1
|
Sigorski D, Sejda A, Abualsaud N, Krawczyk E, Izycka-Swieszewska E, Kitlinska J. Neuropeptide Y in cancer-biological functions and potential clinical implications. Cancer Metastasis Rev 2025; 44:21. [PMID: 39760953 PMCID: PMC11703900 DOI: 10.1007/s10555-024-10237-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 12/20/2024] [Indexed: 01/07/2025]
Abstract
Neuropeptide Y (NPY) is a sympathetic neurotransmitter widely distributed in the peripheral and central nervous system, affecting many physiological functions. Consequently, dysregulation of the NPY system contributes to numerous pathological disorders, including stress, obesity, and cancer. The pleiotropic functions of NPY in humans are mediated by G protein-coupled receptors (Y1R, Y2R, Y5R), which activate several signaling pathways and thereby regulate cell growth, differentiation, apoptosis, proliferation, angiogenesis, and metabolism. These activities of NPY are highly relevant to tumor biology and known hallmarks of cancer, including sustained proliferative potential, resisting cell death, angiogenesis, invasion, and metastases. In this comprehensive review, we describe the cellular functions of NPY and discuss its role in cancer pathobiology, as well as provide the current state of knowledge pertaining to NPY and its receptors in various cancer types. Moreover, we focus on potential clinical applications targeting the NPY system, such as its role as a prognostic and predictive factor, as well as its utility in cancer diagnostics, imaging, and treatment. Altogether, growing evidence supports the significant role of the NPY system in tumor pathobiology and implicates its potential therapeutic and diagnostic value in modern oncology.
Collapse
Affiliation(s)
- Dawid Sigorski
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University Medical Center, BSB 231A, 3900 Reservoir Rd., NW, Washington, DC, 20057, USA
- Department of Oncology, Collegium Medicum, University of Warmia and Mazury, Olsztyn, Poland
| | - Aleksandra Sejda
- Department of Pathomorphology and Forensic Medicine, Collegium Medicum, University of Warmia and Mazury, Olsztyn, Poland
| | - Nouran Abualsaud
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University Medical Center, BSB 231A, 3900 Reservoir Rd., NW, Washington, DC, 20057, USA
- Experimental Medicine Department, King Abdullah International Medical Research Center, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Ewa Krawczyk
- Department of Pathology, Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA
| | - Ewa Izycka-Swieszewska
- Department of Pathology and Neuropathology, Medical University of Gdansk, Gdansk, Poland
- Department of Pathomorphology, Copernicus Hospital, Gdansk, Poland
| | - Joanna Kitlinska
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University Medical Center, BSB 231A, 3900 Reservoir Rd., NW, Washington, DC, 20057, USA.
| |
Collapse
|
2
|
Sigorski D, Wesołowski W, Gruszecka A, Gulczyński J, Zieliński P, Misiukiewicz S, Kitlińska J, Iżycka-Świeszewska E. Neuropeptide Y and its receptors in prostate cancer: associations with cancer invasiveness and perineural spread. J Cancer Res Clin Oncol 2023; 149:5803-5822. [PMID: 36583743 PMCID: PMC10356636 DOI: 10.1007/s00432-022-04540-x] [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: 09/14/2022] [Accepted: 12/16/2022] [Indexed: 12/31/2022]
Abstract
PURPOSE Neuropeptide Y (NPY) is a pleiotropic peptide, which is involved in many biological mechanisms important in regulation of cell growth and survival. The aim of this study was a comprehensive analysis of the NPY system in prostate pathology. METHODS The study was based on immunohistochemical analysis of NPY and its receptors, Y1R, Y2R and Y5R, in tissue samples from benign prostate (BP), primary prostate cancer (PCa) and PCa bone metastases. Tissue microarray (TMA) technique was employed, with analysis of multiple cores from each specimen. Intensity of the immunoreactivity and expression index (EI), as well as distribution of the immunostaining in neoplastic cells and stromal elements were evaluated. Perineural invasion (PNI) and extraprostatic extension (EPE) were areas of special interests. Moreover, a transwell migration assay on the LNCaP PCa cell line was used to assess the chemotactic properties of NPY. RESULTS Morphological analysis revealed homogeneous membrane and cytoplasmic pattern of NPY staining in cancer cells and its membrane localization with apical accentuation in BP glands. All elements of the NPY system were upregulated in pre-invasive prostate intraepithelial neoplasia, PCa and metastases. EI and staining intensity of NPY receptors were significantly higher in PCa then in BP with correlation between Y2R and Y5R. The strength of expression of the NPY system was further increased in the PNI and EPE areas. In bone metastases, Y1R and Y5R presented high expression scores. CONCLUSION The results of our study suggest that the NPY system is involved in PCa, starting from early stages of its development to disseminated states of the disease, and participates in the invasion of PCa into the auto and paracrine matter.
Collapse
Affiliation(s)
- Dawid Sigorski
- Department of Oncology, Collegium Medicum, University of Warmia and Mazury, 10-228, Olsztyn, Poland
- Department of Oncology and Immuno-Oncology, Warmian-Masurian Cancer Center of the Ministry of the Interior and Administration Hospital, 10-228, Olsztyn, Poland
| | | | - Agnieszka Gruszecka
- Department of Radiology Informatics and Statistics, Medical University of Gdansk, 80-210, Gdansk, Poland
| | - Jacek Gulczyński
- Department of Pathology and Neuropathology, Medical University of Gdańsk, 80-210, Gdańsk, Poland
- Department of Pathomorphology, Copernicus Hospital, 80-803, Gdańsk, Poland
| | - Piotr Zieliński
- Division of Tropical and Parasitic Diseases, University Center of Maritime and Tropical Medicine, 81-519, Gdynia, Poland
| | - Sara Misiukiewicz
- Human Science Department, School of Nursing and Health Studies, Georgetown University Medical Center, Washington, DC, USA
| | - Joanna Kitlińska
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC, 20057, USA.
| | - Ewa Iżycka-Świeszewska
- Department of Pathology and Neuropathology, Medical University of Gdańsk, 80-210, Gdańsk, Poland.
- Department of Pathomorphology, Copernicus Hospital, 80-803, Gdańsk, Poland.
| |
Collapse
|
3
|
Kim KH, Nagappan A, Song B, Lim S, Moon Y. Antibiotic-disrupted ribosome biogenesis facilitates tumor chemokine superinduction. Biochem Pharmacol 2022; 206:115303. [DOI: 10.1016/j.bcp.2022.115303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/11/2022] [Accepted: 10/11/2022] [Indexed: 11/26/2022]
|
4
|
Liu Q, Ma Z, Cao Q, Zhao H, Guo Y, Liu T, Li J. Perineural invasion-associated biomarkers for tumor development. Biomed Pharmacother 2022; 155:113691. [PMID: 36095958 DOI: 10.1016/j.biopha.2022.113691] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022] Open
Abstract
Perineural invasion (PNI) is the process of neoplastic invasion of peripheral nerves and is considered to be the fifth mode of cancer metastasis. PNI has been detected in head and neck tumors and pancreatic, prostate, bile duct, gastric, and colorectal cancers. It leads to poor prognostic outcomes and high local recurrence rates. Despite the increasing number of studies on PNI, targeted therapeutic modalities have not been proposed. The identification of PNI-related biomarkers would facilitate the non-invasive and early diagnosis of cancers, the establishment of prognostic panels, and the development of targeted therapeutic approaches. In this review, we compile information on the molecular mediators involved in PNI-associated cancers. The expression and prognostic significance of molecular mediators and their receptors in PNI-associated cancers are analyzed, and the possible mechanisms of action of these mediators in PNI are explored, as well as the association of cells in the microenvironment where PNI occurs.
Collapse
Affiliation(s)
- Qi Liu
- Department of General Surgery, The Second Hospital of Jilin University, Changchun 130041, China
| | - Zhiming Ma
- Department of General Surgery, The Second Hospital of Jilin University, Changchun 130041, China
| | - Qian Cao
- Department of Education, The Second Hospital of Jilin University, Changchun 130041, China
| | - Hongyu Zhao
- Gastroenterology and Center of Digestive Endoscopy, The Second Hospital of Jilin University, Changchun 130041, China
| | - Yu Guo
- Department of General Surgery, The Second Hospital of Jilin University, Changchun 130041, China
| | - Tongjun Liu
- Department of General Surgery, The Second Hospital of Jilin University, Changchun 130041, China
| | - Jiannan Li
- Department of General Surgery, The Second Hospital of Jilin University, Changchun 130041, China.
| |
Collapse
|
5
|
Integration of Transcriptome and Epigenome to Identify and Develop Prognostic Markers for Ovarian Cancer. JOURNAL OF ONCOLOGY 2022; 2022:3744466. [PMID: 36081667 PMCID: PMC9448543 DOI: 10.1155/2022/3744466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/04/2022] [Accepted: 06/29/2022] [Indexed: 11/21/2022]
Abstract
DNA methylation is a widely researched epigenetic modification. It is associated with the occurrence and development of cancer and has helped evaluate patients' prognoses. However, most existing DNA methylation prognosis models have not simultaneously considered the changes of the downstream transcriptome. Methods. The RNA-Sequencing data and DNA methylation omics data of ovarian cancer patients were downloaded from The Cancer Genome Atlas (TCGA) database. The Consensus Cluster Plus algorithm was used to construct the methylated molecular subtypes of the ovary. Lasso regression was employed to build a multi-gene signature. An independent data set was applied to verify the prognostic value of the signature. The Gene Set Variation Analysis (GSVA) was used to carry out the enrichment analysis of the pathways linked to the gene signature. The IMvigor 210 cohort was used to explore the predictive efficacy of the gene signature for immunotherapy response. Results. We distinguished ovarian cancer samples into two subtypes with different prognosis, based on the omics data of DNA methylation. Differentially expressed genes and enrichment analysis among subtypes indicated that DNA methylation was related to fatty acid metabolism and the extracellular matrix (ECM)-receptor. Furthermore, we constructed an 8-gene signature, which proved to be efficient and stable in predicting prognostics in ovarian cancer patients with different data sets and distinctive pathological characteristics. Finally, the 8-gene signature could predict patients' responses to immunotherapy. The polymerase chain reaction experiment was further used to verify the expression of 8 genes. Conclusion. We analyzed the prognostic value of the related genes of methylation in ovarian cancer. The 8-gene signature predicted the prognosis and immunotherapy response of ovarian cancer patients well and is expected to be valuable in clinical application.
Collapse
|
6
|
Liu SQ, Li B, Li JJ, Sun S, Sun SR, Wu Q. Neuroendocrine regulations in tissue-specific immunity: From mechanism to applications in tumor. Front Cell Dev Biol 2022; 10:896147. [PMID: 36072337 PMCID: PMC9442449 DOI: 10.3389/fcell.2022.896147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 07/27/2022] [Indexed: 11/26/2022] Open
Abstract
Immune responses in nonlymphoid tissues play a vital role in the maintenance of homeostasis. Lots of evidence supports that tissue-specific immune cells provide defense against tumor through the localization in different tissue throughout the body, and can be regulated by diverse factors. Accordingly, the distribution of nervous tissue is also tissue-specific which is essential in the growth of corresponding organs, and the occurrence and development of tumor. Although there have been many mature perspectives on the neuroendocrine regulation in tumor microenvironment, the neuroendocrine regulation of tissue-specific immune cells has not yet been summarized. In this review, we focus on how tissue immune responses are influenced by autonomic nervous system, sensory nerves, and various neuroendocrine factors and reversely how tissue-specific immune cells communicate with neuroendocrine system through releasing different factors. Furthermore, we pay attention to the potential mechanisms of neuroendocrine-tissue specific immunity axis involved in tumors. This may provide new insights for the immunotherapy of tumors in the future.
Collapse
Affiliation(s)
- Si-Qing Liu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Bei Li
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Juan-Juan Li
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Si Sun
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Sheng-Rong Sun
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- *Correspondence: Sheng-Rong Sun, ; Qi Wu,
| | - Qi Wu
- Tongji University Cancer Center, Shanghai Tenth People’s Hospital of Tongji University, School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Sheng-Rong Sun, ; Qi Wu,
| |
Collapse
|
7
|
Chakroborty D, Goswami S, Fan H, Frankel WL, Basu S, Sarkar C. Neuropeptide Y, a paracrine factor secreted by cancer cells, is an independent regulator of angiogenesis in colon cancer. Br J Cancer 2022; 127:1440-1449. [PMID: 35902640 PMCID: PMC9553928 DOI: 10.1038/s41416-022-01916-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Resistance to anti-angiogenic therapies targeting vascular endothelial growth factor-A (VEGF-A) stems from VEGF-A independent angiogenesis mediated by other proangiogenic factors. Therefore identifying these factors in colon adenocarcinoma (CA) will reveal new therapeutic targets. METHODS Neuropeptide Y (NPY) and Y2 receptor (Y2R) expressions in CA were studied by immunohistochemical analysis. Orthotopic HT29 with intact VEGF-A gene and VEGF-A knockdown (by CRISPR/Cas9 gene-editing technique) HT29 colon cancer-bearing mice were treated with specific Y2R antagonists, and the effects on angiogenesis and tumour growth were studied. The direct effect of NPY on angiogenesis and the underlying molecular mechanism was elucidated by the modulation of Y2R receptors expressed on colonic endothelial cells (CEC). RESULTS The results demonstrated that NPY and Y2R are overexpressed in human CA, orthotopic HT29, and most interestingly in VEGF-A-depleted orthotopic HT29 tumours. Treatment with Y2R antagonists inhibited angiogenesis and thereby HT29 tumour growth. Blocking /silencing Y2R abrogated NPY-induced angiogenic potential of CEC. Mechanistically, NPY regulated the activation of the ERK/MAPK signalling pathway in CEC. CONCLUSIONS NPY derived from cancer cells independently regulates angiogenesis in CA by acting through Y2R present on CEC. Targeting NPY/Y2R thus emerges as a novel potential therapeutic strategy in CA.
Collapse
Affiliation(s)
- Debanjan Chakroborty
- Department of Pathology, The Ohio State University, Columbus, OH, 43210, USA.,Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA.,Department of Pathology, University of South Alabama, Mobile, AL, 36617, USA.,Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA.,Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL, 36688, USA
| | - Sandeep Goswami
- Department of Pathology, The Ohio State University, Columbus, OH, 43210, USA.,Department of Pathology, University of South Alabama, Mobile, AL, 36617, USA.,Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA
| | - Hao Fan
- Department of Pathology, The Ohio State University, Columbus, OH, 43210, USA
| | - Wendy L Frankel
- Department of Pathology, The Ohio State University, Columbus, OH, 43210, USA.,Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Sujit Basu
- Department of Pathology, The Ohio State University, Columbus, OH, 43210, USA.,Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA.,Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Chandrani Sarkar
- Department of Pathology, The Ohio State University, Columbus, OH, 43210, USA. .,Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA. .,Department of Pathology, University of South Alabama, Mobile, AL, 36617, USA. .,Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA. .,Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL, 36688, USA.
| |
Collapse
|
8
|
Muniyan S, Pothuraju R, Seshacharyulu P, Batra SK. Macrophage inhibitory cytokine-1 in cancer: Beyond the cellular phenotype. Cancer Lett 2022; 536:215664. [PMID: 35351601 PMCID: PMC9088220 DOI: 10.1016/j.canlet.2022.215664] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/20/2022] [Accepted: 03/23/2022] [Indexed: 01/22/2023]
Abstract
Despite technological advances in diagnostic abilities and improved treatment methods, the burden of cancers remains high, leading to significant morbidity and mortality. One primary reason is that cancer cell secretory factors modulate the tumor microenvironment, supporting tumor growth and circumvents anticancer activities of conventional therapies. Macrophage inhibitory cytokine-1 (MIC-1) is a pleiotropic cytokine elevated in various cancers. MIC-1 regulates various cancer hallmarks, including sustained proliferation, tumor-promoting inflammation, avoiding immune destruction, inducing invasion, metastasis, angiogenesis, and resisting cell death. Despite these facts, the molecular regulation and downstream signaling of MIC-1 in cancer remain elusive, partly because its receptor (GFRAL) was unknown until recently. Binding of MIC-1 to GFRAL recruits the coreceptor tyrosine kinase RET to execute its downstream signaling. So far, studies have shown that GFRAL expression is restricted to the brain stem and is responsible for MIC-1/GFRAL/RET-mediated metabolic disorders. Nevertheless, abundant levels of MIC-1 expression have been reported in all cancer types and have been proposed as a surrogate biomarker. Given the ubiquitous expression of MIC-1 in cancers, it is crucial to understand both upstream regulation and downstream MIC-1/GFRAL/RET signaling in cancer hallmark traits.
Collapse
Affiliation(s)
- Sakthivel Muniyan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| | - Ramesh Pothuraju
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Parthasarathy Seshacharyulu
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
9
|
Sigorski D, Gulczyński J, Sejda A, Rogowski W, Iżycka-Świeszewska E. Investigation of Neural Microenvironment in Prostate Cancer in Context of Neural Density, Perineural Invasion, and Neuroendocrine Profile of Tumors. Front Oncol 2021; 11:710899. [PMID: 34277455 PMCID: PMC8281889 DOI: 10.3389/fonc.2021.710899] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 06/21/2021] [Indexed: 12/20/2022] Open
Abstract
Background Cancer stroma contains the neural compartment with specific components and action. Neural microenvironment processing includes among others axonogenesis, perineural invasion (PNI), neurosignaling, and tumor cell neural/neuroendocrine differentiation. Growing data suggest that tumor-neural crosstalk plays an important function in prostate cancer (PCa) biology. However, the mechanisms involved in PNI and axonogenesis, as well as their patho-clinical correlations in this tumor are unclear. Methods The present study was carried out on FFPE samples of 73 PCa and 15 benign prostate (BP) cases. Immunohistochemistry with neural markers PGP9.5, TH, and NFP was performed on constructed TMAs and selected tissue sections. The analyzed parameters of tumor innervation included small nerve density (ND) measured on pan-neural marker (PGP9.5) and TH s4tained slides, as well assessment of PNI presence and morphology. The qualitative and topographic aspects were studied. In addition, the expression of neuroendocrine marker chromogranin and NPY was assessed with dedicated indexes. The correlations of the above parameters with basic patho-clinical data such as patients’ age, tumor stage, grade, angioinvasion, and ERG status were examined. Results The study showed that innervation parameters differed between cancer and BP. The neural network in PCa revealed heterogeneity, and ND PGP9.5 in tumor was significantly lower than in its periphery. The density of sympathetic TH-positive fibers and its proportion to all fibers was lower in cancer than in the periphery and BP samples. Perineural invasion was confirmed in 76% of cases, usually multifocally, occurring more commonly in tumors with a higher grade. NPY expression in PCa cells was common with its intensity often rising towards PNI. ERG+ tumors showed higher ND, more frequent PNI, and a higher stage. Moreover, chromogranin-positive cells were more pronounced in PCa with higher NPY expression. Conclusions The analysis showed an irregular axonal network in prostate cancer with higher neural density (panneural and adrenergic) in the surroundings and the invasive front. ND and PNI interrelated with NPY expression, neuroendocrine differentiation, and ERG status. The above findings support new evidence for the presence of autocrine and paracrine interactions in prostate cancer neural microenvironment.
Collapse
Affiliation(s)
- Dawid Sigorski
- Department of Oncology, Collegium Medicum, University of Warmia and Mazury, Olsztyn, Poland.,Department of Oncology and Immuno-Oncology, Warmian-Masurian Cancer Center of the Ministry of the Interior and Administration Hospital, Olsztyn, Poland
| | - Jacek Gulczyński
- Department of Pathology and Neuropathology, Medical University of Gdańsk, Gdańsk, Poland.,Department of Pathomorphology, Copernicus Hospital, Gdańsk, Poland
| | - Aleksandra Sejda
- Department of Pathomorphology, Collegium Medicum, University of Warmia and Mazury, Olsztyn, Poland
| | - Wojciech Rogowski
- Department of Health, Pomeranian University in Słupsk, Słupsk, Poland.,Department of Oncology, Chemotherapy, Clinical trials, Regional Hospital, Słupsk, Poland
| | - Ewa Iżycka-Świeszewska
- Department of Pathology and Neuropathology, Medical University of Gdańsk, Gdańsk, Poland.,Department of Pathomorphology, Copernicus Hospital, Gdańsk, Poland
| |
Collapse
|
10
|
Ding Y, Lee M, Gao Y, Bu P, Coarfa C, Miles B, Sreekumar A, Creighton CJ, Ayala G. Neuropeptide Y nerve paracrine regulation of prostate cancer oncogenesis and therapy resistance. Prostate 2021; 81:58-71. [PMID: 33022812 PMCID: PMC7756863 DOI: 10.1002/pros.24081] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 04/26/2020] [Indexed: 01/11/2023]
Abstract
BACKGROUND Nerves are key factors in prostate cancer (PCa) progression. Here, we propose that neuropeptide Y (NPY) nerves are key regulators of cancer-nerve interaction. METHODS We used in vitro models for NPY inhibition studies and subsequent metabolomics, apoptotic and migration assays, and nuclear transcription factor-κB (NF-κB) translocation studies. Human naïve and radiated PCa tissues were used for NPY nerve density biomarker studies. Tissues derived from a Botox denervation clinical trial were used to corroborate metabolomic changes in humans. RESULTS Cancer cells increase NPY positive nerves in vitro and in preneoplastic human tissues. NPY-specific inhibition resulted in increased cancer apoptosis, decreased motility, and energetic metabolic pathway changes. A comparison of metabolomic response in NPY-inhibited cells with the transcriptome response in human PCa patients treated with Botox showed shared 13 pathways, including the tricarboxylic acid cycle. We identified that NF-κB is a potential NPY downstream mediator. Using in vitro models and tissues derived from a previous human chemical denervation study, we show that Botox specifically, but not exclusively, inhibits NPY in cancer. Quantification of NPY nerves is independently predictive of PCa-specific death. Finally, NPY nerves might be involved in radiation therapy (RT) resistance, as radiation-induced apoptosis is reduced when PCa cells are cocultured with dorsal root ganglia/nerves and NPY positive nerves are increased in prostates of patients that failed RT. CONCLUSION These data suggest that targeting the NPY neural microenvironment may represent a therapeutic approach for the treatment of PCa and resistance through the regulation of multiple oncogenic mechanisms.
Collapse
Affiliation(s)
- Yi Ding
- Department of Pathology and Laboratory Medicine, McGovern School of MedicineUniversity of Texas Health Sciences Center Medical SchoolHoustonTexasUSA
| | - MinJae Lee
- Biostatistics, Epidemiology, and Research Design (BERD) Core, Department of Internal MedicineUniversity of Texas Health Sciences Center Medical SchoolHoustonTexasUSA
| | - Yan Gao
- Department of Pathology and Laboratory Medicine, McGovern School of MedicineUniversity of Texas Health Sciences Center Medical SchoolHoustonTexasUSA
| | - Ping Bu
- Department of Pathology and Laboratory Medicine, McGovern School of MedicineUniversity of Texas Health Sciences Center Medical SchoolHoustonTexasUSA
| | - Christian Coarfa
- Department of Molecular & Cell BiologyBaylor College of MedicineHoustonTexasUSA
| | - Brian Miles
- Department of UrologyThe Methodist HospitalHoustonTexasUSA
| | - Arun Sreekumar
- Department of Pathology and Laboratory Medicine, McGovern School of MedicineUniversity of Texas Health Sciences Center Medical SchoolHoustonTexasUSA
| | - Chad J. Creighton
- Department of Internal Medicine, Dan L. Duncan Cancer CenterBaylor College of MedicineHoustonTexasUSA
| | - Gustavo Ayala
- Biostatistics, Epidemiology, and Research Design (BERD) Core, Department of Internal MedicineUniversity of Texas Health Sciences Center Medical SchoolHoustonTexasUSA
| |
Collapse
|
11
|
Sejda A, Sigorski D, Gulczyński J, Wesołowski W, Kitlińska J, Iżycka-Świeszewska E. Complexity of Neural Component of Tumor Microenvironment in Prostate Cancer. Pathobiology 2020; 87:87-99. [PMID: 32045912 DOI: 10.1159/000505437] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 12/16/2019] [Indexed: 11/19/2022] Open
Abstract
The tumor microenvironment (TME) plays an essential role in the development and progression of neoplasms. TME consists of the extracellular matrix and numerous specialized cells interacting with cancer cells by paracrine and autocrine mechanisms. Tumor axonogenesis and neoneurogenesis constitute a developing area of investigation. Prostate cancer (PC) is one of the most common malignancies in men worldwide. During the past years, more and more studies have shown that mechanisms leading to the development of PC are not confined only to the epithelial cancer cell, but also involve the tumor stroma. Different nerve types and neurotransmitters present within the TME are thought to be important factors in PC biology. Moreover, perineural invasion, which is a common way of PC spreading, in parallel creates the neural niche for malignant cells. Cancer neurobiology seems to have become a new discipline to explore the contribution of neoplastic cell interactions with the nervous system and the neural TME component, also to search for potential therapeutic targets in malignant tumors such as PC.
Collapse
Affiliation(s)
- Aleksandra Sejda
- Department of Pathomorphology, Collegium Medicum, University of Warmia and Mazury, Olsztyn, Poland,
| | - Dawid Sigorski
- Department of Oncology, Collegium Medicum, University of Warmia and Mazury, Olsztyn, Poland
| | - Jacek Gulczyński
- Department of Pathology and Neuropathology, Medical University of Gdańsk, Gdańsk, Poland
| | | | - Joanna Kitlińska
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Ewa Iżycka-Świeszewska
- Department of Pathology and Neuropathology, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
12
|
Zhang W, Hu C, Wang X, Bai S, Cao S, Kobelski M, Lambert JR, Gu J, Zhan Y. Role of GDF15 in methylseleninic acid-mediated inhibition of cell proliferation and induction of apoptosis in prostate cancer cells. PLoS One 2019; 14:e0222812. [PMID: 31539407 PMCID: PMC6754141 DOI: 10.1371/journal.pone.0222812] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 09/07/2019] [Indexed: 11/19/2022] Open
Abstract
The growth inhibitory efficacy of methylseleninic acid (MSA) in prostate cancer cells has been documented extensively. However, our understanding of the immediate targets that are key to the growth inhibitory effects of MSA remains limited. Here, using multiple preclinical prostate cancer models, we demonstrated in vitro and in vivo that GDF15 is a most highly induced, immediate target of MSA. We further showed that knockdown of GDF15 mitigates MSA inhibition of cell proliferation and induction of apoptosis. Analysis of gene expression data from over 1000 primary and 200 metastatic prostate cancer samples revealed that GDF15 expression is decreased in metastatic prostate cancers compared to primary tumors and that lower GDF15 levels in primary tumors are associated with higher Gleason scores and shorter survival of the patients. Additionally, pathways that are negatively correlated with GDF15 levels in clinical samples are also negatively correlated with MSA treatment in cultured cells. Since most, if not all, of these pathways have been implicated in prostate cancer progression, suppressing their activities by inducing GDF15 is consistent with the anticancer effects of MSA in prostate cancer. Overall, this study provides support for GDF15 as an immediate target of MSA in prostate cancer cells.
Collapse
Affiliation(s)
- Wenbo Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Cheng Hu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Xiaojie Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin, China
- Department of Structural and Cellular Biology, Tulane Cancer Center, School of Medicine, Tulane University, New Orleans, Louisiana, United States of America
| | - Shanshan Bai
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin, China
- Department of Structural and Cellular Biology, Tulane Cancer Center, School of Medicine, Tulane University, New Orleans, Louisiana, United States of America
| | - Subing Cao
- Department of Structural and Cellular Biology, Tulane Cancer Center, School of Medicine, Tulane University, New Orleans, Louisiana, United States of America
| | - Margaret Kobelski
- Department of Structural and Cellular Biology, Tulane Cancer Center, School of Medicine, Tulane University, New Orleans, Louisiana, United States of America
| | - James R. Lambert
- Department of Pathology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Jingkai Gu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Yang Zhan
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin, China
| |
Collapse
|
13
|
Zheng S, Yang L, Dai Y, Jiang L, Wei Y, Wen H, Xu Y. Screening and Survival Analysis of Hub Genes in Gastric Cancer Based on Bioinformatics. J Comput Biol 2019; 26:1316-1325. [PMID: 31233344 DOI: 10.1089/cmb.2019.0119] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Screening for hub genes associated with gastric cancer and elucidating possible molecular mechanisms of gastric cancer. Five gastric cancer-related gene expression profiles were extracted from the GEO database, and differentially expressed genes (DEGs) were obtained using GEO2R. Gene ontology (GO) enrichment analyses were performed by DAVID, and protein-protein interaction (PPI) network of the DEGs was constructed by STRING and Cytoscape software. Survival value for hub gene comes from the Kaplan-Meier plotter platform. In addition, potential miRNAs of hub genes were predicted by miRWalk. Four hundred seventy-six DEGs were identified in the five expression profiles, these genes are mainly involved in extracellular matrix (ECM)-receptor interaction, chemical carcinogenesis, gastric acid secretion, and PI3K-Akt signaling pathway. Combined with the results of the PPI network and CytoHubba, six hub genes were screened: SERPINH1, NPY, PTGDR, GPER, ADHFE1, and AKR1C1. These genes are highly expressed in gastric cancer tissues, and the overexpression level of these genes is associated with poor survival. A series of miRNAs such as hsa-miRNA-92a-1, hsa-miRNA-647, and hsa-miRNA-507 may play a key role in hub gene regulation. Our studies indicate that SERPINH1, NPY, PTGDR, GPER, ADHFE1, and AKR1C1 may be potential biomarkers and therapeutic targets for gastric cancer in the future.
Collapse
Affiliation(s)
- Shunxin Zheng
- Department of Clinical Laboratory, Liuzhou Tanzhong People's Hospital, Liuzhou, P.R. China
| | - Liuhong Yang
- Reproductive Medicine Centre, Liuzhou Maternal and Children Healthcare Hospital, Liuzhou, P.R. China
| | - Yisong Dai
- Department of Clinical Laboratory, Liuzhou Tanzhong People's Hospital, Liuzhou, P.R. China
| | - Lifang Jiang
- Department of Clinical Laboratory, Liuzhou Tanzhong People's Hospital, Liuzhou, P.R. China
| | - Yi Wei
- Department of Clinical Laboratory, Liuzhou Tanzhong People's Hospital, Liuzhou, P.R. China
| | - Hongwei Wen
- Department of Clinical Laboratory, Liuzhou Tanzhong People's Hospital, Liuzhou, P.R. China
| | - Yingfang Xu
- Department of Clinical Laboratory, Liuzhou Tanzhong People's Hospital, Liuzhou, P.R. China
| |
Collapse
|
14
|
Xie R, Yang Y, Zhang H, Liu H, Guo J, Qin H, Ma Y, Goel A, Li X, Wei Q. c-Myb and its Effector COX-2 as an Indicator Associated with Prognosis and Therapeutic Outcome in Colorectal Cancer. J Cancer 2019; 10:1601-1610. [PMID: 31205515 PMCID: PMC6548004 DOI: 10.7150/jca.27261] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 01/04/2019] [Indexed: 02/06/2023] Open
Abstract
Background: One of our previous studies have demonstrated that the cancer suppressor miR-150 regulated the progression of colorectal cancer (CRC) by down-regulating v-myb avian myeloblastosis viral oncogene homolog (c-Myb). The purpose of present study was to evaluate the prognostic value of the expression of c-Myb and its effector, prostaglandin-endoperoxide synthase 2 (COX-2) in patients with CRC. Methods: We used tissue microarrays (containing 202 CRC tissues and matched adjacent normal tissues) and conducted immunohistochemical analysis and western blotting analysis (containing 3 CRC tissues and matched adjacent normal tissues) to detect the expression of c-Myb and COX-2. Results: Compared with the adjacent nontumorous tissues, both the expression levels of c-Myb and COX-2 were higher in the cancer tissues. A statistically significant correlation was found between the expression of c-Myb and COX-2. Elevated c-Myb and COX-2 were associated with more advanced tumor invasion and poorer overall survival by univariate analysis. Higher expression levels of both c-Myb and COX-2 were significantly associated with shorter overall survival for stage II and stage III patients with 5-Fu based chemotherapy. Multivariate analysis identified the lymph node involvement, distant metastatic spread and the elevated c-Myb and COX-2 as independent factors of poor prognosis for CRC. Conclusions: In conclusion, the overexpression of both c-Myb and COX-2 would be of prognostic screening value in patients with CRC.
Collapse
Affiliation(s)
- Ruting Xie
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University, Shanghai200072, P.R. China
| | - Yongzhi Yang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Huizhen Zhang
- Department of Pathology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University, Shanghai200233, P.R. China
| | - Hu Liu
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University, Shanghai200072, P.R. China
| | - Jing Guo
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University, Shanghai200072, P.R. China
| | - Huanlong Qin
- Department of Surgery, Shanghai Tenth People's Hospital Affiliated with Tongji University, Shanghai 200072, P. R. China
| | - Yanlei Ma
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ajay Goel
- Center for Translational Epigenomics and Oncology, Baylor Scott & White Research Institute and Charles A Sammons Cancer Center, Baylor University Medical Center, Dallas, TX, United States
| | - Xinxiang Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Qing Wei
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University, Shanghai200072, P.R. China
| |
Collapse
|
15
|
Liu J, Dong X, Lu J, Zhang Y, Liu K, Liu L, E Q, Lu X, Yin J, Shen Y. Utility of GDF-15 as a diagnostic biomarker in gastric cancer: an investigation combining GEO, TCGA and meta-analysis. FEBS Open Bio 2019; 9:35-42. [PMID: 30652072 PMCID: PMC6325603 DOI: 10.1002/2211-5463.12537] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 09/20/2018] [Accepted: 09/27/2018] [Indexed: 12/17/2022] Open
Abstract
It was recently suggested that growth differentiation factor-15 (GDF-15) is associated with gastric cancer (GC) carcinogenesis. However, the diagnostic potential of GDF-15 for GC remains unclear. To address this issue, we obtained RNA sequencing and microarray data from the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) databases, and searched PubMed, Google Scholar and Web of Science for relevant literature. We then used STATA to perform a meta-analysis. In total, reports of 253 GC patients and 112 healthy controls who contributed peripheral blood samples were taken from the four literature sources, while information on 754 GC tumor and 263 gastric normal tissues was drawn from TCGA and seven GEO datasets. The expression level of GDF-15 mRNA was significantly higher in tumor tissues than in normal tissues, with a standard mean difference (SMD) of 0.79% and a 95% confidence interval (95% CI) of 0.63-0.95. Consistently, the GDF-15 protein in blood was significantly increased in GC patients as compared to controls (SMD = 3.74, 95% CI = 1.81-5.68). In addition, based on information from TCGA and GEO datasets, the expression level of GDF-15 mRNA may be of use for the diagnosis of GC, with a combined sensitivity, specificity and odds ratio of 0.69 (95% CI = 0.58-0.79), 0.90 (95% CI = 0.84-0.93) and 6.32 (95% CI = 4.22-9.49), respectively. The summary receiver operating characteristic curve demonstrated that the area under the curve was 0.90 (95% CI = 0.87-0.93). The results suggest higher levels of GDF-15 may be associated with GC tumorigenesis and may have the potential to be a diagnostic biomarker of GC.
Collapse
Affiliation(s)
- Jie‐yu Liu
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric DiseasesSchool of Public HealthMedical College of Soochow UniversitySuzhouChina
| | - Xing‐xuan Dong
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric DiseasesSchool of Public HealthMedical College of Soochow UniversitySuzhouChina
| | - Jia‐nan Lu
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric DiseasesSchool of Public HealthMedical College of Soochow UniversitySuzhouChina
| | - Yue Zhang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric DiseasesSchool of Public HealthMedical College of Soochow UniversitySuzhouChina
| | - Kai‐fan Liu
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric DiseasesSchool of Public HealthMedical College of Soochow UniversitySuzhouChina
| | - Ling‐feng Liu
- School of Basic MedicineMedical College of Soochow UniversitySuzhouChina
| | - Qing‐zhi E
- School of Basic MedicineMedical College of Soochow UniversitySuzhouChina
| | - Xiao‐jing Lu
- School of Basic MedicineMedical College of Soochow UniversitySuzhouChina
| | - Jie‐yun Yin
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric DiseasesSchool of Public HealthMedical College of Soochow UniversitySuzhouChina
| | - Yue‐ping Shen
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric DiseasesSchool of Public HealthMedical College of Soochow UniversitySuzhouChina
| |
Collapse
|
16
|
Zhang AY, Chiam K, Haupt Y, Fox S, Birch S, Tilley W, Butler LM, Knudsen K, Comstock C, Rasiah K, Grogan J, Mahon KL, Bianco-Miotto T, Ricciardelli C, Böhm M, Henshall S, Delprado W, Stricker P, Horvath LG, Kench JG. An analysis of a multiple biomarker panel to better predict prostate cancer metastasis after radical prostatectomy. Int J Cancer 2018; 144:1151-1159. [PMID: 30288742 DOI: 10.1002/ijc.31906] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 08/22/2018] [Indexed: 12/23/2022]
Abstract
A plethora of individual candidate biomarkers for predicting biochemical relapse in localized prostate cancer (PCa) have been proposed. Combined biomarkers may improve prognostication, and ensuring validation against more clinically relevant endpoints are required. The Australian PCa Research Centre NSW has contributed to numerous studies of molecular biomarkers associated with biochemical relapse. In the current study, these biomarkers were re-analyzed for biochemical relapse, metastatic relapse and PCa death with extended follow-up. Biomarkers of significance were then used to develop a combined prognostic model for clinical outcomes and validated in a large independent cohort. The discovery cohort (n = 324) was based on 12 biomarkers with a median follow-up of 16 years. Seven biomarkers were significantly associated with biochemical relapse. Three biomarkers were associated with metastases: AZGP1, Ki67 and PML. Only AZGP1 was associated with PCa death. In their individual and combinational forms, AZGP1 and Ki67 as a dual BM signature was the most robust predictor of metastatic relapse (AUC 0.762). The AZPG1 and Ki67 signature was validated in an independent cohort of 347 PCa patients. The dual BM signature of AZGP1 and Ki67 predicted metastasis in the univariable (HR 7.2, 95% CI, 1.6-32; p = 0.01) and multivariable analysis (HR 5.4, 95% CI, 1.2-25; p = 0.03). The dual biomarker signature marginally improved risk prediction compared to AZGP1 alone (AUC 0.758 versus 0.738, p < 0.001). Our findings indicate that biochemical relapse is not an adequate surrogate for metastasis or PCa death. The dual biomarker signature of AZGP1 and Ki67 offers a small benefit in predicting metastasis over AZGP1 alone.
Collapse
Affiliation(s)
- Alison Y Zhang
- Cancer Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,Chris O'Brien Lifehouse, Camperdown, NSW, Australia.,University of Sydney, Camperdown, NSW, Australia
| | - Karen Chiam
- Cancer Research Division, Cancer Council New South Wales, Woolloomooloo, NSW, Australia
| | - Ygal Haupt
- Peter MacCallum Cancer Centre, Parkville, VIC, Australia
| | - Stephen Fox
- Peter MacCallum Cancer Centre, Parkville, VIC, Australia.,University of Melbourne, Parkville, VIC, Australia
| | - Simone Birch
- Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Wayne Tilley
- Freemason's Foundation Centre for Men's Health, University of Adelaide, Adelaide, SA, Australia.,South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Lisa M Butler
- Freemason's Foundation Centre for Men's Health, University of Adelaide, Adelaide, SA, Australia.,South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Karen Knudsen
- Sidney Kimmel Cancer Center, Thomas Jefferson University Hospital, Philadelphia, US
| | - Clay Comstock
- Sidney Kimmel Cancer Center, Thomas Jefferson University Hospital, Philadelphia, US
| | | | - Judith Grogan
- Cancer Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Kate L Mahon
- Cancer Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,Chris O'Brien Lifehouse, Camperdown, NSW, Australia.,Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Tina Bianco-Miotto
- School of Agriculture, Food and Wine, University of Adelaide, Adelaide, SA, Australia
| | - Carmela Ricciardelli
- Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Maret Böhm
- Cancer Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Susan Henshall
- Union for International Cancer Control, Geneva, Switzerland
| | - Warick Delprado
- Douglass Hanly Moir Pathology, Macquarie Park, NSW, Australia
| | - Phillip Stricker
- Cancer Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,Department of Urology, St Vincent's Clinic, Darlinghurst, NSW, Australia
| | - Lisa G Horvath
- Cancer Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,Chris O'Brien Lifehouse, Camperdown, NSW, Australia.,University of Sydney, Camperdown, NSW, Australia.,Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - James G Kench
- Cancer Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,University of Sydney, Camperdown, NSW, Australia.,Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| |
Collapse
|
17
|
Kristensen G, Røder MA, Berg KD, Elversang J, Iglesias-Gato D, Moreira J, Toft BG, Brasso K. Predictive value of combined analysis of pro-NPY and ERG in localized prostate cancer. APMIS 2018; 126:804-813. [PMID: 30191621 DOI: 10.1111/apm.12886] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 07/25/2018] [Indexed: 01/04/2023]
Abstract
This study aimed to investigate if combined analysis of pro-Neuropeptide Y (NPY) and ERG expression in tumor tissue are associated with biochemical failure (BF), castration-based treatment, castration-resistant prostate cancer (CRPC), and prostate cancer (PCa)-specific death for men undergoing radical prostatectomy (RP) for PCa. This study included 315 patients, who underwent RP from 2002 to 2005. Both pro-NPY and ERG expression were analyzed using immunohistochemistry and were scored as low or high and negative or positive, respectively. Risk of BF, castration-based treatment, CRPC, and PCa-specific death were analyzed with multiple cause-specific Cox regression analyses and stratified cumulative incidences using competing risk assessment. Median follow-up was 13.0 years (95% CI: 12.7-13.2). In total, 85.7% were pro-NPY high and 14.3% were pro-NPY low. The combined analyses of pro-NPY and ERG expression was not associated with risk of BF (p = 0.7), castration-based treatment (p = 0.8), CRPC (p = 0.4) or PCa-specific death (p = 0.5). In the multiple cause-specific Cox regression analysis, pro-NPY high and ERG positivity was not associated with BF (HR: 1.02; 95% CI 0.6-1.7; p = 0.94). In conclusion the combination of pro-NPY and ERG expression did not show association with risk of BF, castration-based treatment, CRPC, and PCa-specific death following RP.
Collapse
Affiliation(s)
- Gitte Kristensen
- Copenhagen Prostate Cancer Center, Department of Urology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Martin Andreas Røder
- Copenhagen Prostate Cancer Center, Department of Urology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Kasper Drimer Berg
- Copenhagen Prostate Cancer Center, Department of Urology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Johanna Elversang
- Department of Pathology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Diego Iglesias-Gato
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - José Moreira
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | | | - Klaus Brasso
- Copenhagen Prostate Cancer Center, Department of Urology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
18
|
The MIC-1/GDF15-GFRAL Pathway in Energy Homeostasis: Implications for Obesity, Cachexia, and Other Associated Diseases. Cell Metab 2018; 28:353-368. [PMID: 30184485 DOI: 10.1016/j.cmet.2018.07.018] [Citation(s) in RCA: 261] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
MIC-1/GDF15 is a stress response cytokine and a distant member of the transforming growth factor beta (TGFb) superfamily, with no close relatives. It acts via a recently identified receptor called glial-derived neurotrophic factor (GDNF) receptor alpha-like (GFRAL), which is a distant orphan member of the GDNF receptor family that signals through the tyrosine kinase receptor Ret. MIC-1/GDF15 expression and serum levels rise in response to many stimuli that initiate cell stress and as part of a wide variety of disease processes, most prominently cancer and cardiovascular disease. The best documented actions of MIC-1/GDF15 are on regulation of energy homeostasis. When MIC-1/GDF15 serum levels are substantially elevated in diseases like cancer, it subverts a physiological pathway of appetite regulation to induce an anorexia/cachexia syndrome initiated by its actions on hindbrain neurons. These effects make it a potential target for the treatment of both obesity and anorexia/cachexia syndromes, disorders lacking any highly effective, readily accessible therapies.
Collapse
|
19
|
Zhao D, Wang X, Zhang W. GDF15 predict platinum response during first-line chemotherapy and can act as a complementary diagnostic serum biomarker with CA125 in epithelial ovarian cancer. BMC Cancer 2018; 18:328. [PMID: 29580231 PMCID: PMC5870062 DOI: 10.1186/s12885-018-4246-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 03/19/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Growth differentiation factor 15 (GDF15) has attracted much interest as a novel biomarker for epithelial ovarian carcinoma (EOC). Research focus has been directed at GDF15 as a diagnostic detection, while the prognostic determination of GDF15 in EOC patients remains to be clearly elucidated. The present study aimed to investigate GDF15 level relative to clinicopathological characters, chemoresponse, and clinical outcome of EOC patients. METHODS Serum from 122 patients with primary diagnosed EOC were analyzed for GDF15 and serum cancer antigen 125 (CA125). All cases were treated with debulking surgery and first-line chemotherapy, and samples were obtained just before debulking surgical treatment and first-line chemotherapy. Subsequently, clinical characteristics, responses to chemotherapy and progression-free survival (PFS) were recorded. RESULTS Increasing levels of serum GDF15 was significantly associated with FIGO stage and lymphonodus metastasis. GDF15 and CA125 detection are complementary in the diagnosis of EOC and can be simultaneously profiled. The chemo-resistant EOC patients (median, 1225.0 pg/mL) showed significantly higher GDF15 than chemo-sensitive patients (median, 824.2 pg/mL; P = 0.013). Highly expressed GDF15 was an independent negative prognostic indicator in the PFS (P = 0.026) of the 122 EOC cases in the multivariate analysis. Additionally, patients with high level of serum CA125 significantly associated with suboptimal (P = 0.043) debulking surgery. CONCLUSIONS Our results provide valuable evidence that GDF15 is related with first-line chemo-resistance, with highly expressed GDF15 being a strong and an independent indicator of shorter PFS in EOC patients.
Collapse
Affiliation(s)
- Dan Zhao
- Department of Gynecological Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China
| | - Xiaobing Wang
- State Key Lab of Molecular Oncology, Laboratory of Cell and Molecular Biology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Research Building, No.17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China.
| | - Wei Zhang
- Tumor Marker Research Center, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Research Building, No.17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
20
|
Relevance of MIC-1 in the Era of PSA as a Serum Based Predictor of Prostate Cancer: A Critical Evaluation. Sci Rep 2017; 7:16824. [PMID: 29203798 PMCID: PMC5715056 DOI: 10.1038/s41598-017-17207-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 11/21/2017] [Indexed: 01/30/2023] Open
Abstract
To reduce the ambiguity of contradictory observations in different studies regarding the expression level of Macrophage Inhibitory Cytokine-1 (MIC-1) in serum in prostate cancer (PC), benign prostatic hyperplasia (BPH) and healthy controls (HC), we designed this double-blind study. The study comprises 240 sera from PC, BPH and HC subjects. The expression level of MIC-1 in PC, BPH and HC were appraised using Western blot (WB) and ELISA based approach. WB and ELISA appraisal reveals that the expression level of MIC-1 is significantly higher in PC than in HC or BPH subjects. Regression analysis revealed a significant correlation between MIC-1 vs. PSA (r = 0.09; p < 0.001) and MIC-1 vs. GS (r = 0.7; p < 0.001). ROC analysis using discriminant predicted probability revealed that the MIC-1 was better than PSA. Moreover, the combination of MIC-1 and PSA was allowing 99.1% AUC for the differentiation of BPH + PC from HC, 97.9% AUC for differentiation of BPH from HC, 98.6% AUC for differentiation of PC from HC, and 96.7% AUC for the differentiation of PC from BPH. The augmented expression of MIC-1 in PC compared to BPH and HC subjects is in concurrent of the over-expression of MIC-1 in PC reports and confiscates the contradictory findings of other studies.
Collapse
|
21
|
Danta M, Barber DA, Zhang HP, Lee-Ng M, Baumgart SWL, Tsai VWW, Husaini Y, Saxena M, Marquis CP, Errington W, Kerr S, Breit SN, Brown DA. Macrophage inhibitory cytokine-1/growth differentiation factor-15 as a predictor of colonic neoplasia. Aliment Pharmacol Ther 2017; 46:347-354. [PMID: 28569401 DOI: 10.1111/apt.14156] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 04/30/2017] [Accepted: 04/30/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND Serum macrophage inhibitory cytokine-1 (MIC-1/GDF15) concentration has been associated with colonic adenomas and carcinoma. AIMS To determine whether circulating MIC-1/GDF15 serum concentrations are higher in the presence of adenomas and whether the level decreases after excision. METHODS Patients were recruited prospectively from a single centre and stratified into five groups: no polyps (NP); hyperplastic polyps (HP); sessile serrated ademona (SSA); adenomas (AP); and colorectal carcinoma (CRC). Blood samples were collected immediately before and 4 weeks after colonoscopy. MIC-1/GDF15 serum levels were quantified using ELISA. RESULTS Participants (n=301) were stratified as: NP; n=116 (52%), HP; n=37 (12%), SSA; n=19 (7%), AP; n=68 (23%); and CRC; n=3 (1%). Patients were excluded from the study due to nondiagnostic pathology (n=9, 3%) and exclusion criteria (n=20, 6%). In the 272 remaining subjects (M=149; F=123), age (P=.005), history of colonic polyps (P=.003) and family history of colonic polyps (P=.002) were associated with presence of adenomas. Baseline median MIC-1/GDF15 serum levels increased significantly from NP 609 (460-797) pg/mL, HP 582 (466-852) pg/mL, SSA 561 (446-837) pg/mL to AP 723 (602-1122) pg/mL and CRC 1107 (897-1107) pg/mL; (P<.001). In the pre- and postpolypectomy paired adenoma samples median MIC-1/GDF15 reduced significantly from 722 (603-1164) pg/mL to 685 (561-944) pg/mL (P=.002). A ROC analysis for serum MIC-1/GDF15 to identify adenomatous polyps indicated an area under the curve of 0.71. CONCLUSIONS Our data suggest that serum MIC-1/GDF15 has the diagnostic characteristics to increase the detection of colonic neoplasia and improve screening.
Collapse
Affiliation(s)
- M Danta
- St Vincent's Clinical School, Faculty of Medicine, UNSW, Sydney, NSW, Australia.,Department of Gastroenterology, St Vincent's Hospital, Sydney, NSW, Australia
| | - D A Barber
- St Vincent's Clinical School, Faculty of Medicine, UNSW, Sydney, NSW, Australia
| | - H P Zhang
- St Vincent's Centre of Applied Medical Research, St Vincent's Hospital, Sydney, NSW, Australia
| | - M Lee-Ng
- Department of Gastroenterology, St Vincent's Hospital, Sydney, NSW, Australia
| | - S W L Baumgart
- St Vincent's Clinical School, Faculty of Medicine, UNSW, Sydney, NSW, Australia
| | - V W W Tsai
- St Vincent's Clinical School, Faculty of Medicine, UNSW, Sydney, NSW, Australia.,St Vincent's Centre of Applied Medical Research, St Vincent's Hospital, Sydney, NSW, Australia
| | - Y Husaini
- St Vincent's Clinical School, Faculty of Medicine, UNSW, Sydney, NSW, Australia.,St Vincent's Centre of Applied Medical Research, St Vincent's Hospital, Sydney, NSW, Australia
| | - M Saxena
- St Vincent's Centre of Applied Medical Research, St Vincent's Hospital, Sydney, NSW, Australia
| | - C P Marquis
- The Westmead Institute for Medical Research, University of Sydney, Sydney, NSW, Australia
| | - W Errington
- St Vincent's Clinical School, Faculty of Medicine, UNSW, Sydney, NSW, Australia
| | - S Kerr
- Biostatistics, Kirby Institute, UNSW, Sydney, NSW, Australia
| | - S N Breit
- St Vincent's Clinical School, Faculty of Medicine, UNSW, Sydney, NSW, Australia.,St Vincent's Centre of Applied Medical Research, St Vincent's Hospital, Sydney, NSW, Australia
| | - D A Brown
- St Vincent's Clinical School, Faculty of Medicine, UNSW, Sydney, NSW, Australia.,St Vincent's Centre of Applied Medical Research, St Vincent's Hospital, Sydney, NSW, Australia.,The Westmead Institute for Medical Research, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
22
|
Tilan J, Kitlinska J. Neuropeptide Y (NPY) in tumor growth and progression: Lessons learned from pediatric oncology. Neuropeptides 2016; 55:55-66. [PMID: 26549645 PMCID: PMC4755837 DOI: 10.1016/j.npep.2015.10.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 10/25/2015] [Accepted: 10/25/2015] [Indexed: 12/11/2022]
Abstract
Neuropeptide Y (NPY) is a sympathetic neurotransmitter with pleiotropic actions, many of which are highly relevant to tumor biology. Consequently, the peptide has been implicated as a factor regulating the growth of a variety of tumors. Among them, two pediatric malignancies with high endogenous NPY synthesis and release - neuroblastoma and Ewing sarcoma - became excellent models to investigate the role of NPY in tumor growth and progression. The stimulatory effect on tumor cell proliferation, survival, and migration, as well as angiogenesis in these tumors, is mediated by two NPY receptors, Y2R and Y5R, which are expressed in either a constitutive or inducible manner. Of particular importance are interactions of the NPY system with the tumor microenvironment, as hypoxic conditions commonly occurring in solid tumors strongly activate the NPY/Y2R/Y5R axis. This activation is triggered by hypoxia-induced up-regulation of Y2R/Y5R expression and stimulation of dipeptidyl peptidase IV (DPPIV), which converts NPY to a selective Y2R/Y5R agonist, NPY(3-36). While previous studies focused mainly on the effects of NPY on tumor growth and vascularization, they also provided insight into the potential role of the peptide in tumor progression into a metastatic and chemoresistant phenotype. This review summarizes our current knowledge of the role of NPY in neuroblastoma and Ewing sarcoma and its interactions with the tumor microenvironment in the context of findings in other malignancies, as well as discusses future directions and potential clinical implications of these discoveries.
Collapse
Affiliation(s)
- Jason Tilan
- Department of Nursing, School of Nursing and Health Studies, Georgetown University, Washington, DC 20057, USA; Department of Human Science, School of Nursing and Health Studies, Georgetown University, Washington, DC 20057, USA
| | - Joanna Kitlinska
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC 20057, USA.
| |
Collapse
|
23
|
Iglesias-Gato D, Wikström P, Tyanova S, Lavallee C, Thysell E, Carlsson J, Hägglöf C, Cox J, Andrén O, Stattin P, Egevad L, Widmark A, Bjartell A, Collins CC, Bergh A, Geiger T, Mann M, Flores-Morales A. The Proteome of Primary Prostate Cancer. Eur Urol 2015; 69:942-52. [PMID: 26651926 DOI: 10.1016/j.eururo.2015.10.053] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 10/29/2015] [Indexed: 12/15/2022]
Abstract
BACKGROUND Clinical management of the prostate needs improved prognostic tests and treatment strategies. Because proteins are the ultimate effectors of most cellular reactions, are targets for drug actions and constitute potential biomarkers; a quantitative systemic overview of the proteome changes occurring during prostate cancer (PCa) initiation and progression can result in clinically relevant discoveries. OBJECTIVES To study cellular processes altered in PCa using system-wide quantitative analysis of changes in protein expression in clinical samples and to identify prognostic biomarkers for disease aggressiveness. DESIGN, SETTING, AND PARTICIPANTS Mass spectrometry was used for genome-scale quantitative proteomic profiling of 28 prostate tumors (Gleason score 6-9) and neighboring nonmalignant tissue in eight cases, obtained from formalin-fixed paraffin-embedded prostatectomy samples. Two independent cohorts of PCa patients (summing 752 cases) managed by expectancy were used for immunohistochemical evaluation of proneuropeptide-Y (pro-NPY) as a prognostic biomarker. RESULTS AND LIMITATIONS Over 9000 proteins were identified as expressed in the human prostate. Tumor tissue exhibited elevated expression of proteins involved in multiple anabolic processes including fatty acid and protein synthesis, ribosomal biogenesis and protein secretion but no overt evidence of increased proliferation was observed. Tumors also showed increased levels of mitochondrial proteins, which was associated with elevated oxidative phosphorylation capacity measured in situ. Molecular analysis indicated that some of the proteins overexpressed in tumors, such as carnitine palmitoyltransferase 2 (CPT2, fatty acid transporter), coatomer protein complex, subunit alpha (COPA, vesicle secretion), and mitogen- and stress-activated protein kinase 1 and 2 (MSK1/2, protein kinase) regulate the proliferation of PCa cells. Additionally, pro-NPY was found overexpressed in PCa (5-fold, p<0.05), but largely absent in other solid tumor types. Pro-NPY expression, alone or in combination with the ERG status of the tumor, was associated with an increased risk of PCa specific mortality, especially in patients with Gleason score ≤ 7 tumors. CONCLUSIONS This study represents the first system-wide quantitative analysis of proteome changes associated to localized prostate cancer and as such constitutes a valuable resource for understanding the complex metabolic changes occurring in this disease. We also demonstrated that pro-NPY, a protein that showed differential expression between high and low risk tumors in our proteomic analysis, is also a PCa specific prognostic biomarker associated with increased risk for disease specific death in patients carrying low risk tumors. PATIENT SUMMARY The identification of proteins whose expression change in prostate cancer provides novel mechanistic information related to the disease etiology. We hope that future studies will prove the value of this proteome dataset for development of novel therapies and biomarkers.
Collapse
Affiliation(s)
- Diego Iglesias-Gato
- IVS, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Novo Nordisk Foundation Centre for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Danish Cancer Society, Copenhagen, Denmark.
| | - Pernilla Wikström
- Department of Medical Biosciences, Pathology, Umea University, Umea, Sweden
| | - Stefka Tyanova
- Department of Proteomics and Signal Transduction, Max Planck Institute for Biochemistry, Martinsried, Germany
| | - Charlotte Lavallee
- IVS, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Novo Nordisk Foundation Centre for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Danish Cancer Society, Copenhagen, Denmark
| | - Elin Thysell
- Department of Medical Biosciences, Pathology, Umea University, Umea, Sweden
| | - Jessica Carlsson
- School of Health and Medical Sciences, Department of Urology, University of Örebro, Sweden
| | - Christina Hägglöf
- Department of Medical Biosciences, Pathology, Umea University, Umea, Sweden
| | - Jürgen Cox
- Department of Proteomics and Signal Transduction, Max Planck Institute for Biochemistry, Martinsried, Germany
| | - Ove Andrén
- School of Health and Medical Sciences, Department of Urology, University of Örebro, Sweden
| | - Pär Stattin
- Departments of Surgery and Perioperative Sciences, Umea University, Umea, Sweden
| | - Lars Egevad
- Section of Urology, Department of Surgical Science, Karolinska Institutet, Stockholm, Sweden
| | - Anders Widmark
- Department of Radiation Sciences, Oncology, Umea University, Umea, Sweden
| | - Anders Bjartell
- Department of Translational Medicine, Division of Urological Cancers, University of Lund, Lund, Sweden
| | - Colin C Collins
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Anders Bergh
- Department of Medical Biosciences, Pathology, Umea University, Umea, Sweden
| | - Tamar Geiger
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Matthias Mann
- Novo Nordisk Foundation Centre for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Proteomics and Signal Transduction, Max Planck Institute for Biochemistry, Martinsried, Germany
| | - Amilcar Flores-Morales
- IVS, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Novo Nordisk Foundation Centre for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Danish Cancer Society, Copenhagen, Denmark.
| |
Collapse
|
24
|
Schiegnitz E, Kämmerer PW, Rode K, Schorn T, Brieger J, Al-Nawas B. Growth differentiation factor 15 as a radiation-induced marker in oral carcinoma increasing radiation resistance. J Oral Pathol Med 2015; 45:63-9. [PMID: 25880686 DOI: 10.1111/jop.12323] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2015] [Indexed: 12/21/2022]
Abstract
BACKGROUND Growth differentiation factor 15 (GDF15) is involved in tumor pathogenesis of oral squamous cell carcinoma (OSCC). The aim of this study was an investigation of the potential influence of GDF15 on radioresistance of OSCC cells in vitro. METHODS Oral squamous cell carcinoma cell lines were irradiated with 0, 2, or 6 Gy, and GDF15 expression in the supernatant per survived cell colony was examined with ELISA. Non-irradiated and OSCC cell lines irradiated with 6 Gy were evaluated for GDF15 expression using immunofluorescent staining. For further investigation of GDF15 effects on radioresistance, a GDF15 knockdown model in a human OSCC cell line was established, and apoptotic activity after radiation was measured using the Caspase-Glo 3/7 system. RESULTS ELISA and immunofluorescent staining indicated an increased GDF15 expression in 5 OSCC cell lines compared with human gingival epithelial cells. Irradiation with two and six gray resulted in a significant elevation of GDF15 expression per survived cell colony in the irradiated OSCC cell lines (P < 0.001). Furthermore, a dose-dependent expression of GDF15 was seen. Immunofluorescent staining confirmed an elevated GDF15 expression in irradiated OSCC cell lines (n = 10; P ≤ 0.001). Apoptotic activity was significantly increased after irradiation in the GDF15 knockdown group compared with control cells (n = 24; P < 0.001). CONCLUSION This study describes for the first time the vital role of GDF15 both in tumorigenesis and in radioresistance of OSCC cells. With its anti-apoptotic effects, GDF15 possibly promotes tumor progression and might protect carcinoma cells against irradiation effects. Consequently, GDF15 may be a promising therapeutic target in oral cancer.
Collapse
Affiliation(s)
- Eik Schiegnitz
- Department of Oral and Maxillofacial Surgery, Plastic Surgery, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Peer W Kämmerer
- Department of Oral and Maxillofacial Surgery, Plastic Surgery, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany.,Department of Oral and Maxillofacial Surgery, Plastic Surgery, University Medical Centre, Rostock, Germany
| | - Katharina Rode
- Department of Oral and Maxillofacial Surgery, Plastic Surgery, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Thomas Schorn
- Department of Oral and Maxillofacial Surgery, Plastic Surgery, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Jürgen Brieger
- Department of Otorhinolaryngology, Molecular Tumor Biology Laboratory, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Bilal Al-Nawas
- Department of Oral and Maxillofacial Surgery, Plastic Surgery, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
25
|
Li J, Veltri RW, Yuan Z, Christudass CS, Mandecki W. Macrophage inhibitory cytokine 1 biomarker serum immunoassay in combination with PSA is a more specific diagnostic tool for detection of prostate cancer. PLoS One 2015; 10:e0122249. [PMID: 25853582 PMCID: PMC4390224 DOI: 10.1371/journal.pone.0122249] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 02/19/2015] [Indexed: 12/26/2022] Open
Abstract
Background Prostate cancer (PCa) is the most common malignancy among men in the United States. Though highly sensitive, the often-used prostate-specific antigen (PSA) test has low specificity which leads to overdiagnosis and overtreatment of PCa. This paper presents results of a retrospective study that indicates that testing for macrophage inhibitory cytokine 1 (MIC-1) concentration along with the PSA assay could provide much improved specificity to the assay. Methods The MIC-1 serum level was determined by a novel p-Chip-based immunoassay run on 70 retrospective samples. The assay was configured on p-Chips, small integrated circuits (IC) capable of storing in their electronic memories a serial number to identify the molecular probe immobilized on its surface. The distribution of MIC-1 and pre-determined PSA concentrations were displayed in a 2D plot and the predictive power of the dual MIC-1/PSA assay was analyzed. Results MIC-1 concentration in serum was elevated in PCa patients (1.44 ng/ml) compared to normal and biopsy-negative individuals (0.93 ng/ml and 0.88 ng/ml, respectively). In addition, the MIC-1 level was correlated with the progression of PCa. The area under the receiver operator curve (AUC-ROC) was 0.81 providing an assay sensitivity of 83.3% and specificity of 60.7% by using a cutoff of 0.494 for the logistic regression value of MIC-1 and PSA. Another approach, by defining high-frequency PCa zones in a two-dimensional plot, resulted in assay sensitivity of 78.6% and specificity of 89.3%. Conclusions The analysis based on correlation of MIC-1 and PSA concentrations in serum with the patient PCa status improved the specificity of PCa diagnosis without compromising the high sensitivity of the PSA test alone and has potential for PCa prognosis for patient therapy strategies.
Collapse
Affiliation(s)
- Ji Li
- PharmaSeq, Inc., Monmouth Junction, New Jersey, United States of America
| | - Robert W. Veltri
- Johns Hopkins University School of Medicine (JHUSOM), Baltimore, Maryland, United States of America
| | - Zhen Yuan
- PharmaSeq, Inc., Monmouth Junction, New Jersey, United States of America
| | | | - Wlodek Mandecki
- PharmaSeq, Inc., Monmouth Junction, New Jersey, United States of America
- * E-mail:
| |
Collapse
|
26
|
Blanco-Calvo M, Tarrío N, Reboredo M, Haz-Conde M, García J, Quindós M, Figueroa A, Antón-Aparicio L, Calvo L, Valladares-Ayerbes M. Circulating levels of GDF15, MMP7 and miR-200c as a poor prognostic signature in gastric cancer. Future Oncol 2015; 10:1187-202. [PMID: 24947260 DOI: 10.2217/fon.13.263] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
AIM To analyze GDF15 and MMP7 serum levels as diagnostic biomarkers in gastric cancer (GC) patients. The prognostic value of GDF15 and MMP7 serum levels in combination with miR-200c blood expression was also analyzed. PATIENTS & METHODS Fifty-two GC and 23 control samples were included. RESULTS GDF15 and MMP7 proved to be powerful tools for GC diagnosis. Increased levels of GDF15 and MMP7 were associated with shorter progression-free survival and overall survival in univariate analysis. In multivariate analysis, the combination of high levels of GDF15, MMP7 and miR-200c was an independent predictor for death (p = 0.033). CONCLUSION GDF15 and MMP7 serum levels have diagnostic value for GC. The combination marker formed by GDF15, MMP7 and miR-200c is indicative of adverse evolution in GC patients.
Collapse
Affiliation(s)
- Moisés Blanco-Calvo
- Translational Cancer Research Department, La Coruña Biomedical Research Institute (INIBIC), Carretera del Pasaje s/n, 15006 La Coruña, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
The role of inflammation in prostate cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 816:153-81. [PMID: 24818723 DOI: 10.1007/978-3-0348-0837-8_7] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In the United States and in "Westernized" countries, the prevalence of both prostate cancer and prostate inflammation is very high, indicating that the two pathologies could be causally related. Indeed, chronic inflammation is now regarded as an "enabling" characteristic of human cancer. Prostate cancer incidence is thought to be mediated in part by genetics, but also by environmental exposures, including the same exposures that may contribute to the development of prostatic inflammation. As our understanding of the role of inflammation in cancer deepens, it is increasingly apparent that "inflammation" as a whole is a complex entity that does not always play a negative role in cancer etiology. In fact, inflammation can play potentially dichotomous (both pro and antitumorigenic) roles depending on the nature and the cellular makeup of the immune response. This chapter will focus on reviewing the current state of knowledge on the role of innate and adaptive immune cells within the prostate tumor microenvironment and their seemingly complex role in prostate cancer in preventing versus promoting initiation and progression of the disease.
Collapse
|
28
|
Zhou Z, Li W, Song Y, Wang L, Zhang K, Yang J, Zhang W, Su H, Zhang Y. Growth differentiation factor-15 suppresses maturation and function of dendritic cells and inhibits tumor-specific immune response. PLoS One 2013; 8:e78618. [PMID: 24236027 PMCID: PMC3827235 DOI: 10.1371/journal.pone.0078618] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 09/14/2013] [Indexed: 01/24/2023] Open
Abstract
Dendritic cells (DCs) play a key role in the initiation stage of an antigen-specific immune response. A variety of tumor-derived factors (TDFs) can suppress DC maturation and function, resulting in defects in the tumor-specific immune response. To identify unknown TDFs that may suppress DCs maturation and function, we established a high-throughput screening technology based on a human liver tumor T7 phage cDNA library and screened all of the proteins derived from hepatoma cells that potentially interact with immature DCs. Growth/differentiation factor-15 (GDF-15) was detected and chosen for further study. By incubation of DCs cultures with GDF-15, we demonstrate that GDF-15 can inhibit surface protrusion formation during DC maturation; suppress the membrane expression of CD83, CD86 and HLA-DR on DCs; enhance phagocytosis by DCs; reduce IL-12 and elevate TGF-β1 secretion by DCs; inhibit T cell stimulation and cytotoxic T lymphocyte (CTL) activation by DCs. By building tumor-bearing mouse models, we demonstrate that GDF-15 can inhibit the ability of DCs to stimulate a tumor-specific immune response in vivo. These results indicate that GDF-15 may be one of the critical molecules that inhibit DC maturation and function and are involved in tumor immune escape. Thus, GDF-15 may be a novel target in tumor immunotherapy.
Collapse
Affiliation(s)
- Zhizhong Zhou
- The State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
- Department of Biopharmaceutics, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Weina Li
- The State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
- Department of Biopharmaceutics, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yang Song
- Department of Oncology, Tangdu Hospital, Xi'an, Shaanxi, China
| | - Lili Wang
- The State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
- Department of Biopharmaceutics, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Kuo Zhang
- School of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Jing Yang
- Department of Oncology, Tangdu Hospital, Xi'an, Shaanxi, China
| | - Wei Zhang
- The State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
- Department of Biopharmaceutics, Fourth Military Medical University, Xi'an, Shaanxi, China
- * E-mail: (YQZ); (HS); (WZ)
| | - Haichuan Su
- Department of Oncology, Tangdu Hospital, Xi'an, Shaanxi, China
- * E-mail: (YQZ); (HS); (WZ)
| | - Yingqi Zhang
- The State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
- Department of Biopharmaceutics, Fourth Military Medical University, Xi'an, Shaanxi, China
- * E-mail: (YQZ); (HS); (WZ)
| |
Collapse
|
29
|
Ueda K, Tatsuguchi A, Saichi N, Toyama A, Tamura K, Furihata M, Takata R, Akamatsu S, Igarashi M, Nakayama M, Sato TA, Ogawa O, Fujioka T, Shuin T, Nakamura Y, Nakagawa H. Plasma Low-Molecular-Weight Proteome Profiling Identified Neuropeptide-Y as a Prostate Cancer Biomarker Polypeptide. J Proteome Res 2013; 12:4497-506. [DOI: 10.1021/pr400547s] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Koji Ueda
- Laboratory
for Biomarker Development, Center for Genomic Medicine, RIKEN, General
Research Building 6F, Institute of Medical Science, 4-6-1, Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Ayako Tatsuguchi
- Laboratory
for Biomarker Development, Center for Genomic Medicine, RIKEN, General
Research Building 6F, Institute of Medical Science, 4-6-1, Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Naomi Saichi
- Laboratory
for Biomarker Development, Center for Genomic Medicine, RIKEN, General
Research Building 6F, Institute of Medical Science, 4-6-1, Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Atsuhiko Toyama
- Life
Science Research Center, Shimadzu Corporation, 1-3, Nishikicho, Kanda, Chiyoda-ku, Tokyo 101-8448, Japan
| | - Kenji Tamura
- Department
of Urology, Kochi Medical School Hospital, Oko-cho Kohasu, Nankoku-shi, Kochi 783-8505, Japan
| | - Mutsuo Furihata
- Department
of Pathology, Kochi Medical School, Oko-cho Kohasu, Nankoku-shi, Kochi 783-8505, Japan
| | - Ryo Takata
- Department
of Urology, Iwate Medical University, 19-1 Uchimaru, Morioka, Iwate Prefecture 020-0023, Japan
| | - Shusuke Akamatsu
- Department
of Urology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho,
Sakyo-ku, Kyoto 606-8501, Japan
| | - Masahiro Igarashi
- Endoscopy
Division, Gastrointestinal Center, Cancer Institute Hospital, 3-8-31, Ariake, Koto, Tokyo 135-8550, Japan
| | - Masato Nakayama
- Toppan Printing Co., Ltd., 1-5-1, Taito, Taito-ku, Tokyo 110-8560, Japan
| | - Taka-Aki Sato
- Life
Science Research Center, Shimadzu Corporation, 1-3, Nishikicho, Kanda, Chiyoda-ku, Tokyo 101-8448, Japan
| | - Osamu Ogawa
- Department
of Urology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho,
Sakyo-ku, Kyoto 606-8501, Japan
| | - Tomoaki Fujioka
- Department
of Urology, Iwate Medical University, 19-1 Uchimaru, Morioka, Iwate Prefecture 020-0023, Japan
| | - Taro Shuin
- Department
of Urology, Kochi Medical School Hospital, Oko-cho Kohasu, Nankoku-shi, Kochi 783-8505, Japan
| | - Yusuke Nakamura
- Section
of Hematology/Oncology, Department of Medicine Faculty, The University of Chicago, 5841 South Maryland Avenue, Chicago, Illinois, United States
| | - Hidewaki Nakagawa
- Laboratory
for Biomarker Development, Center for Genomic Medicine, RIKEN, General
Research Building 6F, Institute of Medical Science, 4-6-1, Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| |
Collapse
|
30
|
The multiple facets of the TGF-β family cytokine growth/differentiation factor-15/macrophage inhibitory cytokine-1. Cytokine Growth Factor Rev 2013; 24:373-84. [DOI: 10.1016/j.cytogfr.2013.05.003] [Citation(s) in RCA: 193] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 05/21/2013] [Indexed: 12/23/2022]
|
31
|
Massoner P, Kugler KG, Unterberger K, Kuner R, Mueller LAJ, Fälth M, Schäfer G, Seifarth C, Ecker S, Verdorfer I, Graber A, Sültmann H, Klocker H. Characterization of transcriptional changes in ERG rearrangement-positive prostate cancer identifies the regulation of metabolic sensors such as neuropeptide Y. PLoS One 2013; 8:e55207. [PMID: 23390522 PMCID: PMC3563644 DOI: 10.1371/journal.pone.0055207] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 12/27/2012] [Indexed: 12/15/2022] Open
Abstract
ERG gene rearrangements are found in about one half of all prostate cancers. Functional analyses do not fully explain the selective pressure causing ERG rearrangement during the development of prostate cancer. To identify transcriptional changes in prostate cancer, including tumors with ERG gene rearrangements, we performed a meta-analysis on published gene expression data followed by validations on mRNA and protein levels as well as first functional investigations. Eight expression studies (n = 561) on human prostate tissues were included in the meta-analysis. Transcriptional changes between prostate cancer and non-cancerous prostate, as well as ERG rearrangement-positive (ERG+) and ERG rearrangement-negative (ERG−) prostate cancer, were analyzed. Detailed results can be accessed through an online database. We validated our meta-analysis using data from our own independent microarray study (n = 57). 84% and 49% (fold-change>2 and >1.5, respectively) of all transcriptional changes between ERG+ and ERG− prostate cancer determined by meta-analysis were verified in the validation study. Selected targets were confirmed by immunohistochemistry: NPY and PLA2G7 (up-regulated in ERG+ cancers), and AZGP1 and TFF3 (down-regulated in ERG+ cancers). First functional investigations for one of the most prominent ERG rearrangement-associated genes - neuropeptide Y (NPY) - revealed increased glucose uptake in vitro indicating the potential role of NPY in regulating cellular metabolism. In summary, we found robust population-independent transcriptional changes in prostate cancer and first signs of ERG rearrangements inducing metabolic changes in cancer cells by activating major metabolic signaling molecules like NPY. Our study indicates that metabolic changes possibly contribute to the selective pressure favoring ERG rearrangements in prostate cancer.
Collapse
Affiliation(s)
- Petra Massoner
- Division of Experimental Urology, Department of Urology, Innsbruck Medical University, Innsbruck, Austria
- Oncotyrol, Center for Personalized Cancer Medicine GmbH, Innsbruck, Austria
- * E-mail: (PM); (HK)
| | - Karl G. Kugler
- Institute for Bioinformatics and Translational Research, University for Health Sciences, Medical Informatics and Technology (UMIT), Hall in Tirol, Austria
| | - Karin Unterberger
- Oncotyrol, Center for Personalized Cancer Medicine GmbH, Innsbruck, Austria
| | - Ruprecht Kuner
- Unit Cancer Genome Research, Division of Molecular Genetics, German Cancer Research Center and National Center of Tumor Diseases, Heidelberg, Germany
| | - Laurin A. J. Mueller
- Institute for Bioinformatics and Translational Research, University for Health Sciences, Medical Informatics and Technology (UMIT), Hall in Tirol, Austria
| | - Maria Fälth
- Unit Cancer Genome Research, Division of Molecular Genetics, German Cancer Research Center and National Center of Tumor Diseases, Heidelberg, Germany
| | - Georg Schäfer
- Division of Experimental Urology, Department of Urology, Innsbruck Medical University, Innsbruck, Austria
| | - Christof Seifarth
- Division of Experimental Urology, Department of Urology, Innsbruck Medical University, Innsbruck, Austria
| | - Simone Ecker
- Institute for Bioinformatics and Translational Research, University for Health Sciences, Medical Informatics and Technology (UMIT), Hall in Tirol, Austria
| | - Irmgard Verdorfer
- Division of Human Genetics, Department of Medical Genetics, Molecular and Clinical Pharmacology, Innsbruck Medical University, Innsbruck, Austria
| | - Armin Graber
- Institute for Bioinformatics and Translational Research, University for Health Sciences, Medical Informatics and Technology (UMIT), Hall in Tirol, Austria
| | - Holger Sültmann
- Unit Cancer Genome Research, Division of Molecular Genetics, German Cancer Research Center and National Center of Tumor Diseases, Heidelberg, Germany
| | - Helmut Klocker
- Division of Experimental Urology, Department of Urology, Innsbruck Medical University, Innsbruck, Austria
- Oncotyrol, Center for Personalized Cancer Medicine GmbH, Innsbruck, Austria
- * E-mail: (PM); (HK)
| |
Collapse
|
32
|
Macrophage inhibitory cytokine-1 (MIC-1/GDF15) slows cancer development but increases metastases in TRAMP prostate cancer prone mice. PLoS One 2012; 7:e43833. [PMID: 22952779 PMCID: PMC3428289 DOI: 10.1371/journal.pone.0043833] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2012] [Accepted: 07/30/2012] [Indexed: 12/21/2022] Open
Abstract
Macrophage inhibitory cytokine-1 (MIC-1/GDF15), a divergent member of the TGF-β superfamily, is over-expressed by many common cancers including those of the prostate (PCa) and its expression is linked to cancer outcome. We have evaluated the effect of MIC-1/GDF15 overexpression on PCa development and spread in the TRAMP transgenic model of spontaneous prostate cancer. TRAMP mice were crossed with MIC-1/GDF15 overexpressing mice (MIC-1(fms)) to produce syngeneic TRAMP(fmsmic-1) mice. Survival rate, prostate tumor size, histopathological grades and extent of distant organ metastases were compared. Metastasis of TC1-T5, an androgen independent TRAMP cell line that lacks MIC-1/GDF15 expression, was compared by injecting intravenously into MIC-1(fms) and syngeneic C57BL/6 mice. Whilst TRAMP(fmsmic-1) survived on average 7.4 weeks longer, had significantly smaller genitourinary (GU) tumors and lower PCa histopathological grades than TRAMP mice, more of these mice developed distant organ metastases. Additionally, a higher number of TC1-T5 lung tumor colonies were observed in MIC-1(fms) mice than syngeneic WT C57BL/6 mice. Our studies strongly suggest that MIC-1/GDF15 has complex actions on tumor behavior: it limits local tumor growth but may with advancing disease, promote metastases. As MIC-1/GDF15 is induced by all cancer treatments and metastasis is the major cause of cancer treatment failure and cancer deaths, these results, if applicable to humans, may have a direct impact on patient care.
Collapse
|
33
|
Yip PY, Kench JG, Rasiah KK, Benito RP, Lee CS, Stricker PD, Henshall SM, Sutherland RL, Horvath LG. Low AZGP1 expression predicts for recurrence in margin-positive, localized prostate cancer. Prostate 2011; 71:1638-45. [PMID: 21432866 DOI: 10.1002/pros.21381] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Accepted: 02/17/2011] [Indexed: 11/06/2022]
Abstract
BACKGROUND Men with positive margins after radical prostatectomy (RP) for localized prostate cancer (PC) have a 40-50% biochemical relapse rate at 5 years. Adjuvant radiotherapy improves biochemical progression-free and overall survival in men with positive margins, but is associated with increased toxicity. There is an urgent need to identify new prognostic markers to define the group of patients who would benefit from multimodality therapy. METHODS Nuclear β-catenin, membranous secreted frizzled-related protein 4 (sFRP4), zinc-alpha 2-glycoprotein (AZGP1), and macrophage inhibitory cytokine-1 (MIC-1) have previously been identified as molecular markers of outcome in localized PC. From these published studies, we identified a subset of patients with positive margins. The aim of this study was to assess the association between these four molecular markers and outcome in men with margin-positive, localized PC. RESULTS We identified 186 men with positive margins from 330 men with localized PC; 53% had preoperative PSA >10 ng/ml, 72% extraprostatic extension (EPE), 24% seminal vesicles involvement (SVI), and 57% RP Gleason score ≥ 7. AZGP1 (P = 0.009), membranous sFRP4 (P = 0.03) and MIC-1 (P = 0.04) expression predicted for biochemical relapse on univariate analysis. Only absent/low AZGP1 expression (P = 0.01) was an independent predictor of recurrence in margin-positive, localized PC when modeled with preoperative PSA (P = 0.2), EPE (P = 0.2), SVI (P = 0.4), Gleason score ≥ 7 (P = 0.5) and adjuvant treatment (P = 0.4). Furthermore, there was an association between absent/low AZGP1 expression and clinical recurrence (P = 0.007). CONCLUSIONS AZGP1 is a potential molecular marker for biochemical relapse in men with margin-positive, localized PC. Routine assessment of this biomarker may lead to better selection of patients who will benefit from post-RP radiotherapy.
Collapse
Affiliation(s)
- Po Yee Yip
- Department of Medical Oncology, Sydney Cancer Centre, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Breit SN, Johnen H, Cook AD, Tsai VWW, Mohammad MG, Kuffner T, Zhang HP, Marquis CP, Jiang L, Lockwood G, Lee-Ng M, Husaini Y, Wu L, Hamilton JA, Brown DA. The TGF-β superfamily cytokine, MIC-1/GDF15: a pleotrophic cytokine with roles in inflammation, cancer and metabolism. Growth Factors 2011; 29:187-95. [PMID: 21831009 DOI: 10.3109/08977194.2011.607137] [Citation(s) in RCA: 186] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Macrophage inhibitory cytokine-1 (MIC-1/GDF15) is associated with cardiovascular disease, inflammation, body weight regulation and cancer. Its serum levels facilitate the diagnosis and prognosis of cancer and vascular disease. Furthermore, its serum levels are a powerful predictor of all-cause mortality, suggesting a fundamental role in biological processes associated with ageing. In cancer, the data available suggest that MIC-1/GDF15 is antitumorigenic, but this may not always be the case as disease progresses. Cancer promoting effects of MIC-1/GDF15 may be due, in part, to effects on antitumour immunity. This is suggested by the anti-inflammatory and immunosuppressive properties of MIC-1/GDF15 in animal models of atherosclerosis and rheumatoid arthritis. Furthermore, in late-stage cancer, large amounts of MIC-1/GDF15 in the circulation suppress appetite and mediate cancer anorexia/cachexia, which can be reversed by monoclonal antibodies in animals. Available data suggest MIC-1/GDF15 may be an important molecule mediating the interplay between cancer, obesity and chronic inflammation.
Collapse
Affiliation(s)
- Samuel N Breit
- St Vincent's Centre for Applied Medical Research, St Vincent's Hospital and University of New South Wales, Sydney, NSW 2010, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Gilaberte Y, Roca MJ, Garcia-Prats MD, Coscojuela C, Arbues MD, Vera-Alvarez JJ. Neuropeptide Y expression in cutaneous melanoma. J Am Acad Dermatol 2011; 66:e201-8. [PMID: 21620518 DOI: 10.1016/j.jaad.2011.02.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2010] [Revised: 12/25/2010] [Accepted: 02/15/2011] [Indexed: 01/26/2023]
Abstract
BACKGROUND Neuropeptide Y (NPY) is widely found in the nervous system and has a role in numerous physiologic processes. In addition, NPY receptors are expressed in neuroendocrine tumors, breast cancer, prostate cancer, kidney cancer, and some types of sarcomas. Different neuropeptides, particularly α-melanocyte-stimulating hormone (MSH), seem to play a role in the pathogenesis of melanoma. OBJECTIVE We sought to analyze the expression of NPY in cutaneous melanoma, its association with clinical and histologic features, and its correlation with α-MSH. METHODS This was an observational study of the immunohistochemical expression of NPY and α-MSH in tissue samples of cutaneous melanomas, different types of melanocytic nevi, and melanoma metastases diagnosed from 2004 to 2008 in San Jorge Hospital, Huesca, Spain. RESULTS A total of 184 lesions were studied: 49 primary cutaneous melanomas, 12 melanoma metastases (9 cutaneous and 3 lymphatic), and 123 melanocytic nevi. Immunostaining revealed that levels of NPY and α-MSH were significantly higher in melanomas than in melanocytic nevi (P < .001). Melanoma metastases were negative for both neuropeptides. Nodular melanomas showed the highest median percentage of NPY positive cells (75% [20-95]) followed by superficial spreading melanoma (25% [2-92]), whereas lentigo maligna were negative (0% [0-0]). Significant, direct associations between NPY expression and vertical growth (P = .0141) and presence of metastasis (P = .0196) were observed. NPY and α-MSH were positively correlated in cutaneous melanoma (0.49, P < .001). LIMITATIONS The sample size of melanomas was not very large. CONCLUSION Our study demonstrates that NPY is significantly expressed in melanomas, especially the nodular type, being associated with invasiveness independently of proliferative markers such as thickness, ulceration, and mitotic index.
Collapse
|
36
|
Mimeault M, Batra SK. Divergent molecular mechanisms underlying the pleiotropic functions of macrophage inhibitory cytokine-1 in cancer. J Cell Physiol 2010; 224:626-35. [PMID: 20578239 DOI: 10.1002/jcp.22196] [Citation(s) in RCA: 175] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Multifunctional macrophage inhibitory cytokine-1, MIC-1, is a member of the transforming growth factor-beta (TGF-beta) superfamily that plays key roles in the prenatal development and regulation of the cellular responses to stress signals and inflammation and tissue repair after acute injuries in adult life. The stringent control of the MIC-1 expression, secretion, and functions involves complex regulatory mechanisms and the interplay of other growth factor signaling networks that control the cell behavior. The deregulation of MIC-1 expression and signaling pathways has been associated with diverse human diseases and cancer progression. The MIC-1 expression levels substantially increase in cancer cells, serum, and/or cerebrospinal fluid during the progression of diverse human aggressive cancers, such as intracranial brain tumors, melanoma, and lung, gastrointestinal, pancreatic, colorectal, prostate, and breast epithelial cancers. Of clinical interest, an enhanced MIC-1 expression has been positively correlated with poor prognosis and patient survival. Secreted MIC-1 cytokine, like the TGF-beta prototypic member of the superfamily, may provide pleiotropic roles in the early and late stages of carcinogenesis. In particular, MIC-1 may contribute to the proliferation, migration, invasion, metastases, and treatment resistance of cancer cells as well as tumor-induced anorexia and weight loss in the late stages of cancer. Thus, secreted MIC-1 cytokine constitutes a new potential biomarker and therapeutic target of great clinical interest for the development of novel diagnostic and prognostic methods and/or cancer treatment against numerous metastatic, recurrent, and lethal cancers.
Collapse
Affiliation(s)
- Murielle Mimeault
- Department of Biochemistry and Molecular Biology, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska 68198-5870, USA
| | | |
Collapse
|
37
|
Zhao L, Lee BY, Brown DA, Molloy MP, Marx GM, Pavlakis N, Boyer MJ, Stockler MR, Kaplan W, Breit SN, Sutherland RL, Henshall SM, Horvath LG. Identification of candidate biomarkers of therapeutic response to docetaxel by proteomic profiling. Cancer Res 2009; 69:7696-703. [PMID: 19773444 DOI: 10.1158/0008-5472.can-08-4901] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Docetaxel chemotherapy improves symptoms and survival in men with metastatic hormone-refractory prostate cancer (HRPC). However, approximately 50% of patients do not respond to Docetaxel and are exposed to significant toxicity without direct benefit. This study aimed to identify novel therapeutic targets and predictive biomarkers of Docetaxel resistance in HRPC. We used iTRAQ-mass spectrometry analysis to identify proteins associated with the development of Docetaxel resistance using Docetaxel-sensitive PC3 cells and Docetaxel-resistant PC3-Rx cells developed by Docetaxel dose escalation. Functional validation experiments were performed using recombinant protein treatment and siRNA knockdown experiments. Serum/plasma levels of the targets in patient samples were measured by ELISA. The IC(50) for Docetaxel in the PC3-Rx cells was 13-fold greater than the parent PC-3 cell line (P = 0.004). Protein profiling identified MIC-1 and AGR2 as respectively up-regulated and down-regulated in Docetaxel-resistant cells. PC-3 cells treated with recombinant MIC-1 also became resistant to Docetaxel (P = 0.03). Conversely, treating PC3-Rx cells with MIC-1 siRNA restored sensitivity to Docetaxel (P = 0.02). Knockdown of AGR2 expression in PC3 cells resulted in Docetaxel resistance (P = 0.007). Furthermore, increased serum/plasma levels of MIC-1 after cycle one of chemotherapy were associated with progression of the cancer (P = 0.006) and shorter survival after treatment (P = 0.002). These results suggest that both AGR2 and MIC-1 play a role in Docetaxel resistance in HRPC. In addition, an increase in serum/plasma MIC-1 level after cycle one of Docetaxel may be an indication to abandon further treatment. Further investigation of MIC-1 as a biomarker and therapeutic target for Docetaxel resistance in HRPC is warranted.
Collapse
Affiliation(s)
- Liangli Zhao
- Cancer Research Program, Garvan Institute of Medical Research, Sydney, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Proutski I, Stevenson L, Allen WL, McCulla A, Boyer J, McLean EG, Longley DB, Johnston PG. Prostate-derived factor--a novel inhibitor of drug-induced cell death in colon cancer cells. Mol Cancer Ther 2009; 8:2566-74. [PMID: 19723892 DOI: 10.1158/1535-7163.mct-09-0158] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We investigated the role of the divergent transforming growth factor-beta superfamily member, prostate-derived factor (PDF), in regulating response to chemotherapies used in the treatment of colorectal cancer. A clear p53-dependent expression pattern of PDF was shown in a panel of colorectal cancer cell lines following acute exposure to oxaliplatin, 5-fluorouracil, and SN38. PDF gene silencing before chemotherapy treatment significantly sensitized cells expressing wild-type p53, but not p53-null or p53-mutant cells, to drug-induced apoptosis. Similarly, knockdown of PDF expression sensitized HCT116 drug-resistant daughter cell lines to their respective chemotherapies. Inducible PDF expression and treatment with recombinant PDF both significantly attenuated drug-induced apoptosis. Further analysis revealed that PDF activated the Akt but not the extracellular signal-regulated kinase 1/2 signaling pathway. Furthermore, cotreatment with the phosphatidylinositol 3-kinase inhibitor wortmannin abrogated PDF-mediated resistance to chemotherapy-induced apoptosis. Together, these data suggest that PDF may be a novel inhibitor of drug-induced cell death in colorectal cancer cells and that the mature secreted form of the protein activates the phosphatidylinositol 3-kinase/Akt pathway as an acute mechanism of chemoresistance.
Collapse
Affiliation(s)
- Irina Proutski
- Department of Oncology, Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, Northern Ireland, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
39
|
A role for GATA-2 in transition to an aggressive phenotype in prostate cancer through modulation of key androgen-regulated genes. Oncogene 2009; 28:3847-56. [PMID: 19684615 DOI: 10.1038/onc.2009.243] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
GATA-2, a member of the GATA family of transcription factors, is involved in androgen receptor (AR) signaling, however, little is known regarding its role in prostate cancer. Here, we report that GATA-2 is expressed in a substantial proportion of prostate cancers and that high expression of GATA-2 is associated with biochemical recurrence and distant metastatic progression in a validation set of 203 cancers. In vitro data show that GATA-2 is directly recruited to the promoter region of the AR upon androgen stimulation of LNCaP prostate cancer cells with 5alpha-dihydroxytestosterone (DHT) for 24 h. Ectopic GATA-2 expression causes the induction of AR transcript levels under androgen-depleted conditions (P<0.05). The expression of the AR target gene, AZGP1, is induced upon androgen stimulation and this effect is repressed by GATA-2. In contrast, GATA-2 significantly increases transcript levels of KLK2, which increases further in a time-dependent manner on DHT treatment and in the presence of GATA-2. These results indicate that upregulation of GATA-2 may contribute to the progression to aggressive prostate cancer through modulation of expression of AR and key androgen-regulated genes, one of which, AZGP1, is associated with the progression to metastatic disease.
Collapse
|
40
|
McMahon CJ, Bloch BN, Lenkinski RE, Rofsky NM. Dynamic contrast-enhanced MR imaging in the evaluation of patients with prostate cancer. Magn Reson Imaging Clin N Am 2009; 17:363-83. [PMID: 19406364 DOI: 10.1016/j.mric.2009.01.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Prostate cancer is a common tumor among men, with increasing diagnosis at an earlier stage and a lower volume of disease because of screening with prostate-specific antigen (PSA). The need for imaging of the prostate stems from a desire to optimize treatment strategy on a patient and tumor-specific level. The major goals of prostate imaging are (1) staging of known cancer, (2) determination of tumor aggressiveness, (3) diagnosis of cancer in patients who have elevated PSA but a negative biopsy, (4) treatment planning, and (5) the evaluation of therapy response. This article concentrates on the role of dynamic contrast-enhanced MR imaging in the evaluation of patients who have prostate cancer and how it might be used to help achieve the above goals. Various dynamic contrast enhancement approaches (quantitative/semiquantitative/qualitative, high temporal versus high spatial resolution) are summarized with reference to the relevant strengths and compromises of each approach.
Collapse
Affiliation(s)
- Colm J McMahon
- Department of Radiology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02215, USA.
| | | | | | | |
Collapse
|
41
|
Kelly JA, Lucia MS, Lambert JR. p53 controls prostate-derived factor/macrophage inhibitory cytokine/NSAID-activated gene expression in response to cell density, DNA damage and hypoxia through diverse mechanisms. Cancer Lett 2008; 277:38-47. [PMID: 19100681 DOI: 10.1016/j.canlet.2008.11.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2008] [Revised: 11/07/2008] [Accepted: 11/11/2008] [Indexed: 01/02/2023]
Abstract
The p53 tumor suppressor modulates cellular response to stress through both transcriptional and post-transcriptional mechanisms. Elucidation of the downstream targets of p53 following cell stress will aid in our understanding of the pathways involved in cellular adaptation to stressful stimuli. Here, we demonstrate that the TGF-beta superfamily member, and putative tumor suppressor, prostate-derived factor (PDF)/NSAID-activated gene (NAG)-1/macrophage inhibitory cytokine (MIC)-1 is induced in LNCaP human prostate cancer cells following treatment with the DNA-damaging agent, doxorubicin, culture under hypoxic conditions and by the hypoxia mimetic, cobalt chloride. Additionally, PDF expression was induced by increasing cell density. Expression of dominant negative p53 in LNCaP cells blocked induction of PDF mRNA and protein demonstrating the requirement for functional p53 in PDF induction by these stimuli. DNA damage and hypoxia resulted in increased p53 protein accumulation indicating that PDF expression may be controlled by cellular levels of p53. We also show the requirement for de novo protein synthesis in PDF induction by hypoxia and DNA damage. Increased PDF mRNA stability in response to hypoxia and cobalt chloride, but not doxorubicin, indicates that p53-dependent induction of PDF expression occurs via diverse mechanisms. Thus, PDF may represent a novel target of p53 in response to cell stress.
Collapse
Affiliation(s)
- Julie A Kelly
- Department of Pathology, University of Colorado-Denver, 12801 E 17th Avenue, Aurora, CO 80045, USA
| | | | | |
Collapse
|
42
|
An illustration of the potential for mapping MRI/MRS parameters with genetic over-expression profiles in human prostate cancer. MAGNETIC RESONANCE MATERIALS IN PHYSICS BIOLOGY AND MEDICINE 2008; 21:411-21. [PMID: 18752015 DOI: 10.1007/s10334-008-0133-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 01/10/2008] [Revised: 07/24/2008] [Accepted: 07/25/2008] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Magnetic resonance imaging (MRI) and MR spectroscopy can probe a variety of physiological (e.g. blood vessel permeability) and metabolic characteristics of prostate cancer. However, little is known about the changes in gene expression that underlie the spectral and imaging features observed in prostate cancer. Tumor induced changes in vascular permeability and angiogenesis are thought to contribute to patterns of dynamic contrast enhanced (DCE) MRI images of prostate cancer even though the genetic basis of tumor vasculogenesis is complex and the specific mechanisms underlying these DCEMRI features have not yet been determined. MATERIALS AND METHODS In order to identify the changes in gene expression that correspond to MRS and DCEMRI patterns in human prostate cancers, we have utilized tissue print micropeel techniques to generate "whole mount" molecular maps of radical prostatectomy specimens that correspond to pre-surgical MRI/MRS studies. These molecular maps include RNA expression profiles from both Affymetrix GeneChip microarrays and quantitative reverse transcriptase PCR (qrt-PCR) analysis, as well as immunohistochemical studies. RESULTS Using these methods on patients with prostate cancer, we found robust over-expression of choline kinase a in the majority of primary tumors. We also observed overexpression of neuropeptide Y (NPY), a newly identified angiogenic factor, in a subset of prostate cancers, visualized on DCEMRI. CONCLUSION These studies set the stage for establishing MRI/MRS parameters as validated biomarkers for human prostate cancer.
Collapse
|
43
|
Eisermann K, Tandon S, Bazarov A, Brett A, Fraizer G, Piontkivska H. Evolutionary conservation of zinc finger transcription factor binding sites in promoters of genes co-expressed with WT1 in prostate cancer. BMC Genomics 2008; 9:337. [PMID: 18631392 PMCID: PMC2515153 DOI: 10.1186/1471-2164-9-337] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2008] [Accepted: 07/16/2008] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Gene expression analyses have led to a better understanding of growth control of prostate cancer cells. We and others have identified the presence of several zinc finger transcription factors in the neoplastic prostate, suggesting a potential role for these genes in the regulation of the prostate cancer transcriptome. One of the transcription factors (TFs) identified in the prostate cancer epithelial cells was the Wilms tumor gene (WT1). To rapidly identify coordinately expressed prostate cancer growth control genes that may be regulated by WT1, we used an in silico approach. RESULTS Evolutionary conserved transcription factor binding sites (TFBS) recognized by WT1, EGR1, SP1, SP2, AP2 and GATA1 were identified in the promoters of 24 differentially expressed prostate cancer genes from eight mammalian species. To test the relationship between sequence conservation and function, chromatin of LNCaP prostate cancer and kidney 293 cells were tested for TF binding using chromatin immunoprecipitation (ChIP). Multiple putative TFBS in gene promoters of placental mammals were found to be shared with those in human gene promoters and some were conserved between genomes that diverged about 170 million years ago (i.e., primates and marsupials), therefore implicating these sites as candidate binding sites. Among those genes coordinately expressed with WT1 was the kallikrein-related peptidase 3 (KLK3) gene commonly known as the prostate specific antigen (PSA) gene. This analysis located several potential WT1 TFBS in the PSA gene promoter and led to the rapid identification of a novel putative binding site confirmed in vivo by ChIP. Conversely for two prostate growth control genes, androgen receptor (AR) and vascular endothelial growth factor (VEGF), known to be transcriptionally regulated by WT1, regulatory sequence conservation was observed and TF binding in vivo was confirmed by ChIP. CONCLUSION Overall, this targeted approach rapidly identified important candidate WT1-binding elements in genes coordinately expressed with WT1 in prostate cancer cells, thus enabling a more focused functional analysis of the most likely target genes in prostate cancer progression. Identifying these genes will help to better understand how gene regulation is altered in these tumor cells.
Collapse
Affiliation(s)
- Kurtis Eisermann
- School of Biomedical Sciences, Kent State University, Kent, Ohio, USA.
| | | | | | | | | | | |
Collapse
|
44
|
Liu AJ, Furusato B, Ravindranath L, Chen YM, Srikantan V, McLeod DG, Petrovics G, Srivastava S. Quantitative analysis of a panel of gene expression in prostate cancer--with emphasis on NPY expression analysis. J Zhejiang Univ Sci B 2008; 8:853-9. [PMID: 18257117 DOI: 10.1631/jzus.2007.b0853] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE To investigate molecular alterations associating with prostate carcinoma progression and potentially provide information toward more accurate prognosis/diagnosis. METHODS A set of laser captured microdissected (LCM) specimens from 300 prostate cancer (PCa) patients undergoing radical prostatectomy (RP) were defined. Ten patients representing "aggressive" PCa, and 10 representing "non-aggressive" PCa were selected based on prostate-specific antigen (PSA) recurrence, Gleason score, pathological stage and tumor cell differentiation, with matched patient age and race between the two groups. Normal and neoplastic prostate epithelial cells were collected with LCM from frozen tissue slides obtained from the RP specimens. The expressions of a panel of genes, including NPY, PTEN, AR, AMACR, DD3, and GSTP1, were measured by quantitative real-time RT-PCR (TaqMan), and correlation was analyzed with clinicopathological features. RESULTS The expressions of AMACR and DD3 were consistently up-regulated in cancer cells compared to benign prostate epithelial cells in all PCa patients, whereas GSTP1 expression was down regulated in each patient. NPY, PTEN and AR exhibited a striking difference in their expression patterns between aggressive and non-aggressive PCas (P=0.0203, 0.0284, and 0.0378, respectively, Wilcoxon rank sum test). The lower expression of NPY showed association with "aggressive" PCas based on a larger PCa patient cohort analysis (P=0.0037, univariate generalized linear model (GLM) analysis). CONCLUSION Despite widely noted heterogeneous nature of PCa, gene expression alterations of AMACR, DD3, and GSTP1 in LCM-derived PCa epithelial cells suggest for common underlying mechanisms in the initiation of PCa. Lower NPY expression level is significantly associated with more aggressive clinical behavior of PCa; PTEN and AR may have potential in defining PCa with aggressive clinical behavior. Studies along these lines have potential to define PCa-associated gene expression alterations and likely co-regulation of genes/pathways critical in the biology of PCa onset/progression.
Collapse
Affiliation(s)
- Ai-jun Liu
- Department of Pathology, Chinese PLA General Hospital, Beijing 100853, China.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Chattopadhyay I, Kapur S, Purkayastha J, Phukan R, Kataki A, Mahanta J, Saxena S. Gene expression profile of esophageal cancer in North East India by cDNA microarray analysis. World J Gastroenterol 2007; 13:1438-44. [PMID: 17457978 PMCID: PMC4146931 DOI: 10.3748/wjg.v13.i9.1438] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2006] [Revised: 12/29/2006] [Accepted: 01/26/2007] [Indexed: 02/07/2023] Open
Abstract
AIM To identify alterations in genes and molecular functional pathways in esophageal cancer in a high incidence region of India where there is a widespread use of tobacco and betel quid with fermented areca nuts. METHODS Total RNA was isolated from tumor and matched normal tissue of 16 patients with esophageal squamous cell carcinoma. Pooled tumor tissue RNA was labeled with Cy3-dUTP and pooled normal tissue RNA was labeled with Cy5-dUTP by direct labeling method. The labeled probes were hybridized with human 10K cDNA chip and expression profiles were analyzed by Genespring GX V 7.3 (Silicon Genetics). RESULTS Nine hundred twenty three genes were differentially expressed. Of these, 611 genes were upregulated and 312 genes were downregulated. Using stringent criteria (P < or = 0.05 and > or = 1.5 fold change), 127 differentially expressed genes (87 upregulated and 40 downregulated) were identified in tumor tissue. On the basis of Gene Ontology, four different molecular functional pathways (MAPK pathway, G-protein coupled receptor family, ion transport activity, and serine or threonine kinase activity) were most significantly upregulated and six different molecular functional pathways (structural constituent of ribosome, endopeptidase inhibitor activity, structural constituent of cytoskeleton, antioxidant activity, acyl group transferase activity, eukaryotic translation elongation factor activity) were most significantly downregulated. CONCLUSION Several genes that showed alterations in our study have also been reported from a high incidence area of esophageal cancer in China. This indicates that molecular profiles of esophageal cancer in these two different geographic locations are highly consistent.
Collapse
Affiliation(s)
- Indranil Chattopadhyay
- Institute of Pathology, Indian Council of Medical Research, Safdarjang Hospital Campus, Post Box No. 4909, New Delhi 110029, India
| | | | | | | | | | | | | |
Collapse
|
46
|
Ruscica M, Dozio E, Motta M, Magni P. Role of neuropeptide Y and its receptors in the progression of endocrine-related cancer. Peptides 2007; 28:426-34. [PMID: 17204352 DOI: 10.1016/j.peptides.2006.08.045] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2006] [Accepted: 08/20/2006] [Indexed: 10/23/2022]
Abstract
The neuropeptide Y (NPY) family of peptides, in addition to its many physiological actions, has also been involved in the modulation of tumor progression, with specific reference to endocrine-related cancers such as neuroendocrine tumors, breast and prostate cancers. These have been found either to express NPY receptors, or to secrete NPY-related peptides, or both. The study of the role of the NPY family of peptides in the biology of endocrine-related tumors, specifically concerning cell proliferation, angiogenesis, invasion and metastatization, may help to clarify some aspects of tumor pathophysiology, as well as to indicate novel diagnostic markers and therapeutical approaches.
Collapse
Affiliation(s)
- Massimiliano Ruscica
- Center for Endocrinological Oncology, Istituto di Endocrinologia, Università degli Studi di Milano, via Balzaretti 9, Milan 20133, Italy
| | | | | | | |
Collapse
|
47
|
Hayes VM, Severi G, Southey MC, Padilla EJD, English DR, Hopper JL, Giles GG, Sutherland RL. Macrophage inhibitory cytokine-1 H6D polymorphism, prostate cancer risk, and survival. Cancer Epidemiol Biomarkers Prev 2006; 15:1223-5. [PMID: 16775185 DOI: 10.1158/1055-9965.epi-06-0063] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Macrophage inhibitory cytokine-1 (MIC-1), a member of the transforming growth factor-beta superfamily, is important in regulating inflammation. Inflammation of the prostate has been suggested to favor tumor development. A recent study (JNCI 2004, 96:1248-1254) found marginal evidence of an association between the presence of the mature MIC-1 protein nonsynonymous polymorphism H6D C-to-G (rs1058587) with reduced prostate cancer risk [odds ratio, 0.83; 95% confidence interval (95% CI), 0.69-0.99]. We tested this in a population-based study of 819 cases and 731 controls from Australia and found a similar, yet not significant, odds ratio of 0.85 (95% CI, 0.7-1.04; P = 0.11). We also tested the potential association between the H6D variant and disease-specific survival in 640 cases followed-up for an average of 8.2 years. We found that cases carrying the H6D G allele had an increased risk of death from prostate cancer than cases carrying two copies of the C allele (hazard ratio, 1.72; 95% CI, 1.06-2.78; P = 0.03). Our data suggest that the H6D variant in MIC-1 might play a role in prostate cancer, but it is difficult to explain how a variant can be associated with lower risk of developing prostate cancer but more aggressive growth if cancer develops.
Collapse
Affiliation(s)
- Vanessa M Hayes
- Cancer Research Program, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, New South Wales 2010, Australia.
| | | | | | | | | | | | | | | |
Collapse
|