1
|
Chen D, Wang R, Sheng X, Zhao N, Lin Y, Wang Y, Zhu J, Li Y. PDZK1-interacting protein 1(PDZK1IP1) promotes subcutaneous preadipocyte proliferation in goats. Anim Biotechnol 2023; 34:3063-3073. [PMID: 36244042 DOI: 10.1080/10495398.2022.2132952] [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] [Indexed: 11/01/2022]
Abstract
PDZK1-interacting protein 1(PDZK1IP1), also known as MAP17, is encoded by the PDZK1IP1 gene and is a membrane-associated protein. PDZK1IP1 have been proven to be a potent regulator of cancer cell proliferation. However, the role of PDZK1IP1 in regulating goat subcutaneous preadipocyte proliferation is unknown. Here, we cloned the full-length coding sequence of PDZK1IP1 gene, investigated the potential functional of PDZK1IP1 in goat subcutaneous preadipocyte proliferation by gaining or losing function in vitro. Our results indicated that goat PDZK1IP1 gene consists of 345 bp, encoding a protein of 114 amino acids containing a typical PDZK1IP1 (MAP17) super family domain. Overexpression of PDZK1IP1 significantly increased the number of EdU-positive cells and cell viability, and also upregulated mRNA expression of cell proliferation-associated genes including CCND1 and CDK2 in vitro cultured cells. Conversely, knockdown of PDZK1IP1 mediated by siRNA technique significantly inhibited subcutaneous preadipocyte proliferation and downregulated mRNA expression of cell proliferation-associated genes including CCNE1, CCND1 and CDK2. Collectively, these results suggested that PDZK1IP1 can promote proliferation of goat subcutaneous preadipocyte.
Collapse
Affiliation(s)
- Dingshuang Chen
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Education Ministry, Southwest Minzu University, Chengdu, China
- College of Animal Science and Veterinary, Southwest Minzu University, Chengdu, China
| | - Ruilong Wang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Education Ministry, Southwest Minzu University, Chengdu, China
- College of Animal Science and Veterinary, Southwest Minzu University, Chengdu, China
| | - Xueqing Sheng
- College of Animal Science and Veterinary, Southwest Minzu University, Chengdu, China
| | - Nan Zhao
- College of Animal Science and Veterinary, Southwest Minzu University, Chengdu, China
| | - Yaqiu Lin
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Education Ministry, Southwest Minzu University, Chengdu, China
- College of Animal Science and Veterinary, Southwest Minzu University, Chengdu, China
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation of Sichuan Province, Southwest Minzu University, Chengdu, China
| | - Yong Wang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Education Ministry, Southwest Minzu University, Chengdu, China
- College of Animal Science and Veterinary, Southwest Minzu University, Chengdu, China
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation of Sichuan Province, Southwest Minzu University, Chengdu, China
| | - Jiangjiang Zhu
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Education Ministry, Southwest Minzu University, Chengdu, China
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation of Sichuan Province, Southwest Minzu University, Chengdu, China
| | - Yanyan Li
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Education Ministry, Southwest Minzu University, Chengdu, China
- College of Animal Science and Veterinary, Southwest Minzu University, Chengdu, China
| |
Collapse
|
2
|
Wang K, Liu S, Tian Y, Liu C, Gui Z, Yu T, Zhang L. PDZK1 Interacting Protein 1 Promotes the Progression of Papillary Thyroid Cancer. J Clin Endocrinol Metab 2022; 107:2449-2461. [PMID: 35727731 DOI: 10.1210/clinem/dgac376] [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: 04/05/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND The incidence of papillary thyroid cancer (PTC) has increased rapidly in recent decades, and tumor progression events are common in PTC. The purpose of our study is to identify the differentially expressed genes (DEGs) correlated with PTC progression and investigate the function of PDZK1IP1 (PDZK1 interacting protein 1) in PTC. METHODS We first analyzed DEGs associated with PTC progression between paired PTC and normal thyroid tissues in 3 Gene Expression Omnibus data sets (GSE29265, GSE33630, and GSE60542) and The Cancer Genome Atlas (TCGA) database. Data from the TCGA database and our institution were utilized to explore the relationship between PDZK1IP1 expression and clinicopathological characteristics of PTC. The CCK8 cell proliferation assay, clone formation assay, flow cytometry assay, and the xenograft model were used to investigate the function of PDZK1IP1 in PTC. RESULTS Thirty-nine DEGs associated with PTC progression were identified, in which only PDZK1IP1 was upregulated in PTC tissue at both messenger RNA and protein levels. In addition, we found that high expression of PDZK1IP1 in the TCGA database was associated with poor progression-free survival, extrathyroidal extension, high stage, tall cell variant, and BRAFV600E mutation of the PTC (P < 0.001). In our collected samples, high expression of PDZK1IP1 was only related to lymph node metastasis (P < 0.05). Overexpression of PDZK1IP1 significantly promoted proliferation and inhibited apoptosis of PTC cells. Knockdown of PDZK1IP1 significantly inhibited proliferation, promoted apoptosis, and prevented xenograft formation of PTC cells. CONCLUSION PDZK1IP1 is an oncogene for tumorigenesis and development of PTC and might be a potential therapeutic target.
Collapse
Affiliation(s)
- Kun Wang
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- Department of Breast Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Shiyang Liu
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yao Tian
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Chenguang Liu
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Zhengwei Gui
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Tianyao Yu
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Lin Zhang
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| |
Collapse
|
3
|
Sampadi B, Mullenders LHF, Vrieling H. Low and high doses of ionizing radiation evoke discrete global (phospho)proteome responses. DNA Repair (Amst) 2022; 113:103305. [PMID: 35255311 DOI: 10.1016/j.dnarep.2022.103305] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 02/02/2022] [Accepted: 02/22/2022] [Indexed: 12/16/2022]
Abstract
BACKGROUND Although cancer risk is assumed to be linear with ionizing radiation (IR) dose, it is unclear to what extent low doses (LD) of IR from medical and occupational exposures pose a cancer risk for humans. Improved mechanistic understanding of the signaling responses to LD may help to clarify this uncertainty. Here, we performed quantitative mass spectrometry-based proteomics and phosphoproteomics experiments, using mouse embryonic stem cells, at 0.5 h and 4 h after exposure to LD (0.1 Gy) and high doses (HD; 1 Gy) of IR. RESULTS The proteome remained relatively stable (29; 0.5% proteins responded), whereas the phosphoproteome changed dynamically (819; 7% phosphosites changed) upon irradiation. Dose-dependent alterations of 25 IR-responsive proteins were identified, with only four in common between LD and HD. Mitochondrial metabolic proteins and pathways responded to LD, whereas transporter proteins and mitochondrial uncoupling pathways responded to HD. Congruently, mitochondrial respiration increased after LD exposure but decreased after HD exposure. While the bulk of the phosphoproteome response to LD (76%) occurred already at 0.5 h, an equivalent proportion of the phosphosites responded to HD at both time points. Motif, kinome/phosphatome, kinase-substrate, and pathway analyses revealed a robust DNA damage response (DDR) activation after HD exposure but not after LD exposure. Instead, LD-irradiation induced (de)phosphorylation of kinases, kinase-substrates and phosphatases that predominantly respond to reactive oxygen species (ROS) production. CONCLUSION Our analyses identify discrete global proteome and phosphoproteome responses after LD and HD, uncovering novel proteins and protein (de)phosphorylation events involved in the dose-dependent ionizing radiation responses.
Collapse
Affiliation(s)
- Bharath Sampadi
- Department of Human Genetics, Leiden University Medical Center, Einthovenweg 20, 2333ZC Leiden, The Netherlands.
| | - Leon H F Mullenders
- Department of Human Genetics, Leiden University Medical Center, Einthovenweg 20, 2333ZC Leiden, The Netherlands; Department of Genetics, Research Institute of Environmental Medicine (RIeM), Nagoya University, Nagoya, Japan
| | - Harry Vrieling
- Department of Human Genetics, Leiden University Medical Center, Einthovenweg 20, 2333ZC Leiden, The Netherlands.
| |
Collapse
|
4
|
Du J, Gu J, Deng J, Kong L, Guo Y, Jin C, Bao Y, Fu D, Li J. The expression and survival significance of sodium glucose transporters in pancreatic cancer. BMC Cancer 2022; 22:116. [PMID: 35090421 PMCID: PMC8796473 DOI: 10.1186/s12885-021-09060-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 11/25/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Sodium glucose transporters (SGLTs) play vital roles in glucose uptake in many solid cancers, including pancreatic cancer (PC). However, their expression profile in pancreatic cancer and correlation with prognosis are not clear. Thus, we aimed to analyse the expression profile and prognostic significance of SGLT-1 and SGLT-2 in PC. METHODS Eighty-eight patients with pancreatic ductal adenocarcinoma (PDAC) undergoing surgery in Huashan Hospital, Fudan University, from July 2017 to June 2020 were enrolled in the study. Specimens for immunohistochemistry were obtained through surgical resection. Bioinformatics analysis was performed based on the Gene Expression Omnibus (GEO), Oncomine and The Cancer Genome Atlas (TCGA) databases. The statistics were calculated using IBM SPSS Statistics, version 20 and R 4.1.1. P values lower than 0.05 were considered to indicate statistical significance. RESULTS SGLT-1 but not SGLT-2 was significantly overexpressed in PDAC. Survival analysis showed that the median overall survival (OS) and progression-free survival (PFS) of patients with high SGLT-1 expression were significantly longer than that of patients with low SGLT-1 expression. Cox regression indicated that high SGLT-1 expression was an independent predictor for a better prognosis, while residual tumour status (R1 and R2) was an independent risk factor for a poor prognosis. Finally, PDZK1-interacting protein 1 (PDZK1IP1), a protein participating in the generation of reactive oxygen species, was overexpressed in PDAC and its expression was significantly correlated with SGLT-1. CONCLUSIONS SGLT-1 but not SGLT-2 was overexpressed in PDAC, and the overexpression of SGLT-1 could be a predictor of a better prognosis. Residual tumour status (R1 and R2) was a risk factor for poor prognosis and disease progression.
Collapse
Affiliation(s)
- Jiali Du
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Rd, Shanghai, 200040, People's Republic of China
| | - Jichun Gu
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Rd, Shanghai, 200040, People's Republic of China
| | - Junyuan Deng
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Rd, Shanghai, 200040, People's Republic of China
| | - Lei Kong
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Rd, Shanghai, 200040, People's Republic of China
| | - Yujie Guo
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Rd, Shanghai, 200040, People's Republic of China
| | - Chen Jin
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Rd, Shanghai, 200040, People's Republic of China
| | - Yun Bao
- Department of Pathology, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Rd, Shanghai, 200040, People's Republic of China
| | - Deliang Fu
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Rd, Shanghai, 200040, People's Republic of China.
| | - Ji Li
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Rd, Shanghai, 200040, People's Republic of China.
| |
Collapse
|
5
|
Tampakis A, Tampaki EC, Nonni A, Kontos M, Tsourouflis G, Posabella A, Fourie L, Bolli M, Kouraklis G, von Flüe M, Felekouras E, Nikiteas N. MAP17 Expression in Colorectal Cancer Is a Prognostic Factor for Disease Recurrence and Dismal Prognosis Already in Early Stage Disease. Oncology 2021; 99:471-482. [PMID: 33853080 DOI: 10.1159/000515596] [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: 11/26/2020] [Accepted: 02/23/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND Disease recurrence in colorectal cancer constitutes a major cause of significant cancer-associated morbidity and mortality. MAP17 is a small protein, and its overexpression in malignant tumors has been correlated with aggressive tumor phenotypes. The aim of the present study was to investigate the expression patterns of MAP17 in colorectal cancer specimens and to assess its clinical significance. PATIENTS AND METHODS Surgical specimens of 111 patients with primary resectable colorectal cancer constituted the study population. Expression of MAP17 was assessed by immunohistochemistry, and the results were correlated with clinical and survival data. RESULTS MAP17 was expressed in cancer cells and endothelial cells of tumor blood vessels. Expression of MAP17 more than 10% was correlated with advanced disease stage (p < 0.001), higher T classification (p = 0.007), the presence of lymph node metastasis (p < 0.001), vascular (p = 0.013) and perineural invasion (p = 0.012). Patients exhibiting MAP17 expression of more than 30% in cancer cells compared to those expressing MAP17 less than 10% demonstrated a significantly worse 3-year progression-free survival (35.2 vs. 91%, p < 0.001) and 5-year overall survival (40.8 vs. 91%, p < 0.001). Cox regression analysis confirmed MAP17 expression of more than 30% as a prognostic marker of progression free survival (HR 0.136, 95% CI = 0.056-0.329, p < 0.001) and overall survival (HR 0.144 [95% CI) = 0.049-0.419, p < 0.001) independent of other clinicopathological characteristics. Statistically significantly worse 3-year progression-free survival and 5-year overall survival was demonstrated in the subgroup analysis of patients with early stage cancer only and high expression of MAP17. CONCLUSIONS High MAP17 expression in patients with colorectal cancer is a significant risk factor for cancer-associated morbidity and mortality already in early stage disease.
Collapse
Affiliation(s)
- Athanasios Tampakis
- Clarunis, University Center for Gastrointestinal and Liver Disorders, Department of Visceral Surgery, University Hospital of Basel, Basel, Switzerland.,2nd Department of Propedeutic Surgery, Athens University Medical School, Laiko General Hospital, Athens, Greece
| | - Ekaterini Christina Tampaki
- 2nd Department of Propedeutic Surgery, Athens University Medical School, Laiko General Hospital, Athens, Greece
| | - Afroditi Nonni
- 1st Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Michael Kontos
- 1st Department of Surgery, Athens University Medical School, Laiko General Hospital, Athens, Greece
| | - Gerasimos Tsourouflis
- 2nd Department of Propedeutic Surgery, Athens University Medical School, Laiko General Hospital, Athens, Greece
| | - Alberto Posabella
- Clarunis, University Center for Gastrointestinal and Liver Disorders, Department of Visceral Surgery, University Hospital of Basel, Basel, Switzerland
| | - Lana Fourie
- Clarunis, University Center for Gastrointestinal and Liver Disorders, Department of Visceral Surgery, University Hospital of Basel, Basel, Switzerland
| | - Martin Bolli
- Clarunis, University Center for Gastrointestinal and Liver Disorders, Department of Visceral Surgery, University Hospital of Basel, Basel, Switzerland
| | - Gregory Kouraklis
- 2nd Department of Propedeutic Surgery, Athens University Medical School, Laiko General Hospital, Athens, Greece
| | - Markus von Flüe
- Clarunis, University Center for Gastrointestinal and Liver Disorders, Department of Visceral Surgery, University Hospital of Basel, Basel, Switzerland
| | - Evangelos Felekouras
- 1st Department of Surgery, Athens University Medical School, Laiko General Hospital, Athens, Greece
| | - Nikolaos Nikiteas
- 2nd Department of Propedeutic Surgery, Athens University Medical School, Laiko General Hospital, Athens, Greece
| |
Collapse
|
6
|
Liang Q, Zhang H. MAP17 contributes to non-small cell lung cancer progression via suppressing miR-27a-3p expression and p38 signaling pathway. Cancer Biol Ther 2020; 22:19-29. [PMID: 33280497 DOI: 10.1080/15384047.2020.1836948] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
PROBLEM AND AIM The overexpression of MAP17 has been reported in various human carcinomas. However, its molecular mechanism in non-small cell lung cancer (NSCLC) has not been fully understood. Our study aimed to reveal the molecular mechanism of NSCLC that involved MAP17 and identify its target miRNA. METHODS RT-qPCR and immunoblot assays were conducted to measure the expression of mRNA and protein in NSCLC tissues and cell lines. Meanwhile, the A549 cells (an NSCLC cell line) were randomly assigned to the MAP17 overexpression group, the MAP17 knockdown group and negative control group to study the roles of MAP17 in cell viability, cell proliferation, migration, invasion, and apoptosis by performing Trypan blue exclusion, MTT, colony formation, transwell, wound healing and flow-cytometric apoptosis assays. The luciferase reporter assay was conducted to confirm the target relationship between MAP17 and miR-27a-3p. RESULTS The upregulation of MAP17 mRNA and protein was observed in NSCLC tissues and cell lines. In vitro, the positive roles of MAP17 on cell viability, migration, and invasion were confirmed in A549 cells. It was also found that MAP17 could inhibit cell apoptosis by suppressing the activation of the p38 pathway. This research eventually proved the target relationship between MAP17 and miR-27a-3p, and that miR-27a-3p reversed the effects of MAP17 in A549 cells by directly targeting MAP17. CONCLUSIONS MAP17 plays an oncogenic role in NSCLC by suppressing the activation of the p38 pathway. Apart from that, the miR-27a-3p can inhibit the expression of MAP17 to suppress the NSCLC progression.
Collapse
Affiliation(s)
- Qian Liang
- Department of Integrated 2, Affiliated Hospital of Jianghan University , Wuhan, Hubei, China
| | - Huan Zhang
- Department of Integrated 2, Affiliated Hospital of Jianghan University , Wuhan, Hubei, China
| |
Collapse
|
7
|
García-Heredia JM, Otero-Albiol D, Pérez M, Pérez-Castejón E, Muñoz-Galván S, Carnero A. Breast tumor cells promotes the horizontal propagation of EMT, stemness, and metastasis by transferring the MAP17 protein between subsets of neoplastic cells. Oncogenesis 2020; 9:96. [PMID: 33106480 PMCID: PMC7589521 DOI: 10.1038/s41389-020-00280-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 09/29/2020] [Accepted: 10/05/2020] [Indexed: 12/13/2022] Open
Abstract
MAP17 (PDZK1IP1) is a small protein regulating inflammation and tumor progression, upregulated in a broad range of carcinomas. MAP17 levels increase during tumor progression in a large percentage of advanced tumors. In the present work, we explored the role of this protein shaping tumor evolution. Here we show that in breast cancer, cells increased MAP17 levels in tumors by demethylation induced multiple changes in gene expression through specific miRNAs downregulation. These miRNA changes are dependent on Notch pathway activation. As a consequence, epithelial mesenchymal transition (EMT) and stemness are induced promoting the metastatic potential of these cells both in vitro and in vivo. Furthermore, MAP17 increased the exosomes in tumor cells, where MAP17 was released as cargo, and this horizontal propagation also increased the EMT in the recipient cells. Importantly, an antibody against MAP17 in the media reduces the EMT and stemness alterations promoted by the conditioned media from MAP17-expressing cells. Therefore, MAP17 expression promotes the horizontal propagation of EMT and metastasis by transferring the MAP17 protein between subsets of neoplastic cells. Thus, MAP17 can be used to describe a new mechanism for cell malignity at distance, without the involvement of genetic or epigenetic modifications. MAP17 can also be taken in consideration as new target for metastatic high-grade breast tumors.
Collapse
Affiliation(s)
- José Manuel García-Heredia
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Seville, Spain.,Departamento de Bioquímica Vegetal y Biología Molecular, Universidad de Sevilla, Seville, Spain.,CIBER de Cancer, Seville, Spain
| | - Daniel Otero-Albiol
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Seville, Spain.,CIBER de Cancer, Seville, Spain
| | - Marco Pérez
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Seville, Spain.,CIBER de Cancer, Seville, Spain
| | - Elena Pérez-Castejón
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Seville, Spain
| | - Sandra Muñoz-Galván
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Seville, Spain.,CIBER de Cancer, Seville, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Seville, Spain. .,CIBER de Cancer, Seville, Spain.
| |
Collapse
|
8
|
Sarcoma stratification by combined pH2AX and MAP17 (PDZK1IP1) levels for a better outcome on doxorubicin plus olaparib treatment. Signal Transduct Target Ther 2020; 5:195. [PMID: 32963243 PMCID: PMC7508862 DOI: 10.1038/s41392-020-00246-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 05/15/2020] [Accepted: 06/19/2020] [Indexed: 12/13/2022] Open
Abstract
Sarcomas constitute a rare heterogeneous group of tumors, including a wide variety of histological subtypes. Despite advances in our understanding of the pathophysiology of the disease, first-line sarcoma treatment options are still limited and new treatment approaches are needed. Histone H2AX phosphorylation is a sensitive marker for double strand breaks and has recently emerged as biomarker of DNA damage for new drug development. In this study, we explored the role of H2AX phosphorylation at Ser139 alone or in combination with MAP17 protein, an inducer of DNA damage through ROS increase, as prognostic biomarkers in sarcoma tumors. Next, we proposed doxorubicin and olaparib combination as potential therapeutic strategies against sarcomas displaying high level of both markers. We evaluate retrospectively the levels of pH2AX (Ser139) and MAP17 in a cohort of 69 patients with different sarcoma types and its relationship with clinical and pathological features. We found that the levels of pH2AX and MAP17 were related to clinical features and poor survival. Next, we pursued PARP1 inhibition with olaparib to potentiate the antitumor effect of DNA damaging effect of the DNA damaging agent doxorubicin to achieve an optimal synergy in sarcoma. We demonstrated that the combination of olaparib and doxorubicin was synergistic in vitro, inhibiting cell proliferation and enhancing pH2AX intranuclear accumulation, as a result of DNA damage. The synergism was corroborated in patient-derived xenografts (PDX) where the combination was effective in tumors with high levels of pH2AX and MAP17, suggesting that both biomarkers might potentially identify patients who better benefit from this combined therapy.
Collapse
|
9
|
Tampaki EC, Tampakis A, Nonni A, von Flüe M, Patsouris E, Kontzoglou K, Kouraklis G. Combined Fascin-1 and MAP17 Expression in Breast Cancer Identifies Patients with High Risk for Disease Recurrence. Mol Diagn Ther 2020; 23:635-644. [PMID: 31273628 DOI: 10.1007/s40291-019-00411-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND OBJECTIVE Breast cancer stem cells are considered to be a major cause of disease recurrence in breast cancer as they appear to be chemoresistant. Fascin-1 and MAP17 are stem cell markers whose excessive expression in tumors is associated with aggressive tumor phenotypes. The aim of the present study was to investigate the expression patterns of fascin-1 and MAP17 in breast cancer and to assess their clinical significance. METHODS Expression of fascin-1 and MAP17 was assessed via immunohistochemistry in surgical specimens of a cohort comprised of 127 patients with resectable breast cancer. Results were correlated with clinicopathological characteristics and survival data. Progression-free survival (PFS) was defined as the primary outcome of the present study. RESULTS Fascin-1 and MAP17 expression were strongly associated with the presence of triple-negative cancers (p < 0.0001). Tumors displaying high expression of fascin-1 presented correlations with high tumor grade (p = 0.002) and high expression of Ki-67 (p = 0.004). PFS of patients exhibiting high expression of fascin-1 and MAP17 in cancer cells in the first 5 years after surgery was significantly worse than in patients with low expression of the two markers (47.8%, 95% confidence interval [CI] 33-51 vs. 80.5%, 95% CI 47-56; p = 0.012) and independent of other clinicopathological characteristics (hazard ratio 0.171, 95% CI 0.034-0.869; p = 0.033). CONCLUSION Combined expression of fascin-1 and MAP17 in breast cancer cells is associated with a significantly worse 5-year PFS, therefore recognizing a group of patients with high risk for early disease recurrence.
Collapse
Affiliation(s)
- Ekaterini Christina Tampaki
- 2nd Department of Propedeutic Surgery, Athens University Medical School, Laiko General Hospital, 17 Agiou Thoma Street, 11527, Athens, Greece
| | - Athanasios Tampakis
- 2nd Department of Propedeutic Surgery, Athens University Medical School, Laiko General Hospital, 17 Agiou Thoma Street, 11527, Athens, Greece.
- Clarunis, University Center for Gastrointestinal and Liver Disorders, University Hospital of Basel, Spitalstraße 21, 4031, Basel, Switzerland.
| | - Afroditi Nonni
- 1st Department of Pathology, School of Medicine, National University of Athens, Athens, Greece
| | - Markus von Flüe
- Clarunis, University Center for Gastrointestinal and Liver Disorders, University Hospital of Basel, Spitalstraße 21, 4031, Basel, Switzerland
| | - Efstratios Patsouris
- 1st Department of Pathology, School of Medicine, National University of Athens, Athens, Greece
| | - Konstantinos Kontzoglou
- 2nd Department of Propedeutic Surgery, Athens University Medical School, Laiko General Hospital, 17 Agiou Thoma Street, 11527, Athens, Greece
| | - Gregory Kouraklis
- 2nd Department of Propedeutic Surgery, Athens University Medical School, Laiko General Hospital, 17 Agiou Thoma Street, 11527, Athens, Greece
| |
Collapse
|
10
|
Rivero M, Peinado-Serrano J, Muñoz-Galvan S, Espinosa-Sánchez A, Suarez-Martinez E, Felipe-Abrio B, Fernández-Fernández MC, Ortiz MJ, Carnero A. MAP17 (PDZK1IP1) and pH2AX are potential predictive biomarkers for rectal cancer treatment efficacy. Oncotarget 2018; 9:32958-32971. [PMID: 30250642 PMCID: PMC6152481 DOI: 10.18632/oncotarget.26010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 07/13/2018] [Indexed: 12/17/2022] Open
Abstract
Rectal cancer represents approximately 10% of cancers worldwide. Preoperative chemoradiotherapy increases complete pathologic response and local control, although it offers a poor advantage in survivorship and sphincter saving compared with that of radiotherapy alone. After preoperative chemoradiotherapy, approximately 20% of patients with rectal cancer achieve a pathologic complete response to the removed surgical specimen; this response may be related to a better prognosis and an improvement in disease-free survival. However, better biomarkers to predict response and new targets are needed to stratify patients and obtain better response rates. MAP17 (PDZK1IP1) is a small, 17 kDa non-glycosylated membrane protein located in the plasma membrane and Golgi apparatus and is overexpressed in a wide variety of human carcinomas. MAP17 has been proposed as a predictive biomarker for reactive oxygen species, ROS, inducing treatments in cervical tumors or laryngeal carcinoma. Due to the increase in ROS, MAP17 is also associated with the marker of DNA damage, phosphoH2AX (pH2AX). In the present manuscript, we examined the values of MAP17 and pH2AX as surrogate biomarkers of the response in rectal tumors. MAP17 expression after preoperative chemoradiotherapy is able to predict the response to chemoradiotherapy, similar to the increase in pH2AX. Furthermore, we explored whether we can identify molecular targeted therapies that could help improve the response of these tumors to radiotherapy. In this sense, we found that the inhibition of DNA damage with olaparib increased the response to radio- and chemotherapy, specifically in tumors with high levels of pH2AX and MAP17.
Collapse
Affiliation(s)
- Maria Rivero
- Instituto de Biomedicina de Sevilla, HUVR, CSIC, Universidad de Sevilla, Seville, Spain.,Department of Radiation Oncology, HUVR, Seville, Spain.,Department of Pathology, HUVR, Seville, Spain
| | - Javier Peinado-Serrano
- Instituto de Biomedicina de Sevilla, HUVR, CSIC, Universidad de Sevilla, Seville, Spain.,Department of Radiation Oncology, HUVR, Seville, Spain.,CIBER de Cáncer, ISCIII, Madrid, Spain
| | - Sandra Muñoz-Galvan
- Instituto de Biomedicina de Sevilla, HUVR, CSIC, Universidad de Sevilla, Seville, Spain.,CIBER de Cáncer, ISCIII, Madrid, Spain
| | | | - Elisa Suarez-Martinez
- Instituto de Biomedicina de Sevilla, HUVR, CSIC, Universidad de Sevilla, Seville, Spain
| | - Blanca Felipe-Abrio
- Instituto de Biomedicina de Sevilla, HUVR, CSIC, Universidad de Sevilla, Seville, Spain.,CIBER de Cáncer, ISCIII, Madrid, Spain
| | - Maria Carmen Fernández-Fernández
- Instituto de Biomedicina de Sevilla, HUVR, CSIC, Universidad de Sevilla, Seville, Spain.,Department of Pathology, HUVR, Seville, Spain
| | - Maria Jose Ortiz
- Instituto de Biomedicina de Sevilla, HUVR, CSIC, Universidad de Sevilla, Seville, Spain.,Department of Radiation Oncology, HUVR, Seville, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla, HUVR, CSIC, Universidad de Sevilla, Seville, Spain.,CIBER de Cáncer, ISCIII, Madrid, Spain
| |
Collapse
|
11
|
García-Heredia JM, Carnero A. Dr. Jekyll and Mr. Hyde: MAP17's up-regulation, a crosspoint in cancer and inflammatory diseases. Mol Cancer 2018; 17:80. [PMID: 29650022 PMCID: PMC5896160 DOI: 10.1186/s12943-018-0828-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 03/28/2018] [Indexed: 12/14/2022] Open
Affiliation(s)
- José M García-Heredia
- Instituto de Biomedicina de Sevilla, IBIS/Hospital Universitario Virgen del Rocío/ Universidad de Sevilla/Consejo Superior de Investigaciones Científicas, Avda. Manuel Siurot s/n, 41013, Sevilla, Spain.,Department of Vegetal Biochemistry and Molecular Biology, University of Seville, Seville, Spain.,CIBER de Cáncer, Instituto de Salud Carlos III, Pabellón 11, Madrid, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla, IBIS/Hospital Universitario Virgen del Rocío/ Universidad de Sevilla/Consejo Superior de Investigaciones Científicas, Avda. Manuel Siurot s/n, 41013, Sevilla, Spain. .,CIBER de Cáncer, Instituto de Salud Carlos III, Pabellón 11, Madrid, Spain.
| |
Collapse
|
12
|
Shao Y, Lv H, Zhong DS, Zhou QH. EGFR-TKI resistance and MAP17 are associated with cancer stem cell like properties. Oncol Lett 2018; 15:6655-6665. [PMID: 29616128 DOI: 10.3892/ol.2018.8129] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Accepted: 01/19/2018] [Indexed: 02/07/2023] Open
Abstract
Patients with non-small-cell lung cancer (NSCLC) with sensitive epidermal growth factor receptor (EGFR) mutations generally react well to tyrosine kinase inhibitors (TKIs). However acquired resistance eventually occurs. Several mechanisms contribute to the resistance including T790M mutation, c-Met amplification and PIK3CA mutation. In recent years, cancer stem cells (CSCs) have been suggested to be involved in TKI resistance. MAP17 is aberrantly overexpressed in a number of malignancies. However, the expression pattern and function of MAP17 in CSCs are still unclear. The aim the present study was to illustrate the effect of CSC-like cells on the resistance to TKIs in EGFR mutant NSCLC cells and explore the possible role of MAP17 in CSCs. The EGFR mutant cell line PC9 was cultured under serum-deprived undifferentiated conditions. The CSC properties including expression of stem cell markers CD133, CD44, Oct-4 and ABCG2, ability of self-renewal, invasion, proliferation and tumorigenesis were examined. The expression of MAP17 was compared in sphere and parent cells. Sphere cells displayed stem cells phenotypes and were resistant to erlotinib. Sphere cells expressed higher levels of MAP17, and MAP17 was associated with self-renewal and TKI resistance. The function of MAP17 demonstrated to be partially dependent on Na-dependent glucose transporter 1. Collectively these findings suggest that MAP17 serves a role in TKI resistance through regulation of CSCs in lung cancer.
Collapse
Affiliation(s)
- Yi Shao
- Department of Oncology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Hui Lv
- Department of Oncology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Dian-Sheng Zhong
- Department of Oncology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Qing-Hua Zhou
- Tianjin Lung Cancer Center and Institute, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| |
Collapse
|
13
|
Perez M, Peinado-Serrano J, Garcia-Heredia JM, Felipe-Abrio I, Tous C, Ferrer I, Martin-Broto J, Saez C, Carnero A. Efficacy of bortezomib in sarcomas with high levels of MAP17 (PDZK1IP1). Oncotarget 2018; 7:67033-67046. [PMID: 27563810 PMCID: PMC5341855 DOI: 10.18632/oncotarget.11475] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 08/09/2016] [Indexed: 11/25/2022] Open
Abstract
Sarcomas are malignant tumors accounting for a high percentage of cancer morbidity and mortality in children and young adults. Surgery and radiation therapy are the accepted treatments for most sarcomas; however, patients with metastatic disease are treated with systemic chemotherapy. Many tumors display marginal levels of chemoresponsiveness, and new treatment approaches are needed. MAP17 is a small non-glycosylated membrane protein overexpressed in carcinomas. The levels of MAP17 could be used as a prognostic marker to predict the response to bortezomib in hematological malignancies and in breast tumors. Therefore, we analyzed the expression of this oncogene in sarcomas and its relationship with clinico-pathological features, as well as tested whether it can be used as a new biomarker to predict the therapeutic response to bortezomib and new therapies for sarcomas. We found that the levels of MAP17 were related to clinical features and poor survival in a cohort of 69 patients with different sarcoma types, not being restricted to any special subtype of tumor. MAP17 expression is associated with poor overall survival (p<0.001) and worse disease-free survival (p=0.002). Cell lines with high levels of MAP17 show a better response to bortezomib in vitro. Furthermore, patient-derived xenografts (PDX) with high levels of MAP17 respond to bortezomib in vivo. Our results showed that this response is due to the lower levels of NFκB and autophagy activation. Therefore, we suggest that MAP17 is a new biomarker to predict the efficacy of bortezomib as a new therapy for sarcomas.
Collapse
Affiliation(s)
- Marco Perez
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocio, Universidad de Sevilla, Consejo Superior de Investigaciones Cientificas, Seville, Spain
| | - Javier Peinado-Serrano
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocio, Universidad de Sevilla, Consejo Superior de Investigaciones Cientificas, Seville, Spain
| | - Jose Manuel Garcia-Heredia
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocio, Universidad de Sevilla, Consejo Superior de Investigaciones Cientificas, Seville, Spain.,Department of Vegetal Biochemistry and Molecular Biology, University of Seville, Seville, Spain
| | - Irene Felipe-Abrio
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocio, Universidad de Sevilla, Consejo Superior de Investigaciones Cientificas, Seville, Spain
| | - Cristina Tous
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocio, Universidad de Sevilla, Consejo Superior de Investigaciones Cientificas, Seville, Spain
| | - Irene Ferrer
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocio, Universidad de Sevilla, Consejo Superior de Investigaciones Cientificas, Seville, Spain
| | - Javier Martin-Broto
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocio, Universidad de Sevilla, Consejo Superior de Investigaciones Cientificas, Seville, Spain.,Department of Medical Oncology, Virgen del Rocío University Hospital, Seville, Spain
| | - Carmen Saez
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocio, Universidad de Sevilla, Consejo Superior de Investigaciones Cientificas, Seville, Spain.,Department of Pathology, Virgen del Rocío University Hospital, Seville, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocio, Universidad de Sevilla, Consejo Superior de Investigaciones Cientificas, Seville, Spain
| |
Collapse
|
14
|
Verdugo-Sivianes EM, Navas L, Molina-Pinelo S, Ferrer I, Quintanal-Villalonga A, Peinado J, Garcia-Heredia JM, Felipe-Abrio B, Muñoz-Galvan S, Marin JJ, Montuenga L, Paz-Ares L, Carnero A. Coordinated downregulation of Spinophilin and the catalytic subunits of PP1, PPP1CA/B/C, contributes to a worse prognosis in lung cancer. Oncotarget 2017; 8:105196-105210. [PMID: 29285244 PMCID: PMC5739631 DOI: 10.18632/oncotarget.22111] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Accepted: 09/03/2017] [Indexed: 12/20/2022] Open
Abstract
The scaffold protein Spinophilin (Spinophilin, PPP1R9B) is one of the regulatory subunits of phosphatase-1 (PP1), directing it to distinct subcellular locations and targets. The loss of Spinophilin reduces PP1 targeting to pRb, thereby maintaining higher levels of phosphorylated pRb. Spinophilin is absent or reduced in approximately 40% of human lung tumors, correlating with the malignant grade. However, little is known about the relevance of the coordinated activity or presence of Spinophilin and its reported catalytic partners in the prognosis of lung cancer. In the present work, we show that the downregulation of Spinophilin, either by protein or mRNA, is related to a worse prognosis in lung tumors. This effect is more relevant in squamous cell carcinoma, SCC, than in adenocarcinoma. Downregulation of Spinophilin is related to a decrease in the levels of its partners PPP1CA/B/C, the catalytic subunits of PP1. A decrease in these subunits is also related to prognosis in SCC and, in combination with a decrease in Spinophilin, are markers of a poor prognosis in these tumors. The analysis of the genes that correlate to Spinophilin in lung tumors showed clear enrichment in ATP biosynthesis and protein degradation GO pathways. The analysis of the response to several common and pathway-related drugs indicates a direct correlation between the Spinophilin/PPP1Cs ratio and the response to oxaliplatin and bortezomib. This finding indicates that this ratio may be a good predictive biomarker for the activity of the drugs in these tumors with a poor prognosis.
Collapse
Affiliation(s)
- Eva M Verdugo-Sivianes
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Sevilla, Spain.,CIBER de Cáncer, Instituto de Salud Carlos III, Pabellón 11, Planta 0, Madrid, Spain
| | - Lola Navas
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Sevilla, Spain.,CIBER de Cáncer, Instituto de Salud Carlos III, Pabellón 11, Planta 0, Madrid, Spain
| | - Sonia Molina-Pinelo
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Sevilla, Spain.,CIBER de Cáncer, Instituto de Salud Carlos III, Pabellón 11, Planta 0, Madrid, Spain
| | - Irene Ferrer
- CIBER de Cáncer, Instituto de Salud Carlos III, Pabellón 11, Planta 0, Madrid, Spain.,H120-CNIO Lung Cancer Clinical Research Unit, Instituto de Investigación Hospital 12 de Octubre and CNIO, Madrid, Spain
| | - Alvaro Quintanal-Villalonga
- H120-CNIO Lung Cancer Clinical Research Unit, Instituto de Investigación Hospital 12 de Octubre and CNIO, Madrid, Spain
| | - Javier Peinado
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Sevilla, Spain.,Radiation Oncology Department, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - Jose M Garcia-Heredia
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Sevilla, Spain.,CIBER de Cáncer, Instituto de Salud Carlos III, Pabellón 11, Planta 0, Madrid, Spain.,Department of Vegetal Biochemistry and Molecular Biology, University of Seville, Seville, Spain
| | - Blanca Felipe-Abrio
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Sevilla, Spain.,CIBER de Cáncer, Instituto de Salud Carlos III, Pabellón 11, Planta 0, Madrid, Spain
| | - Sandra Muñoz-Galvan
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Sevilla, Spain.,CIBER de Cáncer, Instituto de Salud Carlos III, Pabellón 11, Planta 0, Madrid, Spain
| | - Juan J Marin
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Sevilla, Spain.,CIBER de Cáncer, Instituto de Salud Carlos III, Pabellón 11, Planta 0, Madrid, Spain.,Department of Predictive Medicine and Public Health, Universidad de Sevilla, Sevilla, Spain
| | - Luis Montuenga
- CIBER de Cáncer, Instituto de Salud Carlos III, Pabellón 11, Planta 0, Madrid, Spain.,Program in Solid Tumors and Biomarkers, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Luis Paz-Ares
- CIBER de Cáncer, Instituto de Salud Carlos III, Pabellón 11, Planta 0, Madrid, Spain.,H120-CNIO Lung Cancer Clinical Research Unit, Instituto de Investigación Hospital 12 de Octubre and CNIO, Madrid, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Sevilla, Spain.,CIBER de Cáncer, Instituto de Salud Carlos III, Pabellón 11, Planta 0, Madrid, Spain
| |
Collapse
|
15
|
García-Heredia JM, Carnero A. The cargo protein MAP17 (PDZK1IP1) regulates the immune microenvironment. Oncotarget 2017; 8:98580-98597. [PMID: 29228712 PMCID: PMC5716752 DOI: 10.18632/oncotarget.21651] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 08/25/2017] [Indexed: 02/06/2023] Open
Abstract
Inflammation is a complex defensive response activated after various harmful stimuli allowing the clearance of damaged cells and initiating healing and regenerative processes. Chronic, or pathological, inflammation is also one of the causes of neoplastic transformation and cancer development. MAP17 is a cargo protein that transports membrane proteins from the endoplasmic reticulum. Therefore, its overexpression may be linked to an excess of membrane proteins that may be recognized as an unwanted signal, triggering local inflammation. Therefore, we analyzed whether its overexpression is related to an inflammatory phenotype. In this work, we found a correlation between MAP17 expression and inflammatory phenotype in tumors and in other inflammatory diseases such as Crohn's disease, Barrett's esophagus, COPD or psoriasis. MAP17 expression correlated also with the markers of inflammation HLAs, BBS10, HERC2, ADNP and PYCARD. Furthermore, we found that MAP17 expression directly regulates NFAT2 and IL-6 activation, inducing the differentiation of monocytes to dendritic cells and suggesting a causal role of MAP17 in inflammation. Immunohistochemistry confirms local inflammation, mainly CD45+ cells, at the site of expression of MAP17, at least in tumors, Crohn's and psoriasis. Therefore, our data indicates that the overexpression of the protein MAP17 plays important role in diseases involving chronic inflammation.
Collapse
Affiliation(s)
- José M García-Heredia
- Instituto de Biomedicina de Sevilla, IBIS/Hospital Universitario Virgen del Rocío/Universidad de Sevilla/Consejo Superior de Investigaciones Científicas, Seville, Spain.,Department of Vegetal Biochemistry and Molecular Biology, University of Seville, Seville, Spain.,CIBER de Cáncer, Instituto de Salud Carlos III, Madrid, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla, IBIS/Hospital Universitario Virgen del Rocío/Universidad de Sevilla/Consejo Superior de Investigaciones Científicas, Seville, Spain.,CIBER de Cáncer, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
16
|
Elevation of MAP17 enhances the malignant behavior of cells via the Akt/mTOR pathway in hepatocellular carcinoma. Oncotarget 2017; 8:92589-92603. [PMID: 29190940 PMCID: PMC5696206 DOI: 10.18632/oncotarget.21506] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 08/28/2017] [Indexed: 02/06/2023] Open
Abstract
MAP17, a small non-glycosylated membrane protein, was significantly up-regulated in hepatocellular carcinoma (HCC) tissues in our previous genome-wide microarray analysis. In this study, quantitative real-time RT-PCR and immunohistochemistry were applied to examine MAP17 mRNA and protein expression in primary HCC and matched peritumoral tissues. The disease-free survival (DFS) and overall survival (OS) was estimated using the Kaplan-Meier analysis. The expression of MAP17 was significantly higher in HCC tissues compared to the paired peritumoral tissues at both mRNA and protein levels. High MAP17 expression was positively correlated with gender, distant metastasis, early recurrence (≤ 2 year), and serum alpha-fetoprotein (all p < 0.05). Kaplan-Meier analysis showed that the DFS (p = 0.004) and OS (p = 0.013) in HCC patients with elevated expression of MAP17 were much worse than that in the low expression subgroup. High level of MAP17 was also significantly associated with a high probability of HCC early recurrence after surgical resection (p = 0.005). Cox regression analysis indicated MAP17 was an independent prognostic factor for DFS (HR, 1.710; 95% CI, 1.156-2.449, p = 0.012) and OS (HR, 1.743; 95% CI, 1.152-2.639, p = 0.009) in HCC. Silencing MAP17 significantly inhibited the proliferation, invasion and migration of HCC cells in vitro, and decreased the expression levels of Akt, p-Akt (Ser473), mTOR, p-mTOR (Ser2448) and MMP-9. Suggesting MAP17 was a novel diagnostic and prognostic biomarker for HCC patients and promoted HCC cell proliferation, invasion and migration via the Akt/mTOR pathway.
Collapse
|
17
|
Garcia-Heredia JM, Lucena-Cacace A, Verdugo-Sivianes EM, Pérez M, Carnero A. The Cargo Protein MAP17 (PDZK1IP1) Regulates the Cancer Stem Cell Pool Activating the Notch Pathway by Abducting NUMB. Clin Cancer Res 2017; 23:3871-3883. [PMID: 28153862 DOI: 10.1158/1078-0432.ccr-16-2358] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 12/23/2016] [Accepted: 01/12/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Cancer stem cells (CSC) are self-renewing tumor cells, with the ability to generate diverse differentiated tumor cell subpopulations. They differ from normal stem cells in the deregulation of the mechanisms that normally control stem cell physiology. CSCs are the origin of metastasis and highly resistant to therapy. Therefore, the understanding of the CSC origin and deregulated pathways is important for tumor control.Experimental Design: We have included experiments in vitro, in cell lines and tumors of different origins. We have used patient-derived xenografts (PDX) and public transcriptomic databases of human tumors.Results: MAP17 (PDZKIP1), a small cargo protein overexpressed in tumors, interacts with NUMB through the PDZ-binding domain activating the Notch pathway, leading to an increase in stem cell factors and cancer-initiating-like cells. Identical behavior was mimicked by inhibiting NUMB. Conversely, MAP17 downregulation in a tumor cell line constitutively expressing this gene led to Notch pathway inactivation and a marked reduction of stemness. In PDX models, MAP17 levels directly correlated with tumorsphere formation capability. Finally, in human colon, breast, or lung there is a strong correlation of MAP17 expression with a signature of Notch and stem cell genes.Conclusions: MAP17 overexpression activates Notch pathway by sequestering NUMB. High levels of MAP17 correlated with tumorsphere formation and Notch and Stem gene transcription. Its direct modification causes direct alteration of tumorsphere number and Notch and Stem pathway transcription. This defines a new mechanism of Notch pathway activation and Stem cell pool increase that may be active in a large percentage of tumors. Clin Cancer Res; 23(14); 3871-83. ©2017 AACR.
Collapse
Affiliation(s)
- Jose Manuel Garcia-Heredia
- Instituto de Biomedicina de Sevilla, IBIS/Hospital Universitario Virgen del Rocio/Universidad de Sevilla/Consejo Superior de Investigaciones Cientificas, Seville, Spain
- Department of Vegetal Biochemistry and Molecular Biology, University of Seville, Seville, Spain
- CIBER de Cancer, Seville, Spain
| | - Antonio Lucena-Cacace
- Instituto de Biomedicina de Sevilla, IBIS/Hospital Universitario Virgen del Rocio/Universidad de Sevilla/Consejo Superior de Investigaciones Cientificas, Seville, Spain
- CIBER de Cancer, Seville, Spain
| | - Eva M Verdugo-Sivianes
- Instituto de Biomedicina de Sevilla, IBIS/Hospital Universitario Virgen del Rocio/Universidad de Sevilla/Consejo Superior de Investigaciones Cientificas, Seville, Spain
- CIBER de Cancer, Seville, Spain
| | - Marco Pérez
- Instituto de Biomedicina de Sevilla, IBIS/Hospital Universitario Virgen del Rocio/Universidad de Sevilla/Consejo Superior de Investigaciones Cientificas, Seville, Spain
- CIBER de Cancer, Seville, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla, IBIS/Hospital Universitario Virgen del Rocio/Universidad de Sevilla/Consejo Superior de Investigaciones Cientificas, Seville, Spain.
- CIBER de Cancer, Seville, Spain
| |
Collapse
|
18
|
de Miguel-Luken MJ, Chaves-Conde M, de Miguel-Luken V, Muñoz-Galván S, López-Guerra JL, Mateos JC, Pachón J, Chinchón D, Suarez V, Carnero A. MAP17 (PDZKIP1) as a novel prognostic biomarker for laryngeal cancer. Oncotarget 2016; 6:12625-36. [PMID: 25788275 PMCID: PMC4494962 DOI: 10.18632/oncotarget.3470] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 01/20/2015] [Indexed: 11/25/2022] Open
Abstract
Larynx cancer organ preservation treatments with chemo and radiotherapy have substantially improved laryngoesophageal dysfunction-free survival. However, both of them lead to a high incidence of acute and chronic toxicities and a significant number of patients relapse. To date, there is no evidence available to establish the group of patients that may benefit from preservation approaches and clinical criteria such as primary tumor extension or pretreatment tracheotomy are not validated. MAP17 is a small non-glycosylated membrane protein overexpressed in carcinomas. The tumoral behavior induced by MAP17 is associated with reactive oxygen species production in which SGLT1 seems involved. In this study we found that the levels of MAP17 were related to clinical findings and survival in a cohort of 58 patients with larynx cancer. MAP17 expression is associated with overall survival (p<0.001) and laryngoesophageal dysfunction-free survival (p=0.002). Locoregional control in patients with high MAP17 showed better outcomes than those with low MAP17 (p=0.016). Besides, a positive correlation was observed between MAP17 expression and SGLT (p=0.022) and the combination of high levels of MAP17/SGLT also led to an increased overall survival (p=0,028). These findings suggest that MAP17, alone or in combination with SGLT1, may become a novel predictive biomarker for laryngeal carcinoma.
Collapse
Affiliation(s)
| | - Manuel Chaves-Conde
- Department of Medical Oncology, Hospital Universitario Virgen del Rocío, Seville, Spain
| | | | - Sandra Muñoz-Galván
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Seville, Spain
| | - José Luis López-Guerra
- Department of Radiation Oncology, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Juan C Mateos
- Department of Radiation Oncology, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Jerónimo Pachón
- Department of Radiation Oncology, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - David Chinchón
- Department of Pathology, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Vladimir Suarez
- Department of Radiation Oncology, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Seville, Spain
| |
Collapse
|
19
|
Muñoz-Galván S, Gutierrez G, Perez M, Carnero A. MAP17 (PDZKIP1) Expression Determines Sensitivity to the Proteasomal Inhibitor Bortezomib by Preventing Cytoprotective Autophagy and NFκB Activation in Breast Cancer. Mol Cancer Ther 2015; 14:1454-65. [PMID: 25837675 DOI: 10.1158/1535-7163.mct-14-1053] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 03/24/2015] [Indexed: 11/16/2022]
Abstract
MAP17 is a small nonglycosylated membrane protein that is overexpressed in a high percentage of carcinomas. High levels of MAP17 enhance the tumorigenic properties of tumor cells by increasing oxidative stress, which is dependent on Na(+)-coupled cotransport. Here, we show that MAP17 is associated with proteins involved in protein degradation and that proteasome inhibition induces autophagy. To analyze whether MAP17 could also alter this process, we used the proteasome inhibitor bortezomib (Velcade, PS-341), which is approved for the treatment of multiple myeloma and mantle cell lymphoma, although it has a high rate of resistance emergence and poor efficacy in solid tumors. We provide evidence that bortezomib induces a cytoprotective effect by activating autophagy and NFκB nuclear translocation, responses that are repressed in the presence of high levels of MAP17 both in vitro and in vivo. Indeed, patients with multiple myeloma treated with bortezomib showed higher response rates and a longer time to progression associated with increased levels of MAP17 expression. The MAP17-induced sensitivity to bortezomib is dependent on the oxidative status of the cells and the activity of Na(+)-coupled transporters because treatment with antioxidants or the inhibitor furosemide restores the cytoprotective activity induced by bortezomib. Therefore, bortezomib induces a prosurvival response through cytoprotective autophagy and NFκB nuclear translocation, which is repressed by high levels of MAP17. We propose that the levels of MAP17 could be used as a prognostic marker to predict the response to bortezomib in hematologic malignancies and in other tissues that are not commonly responsive to the drug.
Collapse
Affiliation(s)
- Sandra Muñoz-Galván
- Instituto de Biomedicina de Sevilla, IBIS/Hospital Universitario Virgen del Rocio/Universidad de Sevilla/Consejo Superior de Investigaciones Cientificas, Seville, Spain
| | | | - Marco Perez
- Instituto de Biomedicina de Sevilla, IBIS/Hospital Universitario Virgen del Rocio/Universidad de Sevilla/Consejo Superior de Investigaciones Cientificas, Seville, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla, IBIS/Hospital Universitario Virgen del Rocio/Universidad de Sevilla/Consejo Superior de Investigaciones Cientificas, Seville, Spain.
| |
Collapse
|
20
|
Lai MS, Cheng YS, Chen PR, Tsai SJ, Huang BM. Fibroblast growth factor 9 activates akt and MAPK pathways to stimulate steroidogenesis in mouse leydig cells. PLoS One 2014; 9:e90243. [PMID: 24603862 PMCID: PMC3946167 DOI: 10.1371/journal.pone.0090243] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 01/27/2014] [Indexed: 12/22/2022] Open
Abstract
Fibroblast growth factor 9 (FGF9) is a multifunctional polypeptide belonging to the FGF family and has functions related to bone formation, lens-fiber differentiation, nerve development, gap-junction formation and sex determination. In a previous study, we demonstrated that FGF9 stimulates the production of testosterone in mouse Leydig cells. In the present study, we used both primary mouse Leydig cells and MA-10 mouse Leydig tumor cells to further investigate the molecular mechanism of FGF9-stimulated steroidogenesis. Results showed that FGF9 significantly activated steroidogenesis in both mouse primary and tumor Leydig cells (p<0.05). Furthermore, FGF9 significantly induced the expression of phospho-Akt at 0.5 and 24 hr, phospho-JNK at 0.25, 0.5, and 24 hr, phospho-p38 at 0.5 hr, and phospho-ERK1/2 from 0.25 to 24 hr in primary Leydig cells (p<0.05). Also, FGF9 significantly up-regulated the expression of phospho-Akt at 3 hr, phospho-JNK at 0.25 hr, and phospho-ERK1/2 at 1 and 3 hr in MA-10 cells (p<0.05). Using specific inhibitors of Akt, JNK, p38, and ERK1/2, we further demonstrated that the inhibitors of Akt and ERK1/2 significantly suppressed the stimulatory effect of FGF9 on steroidogenesis in mouse Leydig cells. In conclusion, FGF9 specifically activated the Akt and ERK1/2 in normal mouse Leydig cells and the Akt, JNK and ERK1/2 in MA-10 mouse Leydig tumor cells to stimulate steroidogenesis.
Collapse
Affiliation(s)
- Meng-Shao Lai
- Institute of Basic Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China
| | - Yu-Sheng Cheng
- Department of Urology, National Cheng Kung University Hospital Douliou Branch, Yunlin, Taiwan, Republic of China
| | - Pei-Rong Chen
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China
| | - Shaw-Jenq Tsai
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China
| | - Bu-Miin Huang
- Institute of Basic Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China
- * E-mail:
| |
Collapse
|
21
|
Han BW, Chen YQ. Potential pathological and functional links between long noncoding RNAs and hematopoiesis. Sci Signal 2013; 6:re5. [PMID: 23962981 DOI: 10.1126/scisignal.2004099] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Differential abundance and activity of long noncoding RNAs (lncRNAs) are recognized as the hallmark features in various diseases. We highlight the lncRNAs that play a functional role in the development of blood cells. Many lncRNAs and the protein complexes within which they interact have been implicated in various types of cancers. Multiple lncRNAs participate in normal and may be implicated in malignant hematopoiesis associated with blood cell cancers, such as leukemia, by regulating gene expression through such mechanisms as redirecting chromatin remodeling complexes and activating epigenetic silencing, either of which can inactivate tumor suppressor genes or activate oncogenes. Because of their potential importance in cancers of the blood, lncRNAs may be useful as diagnostic and prognostic markers, and it may be possible to develop lncRNA-mediated therapy.
Collapse
Affiliation(s)
- Bo-Wei Han
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Science, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | | |
Collapse
|
22
|
MAP17 and SGLT1 protein expression levels as prognostic markers for cervical tumor patient survival. PLoS One 2013; 8:e56169. [PMID: 23418532 PMCID: PMC3572015 DOI: 10.1371/journal.pone.0056169] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 01/07/2013] [Indexed: 02/02/2023] Open
Abstract
MAP17 is a membrane-associated protein that is overexpressed in human tumors. Because the expression of MAP17 increases reactive oxygen species (ROS) generation through SGLT1 in cancer cells, in the present work, we investigated whether MAP17 and/or SGLT1 might be markers for the activity of treatments involving oxidative stress, such as cisplatin or radiotherapy. First, we confirmed transcriptional alterations in genes involved in the oxidative stress induced by MAP17 expression in HeLa cervical tumor cells and found that Hela cells expressing MAP17 were more sensitive to therapies that induce ROS than were parental cells. Furthermore, MAP17 increased glucose uptake through SGLT receptors. We then analyzed MAP17 and SGLT1 expression levels in cervical tumors treated with cisplatin plus radiotherapy and correlated the expression levels with patient survival. MAP17 and SGLT1 were expressed in approximately 70% and 50% of cervical tumors of different types, respectively, but they were not expressed in adenoma tumors. Furthermore, there was a significant correlation between MAP17 and SGLT1 expression levels. High levels of either MAP17 or SGLT1 correlated with improved patient survival after treatment. However, the patients with high levels of both MAP17 and SGLT1 survived through the end of this study. Therefore, the combination of high MAP17 and SGLT1 levels is a marker for good prognosis in patients with cervical tumors after cisplatin plus radiotherapy treatment. These results also suggest that the use of MAP17 and SGLT1 markers may identify patients who are likely to exhibit a better response to treatments that boost oxidative stress in other cancer types.
Collapse
|
23
|
Abstract
MAP17 is a small 17 kDa non-glycosylated membrane protein previously identified as being overexpressed in carcinomas. Breast tumor cells that overexpress MAP17 show an increased tumoral phenotype with enhanced proliferative capabilities both in the presence or the absence of contact inhibition, decreased apoptotic sensitivity, and increased migration. MAP17-expressing clones also grow better in nude mice. The increased malignant cell behavior induced by MAP17 is associated with an increase in reactive oxygen species (ROS) production, and the treatment of MAP17-expressing cells with antioxidants results in a reduction in the tumorigenic properties of these cells. The MAP17-dependent increase in ROS and tumorigenesis relies on its PDZ-binding domain because disruption of this sequence by point mutations abolishes the ability of MAP17 to enhance ROS production and tumorigenesis. MAP17 is overexpressed in a great variety of human carcinomas, including breast tumors. Immunohistochemical analysis of MAP17 during cancer progression demonstrates that overexpression of the protein strongly correlates with tumoral progression. Generalized MAP17 overexpression in human carcinomas indicates that MAP17 can be a good marker for tumorigenesis and, especially, for malignant progression.
Collapse
Affiliation(s)
- Amancio Carnero
- Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío, Consejo Superior de Investigaciones Cientificas, Universidad de Sevilla Sevilla, Spain
| |
Collapse
|
24
|
Carnero A. MAP17 and the double-edged sword of ROS. Biochim Biophys Acta Rev Cancer 2012; 1826:44-52. [PMID: 22465409 DOI: 10.1016/j.bbcan.2012.03.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Revised: 03/14/2012] [Accepted: 03/15/2012] [Indexed: 01/18/2023]
Abstract
Reactive oxygen species, ROS, are beneficially involved in many signaling pathways that control development and maintain cellular homeostasis. In physiological conditions, a tightly regulated redox balance protects cells from injurious ROS activity, but if the balance is altered, it promotes various pathological conditions including cancer. Understanding the duality of ROS as cytotoxic molecules and key mediators in signaling cascades may provide novel opportunities for improved cancer therapy. MAP17 is a small 17-kDa non-glycosylated membrane protein that is overexpressed in many tumors of different origins, including carcinomas. Immunohistochemical analysis of MAP17 during cancer progression demonstrates that overexpression of the protein strongly correlates with the progression of most types of tumor. Tumor cells that overexpress MAP17 show an increased tumoral phenotype associated with an increase in ROS. However, in non-tumor cells MAP17 increases ROS, resulting in senescence or apoptosis. Therefore, in tumor cells, MAP17 could be a marker for increased oxidative stress and could define new therapeutic approaches. Here, we review the role of MAP17 as a putative oncogene, as well as its role in cancer and anticancer therapies.
Collapse
Affiliation(s)
- Amancio Carnero
- Instituto de Biomedicina de Sevilla (IBIS), HUVR/CSIC/Universidad de Sevilla, Consejo Superior de Investigaciones Cientificas, Campus Hospital Universitario Virgen del Rocío, Sevilla, Spain.
| |
Collapse
|