1
|
Ravindranathan S, Passang T, Li JM, Wang S, Dhamsania R, Ware MB, Zaidi MY, Zhu J, Cardenas M, Liu Y, Gumber S, Robinson B, Sen-Majumdar A, Zhang H, Chandrakasan S, Kissick H, Frey AB, Thomas SN, El-Rayes BF, Lesinski GB, Waller EK. Targeting vasoactive intestinal peptide-mediated signaling enhances response to immune checkpoint therapy in pancreatic ductal adenocarcinoma. Nat Commun 2022; 13:6418. [PMID: 36302761 PMCID: PMC9613684 DOI: 10.1038/s41467-022-34242-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 10/18/2022] [Indexed: 12/25/2022] Open
Abstract
A paucity of effector T cells within tumors renders pancreatic ductal adenocarcinoma (PDAC) resistant to immune checkpoint therapies. While several under-development approaches target immune-suppressive cells in the tumor microenvironment, there is less focus on improving T cell function. Here we show that inhibiting vasoactive intestinal peptide receptor (VIP-R) signaling enhances anti-tumor immunity in murine PDAC models. In silico data mining and immunohistochemistry analysis of primary tumors indicate overexpression of the neuropeptide vasoactive intestinal peptide (VIP) in human PDAC tumors. Elevated VIP levels are also present in PDAC patient plasma and supernatants of cultured PDAC cells. Furthermore, T cells up-regulate VIP receptors after activation, identifying the VIP signaling pathway as a potential target to enhance T cell function. In mouse PDAC models, VIP-R antagonist peptides synergize with anti-PD-1 antibody treatment in improving T cell recruitment into the tumors, activation of tumor-antigen-specific T cells, and inhibition of T cell exhaustion. In contrast to the limited single-agent activity of anti-PD1 antibodies or VIP-R antagonist peptides, combining both therapies eliminate tumors in up to 40% of animals. Furthermore, tumor-free mice resist tumor re-challenge, indicating anti-cancer immunological memory generation. VIP-R signaling thus represents a tumor-protective immune-modulatory pathway that is targetable in PDAC.
Collapse
Affiliation(s)
- Sruthi Ravindranathan
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA.
- Winship Cancer Institute, Emory University, Atlanta, GA, USA.
| | - Tenzin Passang
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Jian-Ming Li
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Shuhua Wang
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Rohan Dhamsania
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Michael Brandon Ware
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Mohammad Y Zaidi
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Jingru Zhu
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Maria Cardenas
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Yuan Liu
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Sanjeev Gumber
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Brian Robinson
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Hanwen Zhang
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Haydn Kissick
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
- Emory Vaccine Centre, Emory University, Atlanta, GA, USA
| | | | - Susan N Thomas
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Bassel F El-Rayes
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Gregory B Lesinski
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Edmund K Waller
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA.
- Winship Cancer Institute, Emory University, Atlanta, GA, USA.
| |
Collapse
|
2
|
Cytoprotective effect of neuropeptides on cancer stem cells: vasoactive intestinal peptide-induced antiapoptotic signaling. Cell Death Dis 2017; 8:e2844. [PMID: 28569785 PMCID: PMC5520887 DOI: 10.1038/cddis.2017.226] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 04/02/2017] [Accepted: 04/03/2017] [Indexed: 02/07/2023]
Abstract
Cancer stem cells (CSCs) are increasingly considered to be responsible for tumor initiation, metastasis and drug resistance. The drug resistance mechanisms activated in CSCs have not been thoroughly investigated. Although neuropeptides such as vasoactive intestinal peptide (VIP) can promote tumor growth and activate antiapoptotic signaling in differentiated cancer cells, it is not known whether they can activate antiapoptotic mechanisms in CSCs. The objectives of this study are to unravel the cytoprotective effects of neuropeptides and identify antiapoptotic mechanisms activated by neuropeptides in response to anticancer drug treatment in CSCs. We enriched and purified CSCs (CD44+/high/CD24-/low or CD133+ population) from breast and prostate cancer cell lines, and demonstrated their stemness phenotype. Of the several neuropeptides tested, only VIP could protect CSCs from drug-induced apoptosis. A functional correlation was found between drug-induced apoptosis and dephosphorylation of proapoptotic Bcl2 family protein BAD. Similarly, VIP-induced cytoprotection correlated with BAD phosphorylation at Ser112 in CSCs. Using pharmacological inhibitors and dominant-negative proteins, we showed that VIP-induced cytoprotection and BAD phosphorylation are mediated via both Ras/MAPK and PKA pathways in CSCs of prostate cancer LNCaP and C4-2 cells, but only PKA signaling was involved in CSCs of DUVIPR (DU145 prostate cancer cells ectopically expressing VIP receptor) and breast cancer MCF7 cells. As each of these pathways partially control BAD phosphorylation at Ser112, both have to be inhibited to block the cytoprotective effects of VIP. Furthermore, VIP is unable to protect CSCs that express phosphorylation-deficient mutant-BAD, suggesting that BAD phosphorylation is essential. Thus, antiapoptotic signaling by VIP could be one of the drug resistance mechanisms by which CSCs escape from anticancer therapies. Our findings suggest the potential usefulness of VIP receptor inhibition to eliminate CSCs, and that targeting BAD might be an attractive strategy for development of novel therapeutics.
Collapse
|
3
|
Moscona JC, Peters MN, Schally AV, Srivastav S, Delafontaine P, Irimpen A. The effects of a growth hormone-releasing hormone antagonist and a gastrin-releasing peptide antagonist on intimal hyperplasia of the carotid artery after balloon injury in a diabetic rat model☆. Artery Res 2017. [DOI: 10.1016/j.artres.2017.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
4
|
Valdehita A, Carmena MJ, Bajo AM, Prieto JC. RNA interference-directed silencing of VPAC1 receptor inhibits VIP effects on both EGFR and HER2 transactivation and VEGF secretion in human breast cancer cells. Mol Cell Endocrinol 2012; 348:241-6. [PMID: 21896307 DOI: 10.1016/j.mce.2011.08.031] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Revised: 07/28/2011] [Accepted: 08/23/2011] [Indexed: 12/31/2022]
Abstract
We used small-interference RNA (siRNA) to explore the mechanisms of some vasoactive intestinal peptide (VIP) actions on human breast cancer cells. Transfection of estrogen-dependent (T47D) and estrogen-independent (MDA-MB-468) breast cancer cells with VPAC(1)-receptor siRNA completely abolished VIP stimulatory effect on secretion of the main angiogenic factor, vascular endothelial growth factor (VEGF), and transactivation of epidermal growth factor receptor (EGFR or HER1) and HER2, two members of HER family of tyrosine-kinase receptors. The silencing procedure suggested the involvement of EGFR and HER2 transactivation in VIP-stimulated VEGF secretion. It was further supported by blocking tyrosine kinase activity by the selective HER inhibitors AG-1478 (EGFR) and AG-825 (HER2). Results give value to the specific signaling of VIP through VPAC(1) receptor in human breast cancer cells and support the potential use of VPAC(1)-receptor antagonists in combined targeted therapies for breast cancer. Molecular therapies involving RNA interference of VPAC(1)-receptor expression could also be considered.
Collapse
Affiliation(s)
- Ana Valdehita
- Department of Biochemistry and Molecular Biology, University of Alcalá, Alcalá de Henares 28871, Spain
| | | | | | | |
Collapse
|
5
|
Veljkovic M, Dopsaj V, Dopsaj M, Branch DR, Veljkovic N, Sakarellos-Daitsiotis MM, Veljkovic V, Glisic S, Colombatti A. Physical activity and natural anti-VIP antibodies: potential role in breast and prostate cancer therapy. PLoS One 2011; 6:e28304. [PMID: 22140573 PMCID: PMC3227651 DOI: 10.1371/journal.pone.0028304] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Accepted: 11/05/2011] [Indexed: 12/14/2022] Open
Abstract
Background There is convincing evidence from numerous clinical and epidemiological studies that physical activity can reduce the risk for breast and prostate cancer. The biological mechanisms underlying this phenomenon remain elusive. Herein we suggest a role for naturally produced antibodies reactive with the vasoactive intestinal peptide (VIP) in the suppression of breast and prostate cancer, which we believe could offer a possible molecular mechanism underlying control of these cancers by physical exercise. Methodology and Results We found that sera from individuals having breast and prostate cancers have decreased titers of VIP natural antibodies as demonstrated by a lower reactivity against peptide NTM1, having similar informational and structural properties as VIP. In contrast, sera collected from elite athletes, exhibited titers of natural NTM1-reactive antibodies that are significantly increased, suggesting that physical activity boosts production of these antibodies. Significance Presented results suggest that physical exercise stimulates production of natural anti-VIP antibodies and likely results in suppression of VIP. This, in turn, may play a protective role against breast and prostate cancers. Physical exercise should be further investigated as a potential tool in the treatment of these diseases.
Collapse
Affiliation(s)
| | - Violeta Dopsaj
- Institute of Medical Biochemistry, Clinical Centre of Serbia, Belgrade, Serbia
| | - Milivoj Dopsaj
- Faculty of Sport and Physical Education, University of Belgrade, Belgrade, Serbia
| | | | - Nevena Veljkovic
- Center for Multidisciplinary Research, Institute of Nuclear Sciences VINCA, Belgrade, Serbia
| | | | - Veljko Veljkovic
- Center for Multidisciplinary Research, Institute of Nuclear Sciences VINCA, Belgrade, Serbia
- * E-mail:
| | - Sanja Glisic
- Center for Multidisciplinary Research, Institute of Nuclear Sciences VINCA, Belgrade, Serbia
| | - Alfonso Colombatti
- Divisione di Oncologia Sperimentale, Centro di Riferimento Oncologico CRO-IRCCS, Aviano, Italy
| |
Collapse
|
6
|
GHRH antagonists reduce the invasive and metastatic potential of human cancer cell lines in vitro. Cancer Lett 2010; 293:31-40. [PMID: 20064686 DOI: 10.1016/j.canlet.2009.12.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2009] [Revised: 12/13/2009] [Accepted: 12/17/2009] [Indexed: 12/21/2022]
Abstract
We investigated the effect of a GHRH antagonist, MIA-602on the metastatic cascade in vitro of three human cancers, DBTRG-05 glioblastoma, MDA-MB-468 estrogen-independent breast, and ES-2 clear cell ovarian cancer. GHRH receptors and their main splice variant, SV1 were detected on all three cell lines. After treatment with MIA-602, the cell viability decreased significantly, significant inhibition of cell invasion was observed and the release of MMPs was significantly decreased. The attachment of cancer cells to fibronectin and matrigel was severely hindered. Wound-healing experiments demonstrated a reduced cellular motility in all three cell lines. The upregulation of caveolin-1 and E-cadherin,and thepowerful downregulation of NF-kappaB and beta-catenin was detected. Our study suggests that the clinical application of highly potent GHRH antagonists in cancer therapy would be desirable since they inhibit proliferation and metastasis development as well.
Collapse
|
7
|
Valdehita A, Bajo AM, Schally AV, Varga JL, Carmena MJ, Prieto JC. Vasoactive intestinal peptide (VIP) induces transactivation of EGFR and HER2 in human breast cancer cells. Mol Cell Endocrinol 2009; 302:41-8. [PMID: 19101605 DOI: 10.1016/j.mce.2008.11.024] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2008] [Revised: 11/20/2008] [Accepted: 11/21/2008] [Indexed: 01/22/2023]
Abstract
We analyzed the cross-talk between receptors for vasoactive intestinal peptide (VIP) and the human epidermal growth factor family of tyrosine kinase receptors (HER) in oestrogen-dependent (T47D) and oestrogen-independent (MDA-MB-468) human breast cancer cells. VIP treatment slowly increased the expression levels of EGFR but it rapidly augmented phosphorylation of EGFR and HER2 in both cell lines. This pattern of HERs transactivation was blocked by the specific VIP antagonist JV-1-53, supporting the direct involvement of VIP receptors in formation of P-EGFR and P-HER2. VIP-induced transactivation was also abolished by H89 (protein kinase A inhibitor), PP2 (Src inhibitor) or TAPI-1 (inhibitor of matrix metalloproteases), following a differential pattern. These results shed a new light on the specific signalling pathways involved in EGFR/HER2 transactivation by VPAC receptors and suggest the potential usefulness of VIP receptor antagonists together with current antibodies against EGFR/HER2 and/or tyrosine kinase inhibitors for breast cancer therapy.
Collapse
Affiliation(s)
- Ana Valdehita
- Molecular Neuroendocrinology Unit, Department of Biochemistry and Molecular Biology, Alcalá University, 28871 Alcalá de Henares, Spain
| | | | | | | | | | | |
Collapse
|
8
|
Pan CQ, Hamren S, Roczniak S, Tom I, DeRome M. Generation of PEGylated VPAC1-selective antagonists that inhibit proliferation of a lung cancer cell line. Peptides 2008; 29:479-86. [PMID: 17942192 DOI: 10.1016/j.peptides.2007.09.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2007] [Revised: 09/05/2007] [Accepted: 09/06/2007] [Indexed: 11/18/2022]
Abstract
Vasoactive intestinal peptide (VIP) binds to two receptors, VPAC1 and VPAC2. Non-selective VIP antagonists have been shown to inhibit human cancer cell proliferation and reduce tumor growth in mice. Many human cancers over-express VPAC1 but not VPAC2. We show that VPAC1-selective antagonists can inhibit human cancer cell proliferation and identify five positions in the VPAC1-selective antagonist PG 97-269 that may be responsible for VPAC1 selectivity. Position 16 appears to be particularly critical for selectivity, as demonstrated in the replacement of Arg16 of PG 97-269 with the native VIP amino acid; this single change results in greatly reduced VPAC1 binding and selectivity. Finally, we show that site-specific conjugation with a 22kDa polyethylene glycol (PEG) at the C-terminus of VPAC1-selective antagonists further improves VPAC1-selective binding and has minimal effect on antagonistic activity. Our studies have further solidified VPAC1 as a cancer target and offer the possibility of generating highly potent VPAC1-selective antagonists with minimal number of mutations to reduce the risk of immunogenicity and potentially prolonged duration of action to allow more efficient treatment regimen.
Collapse
Affiliation(s)
- Clark Q Pan
- Department of Cancer Biology, Bayer HealthCare Pharmaceuticals, West Haven, CT 06516, USA
| | | | | | | | | |
Collapse
|
9
|
Collado B, Carmena MJ, Clemente C, Prieto JC, Bajo AM. Vasoactive intestinal peptide enhances growth and angiogenesis of human experimental prostate cancer in a xenograft model. Peptides 2007; 28:1896-901. [PMID: 17544169 DOI: 10.1016/j.peptides.2007.04.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2007] [Revised: 04/13/2007] [Accepted: 04/24/2007] [Indexed: 10/23/2022]
Abstract
We show that vasoactive intestinal peptide (VIP) exerts trophic and proangiogenic activities in experimental prostate cancer in vivo. Nude mice were subcutaneously injected with Matrigel impregnated with LNCaP prostate cancer cells. Cell treatment with 100 nM VIP for 1h before xenograft resulted in increased tumor growth after 8 and, more remarkably, 15 days of injection. The same occurred with the mRNA expression of the main angiogenic factor, vascular endothelial growth factor (VEGF), as shown by real-time RT-PCR quantification. The proangiogenic activity of VIP was further established by showing increases of hemoglobin levels, Masson trichromic staining, and immunohistochemical CD34 staining in tumors excised 15 days after subcutaneous injection of VIP-treated cells as compared to control conditions. All these parameters indicate that VIP increases vessel formation. This xenograft model is a useful tool to study in vivo the effects of VIP-related peptides in tumor growth and development of blood supply as well as their therapeutical potential in prostate cancer.
Collapse
Affiliation(s)
- Beatriz Collado
- Department of Biochemistry and Molecular Biology, University of Alcalá, Alcalá de Henares 28871, Spain
| | | | | | | | | |
Collapse
|
10
|
Fernández-Martínez AB, Collado B, Bajo AM, Sánchez-Chapado M, Prieto JC, Carmena MJ. Vasoactive intestinal peptide induces cyclooxygenase-2 expression through nuclear factor-kappaB in human prostate cell lines Differential time-dependent responses in cancer progression. Mol Cell Endocrinol 2007; 270:8-16. [PMID: 17434257 DOI: 10.1016/j.mce.2007.01.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2006] [Revised: 01/11/2007] [Accepted: 01/11/2007] [Indexed: 11/27/2022]
Abstract
The effect of vasoactive intestinal peptide (VIP) on cyclooxygenase-2 (COX-2) expression was analyzed in human prostate non-neoplastic (RWPE-1) as well as cancer androgen-dependent (LNCaP) and independent (PC3) cells. The three cell lines expressed VIP mRNA and VIP peptide, as measured by RT-PCR and immunochemistry, which supports an autocrine/paracrine action of VIP in the prostate gland. VIP levels were progressively higher from non-neoplastic to androgen-dependent and independent cells. Real-time RT-PCR and Western-blotting showed that VIP stimulated both COX-2 mRNA and protein expression in a faster manner as prostate cancer stage progressed (i.e. RWPE1<LNCaP<PC3 cells). Furthermore, VIP induced higher levels of COX-2 protein expression in cancer cells as compared with non-neoplastic cells. The anti-inflammatory agent curcumin blocked VIP-induced COX-2 expression in all cell lines studied supporting the involvement of nuclear factor-kappaB (NFkappaB) in such a response. In fact, VIP increased the translocation of the NFkappaB p50 subunit to the nucleus and the binding of the active form to its target gene promoter, as measured by Western-blotting and ELISA, respectively. VIP provoked faster responses according to the most aggressive status in cancer progression (androgen-independent situation). These results together with the existence of two NFkappaB sites in the COX-2 gene promoter together suggest that COX-2 may be a target for VIP in prostate cancer progression. On the other hand, VIP could be a proinflammatory cytokine acting through the NFkappaB/COX-2 system.
Collapse
Affiliation(s)
- Ana B Fernández-Martínez
- Department of Biochemistry and Molecular Biology, University of Alcalá, 28871 Alcalá de Henares, Spain
| | | | | | | | | | | |
Collapse
|
11
|
Sastry KSR, Smith AJ, Karpova Y, Datta SR, Kulik G. Diverse Antiapoptotic Signaling Pathways Activated by Vasoactive Intestinal Polypeptide, Epidermal Growth Factor, and Phosphatidylinositol 3-Kinase in Prostate Cancer Cells Converge on BAD. J Biol Chem 2006; 281:20891-20901. [PMID: 16728406 DOI: 10.1074/jbc.m602928200] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
It has been demonstrated that vasoactive intestinal polypeptide, epidermal growth factor, and chronic activation of phosphatidylinositol 3-kinase can protect prostate cancer cells from apoptosis; however, the signaling pathways that they use and molecules that they target are unknown. We report that vasoactive intestinal polypeptide, epidermal growth factor, and phosphatidylinositol 3-kinase activate independent signaling pathways that phosphorylate the proapoptotic protein BAD. Vasoactive intestinal polypeptide operated via protein kinase A, epidermal growth factor required Ras activity, and effects of phosphatidylinositol 3-kinase were predominantly mediated by Akt. BAD phosphorylation was critical for the antiapoptotic effects of each signaling pathway. None of these survival signals was able to rescue cells that express BAD with mutations in phosphorylation sites, whereas knockdown of BAD expression with small hairpin RNA rendered cells insensitive to apoptosis. Taken together, these results identify BAD as a convergence point of several antiapoptotic signaling pathways in prostate cells.
Collapse
Affiliation(s)
- Konduru S R Sastry
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, North Carolina 27157
| | - Adrienne Joy Smith
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, North Carolina 27157
| | - Yelena Karpova
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, North Carolina 27157
| | - Sandeep Robert Datta
- Center for Neurobiology and Behavior, Columbia University, New York, New York 10032
| | - George Kulik
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, North Carolina 27157.
| |
Collapse
|
12
|
Collado B, Sánchez MG, Díaz-Laviada I, Prieto JC, Carmena MJ. Vasoactive intestinal peptide (VIP) induces c-fos expression in LNCaP prostate cancer cells through a mechanism that involves Ca2+ signalling. Implications in angiogenesis and neuroendocrine differentiation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2005; 1744:224-33. [PMID: 15921770 DOI: 10.1016/j.bbamcr.2005.04.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2004] [Revised: 03/30/2005] [Accepted: 04/15/2005] [Indexed: 01/05/2023]
Abstract
The effect of vasoactive intestinal peptide (VIP) on intracellular Ca(2+) levels and its relationship with the expression of c-fos and vascular endothelial growth factor (VEGF) as well as with neuroendocrine (NE) differentiation were investigated in human prostate LNCaP cells. VIP induced the expression of c-fos mRNA as studied by reverse transcription polymerase chain reaction (RT-PCR). It was accompanied by VIP stimulation of c-fos protein synthesis, as measured by Western blot analysis. VIP enhanced intracellular Ca(2+) levels as evaluated using the calcium probe fura-2. VIP regulation of c-fos expression depended on [Ca(2+)](i) concentration since the intracellular calcium chelator BAPTA/AM decreased c-fos expression (both mRNA and protein) to basal levels. As shown by means of real-time RT-PCR, VIP stimulated VEGF mRNA expression: the effect was inhibited by 40% in the presence of curcumin (an inhibitor of AP-1 binding), and it was dependent on Ca(2+) since BAPTA/AM inhibited this VIP action by 43%. Similar observations were made on the effects of BAPTA/AM and curcumin on VIP stimulation of VEGF protein expression. Simultaneous treatment of cells with the protein kinase A inhibitor H89 and BAPTA/AM completely blocked this VIP effect, whereas each agent alone led only to a partial inhibition. In addition, the calcium chelator blocked by 37% the ability of VIP to induce NE cell differentiation as estimated by the observation of neurite development. These features support a VIP signalling pathway that could be mediated through both cAMP and [Ca(2+)](i) increase in prostate LNCaP cancer cells. Moreover, our data suggest the implication of c-Fos on the induction of the main angiogenic factor VEGF since the promoter region of the VEGF gene possesses AP-1 (i.e., c-Fos/c-Jun heterodimer) response elements. This feature represents a link between the nuclear oncogene c-fos, angiogenesis and NE differentiation by means of an initiating signal upon VIP receptors.
Collapse
Affiliation(s)
- Beatriz Collado
- Department of Biochemistry and Molecular Biology, Alcalá University, Alcalá de Henares, Spain
| | | | | | | | | |
Collapse
|
13
|
García-Fernández MO, Collado B, Bodega G, Cortés J, Ruíz-Villaespesa A, Carmena MJ, Prieto JC. Pituitary adenylate cyclase-activating peptide/vasoactive intestinal peptide receptors in human normal mammary gland and breast cancer tissue. Gynecol Endocrinol 2005; 20:327-33. [PMID: 16019382 DOI: 10.1080/09513590500098240] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activating peptide (PACAP) bind similarly to VPAC1 and VPAC2 receptors, whereas PACAP binds with higher affinity than VIP to PAC1 receptors. Here we demonstrate by different approaches the expression of the three subclasses of PACAP/VIP receptors in human normal and malignant breast tissue. At the mRNA level, reverse transcription-polymerase chain reaction experiments showed VPAC1 and VPAC2 receptors as well as various isoforms (null, hip/hop) of PAC1 receptors due to alternative splicing. At the protein level, Western blot experiments revealed the three subclasses of receptor although no conclusive differences could be established when comparing control, peritumoral and tumoral tissue samples. Immunohistochemistry showed the distribution of these receptors: they were located at epithelial cells in normal and cancer conditions but also in leukocytes at the stromal level in carcinomatous tissue. A weaker immunostaining of PAC1 receptors in normal tissue and a strong density of the three PACAP/VIP receptor subclasses in cancer tissue may be related to differential expression patterns during breast tumor progression but more samples need to be studied to validate this hypothesis. PAC1, VPAC1 and VPAC2 receptors were functional, as shown by their coupling to adenylate cyclase stimulation: VIP, PACAP-27 and PACAP-38 behaved similarly at this level, whereas both VPAC receptors acted alike as shown by means of specific peptide agonists and antagonists. The present results together with the known presence of PACAP and VIP in the mammary gland support a paracrine/autocrine involvement of both peptides at this level in physiological and pathological conditions, i.e. during malignant transformation.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Breast Neoplasms/metabolism
- Case-Control Studies
- Cell Transformation, Neoplastic
- Female
- Humans
- Immunohistochemistry
- Mammary Glands, Human/metabolism
- Middle Aged
- RNA, Messenger/analysis
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide
- Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide, Type I
- Receptors, Vasoactive Intestinal Peptide/genetics
- Receptors, Vasoactive Intestinal Peptide/metabolism
- Receptors, Vasoactive Intestinal Peptide, Type II
- Receptors, Vasoactive Intestinal Polypeptide, Type I
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
- M Olga García-Fernández
- Department of Biochemistry and Molecular Biology, University of Alcalá, E-28871 Alcalá de Henares, Spain
| | | | | | | | | | | | | |
Collapse
|
14
|
Sion-Vardy N, Tzikinovsky A, Bolotyn A, Segal S, Fishman D. Augmented expression of chromogranin A and serotonin in peri-malignant benign prostate epithelium as compared to adenocarcinoma. Pathol Res Pract 2005; 200:493-9. [PMID: 15462496 DOI: 10.1016/j.prp.2004.06.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
There is a growing body of evidence that the occurrence of neuroendocrine (NE) differentiation in prostate carcinoma correlates with poor prognosis, tumor progression, and androgen-independence. In the present study, the expression of common NE markers, i.e., chromogranin A (ChGA), serotonin (5HT), neuron-specific enolase (NSE) and adrenomedullin (AM), was retrospectively examined in formalin-fixed, paraffin-embedded prostate tissue samples obtained from patients with adenocarcinoma and from patients with nodular hyperplasia of the prostatic gland (NHPG) (33 and 28, respectively). The statistical analysis of the results (tested the equality of matched pairs of observations using the Wilcoxon matched-pairs signed ranks test) revealed a more prominent expression of ChGA in benign epithelial cells adjacent to adenocarcinomatic lesions (Peri-PAC) than in the adenocarcinoma (PAC) (p = 0.0049). A similar pattern of expression was detected for 5HT (p = 0.000). When comparing the expression of ChGA and 5HT in tissue samples originating in cancer patients with those obtained from NHPG samples, more ChGA and 5HT were expressed in Peri-PAC than in NHPG (p = 0.0004 and 0.002, respectively). The results obtained raise the possibility that adenocarcinoma cells urge some adjacent benign epithelial cells to differentiate into NE cells, which, in turn, may promote tumor growth and invasion.
Collapse
Affiliation(s)
- Netta Sion-Vardy
- Pathology Institute, Soroka University Medical Center, Beer-Sheva, Israel
| | | | | | | | | |
Collapse
|
15
|
Hensley HH, Chang WC, Clapper ML. Detection and volume determination of colonic tumors in Min mice by magnetic resonance micro-imaging. Magn Reson Med 2004; 52:524-9. [PMID: 15334570 DOI: 10.1002/mrm.20175] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
We applied MRI to the in vivo detection of spontaneous colorectal tumors in a unique mouse model, the Fox Chase Cancer Center (FCCC) ApcMIN mouse. Unlike other Min (multiple intestinal neoplasia) strains, FCCC ApcMIN animals develop an appreciable number of tumors in the large intestine, which makes them an appropriate mouse model for colon cancer in humans. We describe a method for marking the colon on MRI data sets that involves a bowel-cleansing procedure and the insertion of a polyurethane tube (filled with an MRI contrast agent) fully into the colon. We found that tumors as small as 1.5 mm in diameter can be consistently identified from MRI datasets with a voxel size of 0.1 mm x 0.133 mm x 0.133 mm. Tumor volumes were determined from the MRM data sets with the use of a novel approach to planimetry in 3D data sets. We observed a correlation between tumor volume (as measured from the MRI datasets) and tumor weight of 0.942, and a P-value of 0.008, based on Spearman's test. These data show that MRI can be used to accurately monitor tumor growth in mouse models of colorectal carcinogenesis.
Collapse
|
16
|
Gozes I, Furman S. Clinical endocrinology and metabolism. Potential clinical applications of vasoactive intestinal peptide: a selected update. Best Pract Res Clin Endocrinol Metab 2004; 18:623-40. [PMID: 15533779 DOI: 10.1016/j.beem.2004.08.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Neuropeptides are expressed in neurons innervating endocrine cells or in endocrine cells and cancer cells, and are released on site to act as hormones and growth factors. Vasoactive intestinal peptide (VIP) was first discovered in the early 1970s and has since become the area of research for many laboratories. VIP has a neuroendocrine role as it is intimately involved with the synthesis, secretion and action of other neuroendocrine hormones as well as cytokines and chemokines. Major outcomes of VIP downregulation encompass developmental and behavioral dysfunctions, including impaired diurnal rhythms. Overexpression of VIP has been associated with diarrhea and cancer, and overexpression of VIP receptors is associated with cancerous growth. This short review outlines some of the recent progress made in VIP research.
Collapse
Affiliation(s)
- Illana Gozes
- Department of Clinical Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| | | |
Collapse
|
17
|
Kiaris H, Koutsilieris M, Kalofoutis A, Schally AV. Growth hormone-releasing hormone and extra-pituitary tumorigenesis: therapeutic and diagnostic applications of growth hormone-releasing hormone antagonists. Expert Opin Investig Drugs 2003; 12:1385-94. [PMID: 12882623 DOI: 10.1517/13543784.12.8.1385] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Growth hormone-releasing hormone (GHRH) regulates growth hormone release from the pituitary. However, in addition to this neuroendocrine action, much evidence implies an additional role for GHRH in carcinogenesis in non-pituitary tissues. This role of GHRH in cancer development appears to be due to the operation of several mechanisms, which involve the regulation of the growth hormone-dependent hepatic insulin-like growth factor I (IGFI) production, tumoural IGF-I and IGF-II secretion and direct action of GHRH on tumour cells by autocrine and/or paracrine pathways. This review summarises the available information regarding the role of GHRH in tumorigenesis with special emphasis on the direct action of GHRH in primary and experimental cancers.
Collapse
Affiliation(s)
- Hippokratis Kiaris
- Department of Biological Chemistry, Medical School, University of Athens, 75 Micras Asias, 115 27 Athens, Greece.
| | | | | | | |
Collapse
|
18
|
Gutiérrez-Cañas I, Rodríguez-Henche N, Bolaños O, Carmena MJ, Prieto JC, Juarranz MG. VIP and PACAP are autocrine factors that protect the androgen-independent prostate cancer cell line PC-3 from apoptosis induced by serum withdrawal. Br J Pharmacol 2003; 139:1050-8. [PMID: 12839880 PMCID: PMC1573913 DOI: 10.1038/sj.bjp.0705317] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
1. In the present study, we describe the expression of the neuropeptides vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activating polypeptide (PACAP) as well as their receptors in PC-3 cells, a human prostate cancer cell line. In addition, we have investigated their role in apoptosis induced by serum starvation. 2. By RT-PCR and immunocytochemistry assays, we have demonstrated the production of VIP and PACAP in PC-3 cells. 3. We have demonstrated by RT-PCR and binding assays the expression of common PACAP/VIP (VPAC(1) and VPAC(2)) receptors, but not PACAP-specific (PAC(1)) receptors. The pharmacological profile of [(125)I]-VIP binding assays was as follows: VPAC(1) antagonist=VPAC(1) agonist>VIP>VPAC(2) agonist (IC(50)=1.2, 1.5, 2.3 and 30 nM, respectively). In addition, both receptor subtypes are functional since VIP, PACAP-27 or VPAC(1) and VPAC(2) agonists all increased the intracellular levels of cAMP. 4. The expression of both peptides and their receptors is similar in serum-cultured and serum-deprived PC-3 cells. The treatment of serum-deprived PC-3 cells with exogenous VIP or PACAP-27 increases cell number and viability in a dose-dependent manner, as demonstrated by cellular counting and MTT assays. The increased cell survival is exerted through the VPAC(1) receptor, since a VPAC(1), but not VPAC(2), receptor agonist, mimics the effects and a VPAC(1) receptor antagonist blocks it. Moreover, VIP and PACAP-27 inhibit genomic DNA fragmentation in PC-3 cells triggered by serum starvation, and increase the immunoreactivity of the antiapoptotic protein bcl-2. 5. Our results suggest that VIP and PACAP are autocrine/paracrine factors that protect PC-3 cells from apoptosis through VPAC1 receptors.
Collapse
Affiliation(s)
- Irene Gutiérrez-Cañas
- Department of Biochemistry and Molecular Biology, University of Alcalá, E-28871 Alcalá de Henares, Spain
| | - Nieves Rodríguez-Henche
- Department of Biochemistry and Molecular Biology, University of Alcalá, E-28871 Alcalá de Henares, Spain
| | - Oscar Bolaños
- Department of Biochemistry and Molecular Biology, University of Alcalá, E-28871 Alcalá de Henares, Spain
| | - María J Carmena
- Department of Biochemistry and Molecular Biology, University of Alcalá, E-28871 Alcalá de Henares, Spain
| | - Juan C Prieto
- Department of Biochemistry and Molecular Biology, University of Alcalá, E-28871 Alcalá de Henares, Spain
- Author for correspondence:
| | - María G Juarranz
- Department of Cellular Biology, Faculty of Biology, Complutente University, Madrid, Spain
| |
Collapse
|
19
|
Plonowski A, Schally AV, Busto R, Krupa M, Varga JL, Halmos G. Expression of growth hormone-releasing hormone (GHRH) and splice variants of GHRH receptors in human experimental prostate cancers. Peptides 2002; 23:1127-33. [PMID: 12126741 DOI: 10.1016/s0196-9781(02)00043-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The expression of mRNA for GHRH and splice variants (SVs) of GHRH receptors in LNCaP, MDA-PCa-2b and PC-3 human prostate cancers grown in nude mice was investigated by RT-PCR. The expression of mRNA for GHRH was detected in LNCaP and PC-3, but not in MDA-PCa-2b prostatic carcinoma. RT-PCR analyses of mRNA isolated from LNCaP, MDA-PCa-2b and PC-3 cancers, revealed the presence of 720 and 566 bp products, corresponding to SV(1) and SV(2) isoforms of GHRH receptors. In PC-3 tumor membranes a radiolabeled GHRH antagonist [125I]-JV-1-42 was bound to one class of high-affinity binding sites (K(d)=1.81+/-0.47 nM) and maximum binding capacity of 332.7+/-27.8 fmol/mg membrane protein. The in vivo uptake of [125I]-JV-1-42 was observed in all xenografts of human prostate cancer, the tracer accumulation being the highest in PC-3 tumors. These results indicate that GHRH and SVs of its receptors, different from those found in the pituitary, are present in experimental human prostate cancers and may form a local mitogenic loop. The antiproliferative effects of GHRH antagonists on growth of prostate cancer could be exerted in part by an interference with this local GHRH system.
Collapse
Affiliation(s)
- Artur Plonowski
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, 1601 Perdido Street, New Orleans, LA 70112-1262, USA
| | | | | | | | | | | |
Collapse
|