. Sharov
- Developmental Genomics and Aging Section, Laboratory of Genetics, National Institute on Aging, National Institutes of HealthBaltimore, Maryland, United States of America
| | - Minoru S.H. Ko
- Developmental Genomics and Aging Section, Laboratory of Genetics, National Institute on Aging, National Institutes of HealthBaltimore, Maryland, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
42
|
Vandermoere F, El Yazidi-Belkoura I, Demont Y, Slomianny C, Antol J, Lemoine J, Hondermarck H. Proteomics exploration reveals that actin is a signaling target of the kinase Akt. Mol Cell Proteomics 2006; 6:114-24. [PMID: 17088265 DOI: 10.1074/mcp.m600335-mcp200] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The serine/threonine kinase Akt is a key mediator of cell survival and cell growth that is activated by most growth factors, but its downstream signaling largely remains to be elucidated. To identify signaling partners of Akt, we analyzed proteins co-immunoprecipitated with Akt in MCF-7 breast cancer cells. Mass spectrometry analysis (MALDI-TOF and MS-MS) of SDS-PAGE-separated Akt co-immunoprecipitates allowed the identification of 10 proteins: alpha -actinin, valosin-containing protein, inhibitor kappaB kinase, mortalin, tubulin beta, cytokeratin 8, actin, 14-3-3sigma, proliferating cell nuclear antigen, and heat shock protein HSP27. The identification of these putative Akt binding partners were validated with specific antibodies. Interestingly, the major protein band observed in Akt co-immunoprecipitates was found to be the cytoskeleton protein actin for which a 14-fold increase was observed in Akt-activated compared with non-activated conditions. The interaction between Akt and actin was further confirmed by reverse immunoprecipitation, and confocal microscopy demonstrated a co-localization specifically induced under growth factor stimulation. The use of wortmannin indicated a dependence on the phosphatidylinositol 3-kinase pathway. Using a phospho-Akt substrate antibody, the phosphorylation of actin on an Akt consensus site was detected upon growth factor stimulation, both in cellulo and in vitro, suggesting that actin is a substrate of Akt kinase activity. Interestingly, cortical remodeling of actin associated with cell migration was reversed by small interfering RNA directed against Akt, indicating the involvement of Akt in the dynamic reorganization of actin cytoskeleton germane to breast cancer cell migration. Together these data identify actin as a new functional target of Akt signaling.
Collapse
Affiliation(s)
- Franck Vandermoere
- ERI-8 INSERM (JE-2488), University of Sciences and Technologies Lille, Villeneuve d'Ascq, France
| | | | | | | | | | | | | |
Collapse
|
43
|
Galbaugh T, Cerrito MG, Jose CC, Cutler ML. EGF-induced activation of Akt results in mTOR-dependent p70S6 kinase phosphorylation and inhibition of HC11 cell lactogenic differentiation. BMC Cell Biol 2006; 7:34. [PMID: 16984645 PMCID: PMC1590014 DOI: 10.1186/1471-2121-7-34] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2006] [Accepted: 09/19/2006] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND HC11 mouse mammary epithelial cells differentiate in response to lactogenic hormone resulting in expression of milk proteins including beta-casein. Previous studies have shown that epidermal growth factor (EGF) blocks differentiation not only through activation of the Ras/Mek/Erk pathway but also implicated phosphatidylinositol-3-kinase (PI-3-kinase) signaling. The current study analyzes the mechanism of the PI-3-kinase pathway in an EGF-induced block of HC11 lactogenic differentiation. RESULTS HC11 and HC11-luci cells, which contain luciferase gene under the control of a beta-casein promotor, were treated with specific chemical inhibitors of signal transduction pathways or transiently infected/transfected with vectors encoding dominant negative-Akt (DN-Akt) or conditionally active-Akt (CA-Akt). The expression of CA-Akt inhibited lactogenic differentiation of HC11 cells, and the infection with DN-Akt adenovirus enhanced beta-casein transcription and rescued beta-casein promotor-regulated luciferase activity in the presence of EGF. Treatment of cells with Rapamycin, an inhibitor of mTOR, blocked the effects of EGF on beta-casein promotor driven luciferase activity as effectively as PI-3-kinase inhibitors. While expression of CA-Akt caused a constitutive activation of p70S6 kinase (p70S6K) in HC11 cells, the inhibition of either PI-3-kinase or mTOR abolished the activation of p70S6K by EGF. The activation of p70S6K by insulin or EGF resulted in the phosphorylation of ribosomal protein S6 (RPS6), elongation initiation factor 4E (elF4E) and 4E binding protein1 (4E-BP1). But lower levels of PI-3-K and mTOR inhibitors were required to block insulin-induced phosphorylation of RPS6 than EGF-induced phosphorylation, and insulin-induced phosphorylation of elF4E and 4E-BP1 was not completely mTOR dependent suggesting some diversity of signaling for EGF and insulin. In HC11 cells undergoing lactogenic differentiation the phosphorylation of p70S6K completely diminished by 12 hours, and this was partly attributable to dexamethasone, a component of lactogenic hormone mix. However, p70S6K phosphorylation persisted in the presence of lactogenic hormone and EGF, but the activation could be blocked by a PI-3-kinase inhibitor. CONCLUSION PI-3-kinase signaling contributes to the EGF block of lactogenic differentiation via Akt and p70S6K. The EGF-induced activation of PI-3-kinase-Akt-mTOR regulates phosphorylation of molecules including ribosomal protein S6, eIF4E and 4E-BP1 that influence translational control in HC11 cells undergoing lactogenic differentiation.
Collapse
Affiliation(s)
- Traci Galbaugh
- Department of Pathology, United States Military Cancer Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Maria Grazia Cerrito
- Department of Pathology, United States Military Cancer Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Cynthia C Jose
- Department of Pathology, United States Military Cancer Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Mary Lou Cutler
- Department of Pathology, United States Military Cancer Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| |
Collapse
|
44
|
Lukacova S, Overgaard J, Alsner J, Horsman MR. Strain and tumour specific variations in the effect of hypoxia on osteopontin levels in experimental models. Radiother Oncol 2006; 80:165-71. [PMID: 16920210 DOI: 10.1016/j.radonc.2006.07.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2006] [Revised: 07/17/2006] [Accepted: 07/19/2006] [Indexed: 11/18/2022]
Abstract
PURPOSE To investigate the relationship between tumour hypoxia and serum and tumour osteopontin (OPN) levels. MATERIALS AND METHODS Experiments were performed in CDF1 or C3H/Km mice implanted with a C3H mammary carcinoma (CDF1) or SCCVII squamous cell carcinoma (C3H/Km), respectively. Mice were either untreated or gassed with 10% oxygen for 1-72 h. Serum and tumour OPN levels were measured with an ELISA and tumour OPN mRNA levels using RT-PCR. Tumour oxygenation was estimated using the Eppendorf histograph with the percentage of pO(2) values <or=5 mm Hg (HF5) as the endpoint. RESULTS OPN levels were 50-fold higher in the serum of non-tumour bearing CDF1 mice compared to C3H/Km mice. A tumour related increase in serum OPN levels was observed in CDF1 but not in C3H/Km mice. Low oxygen breathing increased HF5 in both tumour models and in the C3H mammary carcinoma model both serum and tumour OPN decreased after prolonged hypoxia (24h and more). When 12h of hypoxia was followed by 24h reoxygenation there was a twofold increase in serum OPN levels. No changes were observed in the SCCVII model. No changes in tumour OPN mRNA expression were observed during hypoxia and reoxygenation in these tumour models. CONCLUSION Clear strain and tumour specific differences in the effect of hypoxia on OPN levels have been observed in two different mouse tumour models. These data emphasize the complexity in the relationship between poor oxygenation (and/or reoxygenation) of tumours and serum levels of OPN.
Collapse
Affiliation(s)
- Slavka Lukacova
- Department of Experimental Clinical Oncology, Aarhus university Hospital, Denmark
| | | | | | | |
Collapse
|
45
|
Cook AC, Chambers AF, Turley EA, Tuck AB. Osteopontin Induction of Hyaluronan Synthase 2 Expression Promotes Breast Cancer Malignancy. J Biol Chem 2006; 281:24381-9. [PMID: 16807238 DOI: 10.1074/jbc.m602694200] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Osteopontin (OPN) is a tumor-associated, secreted phosphoprotein that has been implicated in breast cancer progression and metastasis. Research concerning how OPN functions in tumor progression has led to the identification of a limited number of genes that contribute functionally to OPN-induced cellular behaviors. Recent microarray analysis, comparing 21NT breast cancer cells transfected to constitutively overexpress OPN with control cells, revealed hyaluronan synthase 2 (HAS2) to be a gene highly up-regulated in OPN-overexpressing cells. In this study, we further examined the relationship between OPN and HAS2. We show that 21NT OPN-transfected cells express high levels of HAS2, which is associated with increased HA production and matrix retention and is necessary for tumor cell adhesion to bone marrow endothelial cells and anchorage-independent growth. Finally, stable transfection of antisense HAS2 into 21NT cells overexpressing OPN resulted in a reduction in HAS2 expression, HA production, and pericellular retention. Antisense-mediated down-regulation of HAS2 also resulted in a significant decrease in cellular proliferation and colony growth in soft agar. To our knowledge, this is the first report of the ability of OPN to regulate HAS2 expression and HA production in breast cancer cells and further illustrates a unique functional relationship by which enhanced HA production facilitates OPN-mediated cell behaviors.
Collapse
Affiliation(s)
- Amy C Cook
- Department of Pathology, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | | | | | | |
Collapse
|
46
|
He B, Mirza M, Weber GF. An osteopontin splice variant induces anchorage independence in human breast cancer cells. Oncogene 2006; 25:2192-202. [PMID: 16288209 DOI: 10.1038/sj.onc.1209248] [Citation(s) in RCA: 145] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In malignant tumors, metastasis genes are typically deregulated by aberrant expression or splicing. Osteopontin is expressed at high levels by various cancers and contributes importantly to their invasive potential. In contrast, osteopontin derived from host cells induces cellular immunity and could bolster antitumor protection by cytotoxic T lymphocytes. Here we show that breast cancer cells express multiple splice variants of osteopontin. According to RT-PCR analysis of human breast tissue specimens, the splice variant osteopontin-c is a highly specific marker for transformed cells, which is not expressed in their surrounding normal tissue. The full-length form of osteopontin aggregates in the presence of physiologic amounts of calcium and, in this state, leads to enhanced cell adhesion. Ostensibly, this effect is inhibitory for tumor cell dissemination. The shortest splice variant, osteopontin-c, does not aggregate in the presence of calcium and enhances clone formation in soft agar. According to microarray analysis, osteopontin-c induces the expression of oxidoreductases, consistent with protection from anoikis during anchorage-independent growth. These studies define a third functional domain of osteopontin, beside the C-terminal CD44-binding site and the central integrin-binding site. They also provide evidence for a bifunctional character of osteopontin, with the soluble form supporting invasiveness and the aggregated form promoting adhesion.
Collapse
Affiliation(s)
- B He
- Molecular Oncology Research Institute, New England Medical Center, Boston, MA, USA
| | | | | |
Collapse
|
47
|
Bilchick KC, Saha SK, Mikolajczyk E, Cope L, Ferguson WJ, Yu W, Girouard S, Kass DA. Differential regional gene expression from cardiac dyssynchrony induced by chronic right ventricular free wall pacing in the mouse. Physiol Genomics 2006; 26:109-15. [PMID: 16670254 DOI: 10.1152/physiolgenomics.00281.2005] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Routine clinical right ventricular pacing generates left ventricular dyssynchrony manifested by early septal shortening followed by late lateral contraction, which, in turn, reciprocally stretches the septum. Dyssynchrony is disadvantageous to cardiac mechanoenergetics and worsens clinical prognosis, yet little is known about its molecular consequences. Here, we report the influence of cardiac dyssynchrony on regional cardiac gene expression in mice. Mice were implanted with a custom-designed miniature cardiac pacemaker and subjected to 1-wk overdrive right ventricular free wall pacing (720 beats/min, baseline heart rate 520-620 beats/min) to generate dyssynchrony (pacemaker: 3-V lithium battery, rate programmable, 1.5 g, bipolar lead). Electrical capture was confirmed by pulsed-wave Doppler and dyssynchrony by echocardiography. Gene expression from the left ventricular septal and lateral wall myocardium was assessed by microarray (dual-dye method, Agilent) using oligonucleotide probes and dye swap. Identical analysis was applied to four synchronously contracting controls. Of the 22,000 genes surveyed, only 18 genes displayed significant (P < 0.01) differential expression between septal/lateral walls >1.5 times that in synchronous controls. Gene changes were confirmed by quantitative PCR with excellent correlations. Most of the genes (n = 16) showed greater septal expression. Of particular interest were seven genes coding proteins involved with stretch responses, matrix remodeling, stem cell differentiation to myocyte lineage, and Purkinje fiber differentiation. One week of iatrogenic cardiac dyssynchrony triggered regional differential expression in relatively few select genes. Such analysis using a murine implantable pacemaker should facilitate molecular studies of cardiac dyssynchrony and help elucidate novel mechanisms by which stress/stretch stimuli due to dyssynchrony impact the normal and failing heart.
Collapse
Affiliation(s)
- Kenneth C Bilchick
- Division of Cardiology, Department of Medicine, School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Zhu Y, Denhardt DT, Cao H, Sutphin PD, Koong AC, Giaccia AJ, Le QT. Hypoxia upregulates osteopontin expression in NIH-3T3 cells via a Ras-activated enhancer. Oncogene 2005; 24:6555-63. [PMID: 16007184 DOI: 10.1038/sj.onc.1208800] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Osteopontin (OPN) is a secreted phosphoglycoprotein that has been linked to tumor progression and survival in several solid tumors, including head and neck cancers. Previous studies showed that OPN expression is induced by tumor hypoxia, and its plasma levels can serve as a surrogate marker for tumor hypoxia and treatment outcome in head and neck cancer patients. In this study, we investigate the transcriptional mechanism by which hypoxia enhances OPN expression. We found that OPN is induced in head and neck squamous cell carcinoma (HNSCC) cell lines and in NIH3T3 cells by hypoxia at both mRNA and protein levels in a time-dependent manner. Actinomycin D chase experiments showed that hypoxic induction of OPN was not due to increased mRNA stability. Deletion analyses of the mouse OPN promoter regions indicated that a ras-activated enhancer (RAE) located at -731 to -712 relative to the transcription start site was essential for hypoxia-enhanced OPN transcription. Using electrophoretic mobility shift assays with the RAE DNA sequence, we found that hypoxia induced sequence-specific DNA-binding complexes. Furthermore, hypoxia and ras exposure resulted in an additive induction of OPN protein and mRNA levels that appeared to be mediated by the RAE. Induction of OPN through the RAE element by hypoxia is mediated by an Akt-kinase signaled pathway as decreasing Akt levels with dominant negative constructs resulted in inhibition of OPN induction by hypoxia. Taken together, these results have identified a new hypoxia responsive transcriptional enhancer that is regulated by Akt signaling.
Collapse
Affiliation(s)
- Yonghua Zhu
- Department of Radiation Oncology, 875 Blake Wilbur Dr, R. CC-G228, Stanford University, Stanford, CA 94305, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Zhang Z, Li M, Rayburn ER, Hill DL, Zhang R, Wang H. Oncogenes as novel targets for cancer therapy (part II): Intermediate signaling molecules. ACTA ACUST UNITED AC 2005; 5:247-57. [PMID: 16078861 DOI: 10.2165/00129785-200505040-00005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
This is the second part of a four-part review on potential therapeutic targeting of oncogenes. The previous part introduced the new technologies responsible for the advancement of oncogene identification, target validation, and drug design. Because of such advances, new specific and more efficient therapeutic agents can be developed for cancer. This part of the review continues the exploration of various oncogenes, which we have grouped within seven categories: growth factors, tyrosine kinases, intermediate signaling molecules, transcription factors, cell cycle regulators, DNA damage repair genes, and genes involved in apoptosis. Part I included a discussion of growth factors and tyrosine kinases. This portion of the review covers intermediate signaling molecules and the various strategies used to inhibit their expression or decrease their activities.
Collapse
Affiliation(s)
- Zhuo Zhang
- Department of Pharmacology and Toxicology and Division of Clinical Pharmacology, Birmingham, Alabama 35294-0019, USA
| | | | | | | | | | | |
Collapse
|
50
|
Borowsky AD, Namba R, Young LJT, Hunter KW, Hodgson JG, Tepper CG, McGoldrick ET, Muller WJ, Cardiff RD, Gregg JP. Syngeneic mouse mammary carcinoma cell lines: two closely related cell lines with divergent metastatic behavior. Clin Exp Metastasis 2005; 22:47-59. [PMID: 16132578 DOI: 10.1007/s10585-005-2908-5] [Citation(s) in RCA: 161] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2004] [Accepted: 02/24/2005] [Indexed: 12/01/2022]
Abstract
Two cell lines, Met-1(fvb2) and DB-7(fvb2), with different metastatic potential, were derived from mammary carcinomas in FVB/N-Tg(MMTV-PyVmT) and FVB/N-Tg(MMTV-PyVmT ( Y315F/Y322F )) mice, transplanted into syngeneic FVB/N hosts and characterized. The lines maintain a stable morphological and biological phenotype after multiple rounds of in vitro culture and in vivo transplantation. The Met-1(fvb2) line derived from a FVB/N-Tg(MMTV-PyVmT) tumor exhibits invasive growth and 100% metastases when transplanted into the females FVB/N mammary fat pad. The DB-7(fvb2) line derived from the FVB/N-Tg(MMTV-PyVmT ( Y315F/Y322F )) with a "double base" modification at Y315F/Y322F exhibits more rapid growth when transplanted into the mammary fat pad, but a lower rate of metastasis (17%). The Met1(fvb2) cells show high activation of AKT, while DB-7(fvb2) cells show very low levels of AKT activation. The DNA content and gene expression levels of both cell lines are stable over multiple generations. Therefore, these two cell lines provide a stable, reproducible resource for the study of metastasis modulators, AKT molecular pathway interactions, and gene target and marker discovery.
Collapse
Affiliation(s)
- Alexander D Borowsky
- Department of Medical Pathology and Laboratory Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|