1
|
Chitca DD, Popescu V, Dumitrescu A, Botezatu C, Mastalier B. Advancing Colorectal Cancer Diagnostics from Barium Enema to AI-Assisted Colonoscopy. Diagnostics (Basel) 2025; 15:974. [PMID: 40310348 PMCID: PMC12026282 DOI: 10.3390/diagnostics15080974] [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: 03/04/2025] [Revised: 03/18/2025] [Accepted: 04/09/2025] [Indexed: 05/02/2025] Open
Abstract
Colorectal cancer (CRC) remains a major global health burden, necessitating continuous advancements in diagnostic methodologies. Traditional screening techniques, including barium enema and fecal occult blood tests, have been progressively replaced by more precise modalities, such as colonoscopy, liquid biopsy, and artificial intelligence (AI)-assisted imaging. Objective: This review explores the evolution of CRC diagnostic tools, from conventional imaging methods to cutting-edge AI-driven approaches, emphasizing their clinical utility, cost-effectiveness, and integration into multidisciplinary healthcare settings. Methods: A comprehensive literature search was conducted using the PubMed, Medline, and Scopus databases, selecting studies that evaluate various CRC diagnostic tools, including endoscopic advancements, liquid biopsy applications, and AI-assisted imaging techniques. Key inclusion criteria include studies on diagnostic accuracy, sensitivity, specificity, clinical outcomes, and economic feasibility. Results: AI-assisted colonoscopy has demonstrated superior adenoma detection rates (ADR), reduced interobserver variability, and enhanced real-time lesion classification, offering a cost-effective alternative to liquid biopsy, particularly in high-volume healthcare institutions. While liquid biopsy provides a non-invasive means of molecular profiling, it remains cost-intensive and requires frequent testing, making it more suitable for post-treatment surveillance and high-risk patient monitoring. Conclusions: The future of CRC diagnostics lies in a hybrid model, leveraging AI-assisted endoscopic precision with molecular insights from liquid biopsy. This integration is expected to revolutionize early detection, risk stratification, and personalized treatment approaches, ultimately improving patient outcomes and healthcare efficiency.
Collapse
Affiliation(s)
- Dumitru-Dragos Chitca
- General Surgery Clinic, Colentina Clinical Hospital, 020125 Bucharest, Romania; (V.P.); (C.B.); (B.M.)
| | - Valentin Popescu
- General Surgery Clinic, Colentina Clinical Hospital, 020125 Bucharest, Romania; (V.P.); (C.B.); (B.M.)
- General Surgery Clinic, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Anca Dumitrescu
- Family Medicine, Vitan Polyclinic, 031087 Bucharest, Romania;
| | - Cristian Botezatu
- General Surgery Clinic, Colentina Clinical Hospital, 020125 Bucharest, Romania; (V.P.); (C.B.); (B.M.)
- General Surgery Clinic, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Bogdan Mastalier
- General Surgery Clinic, Colentina Clinical Hospital, 020125 Bucharest, Romania; (V.P.); (C.B.); (B.M.)
- General Surgery Clinic, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| |
Collapse
|
2
|
Sun Y, Kong D, Zhang Q, Xiang R, Lu S, Feng L, Zhang H. DNA methylation biomarkers for predicting lymph node metastasis in colorectal cancer. Clin Transl Oncol 2025; 27:439-448. [PMID: 39026026 DOI: 10.1007/s12094-024-03601-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 07/06/2024] [Indexed: 07/20/2024]
Abstract
Colorectal cancer is one of the most common cancers worldwide. Lymph node metastasis is an important marker of colorectal cancer progression and plays a key role in the evaluation of patient prognosis. Accurate preoperative assessment of lymph node metastasis is crucial for devising appropriate treatment plans. However, current clinical imaging methods have limitations in many aspects. Therefore, the discovery of a method for accurately predicting lymph node metastasis is crucial clinical decision-making. DNA methylation is a common epigenetic modification that can regulate gene expression, which also has an important impact on the development of colorectal cancer. It is considered to be a promising biomarker with good specificity and stability and has promising application in predicting lymph node metastasis in patients with colorectal cancer. This article reviews the characteristics and limitations of currently available methods for predicting lymph node metastasis in patients with colorectal cancer and discusses the role of DNA methylation as a biomarker.
Collapse
Affiliation(s)
- Yu Sun
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Deyang Kong
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Qi Zhang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Renshen Xiang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shuaibing Lu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lin Feng
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Haizeng Zhang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
3
|
Mills EG, Abbara A, Dhillo WS, Comninos AN. Interactions between kisspeptin and bone: Cellular mechanisms, clinical evidence, and future potential. Ann N Y Acad Sci 2024; 1540:47-60. [PMID: 39269749 DOI: 10.1111/nyas.15213] [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] [Indexed: 09/15/2024]
Abstract
The neuropeptide kisspeptin and its cognate receptor have been extensively studied in reproductive physiology, with diverse and well-established functions, including as an upstream regulator of pubertal onset, reproductive hormone secretion, and sexual behavior. Besides classical reproduction, both kisspeptin and its receptor are extensively expressed in bone-resorbing osteoclasts and bone-forming osteoblasts, which putatively permits direct bone effects. Accordingly, this sets the scene for recent compelling findings derived from in vitro experiments through to in vivo and clinical studies revealing prominent regulatory interactions for kisspeptin signaling in bone metabolism, as well as certain oncological aspects of bone metabolism. Herein, we comprehensively examine the experimental evidence obtained to date supporting the interaction between kisspeptin and bone. A comprehensive understanding of this emerging facet of kisspeptin biology is fundamental to exploiting the future therapeutic potential of kisspeptin-based medicines as a novel strategy for treating bone-related disorders.
Collapse
Affiliation(s)
- Edouard G Mills
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| | - Ali Abbara
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| | - Waljit S Dhillo
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| | - Alexander N Comninos
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, UK
- Endocrine Bone Unit, Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
4
|
Gelman IH. Metastasis suppressor genes in clinical practice: are they druggable? Cancer Metastasis Rev 2023; 42:1169-1188. [PMID: 37749308 PMCID: PMC11629483 DOI: 10.1007/s10555-023-10135-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 09/01/2023] [Indexed: 09/27/2023]
Abstract
Since the identification of NM23 (now called NME1) as the first metastasis suppressor gene (MSG), a small number of other gene products and non-coding RNAs have been identified that suppress specific parameters of the metastatic cascade, yet which have little or no ability to regulate primary tumor initiation or maintenance. MSG can regulate various pathways or cell biological functions such as those controlling mitogen-activated protein kinase pathway mediators, cell-cell and cell-extracellular matrix protein adhesion, cytoskeletal architecture, G-protein-coupled receptors, apoptosis, and transcriptional complexes. One defining facet of this gene class is that their expression is typically downregulated, not mutated, in metastasis, such that any effective therapeutic intervention would involve their re-expression. This review will address the therapeutic targeting of MSG, once thought to be a daunting task only facilitated by ectopically re-expressing MSG in metastatic cells in vivo. Examples will be cited of attempts to identify actionable oncogenic pathways that might suppress the formation or progression of metastases through the re-expression of specific metastasis suppressors.
Collapse
Affiliation(s)
- Irwin H Gelman
- Department of Cancer Genetics & Genomics, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA.
| |
Collapse
|
5
|
Panda SP, Kesharwani A, Singh GD, Prasanth D, Vatchavai BR, Kumari PVK, Panda SK, Mallick SP. Impose of KNDy/GnRH neural circuit in PCOS, ageing, cancer and Alzheimer's disease: StAR actions in prevention of neuroendocrine dysfunction. Ageing Res Rev 2023; 92:102086. [PMID: 37821047 DOI: 10.1016/j.arr.2023.102086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/06/2023] [Accepted: 10/08/2023] [Indexed: 10/13/2023]
Abstract
The Kisspeptin1 (KISS1)/neurokinin B (NKB)/Dynorphin (Dyn) [KNDy] neurons in the hypothalamus regulate the reproduction stage in human beings and rodents. KNDy neurons co-expressed all KISS1, NKB, and Dyn peptides, and hence commonly regarded as KISS1 neurons. KNDy neurons contribute to the "GnRH pulse generator" and are implicated in the regulation of pulsatile GnRH release. The estradiol (E2)-estrogen receptor (ER) interactions over GnRH neurons in the hypothalamus cause nitric oxide (NO) discharge, in addition to presynaptic GABA and glutamate discharge from respective neurons. The released GABA and glutamate facilitate the activity of GnRH neurons via GABAA-R and AMPA/kainate-R. The KISS1 stimulates MAPK/ERK1/2 signaling and cause the release of Ca2+ from intracellular store, which contribute to neuroendocrine function, increase apoptosis and decrease cell proliferation and metastasis. The ageing in women deteriorates KISS1/KISS1R interaction in the hypothalamus which causes lower levels of GnRH. Because examining the human brain is so challenging, decades of clinical research have failed to find the causes of KNDy/GnRH dysfunction. The KISS1/KISS1R interactions in the brain have a neuroprotective effect against Alzheimer's disease (AD). These findings modulate the pathophysiological role of the KNDy/GnRH neural network in polycystic ovarian syndrome (PCOS) associated with ageing and, its protective role in cancer and AD. This review concludes with protecting effect of the steroid-derived acute regulatory enzyme (StAR) against neurotoxicity in the hippocampus, and hypothalamus, and these measures are fundamental for delaying ageing with PCOS. StAR could serve as novel diagnostic marker and therapeutic target for the most prevalent hormone-sensitive breast cancers (BCs).
Collapse
Affiliation(s)
- Siva Prasad Panda
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India.
| | - Adarsh Kesharwani
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | | | - Dsnbk Prasanth
- KVSR Siddhartha College of Pharmaceutical Sciences, Vijayawada, Andhrapradesh, India
| | - Bhaskara Raju Vatchavai
- Sri Vasavi Institute of Pharmaceutical Sciences, Pedatadepalli, Tadepalligudem, Andhrapradesh, India
| | - P V Kamala Kumari
- Vignan Institute of Pharmaceutical Technology, Duvvada, Visakhapatnam, Andhrapradesh, India
| | | | | |
Collapse
|
6
|
Duan H, Ding X, Luo H. KISS-1, Mediated by Promoter Methylation, Suppresses Esophageal Squamous Cell Carcinoma Metastasis via MMP2/9/MAPK Axis. Dig Dis Sci 2022; 67:4780-4796. [PMID: 34993679 DOI: 10.1007/s10620-021-07335-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 11/15/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIMS KISS-1 is an established tumor suppressor that inhibits metastases in various malignancies. However, little is known regarding its role in esophageal squamous cell carcinoma (ESCC). The aim of the present study was to identify the possible mechanisms of KISS-1 in ESCC metastasis. METHODS The expression levels of KISS-1 mRNA and protein in ESCC samples and cell lines were analyzed by qRT-PCR, IHC, and western blotting. Bisulfite sequencing PCR (BSP) and methylation-specific PCR (MSP) were used to analyze the methylation pattern of KISS-1 promoter in ESCC cells with or without 5-Aza-dC treatment. The role of KISS-1 in the progression and metastasis of ESCC was analyzed through in vitro functional assays. RESULTS KISS-1 mRNA and protein were markedly downregulated in ESCC tissues and cell lines compared to the respective controls. Hypermethylation of KISS-1 promoter correlated to its lower expression levels in ESCC, and KISS-1 demethylation inhibited tumor progression. Ectopic KISS-1 overexpression inhibited tumor cell metastasis in vitro. In addition, KISS-1 overexpression downregulated the matrix metalloproteinase 2 and 9 (MMP2 and 9) and inhibited epithelial-mesenchymal transition (EMT). Finally, KISS-1 downregulated phosphorylated extracellular regulated protein kinase 1/2 (ERK1/2) and phosphorylated p38 mitogen-activated protein kinase (MAPK) without affecting their total expression levels in the ESCC cells. MAPK/ERK and p38 MAPK agonists reversed the suppressive effects of KISS-1. CONCLUSIONS The hypermethylation of KISS-1 promoter partly contributed to its downregulation in ESCC. KISS-1 inhibits the metastasis of ESCC cells by targeting the MMP2/9/ERK/p38 MAPK axis.
Collapse
Affiliation(s)
- Houyu Duan
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, People's Republic of China
| | - Xiang Ding
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, People's Republic of China
| | - Hesheng Luo
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, People's Republic of China.
| |
Collapse
|
7
|
Li Z, Liu J, Inuzuka H, Wei W. Functional analysis of the emerging roles for the KISS1/KISS1R signaling pathway in cancer metastasis. J Genet Genomics 2021; 49:181-184. [PMID: 34767970 DOI: 10.1016/j.jgg.2021.10.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/05/2021] [Accepted: 10/06/2021] [Indexed: 02/07/2023]
Abstract
Cancer metastasis, a process that primary tumor cells disseminate to secondary organs, is the most lethal and least effectively treated characteristic of human cancers. Kisspeptins are proteins encoded by the KISS1 gene that was originally described as a melanoma metastasis suppressor gene. Then, Kisspeptins were discovered as the natural ligands of the G-protein-coupled receptor 54 (GPR54) that is also called KISS1R. The KISS1/KISS1R signaling is essential to control GnRH secretion during puberty and to establish mammalian reproductive function through the hypothalamic-pituitary-gonadal (HPG) axis. Although KISS1 primarily plays a suppressive role in the metastasis progression in several cancer types, emerging evidence indicates that the physiological effect of KISS1/KISS1R in cancer metastasis is tissue context-dependent and still controversial. Here, we will discuss the epigenetic mechanism regulation of KISS1 gene expression, the context-dependent role of KISS1/KISS1R, pro-/anti-metastasis signaling pathways of KISS1/KISS1R, and the perspective anti-cancer therapeutics via targeting KISS1/KISS1R.
Collapse
Affiliation(s)
- Zhenxi Li
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Jing Liu
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Hiroyuki Inuzuka
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
8
|
Abstract
The significance of KISS1 goes beyond its original discovery as a metastasis suppressor. Its function as a neuropeptide involved in diverse physiologic processes is more well studied. Enthusiasm regarding KISS1 has cumulated in clinical trials in multiple fields related to reproduction and metabolism. But its cancer therapeutic space is unsettled. This review focuses on collating data from cancer and non-cancer fields in order to understand shared and disparate signaling that might inform clinical development in the cancer therapeutic and biomarker space. Research has focused on amino acid residues 68-121 (kisspeptin 54), binding to the KISS1 receptor and cellular responses. Evidence and counterevidence regarding this canonical pathway require closer look at the covariates so that the incredible potential of KISS1 can be realized.
Collapse
Affiliation(s)
- Thuc Ly
- Department of Cancer Biology, Kansas University Medical Center, 3901 Rainbow Blvd. - MS1071, Kansas City, KS, 66160, USA
| | - Sitaram Harihar
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603203, India
| | - Danny R Welch
- Department of Cancer Biology, Kansas University Medical Center, 3901 Rainbow Blvd. - MS1071, Kansas City, KS, 66160, USA.
- University of Kansas Cancer Center, 3901 Rainbow Blvd., Kansas City, KS, 66160, USA.
| |
Collapse
|
9
|
Reduced Kiss‑1 expression is associated with clinical aggressive feature of gastric cancer patients and promotes migration and invasion in gastric cancer cells. Oncol Rep 2020; 44:1149-1157. [PMID: 32705229 PMCID: PMC7388581 DOI: 10.3892/or.2020.7676] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 06/10/2020] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer (GC) causes high morbidity and mortality in patients largely due to its invasion and metastasis. Kiss‑1 has been shown to be a metastasis suppressor in various malignancies. However, its clinical significance and biological functions in GC have not been thoroughly investigated. The present study investigated the association between Kiss‑1 expression and its methylation status and clinicopathological features in GC. Kiss‑1 expression was reduced in GC and its low expression was associated with poor histological grade, lymph node metastasis and TNM III+IV stage. Kiss‑1 overexpression in AGS GC cells significantly inhibited cell proliferation, migration and invasion in vitro. Kiss‑1 knockdown promoted the proliferation, migration and invasion of HGC‑27 cells. In summary, the data demonstrated that a low expression of Kiss‑1 played a suppressive role for the proliferation, migration and invasion of GC cells. Its expression and methylation levels were associated with the clinical progression of GC. Thus, Kiss‑1 is a potential diagnostic and prognostic marker as well as a new target for the treatment of GC.
Collapse
|
10
|
Lin Y, Chen Z, Zheng Y, Liu Y, Gao J, Lin S, Chen S. MiR-506 Targets UHRF1 to Inhibit Colorectal Cancer Proliferation and Invasion via the KISS1/PI3K/NF-κ B Signaling Axis. Front Cell Dev Biol 2019; 7:266. [PMID: 31803739 PMCID: PMC6873823 DOI: 10.3389/fcell.2019.00266] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 10/18/2019] [Indexed: 12/24/2022] Open
Abstract
Background The UHRF1 gene is an epigenetic modification factor that mediates tumor suppressor gene silencing in a variety of cancers. Related studies have reported that UHRF1 can inhibit the expression of the KISS1 gene. However, the regulatory mechanism underlying UHRF1 expression in colorectal cancer (CRC) is still unclear. The aim of this study was to gain a better understanding of the regulation of UHRF1 expression in CRC and to determine whether it regulates the mechanism by which KISS1 promotes CRC metastasis. Methods In the present study, the levels of miR-506, UHRF1 and KISS1 expression in CRC tissues and in human CRC cell lines were studied using quantitative real-time PCR (qRT-PCR) and Western blotting. Cell proliferation, migration, and invasion assays are used to detect cell proliferation, migration, and invasion. A dual-luciferase reporter system was used to confirm the target gene of miR-506. Results This study found that UHRF1 protein is highly expressed in CRC tissues and negatively correlated with KISS1 protein expression. UHRF1 overexpression activates the PI3K/NF-κB signaling pathway by inhibiting the mRNA expression levels of pathway mediators. Bioinformatics analysis and luciferase reporter gene assays confirmed that miR-506 targets UHRF1. Conclusion This study identified the regulation of UHRF1 expression in CRC and the mechanism of CRC metastasis. UHRF1 may be a new potential target molecule for future CRC metastasis treatment.
Collapse
Affiliation(s)
- Yilin Lin
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Zhihua Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yan Zheng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yisu Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Ji Gao
- School of Nursing, Fujian Medical University, Fuzhou, China
| | - Suyong Lin
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Shaoqin Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
11
|
Motti ML, Meccariello R. Minireview: The Epigenetic Modulation of KISS1 in Reproduction and Cancer. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16142607. [PMID: 31336647 PMCID: PMC6679060 DOI: 10.3390/ijerph16142607] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/04/2019] [Accepted: 07/17/2019] [Indexed: 01/07/2023]
Abstract
Epigenetics describes how both lifestyle and environment may affect human health through the modulation of genome functions and without any change to the DNA nucleotide sequence. The discovery of several epigenetic mechanisms and the possibility to deliver epigenetic marks in cells, gametes, and biological fluids has opened up new perspectives in the prevention, diagnosis, and treatment of human diseases. In this respect, the depth of knowledge of epigenetic mechanisms is fundamental to preserving health status and to developing targeted interventions. In this minireview, we summarize the epigenetic modulation of the KISS1 gene in order to provide an example of epigenetic regulation in health and disease.
Collapse
Affiliation(s)
- Maria Letizia Motti
- Dipartimento di Scienze Motorie e del Benessere, Università di Napoli Parthenope, via Medina 40, 80133 Napoli, Italy
| | - Rosaria Meccariello
- Dipartimento di Scienze Motorie e del Benessere, Università di Napoli Parthenope, via Medina 40, 80133 Napoli, Italy.
| |
Collapse
|
12
|
Wang XQ, Fang PF, Zhang C, Xu YY, Song XB, Liang J, Xia QR. Low KISS1 expression predicts poor prognosis for patients with colorectal cancer: A meta-analysis. Clin Exp Pharmacol Physiol 2019; 46:625-634. [PMID: 30932210 DOI: 10.1111/1440-1681.13093] [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: 10/19/2018] [Revised: 03/22/2019] [Accepted: 03/26/2019] [Indexed: 12/24/2022]
Abstract
KISS1 and KISS1R, a novel pair of metastasis suppressors, are likely to be associated with the prognosis of colorectal cancer (CRC). Here, a meta-analysis was performed to study the role of KISS1 and KISS1R in CRC. Heterogeneity, stability and publication bias were all estimated. Six publications describing a total of 559 CRC patients were included in the present study. Low KISS1 expression predicted 70% higher risk of poor prognosis for general patients (HR, 1.71; 95% CI, 1.28-2.29) and 99% higher risk for East Asian patients (HR, 1.99; 95% CI, 1.46-2.72). Limited evidence indicated that decreased KISS1R expression might predict poor outcome (HR, 2.96; 95% CI, 1.51-5.82). Neither heterogeneity nor publication bias was identified. The current analyses suggest that low KISS1 expression predicts poor overall survival among East Asian patients with CRC. Evidence on other races and KISS1R are still insufficient, and additional studies are required to clarify the risk of CRC associated with KISS1R by race.
Collapse
Affiliation(s)
| | | | - Cheng Zhang
- Anhui Provincial Cancer Institute, Hefei, China.,The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ya-Yun Xu
- Department of Pharmacy, Hefei Fourth People's Hospital, Hefei, China.,Anhui Mental Health Center, Hefei, China
| | | | - Jun Liang
- Department of Pharmacy, Hefei Fourth People's Hospital, Hefei, China.,Anhui Mental Health Center, Hefei, China
| | - Qing-Rong Xia
- Department of Pharmacy, Hefei Fourth People's Hospital, Hefei, China.,Anhui Mental Health Center, Hefei, China
| |
Collapse
|
13
|
Weinman MA, Fischer JA, Jacobs DC, Goodall CP, Bracha S, Chappell PE. Autocrine production of reproductive axis neuropeptides affects proliferation of canine osteosarcoma in vitro. BMC Cancer 2019; 19:158. [PMID: 30777054 PMCID: PMC6379937 DOI: 10.1186/s12885-019-5363-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 02/11/2019] [Indexed: 01/19/2023] Open
Abstract
Background Osteosarcoma strikes hundreds of people each year, of both advanced and younger ages, and is often terminal. Like many tumor types, these bone tumors will frequently undergo a neuroendocrine transition, utilizing autocrine and/or paracrine hormones as growth factors and/or promoters of angiogenesis to facilitate progression and metastasis. While many of these factors and their actions on tumor growth are characterized, some tumor-derived neuropeptides remain unexplored. Methods Using validated canine osteosarcoma cell lines in vitro, as well as cells derived from spontaneous tumors in dogs, we explored the autocrine production of two neuropeptides typically found in the hypothalamus, and most closely associated with reproduction: gonadotropin-releasing hormone (GnRH) and kisspeptin (Kiss-1). We evaluated gene expression and protein secretion of these hormones using quantitative RT-PCR and a sensitive radioimmunoassay, and explored changes in cell proliferation determined by MTS cell viability assays. Results Our current studies reveal that several canine osteosarcoma cell lines (COS, POS, HMPOS, D17, C4) synthesize and secrete GnRH and express the GnRH receptor, while COS and POS also express kiss1 and its cognate receptor. We have further found that GnRH and kisspeptin, exogenously applied to these tumor cells, exert significant effects on both gene expression and proliferation. Of particular interest, kisspeptin exposure stimulated GnRH secretion from COS, similarly to the functional relationship observed within the neuroendocrine reproductive axis. Additionally, GnRH and kisspeptin treatment both increased COS proliferation, which additionally manifested in increased expression of the bone remodeling ligand rankl within these cells. These effects were blocked by treatment with a specific GnRH receptor inhibitor. Both neuropeptides were found to increase expression of the specific serotonin (5HT) receptor htr2a, the activation of which has previously been associated with cellular proliferation, suggesting that production of these factors by osteosarcoma cells may act to sensitize tumors to circulating 5HT of local and/or enteric origin. Conclusions Here we report that kisspeptin and GnRH act as autocrine growth factors in canine osteosarcoma cells in vitro, modulating RANKL and serotonin receptor expression in a manner consistent with pro-proliferative effects. Pharmacological targeting of these hormones may represent new avenues of osteosarcoma treatment. Electronic supplementary material The online version of this article (10.1186/s12885-019-5363-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marcus A Weinman
- Department of Clinical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR, 97331, USA
| | - Jacob A Fischer
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR, 97331, USA
| | - Dakota C Jacobs
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR, 97331, USA
| | - Cheri P Goodall
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR, 97331, USA
| | - Shay Bracha
- Department of Clinical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR, 97331, USA
| | - Patrick E Chappell
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR, 97331, USA.
| |
Collapse
|
14
|
Taniguchi-Ponciano K, Ribas-Aparicio RM, Marrero-Rodríguez D, Arreola-De la Cruz H, Huerta-Padilla V, Muñoz N, Gómez-Ortiz L, Ponce-Navarrete G, Rodríguez-Esquivel M, Mendoza-Rodríguez M, Gómez-Virgilio L, Peralta R, Serna L, Gómez G, Ortiz J, Mantilla A, Hernández D, Hernández Á, Bandala C, Salcedo M. The KISS1 gene overexpression as a potential molecular marker for cervical cancer cells. Cancer Biomark 2018; 22:709-719. [DOI: 10.3233/cbm-181215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Keiko Taniguchi-Ponciano
- Laboratorio de Oncología Genómica, Unidad de Investigación Médica en Enfermedades Oncológicas, Hospital de Oncología, CMN-SXXI, IMSS, Mexico City, Mexico
- Programa de Doctorado en Ciencias en Biomedicina y Biotecnología Molecular, Departamento de Microbiología, Escuela de Ciencias Biológicas Instituto Politécnico Nacional (ENCB-IPN), Mexico City, Mexico
| | - Rosa María Ribas-Aparicio
- Programa de Doctorado en Ciencias en Biomedicina y Biotecnología Molecular, Departamento de Microbiología, Escuela de Ciencias Biológicas Instituto Politécnico Nacional (ENCB-IPN), Mexico City, Mexico
| | - Daniel Marrero-Rodríguez
- Laboratorio de Oncología Genómica, Unidad de Investigación Médica en Enfermedades Oncológicas, Hospital de Oncología, CMN-SXXI, IMSS, Mexico City, Mexico
- Laboratorio 5 Departamento de Biomedicina Molecular, CINVESTAV-IPN, Mexico City, Mexico
| | - Hugo Arreola-De la Cruz
- Laboratorio de Oncología Genómica, Unidad de Investigación Médica en Enfermedades Oncológicas, Hospital de Oncología, CMN-SXXI, IMSS, Mexico City, Mexico
| | - Víctor Huerta-Padilla
- Laboratorio de Oncología Genómica, Unidad de Investigación Médica en Enfermedades Oncológicas, Hospital de Oncología, CMN-SXXI, IMSS, Mexico City, Mexico
| | - Nancy Muñoz
- Laboratorio de Oncología Genómica, Unidad de Investigación Médica en Enfermedades Oncológicas, Hospital de Oncología, CMN-SXXI, IMSS, Mexico City, Mexico
| | - Laura Gómez-Ortiz
- Laboratorio de Oncología Genómica, Unidad de Investigación Médica en Enfermedades Oncológicas, Hospital de Oncología, CMN-SXXI, IMSS, Mexico City, Mexico
| | - Gustavo Ponce-Navarrete
- Laboratorio de Oncología Genómica, Unidad de Investigación Médica en Enfermedades Oncológicas, Hospital de Oncología, CMN-SXXI, IMSS, Mexico City, Mexico
| | - Miriam Rodríguez-Esquivel
- Laboratorio de Oncología Genómica, Unidad de Investigación Médica en Enfermedades Oncológicas, Hospital de Oncología, CMN-SXXI, IMSS, Mexico City, Mexico
| | - Mónica Mendoza-Rodríguez
- Laboratorio de Oncología Genómica, Unidad de Investigación Médica en Enfermedades Oncológicas, Hospital de Oncología, CMN-SXXI, IMSS, Mexico City, Mexico
- Universidad Politécnico de Huatusco, Huatusco, Veracruz, Mexico
| | - Laura Gómez-Virgilio
- Laboratorio 5 Departamento de Biomedicina Molecular, CINVESTAV-IPN, Mexico City, Mexico
| | - Raúl Peralta
- Centro de Investigación en Dinámica Celular, Universidad Autónoma del Estado de Morelos, Cuernavaca, Mexico
| | - Luis Serna
- Clínica de Displasias, Hospital General de México, Mexico City, Mexico
| | - Guillermo Gómez
- Clínica de Displasias, Hospital General de México, Mexico City, Mexico
| | - Jorge Ortiz
- Clínica de Displasias, Hospital General de México, Mexico City, Mexico
| | - Alejandra Mantilla
- Servicio de Patología, Hospital de Oncología CMN-SXXI, IMSS, Mexico City, Mexico
| | - Daniel Hernández
- División de Laboratorios de Vigilancia e Investigación Epidemiológica, IMSS, Mexico City, Mexico
| | - Ángeles Hernández
- División de Laboratorios de Vigilancia e Investigación Epidemiológica, IMSS, Mexico City, Mexico
| | - Cindy Bandala
- Unidad de Apoyo a la Investigación, Instituto Nacional de Rehabilitación, Secretaría de Salud, Mexico City, Mexico
| | - Mauricio Salcedo
- Laboratorio de Oncología Genómica, Unidad de Investigación Médica en Enfermedades Oncológicas, Hospital de Oncología, CMN-SXXI, IMSS, Mexico City, Mexico
| |
Collapse
|
15
|
Shen ZL, Wang B, Jiang KW, Ye CX, Cheng C, Yan YC, Zhang JZ, Yang Y, Gao ZD, Ye YJ, Wang S. Downregulation of miR-199b is associated with distant metastasis in colorectal cancer via activation of SIRT1 and inhibition of CREB/KISS1 signaling. Oncotarget 2018; 7:35092-105. [PMID: 27145368 PMCID: PMC5085212 DOI: 10.18632/oncotarget.9042] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Accepted: 04/11/2016] [Indexed: 12/22/2022] Open
Abstract
The progression of distant metastasis cascade is a multistep and complicated process, frequently leading to a poor prognosis in cancer patients. Recently, growing evidence has indicated that deregulation of microRNAs (miRNAs) contributes to tumorigenesis and tumor progression in colorectal cancer (CRC). In the present study, by comparing the miRNA expression profiles of CRC tissues and corresponding hepatic metastasis tissues, we established the downregulation of miR-199b in CRC metastasis tissues. The decrease in miR-199b expression was significantly correlated to late TNM stage and distant metastasis. Moreover, Kaplan–Meier curves showed that CRC patients with high expression level of miR-199b had a longer median survival. Functional assays results indicated that the restoration of miR-199b considerably reduced cell invasion and migration in vitro and in vivo, and increased the sensitivity to 5-FU and oxaliplatin. Further dual-luciferase reporter gene assays revealed that SIRT1 was the direct target of miR-199b in CRC. The expression of miR-199b was inversely correlated with SIRT1 in CRC specimens. SIRT1 knockdown produced effects on biological behavior that were similar to those of miR-199b overexpression. Furthermore, through Human Tumor Metastasis PCR Array we discovered KISS1 was one of the downstream targets of SIRT1. Silencing of SIRT1 upregulated KISS1 expression by enhancing the acetylation of the transcription factor CREB. The latter was further activated via binding to the promoter of KISS1 to induce transcription. Thus, we concluded that miR-199b regulates SIRT1/CREB/KISS1 signaling pathway and might serve as a prognosis marker or a novel therapeutic target for patients with CRC.
Collapse
Affiliation(s)
- Zhan-Long Shen
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, 100044, PR China.,Laboratory of Surgical Oncology, Peking University People's Hospital, Beijing, 100044, PR China
| | - Bo Wang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, 100044, PR China.,Laboratory of Surgical Oncology, Peking University People's Hospital, Beijing, 100044, PR China
| | - Ke-Wei Jiang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, 100044, PR China.,Laboratory of Surgical Oncology, Peking University People's Hospital, Beijing, 100044, PR China
| | - Chun-Xiang Ye
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, 100044, PR China.,Laboratory of Surgical Oncology, Peking University People's Hospital, Beijing, 100044, PR China
| | - Cheng Cheng
- Department of General Surgery, Tangshan Gongren Hospital, Hebei Medical University, Tangshan, Hebei, 063000, PR China
| | - Yi-Chao Yan
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, 100044, PR China.,Laboratory of Surgical Oncology, Peking University People's Hospital, Beijing, 100044, PR China
| | - Ji-Zhun Zhang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, 100044, PR China.,Laboratory of Surgical Oncology, Peking University People's Hospital, Beijing, 100044, PR China
| | - Yang Yang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, 100044, PR China.,Laboratory of Surgical Oncology, Peking University People's Hospital, Beijing, 100044, PR China
| | - Zhi-Dong Gao
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, 100044, PR China.,Laboratory of Surgical Oncology, Peking University People's Hospital, Beijing, 100044, PR China
| | - Ying-Jiang Ye
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, 100044, PR China.,Laboratory of Surgical Oncology, Peking University People's Hospital, Beijing, 100044, PR China
| | - Shan Wang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, 100044, PR China.,Laboratory of Surgical Oncology, Peking University People's Hospital, Beijing, 100044, PR China
| |
Collapse
|
16
|
Shen ZL, Wang B, Jiang KW, Ye CX, Cheng C, Yan YC, Zhang JZ, Yang Y, Gao ZD, Ye YJ, Wang S. Downregulation of miR-199b is associated with distant metastasis in colorectal cancer via activation of SIRT1 and inhibition of CREB/KISS1 signaling. Oncotarget 2018; 7:49156-49168. [PMID: 27203385 PMCID: PMC5226498 DOI: 10.18632/oncotarget.9402] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 04/22/2016] [Indexed: 12/22/2022] Open
Abstract
Surface CD24 has previously been described, together with CD44 and ESA, for the characterization of putative cancer stem cells in pancreatic ductal adenocarcinoma (PDAC), the most fatal of all solid tumors. CD24 has a variety of biological functions including the regulation of invasiveness and cell proliferation, depending on the tumor entity and subcellular localization. Genetically engineered mouse models (GEMM) expressing oncogenic KrasG12D recapitulate the human disease and develop PDAC. In this study we investigate the function of CD24 using GEMM of endogenous PDAC and a model of cerulein-induced acute pancreatitis. We found that (i) CD24 expression was upregulated in murine and human PDAC and during acute pancreatitis (ii) CD24 was expressed exclusively in differentiated PDAC, whereas CD24 absence was associated with undifferentiated tumors and (iii) membranous CD24 expression determines tumor subpopulations with an epithelial phenotype in grafted models. In addition, we show that CD24 protein is stabilized in response to WNT activation and that overexpression of CD24 in pancreatic cancer cells upregulated β-catenin expression augmenting an epithelial, non-metastatic signature. Our results support a positive feedback model according to which (i) WNT activation and subsequent β-catenin dephosphorylation stabilize CD24 protein expression, and (ii) sustained CD24 expression upregulates β-catenin expression. Eventually, membranous CD24 augments the epithelial phenotype of pancreatic tumors. Thus we link the WNT/β-catenin pathway with the regulation of CD24 in the context of PDAC differentiation.
Collapse
Affiliation(s)
- Zhan-Long Shen
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, 100044, PR China.,Laboratory of Surgical Oncology, Peking University People's Hospital, Beijing, 100044, PR China
| | - Bo Wang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, 100044, PR China.,Laboratory of Surgical Oncology, Peking University People's Hospital, Beijing, 100044, PR China
| | - Ke-Wei Jiang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, 100044, PR China.,Laboratory of Surgical Oncology, Peking University People's Hospital, Beijing, 100044, PR China
| | - Chun-Xiang Ye
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, 100044, PR China.,Laboratory of Surgical Oncology, Peking University People's Hospital, Beijing, 100044, PR China
| | - Cheng Cheng
- Department of General Surgery, Tangshan Gongren Hospital, Hebei Medical University, Tangshan, Hebei, 063000, PR China
| | - Yi-Chao Yan
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, 100044, PR China.,Laboratory of Surgical Oncology, Peking University People's Hospital, Beijing, 100044, PR China
| | - Ji-Zhun Zhang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, 100044, PR China.,Laboratory of Surgical Oncology, Peking University People's Hospital, Beijing, 100044, PR China
| | - Yang Yang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, 100044, PR China.,Laboratory of Surgical Oncology, Peking University People's Hospital, Beijing, 100044, PR China
| | - Zhi-Dong Gao
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, 100044, PR China.,Laboratory of Surgical Oncology, Peking University People's Hospital, Beijing, 100044, PR China
| | - Ying-Jiang Ye
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, 100044, PR China.,Laboratory of Surgical Oncology, Peking University People's Hospital, Beijing, 100044, PR China
| | - Shan Wang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, 100044, PR China.,Laboratory of Surgical Oncology, Peking University People's Hospital, Beijing, 100044, PR China
| |
Collapse
|
17
|
Guzman S, Brackstone M, Radovick S, Babwah AV, Bhattacharya MM. KISS1/KISS1R in Cancer: Friend or Foe? Front Endocrinol (Lausanne) 2018; 9:437. [PMID: 30123188 PMCID: PMC6085450 DOI: 10.3389/fendo.2018.00437] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 07/16/2018] [Indexed: 12/19/2022] Open
Abstract
The KISS1 gene encodes KISS1, a protein that is rapidly processed in serum into smaller but biologically active peptides called kisspeptins (KPs). KISS1 and the KPs signal via the G-protein coupled receptor KISS1R. While KISS1 and KPs are recognized as potent positive regulators of the reproductive neuroendocrine axis in mammals, the first reported role for KISS1 was that of metastasis suppression in melanoma. Since then, it has become apparent that KISS1, KPs, and KISS1R regulate the development and progression of several cancers but interestingly, while these molecules act as suppressors of tumorigenesis and metastasis in many cancers, in breast and liver cancer they function as promoters. Thus, they join a small but growing number of molecules that exhibit dual roles in cancer highlighting the importance of studying cancer in context. Given their roles, KISS1, KPs and KISS1R represent important molecules in the development of novel therapies and/or as prognostic markers in treating cancer. However, getting to that point requires a detailed understanding of the relationship between these molecules and different cancers. The purpose of this review is therefore to highlight and discuss the clinical studies that have begun describing this relationship in varying cancer types including breast, liver, pancreatic, colorectal, bladder, and ovarian. An emerging theme from the reviewed studies is that the relationship between these molecules and a given cancer is complex and affected by many factors such as the micro-environment and steroid receptor status of the cancer cell. Our review and discussion of these important clinical studies should serve as a valuable resource in the successful development of future clinical studies.
Collapse
Affiliation(s)
- Stephania Guzman
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
- Child Health Institute of New Jersey, New Brunswick, NJ, United States
| | - Muriel Brackstone
- Division of Surgical Oncology, The University of Western Ontario, London, ON, Canada
| | - Sally Radovick
- Child Health Institute of New Jersey, New Brunswick, NJ, United States
- Department of Pediatrics, Robert Wood Johnson Medical School, Rutgers University, The State University of New Jersey, New Brunswick, NJ, United States
| | - Andy V. Babwah
- Child Health Institute of New Jersey, New Brunswick, NJ, United States
- Department of Pediatrics, Robert Wood Johnson Medical School, Rutgers University, The State University of New Jersey, New Brunswick, NJ, United States
| | - Moshmi M. Bhattacharya
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
- Child Health Institute of New Jersey, New Brunswick, NJ, United States
- Cancer Institute of New Jersey, New Brunswick, NJ, United States
- *Correspondence: Moshmi M. Bhattacharya
| |
Collapse
|
18
|
Mousavi Ardehaie R, Hashemzadeh S, Behrouz Sharif S, Ghojazadeh M, Teimoori-Toolabi L, Sakhinia E. Aberrant methylated EDNRB can act as a potential diagnostic biomarker in sporadic colorectal cancer while KISS1 is controversial. Bioengineered 2017; 8:555-564. [PMID: 28140749 PMCID: PMC5639868 DOI: 10.1080/21655979.2017.1283458] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 01/08/2017] [Accepted: 01/12/2017] [Indexed: 12/26/2022] Open
Abstract
Cancers are among the most serious threats of human health worldwide. Survival and mortality rates of colorectal cancer (CRC) strongly depend on the early diagnosis. The aberrant methylation pattern of genes as a diagnostic biomarker can serve as a practical option for timely detection and contribute subsequently to the enhancement of survival rate in CRC patients, since methylation changes are not only frequent but also can occur in initial tumorogenesis stages. It has been indicated that EDNRB and KISS1 genes are hypermethylated through progression and development of CRC. In current study, after extraction of genomic DNA from 45 paired tumor and adjacent non-cancerous tissue samples and treatment with bisulfite conversion, the methylation status of EDNRB and KISS1 CpG rich regions were assessed quantitatively using MS-HRM assay to determine practicability of these aberrant methylations for diagnosis of sporadic CRC and its discrimination from corresponding normal tissues. The results showed that the methylation distribution differences, comparing tumor tissues with their adjacent non-cancerous tissues, were statistically significant in all selected locations within EDNRB gene promoter (P < 0.001); they had also some correlations with tumor stage and grade. Nonetheless, methylation distribution in KISS1 gene CpG rich region revealed no statistically significant differences between CRC and adjacent non-cancerous tissues (P = 0.060). Overall, it can be concluded that aberrant methylated EDNRB can be a promising potential diagnostic biomarker for CRC, while KISS1 is controversial and needs to be more investigated.
Collapse
Affiliation(s)
- Reza Mousavi Ardehaie
- Department of Biochemistry and Clinical Laboratory, Division of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Molecular Medicine Department, Pasteur Institute of Iran, Tehran, Iran
| | - Shahriar Hashemzadeh
- Department of General & Vascular Surgery, Tabriz University of Medical Sciences, Tabriz, Iran
- Tuberculosis and lung research center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shahin Behrouz Sharif
- Department of Biochemistry and Clinical Laboratory, Division of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Molecular Medicine Department, Pasteur Institute of Iran, Tehran, Iran
| | - Morteza Ghojazadeh
- Liver and Gastrointestinal Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Ebrahim Sakhinia
- Department of Biochemistry and Clinical Laboratory, Division of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
19
|
Uenoyama Y, Tomikawa J, Inoue N, Goto T, Minabe S, Ieda N, Nakamura S, Watanabe Y, Ikegami K, Matsuda F, Ohkura S, Maeda KI, Tsukamura H. Molecular and Epigenetic Mechanism Regulating Hypothalamic Kiss1 Gene Expression in Mammals. Neuroendocrinology 2016; 103:640-9. [PMID: 26964105 DOI: 10.1159/000445207] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 03/02/2016] [Indexed: 11/19/2022]
Abstract
After the discovery of hypothalamic kisspeptin encoded by the Kiss1 gene, the central mechanism regulating gonadotropin-releasing hormone (GnRH) secretion, and hence gonadotropin secretion, is gradually being unraveled. This has increased our understanding of the central mechanism regulating puberty and subsequent reproductive performance in mammals. Recently, emerging evidence has indicated the molecular and epigenetic mechanism regulating hypothalamic Kiss1 gene expression. Here we compile data regarding DNA and histone modifications in the Kiss1 promoter region and provide a hypothetic scheme of the molecular and epigenetic mechanism regulating Kiss1 gene expression in two populations of hypothalamic kisspeptin neurons, which govern puberty and subsequent reproductive performance via GnRH/gonadotropin secretion.
Collapse
Affiliation(s)
- Yoshihisa Uenoyama
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|