1
|
Pesini C, Artal L, Paúl Bernal J, Sánchez Martinez D, Pardo J, Ramírez-Labrada A. In-depth analysis of the interplay between oncogenic mutations and NK cell-mediated cancer surveillance in solid tumors. Oncoimmunology 2024; 13:2379062. [PMID: 39036370 PMCID: PMC11259085 DOI: 10.1080/2162402x.2024.2379062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 07/08/2024] [Indexed: 07/23/2024] Open
Abstract
Natural killer (NK) cells play a crucial role in antitumoral and antiviral responses. Yet, cancer cells can alter themselves or the microenvironment through the secretion of cytokines or other factors, hindering NK cell activation and promoting a less cytotoxic phenotype. These resistance mechanisms, often referred to as the "hallmarks of cancer" are significantly influenced by the activation of oncogenes, impacting most, if not all, of the described hallmarks. Along with oncogenes, other types of genes, the tumor suppressor genes are frequently mutated or modified during cancer. Traditionally, these genes have been associated with uncontrollable tumor growth and apoptosis resistance. Recent evidence suggests oncogenic mutations extend beyond modulating cell death/proliferation programs, influencing cancer immunosurveillance. While T cells have been more studied, the results obtained highlight NK cells as emerging key protagonists for enhancing tumor cell elimination by modulating oncogenic activity. A few recent studies highlight the crucial role of oncogenic mutations in NK cell-mediated cancer recognition, impacting angiogenesis, stress ligands, and signaling balance within the tumor microenvironment. This review will critically examine recent discoveries correlating oncogenic mutations to NK cell-mediated cancer immunosurveillance, a relatively underexplored area, particularly in the era dominated by immune checkpoint inhibitors and CAR-T cells. Building on these insights, we will explore opportunities to improve NK cell-based immunotherapies, which are increasingly recognized as promising alternatives for treating low-antigenic tumors, offering significant advantages in terms of safety and manufacturing suitability.
Collapse
Affiliation(s)
- Cecilia Pesini
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- Center for Biomedical Research in the Network of Infectious Diseases (CIBERINFEC), Carlos III Health Institute, Zaragoza, Spain
- Department of Microbiology, Radiology, Pediatry and Public Health, University of Zaragoza, Zaragoza, Spain
| | - Laura Artal
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- Institute of Carbochemistry (ICB-CSIC), Zaragoza, Spain
| | - Jorge Paúl Bernal
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
| | - Diego Sánchez Martinez
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- Aragón I + D Foundation (ARAID), Government of Aragon, Zaragoza, Spain
| | - Julián Pardo
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- Center for Biomedical Research in the Network of Infectious Diseases (CIBERINFEC), Carlos III Health Institute, Zaragoza, Spain
- Department of Microbiology, Radiology, Pediatry and Public Health, University of Zaragoza, Zaragoza, Spain
| | - Ariel Ramírez-Labrada
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- Center for Biomedical Research in the Network of Infectious Diseases (CIBERINFEC), Carlos III Health Institute, Zaragoza, Spain
| |
Collapse
|
2
|
Unnikrishnan Meenakshi D, Narde GK, Ahuja A, Al Balushi K, Francis AP, Khan SA. Therapeutic Applications of Nanoformulated Resveratrol and Quercetin Phytochemicals in Colorectal Cancer-An Updated Review. Pharmaceutics 2024; 16:761. [PMID: 38931884 PMCID: PMC11206904 DOI: 10.3390/pharmaceutics16060761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/07/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Natural compounds such as polyphenols play several positive roles in maintaining the oxidative and inflammatory capacity of cells, which leads to their potential use as anticancer therapeutics. There is promising evidence for the in vitro and in vivo anticancer activity of many polyphenols, including resveratrol and quercetin, specifically in the treatment of colorectal cancer (CRC). There is a clear association between resveratrol and quercetin in interfering with the mechanistic pathways involved in CRC, such as Wnt, P13K/AKT, caspase-3, MAPK, NF-κB, etc. These molecular pathways establish the role of resveratrol and quercetin in controlling cancer cell growth, inducing apoptosis, and inhibiting metastasis. The major bottleneck in the progression of the use of resveratrol and quercetin as anticancer therapeutics is their reduced bioavailability in vivo because of their rapid metabolism in humans. Recent advancements in various nanotechnological formulations are promising for overcoming these bioavailability issues. Various nanoformulations of resveratrol and quercetin have shown an optimistic impact on reducing the solubility and improving the stability of resveratrol and quercetin in vivo. A combinatorial approach using nanoformulations of resveratrol with quercetin could potentially increase the impact of resveratrol in controlling CRC cell proliferation. This review discusses the mechanism of resveratrol and quercetin, the two bioactive polyphenolics, in colon cancer, with an emphasis on various types of nanoformulations of the two molecules targeting colon cancer. It also explores the synergistic effect of combining resveratrol and quercetin in various nanoformulations, targeting colon cancer. This research delves into the enhanced pharmacokinetics and potential chemotherapeutic benefits of these bioactive polyphenolics when used together in innovative ways.
Collapse
Affiliation(s)
| | - Gurpreet Kaur Narde
- College of Pharmacy, National University of Science and Technology, Muscat PC 130, Oman; (G.K.N.); (K.A.B.); (S.A.K.)
| | - Alka Ahuja
- College of Pharmacy, National University of Science and Technology, Muscat PC 130, Oman; (G.K.N.); (K.A.B.); (S.A.K.)
| | - Khalid Al Balushi
- College of Pharmacy, National University of Science and Technology, Muscat PC 130, Oman; (G.K.N.); (K.A.B.); (S.A.K.)
| | - Arul Prakash Francis
- Centre of Molecular Medicine and Diagnostics (COMMAND), Saveetha Dental College and Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai 600077, India;
| | - Shah Alam Khan
- College of Pharmacy, National University of Science and Technology, Muscat PC 130, Oman; (G.K.N.); (K.A.B.); (S.A.K.)
| |
Collapse
|
3
|
Prajapati KS, Kumar S. Kurarinone targets JAK2-STAT3 signaling in colon cancer-stem-like cells. Cell Biochem Funct 2024; 42:e3959. [PMID: 38390770 DOI: 10.1002/cbf.3959] [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: 10/24/2023] [Revised: 02/13/2024] [Accepted: 02/14/2024] [Indexed: 02/24/2024]
Abstract
Natural compounds are known to regulate stemness/self-renewal properties in colon cancer cells at molecular level. In the present study, we first time studied the colon cancer stem-like cells targeting potential of Kurarinone (KU) and explored the underlying mechanism. Cytotoxic potential of KU was checked in colon cancer cells. Colonosphere formation assay was performed to check the spheroid formation reduction potential of KU in HCT-116 cells by using phase-contrast microscopy. Stemness/self-renewal marker expression was studied at mRNA and protein levels in colonosphere. The qRT-PCR, western blot analysis, and flow cytometer techniques were used to assess the effect of KU treatment on cell cycle progression and apoptosis induction in colon cancer cells and colonosphere. Further, effect of KU treatment on pSTAT3 status and its nuclear translocation was also studied. KU treatment significantly decreased HCT-116 cell proliferation and reduced sphere formation potential at IC30 (8.71 µM) and IC50 (20.34 µM) concentrations compared to respective vehicle-treated groups, respectively. KU exposure significantly reduced the expression of CD44, c-Myc, Bmi-1, and Sox2 stemness/self-renewal markers in colonosphere in a dose-dependent manner. KU treatment inhibits JAK2-STAT3 signaling pathway by reducing pSTAT3 levels and its nuclear translocation in HCT-116 cells and colonosphere at IC50 concentration. KU treatment significantly decreased the expression of CCND1 and CDK4 cell cycle-specific markers and arrested the HCT-116 cells and colonosphere in G1-phase. Further, KU treatment increased Bax/Bcl-2 ratio, apoptotic cell population, cleaved caspase 3, and PARP-1 in HCT-116 cells and colonosphere. In conclusion, KU treatment decreases stemness/self-renewal, induces cell cycle arrest and apoptosis in HCT-116 colonosphere by down-regulating CD44-JAK2-STAT3 axis. Thus, targeting stemness/self-renewal and other cancer hallmark(s) by KU through CD44/JAK2/STAT3 signaling pathway might be a novel strategy to target colon cancer stem-like cells.
Collapse
Affiliation(s)
- Kumari Sunita Prajapati
- Molecular Signaling & Drug Discovery Laboratory, Department of Biochemistry, Central University of Punjab, Bathinda, Punjab, India
| | - Shashank Kumar
- Molecular Signaling & Drug Discovery Laboratory, Department of Biochemistry, Central University of Punjab, Bathinda, Punjab, India
| |
Collapse
|
4
|
Tani N, Ikeda T, Ishikawa T. Effects of Prolactin on Brain Neurons under Hypoxia. Life (Basel) 2024; 14:152. [PMID: 38276281 PMCID: PMC10817236 DOI: 10.3390/life14010152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/17/2024] [Accepted: 01/20/2024] [Indexed: 01/27/2024] Open
Abstract
The levels and potential role of prolactin (PRL) in the brain under conditions of acute systemic hypoxia were examined, focusing on the accumulation of PRL in cerebrospinal fluid (CSF) and its effects on neuronal activity and injury. The amount of PRL in the brain was investigated using brain tissues from forensic autopsy cases. We counted the number of neurites that formed in human primary neurons (HNs) after the addition of PRL. Furthermore, HNs supplemented with PRL or triiodothyronine (T3) were exposed to hypoxic conditions, and the dead cells were counted. The results showed correlations between brain PRL and CSF PRL levels. Additionally, PRL accumulation in the brain was observed in cases of asphyxia. In vitro experimental findings indicated increased neurite formation in the HNs treated with PRL. Moreover, both PRL and T3 demonstrated neuroprotective effects against hypoxia-induced neuronal cell death, with PRL showing stronger neuroprotective potential than T3. These results suggest that PRL accumulates in the brain during hypoxia, potentially influences neuronal activity, and exhibits neuroprotective properties against hypoxia-induced neuronal injury.
Collapse
Affiliation(s)
- Naoto Tani
- Department of Legal Medicine, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3 Asahi-machi, Abeno, Osaka 545-8585, Japan; (T.I.); (T.I.)
- Forensic Autopsy Section, Medico-Legal Consultation and Postmortem Investigation Support Center, 1-4-3 Asahi-machi, Abeno, Osaka 545-8585, Japan
| | - Tomoya Ikeda
- Department of Legal Medicine, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3 Asahi-machi, Abeno, Osaka 545-8585, Japan; (T.I.); (T.I.)
| | - Takaki Ishikawa
- Department of Legal Medicine, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3 Asahi-machi, Abeno, Osaka 545-8585, Japan; (T.I.); (T.I.)
- Forensic Autopsy Section, Medico-Legal Consultation and Postmortem Investigation Support Center, 1-4-3 Asahi-machi, Abeno, Osaka 545-8585, Japan
| |
Collapse
|
5
|
Zhong C, Wang G, Guo M, Zhu N, Chen X, Yan Y, Li N, Yu W. The Role of Tumor Stem Cells in Colorectal Cancer Drug Resistance. Cancer Control 2024; 31:10732748241274196. [PMID: 39215442 PMCID: PMC11367616 DOI: 10.1177/10732748241274196] [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: 02/24/2024] [Revised: 07/09/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024] Open
Abstract
Background: Colorectal cancer is a major cause of mortality among the prevalent malignant tumors of the gastrointestinal tract. Although chemotherapy is a standard treatment for colorectal cancer, its efficacy is limited by chemoresistance. Recent studies have investigated targeting tumor stem cells as a potential new therapeutic approach for addressing chemoresistance in colorectal cancer. Colorectal cancer frequently relapses, with tumor stem cells often representing one of the leading causes of treatment failure. Purpose: Understanding drug resistance in colorectal cancer stem cells is crucial for improving treatment outcomes. By focusing on developing targeted therapies that specifically address drug resistance in colorectal cancer stem cells, there is potential to make significant advancements in the treatment of colorectal cancer.This approach may lead to more effective and lasting outcomes in patients battling colorectal cancer. Research Design: In this review, a comprehensive overview of recent research on colorectal cancer stem cell treatment resistance is presented.Results: Elucidating the key underlying mechanisms. This review also highlights the potential benefits of targeted therapies in overcoming colorectal cancer resistance to treatment. Conclusions: CCSCs are key players in drug resistance of CRC, indicating their potential as targets for effective therapy. Elucidating their role in this process could aid in discovering tailored treatment strategies.The significance of signaling pathways, TME, and miRNA in regulating drug resistance in CCSCs is been highlighted.
Collapse
Affiliation(s)
- Chen Zhong
- Jiangxi University of Chinese Medicine, Nanchang, China
| | - Guojuan Wang
- Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, China
| | - Min Guo
- Jiangxi University of Chinese Medicine, Nanchang, China
| | - Naicheng Zhu
- Jiangxi University of Chinese Medicine, Nanchang, China
| | - Xiudan Chen
- Jiangxi University of Chinese Medicine, Nanchang, China
| | - Yuwei Yan
- Jiangxi University of Chinese Medicine, Nanchang, China
| | - Nanxin Li
- Jiangxi University of Chinese Medicine, Nanchang, China
| | - Wenyan Yu
- Jiangxi University of Chinese Medicine, Nanchang, China
| |
Collapse
|
6
|
Yang R, Zhang S, Duan C, Guo Y, Shan X, Zhang X, Yue S, Zhang Y, Liu Y. Effect of prolactin on cytotoxicity and oxidative stress in ovine ovarian granulosa cells. PeerJ 2023; 11:e15629. [PMID: 37456891 PMCID: PMC10340108 DOI: 10.7717/peerj.15629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 06/02/2023] [Indexed: 07/18/2023] Open
Abstract
Background Prolactin (PRL) has been reported to be associated with oxidative stress, which is an important contributor leading to cell apoptosis. However, little is known about the mechanisms underlying the effects of PRL on cytotoxicity and oxidative stress in ovine ovarian granulosa cells (GCs). Methods Ovine ovarian GCs were treated with 0, 4, 20, 100 and 500 ng/mL of PRL. Then, the cytotoxicity, cell viability, malondialdehyde (MDA), reactive oxygen species (ROS), superoxide dismutase (SOD) and total antioxidant capacity (T-AOC) of GCs were detected. Additionally, 500 ng/mL PRL was chosen as the high PRL concentration (HPC) due to its high cytotoxicity and oxidative stress. Proteomic and metabonomic were performed to examine the overall difference in proteins and metabolic pathways between C (control: 0 ng/mL PRL) and P groups (500 ng/mL PRL). Results The results indicated that GCs treated with 4 ng/mL PRL significantly decreased (P < 0.05) the cytotoxicity, ROS and MDA, increased (P < 0.05) the cell viability, SOD and T-AOC, and the GCs treated with 500 ng/mL PRL showed the opposite trend (P < 0.05). Supplementation with 500 ng/mL PRL significantly increased the proteins of MT-ND1, MAPK12, UBA52 and BCL2L1, which were enriched in ROS and mitophagy pathways. Pathway enrichment analysis showed that the pentose phosphate pathway was significantly enriched in the P group. Conclusion A low concentration of PRL inhibited cytotoxicity and oxidative stress. HPC induced oxidative stress in ovine ovarian GCs via the pentose phosphate pathway by modulating the associated proteins MT-ND1 in ROS pathway and UBA52, MAPK12 and BCL2L1 in mitophagy pathway, resulting in cytotoxicity.
Collapse
Affiliation(s)
| | - Shuo Zhang
- China Agricultural University, Beijing, China
| | | | - Yunxia Guo
- Hebei Agricultural University, Baoding, China
| | - Xinyu Shan
- Hebei Agricultural University, Baoding, China
| | | | - Sicong Yue
- Hebei Agricultural University, Baoding, China
| | | | - Yueqin Liu
- Hebei Agricultural University, Baoding, China
| |
Collapse
|
7
|
Yang R, Duan C, Zhang S, Liu Y, Zhang Y. Prolactin Regulates Ovine Ovarian Granulosa Cell Apoptosis by Affecting the Expression of MAPK12 Gene. Int J Mol Sci 2023; 24:10269. [PMID: 37373417 DOI: 10.3390/ijms241210269] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Prolactin (PRL) has been reported to influence reproductive performance and cell apoptosis. However, its mechanism remains unclear. Hence, in the present study, ovine ovarian granulosa cells (GCs) were used as a cell model to investigate the relationship between PRL concentration and GC apoptosis, as well as its possible mechanisms. We examined the relationship between serum PRL concentration and follicle counts in sexually mature ewes. GCs were isolated from adult ewes and treated with different concentrations of PRL, while 500 ng/mL PRL was selected as the high concentration of prolactin (HPC). Then, we applied the transcriptome sequencing (RNA-Seq) combined with a gene editing approach to explore the HPC contributing to cell apoptosis and steroid hormones. The apoptosis of GCs gradually increased at PRL concentrations above 20 ng/mL, while 500 ng/mL PRL significantly decreased the secretion of steroid hormones and the expression of L-PRLR and S-PRLR. The results indicated that PRL regulates GC development and steroid hormones mainly through the target gene MAPK12. The expression of MAPK12 was increased after knocked-down L-PRLR and S-PRLR, while it decreased after overexpressed L-PRLR and S-PRLR. Cell apoptosis was inhibited and the secretion of steroid hormones increased after interfering with MAPK12, while the overexpression of MAPK12 showed the opposite trend. Overall, the number of follicles gradually decreased with increasing PRL concentration. HPCs promoted apoptosis and inhibited steroid hormone secretion in GCs by upregulating MAPK12 through reducing L-PRLR and S-PRLR.
Collapse
Affiliation(s)
- Ruochen Yang
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071000, China
| | - Chunhui Duan
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071000, China
| | - Shuo Zhang
- College of Animal Science and Technology, China Agricultural University, Beijing 100089, China
| | - Yueqin Liu
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071000, China
| | - Yingjie Zhang
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071000, China
| |
Collapse
|
8
|
List EO, Berryman DE, List BP, Kopchick JJ. Early Investigations of 20-kDa Human Placental GH Show Promise. Endocr Metab Immune Disord Drug Targets 2023; 23:1674-1677. [PMID: 37190799 PMCID: PMC11483157 DOI: 10.2174/1871530323666230515153130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 03/08/2023] [Accepted: 03/10/2023] [Indexed: 05/17/2023]
Affiliation(s)
- Edward O. List
- Edison Biotechnology Institute, Ohio University, Athens Ohio, 45701, United States
- Department of Specialty Medicine, Heritage College of Osteopathic Medicine, Athens Ohio
| | - Darlene E. Berryman
- Edison Biotechnology Institute, Ohio University, Athens Ohio, 45701, United States
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Athens Ohio
| | - Brian P. List
- Edison Biotechnology Institute, Ohio University, Athens Ohio, 45701, United States
| | - John J. Kopchick
- Edison Biotechnology Institute, Ohio University, Athens Ohio, 45701, United States
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Athens Ohio
| |
Collapse
|
9
|
Hu Q, Bian Q, Rong D, Wang L, Song J, Huang HS, Zeng J, Mei J, Wang PY. JAK/STAT pathway: Extracellular signals, diseases, immunity, and therapeutic regimens. Front Bioeng Biotechnol 2023; 11:1110765. [PMID: 36911202 PMCID: PMC9995824 DOI: 10.3389/fbioe.2023.1110765] [Citation(s) in RCA: 53] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/13/2023] [Indexed: 02/25/2023] Open
Abstract
Janus kinase/signal transduction and transcription activation (JAK/STAT) pathways were originally thought to be intracellular signaling pathways that mediate cytokine signals in mammals. Existing studies show that the JAK/STAT pathway regulates the downstream signaling of numerous membrane proteins such as such as G-protein-associated receptors, integrins and so on. Mounting evidence shows that the JAK/STAT pathways play an important role in human disease pathology and pharmacological mechanism. The JAK/STAT pathways are related to aspects of all aspects of the immune system function, such as fighting infection, maintaining immune tolerance, strengthening barrier function, and cancer prevention, which are all important factors involved in immune response. In addition, the JAK/STAT pathways play an important role in extracellular mechanistic signaling and might be an important mediator of mechanistic signals that influence disease progression, immune environment. Therefore, it is important to understand the mechanism of the JAK/STAT pathways, which provides ideas for us to design more drugs targeting diseases based on the JAK/STAT pathway. In this review, we discuss the role of the JAK/STAT pathway in mechanistic signaling, disease progression, immune environment, and therapeutic targets.
Collapse
Affiliation(s)
- Qian Hu
- Department of Pharmacy, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, China.,Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Qihui Bian
- Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, China
| | - Dingchao Rong
- Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Leiyun Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China.,Department of Pharmacy, Wuhan First Hospital, Wuhan, China
| | - Jianan Song
- Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, China
| | - Hsuan-Shun Huang
- Department of Research, Center for Prevention and Therapy of Gynecological Cancers, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Jun Zeng
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Jie Mei
- Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, China.,Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Peng-Yuan Wang
- Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
10
|
Gu W, Mitsuhashi A, Kobayashi T, Shozu M. Metformin attenuates the production and proliferative effects of prolactin induced by medroxyprogesterone acetate during fertility-sparing treatment for endometrial cancer. BMC Cancer 2022; 22:753. [PMID: 35820883 PMCID: PMC9277913 DOI: 10.1186/s12885-022-09858-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 07/05/2022] [Indexed: 11/18/2022] Open
Abstract
Background Progestin is used for fertility-sparing treatment in cases of endometrial cancer (EC). Progestin can induce hyperprolactinemia by increasing pituitary secretion and endometrial decidualization. However, progestin induces prolactin (PRL) secretion, which stimulates cell proliferation and deleteriously affects treatment. To date, the detrimental effect of PRL, the secretion of which is induced by medroxyprogesterone acetate (MPA) during fertility-sparing treatment, has not yet been fully elucidated. Therefore, we aimed to assess the effects of PRL on EC cells during combined treatment with progestin and metformin. Methods In total, 71 patients with EC/endometrial atypical hyperplasia who underwent fertility-sparing treatment at our institution from 2009–2019 were enrolled. Serum PRL levels were determined using enzyme immunoassays; mRNA levels in endometrial tissues were determined using quantitative reverse-transcription PCR. To evaluate MPA-induced decidualization, cancer-associated stromal cells were enzymatically released from surgically removed specimens of six patients with EC. To examine PRL-induced cell proliferation, the EC cell lines Ishikawa, HEC1B, and HEC265 were used. In vitro cell proliferation was evaluated using the WST assay; protein levels of signaling molecules were determined using western blotting. Results MPA administration significantly increased serum PRL levels at 3 and 6 months and upregulated IGFBP-1 and PRL mRNA expression in tissues at 3 months of fertility-sparing treatment. Metformin significantly reduced MPA-induced IGFBP-1 and PRL mRNA expression during fertility-sparing treatment and significantly inhibited the upregulation of IGFBP-1 and PRL mRNA and PRL levels due to decidualization induced by MPA and cAMP treatment in primary cultured EC stromal cells. In vitro, PRL increased cell proliferation and ERK1/2 phosphorylation levels, whereas metformin attenuated these increases. Conclusions MPA upregulated PRL levels in serum and endometrial tissues during fertility-sparing treatment. Metformin co-administration reduced PRL production and attenuated PRL-induced cell-proliferation activity. This study may provide valuable insights on the application of metformin to improve the outcomes of fertility-sparing treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09858-w.
Collapse
Affiliation(s)
- Wenjing Gu
- Department of Reproductive Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Akira Mitsuhashi
- Department of Reproductive Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan. .,Department of Obstetrics and Gynecology, School of Medicine, Dokkyo Medical University, Tochigi, Japan.
| | - Tatsuya Kobayashi
- Department of Reproductive Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Makio Shozu
- Department of Reproductive Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
11
|
Targeting Inhibition of Notch1 Signaling Pathway on the Study of Human Gastric Cancer Stem Cells with Chemosensitization. COMPUTATIONAL INTELLIGENCE AND NEUROSCIENCE 2022; 2022:1098394. [PMID: 35515501 PMCID: PMC9064537 DOI: 10.1155/2022/1098394] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/25/2022] [Accepted: 03/30/2022] [Indexed: 12/31/2022]
Abstract
Background Gastric cancer is the second most frequent cause of cancer death worldwide, although much geographical variation in incidence exists. Prevention and personalized treatment are regarded as the best options to reduce gastric cancer mortality rates (Hartgrink et al., 2009). Numerous studies have suggested that Notch1 and its ligands are overexpressed in gastric cancer, and its knockdown can inhibit the proliferation and survival of gastric cancer cells. Objective To investigate the effect of Notch1 on the stemness and drug sensitivity of human gastric cancer SGC-7901 cells. Methods Highly expressed Notch1 intracellular domain (NICD1) and Notch1-shRNA lentiviral expression vector were used to infect human gastric cancer SGC-7901 cells cultured in vitro, and western blot and immunofluorescence staining were used to identify highly expressed NICD and Notch1 silenced cells. The percentage of CD133+ cells was analyzed by flow cytometry, the expression of nestin and CFAP by immunofluorescence staining, the formation rate of tumor cell spheres and the tumorigenicity of SCID mice in vivo, and the regulation of cell stemness by Notch1. The sensitivity of each group of cells to the chemotherapeutic drugs teniposide (VM-26) and carmustine (BCNU) was also detected by the MTT method. Results The stemness phenotype of tumor cells with the increased NICD expression was enhanced, such as an increased proportion of CD133+ cells, enhanced nestin expression, decreased GFAP expression, increased tumor cell sphere formation rate and tumorigenic rate of SCID mice implantation, and decreased sensitivity to VM-26 and BCNU. In contrast, the stemness phenotype of tumor cells with downregulated Notch1 gene expression was significantly suppressed, while the sensitivity to VM-26 and BCNU was increased. Conclusion High Notch1 expression increased the stemness of SGC-7901 cells and decreased the sensitivity of SGC-7901 cells to chemotherapeutic drugs.
Collapse
|
12
|
Peng L, Zhao M, Liu T, Chen J, Gao P, Chen L, Xing P, Wang Z, Di J, Xu Q, Qu H, Jiang B, Su X. A stop-gain mutation in GXYLT1 promotes metastasis of colorectal cancer via the MAPK pathway. Cell Death Dis 2022; 13:395. [PMID: 35459861 PMCID: PMC9033806 DOI: 10.1038/s41419-022-04844-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 04/04/2022] [Accepted: 04/06/2022] [Indexed: 12/17/2022]
Abstract
Genomic instability plays a key role in the initiation and progression of colorectal cancer (CRC). Although cancer driver genes in CRC have been well characterized, identifying novel genes associated with carcinogenesis and treatment remains challenging because of tumor heterogeneity. Here, we analyzed the genomic alterations of 45 samples from CRC patients in northern China by whole-exome sequencing. In addition to the identification of six well-known CRC driver genes (APC, TP53, KRAS, FBXW7, PIK3CA, and PABPC), two tumor-related genes (MTCH2 and HSPA6) were detected, along with RRP7A and GXYLT1, which have not been previously linked to cancer. GXYLT1 was mutated in 40% (18/45) of the samples in our cohort. Functionally, GXYLT1 promoted migration and invasion in vitro and metastasis in vivo, while the GXYLT1S212* mutant induced significantly greater effect. Furthermore, both GXYLT1 and GXYLT1S212* interacted with ERK2. GXYLT1 induced metastasis via a mechanism involving the Notch and MAPK pathways, whereas the GXYLT1S212* mutant mainly promoted metastasis by activating the MAPK pathway. We propose that GXYLT1 acts as a novel metastasis-associated driver gene and GXYLT1S212* might serve as a potential indicator for therapies targeting the MAPK pathway in CRC.
Collapse
Affiliation(s)
- Lin Peng
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Min Zhao
- School of Science, Technology and Engineering, University of the Sunshine Coast, Maroochydore DC, QLD, 4556, Australia
| | - Tianqi Liu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Jiangbo Chen
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Pin Gao
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Lei Chen
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Pu Xing
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Zaozao Wang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Jiabo Di
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Qiang Xu
- GenomiCare Biotechnology (Shanghai) Co., Ltd, Shanghai, 201210, China
| | - Hong Qu
- Center for Bioinformatics, State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing, 100871, P. R. China.
| | - Beihai Jiang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| | - Xiangqian Su
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
13
|
Standing D, Dandawate P, Anant S. Prolactin receptor signaling: A novel target for cancer treatment - Exploring anti-PRLR signaling strategies. Front Endocrinol (Lausanne) 2022; 13:1112987. [PMID: 36714582 PMCID: PMC9880166 DOI: 10.3389/fendo.2022.1112987] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 12/22/2022] [Indexed: 01/15/2023] Open
Abstract
Prolactin (PRL) is a peptide hormone mainly secreted from the anterior pituitary gland. PRL is reported to play a role in pregnancy, mammary gland development, immune modulation, reproduction, and differentiation of islet cells. PRL binds to its receptor PRLR, which belongs to a superfamily of the class I cytokine receptor that has no intrinsic kinase activity. In canonical signaling, PRL binding to PRLR induces downstream signaling including JAK-STAT, AKT and MAPK pathways. This leads to increased cell proliferation, stemness, migration, apoptosis inhibition, and resistance to chemotherapy. PRL-signaling is upregulated in numerous hormone-dependent cancers including breast, prostate, ovarian, and endometrial cancer. However, more recently, the pathway has been reported to play a tumor-promoting role in other cancer types such as colon, pancreas, and hepatocellular cancers. Hence, the signaling pathway is an attractive target for drug development with blockade of the receptor being a potential therapeutic approach. Different strategies have been developed to target this receptor including modification of PRL peptides (Del1-9-G129R-hPRL, G129R-Prl), growth hormone receptor/prolactin receptor bispecific antibody antagonist, neutralizing antibody LFA102, an antibody-drug conjugate (ABBV-176) of the humanized antibody h16f (PR-1594804) and pyrrolobenzodiazepine dimer, a bispecific antibody targeting both PRLR and CD3, an in vivo half-life extended fusion protein containing PRLR antagonist PrlRA and albumin binding domain. There have also been attempts to discover and develop small molecular inhibitors targeting PRLR. Recently, using structure-based virtual screening, we identified a few antipsychotic drugs including penfluridol as a molecule that inhibits PRL-signaling to inhibit PDAC tumor progression. In this review, we will summarize the recent advances in the biology of this receptor in cancer and give an account of PRLR antagonist development for the treatment of cancer.
Collapse
|
14
|
Ma TC, Ma YK, Zhang JL, Liu L, Sun J, Guo LN, Liu Q, Sun Y. Integrated Strategy of UHPLC-Q-TOF-MS and Molecular Networking for Identification of Diterpenoids from Euphorbia fischeriana Steud. and Prediction of the Anti-Breast-Cancer Mechanism by the Network Pharmacological Method. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:3829434. [PMID: 34804177 PMCID: PMC8601799 DOI: 10.1155/2021/3829434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 09/09/2021] [Accepted: 10/15/2021] [Indexed: 11/29/2022]
Abstract
Breast cancer is one of the most common malignancies in women worldwide. Traditional Chinese medicine has been used as adjunctive or complementary therapy for breast cancer. Diterpenoids from Euphorbia fischeriana Steud. have been demonstrated to possess anti-breast-cancer activity. This research was aimed to systematically explore the diterpenoids from E. fischeriana and study the multiple mechanisms on breast cancer. The structures of diterpenoids were identified by the integrated strategy of UHPLC-Q-TOF-MS and molecular networking. A total of 177 diterpenoids belonging to 13 types were collected. In silico ADME analysis was performed on these compounds. It indicated that 130 of 177 diterpenoids completely adjusted to Lipinski's rule. The targets of compounds were obtained from PharmMapper. The targets of breast cancer were collected from GeneCards. Then, 197 compounds-related targets and 544 breast cancer-related targets were identified. After the intersection process, 58 overlapping targets between compounds-related targets and breast cancer-related targets were acquired. The STRING database was applied to predict the protein-protein interactions. The GO and KEGG pathway enrichment analysis were performed by using the KOBAS database. It indicated that these predicted pathways were closely related to breast cancer. The treatment effect of E. fischeriana on breast cancer might be performed through signaling pathways, such as IL-17 signaling pathway, MAPK signaling pathway, and PI3K-Akt signaling pathway. The predicted top genes such as EGFR, ESR, MAPK, SRC, CASP3, CDK2, and KDR were involved in cell proliferation, gene transcription, apoptosis, signal transduction, DNA damage and repair, tumor differentiation, metastasis, and cell cycle, which indicated that E. fischeriana might treat breast cancer comprehensively. A compounds-KEGG pathways-related targets network was built by using cytoHubba to analyze the hub compounds and targets. It concluded that E. fischeriana treated breast cancer not only by the main components but also by the microconstituents, which reflected the overall regulatory role of multicomponents treating breast cancer. To estimate the binding affinities, binding sites, and binding postures, molecular docking simulations between 177 diterpenoids and top 19 targets were carried out. The results are basically in line with expectations. In conclusion, these results can serve as references for researchers studying potential targets of diterpenoids from E. fischeriana on breast cancer in the future.
Collapse
Affiliation(s)
- Tian-Cheng Ma
- Research Institute of Medicine and Pharmacy, Qiqihar Medical University, Bukui Road 333, Qiqihar 161006, Heilongjiang, China
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, Liaoning, China
| | - Yu-Kun Ma
- Research Institute of Medicine and Pharmacy, Qiqihar Medical University, Bukui Road 333, Qiqihar 161006, Heilongjiang, China
| | - Jin-Ling Zhang
- Research Institute of Medicine and Pharmacy, Qiqihar Medical University, Bukui Road 333, Qiqihar 161006, Heilongjiang, China
| | - Lei Liu
- Research Institute of Medicine and Pharmacy, Qiqihar Medical University, Bukui Road 333, Qiqihar 161006, Heilongjiang, China
| | - Jia Sun
- Research Institute of Medicine and Pharmacy, Qiqihar Medical University, Bukui Road 333, Qiqihar 161006, Heilongjiang, China
| | - Li-Na Guo
- Research Institute of Medicine and Pharmacy, Qiqihar Medical University, Bukui Road 333, Qiqihar 161006, Heilongjiang, China
| | - Qi Liu
- Research Institute of Medicine and Pharmacy, Qiqihar Medical University, Bukui Road 333, Qiqihar 161006, Heilongjiang, China
| | - Yu Sun
- Research Institute of Medicine and Pharmacy, Qiqihar Medical University, Bukui Road 333, Qiqihar 161006, Heilongjiang, China
| |
Collapse
|
15
|
Huntington KE, Louie A, Zhou L, El-Deiry WS. A high-throughput customized cytokinome screen of colon cancer cell responses to small-molecule oncology drugs. Oncotarget 2021; 12:1980-1991. [PMID: 34611474 PMCID: PMC8487726 DOI: 10.18632/oncotarget.28079] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 09/03/2021] [Indexed: 11/25/2022] Open
Abstract
Inflammatory cytokines, chemokines, and growth factors are molecular messengers that circulate and have the capability to modify the tumor microenvironment and impact therapeutic response. The characterization of soluble mediators as biomarkers for diagnosis and prognosis is of interest in oncology. We utilize the cytokinome to characterize the response of colorectal tumor cell lines to selected small-molecules in oncology as a proof-of-concept dataset with immunomodulatory analyte heat map rankings for drug and cell line combinations. We observed overall trends in drug class effects with MEK-, BRAF-, PARP-inhibitors, and Imipridones in cytokine, chemokine, and growth factor responses that may help guide therapy selection. MEK-inhibitor treatment downregulated analytes VEGF, CXCL9/MIG, and IL-8/CXCL8 and upregulated CXCL14/BRAK, Prolactin, and CCL5/RANTES. BRAF-inhibitor treatment downregulated VEGF and IL-8/CXCL8, while increasing soluble TRAIL-R2. Treatment with PARP-inhibitors decreased CXCL9/MIG, IL-8/CXCL8, CCL3/MIP-1 alpha, VEGF, and CXCL14/BRAK, while treatment increased soluble TRAIL-R2 and prolactin. Treatment with Imipridones decreased CCL3/MIP-1 alpha, VEGF, CXCL14/BRAK, IL-8/CXCL8, and Prolactin and increased CXCL5/ENA-78. We also observed differential responses to therapeutics depending on the mutational profile of the cell line. In the future, a similar but larger dataset may be utilized in the clinic to aid in the prediction of patient response to immunomodulatory therapies based on tumor genotype.
Collapse
Affiliation(s)
- Kelsey E. Huntington
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI 02912, USA
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Pathobiology Graduate Program, Brown University, Providence, RI 02912, USA
| | - Anna Louie
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI 02912, USA
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Department of Surgery, Brown University, Lifespan Health System and Warren, Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Lanlan Zhou
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI 02912, USA
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Wafik S. El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI 02912, USA
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Pathobiology Graduate Program, Brown University, Providence, RI 02912, USA
| |
Collapse
|
16
|
Yang W, Qin C, Han J, Han S, Bai W, Du Y, Xu T. What Mediates Fibrosis in the Tumor Microenvironment of Clear Renal Cell Carcinoma. Front Genet 2021; 12:725252. [PMID: 34539753 PMCID: PMC8446447 DOI: 10.3389/fgene.2021.725252] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/13/2021] [Indexed: 01/31/2023] Open
Abstract
Previous studies have demonstrated that direct targeting of interstitial cancer-associated fibroblasts (CAF) and tumor fibrosis alone seemed to be an unpromising treatment option for malignant tumors. Therefore, it is necessary to further explore the mechanism of the influence of collagen and tumor fibrosis on the biological behavior of malignant tumors. The current study aimed to explore the effect of intratumor fibrosis on the prognosis of renal clear cell carcinoma (ccRCC) and its mechanism. With the bioinformatic analysis of The Cancer Genome Atlas (TCGA) database (n = 537), the study showed that high Collagen type I α 1 (COL1A1) mRNA expression indicated the poor prognosis of ccRCC patients compared with low expression ones. We further used the Two-photon-excited fluorescence (TPEF)/second harmonic generation (SHG) microscopy to determine the intratumor fibrosis of 68 patients with surgical resection of ccRCC and confirmed that a high fibrosis level in the tumor was associated with a poor prognosis compared with patients with low expression (Progression-Free Survival: p = 0.030). We further measured the protein chips of 640 cytokines in ccRCC specimens and found that several cytokines, including prolactin (PRL), were associated with the degree of fibrosis in the tumor, as confirmed by the prolactin receptor (PRLR) immunohistochemical method. In addition, the study showed that PRLR expression decreased significantly in the ccRCC compared with adjacent normal tissue (p < 0.05). Our research shows that low expression of PRLR predicted the poor survival of the patient. We used the Cell Counting Kit-8 experiment, the transwell and the plate clone formation assay to evaluate the role of PRL in the 7860 and the ACHN cell lines. We found that PRL promoted ccRCC cell proliferation and migration. JAK-STAT3 activation was found in the high prolactin expression group by mass spectrum analysis. This study delineated the fibrosis-based tumor microenvironment characteristics of ccRCC. PRL/PRLR may be involved in the fibrosis process and are essential prognostic risk factors for ccRCC.
Collapse
Affiliation(s)
- Wenbo Yang
- Department of Urology, Peking University People's Hospital, Beijing, China
| | - Caipeng Qin
- Department of Urology, Peking University People's Hospital, Beijing, China
| | - Jingli Han
- Department of Urology, Peking University People's Hospital, Beijing, China
| | - Songchen Han
- Department of Urology, Peking University People's Hospital, Beijing, China
| | - Wenjun Bai
- Department of Urology, Peking University People's Hospital, Beijing, China
| | - Yiqing Du
- Department of Urology, Peking University People's Hospital, Beijing, China
| | - Tao Xu
- Department of Urology, Peking University People's Hospital, Beijing, China
| |
Collapse
|
17
|
Kavarthapu R, Anbazhagan R, Dufau ML. Crosstalk between PRLR and EGFR/HER2 Signaling Pathways in Breast Cancer. Cancers (Basel) 2021; 13:4685. [PMID: 34572912 PMCID: PMC8467304 DOI: 10.3390/cancers13184685] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 12/20/2022] Open
Abstract
Prolactin receptor (PRLR) and epidermal growth factor receptor (EGFR/ERBB) signaling pathways activated by prolactin (PRL) and epidermal growth factor (EGF), have a major role in the mammary gland development and in the etiology of breast cancer, respectively. ER+ breast tumors comprise up to 75% of all breast cancers and 10% of these are HER2+. Elevated levels of PRLR in breast tumors, high circulating levels of PRL and increased expression of ERBB1/2 in patients that become resistant to endocrine therapy have shown to be associated with higher risk of cancer progression. In this review, we examine the role of crosstalk between PRLR and ERBB1/2 signaling pathways in the activation of unliganded ERα, cyclin-D1 and other oncogenic factors (MYC, FOS, JUN) in breast cancer. PRL/PRLR and EGF/EGFR induces phosphorylation of ERα through activation of MEK/MAPK and PI3K/AKT signaling pathways. PRL in breast cancer cells via PRLR/JAK2 can also induce phosphorylation of ERBB2/HER2, which in turn activates the downstream RAS/MEK/ERK pathway required for ERα phosphorylation. EGFR, independent of PRL/PRLR, can activate STAT5 indirectly via c-SRC and drive the expression of target genes involved in cell proliferation and survival. The crosstalk between PRLR and HER2, where PRL induces HER2 signaling can be an alternative route for ERα activation to induce transcription of PRLR and other ER target genes. We believe that overexpression of EGFR/HER2 and PRLR in breast tumors could maximize the actions of their ligands, and further induce cell proliferation promoting malignancy. This could also explain the resistance to endocrine therapy resulting in tumor growth.
Collapse
Affiliation(s)
| | | | - Maria L. Dufau
- Section on Molecular Endocrinology, Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA; (R.K.); (R.A.)
| |
Collapse
|
18
|
Flores-Fernández R, Aponte-López A, Suárez-Arriaga MC, Gorocica-Rosete P, Pizaña-Venegas A, Chávez-Sanchéz L, Blanco-Favela F, Fuentes-Pananá EM, Chávez-Rueda AK. Prolactin Rescues Immature B Cells from Apoptosis-Induced BCR-Aggregation through STAT3, Bcl2a1a, Bcl2l2, and Birc5 in Lupus-Prone MRL/lpr Mice. Cells 2021; 10:cells10020316. [PMID: 33557010 PMCID: PMC7913714 DOI: 10.3390/cells10020316] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 01/23/2021] [Accepted: 01/24/2021] [Indexed: 12/30/2022] Open
Abstract
Self-reactive immature B cells are eliminated through apoptosis by tolerance mechanisms, failing to eliminate these cells results in autoimmune diseases. Prolactin is known to rescue immature B cells from B cell receptor engagement-induced apoptosis in lupus-prone mice. The objective of this study was to characterize in vitro prolactin signaling in immature B cells, using sorting, PCR array, RT-PCR, flow cytometry, and chromatin immunoprecipitation. We found that all B cell maturation stages in bone marrow express the prolactin receptor long isoform, in both wild-type and MRL/lpr mice, but its expression increased only in the immature B cells of the latter, particularly at the onset of lupus. In these cells, activation of the prolactin receptor promoted STAT3 phosphorylation and upregulation of the antiapoptotic Bcl2a1a, Bcl2l2, and Birc5 genes. STAT3 binding to the promoter region of these genes was confirmed through chromatin immunoprecipitation. Furthermore, inhibitors of prolactin signaling and STAT3 activation abolished the prolactin rescue of self-engaged MRL/lpr immature B cells. These results support a mechanism in which prolactin participates in the emergence of lupus through the rescue of self-reactive immature B cell clones from central tolerance clonal deletion through the activation of STAT3 and transcriptional regulation of a complex network of genes related to apoptosis resistance.
Collapse
Affiliation(s)
- Rocio Flores-Fernández
- UIM en Inmunologia, Hospital de Pediatría, CMN SIGLO XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (R.F.-F.); (L.C.-S.); (F.B.-F.)
| | - Angélica Aponte-López
- Unidad de Investigación en Virología y Cáncer, Hospital Infantil de Mexico Federico Gómez, Mexico City 06720, Mexico; (A.A.-L.); (M.C.S.-A.)
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de Mexico, Mexico City 04510, Mexico
| | - Mayra C. Suárez-Arriaga
- Unidad de Investigación en Virología y Cáncer, Hospital Infantil de Mexico Federico Gómez, Mexico City 06720, Mexico; (A.A.-L.); (M.C.S.-A.)
- Laboratorio de Biotecnología y Bioinformática Genómica, ENCB, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Patricia Gorocica-Rosete
- Departamento de Investigación en Bioquímica, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosió Villegas”, Mexico City 14080, Mexico;
| | - Alberto Pizaña-Venegas
- Unidad de Investigación y Bioterio, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosió Villegas”, Mexico City 14080, Mexico;
| | - Luis Chávez-Sanchéz
- UIM en Inmunologia, Hospital de Pediatría, CMN SIGLO XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (R.F.-F.); (L.C.-S.); (F.B.-F.)
| | - Francico Blanco-Favela
- UIM en Inmunologia, Hospital de Pediatría, CMN SIGLO XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (R.F.-F.); (L.C.-S.); (F.B.-F.)
| | - Ezequiel M. Fuentes-Pananá
- Unidad de Investigación en Virología y Cáncer, Hospital Infantil de Mexico Federico Gómez, Mexico City 06720, Mexico; (A.A.-L.); (M.C.S.-A.)
- Correspondence: or (E.M.F.-P.); or (A.K.C.-R.); Tel.: +52-5544349663 (E.M.F.-P.); +52-555627694 (A.K.C.-R.)
| | - Adriana K. Chávez-Rueda
- UIM en Inmunologia, Hospital de Pediatría, CMN SIGLO XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (R.F.-F.); (L.C.-S.); (F.B.-F.)
- Correspondence: or (E.M.F.-P.); or (A.K.C.-R.); Tel.: +52-5544349663 (E.M.F.-P.); +52-555627694 (A.K.C.-R.)
| |
Collapse
|
19
|
Bhore N, Wang BJ, Wu PF, Lee YL, Chen YW, Hsu WM, Lee H, Huang YS, Yang DI, Liao YF. Dual-Specificity Phosphatase 15 (DUSP15) Modulates Notch Signaling by Enhancing the Stability of Notch Protein. Mol Neurobiol 2021; 58:2204-2214. [PMID: 33417224 DOI: 10.1007/s12035-020-02254-0] [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: 03/24/2020] [Accepted: 12/08/2020] [Indexed: 11/27/2022]
Abstract
Dual-specificity phosphatases (DUSPs) comprise a unique group of enzymes that dephosphorylate signaling proteins at both phospho-serine/threonine and phospho-tyrosine residues. Since Notch signaling is an essential pathway for neuronal cell fate determination and development that is also upregulated in Alzheimer's disease tissues, we sought to explore whether and how DUSPs may impact Notch processing. Our results show that overexpression of DUSP15 concomitantly and dose-dependently increased the steady-state levels of recombinant Notch (extracellular domain-truncated Notch, NotchΔE) protein and its cleaved product, Notch intracellular domain (NICD). The overall ratio of NotchΔE to NICD was unchanged by overexpression of DUSP15, suggesting that the effect is independent of γ-secretase. Interestingly, overexpression of DUSP15 also dose-dependently increased phosphorylated ERK1/2. Phosphorylated ERK1/2 is known to be positively correlated with Notch protein level, and we found that DUSP15-mediated regulation of Notch was dependent on ERK1/2 activity. Together, our findings reveal the existence of a previously unidentified DUSP15-ERK1/2-Notch signaling axis, which could potentially play a role in neuronal differentiation and neurological disease.
Collapse
Affiliation(s)
- Noopur Bhore
- Laboratory of Molecular Neurobiology, Institute of Cellular and Organismic Biology, Academia Sinica, ICOB 238, 128 Sec. 2 Academia Rd, Taipei, 11529, Taiwan.,Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Yang-Ming University, Academia Sinica, Taipei, Taiwan
| | - Bo-Jeng Wang
- Laboratory of Molecular Neurobiology, Institute of Cellular and Organismic Biology, Academia Sinica, ICOB 238, 128 Sec. 2 Academia Rd, Taipei, 11529, Taiwan
| | - Po-Fan Wu
- Laboratory of Molecular Neurobiology, Institute of Cellular and Organismic Biology, Academia Sinica, ICOB 238, 128 Sec. 2 Academia Rd, Taipei, 11529, Taiwan.,Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Cheng Kung University, Academia Sinica, Taipei, Taiwan
| | - Yen-Lurk Lee
- TIGP in Molecular Medicine, National Yang-Ming University, Academia Sinica, Taipei, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yun-Wen Chen
- Laboratory of Molecular Neurobiology, Institute of Cellular and Organismic Biology, Academia Sinica, ICOB 238, 128 Sec. 2 Academia Rd, Taipei, 11529, Taiwan
| | - Wen-Ming Hsu
- Department of Surgery, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Hsinyu Lee
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Yi-Shuian Huang
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Yang-Ming University, Academia Sinica, Taipei, Taiwan.,Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Cheng Kung University, Academia Sinica, Taipei, Taiwan.,TIGP in Molecular Medicine, National Yang-Ming University, Academia Sinica, Taipei, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Ding-I Yang
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Yang-Ming University, Academia Sinica, Taipei, Taiwan.,Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan
| | - Yung-Feng Liao
- Laboratory of Molecular Neurobiology, Institute of Cellular and Organismic Biology, Academia Sinica, ICOB 238, 128 Sec. 2 Academia Rd, Taipei, 11529, Taiwan. .,Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Yang-Ming University, Academia Sinica, Taipei, Taiwan. .,Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Cheng Kung University, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
20
|
Ramírez-de-Arellano A, Villegas-Pineda JC, Hernández-Silva CD, Pereira-Suárez AL. The Relevant Participation of Prolactin in the Genesis and Progression of Gynecological Cancers. Front Endocrinol (Lausanne) 2021; 12:747810. [PMID: 34745013 PMCID: PMC8566755 DOI: 10.3389/fendo.2021.747810] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/28/2021] [Indexed: 12/28/2022] Open
Abstract
Prolactin (PRL) is a hormone produced by the pituitary gland and multiple non-pituitary sites, vital in several physiological processes such as lactation, pregnancy, cell growth, and differentiation. However, PRL is nowadays known to have a strong implication in oncogenic processes, making it essential to delve into the mechanisms governing these actions. PRL and its receptor (PRLR) activate a series of effects such as survival, cellular proliferation, migration, invasion, metastasis, and resistance to treatment, being highly relevant in developing certain types of cancer. Because women produce high levels of PRL, its influence in gynecological cancers is herein reviewed. It is interesting that, other than the 23 kDa PRL, whose mechanism of action is endocrine, other variants of PRL have been observed to be produced by tumoral tissue, acting in a paracrine/autocrine manner. Because many components, including PRL, surround the microenvironment, it is interesting to understand the hormone's modulation in cancer cells. This work aims to review the most important findings regarding the PRL/PRLR axis in cervical, ovarian, and endometrial cancers and its molecular mechanisms to support carcinogenesis.
Collapse
Affiliation(s)
- Adrián Ramírez-de-Arellano
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - Julio César Villegas-Pineda
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
- Doctorado en Ciencias Biomédicas, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - Christian David Hernández-Silva
- Doctorado en Ciencias Biomédicas, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - Ana Laura Pereira-Suárez
- Doctorado en Ciencias Biomédicas, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
- Departamento de Microbiología y Patología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
- *Correspondence: Ana Laura Pereira-Suárez,
| |
Collapse
|
21
|
Liu Q, Yang X, Long G, Hu Y, Gu Z, Boisclair YR, Long Q. ERAD deficiency promotes mitochondrial dysfunction and transcriptional rewiring in human hepatic cells. J Biol Chem 2020; 295:16743-16753. [PMID: 32978261 PMCID: PMC7864069 DOI: 10.1074/jbc.ra120.013987] [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: 04/21/2020] [Revised: 09/15/2020] [Indexed: 12/19/2022] Open
Abstract
Mitochondrial dysfunction is associated with a variety of human diseases including neurodegeneration, diabetes, nonalcohol fatty liver disease (NAFLD), and cancer, but its underlying causes are incompletely understood. Using the human hepatic cell line HepG2 as a model, we show here that endoplasmic reticulum-associated degradation (ERAD), an ER protein quality control process, is critically required for mitochondrial function in mammalian cells. Pharmacological inhibition or genetic ablation of key proteins involved in ERAD increased cell death under both basal conditions and in response to proinflammatory cytokines, a situation frequently found in NAFLD. Decreased viability of ERAD-deficient HepG2 cells was traced to impaired mitochondrial functions including reduced ATP production, enhanced reactive oxygen species (ROS) accumulation, and increased mitochondrial outer membrane permeability. Transcriptome profiling revealed widespread down-regulation of genes underpinning mitochondrial functions, and up-regulation of genes associated with tumor growth and aggression. These results highlight a critical role for ERAD in maintaining mitochondrial functional and structural integrity and raise the possibility of improving cellular and organismal mitochondrial function via enhancing cellular ERAD capacity.
Collapse
Affiliation(s)
- Qingqing Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Xiaoqin Yang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Guangyu Long
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Yabing Hu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Zhenglong Gu
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA
| | - Yves R Boisclair
- Department of Animal Science, Cornell University, College of Agriculture and Life Sciences, Ithaca, New York, USA
| | - Qiaoming Long
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Medical College of Soochow University, Suzhou, Jiangsu Province, China.
| |
Collapse
|
22
|
Zheng W, Li J, Zhou X, Cui L, Wang Y. The lncRNA XIST promotes proliferation, migration and invasion of gastric cancer cells by targeting miR-337. Arab J Gastroenterol 2020; 21:199-206. [PMID: 32830093 DOI: 10.1016/j.ajg.2020.07.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 07/29/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND STUDY AIMS Gastric cancer (GC) is one of the most common malignant tumours worldwide. Long non-coding RNAs (lncRNAs) and microRNAs regulate the occurrence and development of various cancers and play an important role in GC progression. X-inactive specific transcript (XIST), a carcinogenic lncRNA, is involved in human tumourigenesis and is altered in GC. Janus kinase 2 (JAK2), a transcription factor, is involved in cancer cell metastasis and differentiation. However, the exact mechanism underlying the biological roles of XIST and JAK2 in cancer cells remains unclear. MATERIAL AND METHODS This study was conducted using GES-1, HGC-27, AGS and HEK-293 T cells. Quantitative polymerase chain reaction and western blotting were performed to detect XIST, microRNA-337 (miR-337) and JAK2 expressions. GC cell invasion was investigated by using the Transwell assay. Fluorescein reporter gene detection was used to determine the relationship between JAK2 and XIST. RESULTS Compared with that in GES-1 cells, XIST expression was significantly up-regulated in AGS and HGC-27 cells. miR-337 expression in GC cell lines was decreased. The proliferation, invasion and migration of GC cells were simultaneously inhibited by XIST knockdown, and the relationship between XIST and miR-337 was confirmed by bioinformatics analysis. JAK2 is expected to be the target gene of miR-337. MiR-337 can negatively regulate JAK2 expression in vitro. In addition, si-XIST decreased JAK2 expression by up-regulating miR-337 in vitro, thereby inhibiting GC cell proliferation and migration. Therefore, we speculated that XIST regulates JAK2 by competing with miR-337 as a competitive endogenous lncRNA in GC. CONCLUSION We elucidated the effects of migration and invasion after XIST inhibition, at least in part, by inhibiting miR-337 expression in GC cells to regulate JAK2. These data indicate that a positive feedback loop exists between XIST and JAK2 and suggest that JAK2 and XIST play a vital role in cancer cell migration and invasion.
Collapse
Affiliation(s)
- Wenqi Zheng
- Health Management Center, East Hospital, Qingdao Municipal Hospital, Qingdao 266071, China
| | - Juan Li
- Health Management Center, East Hospital, Qingdao Municipal Hospital, Qingdao 266071, China
| | - Xue Zhou
- Nursing Department, East Hospital, Qingdao Municipal Hospital, Qingdao 266071, China
| | - Li Cui
- Health Management Center, East Hospital, Qingdao Municipal Hospital, Qingdao 266071, China
| | - Yunfang Wang
- Health Care Department(Ⅰ), East Hospital, Qingdao Municipal Hospital, Qingdao 266071, China.
| |
Collapse
|
23
|
List EO, Berryman DE, Basu R, Buchman M, Funk K, Kulkarni P, Duran-Ortiz S, Qian Y, Jensen EA, Young JA, Yildirim G, Yakar S, Kopchick JJ. The Effects of 20-kDa Human Placental GH in Male and Female GH-deficient Mice: An Improved Human GH? Endocrinology 2020; 161:5859553. [PMID: 32556100 PMCID: PMC7375802 DOI: 10.1210/endocr/bqaa097] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 06/11/2020] [Indexed: 11/19/2022]
Abstract
A rare 20K isoform of GH-V (here abbreviated as GHv) was discovered in 1998. To date, only 1 research article has characterized this isoform in vivo, observing that GHv treatment in male high-fat fed rats had several GH-like activities, but unlike GH lacked diabetogenic and lactogenic activities and failed to increase IGF-1 or body length. Therefore, the current study was conducted to further characterize the in vivo activities of GHv in a separate species and in a GH-deficient model (GH-/- mice) and with both sexes represented. GHv-treated GH-/- mice had significant increases to serum IGF-1, femur length, body length, body weight, and lean body mass and reduced body fat mass similar to mice receiving GH treatment. GH treatment increased circulating insulin levels and impaired insulin sensitivity; in contrast, both measures were unchanged in GHv-treated mice. Since GHv lacks prolactin receptor (PRLR) binding activity, we tested the ability of GH and GHv to stimulate the proliferation of human cancer cell lines and found that GHv has a decreased proliferative response in cancers with high PRLR. Our findings demonstrate that GHv can stimulate insulin-like growth factor-1 and subsequent longitudinal body growth in GH-deficient mice similar to GH, but unlike GH, GHv promoted growth without inhibiting insulin action and without promoting the growth of PRLR-positive cancers in vitro. Thus, GHv may represent improvements to current GH therapies especially for individuals at risk for metabolic syndrome or PRLR-positive cancers.
Collapse
Affiliation(s)
- Edward O List
- Edison Biotechnology Institute, Ohio University, Athens, Ohio
- Department of Specialty Medicine, Heritage College of Osteopathic Medicine, Athens, Ohio
- Correspondence: Edward O. List, PhD, Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701. E-mail:
| | - Darlene E Berryman
- Edison Biotechnology Institute, Ohio University, Athens, Ohio
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Athens, Ohio
| | - Reetobrata Basu
- Edison Biotechnology Institute, Ohio University, Athens, Ohio
| | - Mathew Buchman
- Edison Biotechnology Institute, Ohio University, Athens, Ohio
| | - Kevin Funk
- Edison Biotechnology Institute, Ohio University, Athens, Ohio
| | | | | | - Yanrong Qian
- Edison Biotechnology Institute, Ohio University, Athens, Ohio
| | | | | | - Gozde Yildirim
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, New York
| | - Shoshana Yakar
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, New York
| | - John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, Ohio
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Athens, Ohio
| |
Collapse
|
24
|
Dandawate P, Kaushik G, Ghosh C, Standing D, Sayed AAA, Choudhury S, Subramaniam D, Manzardo A, Banerjee T, Santra S, Ramamoorthy P, Butler M, Padhye SB, Baranda J, Kasi A, Sun W, Tawfik O, Coppola D, Malafa M, Umar S, Soares MJ, Saha S, Weir SJ, Dhar A, Jensen RA, Thomas SM, Anant S. Diphenylbutylpiperidine Antipsychotic Drugs Inhibit Prolactin Receptor Signaling to Reduce Growth of Pancreatic Ductal Adenocarcinoma in Mice. Gastroenterology 2020; 158:1433-1449.e27. [PMID: 31786131 PMCID: PMC7103550 DOI: 10.1053/j.gastro.2019.11.279] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 11/04/2019] [Accepted: 11/19/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Prolactin (PRL) signaling is up-regulated in hormone-responsive cancers. The PRL receptor (PRLR) is a class I cytokine receptor that signals via the Janus kinase (JAK)-signal transducer and activator of transcription and mitogen-activated protein kinase pathways to regulate cell proliferation, migration, stem cell features, and apoptosis. Patients with pancreatic ductal adenocarcinoma (PDAC) have high plasma levels of PRL. We investigated whether PRLR signaling contributes to the growth of pancreatic tumors in mice. METHODS We used immunohistochemical analyses to compare levels of PRL and PRLR in multitumor tissue microarrays. We used structure-based virtual screening and fragment-based drug discovery to identify compounds likely to bind PRLR and interfere with its signaling. Human pancreatic cell lines (AsPC-1, BxPC-3, Panc-1, and MiaPaCa-2), with or without knockdown of PRLR (clustered regularly interspaced short palindromic repeats or small hairpin RNA), were incubated with PRL or penfluridol and analyzed in proliferation and spheroid formation. C57BL/6 mice were given injections of UNKC-6141 cells, with or without knockdown of PRLR, into pancreas, and tumor development was monitored for 4 weeks, with some mice receiving penfluridol treatment for 21 days. Human pancreatic tumor tissues were implanted into interscapular fat pads of NSG mice, and mice were given injections of penfluridol daily for 28 days. Nude mice were given injections of Panc-1 cells, xenograft tumors were grown for 2 weeks, and mice were then given intraperitoneal penfluridol for 35 days. Tumors were collected from mice and analyzed by histology, immunohistochemistry, and immunoblots. RESULTS Levels of PRLR were increased in PDAC compared with nontumor pancreatic tissues. Incubation of pancreatic cell lines with PRL activated signaling via JAK2-signal transducer and activator of transcription 3 and extracellular signal-regulated kinase, as well as formation of pancospheres and cell migration; these activities were not observed in cells with PRLR knockdown. Pancreatic cancer cells with PRLR knockdown formed significantly smaller tumors in mice. We identified several diphenylbutylpiperidine-class antipsychotic drugs as agents that decreased PRL-induced JAK2 signaling; incubation of pancreatic cancer cells with these compounds reduced their proliferation and formation of panco spheres. Injections of 1 of these compounds, penfluridol, slowed the growth of xenograft tumors in the different mouse models, reducing proliferation and inducing autophagy of the tumor cells. CONCLUSIONS Levels of PRLR are increased in PDAC, and exposure to PRL increases proliferation and migration of pancreatic cancer cells. Antipsychotic drugs, such as penfluridol, block PRL signaling in pancreatic cancer cells to reduce their proliferation, induce autophagy, and slow the growth of xenograft tumors in mice. These drugs might be tested in patients with PDAC.
Collapse
Affiliation(s)
- Prasad Dandawate
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160
| | - Gaurav Kaushik
- Department of Surgery, University of Kansas Medical Center, Kansas City, KS 66160
| | - Chandrayee Ghosh
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160
| | - David Standing
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160
| | - Afreen Asif Ali Sayed
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160
| | - Sonali Choudhury
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160
| | | | - Ann Manzardo
- Department of Psychiatry and Behavioral Sciences, University of Kansas Medical Center, Kansas City, KS 66160
| | - Tuhina Banerjee
- Department of Chemistry, Pittsburg State University, Pittsburg, KS 66762, USA
| | - Santimukul Santra
- Department of Chemistry, Pittsburg State University, Pittsburg, KS 66762, USA
| | - Prabhu Ramamoorthy
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160
| | - Merlin Butler
- Department of Psychiatry and Behavioral Sciences, University of Kansas Medical Center, Kansas City, KS 66160
| | - Subhash B. Padhye
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, Interdisciplinary Science and Technology Research Academy, Abeda Inamdar College, University of Pune, Pune 411001
| | - Joaquina Baranda
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160
| | - Anup Kasi
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160
| | - Weijing Sun
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160
| | - Ossama Tawfik
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160
| | - Domenico Coppola
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612
| | - Mokenge Malafa
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612
| | - Shahid Umar
- Department of Surgery, University of Kansas Medical Center, Kansas City, KS 66160
| | - Michael J. Soares
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS 66160, Department of Pediatrics, University of Kansas Medical Center, Kansas City, KS 66160, Center for Perinatal Research, Children’s Research Institute, Children’s Mercy-Kansas City, MO 64108
| | - Subhrajit Saha
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, KS 66160
| | - Scott J. Weir
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160
| | - Animesh Dhar
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160
| | - Roy A. Jensen
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160
| | - Sufi Mary Thomas
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, Department of Otolaryngology, University of Kansas Medical Center, Kansas City, KS 66160
| | - Shrikant Anant
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas; Department of Surgery, University of Kansas Medical Center, Kansas City, Kansas; Interdisciplinary Science and Technology Research Academy, Abeda Inamdar College, University of Pune, Pune.
| |
Collapse
|
25
|
Abstract
Hyperprolactinaemia is one of the most common problems in clinical endocrinology. It relates with various aetiologies (physiological, pharmacological, pathological), the clarification of which requires careful history taking and clinical assessment. Analytical issues (presence of macroprolactin or of the hook effect) need to be taken into account when interpreting the prolactin values. Medications and sellar/parasellar masses (prolactin secreting or acting through “stalk effect”) are the most common causes of pathological hyperprolactinaemia. Hypogonadism and galactorrhoea are well-recognized manifestations of prolactin excess, although its implications on bone health, metabolism and immune system are also expanding. Treatment mainly aims at restoration and maintenance of normal gonadal function/fertility, and prevention of osteoporosis; further specific management strategies depend on the underlying cause. In this review, we provide an update on the diagnostic and management approaches for the patient with hyperprolactinaemia and on the current data looking at the impact of high prolactin on metabolism, cardiovascular and immune systems.
Collapse
|
26
|
Basu R, Kopchick JJ. The effects of growth hormone on therapy resistance in cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2019; 2:827-846. [PMID: 32382711 PMCID: PMC7204541 DOI: 10.20517/cdr.2019.27] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pituitary derived and peripherally produced growth hormone (GH) is a crucial mediator of longitudinal growth, organ development, metabolic regulation with tissue specific, sex specific, and age-dependent effects. GH and its cognate receptor (GHR) are expressed in several forms of cancer and have been validated as an anti-cancer target through a large body of in vitro, in vivo and epidemiological analyses. However, the underlying molecular mechanisms of GH action in cancer prognosis and therapeutic response had been sparse until recently. This review assimilates the critical details of GH-GHR mediated therapy resistance across different cancer types, distilling the therapeutic implications based on our current understanding of these effects.
Collapse
Affiliation(s)
- Reetobrata Basu
- Ohio University Heritage College of Osteopathic Medicine (OU-HCOM), Ohio University, Athens, OH 45701, USA.,Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA
| | - John J Kopchick
- Ohio University Heritage College of Osteopathic Medicine (OU-HCOM), Ohio University, Athens, OH 45701, USA.,Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA
| |
Collapse
|
27
|
Shaji SK, Sunilkumar D, Mahalakshmi NV, Kumar GB, Nair BG. Analysis of microarray data for identification of key microRNA signatures in glioblastoma multiforme. Oncol Lett 2019; 18:1938-1948. [PMID: 31423264 PMCID: PMC6614686 DOI: 10.3892/ol.2019.10521] [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: 12/15/2018] [Accepted: 06/06/2019] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most malignant types of glioma known for its reduced survival rate and rapid relapse. Previous studies have shown that the expression patterns of different microRNAs (miRNA/miR) play a crucial role in the development and progression of GBM. In order to identify potential miRNA signatures of GBM for prognostic and therapeutic purposes, we downloaded and analyzed two expression data sets from Gene Expression Omnibus profiling miRNA patterns of GBM compared with normal brain tissues. Validated targets of the deregulated miRNAs were identified using MirTarBase, and were mapped to Search Tool for the Retrieval of Interacting Genes/Proteins, Database for Annotation, Visualization and Integrated Discovery and Kyoto Encyclopedia of Genes and Genomes databases in order to construct interaction networks and identify enriched pathways of target genes. A total of 6 miRNAs were found to be deregulated in both expression datasets studied. Pathway analysis demonstrated that most of the target genes were enriched in signaling cascades connected to cancer development, such as ‘Pathways in cancer’, ‘Focal adhesion’ and ‘PI3K-Akt signaling pathway’. Of the five target genes that were enriched in the glioblastoma pathway, in the WikiPathway database, both HRas proto-oncogene, GTPase and MET proto-oncogene, receptor tyrosine kinase target genes of hsa-miR-139-5p, were found to be significantly associated with patient survival. The present study may thus form the basis for further exploration of hsa-miR-139-5p, not only as a therapeutic agent, but also as a diagnostic biomarker for GBM as well as a predictive marker for patient survival.
Collapse
Affiliation(s)
- Sanu K Shaji
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, Kerala 690525, India
| | - Damu Sunilkumar
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, Kerala 690525, India
| | - N V Mahalakshmi
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, Kerala 690525, India
| | - Geetha B Kumar
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, Kerala 690525, India
| | - Bipin G Nair
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, Kerala 690525, India
| |
Collapse
|
28
|
Tao M, Shi Y, Tang L, Wang Y, Fang L, Jiang W, Lin T, Qiu A, Zhuang S, Liu N. Blockade of ERK1/2 by U0126 alleviates uric acid-induced EMT and tubular cell injury in rats with hyperuricemic nephropathy. Am J Physiol Renal Physiol 2019; 316:F660-F673. [PMID: 30648910 DOI: 10.1152/ajprenal.00480.2018] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Extracellular signal-regulated kinases 1 and 2 (ERK1/2) are serine/threonine kinases and function as regulators of cellular proliferation and differentiation. Recently, we demonstrated that inhibition of ERK1/2 alleviates the development and progression of hyperuricemia nephropathy (HN). However, its potential roles in uric acid-induced tubular epithelial-mesenchymal transition (EMT) and tubular epithelial cell injury are unknown. In this study, we showed that hyperuricemic injury induced EMT as characterized by downregulation of E-cadherin and upregulation of vimentin and Snail1 in a rat model of HN. This was coincident with epithelial cells arrested at the G2/M phase of cell cycle, activation of Notch1/Jagged-1 and Wnt/β-catenin signaling pathways, and upregulation of matrix metalloproteinase-2 (MMP-2) and MMP-9. Administration of U0126, a selective inhibitor of ERK1/2, blocked all these responses. U0126 was also effective in inhibiting renal tubular cell injury, as shown by decreased expression of lipocalin-2 and kidney injury molecule-1 and active forms of caspase-3. U0126 or ERK1/2 siRNA can inhibit tubular cell EMT and cell apoptosis as characterized with decreased expression of cleaved caspase-3. Moreover, ERK1/2 inhibition suppressed hyperuricemic injury-induced oxidative stress as indicated by decreased malondialdehyde and increased superoxide dismutase. Collectively, ERK1/2 inhibition-elicited renal protection is associated with inhibition of EMT through inactivation of multiple signaling pathways and matrix metalloproteinases, as well as attenuation of renal tubule injury by enhancing cellular resistance to oxidative stress.
Collapse
Affiliation(s)
- Min Tao
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine , Shanghai , China
| | - Yingfeng Shi
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine , Shanghai , China
| | - Lunxian Tang
- Emergency Department of Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine , Shanghai , China
| | - Yi Wang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine , Shanghai , China
| | - Lu Fang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine , Shanghai , China
| | - Wei Jiang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine , Shanghai , China
| | - Tao Lin
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine , Shanghai , China
| | - Andong Qiu
- School of Life Science and Technology, Advanced Institute of Translational Medicine, Tongji University , Shanghai , China
| | - Shougang Zhuang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine , Shanghai , China.,Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University , Providence, Rhode Island
| | - Na Liu
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine , Shanghai , China
| |
Collapse
|
29
|
Li D, Wang G, Jin G, Yao K, Zhao Z, Bie L, Guo Y, Li N, Deng W, Chen X, Chen B, Liu Y, Luo S, Guo Z. Resveratrol suppresses colon cancer growth by targeting the AKT/STAT3 signaling pathway. Int J Mol Med 2018; 43:630-640. [PMID: 30387805 DOI: 10.3892/ijmm.2018.3969] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 10/30/2018] [Indexed: 11/06/2022] Open
Abstract
Colon cancer is a common type of cancer worldwide and accounts for a significant number of cancer‑related deaths. Although surgical techniques and treatment strategies for colon cancer have advanced over the past two decades, the prognosis has not improved considerably. Resveratrol, a natural stilbene compound, possesses antioxidant, cardioprotective and anticancer properties. However, the role of resveratrol in colon cancer has not been fully elucidated. The present study demonstrated that resveratrol inhibited cell proliferation and colony growth in DLD1 and HCT15 colon cancer cells, but did not affect normal colon epithelial cells. The resveratrol‑mediated inhibition of cell proliferation correlated with an induction of apoptosis and with G1 phase cell cycle arrest in colon cancer cells. Additionally, resveratrol treatment decreased the protein expression levels of cyclin D1, cyclin E2 and BCL2 apoptosis regulator, while it increased BCL2 associated X and tumor protein p53, all of which are involved in the regulation of cell cycle and apoptosis. Notably, the results obtained from in silico computational screening identified AKT serine/threonine kinase 1 (AKT1) and AKT2 as novel targets of resveratrol. Computational docking suggested that there are three or four possible hydrogen bonds in the active pocket of AKT1 and AKT2 that contribute to the mode of action of resveratrol. The present study confirmed that resveratrol bound to AKT1 and AKT2 with a pull‑down assay. Furthermore, knockdown of AKT1 and AKT2 inhibited cell proliferation and colony growth, by attenuating cell cycle progression and increasing apoptosis in colon cancer cells, effects that were similar to those caused by resveratrol treatment. Taken together, the present results suggest that the targeting effects of resveratrol to AKT1 and AKT2 may be a potent strategy for chemoprevention or therapy for colon cancer.
Collapse
Affiliation(s)
- Dan Li
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| | - Gangcheng Wang
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| | - Guoguo Jin
- Laboratory of Bone Tumor, Henan Luoyang Orthopedic Hospital, Zhengzhou, Henan 450000, P.R. China
| | - Ke Yao
- China‑US (Henan) Hormel Cancer Institute, Zhengzhou, Henan 450003, P.R. China
| | - Zhenjiang Zhao
- Laboratory of Bone Tumor, Henan Luoyang Orthopedic Hospital, Zhengzhou, Henan 450000, P.R. China
| | - Liangyu Bie
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| | - Yongjun Guo
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| | - Ning Li
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| | - Wenying Deng
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| | - Xiaobin Chen
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| | - Beibei Chen
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| | - Yuanyuan Liu
- Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan 450007, P.R. China
| | - Suxia Luo
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| | - Zhiping Guo
- Fu Wai Hua Zhong Vascular Disease Hospital, Zhengzhou, Henan 450018, P.R. China
| |
Collapse
|
30
|
Serum angiogenesis profile in gestational trophoblastic neoplasm using multiplex immunoassay. Life Sci 2018; 211:25-30. [PMID: 30195618 DOI: 10.1016/j.lfs.2018.08.070] [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] [Received: 06/19/2018] [Revised: 08/22/2018] [Accepted: 08/30/2018] [Indexed: 11/23/2022]
Abstract
AIMS Gestational trophoblastic neoplasms (GTN) exemplify a rare, mostly curable but highly aggressive disease. It is often associated with a rapid formation of distant metastases and most likely with an intense neoangiogenesis processes. The aim of the study was to analyze markers in serum of patients with GTN before chemotherapy compared to healthy pregnant women. MAIN METHODS In this study sixteen protein angiogenesis markers were evaluated in serum of 21 patients with GTN before chemotherapy and compared with healthy pregnant women. Markers were measured using BioPlex Pro Human Cancer Biomarker Panel 1 immunoassay. t-Tests and receiver operating characteristic curves were used for statistical analysis. KEY FINDINGS Receiver operator curve analysis identified six proteins (sTIE-2, osteopontin, sIL-6α, sVEGFR-2, sEGFR, PECAM-1) which had sufficient sensitivity and specificity (AUC > 0,70) to distinguish GTN patients before the treatment from pregnant controls. The levels of three proteins (sTIE-2, osteopontin and sIL-6α) were altered in GTN patients before the treatment as compared to healthy controls (p = 0,0112; p = 0,0442; p = 0,0488, respectively) and thereby may serve as potential disease markers. SIGNIFICANCE Serum concentration of proteins related to angiogenesis changes in the course of GTN and may appear useful in the diagnostic process of this disease.
Collapse
|
31
|
Aronchik I, Dai Y, Labenski M, Barnes C, Jones T, Qiao L, Beebe L, Malek M, Elis W, Shi T, Mavrommatis K, Bray GL, Filvaroff EH. Efficacy of a Covalent ERK1/2 Inhibitor, CC-90003, in KRAS-Mutant Cancer Models Reveals Novel Mechanisms of Response and Resistance. Mol Cancer Res 2018; 17:642-654. [DOI: 10.1158/1541-7786.mcr-17-0554] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 05/31/2018] [Accepted: 09/21/2018] [Indexed: 11/16/2022]
|
32
|
Gao S, Hu J, Wu X, Liang Z. PMA treated THP-1-derived-IL-6 promotes EMT of SW48 through STAT3/ERK-dependent activation of Wnt/β-catenin signaling pathway. Biomed Pharmacother 2018; 108:618-624. [PMID: 30243096 DOI: 10.1016/j.biopha.2018.09.067] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 09/09/2018] [Accepted: 09/11/2018] [Indexed: 12/23/2022] Open
Abstract
Colon cancer is one of the most common digestive malignant tumors that leads to high mortality worldwide, and metastasis is the primary cause of cancer-related death. It is well accepted that the epithelial-mesenchymal transition (EMT) plays a key role in the process of metastasis. As a cytokine that macrophage secretes, IL-6 is involved in the progression of tumors, including the invasion and metastasis via kinds of signaling pathways. However, the mechanism of interactions between IL-6, macrophage, EMT and colon cancer is not fully understood. Increased CD68+ macrophages and IL-6 level were found in colon tumor as compared to normal colon tissue. Metastatic lymph node showed even more CD68+ macrophages and higher IL-6 level than the primary tumor. These results suggested that macrophages and IL-6 play an important role in EMT of colon cancer. In order to investigate the effect of macrophage and IL-6 on EMT of colon cancer, we cultured human colon carcinoma cell line SW48 with conditioned medium (CM) from PMA-stimulated monocyte THP-1 cells and tested for IL-6 dependent EMT pathways. Wound healing assay and Transwell assay were used to analyze cell migration and invasion. Results showed that CM-treated SW48 cells increased IL-6 production and displayed elevated capacity of migration and invasion compared to untreated cells. Increased expressions of EMT markers (N-cadherin, Vimentin and β-catenin) and decreased expression of EMT marker(E-cadherin) were found in CM-treated SW48 cells by Western Blot. The addition of an anti-IL-6 antibody significantly inhibited the increase of EMT markers (Vimentin and β-catenin) as well as cell migration and invasion, suggesting that IL-6 played a critical role in promoting EMT of CM-treated SW48 cells. In addition, we found that the levels of p-STAT3 and p-ERK increased in CM-treated SW48 compared to untreated cells, which can be reversed by AG490, an inhibitor of JAK. In the meantime, the suppression of JAK-associated signaling pathways caused a decrease of β-catenin. In summary, our study suggested that macrophage-induced IL-6 promotes migration and invasion of colon cancer cell via Wnt/β-catenin pathway in STAT3/ERK-dependent way.
Collapse
Affiliation(s)
- Sikang Gao
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Junwu Hu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiongwen Wu
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhihui Liang
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
33
|
Shan X, Zhang H, Zhang L, Zhou X, Wang T, Zhang J, Shu Y, Zhu W, Wen W, Liu P. Identification of four plasma microRNAs as potential biomarkers in the diagnosis of male lung squamous cell carcinoma patients in China. Cancer Med 2018; 7:2370-2381. [PMID: 29673101 PMCID: PMC6010830 DOI: 10.1002/cam4.1490] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 03/15/2018] [Accepted: 03/20/2018] [Indexed: 12/19/2022] Open
Abstract
Dysregulated microRNAs (miRNAs) in the plasma of patients with lung squamous cell carcinoma (LSCC) might serve as biomarkers for LSCC diagnosis. The expression of miRNAs was performed using quantitative reverse transcription-polymerase chain reaction (qRT-PCR) on the basis of Exiqon panels in the initial screening phase including three male LSCC pool samples and one normal control (NC) pool sample (per 10 samples were pooled as one pool sample). After the training (32 LSCC vs. 31 NCs), the testing (55 LSCC vs. 55 NCs), and the external validation (15 LSCC vs. 15 NCs) stages via qRT-PCR, a four-miRNA signature (miR-181a-5p, miR-21-5p, miR-106a-5p, and miR-93-5p) was identified for LSCC detection. Areas under the receiver operating characteristic (ROC) curve (AUC) of the four-miRNA panel for the training, the testing, and the external validation phases were 0.795, 0.827, and 0.914, respectively. Then, the four miRNAs were explored in LSCC tissue samples (23 LSCC vs. 23 NCs), and their expression was significantly up-regulated. However, none of the four miRNAs found significantly up-regulated in plasma exosomes expect miR-93-5p with borderline significance (16 LSCC vs. 16 NCs). In summary, our study established a four-miRNA peripheral plasma signature, which contributed to diagnosing male LSCC patients in China to a certain degree.
Collapse
Affiliation(s)
- Xia Shan
- Department of OncologyFirst Affiliated Hospital of Nanjing Medical University300 Guangzhou RoadNanjing210029China
- Department of RespirationThe Affiliated Jiangning Hospital of Nanjing Medical UniversityNanjing210000China
| | - Huo Zhang
- Department of OncologyFirst Affiliated Hospital of Nanjing Medical University300 Guangzhou RoadNanjing210029China
| | - Lan Zhang
- Department of OncologyFirst Affiliated Hospital of Nanjing Medical University300 Guangzhou RoadNanjing210029China
- Department of Radiation OncologySuzhou Municipal HospitalSuzhou Cancer CenterThe Affiliated Suzhou Hospital of Nanjing Medical UniversitySuzhouJiangsu215001China
| | - Xin Zhou
- Department of OncologyFirst Affiliated Hospital of Nanjing Medical University300 Guangzhou RoadNanjing210029China
| | - Tongshan Wang
- Department of OncologyFirst Affiliated Hospital of Nanjing Medical University300 Guangzhou RoadNanjing210029China
| | - JinYing Zhang
- Department of OncologyFirst Affiliated Hospital of Nanjing Medical University300 Guangzhou RoadNanjing210029China
| | - Yongqian Shu
- Department of OncologyFirst Affiliated Hospital of Nanjing Medical University300 Guangzhou RoadNanjing210029China
| | - Wei Zhu
- Department of OncologyFirst Affiliated Hospital of Nanjing Medical University300 Guangzhou RoadNanjing210029China
- Department of OncologyThe Affiliated Jiangsu Shengze Hospital of Nanjing Medical UniversityNo.1399 West RoadWujiang DistrictSuzhou 215000China
| | - Wei Wen
- Department of Thoracic SurgeryFirst Affiliated Hospital of Nanjing Medical University300 Guangzhou RoadNanjing 210029China
| | - Ping Liu
- Department of OncologyFirst Affiliated Hospital of Nanjing Medical University300 Guangzhou RoadNanjing210029China
| |
Collapse
|
34
|
Zuo XL, Chen ZQ, Wang JF, Wang JG, Liang LH, Cai J. miR-337-3p suppresses the proliferation and invasion of hepatocellular carcinoma cells through targeting JAK2. Am J Cancer Res 2018; 8:662-674. [PMID: 29736311 PMCID: PMC5934556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 03/16/2018] [Indexed: 06/08/2023] Open
Abstract
Accumulating evidence has indicated that microRNA (miRNA) dysregulation contributes to hepatocellular carcinoma (HCC) progression. miR-337-3p is downregulated in gastric cancer and neuroblastoma; however, its biological function and underlying mechanism in HCC remain unclear. In this study, we showed that the expression level of miR-337-3p was significantly decreased in HCC, and was associated with several clinicopathological characteristics, including tumor multiplicity, histological differentiation, and Barcelona Clinic Liver Cancer stage. Low expression level of miR-337-3p was associated with poor survival outcomes in HCC patients. Upregulation of miR-337-3p suppressed cell proliferation, migration, and invasion in HCC. Dual luciferase assay demonstrated that JAK2 was a direct downstream target of miR-337-3p. JAK2 reintroduction restored the inhibited proliferation, migration, and invasion of miR-337-3p overexpressed HCC cells. miR-337-3p functioned as a tumor suppressor to modulate the JAK2/STAT3 signaling pathway. The present findings indicate that miR-337-3p could be used as a prognostic predictor and therapeutic candidate for HCC.
Collapse
Affiliation(s)
- Xue-Liang Zuo
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical CollegeWuhu 241000, China
| | - Zhi-Qiang Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory on Living Donor Liver Transplantation, National Health and Family Planning CommissionNanjing 210029, China
| | - Jun-Feng Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical CollegeWuhu 241000, China
| | - Jin-Guo Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical CollegeWuhu 241000, China
| | - Lin-Hu Liang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical CollegeWuhu 241000, China
| | - Juan Cai
- Department of Oncology, The First Affiliated Hospital of Wannan Medical CollegeWuhu 241000, China
| |
Collapse
|
35
|
Tarasiuk A, Mosińska P, Fichna J. The mechanisms linking obesity to colon cancer: An overview. Obes Res Clin Pract 2018; 12:251-259. [PMID: 29428365 DOI: 10.1016/j.orcp.2018.01.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Revised: 01/19/2018] [Accepted: 01/26/2018] [Indexed: 12/16/2022]
Abstract
Obesity, characterised as a chronic low-grade inflammation is a crucial risk factor for colon cancer. The expansion of the adipose tissue is related to elevated triglyceride and low-density lipoprotein (LDL) levels and hyperinsulinemia, which all are presumed mediators of the tumour development. Obesity is also believed to support carcinogenesis by activating the insulin/IGF-1 pathway. Moreover, obesity increases the level of proinflammatory cytokines (e.g. TNF-α, IL-1, and IL-6) and has a significant impact on selected adipokines. This paper briefly outlines the latest evidence of the linkage between the obesity and colon cancer and discusses its possible implication for the improvement of anticancer prevention and treatment strategies connected with nutrition.
Collapse
Affiliation(s)
- Aleksandra Tarasiuk
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Poland
| | - Paula Mosińska
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Poland
| | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Poland.
| |
Collapse
|
36
|
Saby C, Buache E, Brassart-Pasco S, El Btaouri H, Courageot MP, Van Gulick L, Garnotel R, Jeannesson P, Morjani H. Type I collagen aging impairs discoidin domain receptor 2-mediated tumor cell growth suppression. Oncotarget 2018; 7:24908-27. [PMID: 27121132 PMCID: PMC5041879 DOI: 10.18632/oncotarget.8795] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 04/10/2016] [Indexed: 12/26/2022] Open
Abstract
Tumor cells are confronted to a type I collagen rich environment which regulates cell proliferation and invasion. Biological aging has been associated with structural changes of type I collagen. Here, we address the effect of collagen aging on cell proliferation in a three-dimensional context (3D). We provide evidence for an inhibitory effect of adult collagen, but not of the old one, on proliferation of human fibrosarcoma HT-1080 cells. This effect involves both the activation of the tyrosine kinase Discoidin Domain Receptor 2 (DDR2) and the tyrosine phosphatase SHP-2. DDR2 and SHP-2 were less activated in old collagen. DDR2 inhibition decreased SHP-2 phosphorylation in adult collagen and increased cell proliferation to a level similar to that observed in old collagen. In the presence of old collagen, a high level of JAK2 and ERK1/2 phosphorylation was observed while expression of the cell cycle negative regulator p21CIP1 was decreased. Inhibition of DDR2 kinase function also led to an increase in ERK1/2 phosphorylation and a decrease in p21CIP1 expression. Similar signaling profile was observed when DDR2 was inhibited in adult collagen. Altogether, these data suggest that biological collagen aging could increase tumor cell proliferation by reducingthe activation of the key matrix sensor DDR2.
Collapse
Affiliation(s)
- Charles Saby
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) Matrice Extracellulaire et Dynamique Cellulaire, Université de Reims Champagne-Ardenne, Unité de Formation et de Recherche (UFR) Pharmacie, Reims, France
| | - Emilie Buache
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) Matrice Extracellulaire et Dynamique Cellulaire, Université de Reims Champagne-Ardenne, Unité de Formation et de Recherche (UFR) Pharmacie, Reims, France
| | - Sylvie Brassart-Pasco
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) Matrice Extracellulaire et Dynamique Cellulaire, Université de Reims Champagne-Ardenne, Unité de Formation et de Recherche (UFR) Médecine, Reims, France
| | - Hassan El Btaouri
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) Matrice Extracellulaire et Dynamique Cellulaire, Université de Reims Champagne-Ardenne, Unité de Formation et de Recherche (UFR) Sciences Exactes et Naturelles, Reims, France
| | - Marie-Pierre Courageot
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) Matrice Extracellulaire et Dynamique Cellulaire, Université de Reims Champagne-Ardenne, Unité de Formation et de Recherche (UFR) Sciences Exactes et Naturelles, Reims, France
| | - Laurence Van Gulick
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) Matrice Extracellulaire et Dynamique Cellulaire, Université de Reims Champagne-Ardenne, Unité de Formation et de Recherche (UFR) Pharmacie, Reims, France
| | - Roselyne Garnotel
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) Matrice Extracellulaire et Dynamique Cellulaire, Université de Reims Champagne-Ardenne, Unité de Formation et de Recherche (UFR) Médecine, Reims, France
| | - Pierre Jeannesson
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) Matrice Extracellulaire et Dynamique Cellulaire, Université de Reims Champagne-Ardenne, Unité de Formation et de Recherche (UFR) Pharmacie, Reims, France
| | - Hamid Morjani
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) Matrice Extracellulaire et Dynamique Cellulaire, Université de Reims Champagne-Ardenne, Unité de Formation et de Recherche (UFR) Pharmacie, Reims, France
| |
Collapse
|
37
|
Wang Z, Zhao K, Hackert T, Zöller M. CD44/CD44v6 a Reliable Companion in Cancer-Initiating Cell Maintenance and Tumor Progression. Front Cell Dev Biol 2018; 6:97. [PMID: 30211160 PMCID: PMC6122270 DOI: 10.3389/fcell.2018.00097] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 08/08/2018] [Indexed: 12/19/2022] Open
Abstract
Metastasis is the leading cause of cancer death, tumor progression proceeding through emigration from the primary tumor, gaining access to the circulation, leaving the circulation, settling in distant organs and growing in the foreign environment. The capacity of a tumor to metastasize relies on a small subpopulation of cells in the primary tumor, so called cancer-initiating cells (CIC). CIC are characterized by sets of markers, mostly membrane anchored adhesion molecules, CD44v6 being the most frequently recovered marker. Knockdown and knockout models accompanied by loss of tumor progression despite unaltered primary tumor growth unraveled that these markers are indispensable for CIC. The unexpected contribution of marker molecules to CIC-related activities prompted research on underlying molecular mechanisms. This review outlines the contribution of CD44, particularly CD44v6 to CIC activities. A first focus is given to the impact of CD44/CD44v6 to inherent CIC features, including the crosstalk with the niche, apoptosis-resistance, and epithelial mesenchymal transition. Following the steps of the metastatic cascade, we report on supporting activities of CD44/CD44v6 in migration and invasion. These CD44/CD44v6 activities rely on the association with membrane-integrated and cytosolic signaling molecules and proteases and transcriptional regulation. They are not restricted to, but most pronounced in CIC and are tightly regulated by feedback loops. Finally, we discuss on the engagement of CD44/CD44v6 in exosome biogenesis, loading and delivery. exosomes being the main acteurs in the long-distance crosstalk of CIC with the host. In brief, by supporting the communication with the niche and promoting apoptosis resistance CD44/CD44v6 plays an important role in CIC maintenance. The multifaceted interplay between CD44/CD44v6, signal transducing molecules and proteases facilitates the metastasizing tumor cell journey through the body. By its engagement in exosome biogenesis CD44/CD44v6 contributes to disseminated tumor cell settlement and growth in distant organs. Thus, CD44/CD44v6 likely is the most central CIC biomarker.
Collapse
Affiliation(s)
- Zhe Wang
- Department of Oncology, First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong, China
| | - Kun Zhao
- Pancreas Section, University Hospital of Surgery, Heidelberg, Germany
| | - Thilo Hackert
- Pancreas Section, University Hospital of Surgery, Heidelberg, Germany
| | - Margot Zöller
- Pancreas Section, University Hospital of Surgery, Heidelberg, Germany
- *Correspondence: Margot Zöller
| |
Collapse
|
38
|
Amani H, Ajami M, Nasseri Maleki S, Pazoki-Toroudi H, Daglia M, Tsetegho Sokeng AJ, Di Lorenzo A, Nabavi SF, Devi KP, Nabavi SM. Targeting signal transducers and activators of transcription (STAT) in human cancer by dietary polyphenolic antioxidants. Biochimie 2017; 142:63-79. [DOI: 10.1016/j.biochi.2017.08.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 08/08/2017] [Indexed: 12/11/2022]
|
39
|
Tao Y, Yang S, Wu Y, Fang X, Wang Y, Song Y, Han T. MicroRNA-216a inhibits the metastasis of gastric cancer cells by targeting JAK2/STAT3-mediated EMT process. Oncotarget 2017; 8:88870-88881. [PMID: 29179483 PMCID: PMC5687653 DOI: 10.18632/oncotarget.21488] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 08/28/2017] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs), a group of small, non-protein coding, endogenous RNAs, play critical roles in the tumorigenesis and progression of human cancer. miR-216a has recently been reported to play an oncogenic role in human cancer. While, the expression of miR-216a, its biological function and underlying molecular mechanisms in gastric cancer (GC) are largely unknown. In this study, we revealed that miR-216a was underexpressed in GC tissues compared to matched noncancerous tissues. Decreased levels of miR-216a were confirmed in GC cell lines compared with a normal gastric epithelium cell line. miR-216a underexpression was associated with malignant prognostic features including lymph node metastasis, venous infiltration, invasive depth and advanced TNM stage. GC patients with low miR-216a level showed an obvious shorter overall survival. miR-216a overexpression restrained migration and invasion of MGC-803 cells, while its knockdown exerted opposite effects on metastatic behaviors of SGC-7901 cells. In vivo experiments found that miR-216a restoration reduced metastatic nodes of GC cells in nude mice liver. miR-216a notably suppressed epithelial-mesenchymal transition (EMT) of GC cells. Janus kinase 2 (JAK2) was recognized as a direct target and downstream mediator of miR-216a in GC cells. Interestingly, JAK2/signal transducer and activator of transcription 3 (STAT3) pathway was prominently inactivated by miR-216a and probably mediated the role of miR-216a in the regulation of migration, invasion and EMT process of GC cells. In conclusion, these data suggest that miR-216a functions as a tumor suppressive miRNA in the development of GC possibly by targeting JAK2/STAT3-mediated EMT.
Collapse
Affiliation(s)
- Youmao Tao
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province 130033, China
| | - Songbai Yang
- Department of Vascular Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province 130033, China
| | - Yuanyu Wu
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province 130033, China
| | - Xuedong Fang
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province 130033, China
| | - Yannan Wang
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province 130033, China
| | - Yan Song
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province 130033, China
| | - Tao Han
- Department of Vascular Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province 130033, China
| |
Collapse
|
40
|
Kahlert UD, Mooney SM, Natsumeda M, Steiger HJ, Maciaczyk J. Targeting cancer stem-like cells in glioblastoma and colorectal cancer through metabolic pathways. Int J Cancer 2016; 140:10-22. [PMID: 27389307 DOI: 10.1002/ijc.30259] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 06/29/2016] [Accepted: 07/04/2016] [Indexed: 12/12/2022]
Abstract
Cancer stem-like cells (CSCs) are thought to be the main cause of tumor occurrence, progression and therapeutic resistance. Strong research efforts in the last decade have led to the development of several tailored approaches to target CSCs with some very promising clinical trials underway; however, until now no anti-CSC therapy has been approved for clinical use. Given the recent improvement in our understanding of how onco-proteins can manipulate cellular metabolic networks to promote tumorigenesis, cancer metabolism research may well lead to innovative strategies to identify novel regulators and downstream mediators of CSC maintenance. Interfering with distinct stages of CSC-associated metabolics may elucidate novel, more efficient strategies to target this highly malignant cell population. Here recent discoveries regarding the metabolic properties attributed to CSCs in glioblastoma (GBM) and malignant colorectal cancer (CRC) were summarized. The association between stem cell markers, the response to hypoxia and other environmental stresses including therapeutic insults as well as developmentally conserved signaling pathways with alterations in cellular bioenergetic networks were also discussed. The recent developments in metabolic imaging to identify CSCs were also summarized. This summary should comprehensively update basic and clinical scientists on the metabolic traits of CSCs in GBM and malignant CRC.
Collapse
Affiliation(s)
- U D Kahlert
- Department of Neurosurgery, Heinrich-Heine University Medical Center, Düsseldorf, Germany
| | - S M Mooney
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - M Natsumeda
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata, Japan
| | - H-J Steiger
- Department of Neurosurgery, Heinrich-Heine University Medical Center, Düsseldorf, Germany
| | - J Maciaczyk
- Department of Neurosurgery, Heinrich-Heine University Medical Center, Düsseldorf, Germany
| |
Collapse
|
41
|
Basu A, Seth S, Chauhan AK, Bansal N, Arora K, Mahaur A. Comparative study of tumor markers in patients with colorectal carcinoma before and after chemotherapy. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:71. [PMID: 27004218 DOI: 10.3978/j.issn.2305-5839.2016.02.02] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND Colorectal carcinoma (CRC), the second leading cause of cancer-related deaths in US, has a rising time-trend in India. Tumour markers in CRC are extensively researched, and there's still debate on their diagnostic and prognostic values. METHODS In this hospital-based longitudinal study in north India, 51 male diagnosed CRC cases (pre-chemotherapy) were contrasted against 50 age and sex matched controls. Nine biomarkers: carcinoembryonic antigen (CEA), prolactin (PRL), alfa feto protein (AFP), total human chorionic gonadotropin (hCG), cancer antigen-125 (CA-125), serum testosterone, prostate specific antigen (PSA) and ferritin were measured by direct chemiluminescence technique. Further, follow-up was done on 47 cases after treatment with six cycles of 5-flurouracil (5-FU) and oxaliplatin. RESULTS Mean serum CEA (case: 5.94±8.27 ng/mL, control: 2.5±0.79 ng/mL, P<0.05), PRL (case: 28.12±13.39 ng/mL, control: 14.24±13.13 ng/mL, P<0.0001), AFP (case: 10.9±6.65 ng/mL, control: 4.02±1.26 ng/mL, P<0.0001) levels were significantly raised in CRC cases compared to controls. On the contrary, mean testosterone level (P<0.05) was lower among the cases. After chemotherapy, the mean serum CEA (P<0.05), AFP (P<0.0001) and CA-125 (P<0.05) levels among the cases decreased significantly compared to their pretreatment levels. CONCLUSIONS The present study strongly indicates the role of CEA, PRL, AFP, CA-125 and testosterone as important biomarkers in male CRC patients from north India. Further, AFP, CA-125 and CEA may be used to assess the effectiveness of chemotherapy in such patients.
Collapse
Affiliation(s)
- Atreyee Basu
- 1 Department of Biochemistry, Post Graduate Institute of Medical Sciences (PGIMS), Rohtak, Haryana, India ; 2 Department of Radiotherapy, Post Graduate Institute of Medical Sciences (PGIMS), Rohtak, Haryana, India ; 3 Department of Biochemistry, VMMC and Safdarjung Hospital, New Delhi, India
| | - Shashi Seth
- 1 Department of Biochemistry, Post Graduate Institute of Medical Sciences (PGIMS), Rohtak, Haryana, India ; 2 Department of Radiotherapy, Post Graduate Institute of Medical Sciences (PGIMS), Rohtak, Haryana, India ; 3 Department of Biochemistry, VMMC and Safdarjung Hospital, New Delhi, India
| | - Ashok K Chauhan
- 1 Department of Biochemistry, Post Graduate Institute of Medical Sciences (PGIMS), Rohtak, Haryana, India ; 2 Department of Radiotherapy, Post Graduate Institute of Medical Sciences (PGIMS), Rohtak, Haryana, India ; 3 Department of Biochemistry, VMMC and Safdarjung Hospital, New Delhi, India
| | - Nupur Bansal
- 1 Department of Biochemistry, Post Graduate Institute of Medical Sciences (PGIMS), Rohtak, Haryana, India ; 2 Department of Radiotherapy, Post Graduate Institute of Medical Sciences (PGIMS), Rohtak, Haryana, India ; 3 Department of Biochemistry, VMMC and Safdarjung Hospital, New Delhi, India
| | - Kanchan Arora
- 1 Department of Biochemistry, Post Graduate Institute of Medical Sciences (PGIMS), Rohtak, Haryana, India ; 2 Department of Radiotherapy, Post Graduate Institute of Medical Sciences (PGIMS), Rohtak, Haryana, India ; 3 Department of Biochemistry, VMMC and Safdarjung Hospital, New Delhi, India
| | - Anuradha Mahaur
- 1 Department of Biochemistry, Post Graduate Institute of Medical Sciences (PGIMS), Rohtak, Haryana, India ; 2 Department of Radiotherapy, Post Graduate Institute of Medical Sciences (PGIMS), Rohtak, Haryana, India ; 3 Department of Biochemistry, VMMC and Safdarjung Hospital, New Delhi, India
| |
Collapse
|
42
|
HES1 in immunity and cancer. Cytokine Growth Factor Rev 2016; 30:113-7. [PMID: 27066918 DOI: 10.1016/j.cytogfr.2016.03.010] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 03/18/2016] [Accepted: 03/18/2016] [Indexed: 01/06/2023]
Abstract
Hairy and enhancer of split homolog-1 (HES1) is a part of an extensive family of basic helix-loop-helix (bHLH) proteins and plays a crucial role in the control and regulation of cell cycle, proliferation, cell differentiation, survival and apoptosis in neuronal, endocrine, T-lymphocyte progenitors as well as various cancers. HES1 is a transcription factor which is regulated by the NOTCH, Hedgehog and Wnt signalling pathways. Aberrant expression of these pathways is a common feature of cancerous cells. There appears to be a fine and complicated crosstalk at the molecular level between the various signalling pathways and HES1, which contributes to its effects on the immune response and cancers such as leukaemia. Several mechanisms have been proposed, including an enhanced invasiveness and metastasis by inducing epithelial mesenchymal transition (EMT), in addition to its strict requirement for tumour cell survival. In this review, we summarize the current biology and molecular mechanisms as well as its use as a clinical target in cancer therapeutics.
Collapse
|
43
|
Kitayama M, Mizutani K, Maruoka M, Mandai K, Sakakibara S, Ueda Y, Komori T, Shimono Y, Takai Y. A Novel Nectin-mediated Cell Adhesion Apparatus That Is Implicated in Prolactin Receptor Signaling for Mammary Gland Development. J Biol Chem 2016; 291:5817-5831. [PMID: 26757815 PMCID: PMC4786717 DOI: 10.1074/jbc.m115.685917] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 01/08/2016] [Indexed: 11/06/2022] Open
Abstract
Mammary gland development is induced by the actions of various hormones to form a structure consisting of collecting ducts and milk-secreting alveoli, which comprise two types of epithelial cells known as luminal and basal cells. These cells adhere to each other by cell adhesion apparatuses whose roles in hormone-dependent mammary gland development remain largely unknown. Here we identified a novel cell adhesion apparatus at the boundary between the luminal and basal cells in addition to desmosomes. This apparatus was formed by the trans-interaction between the cell adhesion molecules nectin-4 and nectin-1, which were expressed in the luminal and basal cells, respectively. Nectin-4 of this apparatus further cis-interacted with the prolactin receptor in the luminal cells to enhance the prolactin-induced prolactin receptor signaling for alveolar development with lactogenic differentiation. Thus, a novel nectin-mediated cell adhesion apparatus regulates the prolactin receptor signaling for mammary gland development.
Collapse
Affiliation(s)
- Midori Kitayama
- From the Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, 1-5-6 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan and; Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology and; Department of Oral and Maxillofacial Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017, Japan
| | - Kiyohito Mizutani
- From the Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, 1-5-6 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan and; Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology and
| | - Masahiro Maruoka
- From the Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, 1-5-6 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan and
| | - Kenji Mandai
- From the Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, 1-5-6 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan and; Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology and
| | - Shotaro Sakakibara
- From the Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, 1-5-6 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan and
| | - Yuki Ueda
- From the Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, 1-5-6 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan and
| | - Takahide Komori
- Department of Oral and Maxillofacial Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017, Japan
| | - Yohei Shimono
- Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology and
| | - Yoshimi Takai
- From the Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, 1-5-6 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan and; Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology and.
| |
Collapse
|
44
|
Xia L, Yin Z, Mao L, Wang X, Liu J, Jiang X, Zhang Z, Lin K, Chang J, Fang B. Akermanite bioceramics promote osteogenesis, angiogenesis and suppress osteoclastogenesis for osteoporotic bone regeneration. Sci Rep 2016; 6:22005. [PMID: 26911441 PMCID: PMC4766478 DOI: 10.1038/srep22005] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 02/05/2016] [Indexed: 11/09/2022] Open
Abstract
It is a big challenge for bone healing under osteoporotic pathological condition with impaired angiogenesis, osteogenesis and remodeling. In the present study, the effect of Ca, Mg, Si containing akermanite bioceramics (Ca2MgSi2O7) extract on cell proliferation, osteogenic differentiation and angiogenic factor expression of BMSCs derived from ovariectomized rats (BMSCs-OVX) as well as the expression of osteoclastogenic factors was evaluated. The results showed that akermanite could enhance cell proliferation, ALP activity, expression of Runx2, BMP-2, BSP, OPN, OCN, OPG and angiogenic factors including VEGF and ANG-1. Meanwhile, akermanite could repress expression of osteoclastogenic factors including RANKL and TNF-α. Moreover, akermanite could activate ERK, P38, AKT and STAT3 signaling pathways, while crosstalk among these signaling pathways was evident. More importantly, the effect of akermanite extract on RANKL-induced osteoclastogenesis was evaluated by TRAP staining and real-time PCR assay. The results showed that akermanite could suppress osteoclast formation and expression of TRAP, cathepsin K and NFATc1. The in vivo experiments revealed that akermanite bioceramics dramatically stimulated osteogenesis and angiogenesis in an OVX rat critical-sized calvarial defect model. All these results suggest that akermanite bioceramics with the effects of Mg and Si ions on osteogenesis, angiogenesis and osteoclastogenesis are promising biomaterials for osteoporotic bone regeneration.
Collapse
Affiliation(s)
- Lunguo Xia
- Center of Craniofacial Orthodontics, Department of Oral and Cranio-maxillofacial Science, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai 200011, China
| | - Zhilan Yin
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
| | - Lixia Mao
- Center of Craniofacial Orthodontics, Department of Oral and Cranio-maxillofacial Science, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai 200011, China
| | - Xiuhui Wang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
| | - Jiaqiang Liu
- Center of Craniofacial Orthodontics, Department of Oral and Cranio-maxillofacial Science, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai 200011, China
| | - Xinquan Jiang
- Oral Bioengineering and regenerative medicine Lab, Shanghai Research Institute of Stomatology, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Zhiyuan Zhang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Kaili Lin
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China.,School &Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai 200072, China
| | - Jiang Chang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
| | - Bing Fang
- Center of Craniofacial Orthodontics, Department of Oral and Cranio-maxillofacial Science, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai 200011, China
| |
Collapse
|
45
|
Masjkur J, Poser SW, Nikolakopoulou P, Chrousos G, McKay RD, Bornstein SR, Jones PM, Androutsellis-Theotokis A. Endocrine Pancreas Development and Regeneration: Noncanonical Ideas From Neural Stem Cell Biology. Diabetes 2016; 65:314-30. [PMID: 26798118 DOI: 10.2337/db15-1099] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Loss of insulin-producing pancreatic islet β-cells is a hallmark of type 1 diabetes. Several experimental paradigms demonstrate that these cells can, in principle, be regenerated from multiple endogenous sources using signaling pathways that are also used during pancreas development. A thorough understanding of these pathways will provide improved opportunities for therapeutic intervention. It is now appreciated that signaling pathways should not be seen as "on" or "off" but that the degree of activity may result in wildly different cellular outcomes. In addition to the degree of operation of a signaling pathway, noncanonical branches also play important roles. Thus, a pathway, once considered as "off" or "low" may actually be highly operational but may be using noncanonical branches. Such branches are only now revealing themselves as new tools to assay them are being generated. A formidable source of noncanonical signal transduction concepts is neural stem cells because these cells appear to have acquired unusual signaling interpretations to allow them to maintain their unique dual properties (self-renewal and multipotency). We discuss how such findings from the neural field can provide a blueprint for the identification of new molecular mechanisms regulating pancreatic biology, with a focus on Notch, Hes/Hey, and hedgehog pathways.
Collapse
Affiliation(s)
- Jimmy Masjkur
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Steven W Poser
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany
| | | | - George Chrousos
- First Department of Pediatrics, University of Athens Medical School and Aghia Sophia Children's Hospital, Athens, Greece
| | | | - Stefan R Bornstein
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Peter M Jones
- Diabetes Research Group, Division of Diabetes & Nutritional Sciences, King's College London, London, U.K
| | - Andreas Androutsellis-Theotokis
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany Center for Regenerative Therapies Dresden, Dresden, Germany Department of Stem Cell Biology, Centre for Biomolecular Sciences, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, U.K.
| |
Collapse
|
46
|
Abstract
Pathway analysis is a common approach to gain insight from biological experiments. Signaling-pathway impact analysis (SPIA) is one such method and combines both the classical enrichment analysis and the actual perturbation on a given pathway. Because this method focuses on a single pathway, its resolution generally is not very high because the differentially expressed genes may be enriched in a local region of the pathway. In the present work, to identify cancer-related pathways, we incorporated a recent subpathway analysis method into the SPIA method to form the “sub-SPIA method.” The original subpathway analysis uses the k-clique structure to define a subpathway. However, it is not sufficiently flexible to capture subpathways with complex structure and usually results in many overlapping subpathways. We therefore propose using the minimal-spanning-tree structure to find a subpathway. We apply this approach to colorectal cancer and lung cancer datasets, and our results show that sub-SPIA can identify many significant pathways associated with each specific cancer that other methods miss. Based on the entire pathway network in the Kyoto Encyclopedia of Genes and Genomes, we find that the pathways identified by sub-SPIA not only have the largest average degree, but also are more closely connected than those identified by other methods. This result suggests that the abnormality signal propagating through them might be responsible for the specific cancer or disease.
Collapse
|
47
|
Goffin V, Touraine P. The prolactin receptor as a therapeutic target in human diseases: browsing new potential indications. Expert Opin Ther Targets 2015; 19:1229-44. [PMID: 26063597 DOI: 10.1517/14728222.2015.1053209] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Prolactin (PRL) signaling has emerged as a relevant target in breast and prostate cancers. This has encouraged various laboratories to develop compounds targeting the PRL receptor (PRLR). As the latter is widely distributed, it is timely to address whether other conditions could also benefit from such inhibitors. AREAS COVERED The authors briefly overview the two classes of PRLR blockers, which involve: i) PRL-core based analogs that have been validated as competitive antagonists in various preclinical models, and ii) anti-PRLR neutralizing antibodies that are currently in clinical Phase I for advanced breast and prostate cancers. The main purpose of this review is to discuss the multiple organs/diseases that may be considered as potential targets/indications for such inhibitors. This is done in light of reports suggesting that PRLR expression/signaling is increased in disease, and/or that systemic or locally elevated PRL levels correlate with (or promote) organ pathogenesis. EXPERT OPINION The two immediate challenges in the field are i) to provide the scientific community with potent anti-prolactin receptor antibodies to map prolactin receptor expression in target organs, and ii) to take advantage of the availability of functionally validated PRLR blockers to establish the relevance of these potential indications in humans.
Collapse
Affiliation(s)
- Vincent Goffin
- Research Director at Inserm, Head of the 'PRL/GH Pathophysiology: Translational Approaches' Laboratory,University Paris Descartes, Institut Necker Enfants Malades (INEM), Inserm Unit 1151, Faculté de Médecine Paris Descartes , Bâtiment Leriche, 14 Rue Maria Helena Vieira Da Silva, CS61431, 75993 Paris Cedex 14 , France +33 1 72 60 63 68 +33 1 72 60 64 01 ;
| | | |
Collapse
|
48
|
An X, Hou J, Gao T, Lei Y, Li G, Song Y, Wang J, Cao B. Single-nucleotide polymorphisms g.151435C>T and g.173057T>C in PRLR gene regulated by bta-miR-302a are associated with litter size in goats. Theriogenology 2015; 83:1477-1483.e1. [DOI: 10.1016/j.theriogenology.2015.01.030] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 01/19/2015] [Accepted: 01/24/2015] [Indexed: 01/22/2023]
|
49
|
LIU HONGLEI, LEI CHUNLING, LONG KEQIN, YANG XINGUANG, ZHU ZHAOLIANG, ZHANG LIHUA, LIU JUN. Mutant GNAQ promotes cell viability and migration of uveal melanoma cells through the activation of Notch signaling. Oncol Rep 2015; 34:295-301. [DOI: 10.3892/or.2015.3949] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 04/03/2015] [Indexed: 11/05/2022] Open
|
50
|
Mahboob S, Ahn SB, Cheruku HR, Cantor D, Rennel E, Fredriksson S, Edfeldt G, Breen EJ, Khan A, Mohamedali A, Muktadir MG, Ranganathan S, Tan SH, Nice E, Baker MS. A novel multiplexed immunoassay identifies CEA, IL-8 and prolactin as prospective markers for Dukes' stages A-D colorectal cancers. Clin Proteomics 2015; 12:10. [PMID: 25987887 PMCID: PMC4435647 DOI: 10.1186/s12014-015-9081-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 03/04/2015] [Indexed: 12/14/2022] Open
Abstract
Background Current methods widely deployed for colorectal cancers (CRC) screening lack the necessary sensitivity and specificity required for population-based early disease detection. Cancer-specific protein biomarkers are thought to be produced either by the tumor itself or other tissues in response to the presence of cancers or associated conditions. Equally, known examples of cancer protein biomarkers (e.g., PSA, CA125, CA19-9, CEA, AFP) are frequently found in plasma at very low concentration (pg/mL-ng/mL). New sensitive and specific assays are therefore urgently required to detect the disease at an early stage when prognosis is good following surgical resection. This study was designed to meet the longstanding unmet clinical need for earlier CRC detection by measuring plasma candidate biomarkers of cancer onset and progression in a clinical stage-specific manner. EDTA plasma samples (1 μL) obtained from 75 patients with Dukes’ staged CRC or unaffected controls (age and sex matched with stringent inclusion/exclusion criteria) were assayed for expression of 92 human proteins employing the Proseek® Multiplex Oncology I proximity extension assay. An identical set of plasma samples were analyzed utilizing the Bio-Plex Pro™ human cytokine 27-plex immunoassay. Results Similar quantitative expression patterns for 13 plasma antigens common to both platforms endorsed the potential efficacy of Proseek as an immune-based multiplex assay for proteomic biomarker research. Proseek found that expression of Carcinoembryonic Antigen (CEA), IL-8 and prolactin are significantly correlated with CRC stage. Conclusions CEA, IL-8 and prolactin expression were found to identify between control (unaffected), non-malignant (Dukes’ A + B) and malignant (Dukes’ C + D) stages. Electronic supplementary material The online version of this article (doi:10.1186/s12014-015-9081-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sadia Mahboob
- Australian School of Advanced Medicine, Faculty of Medicine and Human Sciences, Macquarie University, Rm1, Level 1, 75 Talavera Road, Sydney, NSW 2109 Australia
| | - Seong Beom Ahn
- Australian School of Advanced Medicine, Faculty of Medicine and Human Sciences, Macquarie University, Rm1, Level 1, 75 Talavera Road, Sydney, NSW 2109 Australia
| | - Harish R Cheruku
- Australian School of Advanced Medicine, Faculty of Medicine and Human Sciences, Macquarie University, Rm1, Level 1, 75 Talavera Road, Sydney, NSW 2109 Australia
| | - David Cantor
- Australian School of Advanced Medicine, Faculty of Medicine and Human Sciences, Macquarie University, Rm1, Level 1, 75 Talavera Road, Sydney, NSW 2109 Australia
| | - Emma Rennel
- Olink Bioscience, Dag Hammarskjölds Väg, 54A, 75183 Uppsala, Sweden
| | | | | | - Edmond J Breen
- Australian Proteome Analysis Facility, Macquarie University, Sydney, NSW 2109 Australia
| | - Alamgir Khan
- Australian Proteome Analysis Facility, Macquarie University, Sydney, NSW 2109 Australia
| | - Abidali Mohamedali
- Department of Chemistry and Biomolecular Sciences, Faculty of Science, Macquarie University, Sydney, NSW 2109 Australia
| | - Md Golam Muktadir
- School of Science and Health, University of Western Sydney, NSW, Australia
| | - Shoba Ranganathan
- Department of Chemistry and Biomolecular Sciences, Faculty of Science, Macquarie University, Sydney, NSW 2109 Australia
| | - Sock-Hwee Tan
- Australian School of Advanced Medicine, Faculty of Medicine and Human Sciences, Macquarie University, Rm1, Level 1, 75 Talavera Road, Sydney, NSW 2109 Australia
| | - Edouard Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton Campus, Melbourne, VIC 3800 Australia
| | - Mark S Baker
- Australian School of Advanced Medicine, Faculty of Medicine and Human Sciences, Macquarie University, Rm1, Level 1, 75 Talavera Road, Sydney, NSW 2109 Australia
| |
Collapse
|