1
|
Schmithals C, Kakoschky B, Denk D, von Harten M, Klug JH, Hintermann E, Dropmann A, Hamza E, Jacomin AC, Marquardt JU, Zeuzem S, Schirmacher P, Herrmann E, Christen U, Vogl TJ, Waidmann O, Dooley S, Finkelmeier F, Piiper A. Tumour-specific activation of a tumour-blood transport improves the diagnostic accuracy of blood tumour markers in mice. EBioMedicine 2024; 105:105178. [PMID: 38889481 PMCID: PMC11237870 DOI: 10.1016/j.ebiom.2024.105178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 04/12/2024] [Accepted: 05/17/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND The accuracy of blood-based early tumour recognition is compromised by signal production at non-tumoral sites, low amount of signal produced by small tumours, and variable tumour production. Here we examined whether tumour-specific enhancement of vascular permeability by the particular tumour homing peptide, iRGD, which carries dual function of binding to integrin receptors overexpressed in the tumour vasculature and is known to promote extravasation via neuropilin-1 receptor upon site-specific cleavage, might be useful to improve blood-based tumour detection by inducing a yet unrecognised vice versa tumour-to-blood transport. METHODS To detect an iRGD-induced tumour-to-blood transport, we examined the effect of intravenously injected iRGD on blood levels of α-fetoprotein (AFP) and autotaxin in several mouse models of hepatocellular carcinoma (HCC) or in mice with chronic liver injury without HCC, and on prostate-specific antigen (PSA) levels in mice with prostate cancer. FINDINGS Intravenously injected iRGD rapidly and robustly elevated the blood levels of AFP in several mouse models of HCC, but not in mice with chronic liver injury. The effect was primarily seen in mice with small tumours and normal basal blood AFP levels, was attenuated by an anti-neuropilin-1 antibody, and depended on the concentration gradient between tumour and blood. iRGD treatment was also able to increase blood levels of autotaxin in HCC mice, and of PSA in mice with prostate cancer. INTERPRETATION We conclude that iRGD induces a tumour-to-blood transport in a tumour-specific fashion that has potential of improving diagnosis of early stage cancer. FUNDING Deutsche Krebshilfe, DKTK, LOEWE-Frankfurt Cancer Institute.
Collapse
Affiliation(s)
- Christian Schmithals
- Goethe University Frankfurt, University Hospital, Medical Clinic 1, Frankfurt am Main, Germany
| | - Bianca Kakoschky
- Goethe University Frankfurt, University Hospital, Medical Clinic 1, Frankfurt am Main, Germany
| | - Dominic Denk
- Goethe University Frankfurt, University Hospital, Medical Clinic 1, Frankfurt am Main, Germany; Frankfurt Cancer Institute, Goethe University Frankfurt, University Hospital, Frankfurt am Main, Germany
| | - Maike von Harten
- Goethe University Frankfurt, University Hospital, Medical Clinic 1, Frankfurt am Main, Germany
| | - Jan Henrik Klug
- Goethe University Frankfurt, University Hospital, Medical Clinic 1, Frankfurt am Main, Germany
| | - Edith Hintermann
- Pharmazentrum Frankfurt / ZAFES, Goethe University Frankfurt, University Hospital, Frankfurt am Main, Germany
| | - Anne Dropmann
- Molecular Hepatology-Alcohol Associated Diseases, Department of Medicine II, Medical Faculty Mannheim, University of Heidelberg, Germany
| | - Eman Hamza
- Goethe University Frankfurt, University Hospital, Medical Clinic 1, Frankfurt am Main, Germany; Suez University, Faculty of Science, Zoology Department, Suez, Egypt
| | - Anne Claire Jacomin
- Frankfurt Cancer Institute, Goethe University Frankfurt, University Hospital, Frankfurt am Main, Germany; Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany
| | - Jens U Marquardt
- Department of Medicine I, University Medical Centre Schleswig-Holstein - Campus Lübeck, Lübeck, Germany
| | - Stefan Zeuzem
- Goethe University Frankfurt, University Hospital, Medical Clinic 1, Frankfurt am Main, Germany; German Cancer Consortium (DKTK), Partner Site Frankfurt/M., a Partnership Between DKFZ and University Hospital Frankfurt/M., Germany
| | | | - Eva Herrmann
- Goethe University Frankfurt, University Hospital, Institute of Biostatistics and Mathematical Modelling, Germany
| | - Urs Christen
- Pharmazentrum Frankfurt / ZAFES, Goethe University Frankfurt, University Hospital, Frankfurt am Main, Germany
| | - Thomas J Vogl
- Goethe University Frankfurt, University Hospital, Institute for Diagnostic and Interventional Radiology, Germany
| | - Oliver Waidmann
- Goethe University Frankfurt, University Hospital, Medical Clinic 1, Frankfurt am Main, Germany; Centrum für Hämatologie und Onkologie Bethanien, Frankfurt/Main, Germany
| | - Steven Dooley
- Molecular Hepatology-Alcohol Associated Diseases, Department of Medicine II, Medical Faculty Mannheim, University of Heidelberg, Germany
| | - Fabian Finkelmeier
- Goethe University Frankfurt, University Hospital, Medical Clinic 1, Frankfurt am Main, Germany; Frankfurt Cancer Institute, Goethe University Frankfurt, University Hospital, Frankfurt am Main, Germany
| | - Albrecht Piiper
- Goethe University Frankfurt, University Hospital, Medical Clinic 1, Frankfurt am Main, Germany; Frankfurt Cancer Institute, Goethe University Frankfurt, University Hospital, Frankfurt am Main, Germany; German Cancer Consortium (DKTK), Partner Site Frankfurt/M., a Partnership Between DKFZ and University Hospital Frankfurt/M., Germany.
| |
Collapse
|
2
|
Hosseiniyan Khatibi SM, Najjarian F, Homaei Rad H, Ardalan M, Teshnehlab M, Zununi Vahed S, Pirmoradi S. Key therapeutic targets implicated at the early stage of hepatocellular carcinoma identified through machine-learning approaches. Sci Rep 2023; 13:3840. [PMID: 36882466 PMCID: PMC9992672 DOI: 10.1038/s41598-023-30720-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 02/28/2023] [Indexed: 03/09/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most frequent type of primary liver cancer. Early-stage detection plays an essential role in making treatment decisions and identifying dominant molecular mechanisms. We utilized machine learning algorithms to find significant mRNAs and microRNAs (miRNAs) at the early and late stages of HCC. First, pre-processing approaches, including organization, nested cross-validation, cleaning, and normalization were applied. Next, the t-test/ANOVA methods and binary particle swarm optimization were used as a filter and wrapper method in the feature selection step, respectively. Then, classifiers, based on machine learning and deep learning algorithms were utilized to evaluate the discrimination power of selected features (mRNAs and miRNAs) in the classification step. Finally, the association rule mining algorithm was applied to selected features for identifying key mRNAs and miRNAs that can help decode dominant molecular mechanisms in HCC stages. The applied methods could identify key genes associated with the early (e.g., Vitronectin, thrombin-activatable fibrinolysis inhibitor, lactate dehydrogenase D (LDHD), miR-590) and late-stage (e.g., SPRY domain containing 4, regucalcin, miR-3199-1, miR-194-2, miR-4999) of HCC. This research could establish a clear picture of putative candidate genes, which could be the main actors at the early and late stages of HCC.
Collapse
Affiliation(s)
- Seyed Mahdi Hosseiniyan Khatibi
- Kidney Research Center, Tabriz University of Medical Sciences, Daneshgah Street, Tabriz, 51665118, Iran.,Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Niyayesh Blvd., Tabriz, Iran.,Rahat Breath and Sleep Research Center, Tabriz University of Medical Science, Tabriz, Iran
| | - Farima Najjarian
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Homaei Rad
- Rahat Breath and Sleep Research Center, Tabriz University of Medical Science, Tabriz, Iran
| | - Mohammadreza Ardalan
- Kidney Research Center, Tabriz University of Medical Sciences, Daneshgah Street, Tabriz, 51665118, Iran
| | - Mohammad Teshnehlab
- Department of Electric and Computer Engineering, K.N. Toosi University of Technology, Tehran, Iran
| | - Sepideh Zununi Vahed
- Kidney Research Center, Tabriz University of Medical Sciences, Daneshgah Street, Tabriz, 51665118, Iran.
| | - Saeed Pirmoradi
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Niyayesh Blvd., Tabriz, Iran.
| |
Collapse
|
3
|
Perrins RD, McCarthy LA, Robinson A, Spry KL, Cognet V, Ferreira A, Porter J, Garcίa CE, Rodriguez MÁ, Lopez D, Perera I, Conlon K, Barrientos A, Coulter T, Pace A, Hale SJM, Ferrari E, Bachrati CZ. Targeting Ultrasmall Gold Nanoparticles with cRGD Peptide Increases the Uptake and Efficacy of Cytotoxic Payload. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:nano12224013. [PMID: 36432299 PMCID: PMC9696180 DOI: 10.3390/nano12224013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/07/2022] [Accepted: 11/14/2022] [Indexed: 05/06/2023]
Abstract
Cyclic arginyl-glycyl-aspartic acid peptide (cRGD) peptides show a high affinity towards αVβ3 integrin, a receptor overexpressed in many cancers. We aimed to combine the versatility of ultrasmall gold nanoparticles (usGNP) with the target selectivity of cRGD peptide for the directed delivery of a cytotoxic payload in a novel design. usGNPs were synthesized with a modified Brust-Schiffrin method and functionalized via amide coupling and ligand exchange and their uptake, intracellular trafficking, and toxicity were characterized. Our cRGD functionalized usGNPs demonstrated increased cellular uptake by αVβ3 integrin expressing cells, are internalized via clathrin-dependent endocytosis, accumulated in the lysosomes, and when loaded with mertansine led to increased cytotoxicity. Targeting via cRGD functionalization provides a mechanism to improve the efficacy, tolerability, and retention of therapeutic GNPs.
Collapse
Affiliation(s)
| | - Lee-Anne McCarthy
- School of Life Sciences, University of Lincoln, Joseph Banks Laboratories, Green Lane, Lincoln LN6 7DL, UK
| | - Angela Robinson
- Midatech Pharma Plc, 1 Caspian Point, Caspian Way, Cardiff CF10 4DQ, UK
| | - Kelly L. Spry
- Midatech Pharma Plc, 1 Caspian Point, Caspian Way, Cardiff CF10 4DQ, UK
| | - Valentin Cognet
- Midatech Pharma Plc, 1 Caspian Point, Caspian Way, Cardiff CF10 4DQ, UK
| | - Avelino Ferreira
- Midatech Pharma Plc, 1 Caspian Point, Caspian Way, Cardiff CF10 4DQ, UK
| | - John Porter
- Midatech Pharma Plc, 1 Caspian Point, Caspian Way, Cardiff CF10 4DQ, UK
| | | | | | - Diana Lopez
- Midatech Pharma Plc, 1 Caspian Point, Caspian Way, Cardiff CF10 4DQ, UK
| | - Ibon Perera
- Midatech Pharma Plc, 1 Caspian Point, Caspian Way, Cardiff CF10 4DQ, UK
| | - Kelly Conlon
- Midatech Pharma Plc, 1 Caspian Point, Caspian Way, Cardiff CF10 4DQ, UK
| | - Africa Barrientos
- Midatech Pharma Plc, 1 Caspian Point, Caspian Way, Cardiff CF10 4DQ, UK
| | - Tom Coulter
- Midatech Pharma Plc, 1 Caspian Point, Caspian Way, Cardiff CF10 4DQ, UK
| | - Alessandro Pace
- Midatech Pharma Plc, 1 Caspian Point, Caspian Way, Cardiff CF10 4DQ, UK
| | - Sarah J. M. Hale
- Midatech Pharma Plc, 1 Caspian Point, Caspian Way, Cardiff CF10 4DQ, UK
| | - Enrico Ferrari
- School of Life Sciences, University of Lincoln, Joseph Banks Laboratories, Green Lane, Lincoln LN6 7DL, UK
| | - Csanad Z. Bachrati
- School of Life Sciences, University of Lincoln, Joseph Banks Laboratories, Green Lane, Lincoln LN6 7DL, UK
- Correspondence: ; Tel.: +44-1522-886787
| |
Collapse
|
4
|
Zhao W, Lv M, Yang X, Zhou J, Xing B, Zhang Z. OUP accepted manuscript. Carcinogenesis 2022; 43:766-778. [PMID: 35436337 DOI: 10.1093/carcin/bgac035] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 03/20/2022] [Accepted: 04/14/2022] [Indexed: 12/24/2022] Open
Affiliation(s)
- Wei Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Cell Biology, Peking University Cancer Hospital and Institute, Beijing 100142, P. R. China
| | - Mengzhu Lv
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Cell Biology, Peking University Cancer Hospital and Institute, Beijing 100142, P. R. China
| | - Xueying Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Cell Biology, Peking University Cancer Hospital and Institute, Beijing 100142, P. R. China
| | - Jing Zhou
- Hemorheology Center, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100083, P. R. China
| | - Baocai Xing
- Department of Hepatobiliary Surgery I, Peking University Cancer Hospital and Institute, Beijing 100142, P. R. China
| | - Zhiqian Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Cell Biology, Peking University Cancer Hospital and Institute, Beijing 100142, P. R. China
| |
Collapse
|
5
|
Expression of Platelet Derived Growth Factor a, Its Receptor, and Integrin Subunit Alpha V in Feline Injection-Site Sarcomas. ACTA VET-BEOGRAD 2021. [DOI: 10.2478/acve-2021-0002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Injection-site sarcomas are considered an important entity in veterinary oncology, especially in cats. The current study investigated the immunohistochemical expression of platelet-derived growth factor A (PDGFA), platelet-derived growth factor receptor-α (PDGFR-α), and integrin alpha subunit v in feline injection-site sarcomas (FISS). A total of 14 paraffin-embedded tissue samples previously diagnosed as soft tissue sarcomas were selected from the departmental archive and sectioned at 5µm using a microtome. Tissue sections were stained with Hematoxylin & Eosin, Alcian blue-Periodic Acid Schiff (AB-PAS), Masson’s Trichrome, and immunohistochemically for PDGFA, PDGR-α, and integrin alpha v. The majority of the tumors were fibrosarcomas (n=13/14), except one case of myxoid liposarcoma. The tumors occurred at sites commonly used for injections or vaccine administration. Histologically, tumors were characterized by peripheral infiltration of mononuclear cells mainly lymphocytes, variable sized central necrotic areas, and many multinucleated tumor giant cells. AB-PAS staining revealed the presence of myxoid or mucinous areas in (8/14) tumors while trichrome staining demonstrated a variable amount of collagenous stroma in 12/14 tumors. Vimentin immunoreactivity was observed in all the tumors while smooth muscle actin and muscle actin staining was noticed in four and two cases, respectively. PDGFA and PDGFR-α immunoexpression was observed in all 14 cases while integrin alpha v in 13/14 cases. The results of the current study indicate that fibrosarcoma is the major morphologic phenotype of FISS. The PDGFA, its receptor, and integrin alpha v immunoexpression are increased in these tumors in cats which elucidates their role in the pathogenesis of FISS.
Collapse
|
6
|
Ahmed I, Sozmen M. Expression of PDGF-A, PDGFRA, integrin subunit alpha V and selectin E is increased in canine cutaneous fibrosarcomas. Biotech Histochem 2020; 96:546-554. [PMID: 33034211 DOI: 10.1080/10520295.2020.1832256] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
We investigated the expression of platelet derived growth factor alpha (PDGFA); its receptor, PDGFRA; integrin subunit alpha V; and selectin E in cutaneous fibrosarcomas in dogs. Ten cases of canine fibrosarcomas were obtained from the archive of the Department of Pathology, Ondokuz Mayis University, Samsun. Tissue sections were cut and stained with hematoxylin and eosin, Alcian blue-periodic acid Schiff, Masson's trichrome, and also immunostained. Eight tumors classified as spontaneous fibrosarcomas exhibited interwoven bundles of spindle shaped cells with oval to plump nuclei and scant cytoplasm, while two tumors exhibited features of injection site fibrosarcoma with peripheral infiltration of mononuclear cells and intratumor necrosis. We found that neoplastic cells from all cases exhibited cytoplasmic expression of PDGFA, and cytoplasmic and nuclear staining for PDGFRA. Integrin subunit alpha V immunostaining was observed in all cases, while selectin E expression was observed in vascular endothelial cells and neoplastic cells. A significant positive correlation was found between the expression of PDGFA and integrin subunit alpha V. Our findings indicate that PDGFA, PDGFRA, integrin subunit alpha V and selectin E are expressed strongly in canine cutaneous fibrosarcomas and may contribute to tumor progression.
Collapse
Affiliation(s)
- Ishtiaq Ahmed
- Department of Pathology, University of Veterinary and Animal Sciences , Lahore, Pakistan
| | - Mahmut Sozmen
- Department of Pathology, Faculty of Veterinary Medicine, Ondokuz Mayis University , Samsun, Turkey
| |
Collapse
|
7
|
Capasso D, Del Gatto A, Comegna D, Russo L, Fattorusso R, Saviano M, Di Gaetano S, Zaccaro L. Selective Targeting of αvβ5 Integrin in HepG2 Cell Line by RGDechi15D Peptide. Molecules 2020; 25:molecules25184298. [PMID: 32961684 PMCID: PMC7570809 DOI: 10.3390/molecules25184298] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 08/25/2020] [Accepted: 09/11/2020] [Indexed: 12/21/2022] Open
Abstract
Recently, the research community has become increasingly concerned with the receptor αvβ5, a member of the well-known integrin family. Different ongoing studies have evidenced that αvβ5 integrin regulates not only physiological processes but also a wide array of pathological events, suggesting the receptor as a valuable biomarker to specifically target for therapeutic/diagnostic purposes. Remarkably, in some tumors the involvement of the receptor in cell proliferation, tumor dissemination and angiogenesis is well-documented. In this scenario, the availability of a selective αvβ5 antagonist without ‘off-target’ protein effects may improve survival rate in patients with highly aggressive tumors, such as hepatocellular carcinoma. We recently reported a cyclic peptide, RGDechi15D, obtained by structure-activity studies. To our knowledge it represents the first peptide-based molecule reported in the literature able to specifically bind αvβ5 integrin and not cross react with αvβ3. Here we demonstrated the ability of the peptide to diminish both adhesion and invasion of HepG2 cells, an in vitro model system for hepatocellular carcinoma, to reduce the cell proliferation through an apoptotic process, and to interfere with the PI3K pathway. The peptide, also decreases the formation of new vessels in endothelial cells. Taken together these results indicate that the peptide can be considered a promising molecule with properties suited to be assessed in the future for its validation as a selective therapeutic/diagnostic weapon in hepatocarcinoma.
Collapse
Affiliation(s)
- Domenica Capasso
- CESTEV, University of Naples “Federico II”, 80145 Naples, Italy;
- CIRPeB, University of Naples “Federico II”, 80134 Naples, Italy; (A.D.G.); (R.F.); (M.S.)
| | - Annarita Del Gatto
- CIRPeB, University of Naples “Federico II”, 80134 Naples, Italy; (A.D.G.); (R.F.); (M.S.)
- Institute of Biostructures and Bioimaging, CNR, 80134 Naples, Italy;
| | - Daniela Comegna
- Institute of Biostructures and Bioimaging, CNR, 80134 Naples, Italy;
| | - Luigi Russo
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy;
| | - Roberto Fattorusso
- CIRPeB, University of Naples “Federico II”, 80134 Naples, Italy; (A.D.G.); (R.F.); (M.S.)
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy;
| | - Michele Saviano
- CIRPeB, University of Naples “Federico II”, 80134 Naples, Italy; (A.D.G.); (R.F.); (M.S.)
- Institute of Crystallography, CNR, 70126 Bari, Italy
| | - Sonia Di Gaetano
- CIRPeB, University of Naples “Federico II”, 80134 Naples, Italy; (A.D.G.); (R.F.); (M.S.)
- Institute of Biostructures and Bioimaging, CNR, 80134 Naples, Italy;
- Correspondence: (S.D.G.); (L.Z.)
| | - Laura Zaccaro
- CIRPeB, University of Naples “Federico II”, 80134 Naples, Italy; (A.D.G.); (R.F.); (M.S.)
- Institute of Biostructures and Bioimaging, CNR, 80134 Naples, Italy;
- Correspondence: (S.D.G.); (L.Z.)
| |
Collapse
|
8
|
Shimagaki T, Yoshio S, Kawai H, Sakamoto Y, Doi H, Matsuda M, Mori T, Osawa Y, Fukai M, Yoshida T, Ma Y, Akita T, Tanaka J, Taketomi A, Hanayama R, Yoshizumi T, Mori M, Kanto T. Serum milk fat globule-EGF factor 8 (MFG-E8) as a diagnostic and prognostic biomarker in patients with hepatocellular carcinoma. Sci Rep 2019; 9:15788. [PMID: 31673081 PMCID: PMC6823494 DOI: 10.1038/s41598-019-52356-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 10/16/2019] [Indexed: 02/07/2023] Open
Abstract
Current serum hepatocellular carcinoma (HCC) biomarkers are insufficient for early diagnosis. We aimed to clarify whether serum MFG-E8 can serve as a diagnostic or prognostic biomarker of HCC. Serum MFG-E8 levels of 282 HCC patients, who underwent primary hepatectomy, were examined by ELISA. We also quantified serum MFG-E8 levels in patients with chronic hepatitis (CH), liver cirrhosis (LC), as well as in healthy volunteers (HVs). Serum MFG-E8 levels were significantly lower in HCC patients than in HVs regardless of the etiology of liver disease (3.6 ± 0.1 vs 5.8 ± 0.2 ng/mL, p < 0.0001), and recovered after treatment of HCC. Serum MFG-E8 levels in CH and LC patients were comparable to those in HVs. Serum MFG-E8 could detect HCCs, even α-fetoprotein (AFP)-negative or des-γ-carboxy prothrombin (DCP)-negative HCCs, in CH and LC patients. Our new HCC prediction model using MFG-E8 and DCP (Logit(p) = 2.619 − 0.809 × serum MFG-E8 + 0.0226 × serum DCP) distinguished HCC patients from CH and LC patients with an area under the curve of 0.923, a sensitivity of 81.1%, and a specificity of 89.8%. Futhermore, low preoperative serum MFG-E8 was an independent predictor of poor overall survival. Thus, serum MFG-E8 could serve as a feasible diagnostic and prognostic biomarker for HCC.
Collapse
Affiliation(s)
- Tomonari Shimagaki
- Department of Liver Disease, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Ichikawa, Japan.,Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Sachiyo Yoshio
- Department of Liver Disease, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Ichikawa, Japan.
| | - Hironari Kawai
- Department of Liver Disease, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Ichikawa, Japan
| | - Yuzuru Sakamoto
- Department of Liver Disease, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Ichikawa, Japan
| | - Hiroyoshi Doi
- Department of Liver Disease, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Ichikawa, Japan
| | - Michitaka Matsuda
- Department of Liver Disease, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Ichikawa, Japan
| | - Taizo Mori
- Department of Liver Disease, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Ichikawa, Japan
| | - Yosuke Osawa
- Department of Liver Disease, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Ichikawa, Japan
| | - Moto Fukai
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Takeshi Yoshida
- Department of Immunology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Yunfei Ma
- Department of Immunology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Tomoyuki Akita
- Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Junko Tanaka
- Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Akinobu Taketomi
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Rikinari Hanayama
- Department of Immunology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Tomoharu Yoshizumi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masaki Mori
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tatsuya Kanto
- Department of Liver Disease, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Ichikawa, Japan.
| |
Collapse
|
9
|
Weidle UH, Epp A, Birzele F, Brinkmann U. The Functional Role of Prostate Cancer Metastasis-related Micro-RNAs. Cancer Genomics Proteomics 2019; 16:1-19. [PMID: 30587496 DOI: 10.21873/cgp.20108] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 11/08/2018] [Accepted: 11/23/2018] [Indexed: 02/06/2023] Open
Abstract
The mortality of patients with hormone-resistant prostate cancer can be ascribed to a large degree to metastasis to distant organs, predominantly to the bones. In this review, we discuss the contribution of micro-RNAs (miRs) to the metastatic process of prostate cancer. The criteria for selection of miRs for this review were the availability of preclinical in vivo metastasis-related data in conjunction with prognostic clinical data. Depending on their function in the metastatic process, the corresponding miRs are up- or down-regulated in prostate cancer tissues when compared to matching normal tissues. Up-regulated miRs preferentially target suppressors of cytokine signaling or tumor suppressor-related genes and metastasis-inhibitory transcription factors. Down-regulated miRs promote epithelial-mesenchymal transition or mesenchymal-epithelial transition and diverse pro-metastatic signaling pathways. Some of the discussed miRs exert their function by simultaneously targeting epigenetic pathways as well as cell-cycle-related, anti-apoptotic and signaling-promoting targets. Finally, we discuss potential therapeutic options for the treatment of prostate cancer-related metastases by substitution or inhibition of miRs.
Collapse
Affiliation(s)
- Ulrich H Weidle
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Alexandra Epp
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Fabian Birzele
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | - Ulrich Brinkmann
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| |
Collapse
|
10
|
Ehrlich L, Scrushy M, Meng F, Lairmore TC, Alpini G, Glaser S. Biliary epithelium: A neuroendocrine compartment in cholestatic liver disease. Clin Res Hepatol Gastroenterol 2018; 42:296-305. [PMID: 29678444 PMCID: PMC6129425 DOI: 10.1016/j.clinre.2018.03.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 03/04/2018] [Accepted: 03/13/2018] [Indexed: 02/07/2023]
Abstract
Hepatic fibrosis is characterized by abnormal accumulation of extracellular matrix (ECM) that can lead to ductopenia, cirrhosis, and even malignant transformation. In this review, we examine cholestatic liver diseases characterized by extensive biliary fibrosis such as primary sclerosing cholangitis (PSC), primary biliary cholangitis (PBC), polycystic liver disease (PLD), and MDR2-/- and BDL mouse models. Following biliary injury, cholangiocytes, the epithelial cells that line the bile ducts, become reactive and adopt a neuroendocrine phenotype in which they secrete and respond to neurohormones and neuropeptides in an autocrine and paracrine fashion. Emerging evidence indicates that cholangiocytes influence and respond to changes in the ECM and stromal cells in the microenvironment. For example, activated myofibroblasts and hepatic stellate cells are major drivers of collagen deposition and biliary fibrosis. Additionally, the liver is richly innervated with adrenergic, cholinergic, and peptidergic fibers that release neurohormones and peptides to maintain homeostasis and can be deranged in disease states. This review summarizes how cholangiocytes interact with their surrounding environment, with particular focus on how autonomic and sensory regulation affects fibrotic pathophysiology.
Collapse
Affiliation(s)
- Laurent Ehrlich
- Department of Medical Physiology, Texas A&M University, College of Medicine, Temple, TX 76504, United States
| | - Marinda Scrushy
- Department of Medical Physiology, Texas A&M University, College of Medicine, Temple, TX 76504, United States
| | - Fanyin Meng
- Research, Central Texas Veterans Health Care System, College of Medicine, Temple, TX 76504, United States; Baylor Scott & White Digestive Disease Research Center, Baylor Scott & White, Baylor Scott & White Health, Temple, TX 76504, United States
| | - Terry C Lairmore
- Department of Surgery, Baylor Scott & White Health and Texas A&M University, College of Medicine, Temple, TX 76504, United States
| | - Gianfranco Alpini
- Research, Central Texas Veterans Health Care System, College of Medicine, Temple, TX 76504, United States; Baylor Scott & White Digestive Disease Research Center, Baylor Scott & White, Baylor Scott & White Health, Temple, TX 76504, United States; Department of Medical Physiology, Texas A&M University, College of Medicine, Temple, TX 76504, United States
| | - Shannon Glaser
- Research, Central Texas Veterans Health Care System, College of Medicine, Temple, TX 76504, United States; Baylor Scott & White Digestive Disease Research Center, Baylor Scott & White, Baylor Scott & White Health, Temple, TX 76504, United States; Department of Medical Physiology, Texas A&M University, College of Medicine, Temple, TX 76504, United States.
| |
Collapse
|
11
|
Huang D, Yuan W, Li H, Li S, Chen Z, Yang H. Identification of key pathways and biomarkers in sorafenib-resistant hepatocellular carcinoma using bioinformatics analysis. Exp Ther Med 2018; 16:1850-1858. [PMID: 30186410 PMCID: PMC6122189 DOI: 10.3892/etm.2018.6427] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 04/26/2018] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignant types of cancer, with a high mortality rate. Sorafenib is the sole approved oral clinical therapy against advanced HCC. However, individual patients exhibit varying responses to sorafenib and the development of sorafenib resistance has been a new challenge for its clinical efficacy. The current study identified gene biomarkers and key pathways in sorafenib-resistant HCC using bioinformatics analysis. Gene dataset GSE73571 was obtained from the Gene Expression Omnibus (GEO) database, including four sorafenib-acquired resistant and three sorafenib-sensitive HCC phenotypes. Differentially expressed genes (DEGs) were identified using the web tool GEO2R. Functional and pathway enrichment of DEGs were analyzed using the Database for Annotation, Visualization and Integrated Discovery and the protein-protein interaction (PPI) network was constructed using the Search Tool for the Retrieval of Interacting Genes/Proteins and Cytoscape. A total of 1,319 DEGs were selected, which included 593 upregulated and 726 downregulated genes. Functional and pathway enrichment analysis revealed DEGs enriched in negative regulation of endopeptidase activity, cholesterol homeostasis, DNA replication and repair, coagulation cascades, insulin resistance, RNA transport, cell cycle and others. Eight hub genes, including kininogen 1, vascular cell adhesion molecule 1, apolipoprotein C3, alpha 2-HS glycoprotein, erb-b2 receptor tyrosine kinase 2, secreted protein acidic and cysteine rich, vitronectin and vimentin were identified from the PPI network. In conclusion, the present study identified DEGs and key genes in sorafenib-resistant HCC, which further the knowledge of potential mechanisms in the development of sorafenib resistance and may provide potential targets for early diagnosis and new treatments for sorafenib-resistant HCC.
Collapse
Affiliation(s)
- Danping Huang
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510000, P.R. China
| | - Weiqu Yuan
- Acupuncture Department, The Fourth Clinical Medical College of Guangzhou University Chinese Medicine, Shenzhen, Guangdong 518000, P.R. China
| | - Hanmin Li
- Hepatopathy Institution, Affiliated Hospital Hubei University Chinese Medicine, Wuhan, Hubei 430061, P.R. China
| | - Shaodong Li
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510000, P.R. China
| | - Zuanguang Chen
- Pharmaceutical Analysis Department, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, P.R. China
| | - Hongzhi Yang
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510000, P.R. China
| |
Collapse
|
12
|
Yan C, Yang Q, Gong Z. Activation of Hepatic Stellate Cells During Liver Carcinogenesis Requires Fibrinogen/Integrin αvβ5 in Zebrafish. Neoplasia 2018; 20:533-542. [PMID: 29649779 PMCID: PMC5915969 DOI: 10.1016/j.neo.2018.02.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 02/07/2018] [Accepted: 02/14/2018] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common cancers and it usually develops from a background of liver fibrosis or inflammation. The crosstalk between tumor cells and stromal cells plays an important and stimulating role during tumor progression. Previously we found in a krasV12-induced zebrafish HCC model that oncogenic hepatocytes activate hepatic stellate cells (HSCs) by up-regulation of serotonin and activate neutrophils and macrophages by up-regulation of cortisol. In the present study, we found a novel signaling transduction mechanism between oncogenic hepatocytes and HSCs. After krasV12 induction, fibrinogen was up-regulated in oncogenic hepatocytes. We reasoned that fibrinogen may bind to integrin αvβ5 on HSCs to activate HSCs. Consistent with this notion, pharmaceutical treatment using an antagonist of integrin αvβ5, cilengitide, significantly blocked HSC activation and function, accompanied by attenuated proliferation of oncogenic hepatocytes and progression of liver fibrosis. On the contrary, adenosine 5'-diphosphate, an agonist of αvβ5, activated HSCs significantly that further stimulated the tumor progression and liver fibrosis. Interestingly, in human liver disease samples, we detected an increased level of fibrinogen during tumor progression which indicated the potential role of fibrinogen signaling in HCC progression. Thus, we concluded a novel interaction between oncogenic hepatocytes and HSCs through the fibrinogen related pathway in both the zebrafish HCC model and human liver disease samples.
Collapse
Key Words
- a-sma, alpha-smooth muscle actin
- dox, doxycycline
- dpf, day post fertilization
- dpi, day post induction
- facs, fluorescence-activated cell sorting
- gfap, glial fibrillary acidic protein
- h&e, hematoxylin and eosin
- hcc, hepatocellular carcinoma
- hsc, hepatic stellate cell
- if, immunofluorescence
- ihc, immunohistochemistry
- oh, oncogenic hepatocyte
- tme, tumor microenvironment
- wt, wild type
Collapse
Affiliation(s)
- Chuan Yan
- Department of Biological Sciences, National University of Singapore, Singapore; National University of Singapore Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore
| | - Qiqi Yang
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Zhiyuan Gong
- Department of Biological Sciences, National University of Singapore, Singapore; National University of Singapore Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore.
| |
Collapse
|
13
|
Barbhuiya MA, Mirando AC, Simons BW, Lemtiri-Chlieh G, Green JJ, Popel AS, Pandey NB, Tran PT. Therapeutic potential of an anti-angiogenic multimodal biomimetic peptide in hepatocellular carcinoma. Oncotarget 2017; 8:101520-101534. [PMID: 29254183 PMCID: PMC5731893 DOI: 10.18632/oncotarget.21148] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 08/26/2017] [Indexed: 12/13/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a major cause of cancer-related death worldwide. Due to inadequate screening methods and the common coexistence of limited functional liver reserves, curative treatment options are limited. Liver transplantation is the only curative treatment modality for early HCC. There are multidisciplinary treatment options like ablative treatments, radiation and systemic therapy available for more advanced patients or those that are inoperable. Treatment resistance and progression is inevitable for these HCC patients. Newer therapeutics need to be explored for better management of HCC. HCC is a hypervascular tumor and many pro-angiogenic proteins are found significantly overexpressed in HCC. Here we explored the therapeutic potential of the anti-angiogenic, anti-lymphangiogenic, and directly anti-tumorigenic biomimetic collagen IV-derived peptide developed by our group. Human HCC cell lines HuH7, Hep3b and HepG2 showed significant disruption of cell adhesion and migration upon treatment with the peptide. Consistent with previously described multimodal inhibitory properties, the peptide was found to inhibit both c-Met and IGF1R signaling in HepG2 cells and blocked HepG2 conditioned media stimulation of microvascular endothelial cell (MEC) tube formation. Furthermore, the peptide treatment of mouse HepG2 tumor xenografts significantly inhibited growth relative to untreated controls. The peptide was also found to improve the survival of autochthonous Myc-induced HCC in a transgenic mouse model. Mechanistically, we found that the peptide treatment reduced microvascular density in the autochthonous liver tumors with increased apoptosis. This study shows the promising therapeutic potential of our biomimetic peptide in the treatment of HCC.
Collapse
Affiliation(s)
- Mustafa A Barbhuiya
- Department of Radiation Oncology and Molecular and Radiation Sciences, Sidney Kimmel Comprehensive Cancer Centre, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Adam C Mirando
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Brian W Simons
- Department of Medical Oncology, Sidney Kimmel Comprehensive Cancer Centre and Department of Urology, The Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ghali Lemtiri-Chlieh
- Department of Radiation Oncology and Molecular and Radiation Sciences, Sidney Kimmel Comprehensive Cancer Centre, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jordan J Green
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Niranjan B Pandey
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Phuoc T Tran
- Department of Radiation Oncology and Molecular and Radiation Sciences, Sidney Kimmel Comprehensive Cancer Centre, Johns Hopkins School of Medicine, Baltimore, MD, USA.,Department of Medical Oncology, Sidney Kimmel Comprehensive Cancer Centre and Department of Urology, The Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
14
|
Rojas A, Zhang P, Wang Y, Foo WC, Muñoz NM, Xiao L, Wang J, Gores GJ, Hung MC, Blechacz B. A Positive TGF-β/c-KIT Feedback Loop Drives Tumor Progression in Advanced Primary Liver Cancer. Neoplasia 2017; 18:371-86. [PMID: 27292026 PMCID: PMC4909706 DOI: 10.1016/j.neo.2016.04.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 04/07/2016] [Indexed: 12/13/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is globally the second most common cause of cancer mortality. The majority of HCC patients are diagnosed at advanced stage disease for which no curative treatments exist. TGF-β has been identified as a potential therapeutic target. However, the molecular mechanisms mediating its functional switch from a tumor suppressor to tumor promoter in HCC and its interactions with other signaling pathways are poorly understood. Here, we demonstrate an aberrant molecular network between the TGF-β and c-KIT pathway that mediates the functional switch of TGF-β to a driver of tumor progression in HCC. TGF-β/SMAD2 signaling transcriptionally regulates expression of the c-KIT receptor ligand (stem cell factor [SCF]) with subsequent auto- and paracrine activation of c-KIT/JAK1/STAT3 signaling. SCF induces TGF-β1 ligand expression via STAT3, thereby forming a positive feedback loop between TGF-β/SMAD and SCF/c-KIT signaling. This network neutralizes TGF-β–mediated cell cycle inhibition and induces tumor cell proliferation, epithelial-to-mesenchymal-transition, migration, and invasion. Disruption of this feedback loop inhibits TGF-β tumor-promoting effects and restores its antiproliferative functions. Consistent with our in vitro data, we demonstrate SCF overexpression and its correlation to SMAD2 and STAT3 activation in human HCC tumors, advanced tumor-node-metastasis stages, and shorter survival. CONCLUSIONS: Canonical TGF-β and c-KIT signaling forms a positive, tumor-promoting feedback loop. Disruption of this loop restores TGF-β tumor suppressor function and provides the rationale for targeting the TGF-β/SCF axis as a novel therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Andres Rojas
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Pingyu Zhang
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ying Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Wai Chin Foo
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Nina M Muñoz
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Lianchun Xiao
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Gregory J Gores
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Mien-Chie Hung
- Department of Molecular Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX; Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX; Graduate Institute of Cancer Biology and Center for Molecular Medicine, China Medical University, Taichung, Taiwan
| | - Boris Blechacz
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
15
|
Naboulsi W, Bracht T, Megger DA, Reis H, Ahrens M, Turewicz M, Eisenacher M, Tautges S, Canbay AE, Meyer HE, Weber F, Baba HA, Sitek B. Quantitative proteome analysis reveals the correlation between endocytosis-associated proteins and hepatocellular carcinoma dedifferentiation. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2016; 1864:1579-85. [PMID: 27519163 DOI: 10.1016/j.bbapap.2016.08.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 07/22/2016] [Accepted: 08/08/2016] [Indexed: 01/04/2023]
Abstract
The majority of poorly differentiated hepatocellular carcinomas (HCCs) develop from well-differentiated tumors. Endocytosis is a cellular function which is likely to take part in this development due to its important role in regulating the abundances of vital signaling receptors. Here, we aimed to investigate the abundance of endocytosis-associated proteins in HCCs with various differentiation grades. Therefore, we analyzed 36 tissue specimens from HCC patients via LC-MS/MS-based label-free quantitative proteomics including 19 HCC tissue samples with different degrees of histological grades and corresponding non-tumorous tissue controls. As a result, 277 proteins were differentially regulated between well-differentiated tumors and controls. In moderately and poorly differentiated tumors, 278 and 1181 proteins, respectively, were significantly differentially regulated compared to non-tumorous tissue. We explored the regulated proteins based on their functions and identified thirty endocytosis-associated proteins, mostly overexpressed in poorly differentiated tumors. These included proteins that have been shown to be up-regulated in HCC like clathrin heavy chain-1 (CLTC) as well as unknown proteins, such as secretory carrier-associated membrane protein 3 (SCAMP3). The abundances of SCAMP3 and CLTC were immunohistochemically examined in tissue sections of 84 HCC patients. We demonstrate the novel association of several endocytosis-associated proteins, in particular, SCAMP3 with HCC progression.
Collapse
Affiliation(s)
- Wael Naboulsi
- Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801 Bochum, Germany.
| | - Thilo Bracht
- Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801 Bochum, Germany
| | - Dominik A Megger
- Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801 Bochum, Germany
| | - Henning Reis
- Institute of Pathology, University of Duisburg-Essen, University Hospital Essen, 45147 Essen, Germany
| | - Maike Ahrens
- Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801 Bochum, Germany
| | - Michael Turewicz
- Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801 Bochum, Germany
| | - Martin Eisenacher
- Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801 Bochum, Germany
| | - Stephanie Tautges
- Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801 Bochum, Germany
| | - Ali E Canbay
- Department of Gastroenterology and Hepatology, University of Duisburg-Essen, University Hospital Essen, 45147 Essen, Germany
| | - Helmut E Meyer
- Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801 Bochum, Germany
| | - Frank Weber
- Department of General, Visceral and Transplantation Surgery, University of Duisburg-Essen, University Hospital Essen, 45147 Essen, Germany
| | - Hideo A Baba
- Institute of Pathology, University of Duisburg-Essen, University Hospital Essen, 45147 Essen, Germany
| | - Barbara Sitek
- Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801 Bochum, Germany.
| |
Collapse
|
16
|
Schmithals C, Köberle V, Korkusuz H, Pleli T, Kakoschky B, Augusto EA, Ibrahim AA, Arencibia JM, Vafaizadeh V, Groner B, Korf HW, Kronenberger B, Zeuzem S, Vogl TJ, Waidmann O, Piiper A. Improving Drug Penetrability with iRGD Leverages the Therapeutic Response to Sorafenib and Doxorubicin in Hepatocellular Carcinoma. Cancer Res 2015; 75:3147-54. [PMID: 26239478 DOI: 10.1158/0008-5472.can-15-0395] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
iRGD is a derivative of the integrin-binding peptide RGD, which selectively increases the penetrability of tumor tissue to various coadministered substances in several preclinical models. In this study, we investigated the ability of iRGD to improve the delivery of sorafenib and doxorubicin therapy in hepatocellular carcinoma (HCC) using established mouse models of the disease. A contrast-enhanced MRI method was developed in parallel to assess the in vivo effects of iRGD in this setting. We found that iRGD improved the delivery of marker substances to the tumors of HCC-bearing mice about three-fold without a parallel increase in normal tissues. Control peptides lacking the critical CendR motif had no effect. Similarly, iRGD also selectively increased the signal intensity from tumors in Gd-DTPA-enhanced MRI. In terms of antitumor efficacy, iRGD coadministration significantly augmented the individual inhibitory effects of sorafenib and doxorubicin without increasing systemic toxicity. Overall, our results offered a preclinical proof of concept for the use of iRGD coadministration as a strategy to widen the therapeutic window for HCC chemotherapy, as monitored by Gd-DTPA-enhanced MRI as a noninvasive, clinically applicable method to identify iRGD-reactive tumors.
Collapse
Affiliation(s)
| | - Verena Köberle
- Department of Medicine 1, University Hospital Frankfurt, Frankfurt, Germany
| | - Hüdayi Korkusuz
- Department of Nuclear Medicine, University Hospital Frankfurt, Frankfurt, Germany
| | - Thomas Pleli
- Department of Medicine 1, University Hospital Frankfurt, Frankfurt, Germany
| | - Bianca Kakoschky
- Department of Medicine 1, University Hospital Frankfurt, Frankfurt, Germany
| | | | - Ahmed Atef Ibrahim
- Department of Medicine 1, University Hospital Frankfurt, Frankfurt, Germany. The Immunology and Infectious Diseases Laboratory, Therapeutic Chemistry Department, The National Research Center, Dokki, Cairo, Egypt
| | - Jose M Arencibia
- Department of Medicine 1, University Hospital Frankfurt, Frankfurt, Germany
| | | | | | - Horst-Werner Korf
- Institute of Anatomy 2, University Hospital Frankfurt, Frankfurt, Germany
| | - Bernd Kronenberger
- Department of Medicine 1, University Hospital Frankfurt, Frankfurt, Germany
| | - Stefan Zeuzem
- Department of Medicine 1, University Hospital Frankfurt, Frankfurt, Germany
| | - Thomas J Vogl
- Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Frankfurt, Germany
| | - Oliver Waidmann
- Department of Medicine 1, University Hospital Frankfurt, Frankfurt, Germany
| | - Albrecht Piiper
- Department of Medicine 1, University Hospital Frankfurt, Frankfurt, Germany.
| |
Collapse
|
17
|
Huang YL, Chu YL, Ho CT, Chung JG, Lai CI, Su YC, Kuo YH, Sheen LY. Antcin K, an Active Triterpenoid from the Fruiting Bodies of Basswood-Cultivated Antrodia cinnamomea, Inhibits Metastasis via Suppression of Integrin-Mediated Adhesion, Migration, and Invasion in Human Hepatoma Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:4561-4569. [PMID: 25911944 DOI: 10.1021/jf5059304] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Previous research demonstrated that the ethyl acetate extract from Antrodia cinnamomea suppresses the invasive potential of human breast and hepatoma cells, but the effective compounds are not identified. The main bioactive compounds of A. cinnamomea are ergostane-type triterpenoids, and the content of antcin K is the highest. The objective of this study was to evaluate the antimetastatic activity and mechanisms of antcin K purified from the fruiting body of basswood-cultivated A. cinnamomea on human liver cancer Hep 3B cells. The results showed that adhesion, migration, and invasion of Hep 3B cells were effectively inhibited by antcin K within 24 h of treatment. Antcin K not only reduced the protein expression and activity of MMP-2 and MMP-9 but also down-regulated vimentin and up-regulated E-cadherin in Hep 3B cells. In depth investigation for the molecular mechanism revealed that antcin K could reduce the protein expression of integrin β1, β3, α5, and αv and suppress phosphorylation of FAK, Src, PI3K, AKT, MEK, ERK, and JNK. These results suggested that antcin K was able to inhibit the metastasis of human hepatoma cells through suppression of integrin-mediated adhesion, migration, and invasion. Coupled with these findings, antcin K has a good potential to reduce the risk of liver cancer metastasis.
Collapse
Affiliation(s)
| | | | - Chi-Tang Ho
- ΔDepartment of Food Science, Rutgers University, New Brunswick, New Jersey, United States
| | - Jing-Gung Chung
- §Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | | | | | - Yueh-Hsiung Kuo
- ΠDepartment of Chinese Pharmaceutical Sciences and Chinese Medicine Resourceσ, China Medical University, Taichung, Taiwan
| | | |
Collapse
|
18
|
Liang J, Wu W, Lai D, Li J, Fang C. Enhanced solubility and targeted delivery of curcumin by lipopeptide micelles. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2015; 26:369-83. [PMID: 25621942 DOI: 10.1080/09205063.2015.1012034] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
A lipopeptide (LP)-containing KKGRGDS as the hydrophilic heads and lauric acid (C12) as the hydrophobic tails has been designed and prepared by standard solid-phase peptide synthesis technique. LP can self-assemble into spherical micelles with the size of ~30 nm in PBS (phosphate buffer saline) (pH 7.4). Curcumin-loaded LP micelles were prepared in order to increase the water solubility, sustain the releasing rate, and improve the tumor targeted delivery of curcumin. Water solubility, cytotoxicity, in vitro release behavior, and intracellular uptake of curcumin-loaded LP micelles were investigated. The results showed that LP micelles can increase the water solubility of curcumin 1.1 × 10(3) times and sustain the release of curcumin in a low rate. Curcumin-loaded LP micelles showed much higher cell inhibition than free curcumin on human cervix carcinoma (HeLa) and HepG2 cells. When incubating these curcumin-loaded micelles with HeLa and COS7 cells, due to the over-expression of integrins on cancer cells, the micelles can efficiently use the tumor-targeting function of RGD (functionalized peptide sequences: Arg-Gly-Asp) sequence to deliver the drug into HeLa cells, and better efficiency of the self-assembled LP micelles for curcumin delivery than crude curcumin was also confirmed by LCSM (laser confocal scanning microscope) assays. Combined with the enhanced solubility and higher cell inhibition, LP micelles reported in this study may be promising in clinical application for targeted curcumin delivery.
Collapse
Affiliation(s)
- Ju Liang
- a Chemical Engineering and Pharmaceutics School , Henan University of Science and Technology , Luoyang 471023 , PR China
| | | | | | | | | |
Collapse
|
19
|
Rajput S, Kumar BNP, Banik P, Parida S, Mandal M. Thymoquinone restores radiation-induced TGF-β expression and abrogates EMT in chemoradiotherapy of breast cancer cells. J Cell Physiol 2015; 230:620-9. [PMID: 25164250 DOI: 10.1002/jcp.24780] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 08/15/2014] [Indexed: 12/16/2022]
Abstract
Radiotherapy remains a prime approach to adjuvant therapies in patients with early and advanced breast cancer. In spite of therapeutic success, metastatic progression in patients undergoing therapy, limits its application. However, effective therapeutic strategies to understand the cellular and molecular machinery in inhibiting radiation-induced metastatic progression, which is poorly understood so far, need to be strengthened. Ionizing radiation was known to prompt cancer cell's metastatic ability by eliciting Transforming Growth Factor-beta (TGF-β), a key regulator in epithelial-mesenchymal transdifferentiation and radio-resistance. In this viewpoint, we employed thymoquinone as a radiosensitizer to investigate its migration and invasion reversal abilities in irradiated breast cancer cell lines by assessing their respective attributes. The role of metastasis regulatory molecules like TGF-β, E-cadherin, and integrin αV and its downstream molecules were determined using RT-PCR, western blotting, immunofluorescence, and extracellular TGF-β levels affirmed through ELISA assays. These studies affirmed the TGF-β restoring ability of thymoquinone in radiation-driven migration and invasion. Also, results demonstrated that the epithelial markers E-cadherin and cytokeratin 19 were downregulated whereas mesenchymal markers like integrin αV, MMP9, and MMP2 were upregulated by irradiation treatment; however thymoquinone pre-sensitization has reverted the expression of these proteins back to control proteins expression. Here, paclitaxel was chosen as an apoptosis inducer in TGF-β restored cells and confirmed its cytotoxic effects in radiation alone and thymoquinone sensitized irradiated cells. We conclude that this therapeutic modality is effective in preventing radiation-induced epithelial-mesenchymal transdifferentiation and concomitant induction of apoptosis in breast cancer.
Collapse
Affiliation(s)
- Shashi Rajput
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, India
| | | | | | | | | |
Collapse
|
20
|
Soejima Y, Inoue M, Takahashi Y, Uozaki H, Sawabe M, Fukusato T. Integrins αvβ6, α6β4 and α3β1 are down-regulated in cholangiolocellular carcinoma but not cholangiocarcinoma. Hepatol Res 2014; 44:E320-34. [PMID: 24552196 DOI: 10.1111/hepr.12312] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2013] [Revised: 02/02/2014] [Accepted: 02/12/2014] [Indexed: 12/16/2022]
Abstract
AIM The aim of this study was to evaluate integrin expression and immunolocalization of the extracellular matrix proteins in cholangiolocellular carcinoma (CoCC). METHODS Tissue specimens of 23 CoCC, 28 cholangiocarcinomas (CCC), 42 hepatocellular carcinomas (HCC) and 11 classical type combined hepatocellular cholangiocarcinomas (CHC) were immunostained for β6, β4 and α3 integrins, fibronectin and laminin. ITGB6, B4 and A3 mRNA levels in six HCC cell lines, five CCC cell lines and two CHC cell lines were quantified by quantitative reverse transcription polymerase chain reaction. RESULTS Little or no positivity for β6, β4 and α3 integrins was shown in 91%, 91% and 52% of CoCC and 100%, 98% and 81% of HCC, respectively, according to immunostaining, whereas intense positive staining for these integrins was demonstrated in 64%, 96% and 75% of CCC, respectively. There was a close correlation between β4 and α3 integrin expression and intracytoplasmic laminin in CoCC, CCC and HCC, but not between β6 expression and its ligand, fibronectin. Integrin mRNA levels were increased in four of five CCC cell lines, but nearly undetectable in five of six HCC cell lines and one CHC cell line. Tubular differentiation of a CHC cell line cultured in collagen gel matrix induced upregulation of these integrins. CONCLUSION Our results first indicated downregulation of αvβ6, α6β4 and α3β1 integrins in CoCC, in contrast to its high expression in CCC, suggesting a diagnostic value of integrins in the differential diagnosis of CoCC and CCC, as well as a useful inducible marker of the intermediate features of CoCC.
Collapse
Affiliation(s)
- Yurie Soejima
- Department of Pathology, Teikyo University School of Medicine, Tokyo, Japan; Department of Molecular Pathology, Graduate School of Health Care Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
21
|
Feng MX, Ma MZ, Fu Y, Li J, Wang T, Xue F, Zhang JJ, Qin WX, Gu JR, Zhang ZG, Xia Q. Elevated autocrine EDIL3 protects hepatocellular carcinoma from anoikis through RGD-mediated integrin activation. Mol Cancer 2014; 13:226. [PMID: 25273699 PMCID: PMC4200221 DOI: 10.1186/1476-4598-13-226] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Accepted: 09/28/2014] [Indexed: 02/20/2023] Open
Abstract
BACKGROUND A remolded microenvironment in hepatocellular carcinoma (HCC) caused by abnormally expressed matricellular proteins could promote HCC progression. The cell-matrix interactions mediated by integrins play an important role in tumor microenvironment. Epidermal Growth Factor-like repeats and Discoidin I-Like Domains 3 (EDIL3), an extracellular matrix (ECM) protein with angiogenic and anti-inflammatory effects, is abnormally highly expressed in HCC. Here we aim to analyze its expression in liver and HCC tissues, investigate the underlined mechanisms accounted for HCC progression. METHODS EDIL3 expression level is examined in normal liver, cirrhotic liver and HCC at both mRNA and protein level. The association between EDIL3 and clinical outcomes is analyzed. The pattern of EDIL3 expression and location is examined using Immunofluorescence and ELISA. Overexpression or knock-down of EDIL3 in a panel of cell lines are subjected to assays related to proliferation, invasion, and anoikis to investigate the mechanisms of this matrix protein in HCC progression. Recombinant EDIL3 treatment is applied to confirm the results. RESULTS Compared with normal liver and cirrhotic liver, EDIL3 is elevated in HCC. High level of EDIL3 protein is much more commonly in patients with larger tumor or portal vein tumor thrombus (PVTT) formation, associated with poor prognosis. EDIL3 is abundantly expressed in HCC cells and secreted by cancer cells. In vitro and in vivo studies indicate that EDIL3, probably in an autocrine manner, inhibits anoikis and promotes anchorage-independent growth of HCC cells. Further mechanistic studies suggest integrin ligation by EDIL3 and thus that the sustained activation of the FAK-Src-AKT signal is responsible for the anoikis resistance and anchorage independence. Both the administration of cilengitide, a RGD-containing integrin antagonist, and silencing of integrin αV, an important RGD-binding integrin, results in the blockade of anoikis-resistance induced by EDIL3. CONCLUSION Our study suggests that high levels of autocrine EDIL3 may contribute to a receptive microenvironment for the survival of detached HCC cells and may involve in cancer cell spreading. We also highlight the importance of interaction between EDIL3 and integrin αV and suggest disrupting the ligation of EDIL3 to integrins via RGD-blocking in selected patients may bear potential therapeutic value.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Zhi-Gang Zhang
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, Shanghai 200127, China.
| | | |
Collapse
|
22
|
Battistini L, Burreddu P, Sartori A, Arosio D, Manzoni L, Paduano L, D’Errico G, Sala R, Reia L, Bonomini S, Rassu G, Zanardi F. Enhancement of the Uptake and Cytotoxic Activity of Doxorubicin in Cancer Cells by Novel cRGD-Semipeptide-Anchoring Liposomes. Mol Pharm 2014; 11:2280-93. [DOI: 10.1021/mp400718j] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Lucia Battistini
- Dipartimento
di Farmacia, Università degli Studi di Parma, Parma 43124, Italy
| | - Paola Burreddu
- Istituto
di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche, Li Punti Sassari 07100, Italy
| | - Andrea Sartori
- Dipartimento
di Farmacia, Università degli Studi di Parma, Parma 43124, Italy
| | - Daniela Arosio
- Istituto
di Scienze e Tecnologie Molecolari, Consiglio Nazionale delle Ricerche, Milano 20133, Italy
| | - Leonardo Manzoni
- Istituto
di Scienze e Tecnologie Molecolari, Consiglio Nazionale delle Ricerche, Milano 20133, Italy
| | - Luigi Paduano
- Dipartimento
di Scienze Chimiche, Università degli Studi di Napoli “Federico II”, Napoli 80126, Italy
- CSGI−Consorzio interuniversitario per lo sviluppo dei Sistemi a Grande Interfase, Sesto Fiorentino 50019, Italy
| | - Gerardino D’Errico
- Dipartimento
di Scienze Chimiche, Università degli Studi di Napoli “Federico II”, Napoli 80126, Italy
- CSGI−Consorzio interuniversitario per lo sviluppo dei Sistemi a Grande Interfase, Sesto Fiorentino 50019, Italy
| | - Roberto Sala
- Dipartimento
di Scienze Biomediche, Biotecnologiche e Traslazionali, Università degli Studi di Parma, Parma 43126, Italy
| | - Laura Reia
- Dipartimento
di Scienze Biomediche, Biotecnologiche e Traslazionali, Università degli Studi di Parma, Parma 43126, Italy
| | - Sabrina Bonomini
- Dipartimento
di Medicina Clinica e Sperimentale, Università degli Studi di Parma, Parma 43126, Italy
| | - Gloria Rassu
- Istituto
di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche, Li Punti Sassari 07100, Italy
| | - Franca Zanardi
- Dipartimento
di Farmacia, Università degli Studi di Parma, Parma 43124, Italy
| |
Collapse
|
23
|
Ha SY, Shin J, Kim JH, Kang MS, Yoo HY, Kim HH, Um SH, Kim SH. Overexpression of integrin αv correlates with poor prognosis in colorectal cancer. J Clin Pathol 2014; 67:576-81. [PMID: 24695839 DOI: 10.1136/jclinpath-2013-202163] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
AIMS Integrin αv subunits are involved in tumour angiogenesis and tumour progression in various types of cancers. Clinical trials evaluating agents targeting integrin αv are ongoing. Integrin αv expression has been reported in several cancers in association with tumour progression or poor survival. However, no study has addressed the prognostic influence of integrin αv expression on survival of patients with colorectal cancer (CRC). METHODS Immunohistochemical staining of integrin αv was performed in 198 CRC samples to evaluate its prognostic significance. RESULTS High expression of integrin αv was observed in 58.1% (115/189) of colorectal adenocarcinoma samples, while only in 11.5% (3/26) of tubular adenoma samples and in none of normal mucosa or hyperplastic polyp samples. It was more frequently found in female patients and less frequently observed in well differentiated tumours. The proportion of cases with high expression of integrin αv showed an increasing trend with increased T stage (p=0.032), N stage (p=0.006) and TNM stage (p=0.001). Patients displaying exuberant expression of integrin αv showed shorter overall survival (p=0.001) and disease-free survival (p=0.004). Elevated integrin αv expression was an independent prognostic factor for overall survival (HR: 2.04, 95% CI 1.16 to 3.56; p=0.013) and disease-free survival (HR: 2.19, 95% CI 1.16 to 4.13; p=0.015). CONCLUSIONS Overexpression of integrin αv is associated with advanced T and N stage and as an independent prognostic factor in CRC.
Collapse
Affiliation(s)
- Sang Yun Ha
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Juyoun Shin
- Division of Cancer Biology, Comparative Biomedicine Research Branch, Research Institute of National Cancer Center, Goyang, Korea
| | - Jeong Hoon Kim
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea Department of Health Sciences and Technology, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Myung Soo Kang
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea Department of Health Sciences and Technology, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hae-Yong Yoo
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea Department of Health Sciences and Technology, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hyeon-Ho Kim
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea Department of Health Sciences and Technology, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sung-Hee Um
- Department of Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Seok-Hyung Kim
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea Department of Health Sciences and Technology, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
24
|
Ferrín G, Ranchal I, Llamoza C, Rodríguez-Perálvarez ML, Romero-Ruiz A, Aguilar-Melero P, López-Cillero P, Briceño J, Muntané J, Montero-Álvarez JL, De la Mata M. Identification of candidate biomarkers for hepatocellular carcinoma in plasma of HCV-infected cirrhotic patients by 2-D DIGE. Liver Int 2014; 34:438-446. [PMID: 23944848 DOI: 10.1111/liv.12277] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Accepted: 06/23/2013] [Indexed: 12/22/2022]
Abstract
BACKGROUND The current methods available for screening and detecting hepatocellular carcinoma (HCC) have insufficient sensitivity and specificity, and only a low percentage of diagnosis of small tumours is based on these assays. Because HCC is usually asymptomatic at potentially curative stages, identification of biomarkers for the early detection of HCC is essential to improve patient survival. AIM The aim of this study was to identify candidate markers for HCC development in the plasma from hepatitis C virus (HCV)-infected cirrhotic patients. METHODS We compared protein expression profiles of plasma samples from HCV-infected cirrhotic patients with and without HCC, using two-dimensional fluorescence difference gel electrophoresis (2-D DIGE) coupled with MALDI-TOF/TOF mass spectrometry. The 2-D DIGE results were analysed statistically using Decyder™ software, and verified by western blot and enzyme-linked immunosorbent assay (ELISA). RESULTS In the plasma of HCV-infected HCC patients, we observed decreased expression of complement component 9, ficolin-3 (FCN3), serum amyloid P component (SAP), fibrinogen-gamma and immunoglobulin gamma-1 chain, and increased expression of vitronectin (VTN) and galectin-3 binding protein (G3BP) by DIGE analysis. ELISA confirmed DIGE results for VTN and G3BP but not for SAP or FCN3 in a larger patient population. CONCLUSIONS The proteins VTN and SAP are candidate biomarkers for HCC development in HCV-infected cirrhotic patients.
Collapse
Affiliation(s)
- Gustavo Ferrín
- Maimónides Institute for Biomedical Research in Córdoba (IMBIC), Reina Sofía University Hospital, University of Córdoba, Córdoba, Spain; Networked Biomedical Research Center, Hepatic and Digestive Diseases (CIBERehd), Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
β3 integrin promotes TGF-β1/H2O2/HOCl-mediated induction of metastatic phenotype of hepatocellular carcinoma cells by enhancing TGF-β1 signaling. PLoS One 2013; 8:e79857. [PMID: 24260309 PMCID: PMC3832483 DOI: 10.1371/journal.pone.0079857] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Accepted: 09/25/2013] [Indexed: 02/06/2023] Open
Abstract
In addition to being an important mediator of migration and invasion of tumor cells, β3 integrin can also enhance TGF-β1 signaling. However, it is not known whether β3 might influence the induction of metastatic phenotype of tumor cells, especially non-metastatic tumor cells which express low level of β3. Here we report that H2O2 and HOCl, the reactive oxygen species produced by neutrophils, could cooperate with TGF-β1 to induce metastatic phenotype of non-metastatic hepatocellular carcinoma (HCC) cells. TGF-β1/H2O2/HOCl, but not TGF-β1 or H2O2/HOCl, induced β3 expression by triggering the enhanced activation of p38 MAPK. Intriguingly, β3 in turn promoted TGF-β1/H2O2/HOCl-mediated induction of metastatic phenotype of HCC cells by enhancing TGF-β1 signaling. β3 promoted TGF-β1/H2O2/HOCl-induced expression of itself via positive feed-back effect on p38 MAPK activation, and also promoted TGF-β1/H2O2/HOCl-induced expression of α3 and SNAI2 by enhancing the activation of ERK pathway, thus resulting in higher invasive capacity of HCC cells. By enhancing MAPK activation, β3 enabled TGF-β1 to augment the promoting effect of H2O2/HOCl on anoikis-resistance of HCC cells. TGF-β1/H2O2/HOCl-induced metastatic phenotype was sufficient for HCC cells to extravasate from circulation and form metastatic foci in an experimental metastasis model in nude mice. Inhibiting the function of β3 could suppress or abrogate the promoting effects of TGF-β1/H2O2/HOCl on invasive capacity, anoikis-resistance, and extravasation of HCC cells. These results suggest that β3 could function as a modulator to promote TGF-β1/H2O2/HOCl-mediated induction of metastatic phenotype of non-metastatic tumor cells, and that targeting β3 might be a potential approach in preventing the induction of metastatic phenotype of non-metastatic tumor cells.
Collapse
|
26
|
Cancer development, progression, and therapy: an epigenetic overview. Int J Mol Sci 2013; 14:21087-113. [PMID: 24152442 PMCID: PMC3821660 DOI: 10.3390/ijms141021087] [Citation(s) in RCA: 213] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 09/27/2013] [Accepted: 10/04/2013] [Indexed: 11/17/2022] Open
Abstract
Carcinogenesis involves uncontrolled cell growth, which follows the activation of oncogenes and/or the deactivation of tumor suppression genes. Metastasis requires down-regulation of cell adhesion receptors necessary for tissue-specific, cell-cell attachment, as well as up-regulation of receptors that enhance cell motility. Epigenetic changes, including histone modifications, DNA methylation, and DNA hydroxymethylation, can modify these characteristics. Targets for these epigenetic changes include signaling pathways that regulate apoptosis and autophagy, as well as microRNA. We propose that predisposed normal cells convert to cancer progenitor cells that, after growing, undergo an epithelial-mesenchymal transition. This process, which is partially under epigenetic control, can create a metastatic form of both progenitor and full-fledged cancer cells, after which metastasis to a distant location may occur. Identification of epigenetic regulatory mechanisms has provided potential therapeutic avenues. In particular, epigenetic drugs appear to potentiate the action of traditional therapeutics, often by demethylating and re-expressing tumor suppressor genes to inhibit tumorigenesis. Epigenetic drugs may inhibit both the formation and growth of cancer progenitor cells, thus reducing the recurrence of cancer. Adopting epigenetic alteration as a new hallmark of cancer is a logical and necessary step that will further encourage the development of novel epigenetic biomarkers and therapeutics.
Collapse
|
27
|
Tilghman SL, Townley I, Zhong Q, Carriere PP, Zou J, Llopis SD, Preyan LC, Williams CC, Skripnikova E, Bratton MR, Zhang Q, Wang G. Proteomic signatures of acquired letrozole resistance in breast cancer: suppressed estrogen signaling and increased cell motility and invasiveness. Mol Cell Proteomics 2013; 12:2440-55. [PMID: 23704778 DOI: 10.1074/mcp.m112.023861] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Aromatase inhibitors, such as letrozole, have become the first-line treatment for postmenopausal women with estrogen-dependent breast cancer. However, acquired resistance remains a major clinical obstacle. Previous studies demonstrated constitutive activation of the MAPK signaling, overexpression of HER2, and down-regulation of aromatase and ERα in letrozole-resistant breast cancer cells. Given the complex signaling network involved in letrozole-refractory breast cancer and the lack of effective treatment for hormone resistance, further investigation of aromatase inhibitor resistance by a novel systems biology approach may reveal previously unconsidered molecular changes that could be utilized as therapeutic targets. This study was undertaken to characterize for the first time global proteomic alterations occurring in a letrozole-resistant cell line. A quantitative proteomic analysis of the whole cell lysates of LTLT-Ca (resistant) versus AC-1 cells (sensitive) was performed to identify significant protein expression changes. A total of 1743 proteins were identified and quantified, of which 411 were significantly up-regulated and 452 significantly down-regulated (p < 0.05, fold change > 1.20). Bioinformatics analysis revealed that acquired letrozole resistance is associated with a hormone-independent, more aggressive phenotype. LTLT-Ca cells exhibited 84% and 138% increase in migration and invasion compared with the control cells. The ROCK inhibitor partially abrogated the enhanced migration and invasion of the letrozole-resistant cells. Flow cytometric analyses also demonstrated an increase in vimentin and twist expression in letrozole-resistance cells, suggesting an onset of epithelial to mesenchymal transition (EMT). Moreover, targeted gene expression arrays confirmed a 28-fold and sixfold up-regulation of EGFR and HER2, respectively, whereas ERα and pS2 were dramatically reduced by 28-fold and 1100-fold, respectively. Taken together, our study revealed global proteomic signatures of a letrozole-resistant cell line associated with hormone independence, enhanced cell motility, EMT and the potential values of several altered proteins as novel prognostic markers or therapeutic targets for letrozole resistant breast cancer.
Collapse
Affiliation(s)
- Syreeta L Tilghman
- Department of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, New Orleans, LA 70125, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Espelt MV, Croci DO, Bacigalupo ML, Carabias P, Manzi M, Elola MT, Muñoz MC, Dominici FP, Wolfenstein-Todel C, Rabinovich GA, Troncoso MF. Novel roles of galectin-1 in hepatocellular carcinoma cell adhesion, polarization, and in vivo tumor growth. Hepatology 2011; 53:2097-2106. [PMID: 21391228 DOI: 10.1002/hep.24294] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
UNLABELLED Galectin-1 (Gal-1), a widely expressed β-galactoside-binding protein, exerts pleiotropic biological functions. Gal-1 is up-regulated in hepatocarcinoma cells, although its role in liver pathophysiology remains uncertain. We investigated the effects of Gal-1 on HepG2 hepatocellular carcinoma (HCC) cell adhesion and polarization. Soluble and immobilized recombinant Gal-1 (rGal-1) promoted HepG2 cell adhesion to uncoated plates and also increased adhesion to laminin. Antibody-mediated blockade experiments revealed the involvement of different integrins as critical mediators of these biological effects. In addition, exposure to rGal-1 markedly accelerated the development of apical bile canaliculi as shown by TRITC-phalloidin labeling and immunostaining for multidrug resistance associated-protein 2 (MRP2). Notably, rGal-1 did not interfere with multidrug resistance protein 1/P-glycoprotein or MRP2 apical localization, neither with transfer nor secretion of 5-chloromethylfluorescein diacetate through MRP2. Stimulation of cell adhesion and polarization by rGal-1 was abrogated in the presence of thiodigalactoside, a galectin-specific sugar, suggesting the involvement of protein-carbohydrate interactions in these effects. Additionally, Gal-1 effects were abrogated in the presence of wortmmanin, PD98059 or H89, suggesting involvement of phosphoinositide 3-kinase (PI3K), mitogen-activated protein kinase and cyclic adenosine monophosphate-dependent protein kinase signaling pathways in these functions. Finally, expression levels of this endogenous lectin correlated with HCC cell adhesion and polarization and up-regulation of Gal-1-favored growth of hepatocarcinoma in vivo. CONCLUSION Our results provide the first evidence of a role of Gal-1 in modulating HCC cell adhesion, polarization, and in vivo tumor growth, with critical implications in liver pathophysiology.
Collapse
Affiliation(s)
- María V Espelt
- Instituto de Química y Fisicoquímica Biológicas, Consejo Nacional de Investigaciones Científicas y Técnicas, Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Argentina
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Lee HJ, Na K, Choi EY, Kim KS, Kim H, Paik YK. Simple Method for Quantitative Analysis of N-Linked Glycoproteins in Hepatocellular Carcinoma Specimens. J Proteome Res 2009; 9:308-18. [DOI: 10.1021/pr900649b] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Hyoung-Joo Lee
- Department of Biochemistry and Biomedical Sciences, College of Life Sciences and Biotechnology, World Class University Program, Yonsei Proteome Research Center and Biomedical Proteome Research Center, Department of Surgery, and Department of Pathology, College of Medicine, Yonsei University, Seoul 120-749, Korea
| | - Keun Na
- Department of Biochemistry and Biomedical Sciences, College of Life Sciences and Biotechnology, World Class University Program, Yonsei Proteome Research Center and Biomedical Proteome Research Center, Department of Surgery, and Department of Pathology, College of Medicine, Yonsei University, Seoul 120-749, Korea
| | - Eun-Young Choi
- Department of Biochemistry and Biomedical Sciences, College of Life Sciences and Biotechnology, World Class University Program, Yonsei Proteome Research Center and Biomedical Proteome Research Center, Department of Surgery, and Department of Pathology, College of Medicine, Yonsei University, Seoul 120-749, Korea
| | - Kyung Sik Kim
- Department of Biochemistry and Biomedical Sciences, College of Life Sciences and Biotechnology, World Class University Program, Yonsei Proteome Research Center and Biomedical Proteome Research Center, Department of Surgery, and Department of Pathology, College of Medicine, Yonsei University, Seoul 120-749, Korea
| | - Hoguen Kim
- Department of Biochemistry and Biomedical Sciences, College of Life Sciences and Biotechnology, World Class University Program, Yonsei Proteome Research Center and Biomedical Proteome Research Center, Department of Surgery, and Department of Pathology, College of Medicine, Yonsei University, Seoul 120-749, Korea
| | - Young-Ki Paik
- Department of Biochemistry and Biomedical Sciences, College of Life Sciences and Biotechnology, World Class University Program, Yonsei Proteome Research Center and Biomedical Proteome Research Center, Department of Surgery, and Department of Pathology, College of Medicine, Yonsei University, Seoul 120-749, Korea
| |
Collapse
|
30
|
Yam JWP, Tse EYT, Ng IOL. Role and significance of focal adhesion proteins in hepatocellular carcinoma. J Gastroenterol Hepatol 2009; 24:520-30. [PMID: 19368632 DOI: 10.1111/j.1440-1746.2009.05813.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Focal adhesions are structural links between the extracellular matrix and actin cytoskeleton. They are important sites where dynamic alterations of proteins in the focal contacts are involved during cell movement. Focal adhesions are composed of diverse molecules, for instance, receptors, structural proteins, adaptors, GTPase, kinases and phosphatases. These molecules play critical roles in normal physiological events such as cellular adhesion, movement, cytoskeletal structure and intracellular signaling pathways. In cancers, aberrant expression and altered functions of focal adhesion proteins contribute to adverse tumor behavior. It is evident that these proteins do not function alone, but rather associate and work together in the process of tumor development and cancer metastasis. Focal adhesion proteins have been shown to play critical roles in hepatocellular carcinoma. Understanding the molecular interactions and mechanisms of the interconnected focal adhesion proteins is of particular importance in understanding mechanisms underlying hepatocellular carcinoma progression and development of potential effective treatment.
Collapse
Affiliation(s)
- Judy Wai Ping Yam
- Liver Cancer and Hepatitis Research Laboratory, Department of Pathology, The University of Hong Kong, Pokfulam, Hong Kong
| | | | | |
Collapse
|
31
|
Lee SK, Kim MH, Cheong JY, Cho SW, Yang SJ, Kwack K. Integrin alpha V polymorphisms and haplotypes in a Korean population are associated with susceptibility to chronic hepatitis and hepatocellular carcinoma. Liver Int 2009; 29:187-95. [PMID: 18694400 DOI: 10.1111/j.1478-3231.2008.01843.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND/AIMS Integrins are cell surface receptors for extracellular matrix (ECM) proteins that initiate signalling pathways that modulate proliferation, survival, invasion or metastasis. Consequently, integrins are potential targets for the treatment of cancer. In this study, we investigated whether single nucleotide polymorphisms (SNPs) in integrin alpha(V) (ITGAV) in a Korean population were associated with chronic hepatitis B virus (HBV) infection and HBV-infected hepatocellular carcinoma (HCC). PATIENTS AND METHODS Thirteen ITGAV SNPs in 111 cases of chronic HBV infection, 86 cases of HBV-infected HCC and 107 cases of acute self-limited HBV infection were genotyped using Illumina's Sentrix array matrix (SAM) chip. RESULTS The ITGAV intron SNPs rs9333289 and rs11685758, the 3'-untranslated region SNP rs1839123 and haplotype 3 (T-T-A) were associated with enhanced susceptibility to HBV-infected HCC (OR=1.75-2.42; P=0.02-0.05), while the intron SNP rs2290083 was associated with both chronic infection and HBV-infected HCC (OR=1.73-2.01; P=0.01-0.04). In addition, both rs2290083 and ht1 (C-C-G) were associated with the age at which chronic infection occurred, as determined by Cox relative hazard analysis (RH=1.39-1.62, P=0.04-0.01) CONCLUSION ITGAV SNPs and haplotypes may be genetic factors that increase the susceptibility of Koreans to chronic HBV infection and HBV-infected HCC.
Collapse
Affiliation(s)
- Seung Ku Lee
- Medical Genomics Laboratory, Graduate School of Life Science and Biotechnology, Pochon CHA University, SeongNam, Korea
| | | | | | | | | | | |
Collapse
|
32
|
Moore NM, Barbour TR, Sakiyama-Elbert SE. Synthesis and Characterization of Four-Arm Poly(ethylene glycol)-Based Gene Delivery Vehicles Coupled to Integrin and DNA-Binding Peptides. Mol Pharm 2007; 5:140-50. [DOI: 10.1021/mp700072n] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Nicole M. Moore
- Department of Energy, Environmental, and Chemical Engineering, Department of Biomedical Engineering and Center for Materials Innovation, Washington University, St. Louis, Missouri 63130
| | - Tiffany R. Barbour
- Department of Energy, Environmental, and Chemical Engineering, Department of Biomedical Engineering and Center for Materials Innovation, Washington University, St. Louis, Missouri 63130
| | - Shelly E. Sakiyama-Elbert
- Department of Energy, Environmental, and Chemical Engineering, Department of Biomedical Engineering and Center for Materials Innovation, Washington University, St. Louis, Missouri 63130
| |
Collapse
|
33
|
Abstract
AIM: To study at transcriptional level the similarities and differences of the physiological and biochemical activities between liver tumor (LT) and regenerating liver cells.
METHODS: LT-associated genes and their expression changes in LT were obtained from databases and scientific articles, and their expression profiles in rat liver regeneration (LR) were detected using Rat Genome 230 2.0 array. Subsequently their expression changes in LT and LR were compared and analyzed.
RESULTS: One hundred and twenty one LT-associated genes were found to be LR-associated. Thirty four genes were up-regulated, and 14 genes were down-regulated in both LT and regenerating liver; 20 genes up-regulated in LT were down-regulated in regenerating liver; 21 up-regulated genes and 16 down-regulated genes in LT were up-regulated at some time points and down-regulated at others during LR.
CONCLUSION: Results suggested that apoptosis activity suppressed in LT was still active in regenerating liver, and there are lots of similarities and differences between the LT and regenerating liver at the aspects of cell growth, proliferation, differentiation, migration and angiogenesis.
Collapse
Affiliation(s)
- Cun-Shuan Xu
- College of Life Science, Henan Normal University, Xinxiang 453007, Henan Province, China.
| | | | | | | |
Collapse
|
34
|
Li J, Tan H, Dong X, Xu Z, Shi C, Han X, Jiang H, Krissansen GW, Sun X. Antisense integrin alphaV and beta3 gene therapy suppresses subcutaneously implanted hepatocellular carcinomas. Dig Liver Dis 2007; 39:557-565. [PMID: 17374519 DOI: 10.1016/j.dld.2007.01.025] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2006] [Revised: 01/26/2007] [Accepted: 01/30/2007] [Indexed: 12/11/2022]
Abstract
BACKGROUND Integrin alphaVbeta3 plays a critical role in tumour angiogenesis and metastasis formation, and is recognized as a key therapeutic target in the treatment of cancer. AIM To investigate whether antisense alphaV and beta3 gene therapy has utility in the treatment of hepatocellular carcinomas. METHODS Antisense expression plasmids targeting integrin alphaV or beta3 were constructed, and examined by immunohistochemistry and Western blot analyses for their ability to inhibit alphaV and beta3 expression. The antisense alphaV and beta3 expression vectors, either alone or in combination, were injected into HepG2 hepatomas established subcutaneously in nude mice and tumour growth, angiogenesis and apoptosis were monitored. RESULTS Antisense alphaV and beta3 downregulated the alphaV and beta3 subunits expressed by human umbilical vein endothelial cells, and the alphaV subunit expressed by HepG2 cells. Gene transfer of antisense alphaV and beta3 expression vectors downregulated alphaV and beta3 in HepG2 tumours established in nude mice, inhibited tumour vascularization and growth, and enhanced tumour cell apoptosis. Antisense alphaV suppressed tumour growth more strongly than antisense beta3; however antisense therapy that simultaneously targeted both integrin subunits was more effective than the respective monotherapies. Antisense alphaV and beta3 inhibited tumour angiogenesis to similar extents, by a process that is independent of vascular endothelial growth factor. CONCLUSIONS Antisense gene therapy targeting alphaV integrins warrants consideration as an approach to treat hepatocellular carcinomas.
Collapse
Affiliation(s)
- J Li
- Department of General Surgery, Shandong Provincial Qianfoshan Hospital, Jinan, China
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Zoppi N, Ritelli M, Salvi A, Colombi M, Barlati S. The FN13 peptide inhibits human tumor cells invasion through the modulation of αvβ3 integrins organization and the inactivation of ILK pathway. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2007; 1773:747-63. [PMID: 17383746 DOI: 10.1016/j.bbamcr.2007.02.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2006] [Revised: 01/29/2007] [Accepted: 02/08/2007] [Indexed: 11/19/2022]
Abstract
We report the effect of the stable expression of a 13 amino acid human fibronectin (FN) peptide (FN13) on the organization of the FN extracellular matrix (ECM) and of FN integrin receptors (FNRs), in relationship with the inhibition of cellular invasion, in three FN-ECM defective human tumor-derived cell lines: SK-Hep1C3, hepatoma, ACN, neuroblastoma, and SK-OV-3, ovary carcinoma. All these cell lines stably expressing the FN13 peptide, organized an FN-ECM, disorganized alpha v beta 1 integrins and inactivated the ILK pathway, with the loss of secretion of MMP-9. This was associated with the inhibition of cell invasion in Matrigel matrix only in SK-Hep1C3 and ACN, but not in SK-OV-3 cells. Analysis of the integrin receptors organization showed that the FN13 expressing cells SK-Hep1C3 and ACN organized alpha v beta 3 integrins, whereas SK-OV-3 organized alpha v beta 5 dimers. The functional block of alpha v beta 5 integrins, with an inactivating anti-alpha v beta 5 antibody, led to the induction of alpha v beta 3 integrins also in SK-OV-3 cells, and to the inhibition of cell invasion. These data show that in the human tumor cells studied FN13 inhibits the in vitro invasion through the dissociation of alpha v beta 1 dimers, leading to ILK pathway inactivation, only when the organization of alpha v beta 3 integrins is induced in the plasma membrane.
Collapse
Affiliation(s)
- Nicoletta Zoppi
- Division of Biology and Genetics, Department of Biomedical Sciences and Biotechnology, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | | | | | | | | |
Collapse
|
36
|
Edwards S, Lalor PF, Tuncer C, Adams DH. Vitronectin in human hepatic tumours contributes to the recruitment of lymphocytes in an alpha v beta3-independent manner. Br J Cancer 2006; 95:1545-54. [PMID: 17088900 PMCID: PMC2360745 DOI: 10.1038/sj.bjc.6603467] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
The degree of lymphocyte infiltration is a prognostic factor in liver cancer, but to date the mechanisms by which lymphocytes infiltrate into and are retained in hepatic tumours are poorly understood. We hypothesised that the extracellular matrix glycoprotein vitronectin, a major component of the stroma of hepatic tumours, might play a role in the recruitment and retention of tumour-infiltrating lymphocytes (TIL). Thus, we investigated the ability of vitronectin to support migration and adhesion of TIL isolated from hepatocellular carcinoma and colorectal hepatic metastases. Soluble vitronectin-induced dose-dependent migration of TIL in in vitro chemotaxis and haptotaxis assays and vitronectin in tissue sections was able to support TIL adhesion to tumour stroma. Neither adhesion nor migration was inhibited by a function blocking mAb against the major vitronectin receptor αvβ3 and we were unable to detect αvβ3 on TIL in vitro or in vivo on tumour tissue. However, TIL did express high levels of urokinase-type plasminogen activator receptor (uPAR) and inhibitory antibodies and amiloride both significantly inhibited TIL adhesion to vitronectin and reduced transendothelial migration of lymphocytes across liver endothelium in vitro. Thus, we provide evidence that vitronectin in liver tumours can support the recruitment and retention of effector lymphocytes by an uPAR-dependent mechanism.
Collapse
Affiliation(s)
- S Edwards
- Liver Research Group, Department of Medicine, 5th Floor, Institute of Biomedical Research, Wolfson Drive, The Medical School, University of Birmingham, Birmingham B15 2TT, UK
| | - P F Lalor
- Liver Research Group, Department of Medicine, 5th Floor, Institute of Biomedical Research, Wolfson Drive, The Medical School, University of Birmingham, Birmingham B15 2TT, UK
- E-mail:
| | - C Tuncer
- Liver Research Group, Department of Medicine, 5th Floor, Institute of Biomedical Research, Wolfson Drive, The Medical School, University of Birmingham, Birmingham B15 2TT, UK
| | - D H Adams
- Liver Research Group, Department of Medicine, 5th Floor, Institute of Biomedical Research, Wolfson Drive, The Medical School, University of Birmingham, Birmingham B15 2TT, UK
| |
Collapse
|
37
|
Abstract
Hepatocellular carcinoma (HCC), one of the most common cancers worldwide, is often diagnosed at an advanced stage when most potentially curative therapies such as resection, transplantation or percutaneous and transarterial interventions are of limited efficacy. The fact that HCC is resistant to conventional chemotherapy, and is rarely amenable to radiotherapy, leaves this disease with no effective therapeutic options and a very poor prognosis. Therefore, the development of more effective therapeutic tools and strategies is much needed. HCCs are phenotypically and genetically heterogeneous tumors that commonly emerge on a background of chronic liver disease. However, in spite of this heterogeneity recent insights into the biology of HCC suggest that certain signaling pathways and molecular alterations are likely to play essential roles in HCC development by promoting cell growth and survival. The identification of such mechanisms may open new avenues for the prevention and treatment of HCC through the development of targeted therapies. In this review we will describe the new potential therapeutic targets and clinical developments that have emerged from progress in the knowledge of HCC biology, In addition, recent advances in gene therapy and combined cell and gene therapy, together with new radiotherapy techniques and immunotherapy in patients with HCC will be discussed.
Collapse
Affiliation(s)
- M A Avila
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | | | | | | |
Collapse
|
38
|
Lacher MD, Tiirikainen MI, Saunier EF, Christian C, Anders M, Oft M, Balmain A, Akhurst RJ, Korn WM. Transforming growth factor-beta receptor inhibition enhances adenoviral infectability of carcinoma cells via up-regulation of Coxsackie and Adenovirus Receptor in conjunction with reversal of epithelial-mesenchymal transition. Cancer Res 2006; 66:1648-57. [PMID: 16452224 DOI: 10.1158/0008-5472.can-05-2328] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Expression of the Coxsackie and Adenovirus Receptor (CAR) is frequently reduced in carcinomas, resulting in decreased susceptibility of such tumors to infection with therapeutic adenoviruses. Because CAR participates physiologically in the formation of tight-junction protein complexes, we examined whether molecular mechanisms known to down-regulate cell-cell adhesions cause loss of CAR expression. Transforming growth factor-beta (TGF-beta)-mediated epithelial-mesenchymal transition (EMT) is a phenomenon associated with tumor progression that is characterized by loss of epithelial-type cell-cell adhesion molecules (including E-cadherin and the tight junction protein ZO-1), gain of mesenchymal biochemical markers, such as fibronectin, and acquisition of a spindle cell phenotype. CAR expression is reduced in tumor cells that have undergone EMT in response to TGF-beta. This down-regulation results from repression of CAR gene transcription, whereas altered RNA stability and increased proteasomal protein degradation play no role. Loss of CAR expression in response to TGF-beta is accompanied by reduced susceptibility to adenovirus infection. Indeed, treatment of carcinoma cells with LY2109761, a specific pharmacologic inhibitor of TGF-beta receptor types I and II kinases, resulted in increased CAR RNA and protein levels as well as improved infectability with adenovirus. This was observed in cells induced to undergo EMT by addition of exogenous TGF-beta and in those that were transformed by endogenous autocrine/paracrine TGF-beta. These findings show down-regulation of CAR in the context of EMT and suggest that combination of therapeutic adenoviruses and TGF-beta receptor inhibitors could be an efficient anticancer strategy.
Collapse
Affiliation(s)
- Markus D Lacher
- Division of Gastroenterology and Hematology/Oncology, University of California-San Francisco Comprehensive Cancer Center, San Francisco, CA 94143, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Varga CM, Tedford NC, Thomas M, Klibanov AM, Griffith LG, Lauffenburger DA. Quantitative comparison of polyethylenimine formulations and adenoviral vectors in terms of intracellular gene delivery processes. Gene Ther 2005; 12:1023-32. [PMID: 15815703 DOI: 10.1038/sj.gt.3302495] [Citation(s) in RCA: 155] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
An objective of designing molecular vehicles exhibiting virus-like transgene delivery capabilities but with low toxicity and immunogenicity continues to drive synthetic vector development. As no single step within the gene delivery pathway represents the critical limiting barrier for all vector types under all circumstances, improvements in synthetic vehicle design may be aided by quantitative analysis of the contributions of each step to the overall delivery process. To our knowledge, however, synthetic and viral gene delivery methods have not yet been explicitly compared in terms of these delivery pathway steps in a quantitative manner. As a first address of this challenge, we compare here quantitative parameters characterizing intracellular gene delivery steps for an E1/E3-deleted adenoviral vector and three polyethylenimine (PEI)-based vector formulations, as well as the liposomal transfection reagent Lipofectamine and naked DNA; the cargo is a plasmid encoding the beta-galactosidase gene under a CMV promoter, and the cell host is the C3A human hepatocellular carcinoma line. The parameters were determined by applying a previously validated mathematical model to transient time-course measurements of plasmid uptake and trafficking (from whole-cell and isolated nuclei lysates, by real-time quantitative PCR), and gene expression levels, enabling discovery of those for which the adenoviral vector manifested superiority. Parameter-sensitivity analysis permitted identification of processes most critically rate-limiting for each vector. We find that the adenoviral vector advantage in delivery appears to reside partially in its import to the nuclear compartment, but that its vast superiority in transgene expression arises predominantly in our situation from postdelivery events: on the basis of per-nuclear plasmid, expression efficiency from adenovirus is superior by orders of magnitude over the PEI vectors. We find that a chemical modification of a PEI-based vector, which substantially improves its performance, appears to do so by enhancing certain trafficking rate parameters, such as binding and uptake, endosomal escape, and binding to nuclear import machinery, but leaves endosomal escape as a barrier over which transgene delivery could be most sensitively increased further for this polymer.
Collapse
Affiliation(s)
- C M Varga
- Biological Engineering Division, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | | | | | | | |
Collapse
|
40
|
Lin KT, Yeh SH, Chen DS, Chen PJ, Jou YS. Epigenetic activation of alpha4, beta2 and beta6 integrins involved in cell migration in trichostatin A-treated Hep3B cells. J Biomed Sci 2005; 12:803-13. [PMID: 16132114 DOI: 10.1007/s11373-005-9005-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2005] [Accepted: 06/14/2005] [Indexed: 10/25/2022] Open
Abstract
The epigenetic modulation by histone deacetylase (HDAC) inhibitors including trichostatin A (TSA) has been known to block cell proliferation, induce apoptosis and inhibit cell migration in human cancer cells that represents the potential therapeutic agents for cancers and fibrosis. However, more than 55% of Hep3B cells remained alive after our initial study of 100 nM TSA treatment. To further study the epigenetic modulation and the biological function of newly activated genes by HDAC inhibitor involved in HCC progression and metastasis, we profiled 23 integrin genes including 15alpha and 8beta in TSA-treated Hep3B cells. Six integrins including three down-regulated alpha6, alpha10, beta8 and three significant up-regulated alpha4, beta2, beta6 integrins were revealed after semi-quantitative RT-PCR. To confirm the epigenetic modulation and explore their biological functions, we selected the three significantly up-regulated integrins for confirmation of protein up-regulation, hyperacetylated-histones by ChIP assays, and functional inhibition by specific neutralizing antibodies of integrins. Our results indicated that epigenetic modulation in TSA-treated Hep3B cells up-regulated new integrins including alpha4, beta2 and beta6 and reduced migration activities by specific neutralizing antibodies to 61.3%, 42.4% and 34.5%, respectively. Our novel findings provided a better understanding of the epigenetic modulation of integrins and suggested that targeting the epigenetic up-regulated integrins to abrogate the migration activity might be a promising strategy to prevent HCC progression.
Collapse
Affiliation(s)
- Kuen-Tyng Lin
- Graduate Institute of Life Sciences, National Defense Medical Center, National Defense University, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
41
|
Mayoral R, Fernández-Martínez A, Boscá L, Martín-Sanz P. Prostaglandin E2 promotes migration and adhesion in hepatocellular carcinoma cells. Carcinogenesis 2005; 26:753-761. [PMID: 15661807 DOI: 10.1093/carcin/bgi022] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The effect of the expression of cyclooxygenase-2 (COX-2) and prostaglandin E2 (PGE2) synthesis on cell migration, the secretion of matrix metalloproteinases (MMPs) and the adhesion of human hepatoma cell lines has been investigated. A close correlation was observed between the expression of COX-2 under basal conditions and the secretion of MMP-2 and MMP-9. Cell migration in HuH-7 cells, which express high constitutive levels of COX-2 was significantly inhibited by selective inhibitors of COX-2 and enhanced by exogenous addition of PGE2. Hepatocellular carcinoma (HCC) cells expressed beta1 and alphaV beta3 integrins, exhibiting an increase in cell adhesion onto fibronectin and vitronectin. Moreover, addition of PGE2 increased the beta1 integrin levels and adhesion on vitronectin in HuH-7 cells. Inhibitors of MEK/ERK, p38 MAPK, protein kinases A and C impaired the migration of HuH-7 cells induced by PGE2, indicating the involvement of multiple pathways in the process. Taken together, these results support the existence of a relationship between COX-2-derived PGE2 synthesis, and migration and adhesion through an integrin-dependent pathway in HCC cells.
Collapse
Affiliation(s)
- Rafael Mayoral
- Instituto de Bioquímica, Centro Mixto CSIC-UCM and Centro Nacional de Investigaciones Cardiovasculares, Facultad de Farmacia, Universidad Complutense, 28040 Madrid, Spain
| | | | | | | |
Collapse
|
42
|
von Sengbusch A, Gassmann P, Fisch KM, Enns A, Nicolson GL, Haier J. Focal adhesion kinase regulates metastatic adhesion of carcinoma cells within liver sinusoids. THE AMERICAN JOURNAL OF PATHOLOGY 2005; 166:585-96. [PMID: 15681841 PMCID: PMC1602334 DOI: 10.1016/s0002-9440(10)62280-8] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Organ-specific tumor cell adhesion to extracellular matrix (ECM) components and cell migration into host organs often involve integrin-mediated cellular processes that can be modified by environmental conditions acting on metastasizing tumor cells, such as shear forces within the blood circulation. Since the focal adhesion kinase (FAK) appears to be essential for the regulation of the integrin-mediated adhesive and migratory properties of tumor cells, its role in early steps of the metastatic cascade was investigated using in vitro and in vivo approaches. Human colon and hepatocellular carcinoma cells were used to study adhesive properties under static conditions and in a parallel plate laminar flow chamber in vitro. In addition, intravital fluorescence microscopy was used to investigate early interactions between circulating tumor cells and the microvasculature of potential target organs in vivo. Shear forces caused by hydrodynamic fluid flow induced Tyr-hyperphosphorylation of FAK in cell monolayers. Reduced expression of FAK or its endogenous inhibition by FAK-related non-kinase (FRNK) interfered with early adhesion events to extracellular matrix components under flow conditions. In contrast, tumor cell adhesion to endothelial cells under these conditions was not affected. Furthermore, down-regulation of FAK inhibited metastatic cell adhesion in vivo within the liver sinusoids. In summary, FAK appears to be involved in early events of integrin-mediated adhesion of circulating carcinoma cells under fluid flow in vitro and in vivo. This kinase may take part in the establishment of definitive adhesive interactions that enable adherent tumor cells to resist fluid shear forces, resulting in an organ-specific formation of distant metastases.
Collapse
Affiliation(s)
- Anke von Sengbusch
- Molecular Biology Lab, Department of General Surgery, University Hospital Münster, Waldeyerstrasse 1, 48149 Münster, Germany
| | | | | | | | | | | |
Collapse
|
43
|
Cowden Dahl KD, Robertson SE, Weaver VM, Simon MC. Hypoxia-inducible factor regulates alphavbeta3 integrin cell surface expression. Mol Biol Cell 2005; 16:1901-12. [PMID: 15689487 PMCID: PMC1073670 DOI: 10.1091/mbc.e04-12-1082] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Hypoxia-inducible factor (HIF)-deficient placentas exhibit a number of defects, including changes in cell fate adoption, lack of fetal angiogenesis, hypocellularity, and poor invasion into maternal tissue. HIF is a heterodimeric transcription factor consisting of alpha and beta aryl hydrocarbon receptor nuclear translocator or ARNT) subunits. We used undifferentiated trophoblast stem (TS) cells to characterize HIF-dependent adhesion, migration, and invasion. Arnt(-/-) and Hifalpha(-/-) TS cells exhibit reduced adhesion and migration toward vitronectin compared with wild-type cells. Furthermore, this defect is associated with decreased cell surface expression of integrin alphavbeta3 and significantly decreased expression of this integrin in focal adhesions. Because of the importance of adhesion and migration in tumor progression (in addition to placental development), we examined the affect of culturing B16F0 melanoma cells in 1.5% oxygen (O(2)). Culturing B16F0 melanoma cells at 1.5% O(2) resulted in increased alphavbeta3 integrin surface expression and increased adhesion to and migration toward vitronectin. Together, these data suggest that HIF and O(2) tension influence placental invasion and tumor migration by increasing cell surface expression of alphavbeta3 integrin.
Collapse
Affiliation(s)
- Karen D Cowden Dahl
- Abramson Family Cancer Research Institute, School of Medicine, University of Pennsylvania, Philadelphia, 19104, USA
| | | | | | | |
Collapse
|
44
|
Paradis V, Degos F, Dargère D, Pham N, Belghiti J, Degott C, Janeau JL, Bezeaud A, Delforge D, Cubizolles M, Laurendeau I, Bedossa P. Identification of a new marker of hepatocellular carcinoma by serum protein profiling of patients with chronic liver diseases. Hepatology 2005; 41:40-7. [PMID: 15690480 DOI: 10.1002/hep.20505] [Citation(s) in RCA: 186] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Surface-enhanced laser desorption ionization time-of-flight mass spectrometry (SELDI-TOF MS) is a proteomic technique that enables the profiling of proteins present in any biological material studied. We used this approach to identify new biomarkers of hepatocellular carcinoma (HCC) in the sera of patients with cirrhosis. Sera from 82 patients with cirrhosis, either without (n = 38) or with (n = 44) HCC, were analyzed by SELDI-TOF MS, and the results of the two groups were compared. The most efficient protein peaks leading to discrimination of patients with HCC were selected (receiver operative characteristic curves). The highest-scoring peak combination was established in a first group of serum samples (multinomial regression) and was tested in an independent group. The protein corresponding to the highest discrimination was purified and characterized further. The intensity of 30 protein peaks significantly differed between cirrhotic patients with and without HCC. An algorithm including the six highest-scoring peaks allowed correct classification (presence or absence of HCC) of 92.5% of patients in the test sample set and 90% in the validation sample set. The highest discriminating peak (8900 Da) was purified further and was characterized as the C-terminal part of the V10 fragment of vitronectin. An in vitro study suggested that the increase of the 8900-Da fragment in the serum of patients with HCC may proceed from the cleavage of native vitronectin with metalloproteases, a family of enzymes whose activity is enhanced in HCC. In conclusion, global protein profiling is an efficient approach that enabled us to identify a catalytic fragment ofvitronectin as a new serum marker of HCC in patients with chronic liver diseases.
Collapse
|
45
|
Abstract
Hepatocellular carcinoma (HCC) is the fifth most common cancer, but the third leading cause of cancer death, in the world, with more than 500,000 fatalities annually. The major etiology of HCC/liver cancer in people is hepatitis B virus (HBV), followed by hepatitis C virus infection (HCV), although nonviral causes also play a role in a minority of cases. Recent molecular studies confirm what was suspected: that HCC tissue from different individuals have many phenotypic differences. However, there are clearly features that unify HCC occurring in a background of viral hepatitis B and C. HCC due to HBV and HCV may be an indirect result of enhanced hepatocyte turnover that occurs in an effort to replace infected cells that have been immunologically attacked. Viral functions may also play a more direct role in mediating oncogenesis. This review considers the molecular and cellular mechanisms involved in primary hepatocellular carcinoma, using a viral perspective.
Collapse
Affiliation(s)
- Timothy M Block
- Department of Molecular Pharmacology and Biochemistry, Jefferson Center for Biomedical Research of Thomas Jefferson University, 700 East Butler Ave., Doylestown, PA 18901, USA.
| | | | | | | |
Collapse
|
46
|
Tan ZJ, Hu XG, Cao GS, Tang Y. Analysis of gene expression profile of pancreatic carcinoma using cDNA microarray. World J Gastroenterol 2003; 9:818-23. [PMID: 12679940 PMCID: PMC4611457 DOI: 10.3748/wjg.v9.i4.818] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To identify new diagnostic markers and drug targets, the gene expression profiles of pancreatic cancer were compared with that of adjacent normal tissues utilizing cDNA microarray analysis.
METHODS: cDNA probes were prepared by labeling mRNA from samples of six pancreatic carcinoma tissues with Cy5-dUTP and mRNA from adjacent normal tissues with Cy3-dUTP respectively through reverse transcription. The mixed probes of each sample were then hybridized with 12800 cDNA arrays (12648 unique human cDNA sequences), and the fluorescent signals were scanned by ScanArray 3000 scanner (General Scanning, Inc.). The values of Cy5-dUTP and Cy3-dUTP on each spot were analyzed and calculated by ImaGene 3.0 software (BioDiscovery, Inc.). Differentially expressed genes were screened according to the criterion that the absolute value of natural logarithm of the ratio of Cy5-dUTP to Cy3-dUTP was greater-than 0.69.
RESULTS: Among 6 samples investigated, 301 genes, which accounted for 2.38% of genes on the microarry slides, exhibited differentially expression at least in 5. There were 166 over-expressed genes including 136 having been registered in Genebank, and 135 under-expressed genes including 79 in Genebank in cancerous tissues.
CONCLUSION: Microarray analysis may provide invaluable information on disease pathology, progression, resistance to treatment, and response to cellular microenvironments of pancreatic carcinoma and ultimately may lead to improving early diagnosis and discovering innovative therapeutic approaches for cancer.
Collapse
Affiliation(s)
- Zhi-Jun Tan
- Department of General Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | | | | | | |
Collapse
|