1
|
Mortezagholi S, Maghsood F, Shojaeian S, Shokri F, Amiri MM, Ghorbani A, Shabani M, Zarnani AH. Production and characterization of a panel of anti-mouse placenta-specific protein 1 (plac1) monoclonal antibodies. Anal Biochem 2025; 696:115682. [PMID: 39332465 DOI: 10.1016/j.ab.2024.115682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/02/2024] [Accepted: 09/24/2024] [Indexed: 09/29/2024]
Abstract
Placenta-Specific Protein 1 (PLAC1) is essential for normal placental and embryonic development. It is widely expressed in various types of cancer cells. We produced a panel of anti-mouse plac1 monoclonal antibodies (mAbs) with different applications. Two recombinant proteins were produced containing either the extracellular domain (ED) plus tetanus toxin P2, P30, pan-DR epitope (PADRE), and KDEL3 (main plac1) or ED plus KDEL3 (control plac1). Recombinant proteins were used for immunization and screening. Positive clones were selected by ELISA and flow cytometry. Purified mAbs were tested by ELISA, WB, flow cytometry, immunohistochemistry (IHC), and immunofluorescent (IF). A combination of bioinformatics tools was used to predict the target epitope(s) of the mAbs. Eight anti-mouse plac1 mAbs (all IgG1/κ1) were generated, all reacting with high affinity in ELISA. Seven clones recognized plac1 in both reduced and non-reduced Western blots, while one only recognized the non-reduced form. Cross-inhibition ELISA revealed that all mAbs recognized overlapping epitopes with a shared motif except for 5C9. Four clones reacted with the native antigen in flow cytometry, but none were functional in IF or IHC staining. The produced multifunctional mAbs can be used to investigate different aspects of PLAC1 biology in reproduction and cancer.
Collapse
Affiliation(s)
- Sahar Mortezagholi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Faezeh Maghsood
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Sorour Shojaeian
- Department of Biochemistry, Alborz University of Medical Sciences, Karaj, Iran
| | - Fazel Shokri
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Mehdi Amiri
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Ghorbani
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Shabani
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Amir-Hassan Zarnani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran; Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran.
| |
Collapse
|
2
|
Farhangnia P, Ghods R, Falak R, Zarnani AH, Delbandi AA. Identification of placenta-specific protein 1 (PLAC-1) expression on human PC-3 cell line-derived prostate cancer stem cells compared to the tumor parental cells. Discov Oncol 2024; 15:251. [PMID: 38943028 PMCID: PMC11213845 DOI: 10.1007/s12672-024-01121-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 06/25/2024] [Indexed: 06/30/2024] Open
Abstract
Placenta-specific protein 1 (PLAC-1) is a gene primarily expressed in the placenta and the testis. Interestingly, it is also found to be expressed in many solid tumors, and it is involved in malignant cell features. However, no evidence has been reported regarding the relationship between PLAC-1 and cancer stem cells (CSCs). In the current research, we explored the expression of the PLAC-1 molecule in prostate cancer stem cells (PCSCs) derived from the human PC-3 cell line. The enrichment of PCSCs was achieved using a three-dimensional cell culture technique known as the sphere-formation assay. To confirm the identity of PCSCs, we examined the expression of genes associated with stemness and pluripotency, such as SOX2, OCT4, Nanog, C-Myc, and KLF-4, as well as stem cell differentiation molecules like CD44 and CD133. These evaluations were conducted in both the PCSCs and the original tumor cells (parental cells) using real-time PCR and flow cytometry. Subsequently, we assessed the expression of the PLAC-1 molecule in both enriched cells and parental tumor cells at the gene and protein levels using the same techniques. The tumor cells from the PC-3 cell line formed spheroids with CSC characteristics in a non-adherent medium. The expression of SOX2, OCT4, Nanog, and C-Myc genes (p < 0.01), and the molecules CD44 and CD133 (p < 0.05) were significantly elevated in PCSCs compared to the parental cells. The expression of the PLAC-1 molecule in PCSCs showed a significant increase compared to the parental cells at both gene (p < 0.01) and protein (p < 0.001) levels. In conclusion, it was indicated for the first time that PLAC-1 is up-regulated in PCSCs derived from human PC-3 cell line. This study may propose PLAC-1 as a potential target in targeted therapies, which should be confirmed through further studies.
Collapse
Affiliation(s)
- Pooya Farhangnia
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Roya Ghods
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Falak
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Amir-Hassan Zarnani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali-Akbar Delbandi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran.
- Reproductive Sciences and Technology Research Center, Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Cagliani R, Fayed B, Jagal J, Shakartalla SB, Soliman SSM, Haider M. Peptide-functionalized zinc oxide nanoparticles for the selective targeting of breast cancer expressing placenta-specific protein 1. Colloids Surf B Biointerfaces 2023; 227:113357. [PMID: 37210795 DOI: 10.1016/j.colsurfb.2023.113357] [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: 03/16/2023] [Revised: 04/20/2023] [Accepted: 05/15/2023] [Indexed: 05/23/2023]
Abstract
Functionalized metal oxide nanoparticles (NPs) have demonstrated specific binding affinity to antigens or receptors presented on the cancer cell surface, favouring selective targeting and minimizing side effects during the chemotherapy. Placenta-specific protein 1 (PLAC-1) is a small cell surface protein overexpressed in certain types of breast cancer (BC); therefore, it can be used as a therapeutic target. The objective of this study is to develop NPs that can bind PLAC-1 and hence can inhibit the progression and metastatic potential of BC cells. Zinc oxide (ZnO) NPs were coated with a peptide (GILGFVFTL), which possesses a strong binding ability to PLAC-1. The physical attachment of the peptide to ZnO NPs was verified through various physicochemical and morphological characterization techniques. The selective cytotoxicity of the designed NPs was investigated using PLAC-1-bearing MDA-MB 231 human BC cell line and compared to LS-180 cells that do not express PLAC-1. The anti-metastatic and pro-apoptotic effects of the functionalized NPs on MDA-MB 231 cells were examined. Confocal microscopy was used to investigate the mechanism of NPs uptake by MDA-MB 231 cells. Compared to non-functionalized NPs, peptide functionalization significantly improved the targeting and uptake of the designed NPs by PLAC-1-expressing cancer cells with significant pro-apoptotic and anti-metastatic effects. The uptake of peptide functionalized ZnO NPs (ZnO-P NPs) occurred via peptide-PLAC1 interaction-assisted clathrin-mediated endocytosis. These findings highlight the potential targeted therapy of ZnO-P NPs against PLAC-1-expressing breast cancer cells.
Collapse
Affiliation(s)
- Roberta Cagliani
- Research Institute of Medical & Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates
| | - Bahgat Fayed
- Research Institute of Medical & Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates; Chemistry of Natural and Microbial Product Department, National Research Centre, Cairo 12622, Egypt
| | - Jayalakshmi Jagal
- Research Institute of Medical & Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates
| | - Sarra B Shakartalla
- Research Institute of Medical & Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates; College of Medicine, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates; Faculty of Pharmacy, University of Gezira, P.O. Box. 21111, Wadmedani, Sudan
| | - Sameh S M Soliman
- Research Institute of Medical & Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates; Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, 27272 Sharjah, United Arab Emirates
| | - Mohamed Haider
- Research Institute of Medical & Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates; Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah, 27272 Sharjah, United Arab Emirates.
| |
Collapse
|
4
|
Peptide-functionalized graphene oxide quantum dots as colorectal cancer theranostics. J Colloid Interface Sci 2023; 630:698-713. [PMID: 36274405 DOI: 10.1016/j.jcis.2022.10.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 09/30/2022] [Accepted: 10/12/2022] [Indexed: 11/11/2022]
|
5
|
Devor EJ, Santillan DA, Warrier A, Scroggins SM, Santillan MK. Placenta-specific protein 1 (PLAC1) expression is significantly down-regulated in preeclampsia via a hypoxia-mediated mechanism. J Matern Fetal Neonatal Med 2022; 35:8419-8425. [PMID: 34565269 PMCID: PMC8959068 DOI: 10.1080/14767058.2021.1977792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 09/03/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Examine a mechanism of PLAC1 regulation and its potential role in preeclampsia (PE). MATERIALS AND METHODS Placental tissue samples and detailed clinical information were obtained through the University of Iowa Maternal Fetal Tissue Bank (IRB# 200910784) from gestational and maternal age-matched control (n = 17) and PE affected pregnancies (n = 12). PLAC1 and PLAC1 promoter-specific expression was measured using quantitative polymerase chain reaction (qPCR) and differences were assessed via the standard ΔΔCt method. In addition, the role of hypoxia in PLAC1 transcription was investigated through the exposure of HTR8/SVneo human trophoblast cells to the hypoxia mimic dimethyloxaloylglycine (DMOG). RESULTS PLAC1 expression is seen to be 8.9-fold lower in human placentas affected by preeclampsia in comparison with controls (p < .05). Further, this decrease is paralleled by a significantly lower expression of the P2 or proximal PLAC1 promoter (p < .05). Expression of mediator complex subunit 1 (MED1), a known hypoxia-sensitive transcription coactivator and PLAC1 effector, is significantly correlated with PLAC 1 expression (r2 = 0.607, p < .001). These data suggest that PLAC1 expression is significantly down-regulated in preeclampsia at least in part via a MED1 hypoxia-mediated mechanism. CONCLUSIONS We confirm that PLAC1 transcription is suppressed in the placentae of women affected by preeclampsia. We further demonstrate that this suppression is driven through the P2 or proximal PLAC1 promoter. This demonstration led to the identification of the MED1-TRAP cofactor complex as the hypoxia-sensitive driver.
Collapse
Affiliation(s)
- Eric J. Devor
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Donna A. Santillan
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Akshaya Warrier
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Sabrina M. Scroggins
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Mark K. Santillan
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| |
Collapse
|
6
|
Rahdan S, Razavi SA, Shojaeian S, Shokri F, Amiri MM, Zarnani AH. Immunization with placenta-specific 1 (plac1) induces potent anti-tumor responses and prolongs survival in a mouse model of melanoma. Adv Med Sci 2022; 67:338-345. [PMID: 36084365 DOI: 10.1016/j.advms.2022.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 05/07/2022] [Accepted: 08/16/2022] [Indexed: 12/30/2022]
Abstract
PURPOSE Melanoma is a malignant and metastatic form of skin cancer, which is not diagnosed in early stages of the disease. Nowadays, immunotherapy is changing the treatment landscape for metastatic melanoma. Placenta-specific1 (PLAC1) is a cancer-testis-placenta (CTP) antigen with differential expression in melanoma tissues. Here, we evaluated the potential of plac1 to induce anti-cancer immune responses as well as to prevent cancer development in a mouse model of melanoma. METHODS Two proteins containing full extracellular domain (ED) of mouse plac1+KDEL3 and full ED of mouse plac1+ tetanus toxin P2 and P30+ pan DR epitope (PADRE) + KDEL3 were produced and injected in mice to evaluate their capacity to induce anti-cancer immune responses as well as their potential to prevent melanoma development. Induction of plac1-specific humoral and cellular responses as well as tumor-associated parameters were tested in a series of 36 mice. RESULTS Sera of mice immunized with ED + P2P30+PADRE + KDEL3 contained antibodies able to react with surface plac1 in B16F10 cells. Both proteins induced proliferative cellular immune responses against B16F10 cells and plac1-specific cytotoxic T cells (CTL) and CD107a + CTL responses, which was higher in mice immunized with ED + P2P30+PADRE + KDEL3. Splenocytes of mice vaccinated with ED + P2P30+PADRE + KDEL3 exerted a significant cytotoxicity against B16F10 cells. Vaccination with ED + P2P30+PADRE + KDEL3 significantly delayed B16F10-induced tumor onset, reduced tumor growth, and increased survival. Tumors induced by B16F10 expressed plac1 in vivo. CONCLUSION Our results pave the way for development of effective melanoma preventive vaccine in humans, although further studies are needed.
Collapse
Affiliation(s)
- Shaghayegh Rahdan
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Alireza Razavi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Sorour Shojaeian
- Department of Biochemistry, School of Medical Sciences, Alborz University of Medical Sciences, Karaj, Iran
| | - Fazel Shokri
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Mehdi Amiri
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir-Hassan Zarnani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran; Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran.
| |
Collapse
|
7
|
Rahat B, Hamid A, Bagga R, Kaur J. Folic Acid Levels During Pregnancy Regulate Trophoblast Invasive Behavior and the Possible Development of Preeclampsia. Front Nutr 2022; 9:847136. [PMID: 35578613 PMCID: PMC9106796 DOI: 10.3389/fnut.2022.847136] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 03/18/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundOne of the unique features of placentation is its similarity to tumorigenesis yet being very well regulated. It allows rapid proliferation, migration, and invasion of mononuclear trophoblast cells into the maternal uterus and remodeling the maternal vasculature. This pseudomalignant nature of trophoblastic cells is strictly regulated and its importance becomes evident in abnormal pregnancies that are characterized by aberrant trophoblast proliferation/invasion like preeclampsia. In addition to this, the importance of folic acid supplementation during pregnancy is well documented. We aimed to analyze the molecular and epigenetic regulation of the pseudomalignant nature of placentation via folic acid levels.MethodsPlacental tissue samples were collected from different pregnancies in three different gestational stages. We estimated the impact of folic acid levels on global methylation, LINE1 methylation, and expression of DNMTs in all three gestational stages in pregnant women and preeclampsia pregnancies. We also analyzed the effect of folic acid supplementation on trophoblastic invasion using placental derived cells viz, JEG-3 and HTR-8/SVneo cell line and verified the molecular and epigenetic mechanisms involved in this regulation.ResultsDevelopment of preeclampsia was observed to be associated with lower folate levels in placental tissue, higher global methylation level, and higher expression of DNMT1and DNMT3A. Folic acid supplementation was found to increase the invasive potential of placental trophoblasts by almost two folds which were associated with the decreased expression of tumor suppressor genes and tissue inhibitors of matrix metalloproteinases; and increased expression of oncogenes, telomerase gene, and matrix metalloproteinases. These folic acid-mediated changes were observed to be regulated by CpG methylation in the case of many genes. Folic acid supplementation was also observed to significantly decrease global methylation in placental trophoblasts related to decreasing expression of DNMT1 and DNMT3A.ConclusionLower folic acid levels are associated with preeclampsia development and folic acid supplementation regulates the invasive potential of placental trophoblasts as mediated by various epigenetic changes in the placenta suggesting the protective effect of folic acid against preeclampsia.
Collapse
Affiliation(s)
- Beenish Rahat
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Abid Hamid
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Rashmi Bagga
- Department of Obstetrics and Gynecology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Jyotdeep Kaur
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh, India
- *Correspondence: Jyotdeep Kaur
| |
Collapse
|
8
|
Alsayed SSR, Suri A, Bailey AW, Lane S, Werry EL, Huang CC, Yu LF, Kassiou M, Sredni ST, Gunosewoyo H. Synthesis and antitumour evaluation of indole-2-carboxamides against paediatric brain cancer cells. RSC Med Chem 2021; 12:1910-1925. [PMID: 34825187 PMCID: PMC8597418 DOI: 10.1039/d1md00065a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 08/05/2021] [Indexed: 11/21/2022] Open
Abstract
Paediatric glioblastomas are rapidly growing, devastating brain neoplasms with an invasive phenotype. Radiotherapy and chemotherapy, which are the current therapeutic adjuvant to surgical resection, are still associated with various toxicity profiles and only marginally improve the course of the disease and life expectancy. A considerable body of evidence supports the antitumour and apoptotic effects of certain cannabinoids, such as WIN55,212-2, against a wide spectrum of cancer cells, including gliomas. In fact, we previously highlighted the potent cytotoxic activity of the cannabinoid ligand 5 against glioblastoma KNS42 cells. Taken together, in this study, we designed, synthesised, and evaluated several indoles and indole bioisosteres for their antitumour activities. Compounds 8a, 8c, 8f, 12c, and 24d demonstrated significant inhibitory activities against the viability (IC50 = 2.34-9.06 μM) and proliferation (IC50 = 2.88-9.85 μM) of paediatric glioblastoma KNS42 cells. All five compounds further retained their antitumour activities against two atypical teratoid/rhabdoid tumour (AT/RT) cell lines. When tested against a medulloblastoma DAOY cell line, only 8c, 8f, 12c, and 24d maintained their viability inhibitory activities. The viability assay against non-neoplastic human fibroblast HFF1 cells suggested that compounds 8a, 8c, 8f, and 12c act selectively towards the panel of paediatric brain tumour cells. In contrast, compound 24d and WIN55,212-2 were highly toxic toward HFF1 cells. Due to their structural resemblance to known cannabimimetics, the most potent compounds were tested in cannabinoid 1 and 2 receptor (CB1R and CB2R) functional assays. Compounds 8a, 8c, and 12c failed to activate or antagonise both CB1R and CB2R, whereas compounds 8f and 24d antagonised CB1R and CB2R, respectively. We also performed a transcriptional analysis on KNS42 cells treated with our prototype compound 8a and highlighted a set of seven genes that were significantly downregulated. The expression levels of these genes were previously shown to be positively correlated with tumour growth and progression, indicating their implication in the antitumour activity of 8a. Overall, the drug-like and selective antitumour profiles of indole-2-carboxamides 8a, 8c, 8f, and 12c substantiate the versatility of the indole scaffold in cancer drug discovery.
Collapse
Affiliation(s)
- Shahinda S R Alsayed
- Curtin Medical School, Faculty of Health Sciences, Curtin University Bentley Perth WA 6102 Australia
| | - Amreena Suri
- Division of Pediatric Neurosurgery, Ann and Robert H. Lurie Children's Hospital of Chicago Chicago IL 60611 USA
| | - Anders W Bailey
- Division of Pediatric Neurosurgery, Ann and Robert H. Lurie Children's Hospital of Chicago Chicago IL 60611 USA
| | - Samuel Lane
- School of Chemistry, The University of Sydney NSW 2006 Australia
| | - Eryn L Werry
- School of Chemistry, The University of Sydney NSW 2006 Australia
- Faculty of Medicine and Health, The University of Sydney NSW 2006 Australia
| | - Chiang-Ching Huang
- Department of Biostatistics, Zilber School of Public Health, University of Wisconsin Milwaukee WI 53205 USA
| | - Li-Fang Yu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University 3663 North Zhongshan Road Shanghai 200062 China
| | - Michael Kassiou
- School of Chemistry, The University of Sydney NSW 2006 Australia
| | - Simone Treiger Sredni
- Division of Pediatric Neurosurgery, Ann and Robert H. Lurie Children's Hospital of Chicago Chicago IL 60611 USA
- Department of Surgery, Northwestern University, Feinberg School of Medicine Chicago IL 60611 USA
| | - Hendra Gunosewoyo
- Curtin Medical School, Faculty of Health Sciences, Curtin University Bentley Perth WA 6102 Australia
| |
Collapse
|
9
|
Devor EJ, Schickling BM, Lapierre JR, Bender DP, Gonzalez-Bosquet J, Leslie KK. The Synthetic Curcumin Analog HO-3867 Rescues Suppression of PLAC1 Expression in Ovarian Cancer Cells. Pharmaceuticals (Basel) 2021; 14:ph14090942. [PMID: 34577642 PMCID: PMC8465575 DOI: 10.3390/ph14090942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/06/2021] [Accepted: 09/09/2021] [Indexed: 12/19/2022] Open
Abstract
Elevated expression of placenta-specific protein 1 (PLAC1) is associated with the increased proliferation and invasiveness of a variety of human cancers, including ovarian cancer. Recent studies have shown that the tumor suppressor p53 directly suppresses PLAC1 transcription. However, mutations in p53 lead to the loss of PLAC1 transcriptional suppression. Small molecules that structurally convert mutant p53 proteins to wild-type conformations are emerging. Our objective was to determine whether the restoration of the wild-type function of mutated p53 could rescue PLAC1 transcriptional suppression in tumors harboring certain TP53 mutations. Ovarian cancer cells OVCAR3 and ES-2, both harboring TP53 missense mutations, were treated with the p53 reactivator HO-3867. Treatment with HO-3867 successfully rescued PLAC1 transcriptional suppression. In addition, cell proliferation was inhibited and cell death through apoptosis was increased in both cell lines. We conclude that the use of HO-3867 as an adjuvant to conventional therapeutics in ovarian cancers harboring TP53 missense mutations could improve patient outcomes. Validation of this conclusion must, however, come from an appropriately designed clinical trial.
Collapse
Affiliation(s)
- Eric J. Devor
- Department of Obstetrics and Gynecology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA; (B.M.S.); (J.R.L.); (D.P.B.); (J.G.-B.); (K.K.L.)
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
- Correspondence:
| | - Brandon M. Schickling
- Department of Obstetrics and Gynecology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA; (B.M.S.); (J.R.L.); (D.P.B.); (J.G.-B.); (K.K.L.)
| | - Jace R. Lapierre
- Department of Obstetrics and Gynecology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA; (B.M.S.); (J.R.L.); (D.P.B.); (J.G.-B.); (K.K.L.)
| | - David P. Bender
- Department of Obstetrics and Gynecology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA; (B.M.S.); (J.R.L.); (D.P.B.); (J.G.-B.); (K.K.L.)
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Jesus Gonzalez-Bosquet
- Department of Obstetrics and Gynecology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA; (B.M.S.); (J.R.L.); (D.P.B.); (J.G.-B.); (K.K.L.)
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Kimberly K. Leslie
- Department of Obstetrics and Gynecology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA; (B.M.S.); (J.R.L.); (D.P.B.); (J.G.-B.); (K.K.L.)
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| |
Collapse
|
10
|
Chen R, Sheng C, Ma R, Zhang L, Yang L, Chen Y. PLAC1 is an independent predictor of poor survival, and promotes cell proliferation and invasion in cervical cancer. Mol Med Rep 2021; 24:800. [PMID: 34523695 PMCID: PMC8456314 DOI: 10.3892/mmr.2021.12440] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 08/11/2021] [Indexed: 11/24/2022] Open
Abstract
Placenta-specific protein 1 (PLAC1) is inversely associated with survival in several types of cancer. However, whether PLAC1 is involved in the progression of cervical cancer (CC) remains to be elucidated. Therefore, the present study aimed to evaluate the prognostic role of PLAC1 in CC by determining the relationship between clinicopathological factors, PLAC1 gene expression and survival prognosis using univariate and multivariate Cox proportional-hazards regression analyses. Similarly, Kaplan-Meier curves were evaluated with the log-rank test. Subsequently, gene set enrichment analysis was performed to compare the high- and low-PLAC1 expression phenotypes. Functional studies were further conducted in PLAC1-overexpressing HeLa cells and PLAC1-silenced MS751 cells, and western blotting was performed to determine whether PLAC1 promoted CC progression via epithelial-mesenchymal transition (EMT). The findings demonstrated that high expression of PLAC1 was associated with American Joint Committee on Cancer metastasis pathological score and suggested a poor overall survival. ‘mTOR complex 1 signaling’, ‘interferon α response’ and ‘hypoxia’ were differentially enriched in the high-PLAC1 phenotype. Furthermore, PLAC1 promoted the invasion of CC cells in vitro. E-cadherin expression was decreased in the PLAC1-overexpressing cells, accompanied by increased expression of the mesenchymal markers, Vimentin, MMP2 and Slug, and the opposite effects were observed in PLAC1-silenced cells. Taken together, the present results indicated that high expression of PLAC1 was associated with poor survival and PLAC1 promoted metastasis via EMT in CC.
Collapse
Affiliation(s)
- Rujun Chen
- Department of Gynaecology and Obstetrics, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, P.R. China
| | - Chan Sheng
- Department of Gynaecology and Obstetrics, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, P.R. China
| | - Ruyue Ma
- Department of Gynaecology and Obstetrics, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, P.R. China
| | - Liwen Zhang
- Department of Gynaecology and Obstetrics, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, P.R. China
| | - Lina Yang
- Department of Gynaecology and Obstetrics, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, P.R. China
| | - Yaping Chen
- Department of Gynaecology and Obstetrics, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, P.R. China
| |
Collapse
|
11
|
Mutated p53 in HGSC-From a Common Mutation to a Target for Therapy. Cancers (Basel) 2021; 13:cancers13143465. [PMID: 34298679 PMCID: PMC8304959 DOI: 10.3390/cancers13143465] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Ovarian high-grade serous cancer (HGSC), the most common and the deadliest subtype of epithelial ovarian cancer, is characterized by frequent mutations in the TP53 tumor suppressor gene, encoding for the p53 protein in nearly 100% of cases. This makes p53 the focus of many studies trying to understand its role in HGSC. The aim of our review paper is to provide updates on the latest findings related to the role of mutant p53 in HGSC. This includes the clinical outcomes of TP53 mutations in HGSC, upstream regulators and downstream effectors of p53, its function in the earliest stages of HGSC development and in the interplay between the tumor cells and their microenvironment. We summarize with the likelihood of p53 mutants to serve as biomarkers for early diagnosis and as targets for therapy in HGSC. Abstract Mutations in tumor suppressor gene TP53, encoding for the p53 protein, are the most ubiquitous genetic variation in human ovarian HGSC, the most prevalent and lethal histologic subtype of epithelial ovarian cancer (EOC). The majority of TP53 mutations are missense mutations, leading to loss of tumor suppressive function of p53 and gain of new oncogenic functions. This review presents the clinical relevance of TP53 mutations in HGSC, elaborating on several recently identified upstream regulators of mutant p53 that control its expression and downstream target genes that mediate its roles in the disease. TP53 mutations are the earliest genetic alterations during HGSC pathogenesis, and we summarize current information related to p53 function in the pathogenesis of HGSC. The role of p53 is cell autonomous, and in the interaction between cancer cells and its microenvironment. We discuss the reduction in p53 expression levels in tumor associated fibroblasts that promotes cancer progression, and the role of mutated p53 in the interaction between the tumor and its microenvironment. Lastly, we discuss the potential of TP53 mutations to serve as diagnostic biomarkers and detail some more advanced efforts to use mutated p53 as a therapeutic target in HGSC.
Collapse
|
12
|
Chen Y, Stagg C, Schlessinger D, Nagaraja R. PLAC1 affects cell to cell communication by interacting with the desmosome complex. Placenta 2021; 110:39-45. [PMID: 34118612 DOI: 10.1016/j.placenta.2021.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 06/01/2021] [Indexed: 11/16/2022]
Abstract
INTRODUCTION X-linked PLAC1 is highly expressed in placenta during embryogenesis, and when ablated in mice, causes aberrant placental cell layer organization. It is also highly expressed in many types of cancer cell-lines. Although it has been shown that it promotes AKT phosphorylation in cancer cells, the exact mechanism by which it influences placental layer differentiation is unclear. METHODS To investigate the mechanism of action of PLAC1 we did cell fractionation and immunoprecipitation of the protein and Mass Spectrometry analysis to identify its interaction partners. The associated proteins were directly tested for interactions by co-transfection with PLAC1 and immunoprecipitation. Mutations in the ZP-N domain of PLAC1 were introduced to assess its involvement in the interactions. RESULTS We provide evidence that Desmoglein-2 (DSG2), a component of the membrane-associated desmosomal complex, directly interacts with PLAC1. Mutations of cysteines in ZP-N domain disrupt the interaction between PLAC1 and DSG-2. DISCUSSION Because desmosomes are responsible for establishing lateral cell-cell junctions, we suggest that direct interaction with the lateral junction protein complex may be implicated in the PLAC1 effects on cell-cell interactions, and thereby on the layer structure of the placenta.
Collapse
Affiliation(s)
- Yaohui Chen
- Laboratory of Genetics and Genomics, National Institute on Aging, Baltimore, MD, 21224, USA
| | - Carole Stagg
- Laboratory of Genetics and Genomics, National Institute on Aging, Baltimore, MD, 21224, USA
| | - David Schlessinger
- Laboratory of Genetics and Genomics, National Institute on Aging, Baltimore, MD, 21224, USA
| | - Ramaiah Nagaraja
- Laboratory of Genetics and Genomics, National Institute on Aging, Baltimore, MD, 21224, USA.
| |
Collapse
|
13
|
Ma J, Li L, Du J, Pan C, Zhang C, Chen Y. Placenta-specific protein 1 enhances liver metastatic potential and is associated with the PI3K/AKT/NF-κB signaling pathway in colorectal cancer. Eur J Cancer Prev 2021; 30:220-231. [PMID: 32701605 PMCID: PMC8011505 DOI: 10.1097/cej.0000000000000611] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 05/25/2020] [Indexed: 11/26/2022]
Abstract
To better explore the underlying mechanism of liver metastatic formation by placenta-specific protein 1 (PLAC1) in human colorectal cancer, we investigated the proliferation, invasion and angiogenic capabilities of human colorectal cancer cells with different liver metastatic potentials as well as the mechanism of action of PLAC1 in the metastatic process. The expression of PLAC1 was detected by reverse transcriptase PCR, western blot, and real-time PCR. The effect of PLAC1 on metastatic potential was determined by proliferation, invasion, and angiogenesis assays, including an in-vitro coculture system consisting of cancer cells and vascular endothelial cells that were used to detect the relationship between cancer cells and angiogenesis. In addition, we also determined PLAC1 downstream targets that preferentially contribute to the metastatic process. PLAC1 was expressed in HT-29, WiDr, and CaCo-2 colorectal cancer cells but not in Colo320 colorectal cancer cells. PLAC1 not only enhanced significantly the proliferation of CoLo320 and human umbilical vein endothelial cells (HUVECs) but also promoted the invasion of CoLo320 cells. The angiogenesis of HUVECs was enhanced by PLAC1 in a dose-dependent manner. In cocultured systems, angiogenesis was significantly increased by coculture with HT-29 cells. In addition, PLAC1 could promote angiogenesis in coculture with HT-29 cells. Furthermore, PLAC1-enhanced metastatic potential of colorectal cancer cells was dependent on the activation of the PI3K/Akt/NF-κB pathway. The activation of PI3K/Akt/NF-κB signaling by PLAC1 may be critical for metastasis of colorectal cancer cells. According to our results, we suggest that modification of PLAC1 function might be a promising new therapeutic approach to inhibit the aggressive spread of colorectal cancer.
Collapse
Affiliation(s)
- Jiachi Ma
- Department of Oncological Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, P.R. China
| | - Lei Li
- Department of Oncological Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, P.R. China
| | - Jun Du
- Department of Oncological Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, P.R. China
| | - Chengwu Pan
- Department of Oncological Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, P.R. China
| | - Chensong Zhang
- Department of Oncological Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, P.R. China
| | - Yuzhong Chen
- Department of Oncological Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, P.R. China
| |
Collapse
|
14
|
Lin C, Qian P, Zhang Y, Liu Z, Dai K, Sun D. Plac1 promotes nasopharyngeal carcinoma cells proliferation, migration and invasion via Furin/NICD/PTEN pathway. Tissue Cell 2021; 69:101480. [PMID: 33418237 DOI: 10.1016/j.tice.2020.101480] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 01/17/2023]
Abstract
Placenta-specific protein 1 (Plac1) has critical functions in multiple human malignancies, but its role in nasopharyngeal carcinoma (NPC) was unclear. Clinical samples of NPC and adjacent normal tissue were collected. Plac1 expressions in both tissues and cells were measured. After cell transfection, NPC cell viability, proliferation, migration and invasion were detected using cell counting kit-8 (CCK-8) assay, colony formation assay, scratch assay and Transwell assay. Relative expressions of Plac1 and proteins related to migration and invasion (E-Cadherin, N-cadherin, Matrix metalloproteinase2 (MMP2), and MMP9), Furin/Notch1 intracellular domain (NICD)/phosphate and tension homology (PTEN) pathway (NICD, PTEN, phosphorylated-Akt (p-Akt), Akt) were quantified by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot as needed. The interaction between Plac1 and Furin, a member of Furin/NICD/PTEN Pathway, was analyzed using co-Immunoprecipitation (co-IP) assay. Plac1 expression was upregulated in both NPC tissue and cells. Overexpressed Plac1 promoted Plac1 and Furin expressions and increased cell viability, proliferation, migration and invasion of NPC cells, while silencing Plac1 showed the opposite effects. Plac1 interacted with Furin, overexpression of Furin reversed the inhibitory effects of silencing Plac1 on NPC cell proliferation, migration, and invasion, and also reversed the effects of silencing Plac1 on Furin/NICD/PTEN pathway-, cell migration-, and invasion-related protein expressions. Plac1 promoted NPC cell proliferation, migration and invasion via Furin/NICD/PTEN Pathway. The findings of this study provide a possible therapeutic method for NPC treatment.
Collapse
Affiliation(s)
- Chuanbao Lin
- Otorhinolaryngology Head and Neck Surgery, Weifang Ruiqing Hospital, Weicheng District, Weifang City, Shandong Province, 261021, China
| | - Pengfei Qian
- Department of Pediatrics, W.F. Maternal and Child Health Hospital, Weicheng District, Weifang City, Shandong Province, 261021, China
| | - Yan Zhang
- Department of Pathology, Z.C. Maternal and Child Health Hospital, Zhucheng City, Weifang City, Shandong Province, 262200, China
| | - Zhihui Liu
- Otorhinolaryngology Head and Neck Surgery, Weifang Ruiqing Hospital, Weicheng District, Weifang City, Shandong Province, 261021, China
| | - Kun Dai
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University, School of Medicine, Chongqing Road, Shanghai, 200025, China
| | - Dawei Sun
- Otorhinolaryngology Head and Neck Surgery, Weifang Ruiqing Hospital, Weicheng District, Weifang City, Shandong Province, 261021, China.
| |
Collapse
|
15
|
Hayashi R, Nagato T, Kumai T, Ohara K, Ohara M, Ohkuri T, Hirata-Nozaki Y, Harabuchi S, Kosaka A, Nagata M, Yajima Y, Yasuda S, Oikawa K, Kono M, Kishibe K, Takahara M, Katada A, Hayashi T, Celis E, Harabuchi Y, Kobayashi H. Expression of placenta-specific 1 and its potential for eliciting anti-tumor helper T-cell responses in head and neck squamous cell carcinoma. Oncoimmunology 2020; 10:1856545. [PMID: 33457076 PMCID: PMC7781841 DOI: 10.1080/2162402x.2020.1856545] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Placenta-specific 1 (PLAC1) is expressed primarily in placental trophoblasts but not in normal tissues and is a targetable candidate for cancer immunotherapy because it is a cancer testis antigen known to be up-regulated in various tumors. Although peptide epitopes capable of stimulating CD8 T cells have been previously described, there have been no reports of PLAC1 CD4 helper T lymphocyte (HTL) epitopes and the expression of this antigen in head and neck squamous cell carcinoma (HNSCC). Here, we show that PLAC1 is highly expressed in 74.5% of oropharyngeal and 51.9% of oral cavity tumors from HNSCC patients and in several HNSCC established cell lines. We also identified an HTL peptide epitope (PLAC131-50) capable of eliciting effective antigen-specific and tumor-reactive T cell responses. Notably, this peptide behaves as a promiscuous epitope capable of stimulating T cells in the context of more than one human leukocyte antigen (HLA)-DR allele and induces PLAC1-specific CD4 T cells that kill PLAC1-positive HNSCC cell lines in an HLA-DR-restricted manner. Furthermore, T-cells reactive to PLAC131-50 peptide were detected in the peripheral blood of HNSCC patients. These findings suggest that PLAC1 represents a potential target antigen for HTL based immunotherapy in HNSCC.
Collapse
Affiliation(s)
- Ryusuke Hayashi
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan.,Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Toshihiro Nagato
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Takumi Kumai
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa, Japan.,Department of Innovative Research for Diagnosis and Treatment of Head and Neck Cancer, Asahikawa Medical University, Asahikawa, Japan
| | - Kenzo Ohara
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Mizuho Ohara
- Department of Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Takayuki Ohkuri
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Yui Hirata-Nozaki
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Shohei Harabuchi
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Akemi Kosaka
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Marino Nagata
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Yuki Yajima
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan.,Department of Oral and Maxillofacial Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Syunsuke Yasuda
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan.,Respiratory and Breast Center, Asahikawa Medical University Hospital, Asahikawa, Japan
| | - Kensuke Oikawa
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Michihisa Kono
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Kan Kishibe
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Miki Takahara
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Akihiro Katada
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Tatsuya Hayashi
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa, Japan.,Department of Innovative Research for Diagnosis and Treatment of Head and Neck Cancer, Asahikawa Medical University, Asahikawa, Japan
| | - Esteban Celis
- Cancer Immunology, Inflammation and Tolerance Program, Augusta University, Georgia Cancer Center, Augusta, GA, USA
| | - Yasuaki Harabuchi
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Hiroya Kobayashi
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| |
Collapse
|
16
|
Mediratta K, El-Sahli S, D’Costa V, Wang L. Current Progresses and Challenges of Immunotherapy in Triple-Negative Breast Cancer. Cancers (Basel) 2020; 12:E3529. [PMID: 33256070 PMCID: PMC7761500 DOI: 10.3390/cancers12123529] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 02/06/2023] Open
Abstract
With improved understanding of the immunogenicity of triple-negative breast cancer (TNBC), immunotherapy has emerged as a promising candidate to treat this lethal disease owing to the lack of specific targets and effective treatments. While immune checkpoint inhibition (ICI) has been effectively used in immunotherapy for several types of solid tumor, monotherapies targeting programmed death 1 (PD-1), its ligand PD-L1, or cytotoxic T lymphocyte-associated protein 4 (CTLA-4) have shown little efficacy for TNBC patients. Over the past few years, various therapeutic candidates have been reviewed, attempting to improve ICI efficacy on TNBC through combinatorial treatment. In this review, we describe the clinical limitations of ICI and illustrate candidates from an immunological, pharmacological, and metabolic perspective that may potentiate therapy to improve the outcomes of TNBC patients.
Collapse
Affiliation(s)
- Karan Mediratta
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (K.M.); (S.E.-S.)
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Sara El-Sahli
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (K.M.); (S.E.-S.)
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Vanessa D’Costa
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (K.M.); (S.E.-S.)
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Lisheng Wang
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (K.M.); (S.E.-S.)
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
17
|
Mahmoudi AR, Ghods R, Rakhshan A, Madjd Z, Bolouri MR, Mahmoudian J, Rahdan S, Shokri MR, Dorafshan S, Shekarabi M, Zarnani AH. Discovery of a potential biomarker for immunotherapy of melanoma: PLAC1 as an emerging target. Immunopharmacol Immunotoxicol 2020; 42:604-613. [PMID: 33106058 DOI: 10.1080/08923973.2020.1837865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
BACKGROUND Melanoma has increased in incidence worldwide prompting investigators to search for new biomarkers for targeted immunotherapy of this disease. Placenta specific 1 (PLAC1) is a new member of cancer-testis antigens with widespread expression in many types of cancer. Here, we aimed to study for the first time the expression pattern of PLAC1 in skin cancer samples including cutaneous melanoma, basal cell carcinoma (BCC), squamous cell carcinoma (SCC) in comparison to normal skin and nevus tissues and potential therapeutic effect of anti-PLAC1 antibody in melanoma cancer cell lines in vitro. MATERIALS AND METHODS Polyclonal and monoclonal antibodies were applied for immunohistochemical profiling of PLAC1 expression using tissue microarray. The cytotoxic action of anti-PLAC1 antibody alone or as an antibody drug conjugate (with anti-neoplastic agent SN38) was investigated in melanoma cell lines. RESULTS We observed that 100% (39 of 39) of melanoma tissues highly expressed PLAC1 with both cytoplasmic and surface expression pattern. Investigation of PLAC1 expression in BCC (n = 110) samples showed negative results. Cancer cells in SCC samples (n = 66) showed very weak staining. Normal skin tissues and nevus samples including congenital melanocytic nevus failed to express PLAC1. Anti-PLAC1-SN38 exerted a specific pattern of cytotoxicity in a dose- and time-dependent manner in melanoma cells expressing surface PLAC1. CONCLUSIONS Our findings re-inforce the concept of re-expression of embryonic/placental tissue antigens in cancer and highlight the possibility of melanoma targeted therapy by employing anti-PLAC1 antibodies. The data presented here should lead to the future research on targeted immunotherapy of patients with melanoma.
Collapse
Affiliation(s)
- Ahmad-Reza Mahmoudi
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran.,Reproductive Immunology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Roya Ghods
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Azadeh Rakhshan
- Department of Pathology, Shohada-e-Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad-Reza Bolouri
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Jafar Mahmoudian
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Shaghayegh Rahdan
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad-Reza Shokri
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Shima Dorafshan
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mehdi Shekarabi
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amir-Hassan Zarnani
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran.,Reproductive Immunology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran.,Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
18
|
High expression of PLAC1 in colon cancer as a predictor of poor prognosis: A study based on TCGA data. Gene 2020; 763:145072. [PMID: 32827679 DOI: 10.1016/j.gene.2020.145072] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/25/2020] [Accepted: 08/17/2020] [Indexed: 01/19/2023]
Abstract
Colon cancer is one of the most common diseases in the world with both a high incidence and high mortality. PLAC1 is activated and expressed in many cancers. We aim to explore the relationship between PLAC1 expression and prognosis in colon cancer patient. The RNA-Seq expression data and clinical information of colon cancer were downloaded from The Cancer Genome Atlas (TCGA) database. Differentially expressed PLAC1 was obtained by the Wilcoxon signed-rank test; the significance difference being that PLAC1 was more highly expressed in tumor rather than normal tissue (p < 0.01). Then patients were classified into high and low risk groups by different risk scores, and the Kaplan-Meier survival analysis showed that colon cancer patients with a high PLAC1 expression had a poorer prognosis than low PLAC1 expression patients (p = 0.0031). Next, in analyzing the clinical pathology associated with PLAC1 expression, logistic regression showed that PLAC1 was expressed high in stage (OR = 4.11 for I vs. IV), lymph nodes (OR = 1.73 for N0 vs. N1+), distant metastasis (OR = 2.8 for M0 vs. M1), and status (OR = 22.81 for normal vs. tumor). Univariate and multivariate cox analyses were employed to identify that PLAC1 could be regarded as an independent prognostic factor. Univariate cox analysis showed PLAC1 had a correlation to overall survival (OS) (HR: 0.46, 95% CI: 0.28-0.77, p = 0.003). Multivariate cox analysis revealed that PLAC1 (HR: 0.51, 95% CI: 0.30-0.86, p = 0.012) could be regarded as an Independent prognostic factor. We also used quantitative real-time polymerase chain reaction (qRT-PCR) to test if PLAC1 was differently expressed in cell lines. The qRT-PCR obtained the significant results that PLAC1 expressed high in colon cancer cell lines (p < 0.05). Finally, Gene Set Enrichment Analysis (GSEA) was utilized to show 14 enriched signaling pathways. Our study discovered that high expression of PLAC1 predicts poor prognosis in colon cancer patients, providing a new biomarker, which can assist physicians in finding new diagnostic and therapy methods for colon cancer.
Collapse
|
19
|
Roldán DB, Grimmler M, Hartmann C, Hubich-Rau S, Beißert T, Paret C, Cagna G, Rohde C, Wöll S, Koslowski M, Türeci Ö, Sahin U. PLAC1 is essential for FGF7/FGFRIIIb-induced Akt-mediated cancer cell proliferation. Oncotarget 2020; 11:1862-1875. [PMID: 32499871 PMCID: PMC7244013 DOI: 10.18632/oncotarget.27582] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 03/14/2020] [Indexed: 12/20/2022] Open
Abstract
PLAC1 (placenta enriched 1) is a mammalian trophoblast-specific protein. Aberrant expression of PLAC1 is observed in various human cancers, where it is involved in the motility, migration, and invasion of tumor cells, which are associated with the phosphoinositide 3-kinase (PI3K)/AKT pathway. We previously demonstrated that AKT activation mediates the downstream effects of PLAC1; however, the molecular mechanisms of PLAC1-induced AKT-mediated tumor-related processes are unclear. We studied human choriocarcinoma and breast cancer cell lines to explore the localization and receptor-ligand interactions, as well as the downstream effects of PLAC1. We show secretion and adherence of PLAC1 to the extracellular matrix, where it forms a trimeric complex with fibroblast growth factor 7 (FGF7) and its receptor, FGF receptor 2 IIIb (FGFR2IIIb). We further show that PLAC1 signaling via FGFR2IIIb activates AKT phosphorylation in cancer cell lines. As the FGF pathway is of major interest in anticancer therapeutic strategies, these data further promote PLAC1 as a promising anticancer drug target.
Collapse
Affiliation(s)
- Diana Barea Roldán
- TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- These authors contributed equally to this work
| | - Matthias Grimmler
- Formerly of TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Current address: DiaSys Diagnostic Systems GmbH, Holzheim, Germany
- These authors contributed equally to this work
| | - Christoph Hartmann
- Formerly of TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Current address: Merck KGaA, Darmstadt, Germany
- These authors contributed equally to this work
| | - Stefanie Hubich-Rau
- TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- These authors contributed equally to this work
| | - Tim Beißert
- TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Claudia Paret
- Formerly of TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Giuseppe Cagna
- Formerly of Ganymed Pharmaceuticals AG, Mainz, Germany
- Current address: Lonza Pharma & Biotech, Cologne, Germany
| | - Christoph Rohde
- Formerly of Ganymed Pharmaceuticals AG, Mainz, Germany
- Current address: Merck KGaA, Darmstadt, Germany
| | - Stefan Wöll
- Formerly of Ganymed Pharmaceuticals AG, Mainz, Germany
- Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, Germany
| | - Michael Koslowski
- Formerly of TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Formerly of Ganymed Pharmaceuticals AG, Mainz, Germany
- Formerly of University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Current address: GammaDelta Therapeutics, London, United Kingdom
| | - Özlem Türeci
- Formerly of TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Formerly of Ganymed Pharmaceuticals AG, Mainz, Germany
- Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, Germany
- Ci3 Cluster for Individualized Immune Intervention, Mainz, Germany
| | - Ugur Sahin
- TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, Germany
- University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
20
|
Yilmaz N, Timur H, Ugurlu EN, Yilmaz S, Ozgu-Erdinc AS, Erkilinc S, Inal HA. Placenta specific protein-1 in recurrent pregnancy loss and in In Vitro Fertilisation failure: a prospective observational case-control study. J OBSTET GYNAECOL 2019; 40:843-848. [PMID: 31791163 DOI: 10.1080/01443615.2019.1674263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Observations from studies have provided evidence that Placenta-specific protein1 (PLAC1) is important for the establishment and maintenance of pregnancy and suggest it as a potential biomarker for gestational pathologies. The aim of this study is to investigate whether maternal serum PLAC1 levels have any impact on etiopathogenesis of recurrent pregnancy loss (RPL) and repeated implantation failure after In Vitro Fertilisation (RIF). We conducted a prospective observational case-control study in a Research Hospital. Twenty-eight patients with RPL (group 1), 30 patients with unexplained infertility and RIF (group 2), 29 fertile patients (group 3) were included. The demographic features and serum PLAC1 levels were compared. There was a significant difference in PLAC1 levels between the groups (group 1 = 19.71 + 16.55 ng/ml; group 2 = 4.82 + 1.44 ng/ml; group 3 = 0.89 + 0.62 ng/ml, respectively) (p=.001). Positive correlation was found between serum PLAC1 levels and abortion rates (r = 0.64; p=.001), a negative correlation was found between serum PLAC1 levels and live birth rates (r = -0.69; p=.001). PLAC1 might have a negative effect on implantation in RPL and RIF. There may be a subgroup of PLAC with different bioactivity. There are no relevant studies conducted among these populations, further large-scale studies are needed to assess the molecular role of PLAC1 on implantation.IMPACT STATEMENTWhat is already known about this subject? PLAC1 (placenta-specific protein-1) gene is located on the X chromosome which encodes for a protein that is thought to be important for placental development although its role has not been clearly defined. Studies in the literature have provided evidence that PLAC1 has an important role in the establishment and maintenance of pregnancy and suggest it as a potential biomarker for gestational pathologies. Several reports over the past few years have demonstrated PLAC1 expression in a variety of human tumours including lung cancers, breast cancer, hepatocellular and colorectal cancers, gastric cancers and uterine cancers.What do the results of this study add? There have been no previous studies conducted among patients with recurrent pregnancy loss (RPL) or repeated implantation failure after In Vitro Fertilisation (RIF) that have searched for any association between PLAC1 levels and implantation failure. This study has demonstrated higher PLAC1 levels in infertile women with RIF and RPL for the first time; suggesting that it could have a negative effect on implantation in these populations. PLAC1 could be detected in the serum as a biomarker that is associated with RIF and RPL. What are the implications of these findings for clinical practice and/or further research? Defining the precise role of PLAC1 during implantation will provide new insight into understanding of poor reproductive outcomes such as RIF and RPL and help in developing treatment strategies. Further large-scale studies with more patients are needed to uncover the clinical value of PLAC1 as a biomarker to predict repeated implantation failure and RPL.
Collapse
Affiliation(s)
- Nafiye Yilmaz
- Dr. Zekai Tahir Burak Women's Health Research and Education Hospital, Ankara, Turkey
| | - Hakan Timur
- Dr. Zekai Tahir Burak Women's Health Research and Education Hospital, Ankara, Turkey
| | - Evin Nil Ugurlu
- Dr. Zekai Tahir Burak Women's Health Research and Education Hospital, Ankara, Turkey
| | - Saynur Yilmaz
- Dr. Zekai Tahir Burak Women's Health Research and Education Hospital, Ankara, Turkey
| | - A Seval Ozgu-Erdinc
- Dr. Zekai Tahir Burak Women's Health Research and Education Hospital, Ankara, Turkey
| | - Selcuk Erkilinc
- Dr. Zekai Tahir Burak Women's Health Research and Education Hospital, Ankara, Turkey
| | - Hasan Ali Inal
- Dr. Zekai Tahir Burak Women's Health Research and Education Hospital, Ankara, Turkey
| |
Collapse
|
21
|
Taheri-Anganeh M, Amiri A, Movahedpour A, Khatami SH, Ghasemi Y, Savardashtaki A, Mostafavi-Pour Z. In silico Evaluation of PLAC1-fliC As a Chimeric Vaccine against Breast Cancer. IRANIAN BIOMEDICAL JOURNAL 2019. [PMID: 31952435 PMCID: PMC7275624 DOI: 10.29252/ibj.24.3.173] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Background Breast cancer is one of the most prevalent cancers among women. Common cancer treatment methods are not effective enough, and there is a need for a more efficient treatment procedure. Cancer vaccine is a novel immunotherapy method that stimulates humoral and/or cellular immunity against cancer. Placenta-specific protein 1 (PLAC1) is a cancer/testis antigen, prevalent in breast cancer and rarely found in normal tissues. FliC, as a bacterial adjuvant, when fused to PLAC1 can elicit humoral and cellular responses. Therefore, PLAC1-fliC is a chimeric protein, which can be considered a suitable candidate against breast cancer. Methods ProtParam was used to evaluate the physicochemical properties of PLAC1-fliC. Second structures were determined using the GOR V server. PLAC1-fliC 3D structure was modeled by Phyre2, and it was refined using GalaxyWEB. The refined model was submitted to RAMPAGE, PROCHECK, and ProSA-web for validation. Antigenicity and allergenicity of the construct were predicted by ANTIGENpro, VaxiJen, AllergenFP, and SDAP databases. Then MHC-I- and MHC-II-binding epitopes of PLAC1-fliC were forecasted by NetMHC 4.0 and NetMHCII 2.3 Servers. Finally, Ellipro and CTLpred were employed to predict B-cell and cytotoxic T lymphocyte epitopes. Results The construct was evaluated as a stable fusion protein, which could be antigenic and could stimulate B and T cells against breast cancer. Conclusion PLAC1-fliC, as a cancer vaccine candidate, might be suitable and specific for breast cancer, which could evoke humoral and cellular immunity against this type of tumor.
Collapse
Affiliation(s)
- Mortaza Taheri-Anganeh
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Amiri
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Movahedpour
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyyed Hossein Khatami
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Younes Ghasemi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.,harmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.,harmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zohreh Mostafavi-Pour
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.,Maternal-Fetal Medicine Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
22
|
GWAS for Meat and Carcass Traits Using Imputed Sequence Level Genotypes in Pooled F2-Designs in Pigs. G3-GENES GENOMES GENETICS 2019; 9:2823-2834. [PMID: 31296617 PMCID: PMC6723123 DOI: 10.1534/g3.119.400452] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In order to gain insight into the genetic architecture of economically important traits in pigs and to derive suitable genetic markers to improve these traits in breeding programs, many studies have been conducted to map quantitative trait loci. Shortcomings of these studies were low mapping resolution, large confidence intervals for quantitative trait loci-positions and large linkage disequilibrium blocks. Here, we overcome these shortcomings by pooling four large F2 designs to produce smaller linkage disequilibrium blocks and by resequencing the founder generation at high coverage and the F1 generation at low coverage for subsequent imputation of the F2 generation to whole genome sequencing marker density. This lead to the discovery of more than 32 million variants, 8 million of which have not been previously reported. The pooling of the four F2 designs enabled us to perform a joint genome-wide association study, which lead to the identification of numerous significantly associated variant clusters on chromosomes 1, 2, 4, 7, 17 and 18 for the growth and carcass traits average daily gain, back fat thickness, meat fat ratio, and carcass length. We could not only confirm previously reported, but also discovered new quantitative trait loci. As a result, several new candidate genes are discussed, among them BMP2 (bone morphogenetic protein 2), which we recently discovered in a related study. Variant effect prediction revealed that 15 high impact variants for the traits back fat thickness, meat fat ratio and carcass length were among the statistically significantly associated variants.
Collapse
|
23
|
Mahmoudian J, Ghods R, Nazari M, Jeddi-Tehrani M, Ghahremani MH, Ghaffari-Tabrizi-Wizsy N, Ostad SN, Zarnani AH. PLAC1: biology and potential application in cancer immunotherapy. Cancer Immunol Immunother 2019; 68:1039-1058. [PMID: 31165204 PMCID: PMC11028298 DOI: 10.1007/s00262-019-02350-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 05/24/2019] [Indexed: 12/29/2022]
Abstract
The emergence of immunotherapy has revolutionized medical oncology with unprecedented advances in cancer treatment over the past two decades. However, a major obstacle in cancer immunotherapy is identifying appropriate tumor-specific antigens to make targeted therapy achievable with fewer normal cells being impaired. The similarity between placentation and tumor development and growth has inspired many investigators to discover antigens for effective immunotherapy of cancers. Placenta-specific 1 (PLAC1) is one of the recently discovered placental antigens with limited normal tissue expression and fundamental roles in placental function and development. There is a growing body of evidence showing that PLAC1 is frequently activated in a wide variety of cancer types and promotes cancer progression. Based on the restricted expression of PLAC1 in testis, placenta and a wide variety of cancers, we have designated this molecule with new terminology, cancer-testis-placenta (CTP) antigen, a feature that PLAC1 shares with many other cancer testis antigens. Recent reports from our lab provide compelling evidence on the preferential expression of PLAC1 in prostate cancer and its potential utility in prostate cancer immunotherapy. PLAC1 may be regarded as a potential CTP antigen for targeted cancer immunotherapy based on the available data on its promoting function in cancer development and also its expression in cancers of different histological origin. In this review, we will summarize current data on PLAC1 with emphasis on its association with cancer development and immunotherapy.
Collapse
Affiliation(s)
- Jafar Mahmoudian
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Roya Ghods
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahboobeh Nazari
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Mahmood Jeddi-Tehrani
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Mohammad Hossein Ghahremani
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Pharmacology Building, Enghelab St., Tehran, 1417614411, Iran
| | | | - Seyed Nasser Ostad
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Pharmacology Building, Enghelab St., Tehran, 1417614411, Iran.
| | - Amir-Hassan Zarnani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Nafisi Building, Enghelab St., Tehran, 1417613151, Iran.
- Reproductive Immunology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran.
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
24
|
Chi Soh JE, Abu N, Jamal R. The potential immune-eliciting cancer testis antigens in colorectal cancer. Immunotherapy 2018; 10:1093-1104. [DOI: 10.2217/imt-2018-0044] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The identification of cancer testis antigens (CTAs) has been an important finding in the search of potential targets for cancer immunotherapy. CTA is one of the subfamilies of the large tumor-associated antigens groups. It is aberrantly expressed in various types of human tumors but is absent in normal tissues except for the testis and placenta. This CTAs-restricted pattern of expression in human malignancies together with its potential immunogenic properties, has stirred the interest of many researchers to use CTAs as one of the ideal targets in cancer immunotherapy. To date, multiple studies have shown that CTAs-based vaccines can elicit clinical and immunological responses in different tumors, including colorectal cancer (CRC). This review details our current understanding of CTAs and CRC in regard to the expression and immunological responses as well as some of the critical hurdles in CTAs-based immunotherapy.
Collapse
Affiliation(s)
- Joanne Ern Chi Soh
- UKM Medical Molecular Biology Institute (UMBI), UKM Medical Center, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Cheras, 56000 Kuala Lumpur, Malaysia
| | - Nadiah Abu
- UKM Medical Molecular Biology Institute (UMBI), UKM Medical Center, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Cheras, 56000 Kuala Lumpur, Malaysia
| | - Rahman Jamal
- UKM Medical Molecular Biology Institute (UMBI), UKM Medical Center, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Cheras, 56000 Kuala Lumpur, Malaysia
| |
Collapse
|
25
|
Mahmoud AM. Cancer testis antigens as immunogenic and oncogenic targets in breast cancer. Immunotherapy 2018; 10:769-778. [PMID: 29926750 PMCID: PMC6462849 DOI: 10.2217/imt-2017-0179] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Accepted: 03/16/2018] [Indexed: 01/07/2023] Open
Abstract
Breast cancer cells frequently express tumor-associated antigens that can elicit immune responses to eradicate cancer. Cancer-testis antigens (CTAs) are a group of tumor-associated antigens that might serve as ideal targets for cancer immunotherapy because of their cancer-restricted expression and robust immunogenicity. Previous clinical studies reported that CTAs are associated with negative hormonal status, aggressive tumor behavior and poor survival. Furthermore, experimental studies have shown the ability of CTAs to induce both cellular and humoral immune responses. They also demonstrated the implication of CTAs in promoting cancer cell growth, inhibiting apoptosis and inducing cancer cell invasion and migration. In the current review, we attempt to address the immunogenic and oncogenic potential of CTAs and their current utilization in therapeutic interventions for breast cancer.
Collapse
Affiliation(s)
- Abeer M Mahmoud
- Department of Physical Therapy, College of Applied Health Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Pathology, South Egypt Cancer Institute, Assiut University, Assiut 71111, Egypt
| |
Collapse
|
26
|
Li Y, Chu J, Li J, Feng W, Yang F, Wang Y, Zhang Y, Sun C, Yang M, Vasilatos SN, Huang Y, Fu Z, Yin Y. Cancer/testis antigen-Plac1 promotes invasion and metastasis of breast cancer through Furin/NICD/PTEN signaling pathway. Mol Oncol 2018; 12:1233-1248. [PMID: 29704427 PMCID: PMC6068355 DOI: 10.1002/1878-0261.12311] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 03/20/2018] [Accepted: 04/04/2018] [Indexed: 12/28/2022] Open
Abstract
Placenta‐specific protein 1 (Plac1) is a cancer/testis antigen that plays a critical role in promoting cancer initiation and progression. However, the clinical significance and mechanism of Plac1 in cancer progression remain elusive. Here, we report that Plac1 is an important oncogenic and prognostic factor, which physically interacts with Furin to drive breast cancer invasion and metastasis. We have shown that Plac1 expression positively correlates with clinical stage, lymph node metastasis, hormone receptor status, and overall patient survival. Overexpression of Plac1 promoted invasion and metastasis of breast cancer cells in vitro and in vivo. Co‐immunoprecipitation and immunofluorescence cell staining assays revealed that interaction of Plac1 and Furin degraded Notch1 and generated Notch1 intracellular domain (NICD) that could inhibit PTEN activity. These findings are consistent with the results of microarray study in MDA‐MB‐231 cells overexpressing Plac1. A rescue study showed that inhibition of Furin and overexpression of PTEN in Plac1 overexpression cells blocked Plac1‐induced tumor cell progression. Taken together, our findings suggest that functional interaction between Plac1 and Furin enhances breast cancer invasion and metastasis and the Furin/NICD/PTEN axis may act as an important therapeutic target for breast cancer treatment.
Collapse
Affiliation(s)
- Yongfei Li
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, China.,Department of Pharmacology and Chemical Biology, UPMC Hillman Cancer Center, University of Pittsburgh, PA, USA
| | - Jiahui Chu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, China
| | - Jun Li
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, China
| | - Wanting Feng
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, China
| | - Fan Yang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, China
| | - Yifan Wang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, China
| | - Yanhong Zhang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, China
| | - Chunxiao Sun
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, China
| | - Mengzhu Yang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, China
| | - Shauna N Vasilatos
- Department of Pharmacology and Chemical Biology, UPMC Hillman Cancer Center, University of Pittsburgh, PA, USA
| | - Yi Huang
- Department of Pharmacology and Chemical Biology, UPMC Hillman Cancer Center, University of Pittsburgh, PA, USA
| | - Ziyi Fu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, China.,Nanjing Maternal and Child Health Medical Institute, Obstetrics and Gynecology Hospital Affiliated of Nanjing Medical University, China
| | - Yongmei Yin
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, China
| |
Collapse
|
27
|
Faramarzi S, Ghafouri-Fard S. Expression analysis of cancer-testis genes in prostate cancer reveals candidates for immunotherapy. Immunotherapy 2018; 9:1019-1034. [PMID: 28971747 DOI: 10.2217/imt-2017-0083] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Prostate cancer is a prevalent disorder among men with a heterogeneous etiological background. Several molecular events and signaling perturbations have been found in this disorder. Among genes whose expressions have been altered during the prostate cancer development are cancer-testis antigens (CTAs). This group of antigens has limited expression in the normal adult tissues but aberrant expression in cancers. This property provides them the possibility to be used as cancer biomarkers and immunotherapeutic targets. Several CTAs have been shown to be immunogenic in prostate cancer patients and some of the have entered clinical trials. Based on the preliminary data obtained from these trials, it is expected that CTA-based therapeutic options are beneficial for at least a subset of prostate cancer patients.
Collapse
Affiliation(s)
- Sepideh Faramarzi
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
28
|
Shi LY, Ma Y, Zhu GY, Liu JW, Zhou CX, Chen LJ, Wang Y, Li RC, Yang ZX, Zhang D. Placenta‐specific 1 regulates oocyte meiosis and fertilization through furin. FASEB J 2018; 32:5483-5494. [DOI: 10.1096/fj.201700922rr] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Li-Ya Shi
- State Key Lab of Reproductive MedicineNanjing Medical UniversityNanjingChina
| | - Yang Ma
- State Key Lab of Reproductive MedicineNanjing Medical UniversityNanjingChina
| | - Gang-Yi Zhu
- State Key Lab of Reproductive MedicineNanjing Medical UniversityNanjingChina
| | - Jin-Wei Liu
- Department of GynecologyZhejiang Provincial People's HospitalHangzhouChina
| | - Chun-Xiang Zhou
- Prenatal Diagnosis Center of Jiangsu ProvinceAffiliated Drum Tower Hospital, Nanjing University Medical SchoolNanjingChina
| | - Liang-Jian Chen
- State Key Lab of Reproductive MedicineNanjing Medical UniversityNanjingChina
| | - Yang Wang
- State Key Lab of Reproductive MedicineNanjing Medical UniversityNanjingChina
| | | | - Zhi-Xia Yang
- State Key Lab of Reproductive MedicineNanjing Medical UniversityNanjingChina
| | - Dong Zhang
- State Key Lab of Reproductive MedicineNanjing Medical UniversityNanjingChina
| |
Collapse
|
29
|
Yuan H, Wang X, Shi C, Jin L, Hu J, Zhang A, Li J, Vijayendra N, Doodala V, Weiss S, Tang Y, Weiner LM, Glazer RI. Plac1 Is a Key Regulator of the Inflammatory Response and Immune Tolerance In Mammary Tumorigenesis. Sci Rep 2018; 8:5717. [PMID: 29632317 PMCID: PMC5890253 DOI: 10.1038/s41598-018-24022-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 03/22/2018] [Indexed: 01/09/2023] Open
Abstract
Plac1 is an X-linked trophoblast gene expressed at high levels in the placenta, but not in adult somatic tissues other than the testis. Plac1 however is re-expressed in several solid tumors and in most human cancer cell lines. To explore the role of Plac1 in cancer progression, Plac1 was reduced by RNA interference in EO771 mammary carcinoma cells. EO771 "knockdown" (KD) resulted in 50% reduction in proliferation in vitro and impaired tumor growth in syngeneic mice; however, tumor growth in SCID mice was equivalent to tumor cells expressing a non-silencing control RNA, suggesting that Plac1 regulated adaptive immunity. Gene expression profiling of Plac1 KD cells indicated reduction in several inflammatory and immune factors, including Cxcl1, Ccl5, Ly6a/Sca-1, Ly6c and Lif. Treatment of mice engrafted with wild-type EO771 cells with a Cxcr2 antagonist impaired tumor growth, reduced myeloid-derived suppressor cells and regulatory T cells, while increasing macrophages, dendritic cells, NK cells and the penetration of CD8+ T cells into the tumor bed. Cxcl1 KD phenocopied the effects of Plac1 KD on tumor growth, and overexpression of Cxcl1 partially rescued Plac1 KD cells. These results reveal that Plac1 modulates a tolerogenic tumor microenvironment in part by modulating the chemokine axis.
Collapse
Affiliation(s)
- Hongyan Yuan
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20007, USA
| | - Xiaoyi Wang
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20007, USA
| | - Chunmei Shi
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20007, USA
| | - Lu Jin
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20007, USA
| | - Jianxia Hu
- Laboratory of Thyroid Diseases, the Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Alston Zhang
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20007, USA
| | - James Li
- Department of Bioinformatics, Biostatistics and Biomathematics, Georgetown University Medical Center, Washington, DC, 20007, USA
| | - Nairuthya Vijayendra
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20007, USA
| | - Venkata Doodala
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20007, USA
| | - Spencer Weiss
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20007, USA
| | - Yong Tang
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20007, USA
| | - Louis M Weiner
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20007, USA
| | - Robert I Glazer
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20007, USA.
| |
Collapse
|
30
|
Sifakis S, Androutsopoulos VP, Pontikaki A, Velegrakis A, Papaioannou GI, Koukoura O, Spandidos DA, Papantoniou N. Placental expression of PAPPA, PAPPA-2 and PLAC-1 in pregnacies is associated with FGR. Mol Med Rep 2018. [PMID: 29532882 PMCID: PMC5928614 DOI: 10.3892/mmr.2018.8721] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Fetal growth restriction (FGR) is a gynecological disorder of varying etiology. In the present study, an expression analysis of pregnancy-associated plasma protein A (PAPPA), pregnancy-associated plasma protein A2 (PAPPA2) and placenta-specific-1 (PLAC-1) was conducted in pregnancies with FGR and control pregnancies. Placental tissues were collected from pregnancies with FGR (n=16) and control pregnancies (n=16) and the expression of the genes of interest was examined by qPCR. The mean expression levels of PAPPA and PAPPA2 were significantly lower (P<0.001) in placental tissues from FGR pregnancies compared with tissues from healthy subjects, whereas the opposite pattern was observed for PLAC-1 (P<0.001). PAPPA and PLAC-1 expression in FGR and control subjects correlated with birth weight (P<0.001). The findings suggest a possible pathophysiological link between the development of FGR and the expression of PAPPA, PAPPA2 and PLAC-1.
Collapse
Affiliation(s)
- Stavros Sifakis
- Department of Obstetrics and Gynecology, University Hospital of Heraklion, 71110 Heraklion, Greece
| | | | - Artemis Pontikaki
- Department of Obstetrics and Gynecology, University Hospital of Heraklion, 71110 Heraklion, Greece
| | - Alexis Velegrakis
- Department of Obstetrics and Gynecology, Venizeleion Hospital, 71409 Heraklion, Greece
| | - George I Papaioannou
- Department of Obstetrics and Gynecology, Attikon University Hospital, University of Athens, 12462 Athens, Greece
| | - Ourania Koukoura
- Department of Obstetrics and Gynecology, University of Thessalia, 41110 Larissa, Greece
| | - Demetrios A Spandidos
- Department of Clinical Virology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Nikos Papantoniou
- Department of Obstetrics and Gynecology, Attikon University Hospital, University of Athens, 12462 Athens, Greece
| |
Collapse
|
31
|
Li Q, Liu M, Wu M, Zhou X, Wang S, Hu Y, Wang Y, He Y, Zeng X, Chen J, Liu Q, Xiao D, Hu X, Liu W. PLAC1-specific TCR-engineered T cells mediate antigen-specific antitumor effects in breast cancer. Oncol Lett 2018; 15:5924-5932. [PMID: 29556312 DOI: 10.3892/ol.2018.8075] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 12/11/2017] [Indexed: 12/30/2022] Open
Abstract
Placenta-specific 1 (PLAC1), a novel cancer-testis antigen (CTA), is expressed in a number of different human malignancies. It is frequently produced in breast cancer, serving a function in tumorigenesis. Adoptive immunotherapy using T cell receptor (TCR)-engineered T cells against CTA mediates objective tumor regression; however, to the best of our knowledge, targeting PLAC1 using engineered T cells has not yet been attempted. In the present study, the cDNAs encoding TCRα- and β-chains specific for human leukocyte antigen (HLA)-A*0201-restricted PLAC1 were cloned from a cytotoxic T-lymphocyte, generated by in vitro by the stimulation of CD8+ T cells using autologous HLA-A2+ dendritic cells loaded with a PLAC1-specific peptide (p28-36, VLCSIDWFM). The TCRα/β-chains were linked by a 2A peptide linker (TCRα-Thosea asigna virus-TCRβ), and the constructs were cloned into the lentiviral vector, followed by transduction into human cytotoxic (CD8+) T cells. The efficiency of transduction was up to 25.16%, as detected by PLAC1 multimers. TCR-transduced CD8+ T cells, co-cultured with human non-metastatic breast cancer MCF-7 cells (PLAC1+, HLA-A2+) and triple-negative breast cancer MDAMB-231 cells (PLAC1+, HLA-A2+), produced interferon γ and tumor necrosis factor α, suggesting TCR activation. Furthermore, the PLAC1 TCR-transduced CD8+ T cells efficiently and specifically identified and annihilated the HLA-A2+/PLAC1+ breast cancer cell lines in a lactate dehydrogenase activity assay. Western blot analysis demonstrated that TCR transduction stimulated the production of mitogen-activated protein kinase signaling molecules, extracellular signal-regulated kinases 1/2 and nuclear factor-κB, through phosphoinositide 3-kinase γ-mediated phosphorylation of protein kinase B in CD8+ T cells. Xenograft mouse assays revealed that PLAC1 TCR-transduced CD8+T cells significantly delayed the tumor progression in mice-bearing breast cancer compared with normal saline or negative control-transduced groups. In conclusion, a novel HLA-A2-restricted and PLAC1-specific TCR was identified. The present study demonstrated PLAC1 to be a potential target for breast cancer treatment; and the usage of PLAC1-specific TCR-engineered T cells may be a novel strategy for PLAC1-positive breast cancer treatment.
Collapse
Affiliation(s)
- Qiongshu Li
- Shenzhen Beike Cell Engineering Research Institute, Shenzhen, Guangdong 518057, P.R. China.,Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Muyun Liu
- Shenzhen Beike Cell Engineering Research Institute, Shenzhen, Guangdong 518057, P.R. China
| | - Man Wu
- Shenzhen Beike Cell Engineering Research Institute, Shenzhen, Guangdong 518057, P.R. China
| | - Xin Zhou
- Shenzhen Beike Cell Engineering Research Institute, Shenzhen, Guangdong 518057, P.R. China
| | - Shaobin Wang
- Interventional and Minimally Invasive Oncology Therapy Department, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Yuan Hu
- Shenzhen Beike Cell Engineering Research Institute, Shenzhen, Guangdong 518057, P.R. China
| | - Youfu Wang
- Shenzhen Beike Cell Engineering Research Institute, Shenzhen, Guangdong 518057, P.R. China
| | - Yixin He
- Shenzhen Beike Cell Engineering Research Institute, Shenzhen, Guangdong 518057, P.R. China
| | - Xiaoping Zeng
- Shenzhen Beike Cell Engineering Research Institute, Shenzhen, Guangdong 518057, P.R. China
| | - Junhui Chen
- Interventional and Minimally Invasive Oncology Therapy Department, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Qubo Liu
- Shenzhen Beike Cell Engineering Research Institute, Shenzhen, Guangdong 518057, P.R. China
| | - Dong Xiao
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Xiang Hu
- Shenzhen Beike Cell Engineering Research Institute, Shenzhen, Guangdong 518057, P.R. China
| | - Weibin Liu
- Shenzhen Beike Cell Engineering Research Institute, Shenzhen, Guangdong 518057, P.R. China
| |
Collapse
|
32
|
Abstract
Placental-specific protein 1 (PLAC1) is an X-linked trophoblast gene that is re-expressed in several malignancies, including breast cancer, and is therefore a potential biomarker to follow disease onset and progression. Sera from 117 preoperative/pretreatment breast cancer patients and 51 control subjects, including those with fibrocystic disease, were analyzed for the presence of PLAC1 protein as well as its expression by IHC in tumor biopsies in a subset of subjects. Serum PLAC1 levels exceeded the mean plus one standard deviation (mean+SD) of the level in control subjects in 67% of subjects with ductal carcinoma in situ (DCIS), 67% with HER2+ tumors, 73% with triple-negative cancer and 73% with ER+/PR+ tumors. Greater sensitivity was achieved using the mean+2 SD of control PLAC1 serum values, where the false positive rate was 3% and was exceeded by 38%, 40%, 60% and 43% of subjects with DCIS, HER2+, TNBC and ER+/PR+/HER2- tumors. PLAC1 was detected in 97% of tumor biopsies, but did not correlate quantitatively with serum levels. There was no significant correlation of serum PLAC1 levels with race, age at diagnosis, body mass index (BMI) or the presence of metastatic disease. It remains to be determined whether PLAC1 serum levels can serve as a diagnostic biomarker for the presence or recurrence of disease post-surgery and/or therapy.
Collapse
|
33
|
Yang L, Zha TQ, He X, Chen L, Zhu Q, Wu WB, Nie FQ, Wang Q, Zang CS, Zhang ML, He J, Li W, Jiang W, Lu KH. Placenta-specific protein 1 promotes cell proliferation and invasion in non-small cell lung cancer. Oncol Rep 2017; 39:53-60. [PMID: 29138842 PMCID: PMC5783604 DOI: 10.3892/or.2017.6086] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 09/26/2017] [Indexed: 01/08/2023] Open
Abstract
Pulmonary carcinoma-associated proteins have emerged as crucial players in governing fundamental biological processes such as cell proliferation, apoptosis and metastasis in human cancers. Placenta-specific protein 1 (PLAC1) is a cancer-related protein, which is activated and upregulated in a variety of malignant tissues, including prostate cancer, gastric adenocarcinoma, colorectal, epithelial ovarian and breast cancer. However, its biological role and clinical significance in non-small cell lung cancer (NSCLC) development and progression are still unknown. In the present study, we found that PLAC1 was significantly upregulated in NSCLC tissues, and its expression level was associated with advanced pathological stage and it was also correlated with shorter progression-free survival of lung cancer patients. Furthermore, knockdown of PLAC1 expression by siRNA inhibited cell proliferation, induced apoptosis and impaired invasive ability in NSCLC cells partly via regulation of epithelial-mesenchymal transition (EMT)-related protein expression. Our findings present that increased PLAC1 could be identified as a negative prognostic biomarker in NSCLC and regulate cell proliferation and invasion. Thus, we conclusively demonstrated that PLAC1 plays a key role in NSCLC development and progression, which may provide novel insights on the function of tumor-related gene-driven tumorigenesis.
Collapse
Affiliation(s)
- Li Yang
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Gulou, Nanjing, Jiangsu 210029, P.R. China
| | - Tian-Qi Zha
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Gulou, Nanjing, Jiangsu 210029, P.R. China
| | - Xiang He
- Department of Digestive, First Affiliated Hospital, Nanjing Medical University, Gulou, Nanjing, Jiangsu 210029, P.R. China
| | - Liang Chen
- Department of Cardiothoracic Surgery, First Affiliated Hospital, Nanjing Medical University, Gulou, Nanjing, Jiangsu 210029, P.R. China
| | - Quan Zhu
- Department of Cardiothoracic Surgery, First Affiliated Hospital, Nanjing Medical University, Gulou, Nanjing, Jiangsu 210029, P.R. China
| | - Wei-Bing Wu
- Department of Cardiothoracic Surgery, First Affiliated Hospital, Nanjing Medical University, Gulou, Nanjing, Jiangsu 210029, P.R. China
| | - Feng-Qi Nie
- Department of Oncology, Second Affiliated Hospital, Nanjing Medical University, Gulou, Nanjing, Jiangsu 210029, P.R. China
| | - Qian Wang
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Gulou, Nanjing, Jiangsu 210029, P.R. China
| | - Chong-Shuang Zang
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Gulou, Nanjing, Jiangsu 210029, P.R. China
| | - Mei-Ling Zhang
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Gulou, Nanjing, Jiangsu 210029, P.R. China
| | - Jing He
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Gulou, Nanjing, Jiangsu 210029, P.R. China
| | - Wei Li
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Gulou, Nanjing, Jiangsu 210029, P.R. China
| | - Wen Jiang
- Department of Biochemistry, Nanjing Medical University, Gulou, Nanjing, Jiangsu 210029, P.R. China
| | - Kai-Hua Lu
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Gulou, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
34
|
Placenta-specific1 (PLAC1) is a potential target for antibody-drug conjugate-based prostate cancer immunotherapy. Sci Rep 2017; 7:13373. [PMID: 29042604 PMCID: PMC5645454 DOI: 10.1038/s41598-017-13682-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 09/26/2017] [Indexed: 11/13/2022] Open
Abstract
Our recent findings strongly support the idea of PLAC1 being as a potential immunotherapeutic target in prostate cancer (PCa). Here, we have generated and evaluated an anti-placenta-specific1 (PLAC1)-based antibody drug conjugate (ADC) for targeted immunotherapy of PCa. Prostate cancer cells express considerable levels of PLAC1. The Anti-PLAC1 clone, 2H12C12, showed high reactivity with recombinant PLAC1 and selectivity recognized PLAC1 in prostate cancer cells but not in LS180 cells, the negative control. PLAC1 binding induced rapid internalization of the antibody within a few minutes which reached to about 50% after 15 min and almost completed within an hour. After SN38 conjugation to antibody, a drug-antibody ratio (DAR) of about 5.5 was achieved without apparent negative effect on antibody affinity to cell surface antigen. The ADC retained intrinsic antibody activity and showed enhanced and selective cytotoxicity with an IC50 of 62 nM which was about 15-fold lower compared to free drug. Anti-PLAC1-ADC induced apoptosis in human primary prostate cancer cells and prostate cell lines. No apparent cytotoxic effect was observed in in vivo animal safety experiments. Our newly developed anti-PLAC1-based ADCs might pave the way for a reliable, efficient, and novel immunotherapeutic modality for patients with PCa.
Collapse
|
35
|
Guo L, Xu D, Lu Y, Peng J, Jiang L. Detection of circulating tumor cells by reverse transcription‑quantitative polymerase chain reaction and magnetic activated cell sorting in the peripheral blood of patients with hepatocellular carcinoma. Mol Med Rep 2017; 16:5894-5900. [PMID: 28849093 PMCID: PMC5865766 DOI: 10.3892/mmr.2017.7372] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 02/23/2017] [Indexed: 01/02/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most lethal malignancies worldwide. Circulating tumor cells (CTCs) are considered a major cause of recurrence and metastasis in cancer; however, the detection of CTCs is challenging owing to their very low numbers in peripheral blood (around 10 CTCs per 1,000,000 erythrocytes). Cancer‑testis antigens (CTAs) are specific tumor markers for CTCs. The present study aimed to evaluate the sensitivity and specificity of reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) for the detection of nine CTAs as well as placenta‑specific antigen 1 (PLAC1) in peripheral blood mononuclear cell (PBMC) samples collected from 51 patients with HCC. The effectiveness of magnetic‑activated cell sorting (MACS) for tumor‑cell enrichment, through the depletion of CD45+ leukocytes in PBMC samples, was also assessed. Immunocytochemistry along with hematoxylin and eosin staining demonstrated that RT‑qPCR achieved an overall positive detection rate for CTAs and PLAC1 of 70.6%; the highest rates were observed for melanoma‑associated antigen A3 (MAGEA3), synovial sarcoma X breakpoint 1, MAGEA1, NY‑ESO‑1, L antigen 1 and PLAC1. MACS‑detected intact CTCs in PBMCs were confirmed by H&E staining and morphological assessment; 12 out of 19 (63.2%) patients were identified as positive for CTAs. Screening for these five CTAs and PLAC1 by RT‑qPCR may offer a potentially valuable prognostic tool with good sensitivity and specificity in patients with HCC that may be enhanced by MACS.
Collapse
Affiliation(s)
- Limin Guo
- Department of Hepatobiliary Surgery, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, P.R. China
| | - Donghai Xu
- Department of Radiology, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, P.R. China
| | - Yan Lu
- Department of Hepatobiliary Surgery, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, P.R. China
| | - Jirun Peng
- Department of Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, P.R. China
| | - Li Jiang
- Department of Hepatobiliary Surgery, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, P.R. China
| |
Collapse
|
36
|
Yin Y, Zhu X, Huang S, Zheng J, Zhang M, Kong W, Chen Q, Zhang Y, Chen X, Lin K, Ouyang X. Expression and clinical significance of placenta-specific 1 in pancreatic ductal adenocarcinoma. Tumour Biol 2017; 39:1010428317699131. [PMID: 28618924 DOI: 10.1177/1010428317699131] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Yin Yin
- Department of Medical Oncology, Fuzhou Dongfang Hospital, Xiamen University, Fuzhou, China
| | - Xu Zhu
- Department of Hepatobiliary Surgery, Fuzhou Dongfang Hospital, Xiamen University, Fuzhou, China
| | - Shanshan Huang
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jiawei Zheng
- Department of Medical Oncology, Fuzhou General Hospital of Nanjing Military Command, Fuzong Clinical College, Fujian Medical University, Fuzhou, China
| | - Mengyun Zhang
- Department of Medical Oncology, Fuzhou Dongfang Hospital, Xiamen University, Fuzhou, China
| | - Wencui Kong
- Department of Medical Oncology, Fuzhou General Hospital of Nanjing Military Command, Fuzong Clinical College, Fujian Medical University, Fuzhou, China
| | - Qun Chen
- Department of Medical Oncology, Fuzhou General Hospital of Nanjing Military Command, Fuzong Clinical College, Fujian Medical University, Fuzhou, China
| | - Yan Zhang
- Department of Medical Oncology, Fuzhou Dongfang Hospital, Xiamen University, Fuzhou, China
| | - Xiong Chen
- Department of Medical Oncology, Fuzhou Dongfang Hospital, Xiamen University, Fuzhou, China
- Department of Medical Oncology, Fuzhou General Hospital of Nanjing Military Command, Fuzong Clinical College, Fujian Medical University, Fuzhou, China
| | - Kerong Lin
- Department of Gastroenterology, Fuzhou Dongfang Hospital, Xiamen University, Fuzhou, China
| | - Xuenong Ouyang
- Department of Medical Oncology, Fuzhou Dongfang Hospital, Xiamen University, Fuzhou, China
- Department of Medical Oncology, Fuzhou General Hospital of Nanjing Military Command, Fuzong Clinical College, Fujian Medical University, Fuzhou, China
| |
Collapse
|
37
|
Alam SMK, Jasti S, Kshirsagar SK, Tannetta DS, Dragovic RA, Redman CW, Sargent IL, Hodes HC, Nauser TL, Fortes T, Filler AM, Behan K, Martin DR, Fields TA, Petroff BK, Petroff MG. Trophoblast Glycoprotein (TPGB/5T4) in Human Placenta: Expression, Regulation, and Presence in Extracellular Microvesicles and Exosomes. Reprod Sci 2017; 25:185-197. [PMID: 28481180 DOI: 10.1177/1933719117707053] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Many parallels exist between growth and development of the placenta and that of cancer. One parallel is shared expression of antigens that may have functional importance and may be recognized by the immune system. Here, we characterize expression and regulation of one such antigen, Trophoblast glycoprotein (TPGB; also called 5T4), in the placenta across gestation, in placentas of preeclamptic (PE) pregnancies, and in purified microvesicles and exosomes. METHODS Trophoblast glycoprotein expression was analyzed by real-time reverse transcription-polymerase chain reaction (RT-PCR), Western blot, and immunohistochemistry. Regulation of 5T4 in cytotrophoblast cells was examined under either differentiating conditions of epidermal growth factor or under varying oxygen conditions. Microvesicles and exosomes were purified from supernatant of cultured and perfused placentas. RESULTS Trophoblast glycoprotein expression was prominent at the microvillus surface of syncytiotrophoblast and on the extravillous trophoblast cells, with minimal expression in undifferentiated cytotrophoblasts and normal tissues. Trophoblast glycoprotein expression was elevated in malignant tumors. In cytotrophoblasts, 5T4 was induced by in vitro differentiation, and its messenger RNA (mRNA) was increased under conditions of low oxygen. PE placentas expressed higher 5T4 mRNA than matched control placentas. Trophoblast glycoprotein was prominent within shed placental microvesicles and exosomes. CONCLUSION Given the potential functional and known immunological importance of 5T4 in cancer, these studies reveal a class of proteins that may influence placental development and/or sensitize the maternal immune system. In extravillous trophoblasts, 5T4 may function in epithelial-to-mesenchymal transition during placentation. The role of syncytiotrophoblast 5T4 is unknown, but its abundance in shed syncytial vesicles may signify route of sensitization of the maternal immune system.
Collapse
Affiliation(s)
- S M K Alam
- 1 Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA.,2 Department of Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | - S Jasti
- 1 Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - S K Kshirsagar
- 3 Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, USA
| | - D S Tannetta
- 4 Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Oxford, UK
| | - R A Dragovic
- 4 Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Oxford, UK
| | - C W Redman
- 4 Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Oxford, UK
| | - I L Sargent
- 4 Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Oxford, UK
| | - H C Hodes
- 5 Center for Women's Health, Overland Park, KS, USA
| | - T L Nauser
- 5 Center for Women's Health, Overland Park, KS, USA
| | - T Fortes
- 6 Sparrow Hospital, Lansing, MI, USA.,7 College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | - A M Filler
- 6 Sparrow Hospital, Lansing, MI, USA.,7 College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | - K Behan
- 7 College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | | | - T A Fields
- 8 Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - B K Petroff
- 3 Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, USA.,9 Veterinary Diagnostic Laboratory, Michigan State University, East Lansing, MI, USA
| | - M G Petroff
- 1 Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA.,3 Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, USA.,10 Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
38
|
Liso A, Massenzio F, Stracci F. PLAC1 immunization does not induce infertility in mice. Immunotherapy 2017; 9:481-486. [DOI: 10.2217/imt-2017-0019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: Placenta specific 1 (PLAC1) is a protein rarely expressed in normal cells, except it is important for placental development, with a possible role in the establishment of the mother–fetus interface. The gene is also highly active in a wide variety of cancers and therefore, immunization with PLAC1 peptides could possibly be part of future immunotherapeutic strategies. We investigated whether vaccination against PLAC1 could induce infertility. Materials & methods: We inoculated female mice with PLAC1 peptides, put them in mating, measured antibody response (ELISA assay) and checked, in immunohistochemistry, binding of the induced antibodies to the native antigen. Results: We demonstrated that mice consistently develop antibody responses. We also demonstrated that female mice, after being inoculated with the PLAC1 peptide mix, do became pregnant and can give birth to normal infants. Conclusion: PLAC1 antigens as a specific anti-cancer vaccine could induce anti-PLAC1 antibodies which do not necessarily cause infertility.
Collapse
Affiliation(s)
- Arcangelo Liso
- Department of Medicine & Surgery, University of Foggia, viale L. Pinto,1 71122 – Foggia, Italy
| | - Francesca Massenzio
- Department of Medicine & Surgery, University of Foggia, viale L. Pinto,1 71122 – Foggia, Italy
| | - Fabrizio Stracci
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
39
|
Devor EJ, Gonzalez-Bosquet J, Warrier A, Reyes HD, Ibik NV, Schickling BM, Newtson A, Goodheart MJ, Leslie KK. p53 mutation status is a primary determinant of placenta-specific protein 1 expression in serous ovarian cancers. Int J Oncol 2017; 50:1721-1728. [PMID: 28339050 PMCID: PMC5403493 DOI: 10.3892/ijo.2017.3931] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 03/16/2017] [Indexed: 12/04/2022] Open
Abstract
Placenta-specific protein 1 (PLAC1) expression is co-opted in numerous human cancers. As a consequence of PLAC1 expression, tumor cells exhibit enhanced proliferation and invasiveness. This characteristic is associated with increased aggressiveness and worse patient outcomes. Recently, the presence of the tumor suppressor p53 was shown in vitro to inhibit PLAC1 transcription by compromising the P1, or distal/cancer, promoter. We sought to determine if this phenomenon occurs in primary patient tumors as well. Furthermore, we wanted to know if p53 mutation influenced PLAC1 expression as compared with wild-type. We chose to study serous ovarian tumors as they are well known to have a high rate of p53 mutation. We report herein that the phenomenon of PLAC1 transcription repression does occur in serous ovarian carcinomas but only when TP53 is wild-type. We find that mutant or absent p53 protein de-represses PLAC1 transcription. We further propose that the inability of mutant p53 to repress PLAC1 transcription is due to the fact that the altered TP53 protein is unable to occupy a putative p53 binding site in the PLAC1 P1 promoter thus allowing transcription to occur. Finally, we show that PLAC1 transcript number is significantly negatively correlated with patient survival in our samples. Thus, we suggest that characterizing tumors for TP53 mutation status, p53 protein status and PLAC1 transcription could be used to predict likely prognosis and inform treatment options in patients diagnosed with serous ovarian cancer.
Collapse
Affiliation(s)
- Eric J Devor
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine and The University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Jesus Gonzalez-Bosquet
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine and The University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Akshaya Warrier
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine and The University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Henry D Reyes
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine and The University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Nonye V Ibik
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine and The University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Brandon M Schickling
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Andreea Newtson
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine and The University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Michael J Goodheart
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine and The University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Kimberly K Leslie
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine and The University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| |
Collapse
|
40
|
Nazari M, Zarnani AH, Ghods R, Emamzadeh R, Najafzadeh S, Minai-Tehrani A, Mahmoudian J, Yousefi M, Vafaei S, Massahi S, Nejadmoghaddam MR. Optimized protocol for soluble prokaryotic expression, purification and structural analysis of human placenta specific-1(PLAC1). Protein Expr Purif 2017; 133:139-151. [PMID: 28315746 DOI: 10.1016/j.pep.2017.03.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Revised: 03/13/2017] [Accepted: 03/13/2017] [Indexed: 12/17/2022]
Abstract
Placenta specific -1 (PLAC1) has been recently introduced as a small membrane-associated protein mainly involved in placental development. Expression of PLAC1 transcript has been documented in almost one hundred cancer cell lines standing for fourteen distinct cancer types. The presence of two disulfide bridges makes difficult to produce functional recombinant PLAC1 in soluble form with high yield. This limitation also complicates the structural studies of PLAC1, which is important for prediction of its physiological roles. To address this issue, we employed an expression matrix consisting of two expression vectors, five different E. coli hosts and five solubilization conditions to optimize production of full and truncated forms of human PLAC1. The recombinant proteins were then characterized using an anti-PLAC1-specific antibody in Western blotting (WB) and enzyme linked immunosorbent assay (ELISA). Structure of full length protein was also investigated using circular dichroism (CD). We demonstrated the combination of Origami™ and pCold expression vector to yield substantial amount of soluble truncated PLAC1 without further need for solubilization step. Full length PLAC1, however, expressed mostly as inclusion bodies with higher yield in Origami™ and Rosetta2. Among solubilization buffers examined, buffer containing Urea 2 M, pH 12 was found to be more effective. Recombinant proteins exhibited excellent reactivity as detected by ELISA and WB. The secondary structure of full length PLAC1 was considered by CD spectroscopy. Taken together, we introduced here a simple, affordable and efficient expression system for soluble PLAC1 production.
Collapse
Affiliation(s)
- Mahboobeh Nazari
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran; Department of Tissue Engineering, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Amir-Hassan Zarnani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran; Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Roya Ghods
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, IUMS, Tehran, Iran
| | - Rahman Emamzadeh
- Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran
| | - Somayeh Najafzadeh
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Arash Minai-Tehrani
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Jafar Mahmoudian
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran; Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Yousefi
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Sedigheh Vafaei
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Sam Massahi
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Mohammad-Reza Nejadmoghaddam
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran; Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
41
|
Li Y, Li J, Wang Y, Zhang Y, Chu J, Sun C, Fu Z, Huang Y, Zhang H, Yuan H, Yin Y. Roles of cancer/testis antigens (CTAs) in breast cancer. Cancer Lett 2017; 399:64-73. [PMID: 28274891 DOI: 10.1016/j.canlet.2017.02.031] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/22/2017] [Accepted: 02/24/2017] [Indexed: 12/19/2022]
Abstract
Breast cancer is the most common cancer diagnosed and is the second leading cause of cancer death among women in the US. For breast cancer, early diagnosis and efficient therapy remains a significant clinical challenge. Therefore, it is necessary to identify novel tumor associated molecules to target for biomarker development and immunotherapy. In this regard, cancer testis antigens (CTAs) have emerged as a potential clinical biomarker targeting immunotherapy for various malignancies due to the nature of its characteristics. CTAs are a group of tumor associated antigens (TAAs) that display normal expression in immune-privileged organs, but display aberrant expression in several types of cancers, particularly in advanced cancers. Investigation of CTAs for the clinical management of breast malignancies indicates that these TAAs have potential roles as novel biomarkers, with increased specificity and sensitivity compared to those currently used in the clinic. Moreover, TAAs could be therapeutic targets for cancer immunotherapy. This review is an attempt to address the promising CTAs in breast cancer and their possible clinical implications as biomarkers and immunotherapeutic targets with particular focus on challenges and future interventions.
Collapse
Affiliation(s)
- Yongfei Li
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University. Nanjing 210004, China
| | - Jun Li
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University. Nanjing 210004, China
| | - Yifan Wang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University. Nanjing 210004, China
| | - Yanhong Zhang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University. Nanjing 210004, China
| | - Jiahui Chu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University. Nanjing 210004, China
| | - Chunxiao Sun
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University. Nanjing 210004, China
| | - Ziyi Fu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University. Nanjing 210004, China; Nanjing Maternity and Child Health Medical Institute, Affiliated Obstetrics and Gynecology Hospital, Nanjing Medical University, Nanjing 210004, China
| | - Yi Huang
- Department of Pharmacology and Chemical Biology, Magee Women's Research Institute, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Hansheng Zhang
- School of Public Health, University of Maryland, College Park, MD 20742, USA
| | - Hongyan Yuan
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Yongmei Yin
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University. Nanjing 210004, China.
| |
Collapse
|
42
|
Li X, Dong W, Qu X, Zhao H, Wang S, Hao Y, Li Q, Zhu J, Ye M, Xiao W. Molecular dysexpression in gastric cancer revealed by integrated analysis of transcriptome data. Oncol Lett 2017; 13:3177-3185. [PMID: 28521423 DOI: 10.3892/ol.2017.5798] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 11/30/2016] [Indexed: 02/07/2023] Open
Abstract
Gastric cancer (GC) is often diagnosed in the advanced stages and is associated with a poor prognosis. Obtaining an in depth understanding of the molecular mechanisms of GC has lagged behind compared with other cancers. This study aimed to identify candidate biomarkers for GC. An integrated analysis of microarray datasets was performed to identify differentially expressed genes (DEGs) between GC and normal tissues. Gene ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were then performed to identify the functions of the DEGs. Furthermore, a protein-protein interaction (PPI) network of the DEGs was constructed. The expression levels of the DEGs were validated in human GC tissues using reverse transcription-quantitative polymerase chain reaction (RT-qPCR). A set of 689 DEGs were identified in GC tissues, as compared with normal tissues, including 202 upregulated DEGs and 487 downregulated DEGs. The KEGG pathway analysis suggested that various pathways may play important roles in the pathology of GC, including pathways related to protein digestion and absorption, extracellular matrix-receptor interaction, and the metabolism of xenobiotics by cytochrome P450. The PPI network analysis indicated that the significant hub proteins consisted of SPP1, TOP2A and ARPC1B. RT-qPCR validation indicated that the expression levels of the top 10 most significantly dysexpressed genes were consistent with the illustration of the integrated analysis. The present study yielded a reference list of reliable DEGs, which represents a robust pool of candidates for further evaluation of GC pathogenesis and treatment.
Collapse
Affiliation(s)
- Xiaomei Li
- Department of Oncology, The First Affiliated Hospital of PLA General Hospital, Beijing 100048, P.R. China.,Department of Oncology, PLA General Hospital, Beijing 100853, P.R. China
| | - Weiwei Dong
- Department of Oncology, The First Affiliated Hospital of PLA General Hospital, Beijing 100048, P.R. China.,Department of Oncology, PLA General Hospital, Beijing 100853, P.R. China
| | - Xueling Qu
- Department of Oncology, The First Affiliated Hospital of PLA General Hospital, Beijing 100048, P.R. China.,Graduate School Department of Oncology, Medical College of Liaoning, Jinzhou, Liaoning 121001, P.R. China
| | - Huixia Zhao
- Department of Oncology, The First Affiliated Hospital of PLA General Hospital, Beijing 100048, P.R. China
| | - Shuo Wang
- Department of Oncology, The First Affiliated Hospital of PLA General Hospital, Beijing 100048, P.R. China
| | - Yixin Hao
- Department of Oncology, The First Affiliated Hospital of PLA General Hospital, Beijing 100048, P.R. China
| | - Qiuwen Li
- Department of Oncology, The First Affiliated Hospital of PLA General Hospital, Beijing 100048, P.R. China
| | - Jianhua Zhu
- Department of Oncology, The First Affiliated Hospital of PLA General Hospital, Beijing 100048, P.R. China
| | - Min Ye
- Department of Oncology, The First Affiliated Hospital of PLA General Hospital, Beijing 100048, P.R. China
| | - Wenhua Xiao
- Department of Oncology, The First Affiliated Hospital of PLA General Hospital, Beijing 100048, P.R. China
| |
Collapse
|
43
|
PPAR δ as a Metabolic Initiator of Mammary Neoplasia and Immune Tolerance. PPAR Res 2016; 2016:3082340. [PMID: 28077942 PMCID: PMC5203902 DOI: 10.1155/2016/3082340] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 11/03/2016] [Indexed: 12/12/2022] Open
Abstract
PPARδ is a ligand-activated nuclear receptor that regulates the transcription of genes associated with proliferation, metabolism, inflammation, and immunity. Within this transcription factor family, PPARδ is unique in that it initiates oncogenesis in a metabolic and tissue-specific context, especially in mammary epithelium, and can regulate autoimmunity in some tissues. This review discusses its role in these processes and how it ultimately impacts breast cancer.
Collapse
|
44
|
Wu Y, Lin X, Di X, Chen Y, Zhao H, Wang X. Oncogenic function of Plac1 on the proliferation and metastasis in hepatocellular carcinoma cells. Oncol Rep 2016; 37:465-473. [PMID: 27878289 DOI: 10.3892/or.2016.5272] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 10/31/2016] [Indexed: 11/06/2022] Open
Abstract
Placenta-specific protein 1 (Plac1), which is selectively expressed in the placental syncytiotrophoblast in adult normal tissues, plays an essential role in normal placental and embryonic development. Accumulating evidence suggests that enhanced Plac1 expression is closely associated with the progression of human cancer. Whether Plac1 contributes to the pathophysiology of hepatocellular carcinoma (HCC) remains unclear. In the present study, our data revealed that the expression of Plac1 in human HCC tissues was upregulated, which significantly correlated with metastasis of HCC. Knockdown of Plac1 by small interfering RNA (siRNA) in Bel-7402 and HepG2 cells resulted in decreasing tumor cell proliferation and increasing apoptosis, which implied the oncogenic potential of Plac1. Moreover, silencing of Plac1 induced G1 cell cycle arrest through suppression of cyclin D1 and CDK4 expression. Furthermore, depletion of Plac1 repressed epithelial-mesenchymal transition (EMT), with decreased cell migration and invasion, supporting upregulated E-cadherin expression and downregulated vimentin, twist and snail expression that characterize EMT. Further study suggested that decreased Plac1 expression attenuated the phosphorylation of Akt. These findings have uncovered that Plac1 plays a pivotal role in the progression of HCC, and may serve as a novel therapeutic target for HCC.
Collapse
Affiliation(s)
- Yuan Wu
- Department of Microbiology and Immunology, Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China
| | - Xiaocong Lin
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China
| | - Xiaoqing Di
- Department of Pathology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Yonghua Chen
- Department of Pathology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Hanning Zhao
- Department of Microbiology and Immunology, Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China
| | - Xin Wang
- Department of Microbiology and Immunology, Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China
| |
Collapse
|
45
|
Chang WL, Wang H, Cui L, Peng NN, Fan X, Xue LQ, Yang Q. PLAC1 is involved in human trophoblast syncytialization. Reprod Biol 2016; 16:218-224. [PMID: 27692364 DOI: 10.1016/j.repbio.2016.07.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 06/30/2016] [Accepted: 07/04/2016] [Indexed: 12/20/2022]
Abstract
Placenta specific protein 1 (PLAC1) is thought to be important for murine and human placentation because of its abundant expression in placenta; however, the trophoblast subtypes that express PLAC1 at the fetomaternal interface and the major role of PLAC1 in placentation are still unclear. This study investigated the expression pattern of PLAC1 at the human fetomaternal interface and its involvement in trophoblast syncytialization. Localization of PLAC1 at the fetomaternal interface was studied using in situ hybridization (ISH) and immunohistochemistry (IHC) assays. Real time RT-PCR and Western Blot were employed to exhibit the expression pattern of PLAC1 during human spontaneous syncytialization of term primary cytotrophoblast cells (CTBs). Spontaneous syncytialization of a primary term CTBs model transfected with siRNA specific to PLAC1 was used to investigate the role of PLAC1 during human trophoblast syncytialization. The results showed that PLAC1 was mainly expressed in the human villous syncytiotrophoblast (STB) layer throughout gestation, and the expression level of PLAC1 was significantly elevated during human trophoblast syncytialization. Down-regulation of PLAC1 via specific PLAC1 siRNA transfection attenuated spontaneous syncytialization of primary term CTBs (p<0.05) as indicated by cell fusion index and the expression patterns of the corresponding markers. These data demonstrate the facilitative role of PLAC1 in normal human trophoblast syncytialization.
Collapse
Affiliation(s)
- Wen-Lin Chang
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China; State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKUHKUST Medical Center, Shenzhen, China
| | - Huiying Wang
- Beijing Shijitan Hospital, Capital Medical University, Beijing, China; Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Lina Cui
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Graduate School of Chinese Academy of Sciences, Beijing, China
| | - Nan-Ni Peng
- Reproductive Medical Center of Luohu Hospital Shenzhen, Shenzhen, Guangdong, China
| | - Xiujun Fan
- Laboratory for Reproductive Health, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Li-Qun Xue
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China.
| | - Qing Yang
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China.
| |
Collapse
|
46
|
Muys BR, Lorenzi JCC, Zanette DL, Bueno RDBLE, de Araújo LF, Dinarte-Santos AR, Alves CP, Ramão A, de Molfetta GA, Vidal DO, Silva WA. Placenta-Enriched LincRNAs MIR503HG and LINC00629 Decrease Migration and Invasion Potential of JEG-3 Cell Line. PLoS One 2016; 11:e0151560. [PMID: 27023770 PMCID: PMC4833476 DOI: 10.1371/journal.pone.0151560] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 03/01/2016] [Indexed: 12/16/2022] Open
Abstract
LINC00629 and MIR503HG are long intergenic non-coding RNAs (lincRNAs) mapped on chromosome X (Xq26), a region enriched for genes associated with human reproduction. Genes highly expressed in normal reproductive tissues and cancers (CT genes) are well known as potential tumor biomarkers. This study aimed to characterize the structure, expression, function and regulation mechanism of MIR503HG and LINC00629 lincRNAs. According to our data, MIR503HG expression was almost exclusive to placenta and LINC00629 was highly expressed in placenta and other reproductive tissues. Further analysis, using a cancer cell lines panel, showed that MIR503HG and LINC00629 were expressed in 50% and 100% of the cancer cell lines, respectively. MIR503HG was expressed predominantly in the nucleus of JEG-3 choriocarcinoma cells. We observed a positively correlated expression between MIR503HG and LINC00629, and between the lincRNAs and neighboring miRNAs. Also, both LINC00629 and MIR503GH could be negatively regulated by DNA methylation in an indirect way. Additionally, we identified new transcripts for MIR503HG and LINC00629 that are relatively conserved when compared to other primates. Furthermore, we found that overexpression of MIR503HG2 and the three-exon LINC00629 new isoforms decreased invasion and migration potential of JEG-3 tumor cell line. In conclusion, our results suggest that lincRNAs MIR503HG and LINC00629 impaired migration and invasion capacities in a choriocarcinoma in vitro model, indicating a potential role in human reproduction and tumorigenesis. Moreover, the MIR503HG expression pattern found here could indicate a putative new tumor biomarker.
Collapse
Affiliation(s)
- Bruna Rodrigues Muys
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Center for Cell-Based Therapy (CEPID/FAPESP), National institute of Science and Technology in Stem Cell and Cell Therapy (INCTC/CNPq), Regional Blood Center of Ribeirão Preto, Riberão Preto, Brazil
- Center for Medical Genomics (HCFMRP/USP), Center for Integrative Systems Biology (CISBi–NAP/USP), Ribeirão Preto, Brazil
| | - Júlio Cesar Cetrulo Lorenzi
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Center for Cell-Based Therapy (CEPID/FAPESP), National institute of Science and Technology in Stem Cell and Cell Therapy (INCTC/CNPq), Regional Blood Center of Ribeirão Preto, Riberão Preto, Brazil
- Center for Medical Genomics (HCFMRP/USP), Center for Integrative Systems Biology (CISBi–NAP/USP), Ribeirão Preto, Brazil
| | | | - Rafaela de Barros Lima e Bueno
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Center for Cell-Based Therapy (CEPID/FAPESP), National institute of Science and Technology in Stem Cell and Cell Therapy (INCTC/CNPq), Regional Blood Center of Ribeirão Preto, Riberão Preto, Brazil
- Center for Medical Genomics (HCFMRP/USP), Center for Integrative Systems Biology (CISBi–NAP/USP), Ribeirão Preto, Brazil
| | - Luíza Ferreira de Araújo
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Center for Cell-Based Therapy (CEPID/FAPESP), National institute of Science and Technology in Stem Cell and Cell Therapy (INCTC/CNPq), Regional Blood Center of Ribeirão Preto, Riberão Preto, Brazil
- Center for Medical Genomics (HCFMRP/USP), Center for Integrative Systems Biology (CISBi–NAP/USP), Ribeirão Preto, Brazil
| | - Anemari Ramos Dinarte-Santos
- Center for Cell-Based Therapy (CEPID/FAPESP), National institute of Science and Technology in Stem Cell and Cell Therapy (INCTC/CNPq), Regional Blood Center of Ribeirão Preto, Riberão Preto, Brazil
- Center for Medical Genomics (HCFMRP/USP), Center for Integrative Systems Biology (CISBi–NAP/USP), Ribeirão Preto, Brazil
| | - Cleidson Pádua Alves
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Center for Cell-Based Therapy (CEPID/FAPESP), National institute of Science and Technology in Stem Cell and Cell Therapy (INCTC/CNPq), Regional Blood Center of Ribeirão Preto, Riberão Preto, Brazil
- Center for Medical Genomics (HCFMRP/USP), Center for Integrative Systems Biology (CISBi–NAP/USP), Ribeirão Preto, Brazil
| | - Anelisa Ramão
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Center for Cell-Based Therapy (CEPID/FAPESP), National institute of Science and Technology in Stem Cell and Cell Therapy (INCTC/CNPq), Regional Blood Center of Ribeirão Preto, Riberão Preto, Brazil
| | - Greice Andreotti de Molfetta
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Center for Cell-Based Therapy (CEPID/FAPESP), National institute of Science and Technology in Stem Cell and Cell Therapy (INCTC/CNPq), Regional Blood Center of Ribeirão Preto, Riberão Preto, Brazil
- Center for Medical Genomics (HCFMRP/USP), Center for Integrative Systems Biology (CISBi–NAP/USP), Ribeirão Preto, Brazil
| | - Daniel Onofre Vidal
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
| | - Wilson Araújo Silva
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Center for Cell-Based Therapy (CEPID/FAPESP), National institute of Science and Technology in Stem Cell and Cell Therapy (INCTC/CNPq), Regional Blood Center of Ribeirão Preto, Riberão Preto, Brazil
- Center for Medical Genomics (HCFMRP/USP), Center for Integrative Systems Biology (CISBi–NAP/USP), Ribeirão Preto, Brazil
| |
Collapse
|
47
|
Kroener L, Wang ET, Pisarska MD. Predisposing Factors to Abnormal First Trimester Placentation and the Impact on Fetal Outcomes. Semin Reprod Med 2015; 34:27-35. [PMID: 26696276 DOI: 10.1055/s-0035-1570029] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Normal placentation during the first trimester sets the stage for the rest of pregnancy and involves a finely orchestrated cellular and molecular interplay of maternal and fetal tissues. The resulting intrauterine environment plays an important role in fetal programming and the future health of the fetus, and is impacted by multiple genetic and epigenetic factors. Abnormalities in placentation and spiral artery invasion can lead to ischemia, placental disease, and adverse obstetrical outcomes including preeclampsia, intrauterine growth restriction, and placental abruption. Although first trimester placentation is affected by multiple factors, preconception environmental influences such as mode of conception, including assisted reproductive technologies which result in fertilization in vitro and intrauterine influences due to sex differences, are emerging as potential significant factors impacting first trimester placentation.
Collapse
Affiliation(s)
- Lindsay Kroener
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Erica T Wang
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Margareta D Pisarska
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, California
| |
Collapse
|
48
|
Ghods R, Ghahremani MH, Madjd Z, Asgari M, Abolhasani M, Tavasoli S, Mahmoudi AR, Darzi M, Pasalar P, Jeddi-Tehrani M, Zarnani AH. High placenta-specific 1/low prostate-specific antigen expression pattern in high-grade prostate adenocarcinoma. Cancer Immunol Immunother 2014; 63:1319-27. [PMID: 25186610 PMCID: PMC11029513 DOI: 10.1007/s00262-014-1594-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2014] [Accepted: 08/05/2014] [Indexed: 11/25/2022]
Abstract
BACKGROUND The scarcity of effective therapeutic approaches for prostate cancer (PCa) has encouraged steadily growing interest for the identification of novel antigenic targets. Placenta-specific 1 (PLAC1) is a novel cancer-testis antigen with reported ectopic expression in a variety of tumors and cancer cell lines. The purpose of the present study was to investigate for the first time the differential expression of PLAC1 in PCa tissues. METHODS We investigated the differential expression of PLAC1 in PCa, high-grade prostatic intraepithelial neoplasia (HPIN), benign prostatic hyperplasia (BPH), and nonneoplastic/nonhyperplastic prostate tissues using microarray-based immunohistochemistry (n = 227). The correlation of PLAC1 expression with certain clinicopathological parameters and expression of prostate-specific antigen (PSA), as a prostate epithelial cell differentiation marker, were investigated. RESULTS Placenta-specific 1 (PLAC1) expression was increased in a stepwise manner from BPH to PCa, which expressed highest levels of this molecule, while in a majority of normal tissues, PLAC1 expression was not detected. Moreover, PLAC1 expression was positively associated with Gleason score (p ≤ 0.001). Interestingly, there was a negative correlation between PLAC1 and PSA expression in patients with PCa and HPIN (p ≤ 0.01). Increment of PLAC1 expression increased the odds of PCa and HPIN diagnosis (OR 49.45, 95 % CI for OR 16.17-151.25). CONCLUSION Our findings on differential expression of PLAC1 in PCa plus its positive association with Gleason score and negative correlation with PSA expression highlight the potential usefulness of PLAC1 for targeted PC therapy especially for patients with advanced disease.
Collapse
Affiliation(s)
- Roya Ghods
- Department of Molecular Medicine, School of Advanced Medical Technologies, Tehran University of Medical Sciences, TUMS, Tehran, Iran
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Mohammad-Hossein Ghahremani
- Department of Molecular Medicine, School of Advanced Medical Technologies, Tehran University of Medical Sciences, TUMS, Tehran, Iran
- Department of Pharmacology-Toxicology, Faculty of Medicine, Tehran University of Medical Sciences, TUMS, Tehran, Iran
- School of Advanced Technologies in Medicine, Eastern side of Tehran University, 88, Italia St, P.O. box: 1417755469, Tehran, Iran
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences, IUMS, Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, IUMS, Tehran, Iran
| | - Mojgan Asgari
- Oncopathology Research Center, Iran University of Medical Sciences, IUMS, Tehran, Iran
- Department of Pathology, Hasheminejad Kidney Center, Iran University of Medical Sciences, IUMS, Tehran, Iran
| | - Maryam Abolhasani
- Oncopathology Research Center, Iran University of Medical Sciences, IUMS, Tehran, Iran
- Department of Pathology, Hasheminejad Kidney Center, Iran University of Medical Sciences, IUMS, Tehran, Iran
| | - Sanaz Tavasoli
- Department of Nutrition, Science and Research Branch, Azad University, Tehran, Iran
| | - Ahmad-Reza Mahmoudi
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Maryam Darzi
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Parvin Pasalar
- Department of Molecular Medicine, School of Advanced Medical Technologies, Tehran University of Medical Sciences, TUMS, Tehran, Iran
| | - Mahmood Jeddi-Tehrani
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Amir-Hassan Zarnani
- Immunology Research Center, Iran University of Medical Sciences, IUMS, Hemmat Highway, P.O. box: 1449614535, Tehran, Iran
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| |
Collapse
|
49
|
Ghafouri-Fard S, Shamsi R, Seifi-Alan M, Javaheri M, Tabarestani S. Cancer-testis genes as candidates for immunotherapy in breast cancer. Immunotherapy 2014; 6:165-79. [PMID: 24491090 DOI: 10.2217/imt.13.165] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cancer-testis (CT) antigens are tumor-associated antigens attracting immunologists for their possible application in the immunotherapy of cancer. Several clinical trials have assessed their therapeutic potentials in cancer patients. Breast cancers, especially triple-negative cancers are among those with significant expression of CT genes. Identification of CT genes with high expression in cancer patients is the prerequisite for any immunotherapeutic approach. CT genes have gained attention not only for immunotherapy of cancer patients, but also for immunoprevention in high-risk individuals. Many CT genes have proved to be immunogenic in breast cancer patients suggesting the basis for the development of polyvalent vaccines.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran 19857-17443, Iran
| | | | | | | | | |
Collapse
|
50
|
Placenta-specific protein 1 is conserved throughout the Placentalia under purifying selection. ScientificWorldJournal 2014; 2014:537356. [PMID: 25180201 PMCID: PMC4142310 DOI: 10.1155/2014/537356] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 07/16/2014] [Indexed: 11/18/2022] Open
Abstract
Placental mammals (Placentalia) are a very successful group that, today, comprise 94% of all mammalian species. Recent phylogenetic analyses, coupled with new, quite complete fossils, suggest that the crown orders were all established rapidly from a common ancestor just after the Cretaceous/Tertiary (K/T) boundary 65 million years ago. Extensive molecular and morphologic evidence has led to a description of the common ancestor of all Placentalia in which a two-horned uterus and a hemochorial placenta are present. Thus, the process of placentation in which the placenta invades and anchors to the uterine epithelium was already established. One factor that has been suggested as a crucial component of this process is placenta-specific protein 1 (PLAC1). A phylogenetic analysis of the PLAC1 protein in 25 placental mammal species, representing nine of the sixteen crown orders of the Placentalia, suggests that this protein was present in the placental common ancestor in the form we see it today, that it evolved in the Placentalia and has been subject to the effects of purifying selection since its appearance.
Collapse
|