1
|
Qasim M, Ricks-Santi LJ, Naab TJ, Rajack F, Beyene D, Abbas M, Kassim OO, Copeland RL, Kanaan Y. Inverse Correlation of KISS1 and KISS1R Expression in Triple-negative Breast Carcinomas from African American Women. Cancer Genomics Proteomics 2022; 19:673-682. [PMID: 36316037 PMCID: PMC9620443 DOI: 10.21873/cgp.20350] [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: 05/23/2022] [Revised: 06/29/2022] [Accepted: 07/21/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND/AIM The kisspeptin 1 (KISS1) gene encodes a precursor polypeptide which after proteolysis forms the kisspeptin-10 (KISS1) protein. KISS1, retains maximum physiological activity when it binds to its receptor (KISS1R), allowing KISS1 to effectively function as a suppressor of metastasis in melanomas and other types of cancer. The goal of this study was to evaluate the expression of KISS1 and KISS1R in breast carcinomas from African American (AA) women and correlate their association with clinicopathological features, including breast cancer subtypes, and outcomes. MATERIALS AND METHODS Tissue microarrays were constructed from formalin-fixed, paraffin-embedded surgical blocks from 216 AA patients. KISS1 and KISS1R expression was assessed using immunohistochemistry. Univariate analysis was used to determine the association between the expression of KISS1 and KISS1R, and clinicopathological characteristics. Pearson correlation was also determined between immunohistochemical H-scores, tumor size, and the number of positive lymph nodes. Kaplan-Meier estimates of overall and disease-free survival were plotted, and log-rank tests were performed to compare estimates among groups. RESULTS KISS1 protein expression was found to be higher in receptor-negative and triple-negative breast cancer (TNBC) compared to other subtypes (p<0.001). However, KISS1R expression was higher in non-TNBC tumors compared to other subtypes (p<0.001). Higher KISS1R expression was marginally negatively correlated with tumor size (p=0.077), and positively correlated with lymph-node positivity (p=0.056), and disease-free survival (p=0.092). CONCLUSION Our study showed a significant inverse correlation between KISS1 and KISS1R in TNBC. This investigation implicates a role for KISS1 and KISS1R in the pathogenesis of TNBCs in AA women.
Collapse
Affiliation(s)
- Mustafa Qasim
- Department of Microbiology, Howard University College of Medicine, Washington, DC, U.S.A
| | - Luisel J Ricks-Santi
- Department of Pharmacotherapy and Translational Research, College of Medicine, University of Florida, Gainesville, FL, U.S.A
| | - Tammey J Naab
- Department of Pathology, Howard University Hospital, Washington, DC, U.S.A
| | - Fareed Rajack
- Department of Pathology, Howard University Hospital, Washington, DC, U.S.A
| | - Desta Beyene
- Howard University Cancer Center, Washington, DC, U.S.A
| | - Muneer Abbas
- Department of Microbiology, Howard University College of Medicine, Washington, DC, U.S.A
| | - Olakunle O Kassim
- Department of Microbiology, Howard University College of Medicine, Washington, DC, U.S.A
| | - Robert L Copeland
- Howard University Cancer Center, Washington, DC, U.S.A
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, U.S.A
| | - Yasmine Kanaan
- Department of Microbiology, Howard University College of Medicine, Washington, DC, U.S.A.;
- Howard University Cancer Center, Washington, DC, U.S.A
| |
Collapse
|
2
|
Akad M, Socolov R, Furnică C, Covali R, Stan CD, Crauciuc E, Pavaleanu I. Kisspeptin Variations in Patients with Polycystic Ovary Syndrome-A Prospective Case Control Study. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58060776. [PMID: 35744039 PMCID: PMC9227115 DOI: 10.3390/medicina58060776] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/02/2022] [Accepted: 06/06/2022] [Indexed: 11/21/2022]
Abstract
Background and objectives: Kisspeptin, also named metastin, showed important roles in initiating the secretion of gonadotropin-releasing hormone (GnRH) and is an essential factor in the development of polycystic ovaries syndrome (PCOS). Several research studies noticed associations between kisspeptin levels and patients with anovulatory cycles due to PCOS with an increased LH/FSH ratio. The aim of our study was to bring scientific evidence regarding the correlation between high kisspeptin and luteinizing hormone values in subfertile women due to PCOS. Materials and Methods: A prospective case-control study was conducted in “Elena Doamna” Hospital of Obstetrics and Gynecology between 4 January 2021 and 1 March 2022. All patients agreed to participate in our study, had ages between 18 and 45 years old, and had a body mass index between 18.5 and 30 kg/m2. The study group consisted of subfertile patients with PCOS and menstrual disturbances, including amenorrhea or oligomenorrhea. The control group consisted of healthy patients with ovulatory cycles and no other reproductive or endocrinology pathologies. During the follicular phase of their menstrual cycle, patients had blood samples taken with the dosage of kisspeptin, LH, FSH, estradiol, insulin, glycemic levels, testosterone, and prolactin. Pelvic ultrasounds and clinical examinations were performed as well. Results: Significant differences were observed in kisspeptin, LH, FSH, and estradiol levels between patients with PCOS and the control group. After the univariate analysis, PCOS was significantly associated with increased kisspeptin, increased LH, and decreased FSH. There was no significant association between PCOS, estradiol, prolactin, and insulin. Conclusions: kisspeptin serum values are higher in subfertile PCOS patients, supporting the hypothesis that an over-stimulation of the KISS1 system might cause the hyper-stimulation of the HPG-axis.
Collapse
Affiliation(s)
- Mona Akad
- Department of Obstetrics and Gynecology, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iași, Romania; (M.A.); (C.F.); (R.C.); (C.D.S.); (E.C.); (I.P.)
- Clinical Hospital of Obstetrics and Pharmacy “Elena Doamna”, 700398 Iași, Romania
| | - Răzvan Socolov
- Department of Obstetrics and Gynecology, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iași, Romania; (M.A.); (C.F.); (R.C.); (C.D.S.); (E.C.); (I.P.)
- Clinical Hospital of Obstetrics and Pharmacy “Elena Doamna”, 700398 Iași, Romania
- Correspondence: ; Tel.: +40-07-2272-8514
| | - Cristina Furnică
- Department of Obstetrics and Gynecology, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iași, Romania; (M.A.); (C.F.); (R.C.); (C.D.S.); (E.C.); (I.P.)
| | - Roxana Covali
- Department of Obstetrics and Gynecology, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iași, Romania; (M.A.); (C.F.); (R.C.); (C.D.S.); (E.C.); (I.P.)
- Clinical Hospital of Obstetrics and Pharmacy “Elena Doamna”, 700398 Iași, Romania
| | - Catalina Daniela Stan
- Department of Obstetrics and Gynecology, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iași, Romania; (M.A.); (C.F.); (R.C.); (C.D.S.); (E.C.); (I.P.)
- Clinical Hospital of Obstetrics and Pharmacy “Elena Doamna”, 700398 Iași, Romania
| | - Eduard Crauciuc
- Department of Obstetrics and Gynecology, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iași, Romania; (M.A.); (C.F.); (R.C.); (C.D.S.); (E.C.); (I.P.)
- Clinical Hospital of Obstetrics and Pharmacy “Elena Doamna”, 700398 Iași, Romania
| | - Ioana Pavaleanu
- Department of Obstetrics and Gynecology, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iași, Romania; (M.A.); (C.F.); (R.C.); (C.D.S.); (E.C.); (I.P.)
- Clinical Hospital of Obstetrics and Pharmacy “Elena Doamna”, 700398 Iași, Romania
| |
Collapse
|
3
|
Guzman S, Dragan M, Kwon H, de Oliveira V, Rao S, Bhatt V, Kalemba KM, Shah A, Rustgi VK, Wang H, Bech PR, Abbara A, Izzi-Engbeaya C, Manousou P, Guo JY, Guo GL, Radovick S, Dhillo WS, Wondisford FE, Babwah AV, Bhattacharya M. Targeting hepatic kisspeptin receptor ameliorates nonalcoholic fatty liver disease in a mouse model. J Clin Invest 2022; 132:145889. [PMID: 35349482 PMCID: PMC9106350 DOI: 10.1172/jci145889] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 03/23/2022] [Indexed: 01/27/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), the most common liver disease, has become a silent worldwide pandemic. The incidence of NAFLD correlates with the rise in obesity, type 2 diabetes, and metabolic syndrome. A hallmark featureof NAFLD is excessive hepatic fat accumulation or steatosis, due to dysregulated hepatic fat metabolism, which can progress to nonalcoholic steatohepatitis (NASH), fibrosis, and cirrhosis. Currently, there are no approved pharmacotherapies to treat this disease. Here, we have found that activation of the kisspeptin 1 receptor (KISS1R) signaling pathway has therapeutic effects in NAFLD. Using high-fat diet-fed mice, we demonstrated that a deletion of hepatic Kiss1r exacerbated hepatic steatosis. In contrast, enhanced stimulation of KISS1R protected against steatosis in wild-type C57BL/6J mice and decreased fibrosis using a diet-induced mouse model of NASH. Mechanistically, we found that hepatic KISS1R signaling activates the master energy regulator, AMPK, to thereby decrease lipogenesis and progression to NASH. In patients with NAFLD and in high-fat diet-fed mice, hepatic KISS1/KISS1R expression and plasma kisspeptin levels were elevated, suggesting a compensatory mechanism to reduce triglyceride synthesis. These findings establish KISS1R as a therapeutic target to treat NASH.
Collapse
Affiliation(s)
- Stephania Guzman
- Department of Medicine, Robert Wood Johnson Medical School, and,Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, New Jersey, USA
| | | | - Hyokjoon Kwon
- Department of Medicine, Robert Wood Johnson Medical School, and
| | | | - Shivani Rao
- Department of Medicine, Robert Wood Johnson Medical School, and
| | - Vrushank Bhatt
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| | | | - Ankit Shah
- Department of Medicine, Robert Wood Johnson Medical School, and
| | - Vinod K. Rustgi
- Department of Medicine, Robert Wood Johnson Medical School, and
| | - He Wang
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, USA
| | - Paul R. Bech
- Section of Endocrinology and Investigative Medicine and
| | - Ali Abbara
- Section of Endocrinology and Investigative Medicine and
| | | | - Pinelopi Manousou
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Jessie Y. Guo
- Department of Medicine, Robert Wood Johnson Medical School, and,Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Grace L. Guo
- Department of Pharmacology and Toxicology, School of Pharmacy, and
| | - Sally Radovick
- Department of Pediatrics, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, USA
| | | | | | - Andy V. Babwah
- Department of Pediatrics, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, USA.,Child Health Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Moshmi Bhattacharya
- Department of Medicine, Robert Wood Johnson Medical School, and,Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, New Jersey, USA.,Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA.,Child Health Institute of New Jersey, New Brunswick, New Jersey, USA
| |
Collapse
|
4
|
Ibrahim SA, Adnan AA, Gahzi ST. Serum Level of Inhibin B and Kisspeptin, as well as Their Correlation with Biochemical Factors in Obese Adult Patients. ARCHIVES OF RAZI INSTITUTE 2022; 77:703-707. [PMID: 36284976 PMCID: PMC9548286 DOI: 10.22092/ari.2022.356954.1945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 01/16/2022] [Indexed: 05/24/2023]
Abstract
Obesity is one of the most important global health problems causing serious health risks and early death in human. It is also associated with disturbance of homeostasis of hormones and immunological biochemical factors inside the human body. This study aimed to evaluate the serum level of inhibin B and kisspeptin among Iraqi obese adult people and other biochemical parameters correlated with obesity. Inhibin B and levels of kisspeptin were evaluated in the samples of serum from 40 Iraqi obese adult patients and 30 healthy non-obese individuals. A significant decrease (P<0.0001) was observed in the kisspeptin level in both males and females, compared to the control group. Moreover, inhibin B decreased significantly in obese females only (P<0.001), while there was no differences between males and the control group in this regard. Finally, body mass index, serum glutamic pyruvic transaminase (SGPT), and leptin showed negative correlation with kisspeptin (0.01, 0.5, and 0.01), respectively. However, a positive association was observed with the level of Ca in the serum. On the other hand, inhibin B confirmed a positive correlation with SGPT. The present study revealed a significant increase in inhibin B and kisspeptin, with SGPT and Ca in the serum of obese patients, which could lead to complications and health problems among these patients.
Collapse
Affiliation(s)
- S A Ibrahim
- Basic Science Department, College of Nursing, University of Baghdad, Baghdad, Iraq
| | - A A Adnan
- Basic Science Department, College of Nursing, University of Baghdad, Baghdad, Iraq
| | - S T Gahzi
- Basic Science Department, College of Nursing, University of Baghdad, Baghdad, Iraq
| |
Collapse
|
5
|
Babwah AV. The wonderful and masterful G protein-coupled receptor (GPCR): A focus on signaling mechanisms and the neuroendocrine control of fertility. Mol Cell Endocrinol 2020; 515:110886. [PMID: 32574585 DOI: 10.1016/j.mce.2020.110886] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/08/2020] [Accepted: 05/20/2020] [Indexed: 12/18/2022]
Abstract
Human GnRH deficiency, both clinically and genetically, is a heterogeneous disorder comprising of congenital GnRH deficiency with anosmia (Kallmann syndrome), or with normal olfaction [normosmic idiopathic hypogonadotropic hypogonadism (IHH)], and adult-onset hypogonadotropic hypogonadism. Our understanding of the neural mechanisms underlying GnRH secretion and GnRH signaling continues to increase at a rapid rate and strikingly, the heterotrimeric guanine nucleotide-binding protein (G protein)-coupled receptors (GPCRs) continue to emerge as essential players in these processes. GPCRs were once viewed as binary on-off switches, where in the "on" state they are bound to their Gα protein, but now we understand that view is overly simplistic and does not adequately characterize GPCRs. Instead, GPCRs have emerged as masterful signaling molecules exploiting different physical conformational states of itself to elicit an array of downstream signaling events via their G proteins and the β-arrestins. The "one receptor-multiple signaling conformations" model is likely an evolved strategy that can be used to our advantage as researchers have shown that targeting specific receptor conformations via biased ligands is proving to be a powerful tool in the effective treatment of human diseases. Can biased ligands be used to selectively modulate signaling by GPCR regulators of the neuroendocrine axis in the treatment of IHH? As discussed in this review, the grand possibility exists. However, while we are still very far from developing these treatments, this exciting likelihood can happen through a much greater mechanistic understanding of how GPCRs signal within the cell.
Collapse
Affiliation(s)
- Andy V Babwah
- Department of Pediatrics, Laboratory of Human Growth and Reproductive Development, 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.
| |
Collapse
|
6
|
Kisspeptin has an independent and direct effect on the pituitary gland in the mare. Theriogenology 2020; 157:199-209. [PMID: 32814247 DOI: 10.1016/j.theriogenology.2020.07.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 06/27/2020] [Accepted: 07/26/2020] [Indexed: 11/22/2022]
Abstract
To more clearly understand the equine gonadotrope response to kisspeptin and gonadotropin releasing hormone (GnRH), peripheral LH and FSH were quantified in diestrous mares after treatment with either equine kisspeptide (eKp-10, 0.5 mg iv), GnRH (25 μg iv), or a combination thereof every 4 h for 3 days. The following observations were made: 1) a diminished LH and FSH response to eKp-10 and GnRH was observed by Day 3, but was not different by treatment, 2) a decrease in basal LH concentration was observed from Day 1 to Day 3 for the eKp-10, but not the GnRH treated mares, 3) there was no change in basal FSH with either treatment. Additionally, pre-treatment with GnRH antagonist (antide 1.0 mg iv) eliminated any measurable change in LH after eKp-10 (1.0 mg iv) treatment. Both GnRH and kisspeptin are Gαq/11 coupled receptors, therefore quantifying the rise in intracellular calcium following treatment with cognate ligand allows simultaneous assessment of receptor activation. Direct stimulation of equine primary pituitary cells with GnRH and/or eKp-10 demonstrates three distinct populations of pituitary cells: one population responded to both eKp-10 and GnRH, a second, independent population, responded to only eKp-10, and a third population responded only to GnRH. These populations were confirmed using co-immunofluorescence of hemipituitaries from mares in diestrus. Although the rise in peripheral LH concentration elicited by eKp-10 is dependent on GnRH, this work suggests that kisspeptin also has a specific and direct effect on the equine gonadotrope, independent of GnRH.
Collapse
|
7
|
Harihar S, Ray S, Narayanan S, Santhoshkumar A, Ly T, Welch DR. Role of the tumor microenvironment in regulating the anti-metastatic effect of KISS1. Clin Exp Metastasis 2020; 37:209-223. [PMID: 32088827 PMCID: PMC7339126 DOI: 10.1007/s10585-020-10030-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 02/19/2020] [Indexed: 12/29/2022]
Abstract
KISS1, a metastasis suppressor gene, has been shown to block metastasis without affecting primary tumor formation. Loss of KISS1 leads to invasion and metastasis in multiple cancers, which is the leading cause of cancer morbidity and mortality. The discovery of KISS1 has provided a ray of hope for early clinical diagnosis and for designing effective treatments targeting metastatic cancer. However, this goal requires greater holistic understanding of its mechanism of action. In this review, we go back into history and highlight some key developments, from the discovery of KISS1 to its role in regulating multiple physiological processes including cancer. We discuss key emerging roles for KISS1, specifically interactions with tissue microenvironment to promote dormancy and regulation of tumor cell metabolism, acknowledged as some of the key players in tumor progression and metastasis. We finally discuss strategies whereby KISS1 might be exploited clinically to treat metastasis.
Collapse
Affiliation(s)
- Sitaram Harihar
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India.
| | - Srijit Ray
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India
| | - Samyukta Narayanan
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India
| | - Anirudh Santhoshkumar
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India
| | - Thuc Ly
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS, 66160, USA
- The University Kansas Cancer Center, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS, 66160, USA
| | - Danny R Welch
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS, 66160, USA
- The University Kansas Cancer Center, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS, 66160, USA
| |
Collapse
|
8
|
Noonan MM, Dragan M, Mehta MM, Hess DA, Brackstone M, Tuck AB, Viswakarma N, Rana A, Babwah AV, Wondisford FE, Bhattacharya M. The matrix protein Fibulin-3 promotes KISS1R induced triple negative breast cancer cell invasion. Oncotarget 2018; 9:30034-30052. [PMID: 30046386 PMCID: PMC6059025 DOI: 10.18632/oncotarget.25682] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 06/13/2018] [Indexed: 12/18/2022] Open
Abstract
Breast cancer is a leading cause of cancer mortality. In particular, triple negative breast cancer (TNBC) comprise a heterogeneous group of basal-like tumors lacking estrogen receptor (ERα), progesterone receptor (PR) and HER2 (ErbB2). TNBC represents 15-20% of all breast cancers and occurs frequently in women under 50 years of age. Unfortunately, these patients lack targeted therapy, are typically high grade and metastatic at time of diagnosis. The mechanisms regulating metastasis remain poorly understood. We have previously shown that the kisspeptin receptor, KISS1R stimulates invasiveness of TNBC cells. In this report, we demonstrate that KISS1R signals via the secreted extracellular matrix protein, fibulin-3, to regulate TNBC invasion. We found that the fibulin-3 gene is amplified in TNBC primary tumors and that plasma fibulin-3 levels are elevated in TNBC patients compared to healthy subjects. In this study, we show that KISS1R activation increases fibulin-3 expression and secretion. We show that fibulin-3 regulates TNBC metastasis in a mouse experimental metastasis xenograft model and signals downstream of KISS1R to stimulate TNBC invasion, by activating matrix metalloproteinase 9 (MMP-9) and the MAPK pathway. These results identify fibulin-3 as a new downstream mediator of KISS1R signaling and as a potential biomarker for TNBC progression and metastasis, thus revealing KISS1R and fibulin-3 as novel drug targets in TNBC.
Collapse
Affiliation(s)
- Michelle M Noonan
- Department of Physiology and Pharmacology, The University of Western Ontario, London, ON, Canada
| | - Magdalena Dragan
- Department of Physiology and Pharmacology, The University of Western Ontario, London, ON, Canada
| | - Michael M Mehta
- Department of Physiology and Pharmacology, The University of Western Ontario, London, ON, Canada
| | - David A Hess
- Department of Physiology and Pharmacology, The University of Western Ontario, London, ON, Canada.,Krembil Centre for Stem Cell Biology, Molecular Medicine Research Group, Robarts Research Institute, London, ON, Canada
| | - Muriel Brackstone
- Department of Oncology, The University of Western Ontario, London, ON, Canada.,Lawson Health Research Institute, The University of Western Ontario, London, ON, Canada.,Division of Surgical Oncology, The University of Western Ontario, London, ON, Canada
| | - Alan B Tuck
- Department of Oncology, The University of Western Ontario, London, ON, Canada.,Department of Pathology, The University of Western Ontario, London, ON, Canada.,The Pamela Greenaway-Kohlmeier Translational Breast Cancer Research Unit, London Regional Cancer Program, London, ON, Canada
| | - Navin Viswakarma
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL, USA
| | - Ajay Rana
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL, USA
| | - Andy V Babwah
- Department of Pediatrics, Child Health Institute of NJ, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Frederic E Wondisford
- Department of Medicine, Child Health Institute of NJ, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Moshmi Bhattacharya
- Department of Physiology and Pharmacology, The University of Western Ontario, London, ON, Canada.,Department of Oncology, The University of Western Ontario, London, ON, Canada.,Lawson Health Research Institute, The University of Western Ontario, London, ON, Canada.,Department of Medicine, Child Health Institute of NJ, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| |
Collapse
|
9
|
Lu G, Wang X, Wang Y, Cheng Z, Zhou L. Value of CagA, HER2, ALDH1, and KiSS-1 in predicting metastasis and prognosis for gastric adenocarcinoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:3628-3637. [PMID: 31949743 PMCID: PMC6962848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 04/28/2018] [Indexed: 06/10/2023]
Abstract
BACKGROUND Cytotoxin-associated gene A (CagA), is able to translocate into gastric epithelial cells. Human epidermal growth factor receptor 2 (also named as HER2, is a proto-oncogene which can encode a transmembrane receptor), Aldehyde dehydrogenase 1 (ALDH1, a biomarker of cancer stem cells), and KiSS-1 (a suppressor gene of cancer metastasis) are all valuably predictive biomarkers for various human cancers. The purpose of this study is to investigate the associations among CagA, HER2, ALDH1, and KiSS-1 in gastric adenocarcinoma (GAC), and their respective associations with clinical characteristics and survival in GAC. METHODS The expression of CagA, HER2, ALDH1, and KiSS-1 in 232 cases of whole GAC tissues were detected by immunohistochemical staining. Patient clinical and survival data were also collected. RESULTS Positive expression of CagA, HER2, and ALDH1 is significantly higher, and positive expression of KiSS-1 is significantly lower, in GAC tissues than in the corresponding normal tissues. Furthermore, the positive expression of CagA, HER2, ALDH1, and KiSS-1 were significantly associated with tumor grade, tumor stage, lymph node metastasis (LNM) stage, and tumor node metastasis (TNM) stages, and with patients' overall survival (OS); whereas the KiSS-1 positive group had longer OS than did the KiSS-1 negative group. In logistic analysis, positive expression of CagA, HER2, ALDH1, and KiSS-1 are significantly associated with LNM of patients with GAC. COX regression analysis indicated that positive expression of CagA, HER2, ALDH1, and KiSS-1, and tumor stages, LNM stages, and TNM stages were independent prognostic factors for patients with GAC. CONCLUSIONS Expression of CagA, HER2, ALDH1, and KiSS-1 should be considered as promising biomarkers for metastasis and prognosis, as well as potential therapeutic targets for GAC.
Collapse
Affiliation(s)
- Guoyu Lu
- Department of Emergency, The First Affiliated Hospital of Bengbu Medical CollegeAnhui, China
| | - Xiaolin Wang
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical CollegeAnhui, China
- Department of Pathology, Bengbu Medical CollegeAnhui, China
| | - Yichao Wang
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical CollegeAnhui, China
- Department of Pathology, Bengbu Medical CollegeAnhui, China
| | - Zenong Cheng
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical CollegeAnhui, China
- Department of Pathology, Bengbu Medical CollegeAnhui, China
| | - Lei Zhou
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical CollegeAnhui, China
- Department of Pathology, Bengbu Medical CollegeAnhui, China
| |
Collapse
|
10
|
Yue X, Han Z, Zhang L, Li J, Gong X. Aberrant expression of ALDH1, MMP9, Integrin αvβ3, and KiSS-1 in invasive ductal carcinoma and their clinical significance. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:3511-3522. [PMID: 31949729 PMCID: PMC6962838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 05/10/2018] [Indexed: 06/10/2023]
Abstract
BACKGROUND Aldehyde dehydrogenase 1 (ALDH1, a biomarker of cancer stem cells), matrix metalloproteinase 9 (MMP9, known as a matrilysin), Integrin αvβ3 (known as a biomarker of cell-matrix adhesion) and KiSS-1 (suppressor gene of tumor metastasis) are all related to cancer invasion and metastasis in many cancers. The purpose of this study was to investigate the expression of ALDH1, MMP9, Integrin αvβ3, and KiSS-1 in invasive ductal carcinoma (IDC), and their respective associations with clinical characteristics and survival in IDC. METHODS Immunohistochemical staining was used to detect the expression of ALDH1, MMP9, Integrin αvβ3, and KiSS-1 in 227 whole IDC tissue specimens. Patients' clinical and demographic data were both collected. RESULTS The expression of ALDH1, MMP9, and Integrin αvβ3 were significantly higher in IDC tissues than in the control tissues. The positive expressions of ALDH1, MMP9, and Integrin αvβ3 were positively associated with tumor grades, lymph node metastasis (LNM), tumor stages, and tumor node metastasis (TNM) stages, and inversely with overall survival (OS) and recurrence-free survival (RFS). Positive expression of KiSS-1 was negatively associated with tumor grades, LNM, tumor stages, and TNM stages, but positively with OS and RFS. A multivariate analysis demonstrated that the positive expression of ALDH1, MMP9, Integrin αvβ3, KiSS-1, ER, and HER-2, as well as TNM stages were independent prognostic factors for OS and RFS in IDC. CONCLUSIONS The expression of ALDH1, MMP9, Integrin αvβ3, and KiSS-11 should represent promising biomarkers in predicting metastasis and prognosis, as well as being potential therapeutic targets for IDC.
Collapse
Affiliation(s)
- Xicheng Yue
- Department of Surgical Oncology, The First Affiliated Hospital of Bengbu Medical UniversityAnhui, China
| | - Zhengquan Han
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical UniversityAnhui, China
| | - Ligong Zhang
- Department of Surgical Oncology, The First Affiliated Hospital of Bengbu Medical UniversityAnhui, China
| | - Jing Li
- Department of Surgical Oncology, The First Affiliated Hospital of Bengbu Medical UniversityAnhui, China
| | - Xiaomeng Gong
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical UniversityAnhui, China
- Department of Pathology, Bengbu Medical UniversityAnhui, China
| |
Collapse
|
11
|
Effect of Kisspeptin on the Developmental Competence and Early Transcript Expression in Porcine Oocytes Parthenogenetically Activated with Different Methods. BIOMED RESEARCH INTERNATIONAL 2018; 2018:3693602. [PMID: 29682539 PMCID: PMC5841116 DOI: 10.1155/2018/3693602] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 01/10/2018] [Accepted: 01/17/2018] [Indexed: 12/02/2022]
Abstract
Recent studies showed the modulatory effect of kisspeptin (KP) on calcium waves through the cell membrane and inside the cell. Spermatozoon can induce similar ooplasmic calcium oscillations at fertilization to trigger meiosis II. Here, we evaluated the effect of KP supplementation with 6-dimethylaminopurine (6-DMAP) for 4 h on embryonic development after oocyte activation with single electric pulse, 5 µM ionomycin, or 8% ethanol. Compared to control nonsupplemented groups, KP significantly improved embryo developmental competence electric- and ethanol-activated oocytes in terms of cleavage (75.3% and 58.6% versus 64% and 48%, respectively, p < 0.05) and blastocyst development (31.3% and 10% versus 19.3% and 4%, respectively, p < 0.05). MOS expression was increased in electrically activated oocytes in presence of KP while it significantly reduced CCNB1 expression. In ionomycin treated group, both MOS and CCNB1 showed significant increase with no difference between KP and control groups. In ethanol-treated group, KP significantly reduced CCNB1 but no effect was observed on MOS expression. The early alterations in MOS and CCNB1 mRNA transcripts caused by KP may explain the significant differences in the developmental competence between the experimental groups. Kisspeptin supplementation may be adopted in protocols for porcine oocyte activation through electric current and ethanol to improve embryonic developmental competence.
Collapse
|
12
|
Mezei Z, Váczi S, Török V, Stumpf C, Ónody R, Földesi I, Szabó G. Effects of kisspeptin on diabetic rat platelets. Can J Physiol Pharmacol 2017; 95:1319-1326. [DOI: 10.1139/cjpp-2017-0036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Hyperglycemia, hyperlipidemia, and free radicals result in platelet activation and atherogenesis. Kisspeptin (KP) is able to regulate metabolism, hemostasis, and the development of atherosclerosis. We examined whether platelet aggregation of streptozotocin-induced diabetic rats depends on the inducer type and if KP-13 and RF-9 (a kisspeptin receptor modifier) can influence platelet function. We measured the speed and the maximum of aggregation, along with the area under the curve. Serum glucose and calcium levels and urine formation of diabetic animals increased, while the body mass and platelet count decreased. Collagen was the most effective inducer of platelet aggregation. The aggregability of nondiabetic platelets was elevated in the presence of 5 × 10−8 mol/L KP-13. This effect was less expressed in diabetic animals. The effectivity of RF-9 was stronger than that of KP-13 in nondiabetic platelets, however it was ineffective in diabetic animals. RF-9 pre-treatment did not change the effects of 5 × 10−8 mol/L KP-13 in either animal group. The in vivo activation of diabetic platelets, which may be due to elevated serum calcium, induces thrombocytopenia and may lead to reduced in vitro aggregability. We could not demonstrate the antagonistic effect of RF-9 against KP-13 in isolated platelets.
Collapse
Affiliation(s)
- Zsófia Mezei
- Department of Pathophysiology, University of Szeged, Semmelweis u. 1, 6725 Szeged, Hungary
| | - Sándor Váczi
- Department of Pathophysiology, University of Szeged, Semmelweis u. 1, 6725 Szeged, Hungary
| | - Viktória Török
- Department of Pathophysiology, University of Szeged, Semmelweis u. 1, 6725 Szeged, Hungary
| | - Csaba Stumpf
- Department of Pathophysiology, University of Szeged, Semmelweis u. 1, 6725 Szeged, Hungary
| | - Rita Ónody
- Institute of Laboratory Medicine, University of Szeged, Semmelweis u. 6, 6725 Szeged, Hungary
| | - Imre Földesi
- Institute of Laboratory Medicine, University of Szeged, Semmelweis u. 6, 6725 Szeged, Hungary
| | - Gyula Szabó
- Department of Pathophysiology, University of Szeged, Semmelweis u. 1, 6725 Szeged, Hungary
| |
Collapse
|
13
|
Yu L, Zhu B, Wu S, Zhou L, Song W, Gong X, Wang D. Evaluation of the correlation of vasculogenic mimicry, ALDH1, KiSS-1, and MACC1 in the prediction of metastasis and prognosis in ovarian carcinoma. Diagn Pathol 2017; 12:23. [PMID: 28253891 PMCID: PMC5335811 DOI: 10.1186/s13000-017-0612-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 02/20/2017] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Recurrence and metastasis are the usual manifestations of treatment failure of epithelial ovarian carcinoma (EOC). Vasculogenic mimicry (VM; blood supply development often seen in highly aggressive cancers), aldehyde dehydrogenase 1 (ALDH1, cancer stem cell biomarker), KiSS-1 (suppressor of tumor metastasis), and metastasis associated in colon cancer-1 (MACC1) are all useful predictive factors for metastasis and prognosis in various cancers. In this study, we analyzed associations among VM, ALDH1, KiSS-1, and MACC1 in EOC, and their respective correlations with clinicopathological characteristics and survival in EOC. METHODS Positive rates of VM, ALDH1, KiSS-1, and MACC1 in 207 whole EOC tissue samples were detected by immunohistochemistry. Patients' clinical data were also collected. RESULTS Levels of VM, ALDH1, and MACC1 were significantly higher, and levels of KiSS-1 significantly lower, in EOC tissues than in benign ovary tumors. Levels of VM, ALDH1, KiSS-1, and MACC1 were associated significantly with tumor/lymph node/metastasis (LNM) grade, implantation, and International Federation of Gynecology and Obstetrics (FIGO) stage, and with patients' overall survival (OS); whereas the KiSS-1+ subgroup had significantly longer OS than did the KiSS-1- subgroup. In multivariate analysis, high VM, ALDH1 or MACC1 levels, FIGO stage, implantation and low KiSS-1 levels were independently associated with shorter OS in patients with EOC. CONCLUSIONS VM and expressions of ALDH1, KiSS-1, and MACC1 represent promising markers for metastasis and prognosis, and potential therapeutic targets for EOC.
Collapse
Affiliation(s)
- Lan Yu
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, No.287, Changhuai Road, Bengbu, 233003, Anhui Province, China
| | - Bo Zhu
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, No.287, Changhuai Road, Bengbu, 233003, Anhui Province, China
| | - Shiwu Wu
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, No.287, Changhuai Road, Bengbu, 233003, Anhui Province, China.
| | - Lei Zhou
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, No.287, Changhuai Road, Bengbu, 233003, Anhui Province, China
| | - Wenqing Song
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, No.287, Changhuai Road, Bengbu, 233003, Anhui Province, China
| | - Xiaomeng Gong
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, No.287, Changhuai Road, Bengbu, 233003, Anhui Province, China
| | - Danna Wang
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, No.287, Changhuai Road, Bengbu, 233003, Anhui Province, China
| |
Collapse
|
14
|
Abstract
Kisspeptins are a group of peptide fragments encoded by the KISS1 gene in humans. They bind to kisspeptin receptors with equal efficacy. Kisspeptins and their receptors are expressed by neurons in the arcuate and anteroventral periventricular nuclei of the hypothalamus. Oestrogen mediates negative feedback of gonadotrophin-releasing hormone secretion via the arcuate nucleus. Conversely, it exerts positive feedback via the anteroventral periventricular nucleus. The sexual dimorphism of these nuclei accounts for the differential behaviour of the hypothalamic-pituitary-gonadal axis between genders. Kisspeptins are essential for reproductive function. Puberty is regulated by the maturation of kisspeptin neurons and by interactions between kisspeptins and leptin. Hence, kisspeptins have potential diagnostic and therapeutic applications. Kisspeptin agonists may be used to localise lesions in cases of hypothalamic-pituitary-gonadal axis dysfunction and evaluate the gonadotrophic potential of subfertile individuals. Kisspeptin antagonists may be useful as contraceptives in women, through the prevention of premature luteinisation during in vitro fertilisation, and in the treatment of sex steroid-dependent diseases and metastatic cancers.
Collapse
Affiliation(s)
- Eng Loon Tng
- Associate Consultant, Department of Medicine, Ng Teng Fong General Hospital, 1 Jurong East Street 21, Singapore 609606
| |
Collapse
|
15
|
Min L, Nie M, Zhang A, Wen J, Noel SD, Lee V, Carroll RS, Kaiser UB. Computational Analysis of Missense Variants of G Protein-Coupled Receptors Involved in the Neuroendocrine Regulation of Reproduction. Neuroendocrinology 2016; 103:230-9. [PMID: 26088945 PMCID: PMC4684493 DOI: 10.1159/000435884] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 06/10/2015] [Indexed: 01/13/2023]
Abstract
INTRODUCTION Many missense variants in G protein-coupled receptors (GPCRs) involved in the neuroendocrine regulation of reproduction have been identified by phenotype-driven or large-scale exome sequencing. Computational functional prediction analysis is commonly performed to evaluate their impact on receptor function. METHODS To assess the performance and outcome of functional prediction analyses for these GPCRs, we performed a statistical analysis of the prediction performance of SIFT and PolyPhen-2 for variants with documented biological function as well as variants retrieved from Ensembl. We obtained missense variants with documented biological function testing from patients with reproductive disorders from a comprehensive literature search. Missense variants from individuals with known reproductive disorders were retrieved from the Human Gene Mutation Database. Missense variants from the general population were retrieved from the Ensembl genome database. RESULTS The accuracies of SIFT and PolyPhen-2 were 83 and 85%, respectively. The performance of both prediction tools was greater in predicting loss-of-function variants (SIFT: 92%; PolyPhen-2: 95%) than in predicting variants that did not affect function (SIFT: 54%; PolyPhen-2: 57%). Concordance between SIFT and PolyPhen-2 did not improve accuracy. Surprisingly, approximately half of the variants retrieved from Ensembl were predicted as loss-of-function variants by SIFT (47%) and PolyPhen-2 (54%). CONCLUSION Our findings provide new guidance for interpreting the results and limitations of computational functional prediction analyses for GPCRs and will help to determine which variants require biological function testing. In addition, our findings raise important questions regarding the link between genotype and phenotype in the general population.
Collapse
Affiliation(s)
- Le Min
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, Boston, MA, 02115 USA
- To whom correspondence and reprint requests should be addressed: Le Min, M.D., Ph.D., Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, 221 Longwood Avenue, Boston, Massachusetts 02115.
| | - Min Nie
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, Boston, MA, 02115 USA
| | - Anna Zhang
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, Boston, MA, 02115 USA
| | - Junping Wen
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, Boston, MA, 02115 USA
| | - Sekoni D. Noel
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, Boston, MA, 02115 USA
| | - Vivian Lee
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, Boston, MA, 02115 USA
| | - Rona S. Carroll
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, Boston, MA, 02115 USA
| | - Ursula B. Kaiser
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, Boston, MA, 02115 USA
| |
Collapse
|
16
|
Prabhu VV, Sakthivel KM, Guruvayoorappan C. Kisspeptins (KiSS-1): essential players in suppressing tumor metastasis. Asian Pac J Cancer Prev 2015; 14:6215-20. [PMID: 24377507 DOI: 10.7314/apjcp.2013.14.11.6215] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Kisspeptins (KPs) encoded by the KiSS-1 gene are C-terminally amidated peptide products, including KP- 10, KP-13, KP-14 and KP-54, which are endogenous agonists for the G-protein coupled receptor-54 (GPR54). Functional analyses have demonstrated fundamental roles of KiSS-1 in whole body homeostasis including sexual differentiation of brain, action on sex steroids and metabolic regulation of fertility essential for human puberty and maintenance of adult reproduction. In addition, intensive recent investigations have provided substantial evidence suggesting roles of Kisspeptin signalling via its receptor GPR54 in the suppression of metastasis with a variety of cancers. The present review highlights the latest studies regarding the role of Kisspeptins and the KiSS-1 gene in tumor progression and also suggests targeting the KiSS-1/GPR54 system may represent a novel therapeutic approach for cancers. Further investigations are essential to elucidate the complex pathways regulated by the Kisspeptins and how these pathways might be involved in the suppression of metastasis across a range of cancers.
Collapse
|
17
|
Pampillo M, Babwah AV. Quantifying GPCR internalization: a focus on the Kisspeptin receptor. Methods Mol Biol 2015; 1272:119-132. [PMID: 25563181 DOI: 10.1007/978-1-4939-2336-6_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
GPCR internalization is a critical regulatory step in determining receptor activity. While internalization terminates G protein-coupled signaling, it might be required for G protein-independent signaling. A large number of clinical therapies are based on preventing or promoting GPCR internalization. Thus, for any given GPCR, it is important to characterize its internalization and understand the factors that regulate such internalization. Here we describe different experimental protocols to evaluate the internalization of any GPCR transiently expressed in HEK 293 cells. The protocols describe the use of immunofluorescence and imaging techniques as well as flow cytometry. The techniques described use the FLAG-tagged kisspeptin receptor (KISS1R) as an example but are equally applicable to any other GPCR.
Collapse
Affiliation(s)
- Macarena Pampillo
- The Children's Health Research Institute, Victoria Research Laboratories, The University of Western Ontario, A4-140, 800 Commissioners Road East, London, ON, Canada, N6C 2V5
| | | |
Collapse
|
18
|
Millar RP, Babwah AV. KISS1R: Hallmarks of an Effective Regulator of the Neuroendocrine Axis. Neuroendocrinology 2015; 101:193-210. [PMID: 25765628 DOI: 10.1159/000381457] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 03/04/2015] [Indexed: 11/19/2022]
Abstract
Kisspeptin (KP) is now well recognized as a potent stimulator of gonadotropin-releasing hormone (GnRH) secretion and thereby a major regulator of the neuroendocrine-reproductive axis. KP signals via KISS1R, a G protein-coupled receptor (GPCR) that activates the G proteins Gαq/11. Modulation of the interaction of KP with KISS1R is therefore a potential new therapeutic target for stimulating (in infertility) or inhibiting (in hormone-dependent diseases) the reproductive hormone cascade. Major efforts are underway to target KISS1R in the treatment of sex steroid hormone-dependent disorders and to stimulate endogenous hormonal responses along the neuroendocrine axis as part of in vitro fertilization protocols. The development of analogs modulating KISS1R signaling will be aided by an understanding of the intracellular pathways and dynamics of KISS1R signaling under normal and pathological conditions. This review focuses on KISS1R recruitment of intracellular signaling (Gαq/11- and β-arrestin-dependent) pathways that mediate GnRH secretion and the respective roles of rapid desensitization, internalization, and recycling of resensitized receptors in maintaining an active population of KISS1R at the cell surface to facilitate prolonged KP signaling. Additionally, this review summarizes and discusses the major findings of an array of studies examining the desensitization of KP signaling in man, domestic and laboratory animals. This discussion highlights the major effects of ligand efficacy and concentration and the physiological, developmental, and metabolic status of the organism on KP signaling. Finally, the potential for the utilization of KP and analogs in stimulating and inhibiting the reproductive hormone cascade as an alternative to targeting the downstream GnRH receptor is discussed.
Collapse
Affiliation(s)
- Robert P Millar
- Mammal Research Institute, University of Pretoria, Pretoria, South Africa
| | | |
Collapse
|
19
|
Ahow M, Min L, Pampillo M, Nash C, Wen J, Soltis K, Carroll RS, Glidewell-Kenney CA, Mellon PL, Bhattacharya M, Tobet SA, Kaiser UB, Babwah AV. KISS1R signals independently of Gαq/11 and triggers LH secretion via the β-arrestin pathway in the male mouse. Endocrinology 2014; 155:4433-46. [PMID: 25147978 PMCID: PMC4197989 DOI: 10.1210/en.2014-1304] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Hypothalamic GnRH is the master regulator of the neuroendocrine reproductive axis, and its secretion is regulated by many factors. Among these is kisspeptin (Kp), a potent trigger of GnRH secretion. Kp signals via the Kp receptor (KISS1R), a Gαq/11-coupled 7-transmembrane-spanning receptor. Until this study, it was understood that KISS1R mediates GnRH secretion via the Gαq/11-coupled pathway in an ERK1/2-dependent manner. We recently demonstrated that KISS1R also signals independently of Gαq/11 via β-arrestin and that this pathway also mediates ERK1/2 activation. Because GnRH secretion is ERK1/2-dependent, we hypothesized that KISS1R regulates GnRH secretion via both the Gαq/11- and β-arrestin-coupled pathways. To test this hypothesis, we measured LH secretion, a surrogate marker of GnRH secretion, in mice lacking either β-arrestin-1 or β-arrestin-2. Results revealed that Kp-dependent LH secretion was significantly diminished relative to wild-type mice (P < .001), thus supporting that β-arrestin mediates Kp-induced GnRH secretion. Based on this, we hypothesized that Gαq/11-uncoupled KISS1R mutants, like L148S, will display Gαq/11-independent signaling. To test this hypothesis, L148S was expressed in HEK 293 cells. and results confirmed that, although strongly uncoupled from Gαq/11, L148S retained the ability to trigger significant Kp-dependent ERK1/2 phosphorylation (P < .05). Furthermore, using mouse embryonic fibroblasts lacking β-arrestin-1 and -2, we demonstrated that L148S-mediated ERK1/2 phosphorylation is β-arrestin-dependent. Overall, we conclude that KISS1R signals via Gαq/11 and β-arrestin to regulate GnRH secretion. This novel and important finding could explain why patients bearing some types of Gαq/11-uncoupled KISS1R mutants display partial gonadotropic deficiency and even a reversal of the condition, idiopathic hypogonadotropic hypogonadism.
Collapse
|
20
|
Beltramo M, Dardente H, Cayla X, Caraty A. Cellular mechanisms and integrative timing of neuroendocrine control of GnRH secretion by kisspeptin. Mol Cell Endocrinol 2014; 382:387-399. [PMID: 24145132 DOI: 10.1016/j.mce.2013.10.015] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Revised: 10/08/2013] [Accepted: 10/11/2013] [Indexed: 01/11/2023]
Abstract
The hypothalamus integrates endogenous and exogenous inputs to control the pituitary-gonadal axis. The ultimate hypothalamic influence on reproductive activity is mediated through timely secretion of GnRH in the portal blood, which modulates the release of gonadotropins from the pituitary. In this context neurons expressing the RF-amide neuropeptide kisspeptin present required features to fulfill the role of the long sought-after hypothalamic integrative centre governing the stimulation of GnRH neurons. Here we focus on the intracellular signaling pathways triggered by kisspeptin through its cognate receptor KISS1R and on the potential role of proteins interacting with this receptor. We then review evidence implicating both kisspeptin and RFRP3--another RF-amide neuropeptide--in the temporal orchestration of both the pre-ovulatory LH surge in female rodents and the organization of seasonal breeding in photoperiodic species.
Collapse
Affiliation(s)
- Massimiliano Beltramo
- UMR Physiologie de la Reproduction et des Comportements (INRA, UMR85, CNRS, UMR7247, Université François Rabelais Tours, IFCE), F-37380 Nouzilly, France.
| | - Hugues Dardente
- UMR Physiologie de la Reproduction et des Comportements (INRA, UMR85, CNRS, UMR7247, Université François Rabelais Tours, IFCE), F-37380 Nouzilly, France
| | - Xavier Cayla
- UMR Physiologie de la Reproduction et des Comportements (INRA, UMR85, CNRS, UMR7247, Université François Rabelais Tours, IFCE), F-37380 Nouzilly, France
| | - Alain Caraty
- UMR Physiologie de la Reproduction et des Comportements (INRA, UMR85, CNRS, UMR7247, Université François Rabelais Tours, IFCE), F-37380 Nouzilly, France
| |
Collapse
|
21
|
Matsui H, Asami T. Effects and therapeutic potentials of kisspeptin analogs: regulation of the hypothalamic-pituitary-gonadal axis. Neuroendocrinology 2014; 99:49-60. [PMID: 24356680 DOI: 10.1159/000357809] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 12/07/2013] [Indexed: 11/19/2022]
Abstract
The hypothalamic peptide kisspeptin (metastin), the endogenous ligand of the G protein-coupled receptor KISS1R, plays a critical role in controlling GnRH release from hypothalamic GnRH neurons and thereby regulates hypothalamic-pituitary-gonadal functions. Although the therapeutic potential of kisspeptin is attractive, its susceptibility to proteolytic degradation limits its utility. To overcome this, KISS1R agonists or antagonists as peptide analogs or small molecules have been investigated. Kisspeptin analogs have been most extensively studied by reducing the length of the peptide from the original 54 amino acids to 10 amino acids or less and by substituting key amino acid residues. Also, 2 investigational kisspeptin agonist analogs have been evaluated in clinical studies in men; in agreement with animal studies, abrupt elevations in gonadotropin and testosterone levels were observed as an acute effect, followed by rapid reductions in these hormones as a chronic effect. Some studies of small-molecule KISS1R antagonists have also been published. In this review, we present a brief overview on kisspeptin/KISS1R physiology in reproductive functions and summarize the available knowledge of both agonists and antagonists. We also focus on the kisspeptin agonist analogs by summarizing key pharmacological findings from both clinical and preclinical studies, and discuss their potential therapeutic utility.
Collapse
Affiliation(s)
- Hisanori Matsui
- Extra Value Generation and General Medicine Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | | |
Collapse
|
22
|
Min L, Soltis K, Reis ACS, Xu S, Kuohung W, Jain M, Carroll RS, Kaiser UB. Dynamic kisspeptin receptor trafficking modulates kisspeptin-mediated calcium signaling. Mol Endocrinol 2013; 28:16-27. [PMID: 24295737 DOI: 10.1210/me.2013-1165] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Kisspeptin receptor (KISS1R) signaling plays a critical role in the regulation of reproduction. We investigated the role of kisspeptin-stimulated KISS1R internalization, recycling, and degradation in the modulation of KISS1R signaling. Kisspeptin stimulation of Chinese hamster ovary or GT1-7 cells expressing KISS1R resulted in a biphasic increase in intracellular Ca(2+) ([Ca(2+)]i), with a rapid acute increase followed by a more sustained second phase. In contrast, stimulation of the TRH receptor, another Gq/11-coupled receptor, resulted in a much smaller second-phase [Ca(2+)]i response. The KISS1R-mediated second-phase [Ca(2+)]i response was abolished by removal of kisspeptin from cell culture medium. Notably, the second-phase [Ca(2+)]i response was also inhibited by dynasore, brefeldin A, and phenylarsine oxide, which inhibit receptor internalization and recycling, suggesting that KISS1R trafficking contributes to the sustained [Ca(2+)]i response. We further demonstrated that KISS1R undergoes dynamic ligand-dependent and -independent recycling. We next investigated the fate of the internalized kisspeptin-KISS1R complex. Most internalized kisspeptin was released extracellularly in degraded form within 1 hour, suggesting rapid processing of the internalized kisspeptin-KISS1R complex. Using a biotinylation assay, we demonstrated that degradation of cell surface KISS1R was much slower than that of the internalized ligand, suggesting dissociated processing of the internalized kisspeptin-KISS1R complex. Taken together, our results suggest that the sustained calcium response to kisspeptin is dependent on the continued presence of extracellular ligand and is the result of dynamic KISS1R trafficking.
Collapse
Affiliation(s)
- Le Min
- Division of Endocrinology, Diabetes and Hypertension (L.M., K.S., A.C.S.R., S.X., W.K., M.J., R.S.C., U.B.K.), Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115; and School of Medicine of Ribeirao Preto (A.C.S.R.), University of Sao Paulo, Brazil 14040-900
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Taylor J, Pampillo M, Bhattacharya M, Babwah AV. Kisspeptin/KISS1R signaling potentiates extravillous trophoblast adhesion to type-I collagen in a PKC- and ERK1/2-dependent manner. Mol Reprod Dev 2013; 81:42-54. [PMID: 24273038 DOI: 10.1002/mrd.22279] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 11/01/2013] [Indexed: 12/16/2022]
Abstract
During the first trimester of human pregnancy, cytotrophoblasts proliferate within the tips of the chorionic villi to form cell columns that anchor the placenta to the uterus. This migration coincides with a widespread change in the adhesion molecule repertoire of these trophoblasts. Kisspeptin and its receptor, KISS1R, are best known as potent triggers of gonadotropin-releasing hormone secretion. The kisspeptin/KISS1R signaling system is also highly expressed in the human placenta, where it was demonstrated to inhibit extra-villous trophoblast (EVT) migration and invasion in vitro. Here we show that kisspeptin, in a dose- and time-dependent manner, induces increased adhesion of human EVTs to type-I collagen, a major component of the human placenta. This increased adhesion was both rapid and transient, suggesting that it likely occurred through the activation of KISS1R secondary effectors such as PKC and ERK, which underwent rapid and transient kisspeptin-dependent activation in EVTs. We then showed that inhibition of both PKC and ERK1/2 attenuated the kisspeptin-dependent increase in EVT adhesion, suggesting that these molecules are key positive regulators of trophoblast adhesion. We therefore propose that kisspeptin/KISS1R signaling potentiates EVT adhesion to type-I collagen via "inside-out signaling." Furthermore, kisspeptin treatment increased mouse blastocyst adhesion to collagen I, suggesting that kisspeptin signaling is a key regulator of trophoblast function during implantation as well as early placentation.
Collapse
Affiliation(s)
- Jay Taylor
- The Children's Health Research Institute, London, Ontario, Canada; Lawson Health Research Institute, London, Ontario, Canada; Department of Physiology and Pharmacology, The University of Western Ontario, London, Ontario, Canada
| | | | | | | |
Collapse
|
24
|
Cvetković D, Babwah AV, Bhattacharya M. Kisspeptin/KISS1R System in Breast Cancer. J Cancer 2013; 4:653-61. [PMID: 24155777 PMCID: PMC3805993 DOI: 10.7150/jca.7626] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 09/22/2013] [Indexed: 01/18/2023] Open
Abstract
Kisspeptins (KP), peptide products of the kisspeptin-1 (KISS1) gene are the endogenous ligands for a G protein-coupled receptor (GPCR) - KP receptor (KISS1R). KISS1R couples to the Gαq/11 signaling pathway. KISS1 is a metastasis suppressor gene and the KP/KISS1R signaling has anti-metastatic and tumor-suppressant effects in numerous human cancers. On the other hand, recent studies indicate that KP/KISS1R pathway plays detrimental roles in breast cancer. In this review, we summarize recent developments in the understanding of the mechanisms regulating KP/KISS1R signaling in breast cancer metastasis.
Collapse
Affiliation(s)
- Donna Cvetković
- 1. Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | | | | |
Collapse
|