1
|
Zhou Y, Luo Z, Guo J, Wu L, Zhou X, Huang JJ, Huang D, Xiao L, Duan Q, Chang J, Gong L, Hang J. Pan-cancer analysis of Sp1 with a focus on immunomodulatory roles in gastric cancer. Cancer Cell Int 2024; 24:338. [PMID: 39402565 PMCID: PMC11476248 DOI: 10.1186/s12935-024-03521-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 10/03/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Sp1, a transcription factor, regulates essential cellular processes and plays important tumorigenic roles across diverse cancers. However, comprehensive pan-cancer analyses of its expression and potential immunomodulatory roles remain unexplored. METHODS Utilizing bioinformatics tools and public datasets, we examined the expression of Sp1 across normal tissues, tumors, and immune cells, and screened for pre- and post-transcriptional modifications, including genetic alterations, DNA methylation, and protein phosphorylation, affecting its expression or function. The association of Sp1 expression with immune cell infiltration, tumor mutational burden, and immune checkpoint signaling was also investigated. Single-cell transcriptome data was used to assess Sp1 expression in immune cells in gastric cancer (GC), and findings were corroborated using immunohistochemistry and multiplex immunofluorescence in an immunotherapy-treated patient cohort. The prognostic value of Sp1 in GC patients receiving immunotherapy was evaluated with Cox regression models. RESULTS Elevated Sp1 levels were observed in various cancers compared to normal tissues, with notable prominence in GC. High Sp1 expression correlated with advanced stage, poor prognosis, elevated tumor mutational burden (TMB), and microsatellite instability (MSI) status, particularly in GC. Significant correlations between Sp1 levels and CD8+ T cell and the M1 phenotype of tumor-associated macrophages were further detected upon multiplex immunofluorescence in GC samples. Interestingly, we verified that GC patients with higher Sp1 levels exhibited improved response to immunotherapy. Moreover, Sp1 emerged as a prognostic and predictive biomarker for GC patients undergoing immunotherapy. CONCLUSIONS Our pan-cancer analysis sheds light on the multifaceted role of Sp1 in tumorigenesis and underscores its potential as a prognostic and predictive biomarker for patients with GC undergoing immunotherapy.
Collapse
Affiliation(s)
- Yang Zhou
- The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213000, China
| | - Zhenzhen Luo
- Department of Oncology, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Shenzhen, 518116, China
| | - Jinfeng Guo
- Department of Oncology, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Shenzhen, 518116, China
| | - Lixia Wu
- Department of Oncology, Shanghai JingAn District ZhaBei Central Hospital, Shanghai, 200070, China
| | - Xiaoli Zhou
- The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213000, China
| | - Jun Jie Huang
- JC School of Public Health and Primary Care, Faculty of Medicine, Centre for Health Education and Health Promotion, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Daijia Huang
- Department of Oncology, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Shenzhen, 518116, China
| | - Li Xiao
- Department of Oncology, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Shenzhen, 518116, China
| | - Qiuhua Duan
- The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213000, China
| | - Jianhua Chang
- Department of Oncology, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Shenzhen, 518116, China.
| | - Libao Gong
- Department of Oncology, The Cancer Center of the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, 519000, China.
| | - Junjie Hang
- Department of Oncology, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Shenzhen, 518116, China.
| |
Collapse
|
2
|
Huang J, Sun C, Teng Liu D, Zhao NN, Shavit JA, Zhu Y, Chen SX. Nuclear Progestin Receptor-mediated Linkage of Blood Coagulation and Ovulation. Endocrinology 2022; 163:bqac057. [PMID: 35511048 PMCID: PMC9653010 DOI: 10.1210/endocr/bqac057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Indexed: 01/22/2023]
Abstract
Ovulation is a dramatic remodeling process that includes rupture of blood capillaries and clotting, but coagulation is not thought to directly regulate this process. Herein, we report remarkable increases of coagulation factors V (f5, ~3145-fold) and tissue factor (f3a, ~120-fold) in zebrafish ovarian follicle cells during ovulation. This increase was mediated through the nuclear progestin receptor (Pgr), which is essential for ovulation in zebrafish, and was totally abolished in ovarian follicular cells from pgr-/- mutants. In addition, promoter activities of f5 and f3a were significantly enhanced by progestin (DHP) via Pgr. Similar regulation of human F5 promoter activity was induced via human PGRB, suggesting a conserved mechanism. Site-directed mutagenesis of the zebrafish f5 promoter further demonstrated a direct regulation of coagulation factors via progestin response elements. Moreover, a stark increase of erythrocytes occurred in capillaries meshed in wild-type preovulatory follicles but was absent in pgr-/- mutants. Interestingly, anticoagulants significantly inhibited ovulation both in vitro and in vivo, respectively. Furthermore, reduced fecundity was observed in f5+/- female zebrafish. Taken together, our study provides plausible evidence for steroid regulation of coagulation factors, and a new hypothesis for blood clotting-triggered ovulation in vertebrates.
Collapse
Affiliation(s)
- Jing Huang
- State Key Laboratory of Marine Environmental Science, College of Ocean and
Earth Sciences, Xiamen University, Xiamen, Fujian
361102, China
| | - Chao Sun
- State Key Laboratory of Marine Environmental Science, College of Ocean and
Earth Sciences, Xiamen University, Xiamen, Fujian
361102, China
| | - Dong Teng Liu
- State Key Laboratory of Marine Environmental Science, College of Ocean and
Earth Sciences, Xiamen University, Xiamen, Fujian
361102, China
| | - Nan Nan Zhao
- State Key Laboratory of Marine Environmental Science, College of Ocean and
Earth Sciences, Xiamen University, Xiamen, Fujian
361102, China
| | - Jordan A Shavit
- Departments of Pediatrics and Human Genetics, University of
Michigan, Ann Arbor, Michigan 48109, USA
| | - Yong Zhu
- State Key Laboratory of Marine Environmental Science, College of Ocean and
Earth Sciences, Xiamen University, Xiamen, Fujian
361102, China
- Department of Biology, East Carolina University,
Greenville, North Carolina 27858, USA
| | - Shi Xi Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean and
Earth Sciences, Xiamen University, Xiamen, Fujian
361102, China
- State-Province Joint Engineering Laboratory of Marine Bioproducts and
Technology, Xiamen University, Xiamen, Fujian
361102, China
| |
Collapse
|
3
|
Abstract
Proton pump inhibitors (PPIs) are the most widely prescribed medications in the world. According to numerous studies, PPIs have been linked to hyperprolactinemia, which can lead to a variety of sexual and reproductive issues. This review summarizes the effects of numerous PPIs on the blood prolactin levels and associated sexual dysfunctions, which have an effect on the patient's life quality and fertility. The study is taken into account all the available resources till January 31, 2021. Out of total 364, only 27 relevant resources were involved in this review. In certain studies, short-term PPIs use has been shown to have little or no effect on the blood prolactin and other reproductive hormones levels. PPIs have been linked to the development of hyperprolactinemia in several case studies with varying degrees of the blood prolactin levels increase seen in individuals taking PPI alone or in combination with medications, like prokinetics. The relative risk of the sexual consequences development, such as gynecomastia, has been documented using lansoprazole and omeprazole in various cohort studies. On the other hand, other bits of data are insufficient to establish a definite relationship that can turn a possibility into certainty. The majority of the literature data is comprising of double-blind, randomized, crossover studies, case reports, and adverse drug reaction incidents reported to various pharmacovigilance centers. To investigate this link, high-quality studies in patients taking PPIs for a longer time period are needed. We conclude this article with a comprehensive discussion of the hyperprolactinemia clinical implications and the PPIs' function.
Collapse
|
4
|
Boonyaratanakornkit V, McGowan EM, Márquez-Garbán DC, Burton LP, Hamilton N, Pateetin P, Pietras RJ. Progesterone Receptor Signaling in the Breast Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1329:443-474. [PMID: 34664251 DOI: 10.1007/978-3-030-73119-9_21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The tumor microenvironment (TME) is a complex infrastructure composed of stromal, epithelial, and immune cells embedded in a vasculature ECM. The microenvironment surrounding mammary epithelium plays a critical role during the development and differentiation of the mammary gland, enabling the coordination of the complex multihormones and growth factor signaling processes. Progesterone/progesterone receptor paracrine signaling interactions in the microenvironment play vital roles in stem/progenitor cell function during normal breast development. In breast cancer, the female sex hormones, estrogen and progesterone, and growth factor signals are altered in the TME. Progesterone signaling modulates not only breast tumors but also the breast TME, leading to the activation of a series of cross-communications that are implicated in the genesis of breast cancers. This chapter reviews the evidence that progesterone and PR signaling modulates not only breast epitheliums but also the breast TME. Furthermore, crosstalk between estrogen and progesterone signaling affecting different cell types within the TME is discussed. A better understanding of how PR and progesterone affect the TME of breast cancer may lead to novel drugs or a therapeutic approach for the treatment of breast cancer shortly.
Collapse
Affiliation(s)
- Viroj Boonyaratanakornkit
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand.
- Age-Related Inflammation and Degeneration Research Unit, Chulalongkorn University, Bangkok, Thailand.
- Graduate Program in Clinical Biochemistry and Molecular Medicine, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand.
| | - Eileen M McGowan
- School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
- Central Laboratory, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Diana C Márquez-Garbán
- UCLA Jonsson Comprehensive Cancer Center and Department of Medicine, Division of Hematology-Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - L P Burton
- UCLA Jonsson Comprehensive Cancer Center and Department of Medicine, Division of Hematology-Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Nalo Hamilton
- UCLA Jonsson Comprehensive Cancer Center and Department of Medicine, Division of Hematology-Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Prangwan Pateetin
- Graduate Program in Clinical Biochemistry and Molecular Medicine, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Richard J Pietras
- UCLA Jonsson Comprehensive Cancer Center and Department of Medicine, Division of Hematology-Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| |
Collapse
|
5
|
Pan L, Yu Y, Yu M, Yao S, Mu Q, Luo G, Xu N. Expression of flTF and asTF splice variants in various cell strains and tissues. Mol Med Rep 2019; 19:2077-2086. [PMID: 30664196 PMCID: PMC6390075 DOI: 10.3892/mmr.2019.9843] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 11/28/2018] [Indexed: 11/10/2022] Open
Abstract
Tissue factor (TF) expressed at the protein level includes two isoforms: The membrane‑bound full‑length TF (flTF) and the soluble alternatively spliced TF (asTF). flTF is the major thrombogenic form of TF, whereas asTF is more closely associated with tumor growth, angiogenesis, metastasis and cell growth. In order to further investigate the different expression and functions of TF splice variants, the expression of these two splice variants were detected in numerous cell strains and tissues in the present study. Quantitative polymerase chain reaction was used to measure the transcript levels of the TF variants in 11 human cell lines, including cervical cancer, breast cancer, hepatoblastoma, colorectal cancer and umbilical vein cells, and five types of tissue specimen, including placenta, esophageal cancer, breast cancer, cervical cancer (alongside normal cervical tissues) and non‑small cell lung cancer (alongside adjacent and normal tissues). Furthermore, the effects of chenodeoxycholic acid (CDCA) and apolipoprotein M (apoM) on the two variants were investigated. The results demonstrated that flTF was the major form of TF, and the mRNA expression levels of flTF were higher than those of asTF in all specimens tested. CDCA significantly upregulated the mRNA expression levels of the two variants. Furthermore, overexpression of apoM promoted the expression levels of asTF in Caco‑2 cells. The mRNA expression levels of asTF in cervical cancer tissues were significantly higher than in the corresponding normal tissues. To the best of our knowledge, the present study is the first to compare the expression of flTF and asTF in various samples. The results demonstrated that CDCA and apoM may modulate TF isoforms in different cell lines, and suggested that asTF may serve a role in the pathophysiological mechanism underlying cervical cancer development. In conclusion, the TF isoforms serve important and distinct roles in pathophysiological processes.
Collapse
Affiliation(s)
- Lili Pan
- Comprehensive Laboratory, Changzhou Key Lab of Individualized Diagnosis and Treatment Associated with High Technology Research, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China
| | - Yang Yu
- Comprehensive Laboratory, Changzhou Key Lab of Individualized Diagnosis and Treatment Associated with High Technology Research, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China
| | - Miaomei Yu
- Comprehensive Laboratory, Changzhou Key Lab of Individualized Diagnosis and Treatment Associated with High Technology Research, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China
| | - Shuang Yao
- Comprehensive Laboratory, Changzhou Key Lab of Individualized Diagnosis and Treatment Associated with High Technology Research, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China
| | - Qinfeng Mu
- Comprehensive Laboratory, Changzhou Key Lab of Individualized Diagnosis and Treatment Associated with High Technology Research, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China
| | - Guanghua Luo
- Comprehensive Laboratory, Changzhou Key Lab of Individualized Diagnosis and Treatment Associated with High Technology Research, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China
| | - Ning Xu
- Division of Clinical Chemistry and Pharmacology, Department of Laboratory Medicine, Lund University Hospital, S-221 85 Lund, Sweden
| |
Collapse
|
6
|
Fabres RB, da Rosa LA, de Souza SK, Cecconello AL, Azambuja AS, Sanches EF, Ribeiro MFM, de Fraga LS. Effects of progesterone on the neonatal brain following hypoxia-ischemia. Metab Brain Dis 2018; 33:813-821. [PMID: 29363039 DOI: 10.1007/s11011-018-0193-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 01/17/2018] [Indexed: 11/25/2022]
Abstract
Progesterone displays a strong potential for the treatment of neonatal hypoxic-ischemic encephalopathy since it has been shown to be beneficial in the treatment of the central nervous system injuries in adult animals. Here, we evaluated the effects of the administration of progesterone (10 mg/kg) in seven-days-old male Wistar rats submitted to neonatal hypoxia-ischemia (HI). Progesterone was administered immediately before ischemia and/or 6 and 24 h after the onset of hypoxia. The body weight of the animals, the volume of brain lesion and the expression of p-Akt and procaspase-3 in the hippocampus were evaluated. All animals submitted to HI showed a reduction in the body weight. However, this reduction was more remarkable in those animals which received progesterone before surgery. Administration of progesterone was unable to reduce the volume of brain damage caused by HI. Moreover, no significant differences were observed in the expression of p-Akt and procaspase-3 in animals submitted to HI and treated with either progesterone or vehicle. In summary, progesterone did not show a neuroprotective effect on the volume of brain lesion in neonatal rats submitted to hypoxia-ischemia. Furthermore, progesterone was unable to modulate p-Akt and procaspase-3 signaling pathways, which may explain the absence of neuroprotection. On the other hand, it seems that administration of progesterone before ischemia exerts some systemic effect, leading to a remarkable reduction in the body weight.
Collapse
Affiliation(s)
- Rafael Bandeira Fabres
- Laboratory of Neurohumoral Interaction, Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, Porto Alegre, RS, 90050-170, Brazil
- Laboratory of Comparative Metabolism and Endocrinology, Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, Porto Alegre, RS, 90050-170, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, Porto Alegre, RS, 90050-170, Brazil
- Hospital de Clínicas de Porto Alegre (HCPA), Ramiro Barcelos, 2350, Porto Alegre, RS, 90035-903, Brazil
| | - Luciana Abreu da Rosa
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, Porto Alegre, RS, 90050-170, Brazil
| | - Samir Khal de Souza
- Laboratory of Comparative Metabolism and Endocrinology, Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, Porto Alegre, RS, 90050-170, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, Porto Alegre, RS, 90050-170, Brazil
| | - Ana Lucia Cecconello
- Laboratory of Neurohumoral Interaction, Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, Porto Alegre, RS, 90050-170, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, Porto Alegre, RS, 90050-170, Brazil
| | - Amanda Stapenhorst Azambuja
- Laboratory of Neurohumoral Interaction, Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, Porto Alegre, RS, 90050-170, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, Porto Alegre, RS, 90050-170, Brazil
| | - Eduardo Farias Sanches
- Laboratory of Cerebral Ischemia, Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos, 2600, Porto Alegre, RS, 90035-000, Brazil
| | - Maria Flavia Marques Ribeiro
- Laboratory of Neurohumoral Interaction, Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, Porto Alegre, RS, 90050-170, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, Porto Alegre, RS, 90050-170, Brazil
| | - Luciano Stürmer de Fraga
- Laboratory of Comparative Metabolism and Endocrinology, Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, Porto Alegre, RS, 90050-170, Brazil.
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, Porto Alegre, RS, 90050-170, Brazil.
- Hospital de Clínicas de Porto Alegre (HCPA), Ramiro Barcelos, 2350, Porto Alegre, RS, 90035-903, Brazil.
| |
Collapse
|
7
|
Sansone A, Romanelli F, Sansone M, Lenzi A, Di Luigi L. Gynecomastia and hormones. Endocrine 2017; 55:37-44. [PMID: 27145756 DOI: 10.1007/s12020-016-0975-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 04/26/2016] [Indexed: 12/28/2022]
Abstract
Gynecomastia-the enlargement of male breast tissue in men-is a common finding, frequently observed in newborns, adolescents, and old men. Physiological gynecomastia, occurring in almost 25 % of cases, is benign and self-limited; on the other hand, several conditions and drugs may induce proliferation of male breast tissue. True gynecomastia is a common feature often related to estrogen excess and/or androgen deficiency as a consequence of different endocrine disorders. Biochemical evaluation should be performed once physiological or iatrogenic gynecomastia has been ruled out. Non-endocrine illnesses, including liver failure and chronic kidney disease, are another cause of gynecomastia which should be considered. Treating the underlying disease or discontinuing medications might resolve gynecomastia, although the psychosocial burden of this condition might require different and careful consideration.
Collapse
Affiliation(s)
- Andrea Sansone
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza, University of Rome, Viale Regina Elena 324, 00161, Rome, Italy.
| | - Francesco Romanelli
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza, University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Massimiliano Sansone
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza, University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Andrea Lenzi
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza, University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Luigi Di Luigi
- Department of Movement, Human and Health Sciences, Unit of Endocrinology, University of Rome "Foro Italico", Largo Lauro de Bosis 15, 00135, Rome, Italy
| |
Collapse
|
8
|
Progesterone receptor (PR) polyproline domain (PPD) mediates inhibition of epidermal growth factor receptor (EGFR) signaling in non-small cell lung cancer cells. Cancer Lett 2016; 374:279-91. [PMID: 26892043 DOI: 10.1016/j.canlet.2016.02.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 02/04/2016] [Accepted: 02/09/2016] [Indexed: 12/31/2022]
Abstract
Recent evidence has suggested a possible role for progesterone receptor (PR) in the progression of non-small cell lung cancer (NSCLC). However, little is known concerning roles of PR in NSCLC. PR contains a polyproline domain (PPD), which directly binds to the SH3 domain of signaling molecules. Because PPD-SH3 interactions are essential for EGFR signaling, we hypothesized that the presence of PR-PPD interfered with EGFR-mediated signaling and cell proliferation. We examined the role of PR-PPD in cell proliferation and signaling by stably expressing PR-B, or PR-B with disrupting mutations in the PPD (PR-BΔSH3), from a tetracycline-regulated promoter in A549 NSCLC cells. PR-B dose-dependently inhibited cell growth in the absence of ligand, and progestin (R5020) treatment further suppressed the growth. Treatment with RU486 abolished PR-B- and R5020-mediated inhibition of cell proliferation. Expression of PR-BΔSH3 and treatment with R5020 or RU486 had no effect on cell proliferation. Furthermore, PR-B expression but not PR-BΔSH3 expression reduced EGF-induced A549 proliferation and activation of ERK1/2, in the absence of ligand. Taken together, our data demonstrated the significance of PR extranuclear signaling through PPD interactions in EGFR-mediated proliferation and signaling in NSCLC.
Collapse
|