1
|
Allen BG, Merlen C, Branco AF, Pétrin D, Hébert TE. Understanding the impact of nuclear-localized GPCRs on cellular signalling. Cell Signal 2024; 123:111358. [PMID: 39181220 DOI: 10.1016/j.cellsig.2024.111358] [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: 07/24/2024] [Revised: 08/14/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024]
Abstract
G protein-coupled receptors (GPCRs) have historically been associated with signalling events driven from the plasma membrane. More recently, signalling from endosomes has been recognized as a feature of internalizing receptors. However, there was little consideration given to the notion that GPCRs can be targeted to distinct subcellular locations that did not involve an initial trafficking to the cell surface. Here, we focus on the evidence for and the potential impact of GPCR signalling specifically initiated from the nuclear membrane. We also discuss the possibilities for selectively targeting this and other internal pools of receptors as novel venues for drug discovery.
Collapse
Affiliation(s)
- Bruce G Allen
- Montreal Heart Institute, Montréal, Québec H1T 1C8, Canada; Departments of Biochemistry and Molecular Medicine, Medicine, Pharmacology and Physiology, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| | | | - Ana F Branco
- Montreal Heart Institute, Montréal, Québec H1T 1C8, Canada
| | - Darlaine Pétrin
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada.
| |
Collapse
|
2
|
Thakur M, Tomar VS, Dale E, Gomella LG, Solomides C, Kolesnikov O, Keith SW, Navarro HT, Dahlgren O, Chaga M, Trabulsi EJ. Targeting genomic receptors in voided urine for confirmation of benign prostatic hyperplasia. BJUI COMPASS 2024; 5:675-680. [PMID: 39022663 PMCID: PMC11250152 DOI: 10.1002/bco2.362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 02/27/2024] [Accepted: 03/18/2024] [Indexed: 07/20/2024] Open
Abstract
Objectives The objective of this study is to validate a hypothesis that a non-invasive optical imaging assay targeting genomic VPAC receptors on malignant cells shed in voided urine will represent either benign prostatic hyperplasia (BPH) or prostatic cancer (PCa). Risk for BPH in men 50-70 years old is 50-70% and PCa is 17%. BPH and PCa can coexist in 20% of men with BPH. Most commonly practiced methods to diagnose BPH do not distinguish BPH from PCa. Patients or Materials and Methods Males with BPH (N = 97, 60.8 ± 6.3 years, prostate-specific antigen 0.7 ± 0.4 ng/mL) and without oncologic disease (N = 35, 63.4 ± 5.8 years, prostate-specific antigen < 1.5 ng/mL) signed informed consent form and provided voided urine. Urine was cytocentrifuged, cells collected on glass slide, fixed, treated with VPAC specific fluorophore TP4303 (Kd 3.1 × 10-8M), washed, incubated with DAPI and observed using a fluorescence microscope. Cells with no VPAC did not fluoresce (BPH) and those with VPAC had red-orange fluorescence (PCa). Real-time polymerase chain reaction analyses for VPAC and NKX3.1 assay for cell origin were performed. Results Eighty-seven subjects were negative for VPAC expression. Positive VPAC expression was noted in 10 subjects. Patient chart review for clinical data on these 10 VPAC positive subjects showed five had nephrolithiasis, three had renal cysts, one had prostatitis and one was being treated with finasteride. Real-time polymerase chain reaction analysis-VPAC expressions for 7 normal and 12 BPH subjects were 1.31 ± 1.26 and 0.94 ± 0.89, respectively (P = 0.46). NKX3.1 showed cells of prostate origin for finasteride-treated patient. Specificity for VPAC urine assay for excluding prostate cancer in this BPH cohort was 88.5%, positive predictive value 0.00% and negative predictive value 100%. Conclusion VPAC assay may contribute extensively for BPH diagnosis and warrant continued investigation.
Collapse
Affiliation(s)
- Mathew Thakur
- Department of Radiology, Urology, Radiation Oncology and S. Kimmel Cancer CenterThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Vivek S. Tomar
- Department of RadiologyThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Emma Dale
- Department of RadiologyThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Leonard G. Gomella
- Department of Urology and S. Kimmel Cancer CenterThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Charalambos Solomides
- Department of Pathology and Genomic MedicineThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Oleksandr Kolesnikov
- Department of Urology and S. Kimmel Cancer CenterThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Scott W. Keith
- Division of Biostatistics and S. Kimmel Cancer CenterThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Hector T. Navarro
- Department of RadiologyThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Olivia Dahlgren
- Department of UrologyThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Michael Chaga
- Department of RadiologyThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Edouard J. Trabulsi
- Department of UrologyJefferson Einstein Medical Center and Thomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
3
|
Witzel R, Block A, Pollmann S, Oetzel L, Fleck F, Bonaterra GA, Kinscherf R, Schwarz A. PACAP regulates VPAC1 expression, inflammatory processes and lipid homeostasis in M1- and M2-macrophages. Front Cardiovasc Med 2023; 10:1264901. [PMID: 37900572 PMCID: PMC10611464 DOI: 10.3389/fcvm.2023.1264901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 09/12/2023] [Indexed: 10/31/2023] Open
Abstract
Background Pituitary adenylate cyclase-activating polypeptide (PACAP) acts as an anti-atherogenic neuropeptide and plays an important role in cytoprotective, as well as inflammatory processes, and cardiovascular regulation. Therefore, the aim of this study is to investigate the regulatory effects of PACAP and its receptor VPAC1 in relation to inflammatory processes and lipid homeostasis in different macrophage (MΦ) subtypes. Methods To investigate the role of PACAP deficiency in the pathogenesis of atherosclerosis under standard chow (SC) or cholesterol-enriched diet (CED) in vivo, PACAP-/- mice were crossbred with ApoE-/- to generate PACAP-/-/ApoE-/- mice. Lumen stenosis in the aortic arch and different MΦ-subtypes were analyzed in atherosclerotic plaques by quantitative immunohistochemistry. Undifferentiated bone marrow-derived cells (BMDC) from 30-weeks-old ApoE-/- and PACAP-/-/ApoE-/- mice were isolated, differentiated into BMDM1- and BMDM2-MΦ, and incubated with oxidized low-density lipoprotein (oxLDL). In addition, PMA-differentiated human THP-1 MΦ were further differentiated into M1-/M2-MΦ and subsequently treated with PACAP38, the VPAC1 agonist [(Ala11,22,28)VIP], the antagonist (PG 97-269), and/or oxLDL. Uptake/accumulation of oxLDL was analyzed by oxLDL-DyLight™488 and Bodipy™ 493/503. The mRNA expression was analyzed by qRT-PCR, protein levels by Western blot, and cytokine release by ELISA. Results In vivo, after 30 weeks of SC, PACAP-/-/ApoE-/- mice showed increased lumen stenosis compared with ApoE-/- mice. In atherosclerotic plaques of PACAP-/-/ApoE-/- mice under CED, immunoreactive areas of VPAC1, CD86, and CD163 were increased compared with ApoE-/- mice. In vitro, VPAC1 protein levels were increased in PACAP-/-/ApoE-/- BMDM compared with ApoE-/- BMDM, resulting in increased TNF-α mRNA expression in BMDM1-MΦ and decreased TNF-α release in BMDM2-MΦ. Concerning lipid homeostasis, PACAP deficiency decreased the area of lipid droplets in BMDM1-/M2-MΦ with concomitant increasing adipose differentiation-related protein level. In THP-1 M1-/M2-MΦ, the VPAC1 antagonist increased the uptake of oxLDL, whereas the VPAC1 agonist decreased the oxLDL-induced intracellular triglyceride content. Conclusion Our data suggest that PACAP via VPAC1 signaling plays an important regulatory role in inflammatory processes in atherosclerotic plaques and in lipid homeostasis in different MΦ-subtypes, thereby affecting foam cell formation. Therefore, VPAC1 agonists or PACAP may represent a new class of anti-atherogenic therapeutics.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Anja Schwarz
- Department of Medical Cell Biology, Institute of Anatomy and Cell Biology, Philipps-University of Marburg, Marburg, Germany
| |
Collapse
|
4
|
Human CD4 +CD45RA + T Cells Behavior after In Vitro Activation: Modulatory Role of Vasoactive Intestinal Peptide. Int J Mol Sci 2022; 23:ijms23042346. [PMID: 35216459 PMCID: PMC8878027 DOI: 10.3390/ijms23042346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/11/2022] [Accepted: 02/17/2022] [Indexed: 11/30/2022] Open
Abstract
Naїve CD4+ T cells, which suffer different polarizing signals during T cell receptor activation, are responsible for an adequate immune response. In this study, we aimed to evaluate the behavior of human CD4+CD45RA+ T cells after in vitro activation by anti-CD3/CD28 bead stimulation for 14 days. We also wanted to check the role of the VIP system during this process. The metabolic biomarker Glut1 was increased, pointing to an increase in glucose requirement whereas Hif-1α expression was higher in resting than in activated cells. Expression of Th1 markers increased at the beginning of activation, whereas Th17-associated biomarkers augmented after that, showing a pathogenic Th17 profile with a possible plasticity to Th17/1. Foxp3 mRNA expression augmented from day 4, but no parallel increases were observed in IL-10, IL-2, or TGFβ mRNA expression, meaning that these potential differentiated Treg could not be functional. Both VIP receptors were located on the plasma membrane, and expression of VPAC2 receptor increased significantly with respect to the VPAC1 receptor from day 4 of CD4+CD45RA+ T activation, pointing to a shift in VPAC receptors. VIP decreased IFNγ and IL-23R expression during the activation, suggesting a feasible modulation of Th17/1 plasticity and Th17 stabilization through both VPAC receptors. These novel results show that, without polarizing conditions, CD4+CD45RA+ T cells differentiate mainly to a pathogenic Th17 subset and an unpaired Treg subset after several days of activation. Moreover, they confirm the important immunomodulatory role of VIP, also on naїve Th cells, stressing the importance of this neuropeptide on lymphocyte responses in different pathological or non-pathological situations.
Collapse
|
5
|
Watson G, Coyne Z, Houlihan E, Leonard G. Exercise oncology: an emerging discipline in the cancer care continuum. Postgrad Med 2021; 134:26-36. [PMID: 34854802 DOI: 10.1080/00325481.2021.2009683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Exercise is an essential component of healthy living and well-being. While there is a global acceptance of the benefits of exercise for the general population, there exists hesitancy and confusion among health-care professionals, particularly oncologists, as to whether these benefits translate to cancer patients. Patient referrals to accessible, structured exercise programs in this setting are often overlooked by physicians when formulating a cancer management plan. There is however increasing awareness and acceptance of cancer survivorship as a part of the cancer care continuum, identifying multiple factors that contribute to well-being beyond just cancer outcomes. Efforts to optimize cancer survivorship have stimulated further academic interest in the benefits of healthy living and particularly exercise oncology. There is now compelling evidence that exercise, which includes daily activities such as walking, as well as structured programs, improves multiple-cancer outcomes such as fatigue, quality of life and likely survival, and warrants consideration in the multidisciplinary care of cancer patients. International guidelines have been established that recommend counseling cancer patients with regard to healthy lifestyle changes including exercise. However, there still remains a reluctance from oncology physicians to prescribe exercise for these patients, largely due to uncertainty with regard to their patients' ability to tolerate such an intervention, coupled with insufficient understanding of the potential benefits of these programs. There also exist patient barriers and attitudes that must be overcome. Exercise strategies and bespoke programs that are tailored to the unique abilities and goals of the patients will enhance participation. To move the field forward and integrate exercise oncology into standard practice, it is imperative to raise awareness of the benefits of exercise to cancer patients and their health-care providers. This will facilitate the prescription of exercise as part of the multimodal treatment plan with the ultimate aim of promoting an active lifestyle to optimize patient care and well-being.
Collapse
Affiliation(s)
- Ga Watson
- Department of Medical Oncology, University Hospital Galway, Galway, Ireland
| | - Zl Coyne
- Department of Medical Oncology, University Hospital Galway, Galway, Ireland
| | - E Houlihan
- Department of Physiotherapy, Cancer Care West, Galway, Ireland
| | - Gd Leonard
- Department of Medical Oncology, University Hospital Galway, Galway, Ireland
| |
Collapse
|
6
|
Nerli RB, Ghagane SC, Rangrez S, Chandra S, Thakur ML, Gomella L. Detection of bladder cancer using voided urine sample and by targeting genomic VPAC receptors. Indian J Urol 2021; 37:345-349. [PMID: 34759527 PMCID: PMC8555562 DOI: 10.4103/iju.iju_132_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 07/12/2021] [Accepted: 09/23/2021] [Indexed: 11/16/2022] Open
Abstract
Introduction: Cells exfoliated into urine from the bladder can help to diagnose the cancer. The objective of this study was to validate the hypothesis that bladder cancer could be detected noninvasively by a simple and reliable assay targeting genomic VPAC (combined vasoactive intestinal peptide and pituitary adenylate cyclase-activating peptide family of cell surface receptors) receptors expressed on the malignant bladder cancer cells shed in the voided urine. Methods: Patients ≥18 years of age with either imaging (ultrasonography/computed tomography [CT])-confirmed bladder tumors or those who have been previously treated for nonmuscle invasive bladder tumors and were visiting the department for check cystoscopy, formed the study group. Freshly voided urine sample was collected from these patients and sent for conventional cytology examination, 5-aminolevulinic acid (ALA) fluorescent urine cytology, and for positivity of VPAC receptors. Results: A total of 103 patients were prospectively included in the study. Of these, 65 patients (Group I) presented with image-diagnosed (ultrasonography and/or CT) bladder cancer. The remaining 38 patients (Group II) were previously diagnosed cases of nonmuscle invasive bladder cancer and presented for follow-up and check cystoscopy. The sensitivity for VPAC receptor positivity was 89.23% compared to conventional cytology (63.07%) and 5-ALA fluorescent urine cytology (87.69%). The specificity of VPAC receptor positivity was 100% compared to conventional cytology (100%) and 5-ALA-induced fluorescent cytology (90.47%). Conclusions: Our preliminary study shows encouraging results with VPAC receptor positivity studies, which has a high sensitivity when compared to the conventional cytology.
Collapse
Affiliation(s)
- Rajendra B Nerli
- Department of Urology, JN Medical College, KLE Academy of Higher Education and Research (Deemed-to-be-University), JNMC Campus, Karnataka, India.,Division of Urologic-Oncology, Urinary Biomarkers Research Centre, KLES Dr. Prabhakar Kore Hospital and Medical Research Centre, Belagavi, Karnataka, India
| | - Shridhar C Ghagane
- Department of Urology, JN Medical College, KLE Academy of Higher Education and Research (Deemed-to-be-University), JNMC Campus, Karnataka, India.,Division of Urologic-Oncology, Urinary Biomarkers Research Centre, KLES Dr. Prabhakar Kore Hospital and Medical Research Centre, Belagavi, Karnataka, India
| | - Shadab Rangrez
- Department of Urology, JN Medical College, KLE Academy of Higher Education and Research (Deemed-to-be-University), JNMC Campus, Karnataka, India
| | - Shreya Chandra
- Division of Urologic-Oncology, Urinary Biomarkers Research Centre, KLES Dr. Prabhakar Kore Hospital and Medical Research Centre, Belagavi, Karnataka, India
| | - Madhukar L Thakur
- Department of Urology and Radiology, Thomas Jefferson University, Philadelphia, PA, USA.,Department of Urology, Thomas Jefferson University, Philadelphia, PA, USA.,The Sidney Kimmel Cancer Centre, Thomas Jefferson University, Philadelphia, PA, USA
| | - Leonard Gomella
- Department of Urology, Thomas Jefferson University, Philadelphia, PA, USA.,The Sidney Kimmel Cancer Centre, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
7
|
Hajdú T, Kovács P, Zsigrai E, Takács R, Vágó J, Cho S, Sasi-Szabó L, Becsky D, Keller-Pinter A, Emri G, Rácz K, Reglodi D, Zákány R, Juhász T. Pituitary Adenylate Cyclase Activating Polypeptide Has Inhibitory Effects on Melanoma Cell Proliferation and Migration In Vitro. Front Oncol 2021; 11:681603. [PMID: 34616669 PMCID: PMC8488289 DOI: 10.3389/fonc.2021.681603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 08/19/2021] [Indexed: 11/13/2022] Open
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) is an endogenous neuropeptide which is distributed throughout the body. PACAP influences development of various tissues and exerts protective function during cellular stress and in some tumour formation. No evidence is available on its role in neural crest derived melanocytes and its malignant transformation into melanoma. Expression of PACAP receptors was examined in human skin samples, melanoma lesions and in a primary melanocyte cell culture. A2058 and WM35 melanoma cell lines, representing two different stages of melanoma progression, were used to investigate the effects of PACAP. PAC1 receptor was identified in melanocytes in vivo and in vitro and in melanoma cell lines as well as in melanoma lesions. PACAP administration did not alter viability but decreased proliferation of melanoma cells. With live imaging random motility, average speed, vectorial distance and maximum distance of migration of cells were reduced upon PACAP treatment. PACAP administration did not alter viability but decreased proliferation capacity of melanoma cells. On the other hand, PACAP administration decreased the migration of melanoma cell lines towards fibronectin chemoattractant in the Boyden chamber. Furthermore, the presence of the neuropeptide inhibited the invasion capability of melanoma cell lines in Matrigel chambers. In summary, we provide evidence that PACAP receptors are expressed in melanocytes and in melanoma cells. Our results also prove that various aspects of the cellular motility were inhibited by this neuropeptide. On the basis of these results, we propose PACAP signalling as a possible target in melanoma progression.
Collapse
Affiliation(s)
- Tibor Hajdú
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Patrik Kovács
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Emese Zsigrai
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Roland Takács
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Judit Vágó
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Sinyoung Cho
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Department of Family Medicine, Seoul National University Hospital, Seoul, South Korea
| | - László Sasi-Szabó
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Dániel Becsky
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Aniko Keller-Pinter
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Gabriella Emri
- Department of Dermatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Kálmán Rácz
- Department of Forensic Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Dora Reglodi
- Department of Anatomy, PTE-MTA PACAP Research Team, Szentagothai Research Center, Medical School, University of Pécs, Pécs, Hungary
| | - Róza Zákány
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tamás Juhász
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
8
|
Gonçalves-Monteiro S, Ribeiro-Oliveira R, Vieira-Rocha MS, Vojtek M, Sousa JB, Diniz C. Insights into Nuclear G-Protein-Coupled Receptors as Therapeutic Targets in Non-Communicable Diseases. Pharmaceuticals (Basel) 2021; 14:439. [PMID: 34066915 PMCID: PMC8148550 DOI: 10.3390/ph14050439] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/27/2021] [Accepted: 04/30/2021] [Indexed: 12/14/2022] Open
Abstract
G-protein-coupled receptors (GPCRs) comprise a large protein superfamily divided into six classes, rhodopsin-like (A), secretin receptor family (B), metabotropic glutamate (C), fungal mating pheromone receptors (D), cyclic AMP receptors (E) and frizzled (F). Until recently, GPCRs signaling was thought to emanate exclusively from the plasma membrane as a response to extracellular stimuli but several studies have challenged this view demonstrating that GPCRs can be present in intracellular localizations, including in the nuclei. A renewed interest in GPCR receptors' superfamily emerged and intensive research occurred over recent decades, particularly regarding class A GPCRs, but some class B and C have also been explored. Nuclear GPCRs proved to be functional and capable of triggering identical and/or distinct signaling pathways associated with their counterparts on the cell surface bringing new insights into the relevance of nuclear GPCRs and highlighting the nucleus as an autonomous signaling organelle (triggered by GPCRs). Nuclear GPCRs are involved in physiological (namely cell proliferation, transcription, angiogenesis and survival) and disease processes (cancer, cardiovascular diseases, etc.). In this review we summarize emerging evidence on nuclear GPCRs expression/function (with some nuclear GPCRs evidencing atypical/disruptive signaling pathways) in non-communicable disease, thus, bringing nuclear GPCRs as targets to the forefront of debate.
Collapse
Affiliation(s)
- Salomé Gonçalves-Monteiro
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.G.-M.); (R.R.-O.); (M.S.V.-R.); (M.V.)
- LAQV/REQUIMTE, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Rita Ribeiro-Oliveira
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.G.-M.); (R.R.-O.); (M.S.V.-R.); (M.V.)
- LAQV/REQUIMTE, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Maria Sofia Vieira-Rocha
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.G.-M.); (R.R.-O.); (M.S.V.-R.); (M.V.)
- LAQV/REQUIMTE, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Martin Vojtek
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.G.-M.); (R.R.-O.); (M.S.V.-R.); (M.V.)
- LAQV/REQUIMTE, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Joana B. Sousa
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.G.-M.); (R.R.-O.); (M.S.V.-R.); (M.V.)
- LAQV/REQUIMTE, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Carmen Diniz
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.G.-M.); (R.R.-O.); (M.S.V.-R.); (M.V.)
- LAQV/REQUIMTE, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
9
|
Tripathi SK, Kean R, Bongiorno E, Hooper DC, Jin YY, Wickstrom E, McCue PA, Thakur ML. Targeting VPAC1 Receptors for Imaging Glioblastoma. Mol Imaging Biol 2021; 22:293-302. [PMID: 31292914 DOI: 10.1007/s11307-019-01388-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE Scintigraphic imaging of malignant glioblastoma (MG) continues to be challenging. We hypothesized that VPAC1 cell surface receptors can be targeted for positron emission tomography (PET) imaging of orthotopically implanted MG in a mouse model, using a VPAC1-specific peptide [64Cu]TP3805. PROCEDURES The expression of VPAC1 in mouse GL261 and human U87 glioma cell lines was determined by western blot. The ability of [64Cu]TP3805 to bind to GL261 and U87 cells was studied by cell-binding. Receptor-blocking studies were performed to validate receptor specificity. GL261 tumors were implanted orthotopically in syngeneic T-bet knockout C57BL/6 mouse brain (N = 15) and allowed to grow for 2-3 weeks. Mice were injected i.v., first with ~ 150 μCi of 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) then 24 h later with ~ 200 μCi of [64Cu]TP3805. In another set of tumor-bearing mice, (N = 5), ionic [64Cu]Cl2 was injected as a control. Mice were imaged at a 2-h post-injection using an Inveon micro-PET/CT, sacrificed and % ID/g of [64Cu]TP3805 and [64Cu]Cl2 were calculated in a tumor, normal brain, and other tissues. For histologic tissue examination, 3-μm thick sections of the tumors and normal brain were prepared, digital autoradiography (DAR) was performed, and then the sections were H&E stained for histologic examination. RESULTS Western blots showed a strong signal for VPAC1 on both cell lines. [64Cu]TP3805 cell-binding was 87 ± 1.5 %. Receptor-blocking reduced cell-binding to 24.3 ± 1.5 % (P < 0.01). PET imaging revealed remarkable accumulation of [64Cu]TP3805 in GL261 MG with a negligible background in the normal brain, as compared to [18F]FDG. Micro-PET/CT image analyses and tissue distribution showed that the brain tumor uptake for [64Cu]TP3805 was 8.2 ± 1.7 % ID/g and for [64Cu]Cl2 2.1 ± 0.5 % ID/g as compared to 1.0 ± 0.3 % ID/g and 1.4 ± 0.3 % ID/g for normal mouse brains, respectively. The high tumor/normal brain ratio for [64Cu]TP3805 (8.1 ± 1.1) allowed tumors to be visualized unequivocally. Histology and [64Cu]TP3805 DAR differentiated malignant tumors from healthy brain and confirmed PET findings. CONCLUSION Targeting VPAC1 receptors using [64Cu]TP3805 for PET imaging of MG is a promising novel approach and calls for further investigation.
Collapse
Affiliation(s)
- Sushil K Tripathi
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Rhonda Kean
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Emily Bongiorno
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Douglas C Hooper
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Eric Wickstrom
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Peter A McCue
- Department of Pathology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Mathew L Thakur
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA.
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, USA.
- Department of Urology, Thomas Jefferson University, Philadelphia, PA, USA.
- Department of Radiology, Laboratories of Radiopharmaceutical Research and Molecular Imaging, 1020 Locust Street, 359-JAH, Philadelphia, PA, 19107, USA.
| |
Collapse
|
10
|
Mohammad Nezhady MA, Rivera JC, Chemtob S. Location Bias as Emerging Paradigm in GPCR Biology and Drug Discovery. iScience 2020; 23:101643. [PMID: 33103080 PMCID: PMC7569339 DOI: 10.1016/j.isci.2020.101643] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
GPCRs are the largest receptor family that are involved in virtually all biological processes. Pharmacologically, they are highly druggable targets, as they cover more than 40% of all drugs in the market. Our knowledge of biased signaling provided insight into pharmacology vastly improving drug design to avoid unwanted effects and achieve higher efficacy and selectivity. However, yet another feature of GPCR biology is left largely unexplored, location bias. Recent developments in this field show promising avenues for evolution of new class of pharmaceuticals with greater potential for higher level of precision medicine. Further consideration and understanding of this phenomenon with deep biochemical and molecular insights would pave the road to success. In this review, we critically analyze this perspective and discuss new avenues of investigation.
Collapse
Affiliation(s)
- Mohammad Ali Mohammad Nezhady
- Programmes en Biologie Moléculaire, Faculté de Médecine, Université de Montréal, Montreal, QC, Canada
- Centre de Recherche du CHU Sainte-Justine, Montreal, QC H3T 1C5, Canada
- Corresponding author
| | | | - Sylvain Chemtob
- Programmes en Biologie Moléculaire, Faculté de Médecine, Université de Montréal, Montreal, QC, Canada
- Centre de Recherche du CHU Sainte-Justine, Montreal, QC H3T 1C5, Canada
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, QC, Canada
| |
Collapse
|
11
|
Martínez C, Juarranz Y, Gutiérrez-Cañas I, Carrión M, Pérez-García S, Villanueva-Romero R, Castro D, Lamana A, Mellado M, González-Álvaro I, Gomariz RP. A Clinical Approach for the Use of VIP Axis in Inflammatory and Autoimmune Diseases. Int J Mol Sci 2019; 21:E65. [PMID: 31861827 PMCID: PMC6982157 DOI: 10.3390/ijms21010065] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 12/16/2019] [Accepted: 12/18/2019] [Indexed: 12/11/2022] Open
Abstract
The neuroendocrine and immune systems are coordinated to maintain the homeostasis of the organism, generating bidirectional communication through shared mediators and receptors. Vasoactive intestinal peptide (VIP) is the paradigm of an endogenous neuropeptide produced by neurons and endocrine and immune cells, involved in the control of both innate and adaptive immune responses. Exogenous administration of VIP exerts therapeutic effects in models of autoimmune/inflammatory diseases mediated by G-protein-coupled receptors (VPAC1 and VPAC2). Currently, there are no curative therapies for inflammatory and autoimmune diseases, and patients present complex diagnostic, therapeutic, and prognostic problems in daily clinical practice due to their heterogeneous nature. This review focuses on the biology of VIP and VIP receptor signaling, as well as its protective effects as an immunomodulatory factor. Recent progress in improving the stability, selectivity, and effectiveness of VIP/receptors analogues and new routes of administration are highlighted, as well as important advances in their use as biomarkers, contributing to their potential application in precision medicine. On the 50th anniversary of VIP's discovery, this review presents a spectrum of potential clinical benefits applied to inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Carmen Martínez
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (Y.J.); (I.G.-C.); (M.C.); (S.P.-G.); (R.V.-R.); (D.C.); (A.L.); (R.P.G.)
| | - Yasmina Juarranz
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (Y.J.); (I.G.-C.); (M.C.); (S.P.-G.); (R.V.-R.); (D.C.); (A.L.); (R.P.G.)
| | - Irene Gutiérrez-Cañas
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (Y.J.); (I.G.-C.); (M.C.); (S.P.-G.); (R.V.-R.); (D.C.); (A.L.); (R.P.G.)
| | - Mar Carrión
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (Y.J.); (I.G.-C.); (M.C.); (S.P.-G.); (R.V.-R.); (D.C.); (A.L.); (R.P.G.)
| | - Selene Pérez-García
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (Y.J.); (I.G.-C.); (M.C.); (S.P.-G.); (R.V.-R.); (D.C.); (A.L.); (R.P.G.)
| | - Raúl Villanueva-Romero
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (Y.J.); (I.G.-C.); (M.C.); (S.P.-G.); (R.V.-R.); (D.C.); (A.L.); (R.P.G.)
| | - David Castro
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (Y.J.); (I.G.-C.); (M.C.); (S.P.-G.); (R.V.-R.); (D.C.); (A.L.); (R.P.G.)
| | - Amalia Lamana
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (Y.J.); (I.G.-C.); (M.C.); (S.P.-G.); (R.V.-R.); (D.C.); (A.L.); (R.P.G.)
| | - Mario Mellado
- Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología (CNB)/CSIC, 28049 Madrid, Spain;
| | - Isidoro González-Álvaro
- Servicio de Reumatología, Instituto de Investigación Médica, Hospital Universitario La Princesa, 28006 Madrid, Spain;
| | - Rosa P. Gomariz
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (Y.J.); (I.G.-C.); (M.C.); (S.P.-G.); (R.V.-R.); (D.C.); (A.L.); (R.P.G.)
| |
Collapse
|
12
|
Mosley RL, Lu Y, Olson KE, Machhi J, Yan W, Namminga KL, Smith JR, Shandler SJ, Gendelman HE. A Synthetic Agonist to Vasoactive Intestinal Peptide Receptor-2 Induces Regulatory T Cell Neuroprotective Activities in Models of Parkinson's Disease. Front Cell Neurosci 2019; 13:421. [PMID: 31619964 PMCID: PMC6759633 DOI: 10.3389/fncel.2019.00421] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 09/02/2019] [Indexed: 12/12/2022] Open
Abstract
A paradigm shift has emerged in Parkinson’s disease (PD) highlighting the prominent role of CD4+ Tregs in pathogenesis and treatment. Bench to bedside research, conducted by others and our own laboratories, advanced a neuroprotective role for Tregs making pharmacologic transformation of immediate need. Herein, a vasoactive intestinal peptide receptor-2 (VIPR2) peptide agonist, LBT-3627, was developed as a neuroprotectant for PD-associated dopaminergic neurodegeneration. Employing both 6-hydroxydopamine (6-OHDA) and α-synuclein (α-Syn) overexpression models in rats, the sequential administration of LBT-3627 increased Treg activity without altering cell numbers both in naïve animals and during progressive nigrostriatal degeneration. LBT-3627 administration was linked to reductions of inflammatory microglia, increased survival of dopaminergic neurons, and improved striatal densities. While α-Syn overexpression resulted in reduced Treg activity, LBT-3627 rescued these functional deficits. This occurred in a dose-dependent manner closely mimicking neuroprotection. Taken together, these data provide the basis for the use of VIPR2 agonists as potent therapeutic immune modulating agents to restore Treg activity, attenuate neuroinflammation, and interdict dopaminergic neurodegeneration in PD. The data underscore an important role of immunity in PD pathogenesis.
Collapse
Affiliation(s)
- R Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE, United States
| | - Yaman Lu
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE, United States
| | - Katherine E Olson
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE, United States
| | - Jatin Machhi
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE, United States
| | - Wenhui Yan
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE, United States
| | - Krista L Namminga
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE, United States
| | - Jenell R Smith
- Longevity Biotech, Inc., Philadelphia, PA, United States
| | | | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
13
|
Kumar P, Tripathi SK, Chen CP, Wickstrom E, Thakur ML. Evaluating Ga-68 Peptide Conjugates for Targeting VPAC Receptors: Stability and Pharmacokinetics. Mol Imaging Biol 2019; 21:130-139. [PMID: 29802552 DOI: 10.1007/s11307-018-1207-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE In recent years, considerable progress has been made in the use of gallium-68 labeled receptor-specific peptides for imaging oncologic diseases. The objective was to examine the stability and pharmacokinetics of [68Ga]NODAGA and DOTA-peptide conjugate targeting VPAC [combined for vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activating peptide (PACAP)] receptors on tumor cells. PROCEDURES A VPAC receptor-specific peptide was chosen as a model peptide and conjugated to NODAGA and DOTA via solid-phase synthesis. The conjugates were characterized by HPLC and MALDI-TOF. Following Ga-68 chelation, the radiochemical purity of Ga-68 labeled peptide conjugate was determined by radio-HPLC. The stability was tested against transmetallation using 100 nM Fe3+/Zn2+/Ca2+ ionic solution and against transchelation using 200 μM DTPA solution. The ex vivo and in vivo stability of the Ga-68 labeled peptide conjugate was tested in mouse plasma and urine. Receptor specificity was determined ex vivo by cell binding assays using human breast cancer BT474 cells. Positron emission tomography (PET)/X-ray computed tomography (CT) imaging, tissue distribution, and blocking studies were performed in mice bearing BT474 xenografts. RESULTS The chemical and radiochemical purity was greater than 95 % and both conjugates were stable against transchelation and transmetallation. Ex vivo stability at 60 min showed that the NODAGA-peptide-bound Ga-68 reduced to 42.1 ± 3.7 % (in plasma) and 37.4 ± 2.9 % (in urine), whereas the DOTA-peptide-bound Ga-68 was reduced to 1.2 ± 0.3 % (in plasma) and 4.2 ± 0.4 % (in urine) at 60 min. Similarly, the in vivo stability for [68Ga]NODAGA-peptide was decreased to 2.1 ± 0.2 % (in plasma) and 2.2 ± 0.4 % (in urine). For [68Ga]DOTA-peptide, it was decreased to 1.4 ± 0.3 % (in plasma) and 1.2 ± 0.4 % (in urine) at 60 min. The specific BT474 cell binding was 53.9 ± 0.8 % for [68Ga]NODAGA-peptide, 25.8 ± 1.4 % for [68Ga]-DOTA-peptide, and 18.8 ± 2.5 % for [68Ga]GaCl3 at 60 min. Inveon microPET/CT imaging at 1 h post-injection showed significantly (p < 0.05) higher tumor to muscle (T/M) ratio for [68Ga]NODAGA-peptide (3.4 ± 0.3) as compared to [68Ga]DOTA-peptide (1.8 ± 0.6). For [68Ga]GaCl3 and blocked mice, their ratios were 1.5 ± 0.6 and 1.5 ± 0.3 respectively. The tissue distributions data were similar to the PET imaging data. CONCLUSION NODAGA is superior to DOTA in terms of radiolabeling kinetics. The method of radiolabeling was reproducible and yielded higher specific activity. Although both agents have relatively low in vivo stability, PET/CT imaging studies delineated BC tumors with [68Ga]NODAGA-peptide, but not with [68Ga]DOTA-peptide.
Collapse
Affiliation(s)
- Pardeep Kumar
- Departments of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Sushil K Tripathi
- Departments of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - C P Chen
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, China
| | - Eric Wickstrom
- Departments of Biochemistry & Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA.,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Mathew L Thakur
- Departments of Radiology, Thomas Jefferson University, Philadelphia, PA, USA. .,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA. .,Departments of Radiation Oncology, Thomas Jefferson University, 1020 Locust Street, JAH Suite 359, Philadelphia, PA, 19107, USA. .,Departments of Urology, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
14
|
Villanueva-Romero R, Gutiérrez-Cañas I, Carrión M, González-Álvaro I, Rodríguez-Frade JM, Mellado M, Martínez C, Gomariz RP, Juarranz Y. Activation of Th lymphocytes alters pattern expression and cellular location of VIP receptors in healthy donors and early arthritis patients. Sci Rep 2019; 9:7383. [PMID: 31089161 PMCID: PMC6517580 DOI: 10.1038/s41598-019-43717-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 04/29/2019] [Indexed: 12/15/2022] Open
Abstract
Vasoactive Intestinal Peptide (VIP) is an important immunomodulator of CD4+ cells in normal and pathological conditions, which exerts its anti-inflammatory and immunomodulatory actions through VPAC receptors, VPAC1 and VPAC2. Only a decrease in the expression of VPAC1 mRNA on Th cells upon activation has been reported. Thus, the deepening in the knowledge of the behavior of these receptors may contribute to the design of new therapies based on their activation and/or blockade. In this study, we describe the expression pattern, cellular location and functional role of VIP receptors during the activation of human Th cells in healthy conditions and in early arthritis (EA). The protein expression pattern of VPAC1 did not change with the activation of Th lymphocytes, whereas VPAC2 was up-regulated. In resting cells, VPAC1 was located on the plasma membrane and nucleus, whereas it only appeared in the nucleus in activated cells. VPAC2 was always found in plasma membrane location. VIP receptors signaled through a PKA-dependent pathway in both conditions, and also by a PKA-independent pathway in activated cells. Both receptors exhibit a potent immunomodulatory capacity by controlling the pathogenic profile and the activation markers of Th cells. These results highlight a novel translational view in inflammatory/autoimmune diseases.
Collapse
Affiliation(s)
- R Villanueva-Romero
- Departamento de Biología Celular, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - I Gutiérrez-Cañas
- Departamento de Biología Celular, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - M Carrión
- Departamento de Biología Celular, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - I González-Álvaro
- Servicio de Reumatología, Instituto de Investigación Sanitaria Hospital La Princesa (IIS-IP), Madrid, Spain
| | - J M Rodríguez-Frade
- Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - M Mellado
- Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - C Martínez
- Departamento de Biología Celular, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - R P Gomariz
- Departamento de Biología Celular, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Y Juarranz
- Departamento de Biología Celular, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain.
| |
Collapse
|
15
|
Gomariz RP, Juarranz Y, Carrión M, Pérez-García S, Villanueva-Romero R, González-Álvaro I, Gutiérrez-Cañas I, Lamana A, Martínez C. An Overview of VPAC Receptors in Rheumatoid Arthritis: Biological Role and Clinical Significance. Front Endocrinol (Lausanne) 2019; 10:729. [PMID: 31695683 PMCID: PMC6817626 DOI: 10.3389/fendo.2019.00729] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 10/09/2019] [Indexed: 12/24/2022] Open
Abstract
The axis comprised by the Vasoactive Intestinal Peptide (VIP) and its G protein-coupled receptors (GPCRs), VPAC1, and VPAC2, belong to the B1 family and signal through Gs or Gq proteins. VPAC receptors seem to preferentially interact with Gs in inflammatory cells, rather than Gq, thereby stimulating adenylate cyclase activity. cAMP is able to trigger various downstream pathways, mainly the canonical PKA pathway and the non-canonical cAMP-activated guanine nucleotide exchange factor (EPAC) pathway. Classically, the presence of VPACs has been confined to the plasma membrane; however, VPAC1 location has been described in the nuclear membrane in several cell types such as activated Th cells, where they are also functional. VPAC receptor signaling modulates a number of biological processes by tipping the balance of inflammatory mediators in macrophages and other innate immune cells, modifying the expression of TLRs, and inhibiting MMPs and the expression of adhesion molecules. Receptor signaling also downregulates coagulation factors and acute-phase proteins, promotes Th2 over Th1, stimulates Treg abundance, and finally inhibits a pathogenic Th17 profile. Thus, the VIP axis signaling regulates both the innate and adaptive immune responses in several inflammatory/autoimmune diseases. Rheumatoid arthritis (RA) is a complex autoimmune disease that develops on a substrate of genetically susceptible individuals and under the influence of environmental factors, as well as epigenetic mechanisms. It is a heterogeneous disease with different pathogenic mechanisms and variable clinical forms between patients with the same diagnosis. The knowledge of VIP signaling generated in both animal models and human ex vivo studies can potentially be translated to clinical reality. Most recently, the beneficial effects of nanoparticles of VIP self-associated with sterically stabilized micelles have been reported in a murine model of RA. Another novel research area is beginning to define the receptors as biomarkers in RA, with their expression levels shown to be associated with the activity of the disease and patients-reported impairment. Therefore, VPAC expression together VIP genetic variants could allow patients to be stratified at the beginning of the disease with the purpose of guiding personalized treatment decisions.
Collapse
Affiliation(s)
- Rosa P. Gomariz
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
- *Correspondence: Rosa P. Gomariz
| | - Yasmina Juarranz
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Mar Carrión
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Selene Pérez-García
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Raúl Villanueva-Romero
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Isidoro González-Álvaro
- Servicio de Reumatología, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria la Princesa (IIS-IP), Madrid, Spain
| | - Irene Gutiérrez-Cañas
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Amalia Lamana
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
- Servicio de Reumatología, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria la Princesa (IIS-IP), Madrid, Spain
| | - Carmen Martínez
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
16
|
Hara M, Takeba Y, Iiri T, Ohta Y, Ootaki M, Watanabe M, Watanabe D, Koizumi S, Otsubo T, Matsumoto N. Vasoactive intestinal peptide increases apoptosis of hepatocellular carcinoma by inhibiting the cAMP/Bcl-xL pathway. Cancer Sci 2018; 110:235-244. [PMID: 30390393 PMCID: PMC6317926 DOI: 10.1111/cas.13861] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 10/25/2018] [Accepted: 11/01/2018] [Indexed: 12/12/2022] Open
Abstract
Vasoactive intestinal peptide (VIP) is a modulator of inflammatory responses. VIP receptors are expressed in several tumor types, such as colorectal carcinoma. The study described herein was conducted to confirm the presence of VIP and its receptors (VPAC1 and VPAC2) in surgically resected hepatocellular carcinoma (HCC) tissues and in the HCC cell line Huh7. The mechanism responsible for apoptosis of HCC cells was then examined because VIP treatment (10-10 M) significantly suppressed proliferation of Huh7 cells. In examining apoptosis-related proteins, we found caspase-3 to be significantly increased and Bcl-xL and cyclic AMP (cAMP) response element-binding protein (CREB) to be significantly decreased in Huh7 cells cultured with VIP. Furthermore, the CREB level and phosphorylation were reduced. These effects were reversed by the addition of VIP receptor antagonist or cAMP antagonist Rp-cAMPS. Pretreatment with cAMP analogue blocked the increased apoptosis, suggesting that VIP induces apoptosis via a PKA-independent signaling mechanism. Our data indicate that VIP prevents the progression of HCC by apoptosis through the cAMP/Bcl-xL pathway.
Collapse
Affiliation(s)
- Masaki Hara
- Division of Gastroenterology and Hepatology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Yuko Takeba
- Department of Pharmacology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Taroh Iiri
- Department of Pharmacology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Yuki Ohta
- Department of Pharmacology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Masanori Ootaki
- Department of Pharmacology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Minoru Watanabe
- Experimental Animals Institution, St. Marianna University Graduate School of Medicine, Kawasaki, Japan
| | - Daiki Watanabe
- Department of Pharmacology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Satoshi Koizumi
- Division of Gastroenterological Surgery, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Takehito Otsubo
- Division of Gastroenterological Surgery, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Naoki Matsumoto
- Department of Pharmacology, St. Marianna University School of Medicine, Kawasaki, Japan
| |
Collapse
|
17
|
Dubuc C, Savard M, Bovenzi V, Lessard A, Côté J, Neugebauer W, Geha S, Chemtob S, Gobeil F. Antitumor activity of cell-penetrant kinin B1 receptor antagonists in human triple-negative breast cancer cells. J Cell Physiol 2018; 234:2851-2865. [PMID: 30132865 DOI: 10.1002/jcp.27103] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 06/28/2018] [Indexed: 12/26/2022]
Abstract
High nuclear expression of G protein-coupled receptors, including kinin B1 receptors (B1R), has been observed in several human cancers, but the clinical significance of this is unknown. We put forward the hypothesis that these "nuclearized" kinin B1R contribute to tumorigenicity and can be a new target in anticancer strategies. Our initial immunostaining and ultrastructural electron microscopy analyses demonstrated high B1R expression predominantly located at internal/nuclear compartments in the MDA-MB-231 triple-negative breast cancer (TNBC) cell line as well as in clinical samples of patients with TNBC. On the basis of these findings, in the present study, we evaluated the anticancer therapeutic potential of newly identified, cell-permeable B1R antagonists in MDA-MB-231 cells (ligand-receptor binding/activity assays and LC-MS/MS analyses). We found that these compounds (SSR240612, NG67, and N2000) were more toxic to MDA-MB-231 cells in comparison with low- or non-B1R expressing MCF-10A normal human mammary epithelial cells and COS-1 cells, respectively (clonogenic, MTT proliferative/cytocidal assays, and fluorescence-activated cell-sorting (FACS)-based apoptosis analyses). By comparison, the peptide B1R antagonist R954 unable to cross cell membrane failed to produce anticancer effects. Furthermore, the putative mechanisms underlying the anticancer activities of cell-penetrant B1R antagonists were assessed by analyzing cell cycle regulation and signaling molecules related to cell survival and apoptosis (FACS and western blot). Finally, drug combination experiments showed that cell-penetrant B1R antagonists can cooperate with suboptimal doses of chemotherapeutic agents (doxorubicin and paclitaxel) to promote TNBC death. This study provides evidence on the potential value of internally acting kinin B1R antagonists in averting growth of breast cancer.
Collapse
Affiliation(s)
- Céléna Dubuc
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Institute of Pharmacology, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Martin Savard
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Institute of Pharmacology, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Veronica Bovenzi
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Institute of Pharmacology, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Andrée Lessard
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jérôme Côté
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Institute of Pharmacology, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Witold Neugebauer
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Institute of Pharmacology, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Sameh Geha
- Department of Pathology, Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Québec, Canada
| | - Sylvain Chemtob
- Department of Pediatrics, Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Center, Montréal, Québec, Canada
| | - Fernand Gobeil
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Institute of Pharmacology, Université de Sherbrooke, Sherbrooke, Québec, Canada
| |
Collapse
|
18
|
Yu R, Liu H, Peng X, Cui Y, Song S, Wang L, Zhang H, Hong A, Zhou T. The palmitoylation of the N-terminal extracellular Cys37 mediates the nuclear translocation of VPAC1 contributing to its anti-apoptotic activity. Oncotarget 2018; 8:42728-42741. [PMID: 28473666 PMCID: PMC5522101 DOI: 10.18632/oncotarget.17449] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 04/14/2017] [Indexed: 12/17/2022] Open
Abstract
VPAC1 is class B G protein-coupled receptors (GPCR) shared by pituitary adenylate cyclase activating polypeptide (PACAP) and vasoactive intestinal peptide (VIP). The first cysteine (Cys37) in the N-terminal extracellular domain of mature VPAC1 is a free Cys not involved in the formation of conserved intramolecular disulfide bonds. In order to investigate the biological role of this Cys37 in VPAC1, the wild-type VPAC1 and Cys37/Ala mutant (VPAC1-C37/A) were expressed stably as fusion proteins with enhanced yellow fluorescent protein (EYFP) respectively in Chinese hamster ovary (CHO) cells. Both VPAC1-EYFP and VPAC1-C37/A-EYFP trafficked to the plasma membrane normally, and CHO cells expressing VPAC1-EYFP displayed higher anti-apoptotic activity against camptothecin (CPT) induced apoptosis than the cells expressing VPAC1-C37/A-EYFP, while VPAC1-C37/A-CHO cells showed higher proliferative activity than VPAC1-CHO cells. Confocal microscopic analysis, western blotting and fluorescence quantification assay showed VPAC1-EYFP displayed significant nuclear translocation while VPAC1-C37/A-EYFP did not transfer into nucleus under the stimulation of VIP (0.1 nM). Acyl-biotin exchange assay and click chemistry-based palmitoylation assay confirmed for the first time the palmitoylation of Cys37, which has been predicted by bioinformatics analysis. And the palmitoylation inhibitor 2-bromopalmitate significantly inhibited the nuclear translocation of VPAC1-EYFP and its anti-apoptotic activity synchronously. These results indicated the palmitoylation of the Cys37 in the N-terminal extracellular domain of VPAC1 mediates the nuclear translocation of VPAC1 contributing to its anti-apoptotic activity. These findings reveal for the first time the lipidation-mediating nuclear translocation of VPAC1 produces a novel anti-apoptotic signal pathway, which may help to promote new drug development strategy targeting VPAC1.
Collapse
Affiliation(s)
- Rongjie Yu
- Institute of Biomedicine, School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China.,National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Hongyu Liu
- Institute of Biomedicine, School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China.,National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Xinhe Peng
- Institute of Biomedicine, School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China.,National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Yue Cui
- Institute of Biomedicine, School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China.,National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Suqin Song
- Institute of Biomedicine, School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China.,National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Like Wang
- Institute of Biomedicine, School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China.,National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Huahua Zhang
- Department of Medical Genetics, Guangdong Medical University, Dongguan, Guangdong, China
| | - An Hong
- Institute of Biomedicine, School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China.,National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Tianhong Zhou
- Department of Bioengineering, School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
19
|
Dubuc C, Savard M, Bovenzi V, Lessard A, Fortier A, Côté J, Neugebauer W, Rizzolio F, Geha S, Giordano A, Chemtob S, Gobeil F. Targeting intracellular B2 receptors using novel cell-penetrating antagonists to arrest growth and induce apoptosis in human triple-negative breast cancer. Oncotarget 2018. [PMID: 29515778 PMCID: PMC5839409 DOI: 10.18632/oncotarget.24009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are integral cell-surface proteins having a central role in tumor growth and metastasis. However, several GPCRs retain an atypical intracellular/nuclear location in various types of cancer. The pathological significance of this is currently unknown. Here we extend this observation by showing that the bradykinin B2R (BK-B2R) is nuclearly expressed in the human triple-negative breast cancer (TNBC) cell line MDA-MB-231 and in human clinical specimens of TNBC. We posited that these “nuclearized” receptors could be involved in oncogenic signaling linked to aberrant growth and survival maintenance of TNBC. We used cell-penetrating BK-B2R antagonists, including FR173657 and novel transducible, cell-permeable forms of the peptide B2R antagonist HOE 140 (NG68, NG134) to demonstrate their superior efficacy over impermeable ones (HOE 140), in blocking proliferation and promoting apoptosis of MDA-MB-231 cells. Some showed an even greater antineoplastic activity over conventional chemotherapeutic drugs in vitro. The cell-permeable B2R antagonists had less to no anticancer effects on B2R shRNA-knockdown or non-B2R expressing (COS-1) cells, indicating specificity in their action. Possible mechanisms of their anticancer effects may involve activation of p38kinase/p27Kip1 pathways. Together, our data support the existence of a possible intracrine signaling pathway via internal/nuclear B2R, critical for the growth of TNBC cells, and identify new chemical entities that enable to target the corresponding intracellular GPCRs.
Collapse
Affiliation(s)
- Céléna Dubuc
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada.,Institute of Pharmacology, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Martin Savard
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada.,Institute of Pharmacology, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Veronica Bovenzi
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada.,Institute of Pharmacology, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Andrée Lessard
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Audrey Fortier
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada.,Institute of Pharmacology, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Jérôme Côté
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada.,Institute of Pharmacology, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Witold Neugebauer
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada.,Institute of Pharmacology, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Flavio Rizzolio
- Department of Biology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA, USA.,Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari Venezia, Mestre-Venezia, Italy
| | - Sameh Geha
- Department of Pathology, Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Québec, Canada
| | - Antonio Giordano
- Department of Biology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA, USA
| | - Sylvain Chemtob
- Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, Québec, Canada
| | - Fernand Gobeil
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada.,Institute of Pharmacology, Université de Sherbrooke, Sherbrooke, Québec, Canada
| |
Collapse
|
20
|
Yamamoto H, Okada R, Tanaka R, Unno K, Iguchi K. Expression of a urokinase-type plasminogen activator during tumor growth leads to angiogenesis via galanin activation in tumor-bearing mice. FEBS Open Bio 2017; 7:1784-1792. [PMID: 29123986 PMCID: PMC5666387 DOI: 10.1002/2211-5463.12318] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 08/18/2017] [Accepted: 09/20/2017] [Indexed: 02/03/2023] Open
Abstract
Small-cell lung carcinoma releases progalanin. The released progalanin is activated via a nonclassical processing pathway, being processed into an active form of galanin (1-20) by plasmin in extracellular components. Plasmin is produced from plasminogen activators. To clarify the regulation of progalanin via plasminogen activation by urokinase and tissue-plasminogen activator (t-PA), we investigated the regulation mechanism for urokinase and t-PA expression and their effect on galanin activation. Additionally, we studied the effect of activated galanin on angiogenesis. To determine the effect of cell density, we measured the expression levels of urokinase and t-PA using real-time PCR and plasminogen/gelatin zymography in a cell culture. The urokinase expression increased under both high cell density and presence of cell membrane fractions. However, urokinase increments induced by conditioned medium were low. These results indicate that expression of plasminogen activators is regulated by cell membrane factors. We used tumor-bearing mice to clarify the expression of plasminogen activators and galanin activation. Real-time PCR showed that urokinase was substantially higher in the central parts of tumors compared to the periphery, and this was confirmed by plasminogen/gelatin zymography. To evaluate the biological effect of plasminogen activators on tumor growth, we used tranexamic acid as a plasminogen inhibitor. Tranexamic acid decreased galanin (1-20) and the hemoglobin content of tumors and suppressed tumor growth. Additionally, galanin had no effect on the hemoglobin content of tumors derived from cells lacking GALR2. These results demonstrate the regulation of urokinase expression in tumors through progalanin activation in extracellular compartments, and confirm that galanin plays a role in angiogenesis.
Collapse
Affiliation(s)
- Hiroyuki Yamamoto
- Nihon Pharmaceutical University Ina-machi, Kitaadachi-gun Japan.,Laboratory of Bioorganic Chemistry School of Pharmaceutical Sciences University of Shizuoka Japan
| | - Rina Okada
- Laboratory of Bioorganic Chemistry School of Pharmaceutical Sciences University of Shizuoka Japan
| | - Rika Tanaka
- Nihon Pharmaceutical University Ina-machi, Kitaadachi-gun Japan
| | - Keiko Unno
- Laboratory of Bioorganic Chemistry School of Pharmaceutical Sciences University of Shizuoka Japan
| | - Kazuaki Iguchi
- Laboratory of Bioorganic Chemistry School of Pharmaceutical Sciences University of Shizuoka Japan
| |
Collapse
|
21
|
Thomas RJ, Kenfield SA, Jimenez A. Exercise-induced biochemical changes and their potential influence on cancer: a scientific review. Br J Sports Med 2017; 51:640-644. [PMID: 27993842 PMCID: PMC5466928 DOI: 10.1136/bjsports-2016-096343] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2016] [Indexed: 12/20/2022]
Abstract
AIM To review and discuss the available international literature regarding the indirect and direct biochemical mechanisms that occur after exercise, which could positively, or negatively, influence oncogenic pathways. METHODS The PubMed, MEDLINE, Embase and Cochrane libraries were searched for papers up to July 2016 addressing biochemical changes after exercise with a particular reference to cancer. The three authors independently assessed their appropriateness for inclusion in this review based on their scientific quality and relevance. RESULTS 168 papers were selected and categorised into indirect and direct biochemical pathways. The indirect effects included changes in vitamin D, weight reduction, sunlight exposure and improved mood. The direct effects included insulin-like growth factor, epigenetic effects on gene expression and DNA repair, vasoactive intestinal peptide, oxidative stress and antioxidant pathways, heat shock proteins, testosterone, irisin, immunity, chronic inflammation and prostaglandins, energy metabolism and insulin resistance. SUMMARY Exercise is one of several lifestyle factors known to lower the risk of developing cancer and is associated with lower relapse rates and better survival. This review highlights the numerous biochemical processes, which explain these potential anticancer benefits.
Collapse
Affiliation(s)
| | - Stacey A Kenfield
- Department of Urology, University of California, San Francisco, California, USA
| | - Alfonso Jimenez
- Centre for Applied Biological and Exercise Sciences, Faculty of Health and Life Sciences, Coventry University, Coventry, UK
| |
Collapse
|
22
|
Dissanayake S, Denny WA, Gamage S, Sarojini V. Recent developments in anticancer drug delivery using cell penetrating and tumor targeting peptides. J Control Release 2017; 250:62-76. [DOI: 10.1016/j.jconrel.2017.02.006] [Citation(s) in RCA: 175] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Revised: 02/02/2017] [Accepted: 02/02/2017] [Indexed: 12/13/2022]
|
23
|
Bhosle VK, Rivera JC, Chemtob S. New insights into mechanisms of nuclear translocation of G-protein coupled receptors. Small GTPases 2017; 10:254-263. [PMID: 28125336 DOI: 10.1080/21541248.2017.1282402] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The G-protein coupled receptor (GPCR) signaling was long believed to involve activation of receptor exclusively at the cell surface, followed by its binding to heterotrimeric G-proteins and arrestins to trigger various intracellular signaling cascades, and termination of signaling by internalization of the receptor. It is now accepted that many GPCRs continue to signal after internalization in the endosomes. Since the breakthrough discoveries of nuclear binding sites for their ligands in 1980s, several GPCRs have been detected at cell nuclei. But mechanisms of nuclear localization of GPCRs, many of whom contain putative nuclear localization signals, remain poorly understood to date. Nevertheless, it is known that subcellular trafficking of GPCRs is regulated by members of Ras superfamily of small GTPases, most notably by Rab and Arf GTPases. In this commentary, we highlight several recent studies which suggest novel roles of small GTPases, importins and sorting nexin proteins in the nuclear translocation of GPCRs via vesicular transport pathways. Taken together with increasing evidence for in vivo functionality of the nuclear GPCRs, better understanding of their trafficking will provide valuable clues in cell biology.
Collapse
Affiliation(s)
- Vikrant K Bhosle
- a Department of Pharmacology and Therapeutics , McGill University , Montréal , Québec , Canada.,b CHU Sainte-Justine Hospital Research Centre , University of Montréal , Montréal , Québec , Canada.,c Maisonneuve-Rosemont Hospital Research Centre , University of Montréal , Montréal , Québec , Canada.,e Cell Biology Program , Peter Gilgan Centre for Research and Learning , Toronto , Ontario , Canada
| | - José Carlos Rivera
- b CHU Sainte-Justine Hospital Research Centre , University of Montréal , Montréal , Québec , Canada.,c Maisonneuve-Rosemont Hospital Research Centre , University of Montréal , Montréal , Québec , Canada
| | - Sylvain Chemtob
- a Department of Pharmacology and Therapeutics , McGill University , Montréal , Québec , Canada.,b CHU Sainte-Justine Hospital Research Centre , University of Montréal , Montréal , Québec , Canada.,c Maisonneuve-Rosemont Hospital Research Centre , University of Montréal , Montréal , Québec , Canada.,d Departments of Pediatrics, Ophthalmology and Pharmacology , University of Montréal , Montréal , Québec , Canada
| |
Collapse
|
24
|
Trabulsi EJ, Tripathi SK, Gomella L, Solomides C, Wickstrom E, Thakur ML. Development of a voided urine assay for detecting prostate cancer non-invasively: a pilot study. BJU Int 2017; 119:885-895. [PMID: 28075510 DOI: 10.1111/bju.13775] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To validate a hypothesis that prostate cancer can be detected non-invasively by a simple and reliable assay by targeting genomic VPAC receptors expressed on malignant prostate cancer cells shed in voided urine. PATIENTS/SUBJECTS AND METHODS VPAC receptors were targeted with a specific biomolecule, TP4303, developed in our laboratory. With an Institutional Review Board exempt approval of use of de-identified discarded samples, an aliquot of urine collected as a standard of care, from patients presenting to the urology clinic (207 patients, 176 men and 31 women, aged ≥21 years) was cytospun. The cells were fixed and treated with TP4303 and 4,6-diamidino-2-phenylindole (DAPI). The cells were then observed under a microscope and cells with TP4303 orange fluorescence around the blue (DAPI) nucleus were considered 'malignant' and those only with a blue nucleus were regarded as 'normal'. VPAC presence was validated using receptor blocking assay and cell malignancy was confirmed by prostate cancer gene profile examination. RESULTS The urine specimens were labelled only with gender and presenting diagnosis, with no personal health identifiers or other clinical data. The assay detected VPAC positive cells in 98.6% of the men with a prostate cancer diagnosis (141), and none of the 10 men with benign prostatic hyperplasia. Of the 56 'normal' patients, 62.5% (35 patients, 10 men and 25 women) were negative for VPAC cells; 19.6% (11, 11 men and no women) had VPAC positive cells; and 17.8% (10, four men and six women) were uninterpretable due to excessive crystals in the urine. Although data are limited, the sensitivity of the assay was 99.3% with a confidence interval (CI) of 96.1-100% and the specificity was 100% with a CI of 69.2-100%. Receptor blocking assay and fluorescence-activated cell sorting (FACS) analyses demonstrated the presence of VPAC receptors and gene profiling examinations confirmed that the cells expressing VPAC receptors were malignant prostate cancer cells. CONCLUSION These preliminary data are highly encouraging and warrant further evaluation of the assay to serve as a simple and reliable tool to detect prostate cancer non-invasively.
Collapse
Affiliation(s)
- Edouard J Trabulsi
- Department of Urology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Sushil K Tripathi
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Leonard Gomella
- Department of Urology, Thomas Jefferson University, Philadelphia, PA, USA.,Department of Pathology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Charalambos Solomides
- Departments of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Eric Wickstrom
- Department of Pathology, Thomas Jefferson University, Philadelphia, PA, USA.,The Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Mathew L Thakur
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA.,Department of Pathology, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
25
|
Wierzbicka JM, Żmijewski MA, Antoniewicz J, Sobjanek M, Slominski AT. Differentiation of Keratinocytes Modulates Skin HPA Analog. J Cell Physiol 2016; 232:154-66. [PMID: 27061711 DOI: 10.1002/jcp.25400] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Accepted: 04/04/2016] [Indexed: 12/16/2022]
Abstract
It is well established, that epidermal keratinocytes express functional equivalent of hypothalamus-pituitary-adrenal axis (HPA) in order to respond to changing environment and maintain internal homeostasis. We are presenting data indicating that differentiation of primary neonatal human keratinocytes (HPEKp), induced by prolonged incubation or calcium is accompanied by significant changes in the expression of the elements of skin analog of HPA (sHPA). Expression of CRF, UCN1-3, POMC, ACTH, CRFR1, CRFR2, MC1R, MC2R, and GR (coded by NR3C1 gene) were observed on gene/protein levels along differentiation of keratinocytes in culture with similar pattern seen by immunohistochemistry on full thickness skin biopsies. Expression of CRF was more pronounced in less differentiated keratinocytes, which corresponded to the detection of CRF immunoreactivity preferentially in the stratum basale. POMC expression was enhanced in more differentiated keratinocytes, which corresponded to detection of ACTH immunoreactivity, predominantly in the stratum spinosum and stratum granulosum. Expression of urocortins was also affected by induction of HPEKp differentiation. Immunohistochemical studies showed high prevalence of CRFR1 in well differentiated keratinocytes, while smaller keratinocytes showed predominantly CRFR2 immunoreactivity. MC2R mRNA levels were elevated from days 4 to 8 of in vitro incubation, while MC2R immunoreactivity was the highest in the upper layers of epidermis. Similar changes in mRNA/protein levels of sHPA elements were observed in HPEKp keratinocytes treated with calcium. Summarizing, preferential expression of CRF and POMC (ACTH) by populations of keratinocytes on different stage of differentiation resembles organization of central HPA axis suggesting their distinct role in physiology and pathology of the epidermis. J. Cell. Physiol. 232: 154-166, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | | | - Jakub Antoniewicz
- Department of Histology, Medical University of Gdańsk, Gdańsk, Poland
| | - Michal Sobjanek
- Department of Dermatology, Venereology and Allergology, Medical University of Gdańsk, Gdańsk, Poland
| | - Andrzej T Slominski
- Department of Dermatology, University of Alabama Birmingham, Birmingham, Alabama.,VA Medical Center, Birmingham, Alabama
| |
Collapse
|
26
|
VPAC1 Targeted (64)Cu-TP3805 Positron Emission Tomography Imaging of Prostate Cancer: Preliminary Evaluation in Man. Urology 2015; 88:111-8. [PMID: 26519886 DOI: 10.1016/j.urology.2015.10.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 09/17/2015] [Accepted: 10/08/2015] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To evaluate (64)Cu-TP3805 as a novel biomolecule, to positron emission tomography (PET) image prostate cancer (PC), at the onset of which VPAC1, the superfamily of G protein-coupled receptors, is expressed in high density on PC cells, but not on normal cells. MATERIALS AND METHODS Twenty-five patients undergoing radical prostatectomy were PET/X-ray computerized tomography imaged preoperatively with (64)Cu-TP3805. Standardized maximum uptake (SUVmax) values were determined and malignant lesions (standardized uptake value > 1.0) counted, and compared with histologic findings. Whole-mount pathology slides from 6 VPAC1 PET imaged patients, 3 benign prostatic hyperplasia patients, 1 malignant and 1 benign lymph node underwent digital autoradiography (DAR) after (64)Cu-TP3805 incubation and were compared to hematoxylin- and eosin-stained slides. RESULTS In 25 patients who underwent PET imaging, 212 prostate gland lesions had SUVmax > 1.0 vs 127 lesions identified by histology of biopsy tissues. The status of the additional 85 PET identified prostate lesions remains to be determined. In 68 histologic slides from 6 PET imaged patients, DAR identified 105 of 107 PC foci, 19 of 19 high-grade prostatic intraepithelial neoplasias, and ejaculatory ducts and verumontanum involved with cancer. Additionally, DAR found 9 PC lesions not previously identified histologically. The positive and negative lymph nodes were correctly identified, and in 3 of 3 benign prostatic hyperplasia patients and 5 of 5 cysts, DAR was negative. CONCLUSION This feasibility study demonstrated that (64)Cu-TP3805 delineates PC in vivo and ex vivo, provided normal images for benign masses, and is worthy of further studies.
Collapse
|
27
|
Capomaccio S, Milanesi M, Bomba L, Cappelli K, Nicolazzi EL, Williams JL, Ajmone-Marsan P, Stefanon B. Searching new signals for production traits through gene-based association analysis in three Italian cattle breeds. Anim Genet 2015; 46:361-70. [DOI: 10.1111/age.12303] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2015] [Indexed: 12/12/2022]
Affiliation(s)
- Stefano Capomaccio
- Istituto di Zootecnica; UCSC; via Emilia Parmense 84 29122 Piacenza Italy
| | - Marco Milanesi
- Istituto di Zootecnica; UCSC; via Emilia Parmense 84 29122 Piacenza Italy
| | - Lorenzo Bomba
- Istituto di Zootecnica; UCSC; via Emilia Parmense 84 29122 Piacenza Italy
| | - Katia Cappelli
- Dipartimento di Medicina Veterinaria; Università di Perugia; Via San Costanzo 4 06100 Perugia Italy
| | | | - John L. Williams
- Parco Tecnologico Padano; Via Einstein; Loc. Cascina Codazza 26900 Lodi Italy
| | | | - Bruno Stefanon
- Dipartimento di Scienze Agrarie e Ambientali; Università di Udine; via delle Scienze 206-33100 Udine Italy
| |
Collapse
|
28
|
Gülçür E, Thaqi M, Khaja F, Kuzmis A, Önyüksel H. Curcumin in VIP-targeted sterically stabilized phospholipid nanomicelles: a novel therapeutic approach for breast cancer and breast cancer stem cells. Drug Deliv Transl Res 2015; 3. [PMID: 24363979 DOI: 10.1007/s13346-013-0167-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Breast cancer is a leading cause of cancer deaths among women in the US, with 40 % chance of relapse after treatment. Recent studies outline the role of cancer stem cells (CSCs) in tumor initiation, propagation, and regeneration of cancer. Moreover, it has been established that breast CSCs reside in a quiescent state that makes them more resistant to conventional cancer therapies than bulk cancer cells resulting in tumor relapse. In this study, we establish that CSCs are associated with the overexpression of vasoactive intestinal peptide (VIP) receptors which can be used to actively target these cells. We investigated the potential of using a novel curcumin nanomedicine (C-SSM) surface conjugated with VIP to target and hinder breast cancer with CSCs. Here, we formulated, characterized, and evaluated the feasibility of C-SSM nanomedicine in vitro. We investigated the cytotoxicity of C-SSM on breast cancer cells and CSCs by tumorsphere formation assay. Our results suggest that curcumin can be encapsulated in SSM up to 200 μg/ml with 1 mM lipid concentration. C-SSM nanomedicine is easy to prepare and maintains its original physicochemical properties after lyophilization, with an IC50 that is significantly improved from that of free curcumin (14.2±1.2 vs. 26.1±3.0 μM). Furthermore, C-SSM-VIP resulted in up to 20 % inhibition of tumorsphere formation at a dose of 5 μM. To this end, our findings demonstrate the feasibility of employing our actively targeted nanomedicine as a potential therapy for CSCs-enriched breast cancer.
Collapse
Affiliation(s)
- Ece Gülçür
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Mentor Thaqi
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Fatima Khaja
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Antonina Kuzmis
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Hayat Önyüksel
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA; Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA; Department of Biopharmaceutical Sciences (M/C 865) College of Pharmacy, University of Illinois at Chicago, 833 South Wood St., Chicago, IL 60612-7231, USA
| |
Collapse
|
29
|
Schulz S, Mann A, Novakhov B, Piggins HD, Lupp A. VPAC2 receptor expression in human normal and neoplastic tissues: evaluation of the novel MAB SP235. Endocr Connect 2015; 4:18-26. [PMID: 25504760 PMCID: PMC4285768 DOI: 10.1530/ec-14-0051] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The vasoactive intestinal peptide receptor 2 (VPAC2) is widely distributed throughout the body and is also overexpressed in a variety of human neoplastic tissues. However, little is known about its precise tissue distribution, regulation and function, which is in part be due to the lack of specific monoclonal anti-VPAC2 antibodies. In this study, we extensively characterised the novel rabbit monoclonal anti-VPAC2 antibody (clone SP235) using transfected cells and mouse, rat and human tissues. SP235 was then subjected to a comparative immunohistochemical study on a series of 167 histological specimens from formalin-fixed, paraffin-embedded human tumours and adjacent normal tissues. SP235 detected a broad band migrating at a molecular weight of 50-70 kDa in western blotting analyses of various mouse tissues as well as VPAC2- but not VPAC1-transfected human embryonic kidney 293 cells. SP235 yielded an efficient immunostaining of distinct cell populations in human tissue samples with a predominance of plasma membrane staining, which was completely abolished by preadsorption with its immunising peptide. SP235 immunohistochemistry detected VPAC2 receptors in lymphocytes present in spleen, tonsils, lymph nodes and Peyer's patches, chief cells of gastric mucosa, exocrine and endocrine pancreas, kidney tubules and blood vessels. In addition, VPAC2 was observed in thyroid, gastric and lung carcinomas, pancreatic adenocarcinomas, sarcomas and neuroendocrine tumours. SP235 may prove of great value in the identification of VPAC2 receptors during routine histopathological examination. VPAC2 visualisation with this simple and rapid immunohistochemical method will facilitate identification of candidate tumours for vasoactive intestinal peptide (VIP)-based diagnostics or therapeutic interventions.
Collapse
Affiliation(s)
- Stefan Schulz
- Institute of Pharmacology and ToxicologyJena University Hospital, Friedrich Schiller University Jena, Drackendorfer Straße 1, D-07747 Jena, GermanyFaculty of Life SciencesUniversity of Manchester, Manchester M13 9PT, UK
| | - Anika Mann
- Institute of Pharmacology and ToxicologyJena University Hospital, Friedrich Schiller University Jena, Drackendorfer Straße 1, D-07747 Jena, GermanyFaculty of Life SciencesUniversity of Manchester, Manchester M13 9PT, UK
| | - Benjamin Novakhov
- Institute of Pharmacology and ToxicologyJena University Hospital, Friedrich Schiller University Jena, Drackendorfer Straße 1, D-07747 Jena, GermanyFaculty of Life SciencesUniversity of Manchester, Manchester M13 9PT, UK
| | - Hugh D Piggins
- Institute of Pharmacology and ToxicologyJena University Hospital, Friedrich Schiller University Jena, Drackendorfer Straße 1, D-07747 Jena, GermanyFaculty of Life SciencesUniversity of Manchester, Manchester M13 9PT, UK
| | - Amelie Lupp
- Institute of Pharmacology and ToxicologyJena University Hospital, Friedrich Schiller University Jena, Drackendorfer Straße 1, D-07747 Jena, GermanyFaculty of Life SciencesUniversity of Manchester, Manchester M13 9PT, UK
| |
Collapse
|
30
|
Fernández-Martínez AB, Carmena MJ, Bajo AM, Vacas E, Sánchez-Chapado M, Prieto JC. VIP induces NF-κB1-nuclear localisation through different signalling pathways in human tumour and non-tumour prostate cells. Cell Signal 2014; 27:236-44. [PMID: 25446255 DOI: 10.1016/j.cellsig.2014.11.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 10/16/2014] [Accepted: 11/08/2014] [Indexed: 01/19/2023]
Abstract
The nuclear factor κB (NF-κB) is a powerful activator of angiogenesis, invasion and metastasis. Transactivation and nuclear localisation of NF-κB is an index of recurrence in prostate cancer. Vasoactive intestinal peptide (VIP) exerts similar effects in prostate cancer models involving increased expression of vascular endothelial growth factor (VEGF) and cyclooxygenase-2 (COX-2) which are related to NF-κB transactivation. Here we studied differential mechanisms of VIP-induced NF-κB transactivation in non-tumour RWPE-1 and tumour LNCaP and PC3 human prostate epithelial cells. Immunofluorescence studies showed that VIP increases translocation of the p50 subunit of NF-κB1 to the nucleus, an effect that was inhibited by curcumin. The signalling transduction pathways involved are different depending on cell transformation degree. In control cells (RWPE1), the effect is mediated by protein kinase A (PKA) activation and does not implicate extracellular signal-regulated kinase (ERK) or phosphoinositide 3-kinase (PI3-K) pathways whereas the opposite is true in tumour LNCaP and PC3 cells. Exchange protein directly activated by cAMP (EPAC) pathway is involved in transformed cells but not in control cells. Curcumin blocks the activating effect of VIP on COX-2 promoter/prostaglandin E2 (PGE2) production and VEGF expression and secretion. The study incorporates direct observation on COX-2 promoter and suggests that VIP effect on VEGF may be indirectly mediated by PGE2 after being synthesised by COX-2, thus amplifying the initial signal. We show that the signalling involved in VIP effects on VEGF is cAMP/PKA in non-tumour cells and cAMP/EPAC/ERK/PI3K in tumour cells which coincides with pathways mediating p50 nuclear translocation. Thus, VIP appears to use different pathways for NF-κB1 (p50) transactivation in prostate epithelial cells depending on whether they are transformed or not. Transformed cells depend on pro-survival and pro-proliferative signalling pathways involving ERK, PI3-K and cAMP/EPAC which supports the potential therapeutic value of these targets in prostate cancer.
Collapse
Affiliation(s)
- Ana B Fernández-Martínez
- Department of Systems Biology, Unit of Biochemistry and Molecular Biology, University of Alcalá, 28871 Alcalá de Henares, Spain
| | - María J Carmena
- Department of Systems Biology, Unit of Biochemistry and Molecular Biology, University of Alcalá, 28871 Alcalá de Henares, Spain
| | - Ana M Bajo
- Department of Systems Biology, Unit of Biochemistry and Molecular Biology, University of Alcalá, 28871 Alcalá de Henares, Spain
| | - Eva Vacas
- Department of Systems Biology, Unit of Biochemistry and Molecular Biology, University of Alcalá, 28871 Alcalá de Henares, Spain
| | - Manuel Sánchez-Chapado
- Department of Surgery and Medical and Social Sciences, University of Alcalá, 28871 Alcalá de Henares, Spain; Department of Urology, Príncipe de Asturias Hospital, 28871 Alcalá de Henares, Spain
| | - Juan C Prieto
- Department of Systems Biology, Unit of Biochemistry and Molecular Biology, University of Alcalá, 28871 Alcalá de Henares, Spain.
| |
Collapse
|
31
|
Barbarin A, Séité P, Godet J, Bensalma S, Muller JM, Chadéneau C. Atypical nuclear localization of VIP receptors in glioma cell lines and patients. Biochem Biophys Res Commun 2014; 454:524-30. [DOI: 10.1016/j.bbrc.2014.10.113] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 10/23/2014] [Indexed: 12/27/2022]
|
32
|
Huang S, Yee C, Ching T, Yu H, Garmire LX. A novel model to combine clinical and pathway-based transcriptomic information for the prognosis prediction of breast cancer. PLoS Comput Biol 2014; 10:e1003851. [PMID: 25233347 PMCID: PMC4168973 DOI: 10.1371/journal.pcbi.1003851] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 08/08/2014] [Indexed: 01/19/2023] Open
Abstract
Breast cancer is the most common malignancy in women worldwide. With the increasing awareness of heterogeneity in breast cancers, better prediction of breast cancer prognosis is much needed for more personalized treatment and disease management. Towards this goal, we have developed a novel computational model for breast cancer prognosis by combining the Pathway Deregulation Score (PDS) based pathifier algorithm, Cox regression and L1-LASSO penalization method. We trained the model on a set of 236 patients with gene expression data and clinical information, and validated the performance on three diversified testing data sets of 606 patients. To evaluate the performance of the model, we conducted survival analysis of the dichotomized groups, and compared the areas under the curve based on the binary classification. The resulting prognosis genomic model is composed of fifteen pathways (e.g. P53 pathway) that had previously reported cancer relevance, and it successfully differentiated relapse in the training set (log rank p-value = 6.25e-12) and three testing data sets (log rank p-value<0.0005). Moreover, the pathway-based genomic models consistently performed better than gene-based models on all four data sets. We also find strong evidence that combining genomic information with clinical information improved the p-values of prognosis prediction by at least three orders of magnitude in comparison to using either genomic or clinical information alone. In summary, we propose a novel prognosis model that harnesses the pathway-based dysregulation as well as valuable clinical information. The selected pathways in our prognosis model are promising targets for therapeutic intervention. With the increasing awareness of heterogeneity in breast cancers, better prediction of breast cancer prognosis is much needed early on for more personalized treatment and management. Towards this goal we propose in this study a novel pathway-based prognosis prediction model, which emphasizes on individualized pathway-based risk measurement using the pathway dysregulation score (PDS). In combination with the L1-LASSO penalized feature selection and the COX-Proportional Hazards regression model, we have identified fifteen cancer relevant pathways using the pathway-based genomic model that successfully differentiated the relapse in the training set as well as three diversified test sets. Moreover, given the debate whether higher-order representative features, such as GO sets, pathways and network modules are superior to the gene-level features in the genomic models, we demonstrate that pathway-based genomic models consistently performed better than gene-based models in all four data sets. Last but not least, we show strong evidence that models that combine genomic information with clinical information improves the prognosis prediction significantly, in comparison to models that use either genomic or clinical information alone.
Collapse
Affiliation(s)
- Sijia Huang
- Molecular Biosciences and Bioengineering Graduate Program, University of Hawaii at Manoa, Honolulu, Hawaii, United States of America
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, United States of America
| | - Cameron Yee
- Neurobiology Program of Biology Department, University of Washington, Seattle, Washington, United States of America
| | - Travers Ching
- Molecular Biosciences and Bioengineering Graduate Program, University of Hawaii at Manoa, Honolulu, Hawaii, United States of America
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, United States of America
| | - Herbert Yu
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, United States of America
| | - Lana X. Garmire
- Molecular Biosciences and Bioengineering Graduate Program, University of Hawaii at Manoa, Honolulu, Hawaii, United States of America
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, United States of America
- * E-mail:
| |
Collapse
|
33
|
Zhang YF, Xu QX, Liao LD, Xu XE, Wu JY, Shen J, Wu ZY, Shen JH, Li EM, Xu LY. κ-Opioid receptor in the nucleus is a novel prognostic factor of esophageal squamous cell carcinoma. Hum Pathol 2013; 44:1756-65. [DOI: 10.1016/j.humpath.2012.11.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 11/28/2012] [Accepted: 11/30/2012] [Indexed: 01/06/2023]
|
34
|
Vacas E, Arenas MI, Muñoz-Moreno L, Bajo AM, Sánchez-Chapado M, Prieto JC, Carmena MJ. Antitumoral effects of vasoactive intestinal peptide in human renal cell carcinoma xenografts in athymic nude mice. Cancer Lett 2013; 336:196-203. [PMID: 23664888 DOI: 10.1016/j.canlet.2013.04.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 04/16/2013] [Accepted: 04/29/2013] [Indexed: 10/26/2022]
Abstract
We studied antitumor effect of VIP in human renal cell carcinoma (RCC) (A498 cells xenografted in immunosuppressed mice). VIP-treated cells gave resulted in p53 upregulation and decreased nuclear β-catenin translocation and NFκB expression, MMP-2 and MMP-9 activities, VEGF levels and CD-34 expression. VIP led to a more differentiated tubular organization in tumours and less metastatic areas. Thus, VIP inhibits growth of A498-cell tumours acting on the major issues involved in RCC progression such as cell proliferation, microenvironment remodelling, tumour invasion, angiogenesis and metastatic ability. These antitumoral effects of VIP offer new therapeutical possibilities in RCC treatment.
Collapse
Affiliation(s)
- Eva Vacas
- Department of Systems Biology, Unit of Biochemistry and Molecular Biology, University of Alcalá, 28871 Alcalá de Henares, Spain
| | | | | | | | | | | | | |
Collapse
|
35
|
Thakur ML, Zhang K, Berger A, Cavanaugh B, Kim S, Channappa C, Frangos AJ, Wickstrom E, Intenzo CM. VPAC1 receptors for imaging breast cancer: a feasibility study. J Nucl Med 2013; 54:1019-25. [PMID: 23651947 DOI: 10.2967/jnumed.112.114876] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED VPAC1 encodes G-protein-coupled receptors expressed on all breast cancer (BC) cells at the onset of the disease, but not on benign lesions. Our extensive preclinical studies have shown that (64)Cu-TP3805 has a high affinity for VPAC1, is stable in vivo, and has the ability to distinguish spontaneously grown malignant BC masses from benign lesions. Our long-term goal is to develop (64)Cu-TP3805 as an agent to perform in vivo histology, to distinguish malignant lesions from benign masses noninvasively and thereby avoid patient morbidity and the excess economic costs of benign biopsies. METHODS (18)F-FDG obtained commercially served as a control. (64)Cu-TP3805 was prepared using a sterile kit containing 20 μg of TP3805. Radiochemical purity and sterility were examined. Nineteen consenting women with histologically proven BC were given 370 MBq of (18)F-FDG. One hour later, 6 of these patients were imaged with PET/CT and 13 with positron emission mammography (PEM). Two to 7 d later, 6 PET/CT patients received 111 MBq (± 10%) (n = 2), 127 MBq (± 10%) (n = 2), or 148 MBq (± 10%) (n = 2) of (64)Cu-TP3805 and were imaged 2 and 4 h later. Thirteen PEM patients received 148 MBq (± 10%) of (64)Cu-TP3805 and were imaged 15 min, 1 h, 2 h, and 4 h later. Standardized uptake value (SUV) was calculated for PET/CT patients, and PUV/BGV (PEM uptake value/background value) was calculated for PEM patients. Tumor volume was also calculated. RESULTS The radiochemical purity of (64)Cu-TP3805 was 97% ± 2%, and specific activity was 44.4 GBq (1.2 Ci)/μmol. In 19 patients, a total of 24 lesions were imaged (15 invasive ductal carcinoma, 1 high-grade mammary carcinoma, 3 lobular carcinoma, 1 invasive papilloma, and 4 sentinel lymph nodes). All lesions were unequivocally detected by (64)Cu-TP3805 and by (18)F-FDG. The average tumor volume as determined by PET/CT with (64)Cu-TP3805 was 90.6% ± 16.1% of that with (18)F-FDG PET/CT, and the average SUV was 92% ± 26.4% of that with (18)F-FDG. For PEM, the tumor volume with (64)Cu-TP3805 was 113% ± 37% of that with (18)F-FDG and the PUV/BGV ratio was 97.7% ± 24.5% of that with (18)F-FDG. CONCLUSION (64)Cu-TP3805 is worthy of further investigation in patients requiring biopsy of suggestive imaging findings, to further evaluate its ability to distinguish malignant lesions from benign masses noninvasively.
Collapse
Affiliation(s)
- Mathew L Thakur
- Department of Radiology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Don-Salu-Hewage AS, Chan SY, McAndrews KM, Chetram MA, Dawson MR, Bethea DA, Hinton CV. Cysteine (C)-x-C receptor 4 undergoes transportin 1-dependent nuclear localization and remains functional at the nucleus of metastatic prostate cancer cells. PLoS One 2013; 8:e57194. [PMID: 23468933 PMCID: PMC3585330 DOI: 10.1371/journal.pone.0057194] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 01/18/2013] [Indexed: 01/01/2023] Open
Abstract
The G-protein coupled receptor (GPCR), Cysteine (C)-X-C Receptor 4 (CXCR4), plays an important role in prostate cancer metastasis. CXCR4 is generally regarded as a plasma membrane receptor where it transmits signals that support transformation, progression and eventual metastasis. Due to the central role of CXCR4 in tumorigenesis, therapeutics approaches such as antagonist and monoclonal antibodies have focused on receptors that exist on the plasma membrane. An emerging concept for G-protein coupled receptors is that they may localize to and associate with the nucleus where they retain function and mediate nuclear signaling. Herein, we demonstrate that CXCR4 associated with the nucleus of malignant prostate cancer tissues. Likewise, expression of CXCR4 was detected in nuclear fractions among several prostate cancer cell lines, compared to normal prostate epithelial cells. Our studies identified a nuclear pool of CXCR4 and we defined a nuclear transport pathway for CXCR4. We reveal a putative nuclear localization sequence (NLS), ‘RPRK’, within CXCR4 that contributed to nuclear localization. Additionally, nuclear CXCR4 interacted with Transportinβ1 and Transportinβ1-binding to CXCR4 promoted its nuclear translocation. Importantly, Gαi immunoprecipitation and calcium mobilization studies indicated that nuclear CXCR4 was functional and participated in G-protein signaling, revealing that the nuclear pool of CXCR4 retained function. Given the suggestion that functional, nuclear CXCR4 may be a mechanism underlying prostate cancer recurrence, increased metastatic ability and poorer prognosis after tumors have been treated with therapy that targets plasma membrane CXCR4, these studies addresses a novel mechanism of nuclear signaling for CXCR4, a novel mechanism of clinical targeting, and demonstrate an active nuclear pool that provides important new information to illuminate what has been primarily clinical reports of nuclear CXCR4.
Collapse
Affiliation(s)
- Ayesha S. Don-Salu-Hewage
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, Georgia, United States of America
- Department of Biological Sciences, Clark Atlanta University, Atlanta, Georgia, United States of America
| | - Siu Yuen Chan
- Department of Paediatrics and Adolescent Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong, PRC
| | - Kathleen M. McAndrews
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Mahandranauth A. Chetram
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, Georgia, United States of America
- Department of Biological Sciences, Clark Atlanta University, Atlanta, Georgia, United States of America
| | - Michelle R. Dawson
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Danaya A. Bethea
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, Georgia, United States of America
| | - Cimona V. Hinton
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
37
|
Vacas E, Bajo AM, Schally AV, Sánchez-Chapado M, Prieto JC, Carmena MJ. Vasoactive intestinal peptide induces oxidative stress and suppresses metastatic potential in human clear cell renal cell carcinoma. Mol Cell Endocrinol 2013; 365:212-22. [PMID: 23123564 DOI: 10.1016/j.mce.2012.10.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Revised: 09/12/2012] [Accepted: 10/20/2012] [Indexed: 12/30/2022]
Abstract
Molecular mechanisms involved in progression of clear-cell renal-cell carcinomas (ccRCCs) are poorly understood. A common genetic mutation found in ccRCC is the loss of the von Hippel-Lindau (VHL) gene, which contributes to cancer progression and metastasis. We investigated VIP effects on metastatic and angiogenic factors in human VHL-null A498 ccRCC and HK2 renal cells. VIP increased adhesion but decreased expression of metalloproteinases, MMP2 and MMP9, as well as cell migration and VEGF expression and secretion in A498 but not in HK2 cells. VIP enhanced ROS levels and decreased nuclear levels of β-catenin and NFκB p50-subunit in A498 cells, suggesting neuropeptide involvement in the observed decrease of metastatic ability in clear-cell carcinoma. VIP effects in A498 cells were blocked by the VPAC(1/2)-receptor antagonist JV-1-53. In conclusion, present data point to a role of VIP in preventing invasion and metastasis in ccRCCs and support its potential therapeutic usefulness in this disease.
Collapse
Affiliation(s)
- Eva Vacas
- Department of Biochemistry and Molecular Biology, University of Alcalá, Alcalá de Henares, Spain
| | | | | | | | | | | |
Collapse
|
38
|
Yamamoto H, Okada R, Iguchi K, Ohno S, Yokogawa T, Nishikawa K, Unno K, Hoshino M, Takeda A. Involvement of plasmin-mediated extracellular activation of progalanin in angiogenesis. Biochem Biophys Res Commun 2012; 430:999-1004. [PMID: 23261456 DOI: 10.1016/j.bbrc.2012.11.124] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 11/17/2012] [Indexed: 10/27/2022]
Abstract
Progalanin is released from the small cell lung carcinoma line SBC-3A and converted to its active form by plasmin. To elucidate the role of progalanin activation in the extracellular compartment, matrix metalloproteinase (MMP) activity was studied in SBC-3A cells treated with progalanin siRNA, and angiogenesis was measured in tumor tissue originating from SBC-3A cell transplantation into mice. Progalanin siRNA caused downregulation of progalanin expression for approximately 8 days. MMP activity and angiogenesis were reduced in tumors induced by transplantation of progalanin siRNA-treated SBC-3A cells. In contrast, MMP-9 and MMP-2 activity and angiogenesis increased in tumors originating from progalanin siRNA-treated SBC-3A cells in the presence of galanin and progalanin. Furthermore, injection of tranexamic acid, a plasmin inhibitor, more markedly reduced MMP-9 and MMP-2 activity and angiogenesis in tumors originating from progalanin siRNA-treated SBC-3A cells and in tumor tissue originating from progalanin siRNA-treated SBC-3A cells in the presence of progalanin. The reduction of MMP-9 and MMP-2 activity with tranexamic acid was restored by galanin, but not by progalanin. Moreover, tranexamic acid reduced angiogenesis in control siRNA-treated SBC-3A cells. These results suggest that the activation of progalanin by plasmin in the extracellular compartment was involved in MMP-9 and MMP-2 activation and in angiogenesis in tumor tissue.
Collapse
Affiliation(s)
- Hiroyuki Yamamoto
- Laboratory of Bioorganic Chemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-Ku, Shizuoka 422-8526, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Vacas E, Bajo AM, Schally AV, Sánchez-Chapado M, Prieto JC, Carmena MJ. Antioxidant activity of vasoactive intestinal peptide in HK2 human renal cells. Peptides 2012; 38:275-81. [PMID: 23000305 DOI: 10.1016/j.peptides.2012.09.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Revised: 09/10/2012] [Accepted: 09/10/2012] [Indexed: 11/21/2022]
Abstract
Oxidative stress is a major mediator of tissue and cell injuries. The injury in chronic nephrotic syndrome, acute renal failure, myeloma kidney injury and other kidney diseases is initiated by oxidative stress. We have previously demonstrated that vasoactive intestinal peptide (VIP) acts as an antiproliferative agent in renal cancer cells. This study was designed to evaluate the renoprotective activity of VIP against H(2)O(2)-induced oxidative damage in a proximal tubule kidney cell line (human, non-tumor, HK2 cells) in order to investigate the potential usefulness of this peptide in the treatment of oxidative-stress related kidney diseases. HK2 cell viability was assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay. Propidium iodide was used to identify cells undergoing apoptosis. Western blotting was performed with anti-Bcl-2, anti-Bax and anti-formyl peptide receptor (low-affinity variant FPRL-1) monoclonal antibodies whereas 2,7-dichlorofluorescein diacetate was used for measurement of levels of intracellular reactive oxygen species (ROS). HK2 cells were injured with H(2)O(2) in order to induce apoptosis: the effect was time- and dose-dependent. VIP increased the levels of the antiapoptotic protein Bcl-2 and decreased those of the proapoptotic protein Bax. VIP decreased the intracellular ROS levels reached by H(2)O(2)-induced oxidative stress. VIP effect on ROS levels involved FPLR-1 but not VPAC(1,2) receptors as evidenced by the use of the respective antagonists WRW4 and JV-1-53. Thus, VIP protects HK2 cells from apoptosis by increasing Bcl-2 levels and this effect is initiated through FPLR1 receptor. In conclusion, VIP might exert a renoprotective effect by the suppression of oxidative stress.
Collapse
Affiliation(s)
- Eva Vacas
- Department of Biochemistry and Molecular Biology, University of Alcalá, 28871 Alcalá de Henares, Spain
| | | | | | | | | | | |
Collapse
|
40
|
Muñoz-Moreno L, Arenas MI, Schally AV, Fernández-Martínez AB, Zarka E, González-Santander M, Carmena MJ, Vacas E, Prieto JC, Bajo AM. Inhibitory effects of antagonists of growth hormone-releasing hormone on growth and invasiveness of PC3 human prostate cancer. Int J Cancer 2012; 132:755-65. [PMID: 22777643 DOI: 10.1002/ijc.27716] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Revised: 06/11/2012] [Accepted: 06/22/2012] [Indexed: 01/28/2023]
Abstract
New approaches are needed to the therapy of advanced prostate cancer. This study determined the effect of growth hormone-releasing hormone (GHRH) antagonists, JMR-132 and JV-1-38 on growth of PC3 tumors as well as on angiogenesis and metastasis through the evaluation of various factors that contribute largely to the progression of prostate cancer. Human PC3 androgen-independent prostate cancer cells were injected subcutaneously into nude mice. The treatment with JMR-132 (10 μg/day) or JV-1-38 (20 μg/day) lasted 41 days. We also evaluated the effects of JMR-132 and JV-1-38 on proliferation, cell adhesion and migration in PC-3 cells in vitro. Several techniques (Western blot, reverse transcription polymerase chain reaction, immunohistochemistry, ELISA and zymography) were used to evaluate the expression levels of GHRH receptors and its splice variants, GHRH, vascular endothelial growth factor (VEGF), hypoxia inducible factor (HIF)-1α, metalloproteinases (MMPs) -2 and -9, β-catenin and E-cadherin. GHRH antagonists suppressed the proliferation of PC-3 cells in vitro and significantly inhibited growth of PC3 tumors. After treatment with these analogues, we found an increase in expression of GHRH receptor accompanied by a decrease of GHRH levels, a reduction in both VEGF and HIF-1α expression and in active forms of MMP-2 and MMP-9, a significant increase in levels of membrane-associated β-catenin and a significant decline in E-cadherin. These results support that the blockade of GHRH receptors can modulate elements involved in angiogenesis and metastasis. Consequently, GHRH antagonists could be considered as suitable candidates for therapeutic trials in the management of androgen-independent prostate cancer.
Collapse
Affiliation(s)
- Laura Muñoz-Moreno
- Molecular Neuroendocrinology Unit, Department of Biochemistry and Molecular Biology, University of Alcalá, Alcalá de Henares, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Vacas E, Fernández-Martínez AB, Bajo AM, Sánchez-Chapado M, Schally AV, Prieto JC, Carmena MJ. Vasoactive intestinal peptide (VIP) inhibits human renal cell carcinoma proliferation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2012; 1823:1676-85. [PMID: 22728770 DOI: 10.1016/j.bbamcr.2012.06.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Revised: 05/29/2012] [Accepted: 06/14/2012] [Indexed: 12/13/2022]
Abstract
Clear renal cell carcinoma (cRCC) is an aggressive and fatal neoplasm. The present work was undertaken to investigate the antiproliferative potential of vasoactive intestinal peptide (VIP) exposure on non-tumoral (HK2) and tumoral (A498, cRCC) human proximal tubular epithelial cell lines. Reverse transcription and semiquantitative PCR was used at the VIP mRNA level whereas enzyme immunoanalysis was performed at the protein level. Both renal cell lines expressed VIP as well as VIP/pituitary adenylate cyclase-activating peptide (VPAC) receptors whereas only HK2 cells expressed formyl peptide receptor-like 1 (FPRL-1). Receptors were functional, as shown by VIP stimulation of adenylyl cyclase activity. Treatment with 0.1μM VIP (24h) inhibited proliferation of A498 but not HK2 cells as based on a reduction in the incorporation of [(3)H]-thymidine and BrdU (5'-Br-2'-deoxyuridine), PCNA (proliferating-cell nuclear antigen) expression and STAT3 (signal transducer and activator of transcription 3) expression and activation. VPAC(1)-receptor participation was established using JV-1-53 antagonist and siRNA transfection. Growth-inhibitory response to VIP was related to the cyclic adenosine monophosphate (cAMP)/exchange protein directly activated by cAMP (EPAC)/phosphoinositide 3-kinase (PI3-K) signaling systems as shown by studies on adenylate cyclase stimulation, and using the EPAC-specific compound 8CPT-2Me-cAMP and specific kinase inhibitors such as H89, wortmannin and PD98059. The efficacy of VIP on the prevention of tumor progression was confirmed in vivo using xenografted athymic mouse. These actions support a potential role of this peptide and its agonists in new therapies for cRCC.
Collapse
Affiliation(s)
- Eva Vacas
- Department of Biochemistry and Molecular Biology, University of Alcalá, Alcalá de Henares, Spain
| | | | | | | | | | | | | |
Collapse
|
42
|
Doan ND, Chatenet D, Létourneau M, Vaudry H, Vaudry D, Fournier A. Receptor-independent cellular uptake of pituitary adenylate cyclase-activating polypeptide. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2012; 1823:940-9. [DOI: 10.1016/j.bbamcr.2012.02.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Revised: 01/31/2012] [Accepted: 02/01/2012] [Indexed: 11/28/2022]
|
43
|
Tadevosyan A, Vaniotis G, Allen BG, Hébert TE, Nattel S. G protein-coupled receptor signalling in the cardiac nuclear membrane: evidence and possible roles in physiological and pathophysiological function. J Physiol 2011; 590:1313-30. [PMID: 22183719 DOI: 10.1113/jphysiol.2011.222794] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
G protein-coupled receptors (GPCRs) play key physiological roles in numerous tissues, including the heart, and their dysfunction influences a wide range of cardiovascular diseases. Recently, the notion of nuclear localization and action of GPCRs has become more widely accepted. Nuclear-localized receptors may regulate distinct signalling pathways, suggesting that the biological responses mediated by GPCRs are not solely initiated at the cell surface but may result from the integration of extracellular and intracellular signalling pathways. Many of the observed nuclear effects are not prevented by classical inhibitors that exclusively target cell surface receptors, presumably because of their structures, lipophilic properties, or affinity for nuclear receptors. In this topical review, we discuss specifically how angiotensin-II, endothelin, β-adrenergic and opioid receptors located on the nuclear envelope activate signalling pathways, which convert intracrine stimuli into acute responses such as generation of second messengers and direct genomic effects, and thereby participate in the development of cardiovascular disorders.
Collapse
Affiliation(s)
- Artavazd Tadevosyan
- Department of Medicine, Université de Montréal, Montréal, Québec, Canada H3C 3J7
| | | | | | | | | |
Collapse
|
44
|
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
|
45
|
Chotani MA, Flavahan NA. Intracellular α(2C)-adrenoceptors: storage depot, stunted development or signaling domain? BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2011; 1813:1495-503. [PMID: 21605601 DOI: 10.1016/j.bbamcr.2011.05.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 05/03/2011] [Accepted: 05/06/2011] [Indexed: 12/11/2022]
Abstract
G-protein coupled receptors (GPCRs) are generally considered to function as cell surface signaling structures that respond to extracellular mediators, many of which do not readily access the cell's interior. Indeed, most GPCRs are preferentially targeted to the plasma membrane. However, some receptors, including α(2C)-Adrenoceptors, challenge conventional concepts of GPCR activity by being preferentially retained and localized within intracellular organelles. This review will address the issues associated with this unusual GPCR localization and discuss whether it represents a novel sub-cellular niche for GPCR signaling, whether these receptors are being stored for rapid deployment to the cell surface, or whether they represent immature or incomplete receptor systems.
Collapse
Affiliation(s)
- Maqsood A Chotani
- Center for Cardiovascular and Pulmonary Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
| | | |
Collapse
|