1
|
Rubino V, Cammarota M, Criscuolo C, Cianflone A, De Martino M, de Rosa V, Esposito F, Abbadessa G, Carriero F, Terrazzano G, Chieffi P, Bonavita S, Bresciamorra V, Annunziato L, Ruggiero G, Boscia F. Modulation of NCX1 expression in monocytes associates with multiple sclerosis progression. Heliyon 2025; 11:e42332. [PMID: 40041001 PMCID: PMC11876900 DOI: 10.1016/j.heliyon.2025.e42332] [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: 05/17/2024] [Revised: 01/16/2025] [Accepted: 01/28/2025] [Indexed: 03/06/2025] Open
Abstract
Ionic imbalance and functional heterogeneity of monocytes play key roles in multiple sclerosis (MS) progression. A better understanding of monocyte response in the context of ionic dysregulation during MS course may have relevant implications for understanding of disease pathogenesis and treatments. The sodium calcium exchanger NCX1 influences monocyte-derived macrophages reactivity under inflammation; however, little is known about its monocyte-specific expression during MS course. By means of RT-PCR, flow cytometry, and confocal analyses, we determined the expression profiling of NCX1 exchanger in monocytes of patients during relapsing-remitting MS (RRMS) and secondary progressive MS (SPMS) course. NCX1 expression was significantly upregulated in monocytes from transitional RRMS subjects. Conversely, it was significantly reduced in all monocyte subsets after RRMS conversion to SPMS. Interestingly, NCX1 levels in monocytes significantly correlated with the percentage and growth ability of the regulatory T cell (Treg) subset, whose derangement underlies MS progression. Perturbation of transcripts encoding the Ca2+-ATPase isoform 1 and 4, the Na+/K+-ATPase α1 subunit, and the long non-coding RNA SLC8A1-AS1 associated with NCX1 changes in peripheral blood mononuclear cells (PBMC) during MS. Our findings demonstrated a stage-specific dysregulation of NCX1 exchanger in monocytes during MS progression and suggested that ionic imbalance in monocytes may influence not only their functional response but also the immune regulatory network during MS course. These data may be relevant for the identification of novel biomarkers and/or therapeutic targets in MS.
Collapse
Affiliation(s)
- Valentina Rubino
- Department of Medical Translational Sciences, School of Medicine, “Federico II” University of Naples, 80131, Naples, Italy
| | - Mariarosaria Cammarota
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, “Federico II” University of Naples, 80131, Naples, Italy
| | - Chiara Criscuolo
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, “Federico II” University of Naples, 80131, Naples, Italy
- CDCD Neurology, “Federico II” University Hospital, Naples, Italy
| | - Alessandra Cianflone
- Clinical and Translational Research Unit, Santobono-Pausilipon Children's Hospital, 80129, Naples, Italy
| | - Marco De Martino
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138, Naples, Italy
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Naples, Italy
| | - Valeria de Rosa
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, “Federico II” University of Naples, 80131, Naples, Italy
| | - Francesco Esposito
- Institute of Experimental Endocrinology and Oncology (IEOS) “G. Salvatore”, National Research Council (CNR), 80131, Naples, Italy
| | - Gianmarco Abbadessa
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138, Naples, Italy
| | - Flavia Carriero
- Department of Health Science, University of Basilicata, 85100, Potenza, Italy
| | - Giuseppe Terrazzano
- Department of Health Science, University of Basilicata, 85100, Potenza, Italy
| | - Paolo Chieffi
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138, Naples, Italy
| | - Simona Bonavita
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138, Naples, Italy
| | - Vincenzo Bresciamorra
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, “Federico II” University of Naples, 80131, Naples, Italy
- Multiple Sclerosis Unit, Policlinico “Federico II” University Hospital, 80131, Naples, Naples, Italy
| | | | - Giuseppina Ruggiero
- Department of Medical Translational Sciences, School of Medicine, “Federico II” University of Naples, 80131, Naples, Italy
| | - Francesca Boscia
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, “Federico II” University of Naples, 80131, Naples, Italy
| |
Collapse
|
2
|
Macheroni C, Souza DS, Porto CS, Vicente CM. Estrogen receptor activates SRC and ERK1/2 and promotes tumorigenesis in human testicular embryonic carcinoma cells NT2/D1. Exp Cell Res 2024; 442:114282. [PMID: 39413983 DOI: 10.1016/j.yexcr.2024.114282] [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: 07/04/2024] [Revised: 08/26/2024] [Accepted: 10/12/2024] [Indexed: 10/18/2024]
Abstract
Testicular germ cell tumors have the highest incidence in young men (between 15 and 44 years of age) and its etiology is still unclear, but its emergence on puberty suggests a hormone-dependent mechanism for the development of these tumors and their progression. We previously identified the estrogen receptor ESR1, ESR2, GPER and an isoform of ESR1, the ESR1-36 in human testicular embryonic carcinoma NT2/D1 cells, and the activation of SRC induced by ESR1 and ESR2 in these cells. Therefore, this study aimed to analyze the role of ER in the activation of ERK1/2, and the involvement of SRC and ERK1/2 on proliferation, migration, and invasion of the NT2/D1 cells. Our results showed that the activation of ESR1 (using ESR1-selective agonist PPT) or ESR2 (using ESR2-selective agonist DPN) increased phosphorylation of ERK1/2 in NT2/D1 cells. In the presence of the selective inhibitor for SRC-family kinases PP2, or the MEK specific inhibitor U0126, the effects of 17β-estradiol (E2) or PPT were blocked on proliferation and invasion of NT2/D1 cells. Finally, the proliferation, migration, and invasion of NT2/D1 cells simulated by E2 or ESR2 were also blocked by PP2 and U0126. This study provides novel insights into molecular mechanisms of ER in NT2/D1 cells by demonstrating that ER activates rapid responses molecules, including SRC and ERK1/2, which enhance the tumorigenic potential of testicular cancer cells.
Collapse
Affiliation(s)
- Carla Macheroni
- Laboratory of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP, 04039-032, Brazil
| | - Deborah Simão Souza
- Laboratory of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP, 04039-032, Brazil
| | - Catarina Segreti Porto
- Laboratory of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP, 04039-032, Brazil
| | - Carolina Meloni Vicente
- Laboratory of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP, 04039-032, Brazil.
| |
Collapse
|
3
|
Sinisi AA, Rossi V, Martino MD, Esposito F, Chieffi P. The role of endocrine gland derived vascular growth factor/ Prokineticin-1 in human prostate cells. GHM OPEN 2024; 4:37-41. [PMID: 40144745 PMCID: PMC11933967 DOI: 10.35772/ghmo.2023.01021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/07/2023] [Accepted: 12/26/2023] [Indexed: 03/28/2025]
Abstract
In steroidogenic tissues, a novel class of angiogenic molecules known as endocrine gland-derived vascular endothelial growth factors (EG-VEGF)/prokineticins are primarily produced. Here, we investigated how EG-VEGF/ PROK1, a member of PROKs family, and its receptor are able to affect cellular motility in both non-neoplastic and cancerous human prostate cells. Using Western blot and motility test studies, EPN cells, a non-transformed cell line and Cancer Epithelial Prostatic Cells (CEPC) were employed as cellular models in the current investigation. Western blot examination of EPN normal prostate cells treated with EG-VEGF/PROK1 revealed that ERK1/2 was rapidly phosphorylated within 5, 10, and 20 minutes, while CEPC had high and sustained ERK1/2 activity at the same periods. Then, compared to normal EPN prostate cells, CEPC treated with EG-VEGF/PROK1 for up to 72 hours demonstrated enhanced cell motility. Based on our findings, EG-VEGF/PROK1 may play a role in prostate cancer progression by controlling angiogenesis and the motility of metastatic cells in CEPC cells, likely as a consequence of ERK1/2 activation, as contrasted to EPN normal prostate cells.
Collapse
Affiliation(s)
- Antonio Agostino Sinisi
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Valentina Rossi
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Marco De Martino
- Institute of Endocrinology and Experimental Oncology (IEOS) "G. Salvatore", National Research Council (CNR), Naples, Italy
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Francesco Esposito
- Institute of Endocrinology and Experimental Oncology (IEOS) "G. Salvatore", National Research Council (CNR), Naples, Italy
| | - Paolo Chieffi
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
4
|
Macheroni C, Leite GGF, Souza DS, Vicente CM, Lacerda JT, Moraes MN, Juliano MA, Porto CS. Activation of estrogen receptor induces differential proteomic responses mainly involving migration, invasion, and tumor development pathways in human testicular embryonal carcinoma NT2/D1 cells. J Steroid Biochem Mol Biol 2024; 237:106443. [PMID: 38092129 DOI: 10.1016/j.jsbmb.2023.106443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/27/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023]
Abstract
The aims of the present study were to investigate the global changes on proteome of human testicular embryonal carcinoma NT2/D1 cells treated with 17β-estradiol (E2), and the effects of this hormone on migration, invasion, and colony formation of these cells. A quantitative proteomic analysis identified the presence of 1230 proteins in both E2-treated and control cells. The analysis revealed 75 differentially abundant proteins (DAPs), out of which 43 proteins displayed a higher abundance and, 30 proteins showed a lower abundance in E2-treated NT2/D1 cancer cells. Functional analysis using IPA highlighted some activation processes such as migration, invasion, metastasis, and tumor growth. Interestingly, the treatment with E2 and ERβ-selective agonist DPN increased the migration of NT2/D1 cells. On the other hand, ERα-selective agonist PPT did not modify cell migration, indicating that ERβ is the upstream receptor involved in this process. The activation of ERβ increased the invasion and anchorage‑independent growth of NT2/D1 cells more intensely than ERα. ERα and ERβ may play overlapping roles on invasion and colony formation of these cells. Further studies are required to clarify the mechanism underlying these effects. The molecular mechanisms revealed by proteomic and functional studies might also guide the development of potential targets for a better understanding of the biology of these cells and novel treatments for non-seminoma in the future.
Collapse
Affiliation(s)
- Carla Macheroni
- Laboratory of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP 04039-032, Brazil
| | - Giuseppe Gianini Figueirêdo Leite
- Division of Infectious Diseases, Department of Medicine, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP 04039-032, Brazil
| | - Deborah Simão Souza
- Laboratory of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP 04039-032, Brazil
| | - Carolina Meloni Vicente
- Laboratory of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP 04039-032, Brazil
| | - José Thalles Lacerda
- Department of Physiology, Instituto de Biociências, Universidade de São Paulo, Rua do Matão 277, Butantã, São Paulo, SP 05508-090, Brazil
| | - Maria Nathália Moraes
- Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Universidade Federal de São Paulo, Av. Conceição 515, Diadema, São Paulo, SP, 09920-000, Brazil
| | - Maria Aparecida Juliano
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Três de Maio, 100, Vila Clementino, São Paulo, SP 04044-020, Brazil
| | - Catarina Segreti Porto
- Laboratory of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP 04039-032, Brazil.
| |
Collapse
|
5
|
Yodkhunnatham N, Pandit K, Puri D, Yuen KL, Bagrodia A. MicroRNAs in Testicular Germ Cell Tumors: The Teratoma Challenge. Int J Mol Sci 2024; 25:2156. [PMID: 38396829 PMCID: PMC10889716 DOI: 10.3390/ijms25042156] [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: 01/03/2024] [Revised: 02/05/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
Testicular germ cell tumors (TGCTs) are relatively common in young men, making accurate diagnosis and prognosis assessment essential. MicroRNAs (miRNAs), including microRNA-371a-3p (miR-371a-3p), have shown promise as biomarkers for TGCTs. This review discusses the recent advancements in the use of miRNA biomarkers in TGCTs, with a focus on the challenges surrounding the noninvasive detection of teratomas. Circulating miR-371a-3p, which is expressed in undifferentiated TGCTs but not in teratomas, is a promising biomarker for TGCTs. Its detection in serum, plasma, and, potentially, cystic fluid could be useful for TGCT diagnosis, surveillance, and monitoring of therapeutic response. Other miRNAs, such as miR-375-3p and miR-375-5p, have been investigated to differentiate between TGCT subtypes (teratoma, necrosis/fibrosis, and viable tumors), which can aid in treatment decisions. However, a reliable marker for teratoma has yet to be identified. The clinical applications of miRNA biomarkers could spare patients from unnecessary surgeries and allow for more personalized therapeutic approaches. Particularly in patients with residual masses larger than 1 cm following chemotherapy, it is critical to differentiate between viable tumors, teratomas, and necrosis/fibrosis. Teratomas, which mimic somatic tissues, present a challenge in differentiation and require a comprehensive diagnostic approach. The combination of miR-371 and miR-375 shows potential in enhancing diagnostic precision, aiding in distinguishing between teratomas, viable tumors, and necrosis. The implementation of miRNA biomarkers in TGCT care could improve patient outcomes, reduce overtreatment, and facilitate personalized therapeutic strategies. However, a reliable marker for teratoma is still lacking. Future research should focus on the clinical validation and standardization of these biomarkers to fully realize their potential.
Collapse
Affiliation(s)
- Nuphat Yodkhunnatham
- Department of Urology, University of California San Diego School of Medicine, La Jolla, CA 92093, USA; (N.Y.); (K.P.); (D.P.); (K.L.Y.)
| | - Kshitij Pandit
- Department of Urology, University of California San Diego School of Medicine, La Jolla, CA 92093, USA; (N.Y.); (K.P.); (D.P.); (K.L.Y.)
| | - Dhruv Puri
- Department of Urology, University of California San Diego School of Medicine, La Jolla, CA 92093, USA; (N.Y.); (K.P.); (D.P.); (K.L.Y.)
| | - Kit L. Yuen
- Department of Urology, University of California San Diego School of Medicine, La Jolla, CA 92093, USA; (N.Y.); (K.P.); (D.P.); (K.L.Y.)
| | - Aditya Bagrodia
- Department of Urology, University of California San Diego School of Medicine, La Jolla, CA 92093, USA; (N.Y.); (K.P.); (D.P.); (K.L.Y.)
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
6
|
Pagliuca F, Lucà S, De Sio M, Arcaniolo D, Facchini G, De Martino M, Esposito F, DE Vita F, Chieffi P, Franco R. Testicular/paratesticular mesothelial tumours: Uncommon histopathologic entities in a very complex anatomical site. Pathol Res Pract 2024; 253:155069. [PMID: 38181581 DOI: 10.1016/j.prp.2023.155069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 12/28/2023] [Indexed: 01/07/2024]
Abstract
Mesothelial tumours of the testicular/paratesticular region are uncommon, poorly characterised and difficult-to-diagnose lesions. They encompass entirely benign proliferations (adenomatoid tumour) and malignant, very aggressive tumours (mesothelioma) whose morphological features can be overlapping, highly variable and confounding. Moreover, testicular/paratesticular mesothelial tumours comprise relatively new entities with indolent behaviour (well-differentiated papillary mesothelial tumour) as well as tumours which cannot be correctly included in any of the aforementioned categories and whose classification is still controversial. The molecular profile of such tumours represents an open issue. In fact, despite the recent discoveries about the genomic landscape of mesothelial proliferations at other sites (pleura, peritoneum), testicular/paratesticular mesothelial tumours, and namely mesotheliomas, are too rare to be extensively studied on large case series and they could arguably hide relevant differences in their molecular background when compared to the more common pleural/peritoneal counterparts.The aim of this review is to provide a guide for the pathological assessment of testicular/paratesticular mesothelial tumours. Herein, we describe the most recent updates on this topic according to the latest (year 2022) World Health Organisation Classification of Urinary and Male Genital Tumours (5th edition) and current literature. The diagnostic criteria, the main differentials and the role of ancillary techniques in the diagnosis of mesothelial testicular/paratesticular tumours are discussed.
Collapse
Affiliation(s)
| | - Stefano Lucà
- Department of Mental and Physical Health and Preventive Medicine, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Marco De Sio
- Department of Woman, Child and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", 80131 Naples, Italy; Urology Unit, Azienda Policlinico Unversità 'L. Vanvitelli', 80131 Naples, Italy
| | - Davide Arcaniolo
- Department of Woman, Child and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", 80131 Naples, Italy; Urology Unit, Azienda Policlinico Unversità 'L. Vanvitelli', 80131 Naples, Italy
| | - Gaetano Facchini
- Oncology Unit, "S. Maria delle Grazie" Hospital, ASL NA2 NORD, 80078 Pozzuoli, Naples, Italy
| | - Marco De Martino
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138 Naples, Italy; Institute of Endocrinology and Experimental Oncology of the CNR, 80131 Naples, Italy
| | - Francesco Esposito
- Institute of Endocrinology and Experimental Oncology of the CNR, 80131 Naples, Italy
| | - Ferdinando DE Vita
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Paolo Chieffi
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138 Naples, Italy.
| | - Renato Franco
- Pathology Unit, Azienda Policlinico Unversità 'L. Vanvitelli, Italy; Department of Mental and Physical Health and Preventive Medicine, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| |
Collapse
|
7
|
Cammarota M, Boscia F. Contribution of Oligodendrocytes, Microglia, and Astrocytes to Myelin Debris Uptake in an Explant Model of Inflammatory Demyelination in Rats. Cells 2023; 12:2203. [PMID: 37681935 PMCID: PMC10486984 DOI: 10.3390/cells12172203] [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: 08/05/2023] [Revised: 08/30/2023] [Accepted: 08/31/2023] [Indexed: 09/09/2023] Open
Abstract
The internalization and degradation of myelin in glia contributes to the resolution of neuroinflammation and influences disease progression. The identification of a three-dimensional experimental model to study myelin processing under neuroinflammation will offer a novel approach for studying treatment strategies favoring inflammation resolution and neuroprotection. Here, by using a model of neuroinflammation in hippocampal explants, we show that myelin debris accumulated immediately after insult and declined at 3 days, a time point at which tentative repair processes were observed. Olig2+ oligodendrocytes upregulated the LRP1 receptor and progressively increased MBP immunoreactivity both at peri-membrane sites and within the cytosol. Oligodendrocyte NG2+ precursors increased in number and immunoreactivity one day after insult, and moderately internalized MBP particles. Three days after insult MBP was intensely coexpressed by microglia and, to a much lesser extent, by astrocytes. The engulfment of both MBP+ debris and whole MBP+ cells contributed to the greatest microglia response. In addition to improving our understanding of the spatial-temporal contribution of glial scarring to myelin uptake under neuroinflammation, our findings suggest that the exposure of hippocampal explants to LPS + IFN-γ-induced neuroinflammation may represent a valuable demyelination model for studying both the extrinsic and intrinsic myelin processing by glia under neuroinflammation.
Collapse
Affiliation(s)
| | - Francesca Boscia
- Division of Pharmacology, Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, Federico II University of Naples, 80131 Naples, Italy;
| |
Collapse
|
8
|
Montella M, Errico ME, Ronchi A, Zannini G, Donofrio V, Savarese G, Sirica R, Esposito F, Martino MD, Papparella A, Franco R, Chieffi P, Marino FZ. Analysis of microsatellite instability (MSI) in pediatric gonadal and extra-gonadal germ cell tumors. Intractable Rare Dis Res 2023; 12:191-197. [PMID: 37662626 PMCID: PMC10468407 DOI: 10.5582/irdr.2023.01039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/27/2023] [Accepted: 08/15/2023] [Indexed: 09/05/2023] Open
Abstract
Gonadal and extragonadal pediatric germ cell tumors (GCTs) are rare neoplasms with different clinical behavior. Although surgery and cisplatin-based chemotherapy are resolutive in most cases, some patients do not respond to chemotherapy and have a worse outcome. Microsatellite instability (MSI) was correlated to resistance to chemotherapy and sensitivity to immunotherapy in different neoplasms. A series of 21 pediatric GCTs were tested by immuno-histochemistry and PCR to evaluate MSI status. Next generation sequencing was applied to further evaluate cases with discordant results between immunohistochemistry and PCR. Twenty-one cases of pediatric GCT were included in the series. The mean age ranged between 1 and 10 years. Nine cases were gonadal GCTs and the remaining 12 were extra-gonadal GCTs. By immunohistochemistry, one case showed a deficit of Mismatch repair (MMR) proteins. This case was a 1-year-old children affected by gonadal yolk sac tumor. However, all cases resulted microsatellite stable (MSS) by PCR and NGS. MSI was not detected in our series of pediatric GCTs, as well as the data present in literature about adult patients with GCTs. Molecular techniques could have a role to confirm the MSI status in case of dMMR by immunohistochemistry.
Collapse
Affiliation(s)
- Marco Montella
- Pathology Unit, Department of Mental Health and Physic and Preventive Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Maria Elena Errico
- Pathology Unit Department of Pathology, Santobono-Pausilipon Children's Hospital, Naples, Italy
| | - Andrea Ronchi
- Pathology Unit, Department of Mental Health and Physic and Preventive Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giuseppa Zannini
- Pathology Unit, Department of Mental Health and Physic and Preventive Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Vittoria Donofrio
- Pathology Unit Department of Pathology, Santobono-Pausilipon Children's Hospital, Naples, Italy
| | - Giovanni Savarese
- AMES, Centro Polidiagnostico Strumentale srl, Casalnuovo, Naples, Italy
| | - Roberto Sirica
- AMES, Centro Polidiagnostico Strumentale srl, Casalnuovo, Naples, Italy
| | - Francesco Esposito
- Institute of Experimental Endocrinology and Oncology (IEOS) "G. Salvatore", National Research Council (CNR), Naples, Italy
| | - Marco De Martino
- Institute of Experimental Endocrinology and Oncology (IEOS) "G. Salvatore", National Research Council (CNR), Naples, Italy
| | - Alfonso Papparella
- Department of Child, Women, General and Specialized Surgery, University of Campania, Naples, Italy
| | - Renato Franco
- Pathology Unit, Department of Mental Health and Physic and Preventive Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Paolo Chieffi
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Federica Zito Marino
- Pathology Unit, Department of Mental Health and Physic and Preventive Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
9
|
Cammarota M, Ferlenghi F, Vacondio F, Vincenzi F, Varani K, Bedini A, Rivara S, Mor M, Boscia F. Combined targeting of fatty acid amide hydrolase and melatonin receptors promotes neuroprotection and stimulates inflammation resolution in rats. Br J Pharmacol 2022; 180:1316-1338. [PMID: 36526591 DOI: 10.1111/bph.16014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 11/09/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND AND PURPOSE Devising novel strategies to therapeutically favour inflammation resolution and provide neuroprotection is an unmet clinical need. Enhancing endocannabinoid tone by inhibiting the catabolic enzyme fatty acid amide hydrolase (FAAH), or stimulating melatonin receptors has therapeutic potential to treat neuropathological states in which neuroinflammation plays a central role. EXPERIMENTAL APPROACH A rodent hippocampal explant model of inflammatory injury was used to assess the effects of UCM1341, a dual-acting compound with FAAH inhibitory action and agonist activity at melatonin receptors, against neuroinflammatory damage. FAAH activity was measured by a radiometric assay, and N-acylethanolamine levels were assessed by HPLC-MS/MS methods. FAAH distribution, evolution of inflammation and the contribution of UCM1341 to the expression of proteins controlling macrophage behaviour were investigated by biochemical and confocal analyses. KEY RESULTS UCM1341 exhibited greater neuroprotection against neuroinflammatory degeneration, compared with the reference compounds URB597 (FAAH inhibitor) and melatonin. During neuroinflammation, UCM1341 augmented the levels of anandamide and N-oleoylethanolamine, but not N-palmitoylethanolamine, up-regulated PPAR-α levels, attenuated demyelination and prevented the release of TNF-α. UCM1341 modulated inflammatory responses by contributing to microglia/macrophage polarization, stimulating formation of lipid-laden macrophages and regulating expression of proteins controlling cholesterol metabolism and efflux. The neuroprotective effects of UCM1341 were prevented by PPARα, TRPV1 and melatonin receptor antagonists. CONCLUSION AND IMPLICATIONS UCM1341, by enhancing endocannabinoid and melatoninergic signalling, benefits neuroprotection and stimulates inflammation resolution pathways. Our findings provide an encouraging prospect of therapeutically targeting endocannabinoid and melatoninergic systems in inflammatory demyelinating states in the CNS.
Collapse
Affiliation(s)
- Mariarosaria Cammarota
- Division of Pharmacology, Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, Federico II University of Naples, Naples, Italy
| | | | | | - Fabrizio Vincenzi
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Katia Varani
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Annalida Bedini
- Department of Biomolecular Sciences, University of Urbino 'Carlo Bo', Urbino, Italy
| | - Silvia Rivara
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Marco Mor
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Francesca Boscia
- Division of Pharmacology, Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, Federico II University of Naples, Naples, Italy
| |
Collapse
|
10
|
Muhammad A, Forcados GE, Yusuf AP, Abubakar MB, Sadiq IZ, Elhussin I, Siddique MAT, Aminu S, Suleiman RB, Abubakar YS, Katsayal BS, Yates CC, Mahavadi S. Comparative G-Protein-Coupled Estrogen Receptor (GPER) Systems in Diabetic and Cancer Conditions: A Review. Molecules 2022; 27:molecules27248943. [PMID: 36558071 PMCID: PMC9786783 DOI: 10.3390/molecules27248943] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/05/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022] Open
Abstract
For many patients, diabetes Mellitus and Malignancy are frequently encountered comorbidities. Diabetes affects approximately 10.5% of the global population, while malignancy accounts for 29.4 million cases each year. These troubling statistics indicate that current treatment approaches for these diseases are insufficient. Alternative therapeutic strategies that consider unique signaling pathways in diabetic and malignancy patients could provide improved therapeutic outcomes. The G-protein-coupled estrogen receptor (GPER) is receiving attention for its role in disease pathogenesis and treatment outcomes. This review aims to critically examine GPER' s comparative role in diabetes mellitus and malignancy, identify research gaps that need to be filled, and highlight GPER's potential as a therapeutic target for diabetes and malignancy management. There is a scarcity of data on GPER expression patterns in diabetic models; however, for diabetes mellitus, altered expression of transport and signaling proteins has been linked to GPER signaling. In contrast, GPER expression in various malignancy types appears to be complex and debatable at the moment. Current data show inconclusive patterns of GPER expression in various malignancies, with some indicating upregulation and others demonstrating downregulation. Further research should be conducted to investigate GPER expression patterns and their relationship with signaling pathways in diabetes mellitus and various malignancies. We conclude that GPER has therapeutic potential for chronic diseases such as diabetes mellitus and malignancy.
Collapse
Affiliation(s)
- Aliyu Muhammad
- Center for Cancer Research, Department of Biology, Tuskegee University, Tuskegee, AL 36088, USA
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria P.M.B. 1044, Nigeria
| | | | - Abdurrahman Pharmacy Yusuf
- Department of Biochemistry, School of Life Sciences, Federal University of Technology, Minna P.M.B. 65, Nigeria
| | - Murtala Bello Abubakar
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Usmanu Danfodiyo University, Sokoto P.M.B. 2254, Nigeria
- Centre for Advanced Medical Research & Training (CAMRET), Usmanu Danfodiyo University, Sokoto P.M.B. 2254, Nigeria
| | - Idris Zubairu Sadiq
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria P.M.B. 1044, Nigeria
| | - Isra Elhussin
- Center for Cancer Research, Department of Biology, Tuskegee University, Tuskegee, AL 36088, USA
| | - Md Abu Talha Siddique
- Center for Cancer Research, Department of Biology, Tuskegee University, Tuskegee, AL 36088, USA
| | - Suleiman Aminu
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria P.M.B. 1044, Nigeria
| | - Rabiatu Bako Suleiman
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria P.M.B. 1044, Nigeria
| | - Yakubu Saddeeq Abubakar
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria P.M.B. 1044, Nigeria
| | - Babangida Sanusi Katsayal
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria P.M.B. 1044, Nigeria
| | - Clayton C Yates
- Center for Cancer Research, Department of Biology, Tuskegee University, Tuskegee, AL 36088, USA
| | - Sunila Mahavadi
- Center for Cancer Research, Department of Biology, Tuskegee University, Tuskegee, AL 36088, USA
| |
Collapse
|
11
|
Macheroni C, Gameiro Lucas TF, Souza DS, Vicente CM, Pereira GJDS, Junior IDSV, Juliano MA, Porto CS. Activation of estrogen receptor ESR1 and ESR2 induces proliferation of the human testicular embryonal carcinoma NT2/D1 cells. Mol Cell Endocrinol 2022; 554:111708. [PMID: 35792284 DOI: 10.1016/j.mce.2022.111708] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 06/14/2022] [Accepted: 06/20/2022] [Indexed: 12/01/2022]
Abstract
The aims of the present study were to investigate the expression of the classic estrogen receptors ESR1 and ESR2, the splicing variant ESR1-36 and GPER in human testicular embryonal carcinoma NT2/D1 cells, and the effects of the activation of the ESR1 and ESR2 on cell proliferation. Immunostaining of ESR1, ESR2, and GPER were predominantly found in the nuclei, and less abundant in the cytoplasm. ESR1-36 isoform was predominantly expressed in the perinuclear region and cytoplasm, and some weakly immunostained in the nuclei. In nonstimulated NT2/D1 cells (control), proteins of the cell cycle CCND1, CCND2, CCNE1 and CDKN1B are present. Activation of ESR1 and ESR2 increases, respectively, CCND2 and CCNE1 expression, but not CCND1. Activation of ESR2 also mediates upregulation of the cell cycle inhibitor CDKN1B. This protein co-immunoprecipitated with CCND2. Also, E2 induces an increase in the number and viability of the NT2/D1 cells. These effects are blocked by simultaneous pretreatment with ESR1-and ESR2-selective antagonists, confirming that both estrogen receptors regulate NT2/D1 cell proliferation. In addition, E2 increases SRC phosphorylation, and SRC mediates cell proliferation. Our study provides novel insights into the signatures and molecular mechanisms of estrogen receptor in NT2/D1 cells.
Collapse
Affiliation(s)
- Carla Macheroni
- Laboratory of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP, 04039-032, Brazil
| | - Thaís Fabiana Gameiro Lucas
- Laboratory of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP, 04039-032, Brazil
| | - Deborah Simão Souza
- Laboratory of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP, 04039-032, Brazil
| | - Carolina Meloni Vicente
- Laboratory of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP, 04039-032, Brazil
| | - Gustavo José da Silva Pereira
- Laboratory of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP, 04039-032, Brazil
| | - Itabajara da Silva Vaz Junior
- Centro de Biotecnologia e Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Av. Bento Goncalves 9500, Porto Alegre, RS, 91501-970, Brazil
| | - Maria Aparecida Juliano
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Três de Maio, 100, Vila Clementino, São Paulo, SP, 04044-020, Brazil
| | - Catarina Segreti Porto
- Laboratory of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP, 04039-032, Brazil.
| |
Collapse
|
12
|
Walczak-Jędrzejowska R, Forma E, Oszukowska E, Bryś M, Marchlewska K, Kula K, Słowikowska-Hilczer J. Expression of G-Protein-Coupled Estrogen Receptor ( GPER) in Whole Testicular Tissue and Laser-Capture Microdissected Testicular Compartments of Men with Normal and Aberrant Spermatogenesis. BIOLOGY 2022; 11:biology11030373. [PMID: 35336747 PMCID: PMC8945034 DOI: 10.3390/biology11030373] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 02/21/2022] [Accepted: 02/24/2022] [Indexed: 01/23/2023]
Abstract
Simple Summary Nowadays, there is no doubt that estrogens play an important role in male reproduction, affecting testicular cell differentiation, proliferation, apoptosis and metabolism. It is also widely believed that intratesticular balance of androgens and estrogens is crucial for the testicular development and function and that the increased testicular estrogen production may be associated with spermatogenic failure. There is also growing epidemiological evidence that the exposure of men to endocrine disruptors demonstrating estrogenic activity (xenoestrogens) may lead to impairment of male fertility via interference with estrogen signaling pathways. Besides the two classical nuclear estrogen receptors, the membrane-bound G protein-coupled estrogen receptor (GPER) was described in human testicular tissue. However, there are little data on its expression in testes with disturbed spermatogenesis. In this study, we investigated the GPER expression pattern in biopsies of azoospermic men with complete and aberrant spermatogenesis. Our results showed an increased expression of the GPER in testes with impaired spermatogenesis. Moreover, they indicate a possible involvement of estrogen signaling through GPER in disturbed function of Sertoli cells—the cells that support spermatogenic process. Abstract In this study, we retrospectively investigated GPER expression in biopsies of azoospermic men with complete (obstructive azoospermia—OA) and aberrant spermatogenesis (nonobstructive azoospermia—NOA). Each biopsy was histologically evaluated with morphometry. The testicular GPER expression was analyzed by the immunohistochemistry and RT-PCR technique in the whole testicular tissue and in seminiferous tubules and Leydig cells after laser-capture microdissection. In laser-microdissected compartments, we also analyzed transcriptional expression of selected Leydig (CYP17A1, HSD17B3, StAR) and Sertoli cell (AMH, SCF, BMP4) function markers. Immunohistochemical staining revealed expression of GPER in the cytoplasm of Leydig and Sertoli cells. Its stronger intensity was observed in Sertoli cells of NOA biopsies. The RT-PCR analysis of the GPER mRNA level unequivocally showed its increased expression in seminiferous tubules (i.e., Sertoli cells), not Leydig cells in NOA biopsies. This increased expression correlated positively with the transcriptional level of AMH—a marker of Sertoli cell immaturity, as well as FSH serum level in NOA but not in the OA group. Our results clearly demonstrate altered GPER expression in testes with primary spermatogenic impairment that might be related to Sertoli cell maturity/function.
Collapse
Affiliation(s)
- Renata Walczak-Jędrzejowska
- Department of Andrology and Reproductive Endocrinology, Medical University of Lodz, Pomorska Str. 251, 92-213 Lodz, Poland; (K.M.); (K.K.); (J.S.-H.)
- Correspondence: ; Tel.: +48-42-272-53-91
| | - Ewa Forma
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska Str. 141/143, 90-236 Lodz, Poland; (E.F.); (M.B.)
| | - Elżbieta Oszukowska
- II Clinic of Urology, Medical University of Lodz, Pabianicka Str. 62, 93-513 Lodz, Poland;
| | - Magdalena Bryś
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska Str. 141/143, 90-236 Lodz, Poland; (E.F.); (M.B.)
| | - Katarzyna Marchlewska
- Department of Andrology and Reproductive Endocrinology, Medical University of Lodz, Pomorska Str. 251, 92-213 Lodz, Poland; (K.M.); (K.K.); (J.S.-H.)
| | - Krzysztof Kula
- Department of Andrology and Reproductive Endocrinology, Medical University of Lodz, Pomorska Str. 251, 92-213 Lodz, Poland; (K.M.); (K.K.); (J.S.-H.)
| | - Jolanta Słowikowska-Hilczer
- Department of Andrology and Reproductive Endocrinology, Medical University of Lodz, Pomorska Str. 251, 92-213 Lodz, Poland; (K.M.); (K.K.); (J.S.-H.)
| |
Collapse
|
13
|
Chimento A, De Luca A, Avena P, De Amicis F, Casaburi I, Sirianni R, Pezzi V. Estrogen Receptors-Mediated Apoptosis in Hormone-Dependent Cancers. Int J Mol Sci 2022; 23:1242. [PMID: 35163166 PMCID: PMC8835409 DOI: 10.3390/ijms23031242] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 02/04/2023] Open
Abstract
It is known that estrogen stimulates growth and inhibits apoptosis through estrogen receptor(ER)-mediated mechanisms in many cancer cell types. Interestingly, there is strong evidence that estrogens can also induce apoptosis, activating different ER isoforms in cancer cells. It has been observed that E2/ERα complex activates multiple pathways involved in both cell cycle progression and apoptotic cascade prevention, while E2/ERβ complex in many cases directs the cells to apoptosis. However, the exact mechanism of estrogen-induced tumor regression is not completely known. Nevertheless, ERs expression levels of specific splice variants and their cellular localization differentially affect outcome of estrogen-dependent tumors. The goal of this review is to provide a general overview of current knowledge on ERs-mediated apoptosis that occurs in main hormone dependent-cancers. Understanding the molecular mechanisms underlying the induction of ER-mediated cell death will be useful for the development of specific ligands capable of triggering apoptosis to counteract estrogen-dependent tumor growth.
Collapse
Affiliation(s)
- Adele Chimento
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Arianna De Luca
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Paola Avena
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Francesca De Amicis
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Ivan Casaburi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Rosa Sirianni
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Vincenzo Pezzi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, Arcavacata di Rende, 87036 Cosenza, Italy
| |
Collapse
|
14
|
Chieffi P. GPR30: A new potential therapeutic target in human testicular germ cell tumors. Intractable Rare Dis Res 2021; 10:292-293. [PMID: 34877243 PMCID: PMC8630463 DOI: 10.5582/irdr.2021.01103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/07/2021] [Accepted: 09/21/2021] [Indexed: 11/05/2022] Open
Abstract
The G protein-coupled estrogen receptor (GPR30) is suggested to exert a role in non-nuclear estrogen signalling and is over-expressed in a variety of hormone dependent tumors. It is well known that estrogens and xenoestrogens are involved in testicular germ cell tumorigenesis. Different studies show that down regulation of estrogen receptor β (ERβ) associates with GPR30 over-expression both in human testicular carcinoma in situ (CIS) and seminomas and that the mitogenic role exerted by 17β-oestradiol induces the activation of extracellular signal-regulated kinase 1/2 (ERK1/2) through GPR30. In conclusion, the exposure to oestrogens or oestrogen-mimics, in some as of yet undefined manner, diminishes the ERβ-mediated growth restraint in CIS and in human testicular seminoma, indicating that GPR30 could be considered a potential therapeutic target to design specific inhibitors.
Collapse
Affiliation(s)
- Paolo Chieffi
- Address correspondence to:Paolo Chieffi, Dipartimento di Psicologia, Viale Ellittico, 31 81100 Caserta, Italy. E-mail:
| |
Collapse
|
15
|
Cammarota M, de Rosa V, Pannaccione A, Secondo A, Tedeschi V, Piccialli I, Fiorino F, Severino B, Annunziato L, Boscia F. Rebound effects of NCX3 pharmacological inhibition: A novel strategy to accelerate myelin formation in oligodendrocytes. Biomed Pharmacother 2021; 143:112111. [PMID: 34481380 DOI: 10.1016/j.biopha.2021.112111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 08/20/2021] [Accepted: 08/23/2021] [Indexed: 11/18/2022] Open
Abstract
The Na+/Ca2+ exchanger NCX3 is an important regulator of sodium and calcium homeostasis in oligodendrocyte lineage. To date, no information is available on the effects resulting from prolonged exposure to NCX3 blockers and subsequent drug washout in oligodendroglia. Here, we investigated, by means of biochemical, morphological and functional analyses, the pharmacological effects of the NCX3 inhibitor, the 5-amino-N-butyl-2-(4-ethoxyphenoxy)-benzamide hydrochloride (BED), on NCXs expression and activity, as well as intracellular [Na+]i and [Ca2+]i levels, during treatment and following drug washout both in human MO3.13 oligodendrocytes and rat primary oligodendrocyte precursor cells (OPCs). BED exposure antagonized NCX activity, induced OPCs proliferation and [Na+]i accumulation. By contrast, 2 days of BED washout after 4 days of treatment significantly upregulated low molecular weight NCX3 proteins, reversed NCX activity, and increased intracellular [Ca2+]i. This BED-free effect was accompanied by an upregulation of NCX3 expression in oligodendrocyte processes and accelerated expression of myelin markers in rat primary oligodendrocytes. Collectively, our findings show that the pharmacological inhibition of the NCX3 exchanger with BED blocker maybe followed by a rebound increase in NCX3 expression and reversal activity that accelerate myelin sheet formation in oligodendrocytes. In addition, they indicate that a particular attention should be paid to the use of NCX inhibitors for possible rebound effects, and suggest that further studies will be necessary to investigate whether selective pharmacological modulation of NCX3 exchanger may be exploited to benefit demyelination and remyelination in demyelinating diseases.
Collapse
Affiliation(s)
- Mariarosaria Cammarota
- Division of Pharmacology, Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, Federico II University of Naples, Naples, Italy
| | - Valeria de Rosa
- Division of Pharmacology, Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, Federico II University of Naples, Naples, Italy
| | - Anna Pannaccione
- Division of Pharmacology, Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, Federico II University of Naples, Naples, Italy
| | - Agnese Secondo
- Division of Pharmacology, Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, Federico II University of Naples, Naples, Italy
| | - Valentina Tedeschi
- Division of Pharmacology, Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, Federico II University of Naples, Naples, Italy
| | - Ilaria Piccialli
- Division of Pharmacology, Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, Federico II University of Naples, Naples, Italy
| | | | - Beatrice Severino
- Department of Pharmacy, Federico II University of Naples, Naples, Italy
| | | | - Francesca Boscia
- Division of Pharmacology, Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, Federico II University of Naples, Naples, Italy.
| |
Collapse
|
16
|
Falvo S, Rosati L, Di Fiore MM, Di Giacomo Russo F, Chieffi Baccari G, Santillo A. Proliferative and Apoptotic Pathways in the Testis of Quail Coturnix coturnix during the Seasonal Reproductive Cycle. Animals (Basel) 2021; 11:ani11061729. [PMID: 34207904 PMCID: PMC8226535 DOI: 10.3390/ani11061729] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 05/27/2021] [Accepted: 05/31/2021] [Indexed: 11/30/2022] Open
Abstract
Simple Summary The quail Coturnix coturnix exhibits an annual cycle of testis size, sexual steroid production, and spermatogenesis. The testicular levels of both 17β-estradiol (E2) and androgens are higher during the reproductive period compared to the non-reproductive period, suggesting that estrogens act in synergy with the androgens for the initiation of spermatogenesis. Therefore, the present study aimed to investigate the estrogen responsive system in quail testis in relation to the reproduction seasons, with a focus on the molecular pathways activated in both active and regressive quail testes. The results indicated that estrogens participated in the activation of mitotic and meiotic events during the reproductive period by activating the ERK1/2 and Akt-1 pathways. In the non-reproductive period, when the E2/ERα levels are low, ERK1/2 and Akt-1 pathways remain inactive and apoptotic events occur. Our results suggest that the activation or inhibition of these molecular pathways plays a crucial role in the physiological switch “on/off” of the testicular activity in male quail during the seasonal reproductive cycle. Abstract The quail Coturnix coturnix is a seasonal breeding species, with the annual reproductive cycle of its testes comprising an activation phase and a regression phase. Our previous results have proven that the testicular levels of both 17β-estradiol (E2) and androgens are higher during the reproductive period compared to the non-reproductive period, which led us to hypothesize that estrogens and androgens may act synergistically to initiate spermatogenesis. The present study was, therefore, aimed to investigate the estrogen responsive system in quail testis in relation to the reproduction seasonality, with a focus on the molecular pathways elicited in both active and regressive quail testes. Western blotting and immunohistochemistry analysis revealed that the expression of ERα, which is the predominant form of estrogen receptors in quail testis, was correlated with E2 concentration, suggesting that increased levels of E2-induced ERα could play a key role in the resumption of spermatogenesis during the reproductive period, when both PCNA and SYCP3, the mitotic and meiotic markers, respectively, were also increased. In the reproductive period we also found the activation of the ERK1/2 and Akt-1 kinase pathways and an increase in second messengers cAMP and cGMP levels. In the non-reproductive phase, when the E2/ERα levels were low, the inactivation of ERK1/2 and Akt-1 pathways favored apoptotic events due to an increase in the levels of Bax and cytochrome C, with a consequent regression of the gonad.
Collapse
Affiliation(s)
- Sara Falvo
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università degli Studi della Campania “Luigi Vanvitelli”, 81100 Caserta, Italy; (S.F.); (M.M.D.F.); (F.D.G.R.); (G.C.B.)
| | - Luigi Rosati
- Dipartimento di Biologia, Università degli Studi di Napoli “Federico II”, 80138 Napoli, Italy;
| | - Maria Maddalena Di Fiore
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università degli Studi della Campania “Luigi Vanvitelli”, 81100 Caserta, Italy; (S.F.); (M.M.D.F.); (F.D.G.R.); (G.C.B.)
| | - Federica Di Giacomo Russo
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università degli Studi della Campania “Luigi Vanvitelli”, 81100 Caserta, Italy; (S.F.); (M.M.D.F.); (F.D.G.R.); (G.C.B.)
| | - Gabriella Chieffi Baccari
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università degli Studi della Campania “Luigi Vanvitelli”, 81100 Caserta, Italy; (S.F.); (M.M.D.F.); (F.D.G.R.); (G.C.B.)
| | - Alessandra Santillo
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università degli Studi della Campania “Luigi Vanvitelli”, 81100 Caserta, Italy; (S.F.); (M.M.D.F.); (F.D.G.R.); (G.C.B.)
- Correspondence:
| |
Collapse
|
17
|
miRNAs and Biomarkers in Testicular Germ Cell Tumors: An Update. Int J Mol Sci 2021; 22:ijms22031380. [PMID: 33573132 PMCID: PMC7866514 DOI: 10.3390/ijms22031380] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 01/21/2021] [Accepted: 01/27/2021] [Indexed: 12/12/2022] Open
Abstract
Testicular germ cell tumors (TGCTs) are the leading form of solid cancer and death affecting males between the ages of 20 and 40. Today, their surgical resection and chemotherapy are the treatments of first choice, even if sometimes this is not enough to save the lives of patients with TGCT. As seen for several tumors, the deregulation of microRNAs (miRNAs) is also a key feature in TGCTs. miRNAs are small molecules of RNA with biological activity that are released into biological fluids by testicular cancer cells. Their presence, therefore, can be detected and monitored by considering miRNAs as diagnostic and prognostic markers for TGCTs. The purpose of this review is to collect all the studies executed on miRNAs that have a potential role as biomarkers for testicular tumors.
Collapse
|
18
|
Chimento A, De Luca A, Nocito MC, Avena P, La Padula D, Zavaglia L, Pezzi V. Role of GPER-Mediated Signaling in Testicular Functions and Tumorigenesis. Cells 2020; 9:cells9092115. [PMID: 32957524 PMCID: PMC7563107 DOI: 10.3390/cells9092115] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/15/2020] [Accepted: 09/15/2020] [Indexed: 12/11/2022] Open
Abstract
Estrogen signaling plays important roles in testicular functions and tumorigenesis. Fifteen years ago, it was discovered that a member of the G protein-coupled receptor family, GPR30, which binds also with high affinity to estradiol and is responsible, in part, for the rapid non-genomic actions of estrogens. GPR30, renamed as GPER, was detected in several tissues including germ cells (spermatogonia, spermatocytes, spermatids) and somatic cells (Sertoli and Leydig cells). In our previous review published in 2014, we summarized studies that evidenced a role of GPER signaling in mediating estrogen action during spermatogenesis and testis development. In addition, we evidenced that GPER seems to be involved in modulating estrogen-dependent testicular cancer cell growth; however, the effects on cell survival and proliferation depend on specific cell type. In this review, we update the knowledge obtained in the last years on GPER roles in regulating physiological functions of testicular cells and its involvement in neoplastic transformation of both germ and somatic cells. In particular, we will focus our attention on crosstalk among GPER signaling, classical estrogen receptors and other nuclear receptors involved in testis physiology regulation.
Collapse
Affiliation(s)
- Adele Chimento
- Correspondence: (A.C.); (V.P.); Tel.: +39-0984-493184 (A.C.); +39-0984-493148 (V.P.)
| | | | | | | | | | | | - Vincenzo Pezzi
- Correspondence: (A.C.); (V.P.); Tel.: +39-0984-493184 (A.C.); +39-0984-493148 (V.P.)
| |
Collapse
|
19
|
De Martino M, Esposito F, Chieffi P. An update on microRNAs as potential novel therapeutic targets in testicular germ cell tumors. Intractable Rare Dis Res 2020; 9:184-186. [PMID: 32844079 PMCID: PMC7441029 DOI: 10.5582/irdr.2020.03025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Testicular germ cell tumors (TGCTs) are the most frequent solid malignant tumors in men 20- 40 years of age and the most frequent cause of death from solid tumors in this age group. Recent studies have underscored the fact that miRNA deregulation is a feature of carcinogenesis, including TGCT development and progression. MiRNAs are a group of small noncoding RNAs that bind to the 3'-untranslated region (UTR) of the targeted mRNAs, thus causing mRNA degradation or the inhibition of its translation, regulating gene expression in a temporal and tissue-specific manner. However, few miRNAs have been found to play key roles in TGCTs; recently, other miRNAs have been identified, representing novel potential therapeutic targets.
Collapse
Affiliation(s)
- Marco De Martino
- Dipartimento di Psicologia, Università della Campania "Luigi Vanvitelli", Caserta, Italy
- Istituto di Endocrinologia ed Oncologia Sperimentale-CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", Napoli, Italy
| | - Francesco Esposito
- Istituto di Endocrinologia ed Oncologia Sperimentale-CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", Napoli, Italy
| | - Paolo Chieffi
- Dipartimento di Psicologia, Università della Campania "Luigi Vanvitelli", Caserta, Italy
- Address correspondence to:Paolo Chieffi, Dipartimento di Psicologia, Università della Campania "Luigi Vanvitelli", Caserta, 31 81100 Caserta, Italy. E-mail:
| |
Collapse
|
20
|
Pan MX, Li J, Ma C, Fu K, Li ZQ, Wang ZF. Sex-dependent effects of GPER activation on neuroinflammation in a rat model of traumatic brain injury. Brain Behav Immun 2020; 88:421-431. [PMID: 32272225 DOI: 10.1016/j.bbi.2020.04.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 04/03/2020] [Accepted: 04/04/2020] [Indexed: 02/06/2023] Open
Abstract
The G protein-coupled estrogen receptor (GPER) plays a role in estrogen-mediated neuroprotection and has been considered a potential therapeutic target for treating various neurological diseases. It is increasingly recognized that sex is a biological variable affecting treatment outcomes and efficacy, and that neuroinflammation is a key secondary injury mechanism following brain injury, though it is unknown whether the neuroprotective effects exerted by GPER involve modulation of inflammatory processes. The aim of this study was to investigate whether activation of GPER has a sex-dependent effect on neuroinflammation following traumatic brain injury (TBI), a sexually dimorphic disease. In male and ovariectomized (OVX) female rats, the GPER agonist, G1, inhibited the upregulated expression of pro-inflammatory cytokines (IL-1β, IL-6, and TNF-α), increased the expression of the anti-inflammatory cytokine IL-4, and shifted microglia/macrophage polarization toward the M2 phenotype. In gonadally-intact females, G1 caused more pro-inflammatory (IL-6 and TNF-α) and less anti-inflammatory cytokine (IL-4) production, without altering microglia/macrophage polarization. Estradiol supplementation blocked the effects of G1 in OVX females. We also found that post-injury GPER expression was increased in males and OVX females but not in intact females. G1 administration increased Akt phosphorylation in males and OVX females, but had no significant effect in intact females, while Akt inhibition blocked the effects of G1 in males and OVX females. These results indicate that G1 exerts anti-inflammatory effects in males and OVX females but not in intact females; these sex-specific effects are dependent on circulating estrogen levels and are partially mediated through Akt signaling. Future studies are needed to elucidate the relevant molecular mechanisms, especially in females. A better understanding of the sex differences in treatment efficacy with GPER agonists may help improve personalized therapeutic strategies for males and pre- and postmenopausal females with TBI.
Collapse
Affiliation(s)
- Meng-Xian Pan
- Department of Physiology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Jie Li
- Department of Physiology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Chao Ma
- Department of Neurosurgery, Wuhan University Zhongnan Hospital, Wuhan 430071, China
| | - Kai Fu
- Department of Neurosurgery, Wuhan University Zhongnan Hospital, Wuhan 430071, China
| | - Zhi-Qiang Li
- Department of Neurosurgery, Wuhan University Zhongnan Hospital, Wuhan 430071, China.
| | - Ze-Fen Wang
- Department of Physiology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China.
| |
Collapse
|
21
|
HMGA1-Regulating microRNAs Let-7a and miR-26a are Downregulated in Human Seminomas. Int J Mol Sci 2020; 21:ijms21083014. [PMID: 32344629 PMCID: PMC7215726 DOI: 10.3390/ijms21083014] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 04/21/2020] [Accepted: 04/22/2020] [Indexed: 02/08/2023] Open
Abstract
Background: Recent studies have underlined HMGA protein’s key role in the onset of testicular germ cell tumors, where HMGA1 is differently expressed with respect to the state of differentiation, suggesting its fine regulation as master regulator in testicular tumorigenesis. Several studies have highlighted that the HMGA1 transcript is strictly regulated by a set of inhibitory microRNAs. Thus, the aim of this study is to test whether HMGA1 overexpression in human seminomas may be induced by the deregulation of miR-26a and Let-7a—two HMGA1-targeting microRNAs. Methods: HMGA1 mRNA and Let-7a and miR-26a levels were measured in a seminoma dataset available in the Cancer Genome Atlas database and confirmed in a subset of seminomas by qRT-PCR and western blot. A TCam-2 seminoma cell line was then transfected with Let-7a and miR-26a and tested for proliferation and motility abilities. Results: an inverse correlation was found between the expression of miR-26a and Let-7a and HMGA1 expression levels in seminomas samples, suggesting a critical role of these microRNAs in HMGA1 levels regulation. Accordingly, functional studies showed that miR-26a and Let-7a inhibited the proliferation, migration and invasion capabilities of the human seminoma derived cell line TCam-2. Conclusions: these data strongly support that the upregulation of HMGA1 levels occurring in seminoma is—at least in part—due to the downregulation of HMGA1-targeting microRNAs.
Collapse
|
22
|
Kotula-Balak M, Duliban M, Pawlicki P, Tuz R, Bilinska B, Płachno BJ, Arent ZJ, Krakowska I, Tarasiuk K. The meaning of non-classical estrogen receptors and peroxisome proliferator-activated receptor for boar Leydig cell of immature testis. Acta Histochem 2020; 122:151526. [PMID: 32094002 DOI: 10.1016/j.acthis.2020.151526] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 02/10/2020] [Accepted: 02/10/2020] [Indexed: 01/09/2023]
Abstract
Communication in biological systems involves diverse-types of cell-cell interaction including cross-talk between receptors expressed by the target cells. Recently, novel sort of estrogen receptors (G protein - coupled estrogen receptor; GPER and estrogen-related receptor; ERR) that signal directly via estrogen binding and/or via mutual interaction-regulated estrogen signaling were reported in various organs including testis. Peroxisome proliferator - activated receptor (PPAR) is responsible for maintaining of lipid homeostasis that is critical for sex steroid production in the testis. Here, we investigated the role of interaction between GPER, ERRβ and PPARγ in steroidogenic Leydig cells of immature boar testis. Testicular fragments cultured ex vivo were treated with GPER or PPARγ antagonists. Then, cell ultrastructure, expression and localization of GPER, ERRβ, PPARγ together with the molecular receptor mechanism, through cyclic AMP and Raf/Ras/extracellular signal activated kinases (ERK), in the control of cholesterol concentration and estrogen production by Leydig cells were studied. In the ultrastructure of antagonist-treated Leydig cells, mitochondria were not branched and not bifurcated as they were found in control. Additionally, in PPARγ-blocked Leydig cells changes in the number of lipid droplets were revealed. Independent of used antagonist, western blot revealed decreased co-expression of GPER, ERRβ, PPARγ with exception of increased expression of ERRβ after PPARγ blockage. Immunohistochemistry confirmed presence of all receptors partially located in the nucleus or cytoplasm of Leydig cells of both control and treated testes. Changes in receptor expression, decreased cholesterol and increased estradiol tissue concentrations occurred through decreased cAMP level (with exception after GPER blockage) as well as Raf/Ras/ERK pathway expression. These all findings indicate that GPER-ERRβ-PPARγ interaction exists in immature boar testis and regulates Leydig cell function. Further detailed studies and considerations on GPER-ERRβ-PPARγ as possible diagnosis/therapy target in disturbances of testis steroidogenic function are needed.
Collapse
Affiliation(s)
- M Kotula-Balak
- University Centre of Veterinary Medicine, University of Agriculture in Krakow, Mickiewicza 24/28, 30-059, Krakow, Poland.
| | - M Duliban
- Department of Endocrinology, Institute of Zoology, Jagiellonian University in Krakow, Gronostajowa 9, 30-387 Krakow, Poland
| | - P Pawlicki
- Department of Endocrinology, Institute of Zoology, Jagiellonian University in Krakow, Gronostajowa 9, 30-387 Krakow, Poland
| | - R Tuz
- Department of Swine and Small Animal Breeding, Institute of Animal Sciences, University of Agriculture in Krakow, Mickiewicza 24/28, 30-059 Krakow, Poland
| | - B Bilinska
- Department of Endocrinology, Institute of Zoology, Jagiellonian University in Krakow, Gronostajowa 9, 30-387 Krakow, Poland
| | - B J Płachno
- Department of Plant Cytology and Embryology, Institute of Botany, Jagiellonian University in Kraków, Poland, Gronostajowa 9, 30-387 Krakow, Poland
| | - Z J Arent
- University Centre of Veterinary Medicine, University of Agriculture in Krakow, Mickiewicza 24/28, 30-059, Krakow, Poland
| | - I Krakowska
- University Centre of Veterinary Medicine, University of Agriculture in Krakow, Mickiewicza 24/28, 30-059, Krakow, Poland
| | - K Tarasiuk
- University Centre of Veterinary Medicine, University of Agriculture in Krakow, Mickiewicza 24/28, 30-059, Krakow, Poland
| |
Collapse
|
23
|
Chieffi P, De Martino M, Esposito F. Further insights into testicular germ cell tumor oncogenesis: potential therapeutic targets. Expert Rev Anticancer Ther 2020; 20:189-195. [PMID: 32164473 DOI: 10.1080/14737140.2020.1736566] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Testicular germ cell tumors (TGCTs) are the most common neoplasia in the young male population, and the incidence has been constantly increasing in many parts of the world. These tumors are classified into seminomas and non-seminomas, and those divided, in turn, into yolk sac tumors, embryonal cell carcinomas, choriocarcinomas, and teratomas. Although therapeutic approaches have improved, approximately 25% of the patients relapse or, in a small number of cases, show platinum-resistant disease.Areas covered: We review several molecular targets that have recently emerged as powerful tools for both diagnosis and therapy of TGCTs. Moreover, we reviewed the most frequent deregulated pathways involved in TGCT tumorigenesis, reporting drugs that may emerge as novel therapeutic agents.Expert opinion: TGCT treatment is mainly based on platinum-derivative therapy with high cure rates. However, in the refractory patients, there are few alternative treatments. Thus, different pharmacological approaches have to be thoroughly investigated to shed new light on TGCT pathogenesis and treatment.
Collapse
Affiliation(s)
- Paolo Chieffi
- Dipartimento di Psicologia, Università della Campania, Caserta, Italy
| | - Marco De Martino
- Dipartimento di Psicologia, Università della Campania, Caserta, Italy.,Istituto di Endocrinologia ed Oncologia Sperimentale del CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Scuola di Medicina e Chirurgia di Napoli, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Francesco Esposito
- Istituto di Endocrinologia ed Oncologia Sperimentale del CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Scuola di Medicina e Chirurgia di Napoli, Università degli Studi di Napoli 'Federico II', Naples, Italy
| |
Collapse
|
24
|
Chevalier N, Hinault C, Clavel S, Paul-Bellon R, Fenichel P. GPER and Testicular Germ Cell Cancer. Front Endocrinol (Lausanne) 2020; 11:600404. [PMID: 33574796 PMCID: PMC7870790 DOI: 10.3389/fendo.2020.600404] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 12/07/2020] [Indexed: 12/12/2022] Open
Abstract
The G protein-coupled estrogen receptor (GPER), also known as GPR30, is a widely conserved 7-transmembrane-domain protein which has been identified as a novel 17β-estradiol-binding protein that is structurally distinct from the classic oestrogen receptors (ERα and ERβ). There are still conflicting data regarding the exact role and the natural ligand of GPER/GPR30 in reproductive tracts as both male and female knock-out mice are fertile and have no abnormalities of reproductive organs. Testicular germ cell cancers (TGCCs) are the most common malignancy in young males and the most frequent cause of death from solid tumors in this age group. Clinical and experimental studies suggested that estrogens participate in the physiological and pathological control of male germ cell proliferation. In human seminoma cell line, while 17β-estradiol (E2) inhibits in vitro cell proliferation through an ERβ-dependent mechanism, an impermeable E2 conjugate (E2 coupled to BSA), in vitro cell proliferation is stimulated by activating ERK1/2 and protein kinase A through a membrane GPCR that we further identified as GPER/GPR30. The same effect was observed with low but environmentally relevant doses of BPA, an estrogenic endocrine disrupting compound. Furthermore, GPER/GPR30 is specifically overexpressed in seminomas but not in non-seminomas and this overexpression is correlated with an ERβ-downregulation. This GPER/GPR30 overexpression could be linked to some genetic variations, as single nucleotide polymorphisms, which was also reported in other hormone-dependent cancers. We will review here the implication of GPER/GPR30 in TGCCs pathophysiology and the arguments to consider GPER/GPR30 as a potential therapeutic target in humans.
Collapse
Affiliation(s)
- Nicolas Chevalier
- Université Côte d’Azur, CHU, INSERM U1065, C3M, Nice, France
- Université Côte d’Azur, INSERM U1065, C3M, Nice, France
- *Correspondence: Nicolas Chevalier, ;
| | - Charlotte Hinault
- Université Côte d’Azur, CHU, INSERM U1065, C3M, Nice, France
- Université Côte d’Azur, INSERM U1065, C3M, Nice, France
| | | | | | - Patrick Fenichel
- Université Côte d’Azur, CHU, INSERM U1065, C3M, Nice, France
- Université Côte d’Azur, INSERM U1065, C3M, Nice, France
| |
Collapse
|
25
|
Fénichel P, Chevalier N. Is Testicular Germ Cell Cancer Estrogen Dependent? The Role of Endocrine Disrupting Chemicals. Endocrinology 2019; 160:2981-2989. [PMID: 31617897 DOI: 10.1210/en.2019-00486] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/11/2019] [Indexed: 11/19/2022]
Abstract
Testicular germ cell cancer (TGCC) is the most frequent cancer of the young male, with an increasing incidence worldwide. The pathogenesis and reasons for this increase remain unknown. However, epidemiological and experimental data have suggested that, similar to genital malformations and sperm impairment, it could result from the interaction of genetic and environmental factors including fetal exposure to endocrine-disrupting chemicals (EDCs) with estrogenic effects. In this review, we analyze the expression of classic and nonclassic estrogen receptors by TGCC cells, the way they may influence germ cell proliferation induced by EDCs, and discuss how this estrogen dependency supports the developmental and environmental hypothesis.
Collapse
Affiliation(s)
- Patrick Fénichel
- Université Côte d'Azur, CHU de Nice, Service d'Endocrinologie, Diabétologie et Médecine de la Reproduction, Hôpital de l'Archet, France
- Université Côte d'Azur, Centre Hospitalier Universitaire de Nice, INSERM U1065/UNS, Centre Méditerranéen de Médecine Moléculaire, Equipe, France
| | - Nicolas Chevalier
- Université Côte d'Azur, CHU de Nice, Service d'Endocrinologie, Diabétologie et Médecine de la Reproduction, Hôpital de l'Archet, France
- Université Côte d'Azur, Centre Hospitalier Universitaire de Nice, INSERM U1065/UNS, Centre Méditerranéen de Médecine Moléculaire, Equipe, France
| |
Collapse
|
26
|
Milon A, Kaczmarczyk M, Pawlicki P, Bilinska B, Duliban M, Gorowska-Wojtowicz E, Tworzydlo W, Kotula-Balak M. Do estrogens regulate lipid status in testicular steroidogenic Leydig cell? Acta Histochem 2019; 121:611-618. [PMID: 31126612 DOI: 10.1016/j.acthis.2019.04.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/26/2019] [Accepted: 04/26/2019] [Indexed: 12/24/2022]
Abstract
In this study mouse Leydig cell (MA-10) were treated with G-protein coupled membrane estrogen receptor antagonist (G-15; 10 nM). Cells were analyzed by Western blotting for expression of estrogen-related receptors (ERRα, β and γ), steroidogenic markers (lutropin receptor; LHR and 3β-hydroxysteroid dehydrogenase; 3β-HSD) and lipid droplet markers (perilipin; PLIN and microtubule-associated protein 1 A/1B-light chain 3; LC3). Concomitantly, microscopic analyses by light microscope (immunofluorescent staining for lipid droplets, PLIN and LC3) as well as by electron microscope (for lipid droplet ultrastructure) were utilized. For analysis of cholesterol content, cAMP level and progesterone secretion, G-15, estrogen receptor (ER) antagonist (ICI 182,780; 10 μM), 17β-estradiol (10 mM) and, bisphenol A (BPA; 10 nM) were used alone or in combinations. We revealed no changes in ERRs expression but alterations in ERRβ and γ localization in G-15-treated cells when compared to control. Partial translocation of ERRβ and γ from the cell nucleus to cytoplasm was observed. Decreased expression of LHR, 3β-HSD, PLIN and LC3 was detected. Moreover, in treated cells large lipid droplets and differences in their distribution were found. Very strong signal of co-localization for PLIN and LC3 was found in treated cells when compared to control. In ultrastructure of treated cells, degenerating lipid droplets and double membrane indicating on presence of lipophagosome were observed. We found, that only (i) BPA and G-15 did not effect on cholesterol content, (ii) BPA, G-15 and ICI did not effect on cAMP level and (iii) BPA, ICI alone and in combination, and BPA with G-15 did not modulate progesterone secretion. These findings showed complex and diverse estrogen effects on mouse Leydig cells at various steps of steroid hormone production (cholesterol storage, release and processing). Lipid homeostasis and metabolism in these cells were affected by endogenous and exogenous estrogen, interactions of receptors (GPER, ER and ERR) and GPER and ER antagonists.
Collapse
Affiliation(s)
- A Milon
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Gronostajowa 9, 30-387, Krakow, Poland
| | - M Kaczmarczyk
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Gronostajowa 9, 30-387, Krakow, Poland
| | - P Pawlicki
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Gronostajowa 9, 30-387, Krakow, Poland
| | - B Bilinska
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Gronostajowa 9, 30-387, Krakow, Poland
| | - M Duliban
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Gronostajowa 9, 30-387, Krakow, Poland
| | - E Gorowska-Wojtowicz
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Gronostajowa 9, 30-387, Krakow, Poland
| | - W Tworzydlo
- Department of Developmental Biology and Invertebrate Morphology Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Gronostajowa 9, 30-387, Krakow, Poland
| | - M Kotula-Balak
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Gronostajowa 9, 30-387, Krakow, Poland.
| |
Collapse
|
27
|
Abstract
Testicular germ cell tumors (TGCTs) are the most frequent solid malignant tumors in men 20-34 years of age and the most frequent cause of death from solid tumors in this age group. In addition, the incidence of these tumors has significantly increased over the last few decades. Testicular germ cell tumors are classified into seminoma and nonseminoma germ cell tumors (NSGCTs). NSGCTs can be further divided into embryonal carcinoma, Teratoma, yolk sac tumor, and choriocarcinoma. There are noteworthy differences about therapy and prognosis of seminomas and nonseminoma germ cell tumors, even though both share characteristics of the primordial germ cells (PGCs). Many discovered biomarkers including HMGA1, GPR30, Aurora-B, estrogen receptor β, and others have given further advantage to discriminate between histological subgroups and could represent useful molecular therapeutic targets.
Collapse
Affiliation(s)
- Paolo Chieffi
- Dipartimento di Psicologia, Università della Campania, Caserta, Italy
- Address correspondence to:Dr. Paolo Chieffi, Dipartimento di Psicologia, Università della Campania, Viale Ellittico, 3181100 Caserta, Italy. E-mail:
| |
Collapse
|
28
|
Chieffi P, De Martino M, Esposito F. New Anti-Cancer Strategies in Testicular Germ Cell Tumors. Recent Pat Anticancer Drug Discov 2019; 14:53-59. [DOI: 10.2174/1574892814666190111120023] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/10/2018] [Accepted: 01/02/2019] [Indexed: 11/22/2022]
Abstract
Background: The most common solid malignancy of young men aged 20 to 34 years is testicular germ cell tumor. In addition, the incidence of these tumors has significantly increased throughout the last years. Testicular germ cell tumors are classified into seminoma and nonseminoma germ cell tumors, which take in yolk sac tumor, embryonal cell carcinoma, choriocarcinoma, and teratoma. There are noteworthy differences about therapy and prognosis of seminomas and nonseminoma germ cell tumors, even though both share characteristics of the primordial germ cells. </P><P> Objectives: The study is focused on different molecular mechanisms strongly involved in testicular germ cell line tumors underlying new strategies to treat this human neoplasia.Methods:Bibliographic data from peer-reviewed research, patent and clinical trial literature, and around eighty papers and patents have been included in this review.Results:Our study reveals that several biomarkers are usefully utilized to discriminate among different histotypes. Moreover, we found new patents regarding testicular germ cell tumor treatments such as the expression of claudin 6, monoclonal antibody (Brentuximab Vedotin), immune checkpoint blockade (ICB) with the FDA-approved drugs pembrolizumab and nivolumab or the oncolytic virus Pelareorep, the combination of selective inhibitors of Aurora kinase.Conclusion:Finally, the pathogenesis of testicular germ cell tumor needs to be deeply understood so that it will improve data on stem cells, tumorigenesis and disease tumor management by more selective treatment.
Collapse
Affiliation(s)
- Paolo Chieffi
- Department of Psychology, University of Campania, 81100 Caserta, Italy
| | - Marco De Martino
- Department of Psychology, University of Campania, 81100 Caserta, Italy
| | - Francesco Esposito
- Institute of Endocrinology and Experimental Oncology of the CNR c / o Department of Molecular Medicine and Medical Biotechnology, School of Medicine and Surgery of Naples, University of Naples 'Federico II', Naples, Italy
| |
Collapse
|
29
|
Pawlicki P, Hejmej A, Milon A, Lustofin K, Płachno BJ, Tworzydlo W, Gorowska-Wojtowicz E, Pawlicka B, Kotula-Balak M, Bilinska B. Telocytes in the mouse testicular interstitium: implications of G-protein-coupled estrogen receptor (GPER) and estrogen-related receptor (ERR) in the regulation of mouse testicular interstitial cells. PROTOPLASMA 2019; 256:393-408. [PMID: 30187340 PMCID: PMC6510843 DOI: 10.1007/s00709-018-1305-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 08/24/2018] [Indexed: 05/12/2023]
Abstract
Telocytes (TCs), a novel type of interstitial cells, are involved in tissue homeostasis maintenance. This study aimed to investigate TC presence in the interstitium of mouse testis. Additionally, inactivation of the G-coupled membrane estrogen receptor (GPER) in the testis was performed to obtain insight into TC function, regulation, and interaction with other interstitial cells. Mice were injected with a GPER antagonist (G-15; 50 μg/kg bw), and the GPER-signaling effect on TC distribution, ultrastructure, and function, as well as the interstitial tissue interaction of GPER with estrogen-related receptors (ERRs), was examined. Microscopic observations of TC morphology were performed with the use of scanning and transmission electron microscopes. Telocyte functional markers (CD34; c-kit; platelet-derived growth factor receptors α and β, PDGFRα and β; vascular endothelial growth factor, VEGF; and vimentin) were analyzed by immunohistochemistry/immunofluorescence and Western blot. mRNA expression of CD34 as well as ERR α, β, and γ was measured by qRT-PCR. Relaxin and Ca2+ concentrations were analyzed by immunoenzymatic and colorimetric assays, respectively. For the first time, we reveal the presence of TCs in the interstitium together with the peritubular area of mouse testis. Telocytes were characterized by specific features such as a small cell body and extremely long prolongations, constituting a three-dimensional network mainly around the interstitial cells. Expression of all TC protein markers was confirmed. Based on scanning electron microscopic observation in GPER-blocked testis, groups of TCs were frequently seen. No changes were found in TC ultrastructure in GPER-blocked testis when compared to the control. However, tendency to TC number change (increase) after the blockage was observed. Concomitantly, no changes in mRNA CD34 expression and increase in ERR expression were detected in GPER-blocked testes. In addition, Ca2+ was unchanged; however, an increase in relaxin concentration was observed. Telocytes are an important component of the mouse testicular interstitium, possibly taking part in maintaining its microenvironment as well as contractile and secretory functions (via themselves or via controlling of other interstitial cells). These cells should be considered a unique and useful target cell type for the prevention and treatment of testicular interstitial tissue disorders based on estrogen-signaling disturbances.
Collapse
Affiliation(s)
- Piotr Pawlicki
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Gronostajowa 9, 30-387, Krakow, Poland
| | - Anna Hejmej
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Gronostajowa 9, 30-387, Krakow, Poland
| | - Agnieszka Milon
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Gronostajowa 9, 30-387, Krakow, Poland
| | - Krzysztof Lustofin
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Gronostajowa 9, 30-387, Krakow, Poland
| | - Bartosz J Płachno
- Department of Plant Cytology and Embryology, Institute of Botany, Jagiellonian University in Kraków, Gronostajowa 9, 30-387, Krakow, Poland
| | - Waclaw Tworzydlo
- Department of Developmental Biology and Invertebrate Morphology, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Gronostajowa 9, 30-387, Krakow, Poland
| | - Ewelina Gorowska-Wojtowicz
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Gronostajowa 9, 30-387, Krakow, Poland
| | - Bernadetta Pawlicka
- Department of Genetics and Evolution, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Gronostajowa 9, 30-387, Krakow, Poland
| | - Malgorzata Kotula-Balak
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Gronostajowa 9, 30-387, Krakow, Poland.
| | - Barbara Bilinska
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Gronostajowa 9, 30-387, Krakow, Poland
| |
Collapse
|
30
|
Abstract
A critical step for maintenance of genetic stability is chromosome segregation, which requires a high coordination of cellular processes. Loss of mitotic regulation is a possible cause of aneuploidy in human epithelial malignancy and it is thought to create an abnormal nuclear morphology in cancer cells. Serine/threonine protein kinase Aurora B gene plays a regulatory role from G2 to cytokinesis, encompassing key cell cycle events such as centrosome duplication, chromosome bi-orientation, and segregation. The overexpression of Aurora B has been observed in several tumour types, and has been linked with a poor prognosis for cancer patients. Therapeutic inhibition of Aurora kinase showed great promise as a probable anticancer regime because of its important role during cell division.
Collapse
Affiliation(s)
- Paolo Chieffi
- Dipartimento di Psicologia, Università della Campania, Caserta, Italy
- Address correspondence to:Dr. Paolo Chieffi, Dipartimento di Psicologia, Università della Campania, Viale Ellittico, 31 81100 Caserta, Italy. E-mail:
| |
Collapse
|
31
|
Hadjimarkou MM, Vasudevan N. GPER1/GPR30 in the brain: Crosstalk with classical estrogen receptors and implications for behavior. J Steroid Biochem Mol Biol 2018; 176:57-64. [PMID: 28465157 DOI: 10.1016/j.jsbmb.2017.04.012] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 04/16/2017] [Accepted: 04/23/2017] [Indexed: 12/31/2022]
Abstract
The GPER1/GPR30 is a membrane estrogen receptor (mER) that binds 17β-estradiol (17β-E) with high affinity and is thought to play a role in cancer progression and cardiovascular health. Though widespread in the central nervous system, less is known about this receptor's function in the brain. GPER1 has been shown to activate kinase cascades and calcium flux within cells rapidly, thus fitting in with the idea of being a mER that mediates non-genomic signaling by estrogens. Signaling from GPER1 has been shown to improve spatial memory, possibly via release of neurotransmitters and generation of new spines on neurons in the hippocampus. In addition, GPER1 activation contributes to behaviors that denote anxiety and to social behaviors such as social memory and lordosis behavior in mice. In the male hippocampus, GPER1 activation has also been shown to phosphorylate the classical intracellular estrogen receptor (ER)α, suggesting that crosstalk with ERα is important in the display of these behaviors, many of which are absent in ERα-null mice. In this review, we present a number of categories of such crosstalk, using examples from literature. The function of GPER1 as an ERα collaborator or as a mER in different tissues is relevant to understanding both normal physiology and abnormal pathology, mediated by estrogen signaling.
Collapse
Affiliation(s)
- Maria M Hadjimarkou
- School of Humanities and Social Sciences, University of Nicosia, 1700 Nicosia, Cyprus.
| | - Nandini Vasudevan
- School of Biological Sciences, University of Reading, Reading, United Kingdom RG6 6AS, United Kingdom.
| |
Collapse
|
32
|
Abstract
Testicular germ cell tumor (TGCT) is the most common solid malignancy occurring in young men between 20 and 34 years of age, and its incidence has increased significantly over the last decades. Clinically several types of immunohistochemical markers are useful and sensitive. These new biomarkers are genes expressed in primordial germ cells/gonocytes and embryonic pluripotency-related cells but not in normal adult germ cells and they include OCT3/4, HMGA1 and 2, NANOG, SOX2, and LIN28. Gene expression in TGCT is regulated, at least in part, by DNA and histone modifications, and the epigenetic profile of these tumours is characterised by genome-wide demethylation. There are different epigenetic modifications in TGCT subtypes that reflect the normal developmental switch in primordial germ cells from an under to normally methylated genome.
Collapse
Affiliation(s)
- Paolo Chieffi
- Dipartimento di Psicologia, Università della Campania, Caserta, Italy
- Address correspondence to: Dr. Paolo Chieffi, Dipartimento di Psicologia, Viale Ellittico, 31 81100 Caserta, Italy. E-mail:
| |
Collapse
|
33
|
Fábián M, Rencz F, Krenács T, Brodszky V, Hársing J, Németh K, Balogh P, Kárpáti S. Expression of G protein-coupled oestrogen receptor in melanoma and in pregnancy-associated melanoma. J Eur Acad Dermatol Venereol 2017; 31:1453-1461. [PMID: 28467693 DOI: 10.1111/jdv.14304] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 03/29/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND The hormone sensitivity of melanoma and the role of 'classical' oestrogen receptor (ER) α and β in tumour progression have been intensively studied with rather contradictory results. The presence of 'non-classical' G protein-coupled oestrogen receptor (GPER) has not been investigated on human melanoma tissues. OBJECTIVE To analyse the expression of GPER, ERα and ERβ in pregnancy-associated (PAM) and in non-pregnancy-associated (NPAM) melanomas in correlation with traditional prognostic markers and disease-free survival (DFS). METHODS Receptor protein levels were tested using immunohistochemistry in 81 formalin-fixed paraffin-embedded melanoma tissues. PAMs (n = 38) were compared with age- and Breslow thickness-matched cases (n = 43) including non-pregnant women (NPAM-W) (n = 22) and men (NPAM-M) (n = 21). The association between receptor expression and DFS was analysed by uni- and multivariate Cox proportional hazards regression. RESULTS G protein-coupled oestrogen receptor was detected both in PAMs and NPAMs. In 39 of the 41 (95.1%) GPER-positive melanomas, GPER and ERβ were co-expressed. GPER/ERβ-positive melanomas were significantly more common in PAM compared to NPAM (P = 0.0001) with no significant difference between genders (P = 0.4383). In PAMs, the distribution of GPER and ERβ was similar (78.4% vs. 81.6%; P = 0.8504), while in NPAM, ERβ was the representative ER (60.5% vs. 27.9%; P = 0.0010) without gender difference (59.1% vs. 61.9%). GPER-/ERβ-positive melanomas were associated with lower Breslow thickness, lower mitotic rate and higher presence of peritumoral lymphocyte infiltration (PLI) compared to GPER-/ERβ-negative cases (P = 0.0156, P = 0.0036 and P = 0.0001) predicting a better DFS (HR = 0.785, 95% CI 0.582-1.058). Despite the significantly higher frequency of GPER and ERβ expression in PAM, no significant difference was found in DFS between PAM and NPAM. All but one case failed to show ERα expression. CONCLUSIONS The presence of GPER and its simultaneous expression with ERβ can serve as a new prognostic indicator in a significant subpopulation of melanoma patients.
Collapse
Affiliation(s)
- M Fábián
- Department of Dermatology, Venereology and Dermatooncology, Semmelweis University, Budapest, Hungary.,Doctoral School of Clinical Medicine, Semmelweis University, Budapest, Hungary
| | - F Rencz
- Department of Health Economics, Corvinus University of Budapest, Budapest, Hungary
| | - T Krenács
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary.,MTA-SE Tumour Progression Research Group, Budapest, Hungary
| | - V Brodszky
- Department of Health Economics, Corvinus University of Budapest, Budapest, Hungary
| | - J Hársing
- Department of Dermatology, Venereology and Dermatooncology, Semmelweis University, Budapest, Hungary
| | - K Németh
- Department of Dermatology, Venereology and Dermatooncology, Semmelweis University, Budapest, Hungary
| | - P Balogh
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - S Kárpáti
- Department of Dermatology, Venereology and Dermatooncology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
34
|
Chieffi S, Carotenuto M, Monda V, Valenzano A, Villano I, Precenzano F, Tafuri D, Salerno M, Filippi N, Nuccio F, Ruberto M, De Luca V, Cipolloni L, Cibelli G, Mollica MP, Iacono D, Nigro E, Monda M, Messina G, Messina A. Orexin System: The Key for a Healthy Life. Front Physiol 2017; 8:357. [PMID: 28620314 PMCID: PMC5450021 DOI: 10.3389/fphys.2017.00357] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 05/15/2017] [Indexed: 11/30/2022] Open
Abstract
The orexin-A/hypocretin-1 and orexin-B/hypocretin-2 are neuropeptides synthesized by a cluster of neurons in the lateral hypothalamus and perifornical area. Orexin neurons receive a variety of signals related to environmental, physiological and emotional stimuli, and project broadly to the entire CNS. Orexin neurons are “multi-tasking” neurons regulating a set of vital body functions, including sleep/wake states, feeding behavior, energy homeostasis, reward systems, cognition and mood. Furthermore, a dysfunction of orexinergic system may underlie different pathological conditions. A selective loss orexin neurons was found in narcolepsia, supporting the crucial role of orexins in maintaining wakefulness. In animal models, orexin deficiency lead to obesity even if the consume of calories is lower than wildtype counterpart. Reduced physical activity appears the main cause of weight gain in these models resulting in energy imbalance. Orexin signaling promotes obesity resistance via enhanced spontaneous physical activity and energy expenditure regulation and the deficiency/dysfunction in orexins system lead to obesity in animal models despite of lower calories intake than wildtype associated with reduced physical activity. Interestingly, orexinergic neurons show connections to regions involved in cognition and mood regulation, including hippocampus. Orexins enhance hippocampal neurogenesis and improve spatial learning and memory abilities, and mood. Conversely, orexin deficiency results in learning and memory deficits, and depression.
Collapse
Affiliation(s)
- Sergio Chieffi
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetic and Sport Medicine, Università degli Studi della Campania "Luigi Vanvitelli"Naples, Italy
| | - Marco Carotenuto
- Department of Mental Health, Physical and Preventive Medicine, Clinic of Child and Adolescent Neuropsychiatry, Università degli Studi della Campania "Luigi Vanvitelli"Naples, Italy
| | - Vincenzo Monda
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetic and Sport Medicine, Università degli Studi della Campania "Luigi Vanvitelli"Naples, Italy
| | - Anna Valenzano
- Department of Clinical and Experimental Medicine, University of FoggiaFoggia, Italy
| | - Ines Villano
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetic and Sport Medicine, Università degli Studi della Campania "Luigi Vanvitelli"Naples, Italy
| | - Francesco Precenzano
- Department of Mental Health, Physical and Preventive Medicine, Clinic of Child and Adolescent Neuropsychiatry, Università degli Studi della Campania "Luigi Vanvitelli"Naples, Italy
| | - Domenico Tafuri
- Department of Motor Sciences and Wellness, University of Naples "Parthenope"Naples, Italy
| | - Monica Salerno
- Department of Mental Health, Physical and Preventive Medicine, Clinic of Child and Adolescent Neuropsychiatry, Università degli Studi della Campania "Luigi Vanvitelli"Naples, Italy
| | - Nicola Filippi
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetic and Sport Medicine, Università degli Studi della Campania "Luigi Vanvitelli"Naples, Italy
| | - Francesco Nuccio
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetic and Sport Medicine, Università degli Studi della Campania "Luigi Vanvitelli"Naples, Italy
| | - Maria Ruberto
- Department of Medical-Surgical and Dental Specialties, Università degli Studi della Campania "Luigi Vanvitelli"Naples, Italy
| | - Vincenzo De Luca
- Department of Psychiatry, University of TorontoToronto, ON, Canada
| | - Luigi Cipolloni
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Università degli Studi di Roma La SapienzaRome, Italy
| | - Giuseppe Cibelli
- Department of Clinical and Experimental Medicine, University of FoggiaFoggia, Italy
| | - Maria P Mollica
- Department of Biology Università degli Studi di Napoli Federico IINaples, Italy
| | - Diego Iacono
- Neurodevelopmental Research Lab, Biomedical Research Institute of New JerseyMorristown, NJ, United States.,Neuroscience Research, MidAtlantic Neonatology Associates, Atlantic Health SystemMorristown, NJ, United States.,Neuropathology Research, MANA/Biomedical Research Institute of New JerseyMorristown, NJ, United States
| | | | - Marcellino Monda
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetic and Sport Medicine, Università degli Studi della Campania "Luigi Vanvitelli"Naples, Italy
| | - Giovanni Messina
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetic and Sport Medicine, Università degli Studi della Campania "Luigi Vanvitelli"Naples, Italy.,Department of Clinical and Experimental Medicine, University of FoggiaFoggia, Italy
| | - Antonietta Messina
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetic and Sport Medicine, Università degli Studi della Campania "Luigi Vanvitelli"Naples, Italy
| |
Collapse
|
35
|
Chieffi S, Messina G, Villano I, Messina A, Valenzano A, Moscatelli F, Salerno M, Sullo A, Avola R, Monda V, Cibelli G, Monda M. Neuroprotective Effects of Physical Activity: Evidence from Human and Animal Studies. Front Neurol 2017; 8:188. [PMID: 28588546 PMCID: PMC5439530 DOI: 10.3389/fneur.2017.00188] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 04/19/2017] [Indexed: 11/13/2022] Open
Abstract
In the present article, we provide a review of current knowledge regarding the role played by physical activity (PA) in preventing age-related cognitive decline and reducing risk of dementia. The cognitive benefits of PA are highlighted by epidemiological, neuroimaging and behavioral studies. Epidemiological studies identified PA as an influential lifestyle factor in predicting rates of cognitive decline. Individuals physically active from midlife show a reduced later risk of cognitive impairment. Neuroimaging studies documented attenuation of age-related brain atrophy, and also increase of gray matter and white matter of brain areas, including frontal and temporal lobes. These structural changes are often associated with improved cognitive performance. Importantly, the brain regions that benefit from PA are also those regions that are often reported to be severely affected in dementia. Animal model studies provided significant information about biomechanisms that support exercise-enhanced neuroplasticity, such as angiogenesis and upregulation of growth factors. Among the growth factors, the brain-derived neurotrophic factor seems to play a significant role. Another putative factor that might contribute to beneficial effects of exercise is the neuropeptide orexin-A. The beneficial effects of PA may represent an important resource to hinder the cognitive decline associated with aging.
Collapse
Affiliation(s)
- Sergio Chieffi
- Department of Experimental Medicine, Università degli Studi della Campania "Luigi Vanvitelli", Naples, Italy
| | - Giovanni Messina
- Department of Experimental Medicine, Università degli Studi della Campania "Luigi Vanvitelli", Naples, Italy.,Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Ines Villano
- Department of Experimental Medicine, Università degli Studi della Campania "Luigi Vanvitelli", Naples, Italy
| | - Antonietta Messina
- Department of Experimental Medicine, Università degli Studi della Campania "Luigi Vanvitelli", Naples, Italy
| | - Anna Valenzano
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Fiorenzo Moscatelli
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Monica Salerno
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Alessio Sullo
- Department of Experimental Medicine, Università degli Studi della Campania "Luigi Vanvitelli", Naples, Italy
| | - Roberto Avola
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Vincenzo Monda
- Department of Experimental Medicine, Università degli Studi della Campania "Luigi Vanvitelli", Naples, Italy
| | - Giuseppe Cibelli
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Marcellino Monda
- Department of Experimental Medicine, Università degli Studi della Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
36
|
Chieffi S, Messina G, Villano I, Messina A, Esposito M, Monda V, Valenzano A, Moscatelli F, Esposito T, Carotenuto M, Viggiano A, Cibelli G, Monda M. Exercise Influence on Hippocampal Function: Possible Involvement of Orexin-A. Front Physiol 2017; 8:85. [PMID: 28261108 PMCID: PMC5306252 DOI: 10.3389/fphys.2017.00085] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 01/30/2017] [Indexed: 01/24/2023] Open
Abstract
In the present article, we provide a brief review of current knowledge regarding the effects induced by physical exercise on hippocampus. Research involving animals and humans supports the view that physical exercise, enhancing hippocampal neurogenesis and function, improves cognition, and regulates mood. These beneficial effects depend on the contribute of more factors including the enhancement of vascularization and upregulation of growth factors. Among these, the BDNF seems to play a significant role. Another putative factor that might contribute to beneficial effects of exercise is the orexin-A. In support of this hypothesis there are the following observations: (1) orexin-A enhances hippocampal neurogenesis and function and (2) the levels of orexin-A increase with physical exercise. The beneficial effects of exercise may represent an important resource to hinder the cognitive decline associated with the aging-related hippocampal deterioration and ameliorate depressive symptoms.
Collapse
Affiliation(s)
- Sergio Chieffi
- Section of Human Physiology and Unit of Dietetic and Sport Medicine, Department of Experimental Medicine, Second University of Naples Naples, Italy
| | - Giovanni Messina
- Section of Human Physiology and Unit of Dietetic and Sport Medicine, Department of Experimental Medicine, Second University of NaplesNaples, Italy; Department of Clinical and Experimental Medicine, University of FoggiaFoggia, Italy
| | - Ines Villano
- Section of Human Physiology and Unit of Dietetic and Sport Medicine, Department of Experimental Medicine, Second University of Naples Naples, Italy
| | - Antonietta Messina
- Section of Human Physiology and Unit of Dietetic and Sport Medicine, Department of Experimental Medicine, Second University of Naples Naples, Italy
| | - Maria Esposito
- Department of Mental Health, Physical and Preventive Medicine, Clinic of Child and Adolescent Neuropsychiatry, Center for Childhood Headache, Second University of Naples Naples, Italy
| | - Vincenzo Monda
- Section of Human Physiology and Unit of Dietetic and Sport Medicine, Department of Experimental Medicine, Second University of Naples Naples, Italy
| | - Anna Valenzano
- Department of Clinical and Experimental Medicine, University of Foggia Foggia, Italy
| | - Fiorenzo Moscatelli
- Department of Clinical and Experimental Medicine, University of Foggia Foggia, Italy
| | - Teresa Esposito
- Section of Human Physiology and Unit of Dietetic and Sport Medicine, Department of Experimental Medicine, Second University of Naples Naples, Italy
| | - Marco Carotenuto
- Department of Mental Health, Physical and Preventive Medicine, Clinic of Child and Adolescent Neuropsychiatry, Center for Childhood Headache, Second University of Naples Naples, Italy
| | - Andrea Viggiano
- Department of Medicine and Surgery, University of Salerno Salerno, Italy
| | - Giuseppe Cibelli
- Section of Human Physiology and Unit of Dietetic and Sport Medicine, Department of Experimental Medicine, Second University of Naples Naples, Italy
| | - Marcellino Monda
- Department of Clinical and Experimental Medicine, University of Foggia Foggia, Italy
| |
Collapse
|
37
|
Chieffi P. An Overview on Predictive Biomarkers of Testicular Germ Cell Tumors. J Cell Physiol 2016; 232:276-280. [DOI: 10.1002/jcp.25482] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 07/11/2016] [Indexed: 02/06/2023]
Affiliation(s)
- Paolo Chieffi
- Dipartimento di Psicologia; Seconda Università di Napoli; Caserta Italy
| |
Collapse
|
38
|
Function of G-Protein-Coupled Estrogen Receptor-1 in Reproductive System Tumors. J Immunol Res 2016; 2016:7128702. [PMID: 27314054 PMCID: PMC4903118 DOI: 10.1155/2016/7128702] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 04/08/2016] [Accepted: 05/08/2016] [Indexed: 01/13/2023] Open
Abstract
The G-protein-coupled estrogen receptor-1 (GPER-1), also known as GPR30, is a novel estrogen receptor mediating estrogen receptor signaling in multiple cell types. The progress of estrogen-related cancer is promoted by GPER-1 activation through mitogen-activated protein kinases (MAPK), phosphoinositide 3-kinase (PI3K), and phospholipase C (PLC) signaling pathways. However, this promoting effect of GPER-1 is nonclassic estrogen receptor (ER) dependent manner. In addition, clinical evidences revealed that GPER-1 is associated with estrogen resistance in estrogen-related cancer patients. These give a hint that GPER-1 may be a novel therapeutic target for the estrogen-related cancers. However, preclinical studies also found that GPER-1 activation of its special agonist G-1 inhibits cancer cell proliferation. This review aims to summarize the characteristics and complex functions of GPER-1 in cancers.
Collapse
|
39
|
Chieffi P. New perspective on molecular markers as promising therapeutic targets in germ cell tumors. Intractable Rare Dis Res 2016; 5:137-9. [PMID: 27195201 PMCID: PMC4869583 DOI: 10.5582/irdr.2016.01007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 02/26/2016] [Indexed: 01/28/2023] Open
Abstract
Testicular germ cell tumors (TGCTs) are the most frequent solid malignant tumors in men 20-40 years of age and the most frequent cause of death from solid tumors in this age group. TGCTs comprise two major histologic groups: seminomas and non-seminomas germ cell tumors (NSGCTs). NSGCTs can be further divided into embryonal carcinoma, Teratoma, yolk sac tumor, and choriocarcinoma. Seminomas and NSGCTs present significant differences in clinical features, therapy, and prognosis, and both show characteristics of the Primordial Germ Cells (PGCs). Many discovered biomarkers including HMGA1, GPR30, Aurora-B, estrogen receptor β, and others have given further advantages to discriminate between histological subgroups and could represent useful therapeutic targets.
Collapse
Affiliation(s)
- Paolo Chieffi
- Dipartimento di Psicologia, Seconda Università di Napoli, Caserta, Italy
| |
Collapse
|
40
|
Santillo A, Falvo S, Chieffi P, Di Fiore MM, Senese R, Chieffi Baccari G. D-Aspartate Induces Proliferative Pathways in Spermatogonial GC-1 Cells. J Cell Physiol 2016; 231:490-5. [PMID: 26189884 DOI: 10.1002/jcp.25095] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 07/17/2015] [Indexed: 01/10/2023]
Abstract
D-aspartate (D-Asp) is an endogenous amino acid present in vertebrate tissues, with particularly high levels in the testis. In vivo studies indicate that D-Asp indirectly stimulates spermatogenesis through the hypothalamic-pituitary-gonadal axis. Moreover, in vitro studies have demonstrated that D-Asp up-regulates testosterone production in Leydig cells by enhancing expression of the steroidogenic acute regulatory protein. In this study, a cell line derived from immortalized type-B mouse spermatogonia retaining markers of mitotic germ cells (GC-1) was employed to explore more direct involvement of D-Asp in spermatogenesis. Activity and protein expression of markers of cell proliferation were determined at intervals during incubation in D-Asp-containing medium. D-Asp induced phosphorylation of ERK and Akt proteins, stimulated expression of PCNA and Aurora B, and enhanced mRNA synthesis and protein expression of P450 aromatase and protein expression of Estrogen Receptor β (ERβ). These results are the first demonstration of a direct effect of D-Asp on spermatogonial mitotic activity. Considering that spermatogonia express the NR1 subunit of the N-Methyl-D-Aspartic Acid receptor (NMDAR), we suggest that their response to D-Asp depends on NMDAR-mediated activation of the ERK and Akt pathways and is further enhanced by activation of the P450 aromatase/ERβ pathway.
Collapse
Affiliation(s)
- Alessandra Santillo
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Seconda Università di Napoli, Caserta, Italy
| | - Sara Falvo
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Seconda Università di Napoli, Caserta, Italy
| | - Paolo Chieffi
- Dipartimento di Psicologia, Seconda Università di Napoli, Caserta, Italy
| | - Maria Maddalena Di Fiore
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Seconda Università di Napoli, Caserta, Italy
| | - Rosalba Senese
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Seconda Università di Napoli, Caserta, Italy
| | - Gabriella Chieffi Baccari
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Seconda Università di Napoli, Caserta, Italy
| |
Collapse
|
41
|
Zarzycka M, Gorowska-Wojtowicz E, Tworzydlo W, Klak A, Kozub K, Hejmej A, Bilinska B, Kotula-Balak M. Are aryl hydrocarbon receptor and G-protein-coupled receptor 30 involved in the regulation of seasonal testis activity in photosensitive rodent-the bank vole (Myodes glareolus)? Theriogenology 2016; 86:674-686.e1. [PMID: 27004452 DOI: 10.1016/j.theriogenology.2016.02.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 12/14/2015] [Accepted: 02/22/2016] [Indexed: 02/03/2023]
Abstract
Within the reproductive system both aryl hydrocarbon receptor (AHR) and G-protein-coupled receptor 30 (GPR30) contribute to estrogen signaling and controlling of reproductive physiology. The specific question is whether and how AHR and GPR30 are involved in regulation of testis function in seasonally breeding rodents. Bank vole testes were obtained from animals reared under 18 hours light:6 hours dark (LD) and 6 hours light:18 hours dark (SD) conditions. Aryl hydrocarbon receptor and GPR30 expression were analyzed by quantitative reverse transcriptase-polymerase chain reaction, Western blot, and immunohistochemistry and/or immunofluorescent staining. In addition, the activity of enzymes involved in the intracellular signal transduction; extracellular signal-regulated kinase (ERK), protein kinase (PKA), matrix metalloproteinase 9 (MMP 9) and the concentrations of cyclic adenosine monophosphate (cAMP), cyclic guanosine monophosphate (cGMP), and calcium (Ca(2+)) were examined by immunohistochemical, immunoenzymatic, and colorimetric assays, respectively. Aryl hydrocarbon receptor and GPR30 were expressed in testes of actively reproducing voles and regressed ones although their expression at the messenger RNA and AHR also at protein level appeared to be photoperiod-dependent. A specific cellular localization and expression of AHR and GPR30 correlated with the expression of ERK, PKA, and MMP 9. Moreover, we found robust differences in the levels of cAMP, cGMP, and Ca(2+) in testicular homogenates between LD and SD voles. In the testes of LD voles, the levels of second messengers were always higher compared to SD. In vole testis, AHR and GPR30 can induce signaling pathways that involve ERK, PKA, MMP 9 and cAMP, cGMP, Ca(2+). In addition, in AHR, signaling the engagement of both photoperiod and estrogens, whereas in GPR30, signaling only estrogens is reported. It is likely that in vole, because of a differential activity of signaling molecules, signal transduction via AHR rather than through GPR30 plays a role in regulation of seasonal changes of testis physiology.
Collapse
Affiliation(s)
- Marta Zarzycka
- Department of Endocrinology, Institute of Zoology, Jagiellonian University, Krakow, Poland
| | | | - Waclaw Tworzydlo
- Department of Developmental Biology and Morphology of Invertebrates, Institute of Zoology, Jagiellonian University, Krakow, Poland
| | - Aleksandra Klak
- Department of Endocrinology, Institute of Zoology, Jagiellonian University, Krakow, Poland
| | - Klaudia Kozub
- Department of Endocrinology, Institute of Zoology, Jagiellonian University, Krakow, Poland
| | - Anna Hejmej
- Department of Endocrinology, Institute of Zoology, Jagiellonian University, Krakow, Poland
| | - Barbara Bilinska
- Department of Endocrinology, Institute of Zoology, Jagiellonian University, Krakow, Poland
| | | |
Collapse
|
42
|
Chieffi P, Boscia F. New discovered molecular markers as promising therapeutic targets in germ cell tumors. Expert Opin Orphan Drugs 2015. [DOI: 10.1517/21678707.2015.1074070] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|