1
|
Basu R, Boguszewski CL, Kopchick JJ. Growth Hormone Action as a Target in Cancer: Significance, Mechanisms, and Possible Therapies. Endocr Rev 2025; 46:224-280. [PMID: 39657053 DOI: 10.1210/endrev/bnae030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 08/29/2024] [Accepted: 12/03/2024] [Indexed: 12/17/2024]
Abstract
Growth hormone (GH) is a pituitary-derived endocrine hormone required for normal postnatal growth and development. Hypo- or hypersecretion of endocrine GH results in 2 pathologic conditions, namely GH deficiency (GHD) and acromegaly. Additionally, GH is also produced in nonpituitary and tumoral tissues, where it acts rather as a cellular growth factor with an autocrine/paracrine mode of action. An increasingly persuasive and large body of evidence over the last 70 years concurs that GH action is implicit in escalating several cancer-associated events, locally and systemically. This pleiotropy of GH's effects is puzzling, but the association with cancer risk automatically raises a concern for patients with acromegaly and for individuals treated with GH. By careful assessment of the available knowledge on the fundamental concepts of cancer, suggestions from epidemiological and clinical studies, and the evidence from specific reports, in this review we aimed to help clarify the distinction of endocrine vs autocrine/paracrine GH in promoting cancer and to reconcile the discrepancies between experimental and clinical data. Along this discourse, we critically weigh the targetability of GH action in cancer-first by detailing the molecular mechanisms which posit GH as a critical node in tumor circuitry; and second, by enumerating the currently available therapeutic options targeting GH action. On the basis of our discussion, we infer that a targeted intervention on GH action in the appropriate patient population can benefit a sizable subset of current cancer prognoses.
Collapse
Affiliation(s)
- Reetobrata Basu
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA
- Department of Biomedical Sciences, Ohio University Heritage College of Osteopathic Medicine (OU-HCOM), Athens, OH 45701, USA
- Diabetes Institute, Ohio University Heritage College of Osteopathic Medicine (OU-HCOM), Athens, OH 45701, USA
| | - Cesar L Boguszewski
- SEMPR, Endocrine Division, Department of Internal Medicine, Federal University of Parana, Curitiba 80060-900, Brazil
| | - John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA
- Department of Biomedical Sciences, Ohio University Heritage College of Osteopathic Medicine (OU-HCOM), Athens, OH 45701, USA
- Diabetes Institute, Ohio University Heritage College of Osteopathic Medicine (OU-HCOM), Athens, OH 45701, USA
- Molecular and Cellular Biology Program, Ohio University, Athens, OH 45701, USA
| |
Collapse
|
2
|
Luo H, Huang Z, Mo X, Long C, Wang K, Deng R, Zhu X, Zeng Z. Synthesis of fluorinated tubastatin A derivatives with bi-, tri-, and tetracyclic cap groups: molecular docking with HDAC6 and evaluation of in vitro antitumor activity. RSC Med Chem 2025:d4md00898g. [PMID: 40027347 PMCID: PMC11865918 DOI: 10.1039/d4md00898g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 01/30/2025] [Indexed: 03/05/2025] Open
Abstract
Herein, we report the synthesis of 16 tubastatin A derivatives with fluorinated bi-, tri-, and tetracyclic cap groups. Most derivatives show strong in vitro antitumor activity, achieving micromolar or sub-micromolar efficacy. The most promising compound, 4-(6-bromo-3,3-difluoro-1,2,3,4-tetrahydro-9H-carbozol-9-yl)methyl)-N-hydroxybenzamide (14f), demonstrated potent anti-proliferative effects against human nasopharyngeal carcinoma cells (SUNE1) and human breast cancer cells (MDA-MB-231), with IC50 values of 0.51 μM and 0.52 μM, respectively. Notably, compound 4-((8-fluoroindeno[2,1-b]indol-5(6H)-yl)-N-hydroxybenzamide (13c) exhibited significant anti-proliferative activity against pancreatic cancer cells (SW1990), with an IC50 of 2.06 μM and low cytotoxicity to normal cells. Overall, variations in the cap group from bi- to tri-, then to tetracyclic, and the introduction of fluorinated groups enhance the antitumor activity of these derivatives. Among them, difluoromethyl-modified tricyclic derivatives show a broad spectrum in vitro antitumor effect. Molecular docking studies indicate that these derivatives bind to Histone Deacetylase 6 (HDAC6) at low binding energies, ranging from -6.54 to -9.84 kcal mol-1, through metal complexation, hydrogen bonding, π-π stacking, and π-cation interactions, which correlates with their good antitumor activity. Compound 4-((2-fluoro-5,6-dihydro-7H-benzo[c]carbazol-7-yl)methyl)-N-hydroxybenzamide (13a) with the lowest binding energy of -9.84 kcal mol-1 exhibited the best in vitro antitumor activity against MCF-7, with IC50 of 1.98 μM.
Collapse
Affiliation(s)
- Huaxin Luo
- School of Chemistry, South China Normal University Guangzhou 510006 People's Republic of China
| | - Zheng Huang
- School of Chemistry, South China Normal University Guangzhou 510006 People's Republic of China
| | - Xiangdong Mo
- School of Chemistry, South China Normal University Guangzhou 510006 People's Republic of China
| | - Chunmei Long
- School of Chemistry, South China Normal University Guangzhou 510006 People's Republic of China
| | - Kaiyuan Wang
- School of Chemistry, South China Normal University Guangzhou 510006 People's Republic of China
| | - Rong Deng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center Guangzhou 510060 China
| | - Xiaofeng Zhu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center Guangzhou 510060 China
| | - Zhuo Zeng
- School of Chemistry, South China Normal University Guangzhou 510006 People's Republic of China
| |
Collapse
|
3
|
Cortesi M, Bravaccini S, Ravaioli S, Petracci E, Angeli D, Tumedei MM, Balzi W, Pirini F, Zanoni M, Possanzini P, Rocca A, Palleschi M, Ulivi P, Martinelli G, Maltoni R. HDAC6 as a Prognostic Factor and Druggable Target in HER2-Positive Breast Cancer. Cancers (Basel) 2024; 16:3752. [PMID: 39594707 PMCID: PMC11591923 DOI: 10.3390/cancers16223752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 10/27/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Adjuvant trastuzumab is the standard of care for HER2+ breast cancer (BC) patients. However, >50% of patients become resistant. This study aimed at the identification of the molecular factors associated with disease relapse and their further investigation as therapeutically exploitable targets. METHODS Analyses were conducted on formalin-fixed paraffin-embedded tissues of the primary tumors of relapsed (cases) and not relapsed (controls) HER2+ BC patients treated with adjuvant trastuzumab. The nCounter Human Breast Cancer Panel 360 was used. Logistic regression and partitioning around medoids were employed to identify the genes associated with disease recurrence. Cytotoxicity experiments using trastuzumab-resistant cell lines and a network pharmacology approach were carried out to investigate drug efficacy. RESULTS A total of 52 patients (26 relapsed and 26 not relapsed) were analyzed. We found that a higher expression of HDAC6 was significantly associated with an increased risk of recurrence, with an adjusted OR of 3.20 (95% CI 1.38-9.91, p = 0.016). Then, we investigated the cytotoxic activity of the selective HDAC6 inhibitor Nexturastat A (NextA) on HER2+ cell lines, which were both sensitive and trastuzumab-resistant. A sub-cytotoxic concentration of NextA, combined with trastuzumab, showed a synergistic effect on BC cell lines. Finally, using a network pharmacology approach, we identified HSP90AA1 as the putative molecular candidate responsible for the synergism observed in vitro. CONCLUSIONS Our findings encourage the exploration of the role of HDAC6 as a prognostic factor and the combinatorial use of HDAC6 selective inhibitors combined with trastuzumab in HER2+ BC, in particular for those patients experiencing drug resistance.
Collapse
Affiliation(s)
- Michela Cortesi
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.C.); (R.M.)
| | - Sara Bravaccini
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.C.); (R.M.)
| | - Sara Ravaioli
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.C.); (R.M.)
| | - Elisabetta Petracci
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.C.); (R.M.)
| | - Davide Angeli
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.C.); (R.M.)
| | - Maria Maddalena Tumedei
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.C.); (R.M.)
| | - William Balzi
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.C.); (R.M.)
| | - Francesca Pirini
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.C.); (R.M.)
| | - Michele Zanoni
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.C.); (R.M.)
| | - Paola Possanzini
- Pathology Unit, Morgagni-Pierantoni Hospital, 47121 Forlì, Italy
| | - Andrea Rocca
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy
| | - Michela Palleschi
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.C.); (R.M.)
| | - Paola Ulivi
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.C.); (R.M.)
| | - Giovanni Martinelli
- Department of Hematology and Sciences Oncology, Institute of Haematology “L. and A. Seràgnoli”, S. Orsola University Hospital, 40138 Bologna, Italy
| | - Roberta Maltoni
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.C.); (R.M.)
| |
Collapse
|
4
|
Peng J, Liu H, Liu Y, Liu J, Zhao Q, Liu W, Niu H, Xue H, Sun J, Wu J. HDAC6 mediates tumorigenesis during mitosis and the development of targeted deactivating agents. Bioorg Chem 2024; 153:107818. [PMID: 39288633 DOI: 10.1016/j.bioorg.2024.107818] [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: 07/09/2024] [Revised: 08/29/2024] [Accepted: 09/08/2024] [Indexed: 09/19/2024]
Abstract
Epigenetics, particularly deacetylation, plays a critical role in tumorigenesis as many carcinogens are under tight control by post-translational modification. HDAC6, an important and special histone deacetylase (HDAC) family member, has been indicated to increase carcinogenesis through various functions. Recent studies demonstrated the effects of HDAC6 inhibitors in mitotic arrest, however, detailed mechanisms still remain unknown. Herein, we review and summarize HDAC6-associated proteins that have been implicated in important roles in mitosis. We also discuss the development of medicinal agents targeting HDAC6.
Collapse
Affiliation(s)
- Jie Peng
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Hongyan Liu
- The People's Hospital of Zhaoyuan City, No. 168 Yingbin Road, Zhaoyuan 265400, Shandong Province, PR China
| | - Yujing Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Jingqian Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Qianlong Zhao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Wenjia Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Haoqian Niu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Haoyu Xue
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Jie Sun
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Jingde Wu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China.
| |
Collapse
|
5
|
Chesnokova V, Zonis S, Apaydin T, Barrett R, Melmed S. Non-pituitary growth hormone enables colon cell senescence evasion. Aging Cell 2024; 23:e14193. [PMID: 38724466 PMCID: PMC11320355 DOI: 10.1111/acel.14193] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 03/18/2024] [Accepted: 04/02/2024] [Indexed: 08/15/2024] Open
Abstract
DNA damage-induced senescence is initially sustained by p53. Senescent cells produce a senescence-associated secretory phenotype (SASP) that impacts the aging microenvironment, often promoting cell transformation. Employing normal non-tumorous human colon cells (hNCC) derived from surgical biopsies and three-dimensional human intestinal organoids, we show that local non-pituitary growth hormone (npGH) induced in senescent cells is a SASP component acting to suppress p53. npGH autocrine/paracrine suppression of p53 results in senescence evasion and cell-cycle reentry, as evidenced by increased Ki67 and BrdU incorporation. Post-senescent cells exhibit activated epithelial-to-mesenchymal transition (EMT), and increased cell motility. Nu/J mice harboring GH-secreting HCT116 xenografts with resultant high GH levels and injected intrasplenic with post-senescent hNCC developed fourfold more metastases than did mice harboring control xenografts, suggesting that paracrine npGH enables post-senescent cell transformation. By contrast, senescent cells with suppressed npGH exhibit downregulated Ki67 and decreased soft agar colony formation. Mechanisms underlying these observations include npGH induction by the SASP chemokine CXCL1, which attracts immune effectors to eliminate senescent cells; GH, in turn, suppresses CXCL1, likely by inhibiting phospho-NFκB, resulting in SASP cytokine downregulation. Consistent with these findings, GH-receptor knockout mice exhibited increased colon phospho-NFκB and CXCL1, while GH excess decreased colon CXCL1. The results elucidate mechanisms for local hormonal regulation of microenvironmental changes in DNA-damaged non-tumorous epithelial cells and portray a heretofore unappreciated GH action favoring age-associated epithelial cell transformation.
Collapse
Affiliation(s)
- Vera Chesnokova
- Department of MedicineCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Svetlana Zonis
- Department of MedicineCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Tugce Apaydin
- Department of MedicineCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Robert Barrett
- Board of Governors Regenerative Medicine InstituteCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Shlomo Melmed
- Department of MedicineCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| |
Collapse
|
6
|
Kulkarni P, Basu R, Bonn T, Low B, Mazurek N, Kopchick JJ. Growth Hormone Upregulates Melanoma Drug Resistance and Migration via Melanoma-Derived Exosomes. Cancers (Basel) 2024; 16:2636. [PMID: 39123364 PMCID: PMC11311539 DOI: 10.3390/cancers16152636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
Drug resistance in melanoma is a major hindrance in cancer therapy. Growth hormone (GH) plays a pivotal role in contributing to the resistance to chemotherapy. Knocking down or blocking the GH receptor has been shown to sensitize the tumor cells to chemotherapy. Extensive studies have demonstrated that exosomes, a subset of extracellular vesicles, play an important role in drug resistance by transferring key factors to sensitize cancer cells to chemotherapy. In this study, we explore how GH modulates exosomal cargoes from melanoma cells and their role in drug resistance. We treated the melanoma cells with GH, doxorubicin, and the GHR antagonist, pegvisomant, and analyzed the exosomes released. Additionally, we administered these exosomes to the recipient cells. The GH-treated melanoma cells released exosomes with elevated levels of ABC transporters (ABCC1 and ABCB1), N-cadherin, and MMP2, enhancing drug resistance and migration in the recipient cells. GHR antagonism reduced these exosomal levels, restoring drug sensitivity and attenuating migration. Overall, our findings highlight a novel role of GH in modulating exosomal cargoes that drive chemoresistance and metastasis in melanoma. This understanding provides insights into the mechanisms of GH in melanoma chemoresistance and suggests GHR antagonism as a potential therapy to overcome chemoresistance in melanoma treatment.
Collapse
Affiliation(s)
- Prateek Kulkarni
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; (P.K.); (R.B.); (T.B.); (B.L.); (N.M.)
- Molecular and Cellular Biology Program, Ohio University, Athens, OH 45701, USA
- Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Reetobrata Basu
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; (P.K.); (R.B.); (T.B.); (B.L.); (N.M.)
| | - Taylor Bonn
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; (P.K.); (R.B.); (T.B.); (B.L.); (N.M.)
- Department of Nutrition, Ohio University, Athens, OH 45701, USA
| | - Beckham Low
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; (P.K.); (R.B.); (T.B.); (B.L.); (N.M.)
- Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Nathaniel Mazurek
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; (P.K.); (R.B.); (T.B.); (B.L.); (N.M.)
- Environmental and Plant Biology, Ohio University, Athens, OH 45701, USA
| | - John J. Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; (P.K.); (R.B.); (T.B.); (B.L.); (N.M.)
- Molecular and Cellular Biology Program, Ohio University, Athens, OH 45701, USA
- Department of Biomedical Sciences, Ohio University, Athens, OH 45701, USA
| |
Collapse
|
7
|
Basu R, Kulkarni P, Swegan D, Duran-Ortiz S, Ahmad A, Caggiano LJ, Davis E, Walsh C, Brenya E, Koshal A, Brody R, Sandbhor U, Neggers SJCMM, Kopchick JJ. Growth Hormone Receptor Antagonist Markedly Improves Gemcitabine Response in a Mouse Xenograft Model of Human Pancreatic Cancer. Int J Mol Sci 2024; 25:7438. [PMID: 39000545 PMCID: PMC11242728 DOI: 10.3390/ijms25137438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/25/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
Chemotherapy treatment against pancreatic ductal adenocarcinoma (PDAC) is thwarted by tumoral activation of multiple therapy resistance pathways. The growth hormone (GH)-GH receptor (GHR) pair is a covert driver of multimodal therapy resistance in cancer and is overexpressed in PDAC tumors, yet the therapeutic potential of targeting the same has not been explored. Here, we report that GHR expression is a negative prognostic factor in patients with PDAC. Combinations of gemcitabine with different GHR antagonists (GHRAs) markedly improve therapeutic outcomes in nude mice xenografts. Employing cultured cells, mouse xenografts, and analyses of the human PDAC transcriptome, we identified that attenuation of the multidrug transporter and epithelial-to-mesenchymal transition programs in the tumors underlie the observed augmentation of chemotherapy efficacy by GHRAs. Moreover, in human PDAC patients, GHR expression strongly correlates with a gene signature of tumor promotion and immune evasion, which corroborate with that in syngeneic tumors in wild-type vs. GH transgenic mice. Overall, we found that GH action in PDAC promoted a therapy-refractory gene signature in vivo, which can be effectively attenuated by GHR antagonism. Our results collectively present a proof of concept toward considering GHR antagonists to improve chemotherapeutic outcomes in the highly chemoresistant PDAC.
Collapse
MESH Headings
- Animals
- Gemcitabine
- Humans
- Deoxycytidine/analogs & derivatives
- Deoxycytidine/pharmacology
- Deoxycytidine/therapeutic use
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/genetics
- Mice
- Xenograft Model Antitumor Assays
- Receptors, Somatotropin/metabolism
- Receptors, Somatotropin/antagonists & inhibitors
- Receptors, Somatotropin/genetics
- Carcinoma, Pancreatic Ductal/drug therapy
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Carcinoma, Pancreatic Ductal/genetics
- Cell Line, Tumor
- Mice, Nude
- Drug Resistance, Neoplasm/drug effects
- Gene Expression Regulation, Neoplastic/drug effects
- Female
Collapse
Affiliation(s)
- Reetobrata Basu
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; (R.B.); (P.K.); (D.S.); (S.D.-O.); (A.A.); (L.J.C.); (E.D.); (C.W.); (E.B.)
- Diabetes Institute, Ohio University, Athens, OH 45701, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Prateek Kulkarni
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; (R.B.); (P.K.); (D.S.); (S.D.-O.); (A.A.); (L.J.C.); (E.D.); (C.W.); (E.B.)
- Molecular and Cellular Biology Program, Ohio University, Athens, OH 45701, USA
- Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Deborah Swegan
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; (R.B.); (P.K.); (D.S.); (S.D.-O.); (A.A.); (L.J.C.); (E.D.); (C.W.); (E.B.)
- Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Silvana Duran-Ortiz
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; (R.B.); (P.K.); (D.S.); (S.D.-O.); (A.A.); (L.J.C.); (E.D.); (C.W.); (E.B.)
| | - Arshad Ahmad
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; (R.B.); (P.K.); (D.S.); (S.D.-O.); (A.A.); (L.J.C.); (E.D.); (C.W.); (E.B.)
- Translational Biomedical Sciences Program, Ohio University, Athens, OH 45701, USA
| | - Lydia J. Caggiano
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; (R.B.); (P.K.); (D.S.); (S.D.-O.); (A.A.); (L.J.C.); (E.D.); (C.W.); (E.B.)
- Honors Tutorial College, Ohio University, Athens, OH 45701, USA
| | - Emily Davis
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; (R.B.); (P.K.); (D.S.); (S.D.-O.); (A.A.); (L.J.C.); (E.D.); (C.W.); (E.B.)
- Molecular and Cellular Biology Program, Ohio University, Athens, OH 45701, USA
- Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Christopher Walsh
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; (R.B.); (P.K.); (D.S.); (S.D.-O.); (A.A.); (L.J.C.); (E.D.); (C.W.); (E.B.)
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- Translational Biomedical Sciences Program, Ohio University, Athens, OH 45701, USA
| | - Edward Brenya
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; (R.B.); (P.K.); (D.S.); (S.D.-O.); (A.A.); (L.J.C.); (E.D.); (C.W.); (E.B.)
- Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Adeel Koshal
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA;
| | - Rich Brody
- InfinixBio LLC, Columbus, OH 43212, USA; (R.B.); (U.S.)
| | - Uday Sandbhor
- InfinixBio LLC, Columbus, OH 43212, USA; (R.B.); (U.S.)
| | | | - John J. Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; (R.B.); (P.K.); (D.S.); (S.D.-O.); (A.A.); (L.J.C.); (E.D.); (C.W.); (E.B.)
- Diabetes Institute, Ohio University, Athens, OH 45701, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- Molecular and Cellular Biology Program, Ohio University, Athens, OH 45701, USA
- Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
- Translational Biomedical Sciences Program, Ohio University, Athens, OH 45701, USA
| |
Collapse
|
8
|
Stark MS, Sturm RA, Pan Y, Smit DJ, Kommajosyula V, Lee KJ, Jagirdar K, McLean C, Duffy DL, Soyer HP, Mar VJ. Assessing the genetic risk of nodular melanoma using a candidate gene approach. Br J Dermatol 2024; 190:199-206. [PMID: 37766469 DOI: 10.1093/bjd/ljad365] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/28/2023] [Accepted: 09/21/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND Nodular melanoma (NM) is a challenge to diagnose early due to its rapid growth and more atypical clinical presentation, making it the largest contributor to melanoma mortality. OBJECTIVES Our study aim was to perform a rare-variant allele (RVA) analysis of whole-exome sequencing of patients with NM and non-NM (minor allele frequency ≤ 1% non-Finnish European) for a set of 500 candidate genes potentially implicated in melanoma. METHODS This study recruited 131 participants with NM and 194 with non-NM from South-east Queensland and patients with NM from Victoria to perform a comparative analysis of possible genetic differences or similarities between the two melanoma cohorts. RESULTS Phenotypic analysis revealed that a majority of patients diagnosed with NM were older males with a higher frequency of fair skin and red hair than is seen in the general population. The distribution of common melanoma polygenic risk scores was similar in patients with NM and non-NM, with over 28% in the highest quantile of scores. There was also a similar frequency of carriage of familial/high-penetrant melanoma gene and loss-of-function variants. We identified 39 genes by filtering 500 candidate genes based on the greatest frequency in NM compared with non-NM cases. The genes with RVAs of greatest frequency in NM included PTCH1, ARID2 and GHR. Rare variants in the SMO gene, which interacts with PTCH1 as ligand and receptor, were also identified, providing evidence that the Hedgehog pathway may contribute to NM risk. There was a cumulative effect in carrying multiple rare variants in the NM-associated genes. A 14.8-fold increased ratio for NM compared with non-NM was seen when two RVAs of the 39 genes were carried by a patient. CONCLUSIONS This study highlights the importance of considering frequency of RVA to identify those at risk of NM in addition to known high penetrance genes.
Collapse
Affiliation(s)
- Mitchell S Stark
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Brisbane, Qld, Australia
| | - Richard A Sturm
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Brisbane, Qld, Australia
| | - Yan Pan
- Victorian Melanoma Service, The Alfred Hospital, Melbourne, Vic, Australia
- Central Clinical School, Faculty of Medicine, Nursing and Health Sciences
| | - Darren J Smit
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Brisbane, Qld, Australia
| | - Varsha Kommajosyula
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Brisbane, Qld, Australia
| | - Katie J Lee
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Brisbane, Qld, Australia
| | - Kasturee Jagirdar
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Brisbane, Qld, Australia
| | - Catriona McLean
- Victorian Melanoma Service, The Alfred Hospital, Melbourne, Vic, Australia
- Central Clinical School, Faculty of Medicine, Nursing and Health Sciences
| | - David L Duffy
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Brisbane, Qld, Australia
- QIMR Berghofer Medical Research Institute, Brisbane, Qld, Australia
| | - H Peter Soyer
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Brisbane, Qld, Australia
- Dermatology Department, Princess Alexandra Hospital, Brisbane, Qld, Australia
| | - Victoria J Mar
- Victorian Melanoma Service, The Alfred Hospital, Melbourne, Vic, Australia
- School of Public Health and Preventive Medicine; Monash University, Melbourne, Vic, Australia
| |
Collapse
|
9
|
Poonia P, Sharma M, Jha P, Chopra M. Pharmacophore-based virtual screening of ZINC database, molecular modeling and designing new derivatives as potential HDAC6 inhibitors. Mol Divers 2023; 27:2053-2071. [PMID: 36214962 DOI: 10.1007/s11030-022-10540-3] [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: 09/10/2022] [Accepted: 09/30/2022] [Indexed: 11/25/2022]
Abstract
To date, many HDAC6 inhibitors have been identified and developed but none is clinically approved as of now. Through this study, we aim to obtain novel HDAC6 selective inhibitors and provide new insights into the detailed structural design of potential HDAC6 inhibitors. A HypoGen-based 3D QSAR HDAC6 pharmacophore was built and used as a query model to screen approximately 8 million ZINC database compounds. First, the ZINC Database was filtered using ADMET, followed by pharmacophore-based library screening. Using fit value and estimated activity cutoffs, a final set of 54 ZINC hits was obtained that were further investigated using molecular docking with the crystal structure of human histone deacetylase 6 catalytic domain 2 in complex with Trichostatin A (PDB ID: 5EDU). Through detailed in silico screening of the ZINC database, we shortlisted three hits as the lead molecules for designing novel HDAC6 inhibitors with better efficacy. Docking with 5EDU, followed by ADMET and TOPKAT analysis of modified ZINC hits provided 9 novel potential HDAC6 inhibitors that possess better docking scores and 2D interactions as compared to the control ZINC hit molecules. Finally, a 50 ns MD analysis run followed by Protein-Ligand Interaction Energy (PLIE) analysis of the top scored hits provided a novel molecule N1 that showed promisingly similar results to that of Ricolinostat (a known HDAC6 inhibitor). The comparable result of the designed hits to established HDAC6 inhibitors suggests that these compounds might prove to be successful HDAC6 inhibitors in future. Designed novel hits that might act as good HDAC6 inhibitors derived from ZINC database using combined molecular docking and modeling approaches.
Collapse
Affiliation(s)
- Priya Poonia
- Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110036, India
| | - Monika Sharma
- Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110036, India
| | - Prakash Jha
- Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110036, India
| | - Madhu Chopra
- Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110036, India.
| |
Collapse
|
10
|
Peng J, Xie F, Qin P, Liu Y, Niu H, Sun J, Xue H, Zhao Q, Liu J, Wu J. Recent development of selective inhibitors targeting the HDAC6 as anti-cancer drugs: Structure, function and design. Bioorg Chem 2023; 138:106622. [PMID: 37244230 DOI: 10.1016/j.bioorg.2023.106622] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/09/2023] [Accepted: 05/19/2023] [Indexed: 05/29/2023]
Abstract
HDAC6, a member of the histone deacetylase family, mainly is a cytosolic protein and regulates cell growth by acting on non-histone substrates, such as α -tubulin, cortactin, heat shock protein HSP90, programmed death 1 (PD-1) and programmed death ligand 1 (PD-L1), that are closely related to the proliferation, invasion, immune escape and angiogenesis of cancer tissues. The approved drugs targeting the HDACs are all pan-inhibitors and have many side effects due to their lack of selectivity. Therefore, development of selective inhibitors of HDAC6 has attracted much attention in the field of cancer therapy. In this review, we will summarize the relationship between HDAC6 and cancer, and discuss the design strategies of HDAC6 inhibitors for cancer treatment in recent years.
Collapse
Affiliation(s)
- Jie Peng
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Fei Xie
- Department of Pharmacy, Qilu Hospital of Shandong University, Jinan 250012, PR China
| | - Pengxia Qin
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Yujing Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Haoqian Niu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Jie Sun
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Haoyu Xue
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Qianlong Zhao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Jingqian Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Jingde Wu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China.
| |
Collapse
|
11
|
Basu R, Brody R, Sandbhor U, Kulkarni P, Davis E, Swegan D, Caggiano LJ, Brenya E, Neggers S, Kopchick JJ. Structure and function of a dual antagonist of the human growth hormone and prolactin receptors with site-specific PEG conjugates. J Biol Chem 2023; 299:105030. [PMID: 37442239 PMCID: PMC10410519 DOI: 10.1016/j.jbc.2023.105030] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 06/06/2023] [Accepted: 06/08/2023] [Indexed: 07/15/2023] Open
Abstract
Human growth hormone (hGH) is a pituitary-derived endocrine protein that regulates several critical postnatal physiologic processes including growth, organ development, and metabolism. Following adulthood, GH is also a regulator of multiple pathologies like fibrosis, cancer, and diabetes. Therefore, there is a significant pharmaceutical interest in developing antagonists of hGH action. Currently, there is a single FDA-approved antagonist of the hGH receptor (hGHR) prescribed for treating patients with acromegaly and discovered in our laboratory almost 3 decades ago. Here, we present the first data on the structure and function of a new set of protein antagonists with the full range of hGH actions-dual antagonists of hGH binding to the GHR as well as that of hGH binding to the prolactin receptor. We describe the site-specific PEG conjugation, purification, and subsequent characterization using MALDI-TOF, size-exclusion chromatography, thermostability, and biochemical activity in terms of ELISA-based binding affinities with GHR and prolactin receptor. Moreover, these novel hGHR antagonists display distinct antagonism of GH-induced GHR intracellular signaling in vitro and marked reduction in hepatic insulin-like growth factor 1 output in vivo. Lastly, we observed potent anticancer biological efficacies of these novel hGHR antagonists against human cancer cell lines. In conclusion, we propose that these new GHR antagonists have potential for development towards multiple clinical applications related to GH-associated pathologies.
Collapse
Affiliation(s)
- Reetobrata Basu
- Edison Biotechnology Institute, Ohio University, Athens, Ohio, USA
| | | | | | - Prateek Kulkarni
- Edison Biotechnology Institute, Ohio University, Athens, Ohio, USA; Molecular and Cellular Biology Program, Ohio University, Athens, Ohio, USA; Department of Biological Sciences, Ohio University, Athens, Ohio, USA
| | - Emily Davis
- Edison Biotechnology Institute, Ohio University, Athens, Ohio, USA; Molecular and Cellular Biology Program, Ohio University, Athens, Ohio, USA; Department of Biological Sciences, Ohio University, Athens, Ohio, USA
| | - Deborah Swegan
- Edison Biotechnology Institute, Ohio University, Athens, Ohio, USA; Department of Biological Sciences, Ohio University, Athens, Ohio, USA
| | - Lydia J Caggiano
- Edison Biotechnology Institute, Ohio University, Athens, Ohio, USA; Honors Tutorial College, Ohio University, Athens, Ohio, USA
| | - Edward Brenya
- Edison Biotechnology Institute, Ohio University, Athens, Ohio, USA; Department of Biological Sciences, Ohio University, Athens, Ohio, USA
| | - Sebastian Neggers
- Department of Medicine, Endocrinology, Erasmus Medical Centre, Rotterdam, Netherlands
| | - John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, Ohio, USA; Molecular and Cellular Biology Program, Ohio University, Athens, Ohio, USA; Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA.
| |
Collapse
|
12
|
Shaker BT, Ismail AA, Salih R, Hadj Kacem H, Rahmani M, Struman I, Bajou K. The 14-Kilodalton Human Growth Hormone Fragment a Potent Inhibitor of Angiogenesis and Tumor Metastasis. Int J Mol Sci 2023; 24:ijms24108877. [PMID: 37240223 DOI: 10.3390/ijms24108877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/02/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
The 14-kilodalton human growth hormone (14 kDa hGH) N-terminal fragment derived from the proteolytic cleavage of its full-length counterpart has been shown to sustain antiangiogenic potentials. This study investigated the antitumoral and antimetastatic effects of 14 kDa hGH on B16-F10 murine melanoma cells. B16-F10 murine melanoma cells transfected with 14 kDa hGH expression vectors showed a significant reduction in cellular proliferation and migration associated with an increase in cell apoptosis in vitro. In vivo, 14 kDa hGH mitigated tumor growth and metastasis of B16-F10 cells and was associated with a significant reduction in tumor angiogenesis. Similarly, 14 kDa hGH expression reduced human brain microvascular endothelial (HBME) cell proliferation, migration, and tube formation abilities and triggered apoptosis in vitro. The antiangiogenic effects of 14 kDa hGH on HBME cells were abolished when we stably downregulated plasminogen activator inhibitor-1 (PAI-1) expression in vitro. In this study, we showed the potential anticancer role of 14 kDa hGH, its ability to inhibit primary tumor growth and metastasis establishment, and the possible involvement of PAI-1 in promoting its antiangiogenic effects. Therefore, these results suggest that the 14 kDa hGH fragment can be used as a therapeutic molecule to inhibit angiogenesis and cancer progression.
Collapse
Affiliation(s)
- Baraah Tariq Shaker
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Human Genetics & Stem Cells Research Group, Research Institute of Sciences & Engineering, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Asmaa Anwar Ismail
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Human Genetics & Stem Cells Research Group, Research Institute of Sciences & Engineering, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Rawan Salih
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Human Genetics & Stem Cells Research Group, Research Institute of Sciences & Engineering, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Hassen Hadj Kacem
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Human Genetics & Stem Cells Research Group, Research Institute of Sciences & Engineering, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Mohamed Rahmani
- Department of Molecular Biology and Genetics, College of Medicine & Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
- Center for Biotechnology, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Ingrid Struman
- Laboratory of Molecular Angiogenesis, GIGA Research Center, University of Liège, 4000 Liège, Belgium
| | - Khalid Bajou
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Human Genetics & Stem Cells Research Group, Research Institute of Sciences & Engineering, University of Sharjah, Sharjah 27272, United Arab Emirates
| |
Collapse
|
13
|
Kopchick JJ, Basu R, Berryman DE, Jorgensen JOL, Johannsson G, Puri V. Covert actions of growth hormone: fibrosis, cardiovascular diseases and cancer. Nat Rev Endocrinol 2022; 18:558-573. [PMID: 35750929 PMCID: PMC9703363 DOI: 10.1038/s41574-022-00702-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/19/2022] [Indexed: 12/20/2022]
Abstract
Since its discovery nearly a century ago, over 100,000 studies of growth hormone (GH) have investigated its structure, how it interacts with the GH receptor and its multiple actions. These include effects on growth, substrate metabolism, body composition, bone mineral density, the cardiovascular system and brain function, among many others. Recombinant human GH is approved for use to promote growth in children with GH deficiency (GHD), along with several additional clinical indications. Studies of humans and animals with altered levels of GH, from complete or partial GHD to GH excess, have revealed several covert or hidden actions of GH, such as effects on fibrosis, cardiovascular function and cancer. In this Review, we do not concentrate on the classic and controversial indications for GH therapy, nor do we cover all covert actions of GH. Instead, we stress the importance of the relationship between GH and fibrosis, and how fibrosis (or lack thereof) might be an emerging factor in both cardiovascular and cancer pathologies. We highlight clinical data from patients with acromegaly or GHD, alongside data from cellular and animal studies, to reveal novel phenotypes and molecular pathways responsible for these actions of GH in fibrosis, cardiovascular function and cancer.
Collapse
Affiliation(s)
- John J Kopchick
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA.
- The Diabetes Institute, Ohio University, Athens, OH, USA.
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA.
| | - Reetobrata Basu
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
- The Diabetes Institute, Ohio University, Athens, OH, USA
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
| | - Darlene E Berryman
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
- The Diabetes Institute, Ohio University, Athens, OH, USA
| | - Jens O L Jorgensen
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Gudmundur Johannsson
- Department of Endocrinology, Sahlgrenska University Hospital, Sahlgrenska Academy, University of Göteborg, Gothenburg, Sweden
| | - Vishwajeet Puri
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
- The Diabetes Institute, Ohio University, Athens, OH, USA
| |
Collapse
|
14
|
Verreault M, Segoviano Vilchis I, Rosenberg S, Lemaire N, Schmitt C, Guehennec J, Royer-Perron L, Thomas JL, Lam TT, Dingli F, Loew D, Ducray F, Paris S, Carpentier C, Marie Y, Laigle-Donadey F, Rousseau A, Pigat N, Boutillon F, Bielle F, Mokhtari K, Frank SJ, de Reyniès A, Hoang-Xuan K, Sanson M, Goffin V, Idbaih A. Identification of growth hormone receptor as a relevant target for precision medicine in low-EGFR expressing glioblastoma. Clin Transl Med 2022; 12:e939. [PMID: 35808822 PMCID: PMC9270581 DOI: 10.1002/ctm2.939] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 05/30/2022] [Accepted: 06/05/2022] [Indexed: 11/11/2022] Open
Abstract
Objective New therapeutic approaches are needed to improve the prognosis of glioblastoma (GBM) patients. Methods With the objective of identifying alternative oncogenic mechanisms to abnormally activated epidermal growth factor receptor (EGFR) signalling, one of the most common oncogenic mechanisms in GBM, we performed a comparative analysis of gene expression profiles in a series of 54 human GBM samples. We then conducted gain of function as well as genetic and pharmocological inhibition assays in GBM patient‐derived cell lines to functionnally validate our finding. Results We identified that growth hormone receptor (GHR) signalling defines a distinct molecular subset of GBMs devoid of EGFR overexpression. GHR overexpression was detected in one third of patients and was associated with low levels of suppressor of cytokine signalling 2 (SOCS2) expression due to SOCS2 promoter hypermethylation. In GBM patient‐derived cell lines, GHR signalling modulates the expression of proteins involved in cellular movement, promotes cell migration, invasion and proliferation in vitro and promotes tumourigenesis, tumour growth, and tumour invasion in vivo. GHR genetic and pharmacological inhibition reduced cell proliferation and migration in vitro. Conclusion This study pioneers a new field of investigation to improve the prognosis of GBM patients.
Collapse
Affiliation(s)
- Maïté Verreault
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Irma Segoviano Vilchis
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Shai Rosenberg
- Laboratory for Cancer Computational Biology & Gaffin Center for Neuro-Oncology, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Nolwenn Lemaire
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Charlotte Schmitt
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Jérémy Guehennec
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Louis Royer-Perron
- DMU Neurosciences, Service de Neurologie 2-Mazarin, Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Jean-Léon Thomas
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France.,Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - TuKiet T Lam
- Mass Spectrometry & Proteomics Resource, Keck Biotechnology Resource Laboratory, New Haven, Connecticut, USA.,Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut, USA
| | - Florent Dingli
- Institut Curie, Centre de Recherche, PSL Research University, Laboratoire de Spectrométrie de Masse Protéomique, Paris, France
| | - Damarys Loew
- Institut Curie, Centre de Recherche, PSL Research University, Laboratoire de Spectrométrie de Masse Protéomique, Paris, France
| | | | - Sophie Paris
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Catherine Carpentier
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Yannick Marie
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Florence Laigle-Donadey
- DMU Neurosciences, Service de Neurologie 2-Mazarin, Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Audrey Rousseau
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France.,DMU Neurosciences, Service de Neurologie 2-Mazarin, Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Natascha Pigat
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Paris, France
| | - Florence Boutillon
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Paris, France
| | - Franck Bielle
- DMU Neurosciences, Service de Neurologie 2-Mazarin, Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Karima Mokhtari
- DMU Neurosciences, Service de Neurologie 2-Mazarin, Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Stuart J Frank
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Alabama, Birmingham, Alabama, USA.,Endocrinology Section, Medical Service, Birmingham VA Medical Center, Birmingham, Alabama, USA
| | - Aurélien de Reyniès
- Programme Cartes d'Identité des Tumeurs (CIT), Ligue Nationale Contre le Cancer, Service de Bioinformatique, Paris, France
| | - Khê Hoang-Xuan
- DMU Neurosciences, Service de Neurologie 2-Mazarin, Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Marc Sanson
- DMU Neurosciences, Service de Neurologie 2-Mazarin, Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Vincent Goffin
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Paris, France
| | - Ahmed Idbaih
- DMU Neurosciences, Service de Neurologie 2-Mazarin, Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| |
Collapse
|
15
|
van der Velden LM, Maas P, van Amersfoort M, Timmermans-Sprang EPM, Mensinga A, van der Vaart E, Malergue F, Viëtor H, Derksen PWB, Klumperman J, van Agthoven A, Egan DA, Mol JA, Strous GJ. Small molecules to regulate the GH/IGF1 axis by inhibiting the growth hormone receptor synthesis. Front Endocrinol (Lausanne) 2022; 13:926210. [PMID: 35966052 PMCID: PMC9365994 DOI: 10.3389/fendo.2022.926210] [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] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
Growth hormone (GH) and insulin-like growth factor-1 (IGF1) play an important role in mammalian development, cell proliferation and lifespan. Especially in cases of tumor growth there is an urgent need to control the GH/IGF1 axis. In this study we screened a 38,480-compound library, and in two consecutive rounds of analogues selection, we identified active lead compounds based on the following criteria: inhibition the GH receptor (GHR) activity and its downstream effectors Jak2 and STAT5, and inhibition of growth of breast and colon cancer cells. The most active small molecule (BM001) inhibited both the GH/IGF1 axis and cell proliferation with an IC50 of 10-30 nM of human cancer cells. BM001 depleted GHR in human lymphoblasts. In preclinical xenografted experiments, BM001 showed a strong decrease in tumor volume in mice transplanted with MDA-MB-231 breast cancer cells. Mechanistically, the drug acts on the synthesis of the GHR. Our findings open the possibility to inhibit the GH/IGF1 axis with a small molecule.
Collapse
Affiliation(s)
- Lieke M. van der Velden
- Department of Cell Biology, Centre for Molecular Medicine, University Medical Center (UMC) Utrecht, Utrecht, Netherlands
| | - Peter Maas
- Specs Compound Handling, Zoetermeer, Netherlands
- *Correspondence: Ger J. Strous, ; Jan A. Mol, ; Peter Maas,
| | | | | | - Anneloes Mensinga
- Department of Cell Biology, Centre for Molecular Medicine, University Medical Center (UMC) Utrecht, Utrecht, Netherlands
| | - Elisabeth van der Vaart
- Department of Cell Biology, Centre for Molecular Medicine, University Medical Center (UMC) Utrecht, Utrecht, Netherlands
| | - Fabrice Malergue
- Department of Research and Development, Beckman Coulter Life Science, Immunotech Marseille, Marseille, France
| | - Henk Viëtor
- Drug Discovery Factory (DDF) Ventures, Breukelen, Netherlands
| | - Patrick W B. Derksen
- Department of Pathology, University Medical Center (UMC) Utrecht, Utrecht, Netherlands
| | - Judith Klumperman
- Department of Cell Biology, Centre for Molecular Medicine, University Medical Center (UMC) Utrecht, Utrecht, Netherlands
| | - Andreas van Agthoven
- Department of Research and Development, Beckman Coulter Life Science, Immunotech Marseille, Marseille, France
| | - David A. Egan
- Cell Screening Core, Department of Cell Biology, Center for Molecular Medicine, University Medical Center, Utrecht, Netherlands
| | - Jan A. Mol
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
- *Correspondence: Ger J. Strous, ; Jan A. Mol, ; Peter Maas,
| | - Ger J. Strous
- Department of Cell Biology, Centre for Molecular Medicine, University Medical Center (UMC) Utrecht, Utrecht, Netherlands
- *Correspondence: Ger J. Strous, ; Jan A. Mol, ; Peter Maas,
| |
Collapse
|
16
|
HDAC6 inhibitor WT161 performs anti-tumor effect on osteosarcoma and synergistically interacts with 5-FU. Biosci Rep 2021; 41:228382. [PMID: 33860796 PMCID: PMC8150159 DOI: 10.1042/bsr20203905] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 04/02/2021] [Accepted: 04/13/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND WT161, as a selective HDAC6 inhibitor, has been shown to play anti-tumor effects on several kinds of cancers. The aim of the present study is to explore the roles of WT161 in osteosarcoma and its underlying mechanisms. METHODS The anti-proliferative effect of WT161 on osteosarcoma cells was examined using MTT assay and colony formation assay. Cell apoptosis was analyzed using flow cytometer. The synergistic effect was evaluated by isobologram analysis using CompuSyn software. The osteosarcoma xenograft models were established to evaluate the anti-proliferative effect of WT161 in vivo. RESULTS WT161 suppressed the cell growth and induced apoptosis of osteosarcoma cells in a dose- and time-dependent manner. Mechanistically, we found that WT161 treatment obviously increased the protein level of PTEN and decreased the phosphorylation level of protein kinase-B (AKT). More importantly, WT161 showed synergistic inhibition with 5-FU on osteosarcoma cells in vitro and in vivo. CONCLUSIONS These results indicate that WT161 inhibits the growth of osteosarcoma through PTEN and has a synergistic efficiency with 5-FU.
Collapse
|
17
|
Lin CC, Liu TW, Yeh ML, Tsai YS, Tsai PC, Huang CF, Huang JF, Chuang WL, Dai CY, Yu ML. Significant down-regulation of growth hormone receptor expression revealed as a new unfavorable prognostic factor in hepatitis C virus-related hepatocellular carcinoma. Clin Mol Hepatol 2021; 27:313-328. [PMID: 33317258 PMCID: PMC8046631 DOI: 10.3350/cmh.2020.0247] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/19/2020] [Accepted: 12/10/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND/AIMS Growth hormone (GH) is the main regulator of somatic growth, metabolism, and gender dimorphism in the liver. GH receptor (GHR) signaling in cancer is derived from a large body of evidence, although the GHR signaling pathway involved in the prognosis of hepatocellular carcinoma (HCC) in patients with hepatitis C virus (HCV)-related HCC, remains unclear. We aimed to explore the expression of GHR and analyze its association with clinicopathologic features and prognosis of patients with chronic hepatitis C and HCC. METHODS The expression of GHR mRNA was investigated by quantitative real-time polymerase chain reaction in paired tumors and adjacent non-tumorous (ANT) liver tissues of 200 patients with chronic hepatitis C and HCC. Western blotting and immunofluorescence assays using the HCV-infected Huh7.5.1 cell model was performed. RESULTS GHR mRNA was significantly lower in HCV-HCC tissues than in corresponding ANT liver tissues. GHR mRNA and protein levels also decreased in the HCV-infected Huh7.5.1 cell model. Notably, lower GHR expression was associated with age of >60 years (P=0.0111) and worse clinicopathologic characteristics, including alpha-fetoprotein >100 ng/mL (P=0.0403), cirrhosis (P=0.0075), vascular invasion (P=0.0052), pathological stage II-IV (P=0.0002), and albumin ≤4.0 g/dL (P=0.0055), which were linked with poor prognosis of HCC. Most importantly, the high incidence of recurrence and poor survival rates in patients with a low ratio of tumor/ANT GHR (≤0.1) were observed, indicating that low expression levels of GHR had great risk for development of HCC in patients with chronic hepatitis C. CONCLUSION Our study demonstrates a significant down-regulation of GHR expression as a new unfavorable independent prognostic factor in patients with chronic hepatitis C and HCC.
Collapse
Affiliation(s)
- Ching-Chih Lin
- Division of Hepatobiliary, Department of Internal Medicine, Hepatitis Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Center for Liquid Biopsy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ta-Wei Liu
- Division of Hepatobiliary, Department of Internal Medicine, Hepatitis Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Center for Liquid Biopsy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ming-Lun Yeh
- Division of Hepatobiliary, Department of Internal Medicine, Hepatitis Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- School of Medicine and Hepatitis Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center for Cancer Research and Center for Liquid Biopsy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Shan Tsai
- Division of Hepatobiliary, Department of Internal Medicine, Hepatitis Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Pei-Chien Tsai
- Division of Hepatobiliary, Department of Internal Medicine, Hepatitis Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Chung-Feng Huang
- Division of Hepatobiliary, Department of Internal Medicine, Hepatitis Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- School of Medicine and Hepatitis Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center for Cancer Research and Center for Liquid Biopsy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jee-Fu Huang
- Division of Hepatobiliary, Department of Internal Medicine, Hepatitis Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- School of Medicine and Hepatitis Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center for Cancer Research and Center for Liquid Biopsy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wan-Long Chuang
- Division of Hepatobiliary, Department of Internal Medicine, Hepatitis Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- School of Medicine and Hepatitis Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center for Cancer Research and Center for Liquid Biopsy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chia-Yen Dai
- Division of Hepatobiliary, Department of Internal Medicine, Hepatitis Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- School of Medicine and Hepatitis Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center for Cancer Research and Center for Liquid Biopsy, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan
| | - Ming-Lung Yu
- Division of Hepatobiliary, Department of Internal Medicine, Hepatitis Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- School of Medicine and Hepatitis Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center for Cancer Research and Center for Liquid Biopsy, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan
| |
Collapse
|
18
|
A novel peptide antagonist of the human growth hormone receptor. J Biol Chem 2021; 296:100588. [PMID: 33774052 PMCID: PMC8086144 DOI: 10.1016/j.jbc.2021.100588] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 03/09/2021] [Accepted: 03/23/2021] [Indexed: 12/11/2022] Open
Abstract
Excess circulating human growth hormone (hGH) in vivo is linked to metabolic and growth disorders such as cancer, diabetes, and acromegaly. Consequently, there is considerable interest in developing antagonists of hGH action. Here, we present the design, synthesis, and characterization of a 16-residue peptide (site 1-binding helix [S1H]) that inhibits hGH-mediated STAT5 phosphorylation in cultured cells. S1H was designed as a direct sequence mimetic of the site 1 mini-helix (residues 36-51) of wild-type hGH and acts by inhibiting the interaction of hGH with the human growth hormone receptor (hGHR). In vitro studies indicated that S1H is stable in human serum and can adopt an α-helix in solution. Our results also show that S1H mitigates phosphorylation of STAT5 in cells co-treated with hGH, reducing intracellular STAT5 phosphorylation levels to those observed in untreated controls. Furthermore, S1H was found to attenuate the activity of the hGHR and the human prolactin receptor, suggesting that this peptide acts as an antagonist of both lactogenic and somatotrophic hGH actions. Finally, we used alanine scanning to determine how discrete amino acids within the S1H sequence contribute to its structural organization and biological activity. We observed a strong correlation between helical propensity and inhibitory effect, indicating that S1H-mediated antagonism of the hGHR is largely dependent on the ability for S1H to adopt an α-helix. Taken together, these results show that S1H not only acts as a novel peptide-based antagonist of the hGHR but can also be applied as a chemical tool to study the molecular nature of hGH-hGHR interactions.
Collapse
|
19
|
Growth Hormone Upregulates Mediators of Melanoma Drug Efflux and Epithelial-to-Mesenchymal Transition In Vitro and In Vivo. Cancers (Basel) 2020; 12:cancers12123640. [PMID: 33291663 PMCID: PMC7761932 DOI: 10.3390/cancers12123640] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/24/2020] [Accepted: 12/02/2020] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Growth hormone (GH) action is strongly implicated in the progression and therapy resistance in several types of solid tumors which overexpress the GH receptor (GHR). The aim of our study was to characterize the effects of GH and its downstream effector insulin-like growth factor 1 (IGF-1) on melanoma using in vitro and in vivo models. We confirmed an IGF-1-independent role of elevated circulating GH in upregulating key mechanisms of therapy resistance and malignancy with analyses conducted at the molecular and cellular level. We identified that GH upregulates key mechanisms of therapy resistance and metastases in melanoma tumors in an IGF-1 dependent and independent manner by upregulating multidrug efflux pumps and EMT transcription factors. Our study reveals that GH action renders an intrinsic drug resistance phenotype to the melanoma tumors—a clinically crucial property of GH verifiable in other human cancers with GHR expression. Abstract Growth hormone (GH) and the GH receptor (GHR) are expressed in a wide range of malignant tumors including melanoma. However, the effect of GH/insulin-like growth factor (IGF) on melanoma in vivo has not yet been elucidated. Here we assessed the physical and molecular effects of GH on mouse melanoma B16-F10 and human melanoma SK-MEL-30 cells in vitro. We then corroborated these observations with syngeneic B16-F10 tumors in two mouse lines with different levels of GH/IGF: bovine GH transgenic mice (bGH; high GH, high IGF-1) and GHR gene-disrupted or knockout mice (GHRKO; high GH, low IGF-1). In vitro, GH treatment enhanced mouse and human melanoma cell growth, drug retention and cell invasion. While the in vivo tumor size was unaffected in both bGH and GHRKO mouse lines, multiple drug-efflux pumps were up regulated. This intrinsic capacity of therapy resistance appears to be GH dependent. Additionally, epithelial-to-mesenchymal transition (EMT) gene transcription markers were significantly upregulated in vivo supporting our current and recent in vitro observations. These syngeneic mouse melanoma models of differential GH/IGF action can be valuable tools in screening for therapeutic options where lowering GH/IGF-1 action is important.
Collapse
|
20
|
Strous GJ, Almeida ADS, Putters J, Schantl J, Sedek M, Slotman JA, Nespital T, Hassink GC, Mol JA. Growth Hormone Receptor Regulation in Cancer and Chronic Diseases. Front Endocrinol (Lausanne) 2020; 11:597573. [PMID: 33312162 PMCID: PMC7708378 DOI: 10.3389/fendo.2020.597573] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/14/2020] [Indexed: 12/14/2022] Open
Abstract
The GHR signaling pathway plays important roles in growth, metabolism, cell cycle control, immunity, homeostatic processes, and chemoresistance via both the JAK/STAT and the SRC pathways. Dysregulation of GHR signaling is associated with various diseases and chronic conditions such as acromegaly, cancer, aging, metabolic disease, fibroses, inflammation and autoimmunity. Numerous studies entailing the GHR signaling pathway have been conducted for various cancers. Diverse factors mediate the up- or down-regulation of GHR signaling through post-translational modifications. Of the numerous modifications, ubiquitination and deubiquitination are prominent events. Ubiquitination by E3 ligase attaches ubiquitins to target proteins and induces proteasomal degradation or starts the sequence of events that leads to endocytosis and lysosomal degradation. In this review, we discuss the role of first line effectors that act directly on the GHR at the cell surface including ADAM17, JAK2, SRC family member Lyn, Ubc13/CHIP, proteasome, βTrCP, CK2, STAT5b, and SOCS2. Activity of all, except JAK2, Lyn and STAT5b, counteract GHR signaling. Loss of their function increases the GH-induced signaling in favor of aging and certain chronic diseases, exemplified by increased lung cancer risk in case of a mutation in the SOCS2-GHR interaction site. Insight in their roles in GHR signaling can be applied for cancer and other therapeutic strategies.
Collapse
Affiliation(s)
- Ger J. Strous
- Department of Cell Biology, Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, Netherlands
- BIMINI Biotech B.V., Leiden, Netherlands
| | - Ana Da Silva Almeida
- Department of Cell Biology, Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Joyce Putters
- Department of Cell Biology, Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Julia Schantl
- Department of Cell Biology, Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Magdalena Sedek
- Department of Cell Biology, Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Johan A. Slotman
- Department of Cell Biology, Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Tobias Nespital
- Department of Cell Biology, Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Gerco C. Hassink
- Department of Cell Biology, Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Jan A. Mol
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
21
|
Iwase H, Ball S, Adams K, Eyestone W, Walters A, Cooper DKC. Growth hormone receptor knockout: Relevance to xenotransplantation. Xenotransplantation 2020; 28:e12652. [PMID: 33058285 DOI: 10.1111/xen.12652] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/03/2020] [Accepted: 09/15/2020] [Indexed: 02/06/2023]
Abstract
Xenotransplantation research has made considerable progress in recent years, largely through the increasing availability of pigs with multiple genetic modifications, effective immunosuppressive therapy, and anti-inflammatory therapy to protect pig tissues from the primate immune and inflammatory responses and correct molecular incompatibilities. Further study is required regarding identification and investigation of physiological incompatibilities. Although the exact cause remains uncertain, we and others have observed relatively rapid growth of kidney xenografts after transplantation into nonhuman primates (NHPs). There has also been some evidence of growth, or at least ventricular hypertrophy, of the pig heart after orthotopic transplantation into NHPs. Rapid growth could be problematic, particularly with regard to the heart within the relatively restricted confines of the chest. It has been suggested that the problem of rapid growth of the pig organ after transplantation could be resolved by growth hormone receptor (GHR) gene knockout in the pig. The GHR, although most well-known for regulating growth, has many other biological functions, including regulating metabolism and controlling physiological processes. Genetically modified GHRKO pigs have recently become available. We provide data on their growth compared to comparable pigs that do not include GHRKO, and we have reviewed the literature regarding the effect of GHRKO, and its relevance to xenotransplantation.
Collapse
Affiliation(s)
- Hayato Iwase
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | | | | | - David K C Cooper
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
22
|
MicroRNA-21-Enriched Exosomes as Epigenetic Regulators in Melanomagenesis and Melanoma Progression: The Impact of Western Lifestyle Factors. Cancers (Basel) 2020; 12:cancers12082111. [PMID: 32751207 PMCID: PMC7464294 DOI: 10.3390/cancers12082111] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 07/16/2020] [Accepted: 07/24/2020] [Indexed: 02/06/2023] Open
Abstract
DNA mutation-induced activation of RAS-BRAF-MEK-ERK signaling associated with intermittent or chronic ultraviolet (UV) irradiation cannot exclusively explain the excessive increase of malignant melanoma (MM) incidence since the 1950s. Malignant conversion of a melanocyte to an MM cell and metastatic MM is associated with a steady increase in microRNA-21 (miR-21). At the epigenetic level, miR-21 inhibits key tumor suppressors of the RAS-BRAF signaling pathway enhancing proliferation and MM progression. Increased MM cell levels of miR-21 either result from endogenous upregulation of melanocytic miR-21 expression or by uptake of miR-21-enriched exogenous exosomes. Based on epidemiological data and translational evidence, this review provides deeper insights into environmentally and metabolically induced exosomal miR-21 trafficking beyond UV-irradiation in melanomagenesis and MM progression. Sources of miR-21-enriched exosomes include UV-irradiated keratinocytes, adipocyte-derived exosomes in obesity, airway epithelium-derived exosomes generated by smoking and pollution, diet-related exosomes and inflammation-induced exosomes, which may synergistically increase the exosomal miR-21 burden of the melanocyte, the transformed MM cell and its tumor environment. Several therapeutic agents that suppress MM cell growth and proliferation attenuate miR-21 expression. These include miR-21 antagonists, metformin, kinase inhibitors, beta-blockers, vitamin D, and plant-derived bioactive compounds, which may represent new options for the prevention and treatment of MM.
Collapse
|
23
|
ElHady AK, Shih SP, Chen YC, Liu YC, Ahmed NS, Keeton AB, Piazza GA, Engel M, Abadi AH, Abdel-Halim M. Extending the use of tadalafil scaffold: Development of novel selective phosphodiesterase 5 inhibitors and histone deacetylase inhibitors. Bioorg Chem 2020; 98:103742. [PMID: 32199305 DOI: 10.1016/j.bioorg.2020.103742] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/27/2020] [Accepted: 03/07/2020] [Indexed: 12/17/2022]
Abstract
Herein we present the synthesis and characterization of a novel chemical series of tadalafil analogues that display different pharmacological profiles. Compounds that have the 6R, 12aR configuration and terminal carboxylic acid group at the side chain arising from the piperazinedione nitrogen were potent PDE5 inhibitors, with compound 11 having almost equal potency to tadalafil and superior selectivity over PDE11, the most common off-target for tadalafil. Modifying the stereochemistry into 6S, 12aS configuration and adopting the hydroxamic acid moiety as a terminal group gave rise to compounds that only inhibited HDAC. Dual PDE5/HDAC inhibition could be achieved with compounds having 6R, 12aR configuration and hydroxamic acid moiety as a terminal group. The anticancer activity of the synthesized compounds was evaluated against a diverse number of cell lines of different origin. The compounds elicited anticancer activity against cell lines belonging to lymphoproliferative cancer as well as solid tumors. Despite the previous reports suggesting anticancer activity of PDE5 inhibitors, the growth inhibitory activity of the compounds seemed to be solely dependent on HDAC inhibition. Compound 26 (pan HDAC IC50 = 14 nM, PDE5 IC50 = 46 nM) displayed the most potent anticancer activity in the present series and was shown to induce apoptosis in Molt-4 cells. HDAC isoform selectivity testing for compound 26 showed that it is more selective for HDAC6 and 8 over HDAC1 by more than 20-fold.
Collapse
Affiliation(s)
- Ahmed K ElHady
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Shou-Ping Shih
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University, 70 Lien-Hai Road, Kaohsiung 804, Taiwan; Doctoral Degree Program in Marine Biotechnology, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 115, Taiwan
| | - Yu-Cheng Chen
- The Ph.D. Program for Cancer Biology and Drug Discovery, China Medical University and Academia Sinica, Taichung 40402, Taiwan
| | - Yi-Chang Liu
- Division of Hematology-Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; Department of Internal Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Nermin S Ahmed
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Adam B Keeton
- Department of Oncologic Sciences and Pharmacology, Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36608, USA
| | - Gary A Piazza
- Department of Oncologic Sciences and Pharmacology, Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36608, USA
| | - Matthias Engel
- Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C2.3, D-66123 Saarbrücken, Germany
| | - Ashraf H Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Mohammad Abdel-Halim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt.
| |
Collapse
|
24
|
Buckels A, Zhang Y, Jiang J, Athar M, Afaq F, Shevde-Samant L, Frank SJ. Autocrine/paracrine actions of growth hormone in human melanoma cell lines. Biochem Biophys Rep 2020; 21:100716. [PMID: 31890904 PMCID: PMC6928330 DOI: 10.1016/j.bbrep.2019.100716] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 12/02/2019] [Accepted: 12/06/2019] [Indexed: 12/30/2022] Open
Abstract
Melanoma is the most aggressive skin cancer. Its aggressiveness is most commonly attributed to ERK pathway mutations leading to constitutive signaling. Though initial tumor regression results from targeting this pathway, resistance often emerges. Interestingly, interrogation of the NCI-60 database indicates high growth hormone receptor (GHR) expression in melanoma cell lines. To further characterize melanoma, we tested responsiveness to human growth hormone (GH). GH treatment resulted in GHR signaling and increased invasion and migration, which was inhibited by a GHR monoclonal antibody (mAb) antagonist in WM35, SK-MEL 5, SK-MEL 28 and SK-MEL 119 cell lines. We also detected GH in the conditioned medium (CM) of human melanoma cell lines. GHR, JAK2 and STAT5 were basally phosphorylated in these cell lines, consistent with autocrine/paracrine GH production. Together, our results suggest that melanomas are enriched in GHR and produce GH that acts in an autocrine/paracrine manner. We suggest that GHR may constitute a therapeutic target in melanoma.
Collapse
Affiliation(s)
- Ashiya Buckels
- Department of Medicine Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Yue Zhang
- Department of Medicine Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jing Jiang
- Department of Medicine Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mohammad Athar
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Farrukh Afaq
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lalita Shevde-Samant
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Stuart J. Frank
- Department of Medicine Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Medical Service, Veterans Affairs Medical Center, Birmingham, AL, USA
| |
Collapse
|
25
|
Wang X, Wang S, Wu H, Jiang M, Xue H, Zhu Y, Wang C, Zha X, Wen Y. Human growth hormone level decreased in women aged <60 years but increased in men aged >50 years. Medicine (Baltimore) 2020; 99:e18440. [PMID: 31914017 PMCID: PMC6959966 DOI: 10.1097/md.0000000000018440] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
To investigate the relationship amongst human growth hormone (HGH), sex, and age groups.A cross-sectional study was conducted on a health check-up population from Wannan area of China from 2014 to 2016. The study involved 6843 individuals aged 23 to 85 years. Logistic regression analysis and smooth curve were applied to determine the relationship amongst age, sex, and HGH.The average level of HGH in the population was 0.37 ± 0.59 ng/mL. There were significant differences in sex, age, body mass index (BMI), triglycerides (TG), total cholesterol (TC), systolic blood pressure (SBP), diastolic blood pressure (DBP), and glucose (GLU) amongst different quartiles of HGH (P < .001). A U-shape relationship was established between HGH and age. After sex stratification, the results showed that the thresholds of age were 60 years in women, and 50 years in men, after adjusting for body mass index, triglycerides, total cholesterol, blood pressure, and blood glucose. Logistic regression showed that HGH level decreased in women aged <60 years (OR = 1.472, P < .001) and increased in men aged >50 years (OR = 0.711, P < .001). So the distributive characteristics of HGH concentration vary with sex and age group.
Collapse
Affiliation(s)
| | | | - Huan Wu
- School of Laboratory Medicine
| | | | | | | | | | - Xiaojuan Zha
- First Affiliated Hospital, Wannan Medical College, Wuhu City, Anhui Province, China
| | | |
Collapse
|
26
|
Chesnokova V, Melmed S. Growth hormone in the tumor microenvironment. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2019; 63:568-575. [PMID: 31939481 PMCID: PMC7025769 DOI: 10.20945/2359-3997000000186] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 09/27/2019] [Indexed: 12/20/2022]
Abstract
Tumor development is a multistep process whereby local mechanisms enable somatic mutations during preneoplastic stages. Once a tumor develops, it becomes a complex organ composed of multiple cell types. Interactions between malignant and non-transformed cells and tissues create a tumor microenvironment (TME) comprising epithelial cancer cells, cancer stem cells, non-tumorous cells, stromal cells, immune-inflammatory cells, blood and lymphatic vascular network, and extracellular matrix. We review reports and present a hypothesis that postulates the involvement of growth hormone (GH) in field cancerization. We discuss GH contribution to TME, promoting epithelial-to-mesenchymal transition, accumulation of unrepaired DNA damage, tumor vascularity, and resistance to therapy. Arch Endocrinol Metab. 2019;63(6):568-75.
Collapse
Affiliation(s)
- Vera Chesnokova
- Pituitary CenterDepartment of MedicineCedars-Sinai Medical CenterLos AngelesCAUSAPituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Shlomo Melmed
- Pituitary CenterDepartment of MedicineCedars-Sinai Medical CenterLos AngelesCAUSAPituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
27
|
Basu R, Kopchick JJ. The effects of growth hormone on therapy resistance in cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2019; 2:827-846. [PMID: 32382711 PMCID: PMC7204541 DOI: 10.20517/cdr.2019.27] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pituitary derived and peripherally produced growth hormone (GH) is a crucial mediator of longitudinal growth, organ development, metabolic regulation with tissue specific, sex specific, and age-dependent effects. GH and its cognate receptor (GHR) are expressed in several forms of cancer and have been validated as an anti-cancer target through a large body of in vitro, in vivo and epidemiological analyses. However, the underlying molecular mechanisms of GH action in cancer prognosis and therapeutic response had been sparse until recently. This review assimilates the critical details of GH-GHR mediated therapy resistance across different cancer types, distilling the therapeutic implications based on our current understanding of these effects.
Collapse
Affiliation(s)
- Reetobrata Basu
- Ohio University Heritage College of Osteopathic Medicine (OU-HCOM), Ohio University, Athens, OH 45701, USA.,Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA
| | - John J Kopchick
- Ohio University Heritage College of Osteopathic Medicine (OU-HCOM), Ohio University, Athens, OH 45701, USA.,Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA
| |
Collapse
|
28
|
Basu R, Kulkarni P, Qian Y, Walsh C, Arora P, Davis E, Duran-Ortiz S, Funk K, Ibarra D, Kruse C, Mathes S, McHugh T, Brittain A, Berryman DE, List EO, Okada S, Kopchick JJ. Growth Hormone Upregulates Melanocyte-Inducing Transcription Factor Expression and Activity via JAK2-STAT5 and SRC Signaling in GH Receptor-Positive Human Melanoma. Cancers (Basel) 2019; 11:E1352. [PMID: 31547367 PMCID: PMC6769493 DOI: 10.3390/cancers11091352] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 08/30/2019] [Accepted: 09/09/2019] [Indexed: 02/07/2023] Open
Abstract
Growth hormone (GH) facilitates therapy resistance in the cancers of breast, colon, endometrium, and melanoma. The GH-stimulated pathways responsible for this resistance were identified as suppression of apoptosis, induction of epithelial-to-mesenchymal transition (EMT), and upregulated drug efflux by increased expression of ATP-binding cassette containing multidrug efflux pumps (ABC-transporters). In extremely drug-resistant melanoma, ABC-transporters have also been reported to mediate drug sequestration in intracellular melanosomes, thereby reducing drug efficacy. Melanocyte-inducing transcription factor (MITF) is the master regulator of melanocyte and melanoma cell fate as well as the melanosomal machinery. MITF targets such as the oncogene MET, as well as MITF-mediated processes such as resistance to radiation therapy, are both known to be upregulated by GH. Therefore, we chose to query the direct effects of GH on MITF expression and activity towards conferring chemoresistance in melanoma. Here, we demonstrate that GH significantly upregulates MITF as well as the MITF target genes following treatment with multiple anticancer drug treatments such as chemotherapy, BRAF-inhibitors, as well as tyrosine-kinase inhibitors. GH action also upregulated MITF-regulated processes such as melanogenesis and tyrosinase activity. Significant elevation in MITF and MITF target gene expression was also observed in mouse B16F10 melanoma cells and xenografts in bovine GH transgenic (bGH) mice compared to wild-type littermates. Through pathway inhibitor analysis we identified that both the JAK2-STAT5 and SRC activities were critical for the observed effects. Additionally, a retrospective analysis of gene expression data from GTEx, NCI60, CCLE, and TCGA databases corroborated our observed correlation of MITF function and GH action. Therefore, we present in vitro, in vivo, and in silico evidence which strongly implicates the GH-GHR axis in inducing chemoresistance in human melanoma by driving MITF-regulated and ABC-transporter-mediated drug clearance pathways.
Collapse
Affiliation(s)
- Reetobrata Basu
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA.
| | - Prateek Kulkarni
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA.
- Molecular and Cellular Biology (MCB) Program, Department of Biological Sciences, Ohio University, Athens, OH 45701, USA.
| | - Yanrong Qian
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA.
| | - Christopher Walsh
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA.
| | - Pranay Arora
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA.
| | - Emily Davis
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA.
- Department of Chemistry and Biochemistry, Ohio University, Athens, OH 45701, USA.
| | - Silvana Duran-Ortiz
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA.
- Molecular and Cellular Biology (MCB) Program, Department of Biological Sciences, Ohio University, Athens, OH 45701, USA.
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA.
| | - Kevin Funk
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA.
- Molecular and Cellular Biology (MCB) Program, Department of Biological Sciences, Ohio University, Athens, OH 45701, USA.
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA.
| | - Diego Ibarra
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA.
- Department of Chemistry and Biochemistry, Ohio University, Athens, OH 45701, USA.
| | - Colin Kruse
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA.
- Molecular and Cellular Biology (MCB) Program, Department of Biological Sciences, Ohio University, Athens, OH 45701, USA.
| | - Samuel Mathes
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA.
| | - Todd McHugh
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA.
| | - Alison Brittain
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA.
- Molecular and Cellular Biology (MCB) Program, Department of Biological Sciences, Ohio University, Athens, OH 45701, USA.
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA.
| | - Darlene E Berryman
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA.
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA.
- The Diabetes Institute, Ohio University, Athens, OH 45701, USA.
| | - Edward O List
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA.
| | - Shigeru Okada
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA.
- The Diabetes Institute, Ohio University, Athens, OH 45701, USA.
- Department of Pediatrics, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA.
| | - John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA.
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA.
| |
Collapse
|
29
|
Vena GA, Cassano N, Caccavale S, Argenziano G. Association Between Melanoma Risk and Height: A Narrative Review. Dermatol Pract Concept 2019; 9:82-89. [PMID: 31106009 DOI: 10.5826/dpc.0902a02] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2019] [Indexed: 02/02/2023] Open
Abstract
The link between anthropometric indices, including height, and cancer risk and/or progression has attracted considerable interest in recent years. Adult height results from the complex interplay between genetic, hormonal, nutritional, and other environmental factors and has been found to contribute to the risk of several selected malignancies, although it has not been implicated as a real cause per se. A number of studies have investigated the height-melanoma relationship, showing controversial results so far. In this review, we summarize the epidemiological data regarding the association between height and melanoma risk and analyze the potential underlying mechanisms.
Collapse
Affiliation(s)
- Gino A Vena
- Dermatology and Venereology Private Practice, Bari, Italy.,Dermatology and Venereology Private Practice, Barletta, Italy
| | - Nicoletta Cassano
- Dermatology and Venereology Private Practice, Bari, Italy.,Dermatology and Venereology Private Practice, Barletta, Italy
| | - Stefano Caccavale
- Dermatology Unit, University of Campania Luigi Vanvitelli, Naples, Italy
| | | |
Collapse
|
30
|
Boguszewski CL, Boguszewski MCDS. Growth Hormone's Links to Cancer. Endocr Rev 2019; 40:558-574. [PMID: 30500870 DOI: 10.1210/er.2018-00166] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 10/23/2018] [Indexed: 12/13/2022]
Abstract
Several components of the GH axis are involved in tumor progression, and GH-induced intracellular signaling has been strongly associated with breast cancer susceptibility in genome-wide association studies. In the general population, high IGF-I levels and low IGF-binding protein-3 levels within the normal range are associated with the development of common malignancies, and components of the GH-IGF signaling system exhibit correlations with clinical, histopathological, and therapeutic parameters in cancer patients. Despite promising findings in preclinical studies, anticancer therapies targeting the GH-IGF signaling system have led to disappointing results in clinical trials. There is substantial evidence for some degree of protection against tumor development in several animal models and in patients with genetic defects associated with GH deficiency or resistance. In contrast, the link between GH excess and cancer risk in acromegaly patients is much less clear, and cancer screening in acromegaly has been a highly controversial issue. Recent studies have shown that increased life expectancy in acromegaly patients who attain normal GH and IGF-I levels is associated with more deaths due to age-related cancers. Replacement GH therapy in GH deficiency hypopituitary adults and short children has been shown to be safe when no other risk factors for malignancy are present. Nevertheless, the use of GH in cancer survivors and in short children with RASopathies, chromosomal breakage syndromes, or DNA-repair disorders should be carefully evaluated owing to an increased risk of recurrence, primary cancer, or second neoplasia in these individuals.
Collapse
Affiliation(s)
- Cesar Luiz Boguszewski
- Department of Internal Medicine, Endocrine Division (SEMPR), University Hospital, Federal University of Parana, Curitiba, Brazil
| | | |
Collapse
|
31
|
Barry S, Carlsen E, Marques P, Stiles CE, Gadaleta E, Berney DM, Roncaroli F, Chelala C, Solomou A, Herincs M, Caimari F, Grossman AB, Crnogorac-Jurcevic T, Haworth O, Gaston-Massuet C, Korbonits M. Tumor microenvironment defines the invasive phenotype of AIP-mutation-positive pituitary tumors. Oncogene 2019; 38:5381-5395. [PMID: 30867568 PMCID: PMC6755983 DOI: 10.1038/s41388-019-0779-5] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 12/07/2018] [Accepted: 01/18/2019] [Indexed: 12/17/2022]
Abstract
The molecular mechanisms leading to aryl hydrocarbon receptor interacting protein (AIP) mutation-induced aggressive, young-onset growth hormone-secreting pituitary tumors are not fully understood. In this study, we have identified that AIP-mutation-positive tumors are infiltrated by a large number of macrophages compared to sporadic tumors. Tissue from pituitary-specific Aip-knockout (AipFlox/Flox;Hesx1Cre/+) mice recapitulated this phenotype. Our human pituitary tumor transcriptome data revealed the "epithelial-to-mesenchymal transition (EMT) pathway" as one of the most significantly altered pathways in AIPpos tumors. Our in vitro data suggest that bone marrow-derived macrophage-conditioned media induces more prominent EMT-like phenotype and enhanced migratory and invasive properties in Aip-knockdown somatomammotroph cells compared to non-targeting controls. We identified that tumor-derived cytokine CCL5 is upregulated in AIP-mutation-positive human adenomas. Aip-knockdown GH3 cell-conditioned media increases macrophage migration, which is inhibited by the CCL5/CCR5 antagonist maraviroc. Our results suggest that a crosstalk between the tumor and its microenvironment plays a key role in the invasive nature of AIP-mutation-positive tumors and the CCL5/CCR5 pathway is a novel potential therapeutic target.
Collapse
Affiliation(s)
- Sayka Barry
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
| | | | - Pedro Marques
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Craig E Stiles
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Emanuela Gadaleta
- Molecular Oncology, Barts Cancer Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Dan M Berney
- Molecular Oncology, Barts Cancer Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Federico Roncaroli
- Division of Neuroscience & Experimental Psychology, University of Manchester, Manchester, M13 9PL, UK
| | - Claude Chelala
- Molecular Oncology, Barts Cancer Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Antonia Solomou
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Maria Herincs
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Francisca Caimari
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Ashley B Grossman
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Tatjana Crnogorac-Jurcevic
- Molecular Oncology, Barts Cancer Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Oliver Haworth
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Carles Gaston-Massuet
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Márta Korbonits
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK.
| |
Collapse
|
32
|
Palladium based nanoparticles for the treatment of advanced melanoma. Sci Rep 2019; 9:3255. [PMID: 30824801 PMCID: PMC6397149 DOI: 10.1038/s41598-019-40258-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 01/16/2019] [Indexed: 12/19/2022] Open
Abstract
IGF1R and CD44 are overexpressed in most advanced melanomas so we designed chemotherapeutic nanoparticles to target those receptors. Tris(dibenzylideneacetone)dipalladium (Tris DBA-Pd) is a novel inhibitor of N-myristoyltransferase 1 (NMT-1) and has proven in vivo activity against melanoma. However, poor solubility impairs its effectiveness. To improve its therapeutic efficacy and overcome drug resistance in advanced melanomas, we synthesized Tris DBA-Pd hyaluronic acid nanoparticles (Tris DBA-Pd HANP) and evaluated them against in vivo xenografts of LM36R, an aggressive BRAF mutant human melanoma resistant to BRAF inhibitors. We treated xenografted mice in four arms: empty HANPs, free Tris DBA-Pd, Tris DBA-Pd HANPs, and Tris DBA-Pd HANPs with IGF1R antibody. The Tris DBA-Pd HANP group was the most responsive to treatment and showed the greatest depletion of CD44-positive cells on IHC. Surprisingly, the HANP containing IGF1R antibody was less effective than particles without antibody, possibly due to steric hindrance of IGF1R and CD44 binding. Tris DBA-Pd nanoparticles are an effective therapy for CD44-positive tumors like melanoma, and further development of these nanoparticles should be pursued.
Collapse
|
33
|
Lu M, Flanagan JU, Langley RJ, Hay MP, Perry JK. Targeting growth hormone function: strategies and therapeutic applications. Signal Transduct Target Ther 2019; 4:3. [PMID: 30775002 PMCID: PMC6367471 DOI: 10.1038/s41392-019-0036-y] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 12/10/2018] [Accepted: 12/12/2018] [Indexed: 01/12/2023] Open
Abstract
Human growth hormone (GH) is a classical pituitary endocrine hormone that is essential for normal postnatal growth and has pleiotropic effects across multiple physiological systems. GH is also expressed in extrapituitary tissues and has localized autocrine/paracrine effects at these sites. In adults, hypersecretion of GH causes acromegaly, and strategies that block the release of GH or that inhibit GH receptor (GHR) activation are the primary forms of medical therapy for this disease. Overproduction of GH has also been linked to cancer and the microvascular complications that are associated with diabetes. However, studies to investigate the therapeutic potential of GHR antagonism in these diseases have been limited, most likely due to difficulty in accessing therapeutic tools to study the pharmacology of the receptor in vivo. This review will discuss current and emerging strategies for antagonizing GH function and the potential disease indications. Emerging therapies are offering an expanded toolkit for combatting the effects of human growth hormone overproduction. Human growth hormone (GH) is a major driver of postnatal growth; however, systemic or localized overproduction is implicated in the aberrant growth disease acromegaly, cancer, and diabetes. In this review, researchers led by Jo Perry, from the University of Auckland, New Zealand, discuss strategies that either inhibit GH production, block its systemic receptor, or interrupt its downstream signaling pathways. The only licensed GH receptor blocker is pegvisomant, but therapies are in development that include long-acting protein and antibody-based blockers, and nucleotide complexes that degrade GHR production have also shown promise. Studies investigating GHR antagonism are limited, partly due to difficulty in accessing therapeutic tools which block GHR function, but overcoming these obstacles may yield advances in alleviating chronic disease.
Collapse
Affiliation(s)
- Man Lu
- 1Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Jack U Flanagan
- 2Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand.,3Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Ries J Langley
- 3Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand.,4Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Michael P Hay
- 2Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand.,3Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Jo K Perry
- 1Liggins Institute, University of Auckland, Auckland, New Zealand.,3Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| |
Collapse
|
34
|
Chesnokova V, Zonis S, Barrett R, Kameda H, Wawrowsky K, Ben-Shlomo A, Yamamoto M, Gleeson J, Bresee C, Gorbunova V, Melmed S. Excess growth hormone suppresses DNA damage repair in epithelial cells. JCI Insight 2019; 4:e125762. [PMID: 30728323 PMCID: PMC6413789 DOI: 10.1172/jci.insight.125762] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 12/20/2018] [Indexed: 12/18/2022] Open
Abstract
Growth hormone (GH) decreases with age, and GH therapy has been advocated by some to sustain lean muscle mass and vigor in aging patients and advocated by athletes to enhance performance. Environmental insults and aging lead to DNA damage, which - if unrepaired - results in chromosomal instability and tumorigenesis. We show that GH suppresses epithelial DNA damage repair and blocks ataxia telangiectasia mutated (ATM) kinase autophosphorylation with decreased activity. Decreased phosphorylation of ATM target proteins p53, checkpoint kinase 2 (Chk2), and histone 2A variant led to decreased DNA repair by nonhomologous end-joining. In vivo, prolonged high GH levels resulted in a 60% increase in unrepaired colon epithelial DNA damage. GH suppression of ATM was mediated by induced tripartite motif containing protein 29 (TRIM29) and attenuated tat interacting protein 60 kDa (Tip60). By contrast, DNA repair was increased in human nontumorous colon cells (hNCC) where GH receptor (GHR) was stably suppressed and in colon tissue derived from GHR-/- mice. hNCC treated with etoposide and GH showed enhanced transformation, as evidenced by increased growth in soft agar. In mice bearing human colon GH-secreting xenografts, metastatic lesions were increased. The results elucidate a mechanism underlying GH-activated epithelial cell transformation and highlight an adverse risk for inappropriate adult GH treatment.
Collapse
Affiliation(s)
| | | | - Robert Barrett
- Board of Governors Regenerative Medicine Institute
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Department of Medicine, and
| | | | | | | | | | - John Gleeson
- Board of Governors Regenerative Medicine Institute
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Department of Medicine, and
| | - Catherine Bresee
- Biostatistics and Bioinformatics Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Vera Gorbunova
- Department of Biology, University of Rochester, Rochester, New York, USA
| | | |
Collapse
|
35
|
Gadelha MR, Kasuki L, Lim DST, Fleseriu M. Systemic Complications of Acromegaly and the Impact of the Current Treatment Landscape: An Update. Endocr Rev 2019; 40:268-332. [PMID: 30184064 DOI: 10.1210/er.2018-00115] [Citation(s) in RCA: 206] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 07/26/2018] [Indexed: 12/19/2022]
Abstract
Acromegaly is a chronic systemic disease with many complications and is associated with increased mortality when not adequately treated. Substantial advances in acromegaly treatment, as well as in the treatment of many of its complications, mainly diabetes mellitus, heart failure, and arterial hypertension, were achieved in the last decades. These developments allowed change in both prevalence and severity of some acromegaly complications and furthermore resulted in a reduction of mortality. Currently, mortality seems to be similar to the general population in adequately treated patients with acromegaly. In this review, we update the knowledge in complications of acromegaly and detail the effects of different acromegaly treatment options on these complications. Incidence of mortality, its correlation with GH (cumulative exposure vs last value), and IGF-I levels and the shift in the main cause of mortality in patients with acromegaly are also addressed.
Collapse
Affiliation(s)
- Mônica R Gadelha
- Neuroendocrinology Research Center/Endocrine Section and Medical School, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Neuroendocrine Section, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde do Rio de Janeiro, Rio de Janeiro, Brazil.,Neuropathology and Molecular Genetics Laboratory, Instituto Estadual do Cérebro Paulo Niemeyer, Rio de Janeiro, Brazil
| | - Leandro Kasuki
- Neuroendocrinology Research Center/Endocrine Section and Medical School, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Neuroendocrine Section, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde do Rio de Janeiro, Rio de Janeiro, Brazil.,Endocrine Unit, Hospital Federal de Bonsucesso, Rio de Janeiro, Brazil
| | - Dawn S T Lim
- Department of Endocrinology, Singapore General Hospital, Singapore, Singapore
| | - Maria Fleseriu
- Department of Endocrinology, Diabetes and Metabolism, Oregon Health and Science University, Portland, Oregon.,Department of Neurological Surgery, Oregon Health and Science University, Portland, Oregon.,Northwest Pituitary Center, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
36
|
Duran-Ortiz S, Bell S, Kopchick JJ. Standardizing protocols dealing with growth hormone receptor gene disruption in mice using the Cre-lox system. Growth Horm IGF Res 2018; 42-43:52-57. [PMID: 30195091 PMCID: PMC9704043 DOI: 10.1016/j.ghir.2018.08.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 07/19/2018] [Accepted: 08/27/2018] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Mice and humans with reduced growth hormone (GH) action before birth are conferred positive health- and life-span advantages. However, little work has been performed to study the effect of conditional disruption of GH action in adult life. With this as our objective, we sought to elucidate a reproducible protocol that allows generation of adult mice with a global disruption of the GH receptor (Ghr) gene, using the tamoxifen (TAM)-inducible Cre-lox system, driven by the ROSA26 enhancer/promoter. Here we report the optimum conditions for the gene disruption. DESIGN Six month old mice, homozygous for the ROSA26-Cre and the Ghr-floxed gene, were injected, once daily for five days with four distinct TAM doses (from 0.08 to 0.32 mg of TAM/g of body weight). To evaluate the most effective TAM dose that leads to global disruption of the GHR, mRNA expression of the Ghr and insulin growth factor-1 (Igf1) genes were assessed in liver, adipose tissue, kidney, and skeletal and cardiac muscles of experimental and control mice. Additionally, serum GH and IGF-1 levels were evaluated one month after TAM injections in both, TAM-treated and TAM-untreated control mice. RESULTS A dose of 0.25 mg of TAM/g of body weight was sufficient to significantly reduce the Ghr and Igf1 expression levels in the liver, fat, kidney, and skeletal and cardiac muscle of six-month old mice that are homozygous for the Ghr floxed gene and Cre recombinase. The reduction of the Ghr mRNA levels of the TAM-treated mice was variable between tissues, with liver and adipose tissue showing the lowest and skeletal and cardiac muscle the highest levels of Ghr gene expression when compared to control mice. Moreover, liver tissue showed the 'best' Ghr gene disruption, resulting in decreased total circulating IGF-1 levels while GH levels were increased versus control mice. CONCLUSION The results show that in mice at six months of age, a total TAM dose of at least 0.25 mg of TAM/g of body weight is needed for a global downregulation of Ghr gene expression with a regimen of 100 μL intraperitoneal (ip) TAM injections, once daily for five consecutive days. Furthermore, we found that even though this system does not achieve an equivalent disruption of the Ghr between tissues, the circulating IGF-1 is >95% decreased. This work helped to create adult mice with a global GHR knockdown.
Collapse
Affiliation(s)
- Silvana Duran-Ortiz
- Edison Biotechnology Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, United States; Department of Biological Sciences, College of Arts and Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, United States; Molecular and Cellular Biology Program, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, United States.
| | - Stephen Bell
- Edison Biotechnology Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, United States.
| | - John J Kopchick
- Edison Biotechnology Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, United States; Molecular and Cellular Biology Program, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, United States; Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, United States.
| |
Collapse
|
37
|
Li T, Zhang C, Hassan S, Liu X, Song F, Chen K, Zhang W, Yang J. Histone deacetylase 6 in cancer. J Hematol Oncol 2018; 11:111. [PMID: 30176876 PMCID: PMC6122547 DOI: 10.1186/s13045-018-0654-9] [Citation(s) in RCA: 221] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 08/22/2018] [Indexed: 12/15/2022] Open
Abstract
Histone acetylation and deacetylation are important epigenetic mechanisms that regulate gene expression and transcription. Histone deacetylase 6 (HDAC6) is a unique member of the HDAC family that not only participates in histone acetylation and deacetylation but also targets several nonhistone substrates, such as α-tubulin, cortactin, and heat shock protein 90 (HSP90), to regulate cell proliferation, metastasis, invasion, and mitosis in tumors. Furthermore, HDAC6 also upregulates several critical factors in the immune system, such as program death receptor-1 (PD-1) and program death receptor ligand-1 (PD-L1) receptor, which are the main targets for cancer immunotherapy. Several selective HDAC6 inhibitors are currently in clinical trials for cancer treatment and bring hope for patients with malignant tumors. A fuller understanding of HDAC6 as a critical regulator of many cellular pathways will help further the development of targeted anti-HDAC6 therapies. Here, we review the unique features of HDAC6 and its role in cancer, which make HDAC6 an appealing drug target.
Collapse
Affiliation(s)
- Ting Li
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, People's Republic of China.,National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, People's Republic of China
| | - Chao Zhang
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, People's Republic of China.,National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, People's Republic of China
| | - Shafat Hassan
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, People's Republic of China.,National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, People's Republic of China.,International Medical School, Tianjin Medical University, Tianjin, 300061, People's Republic of China
| | - Xinyue Liu
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, People's Republic of China.,National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, People's Republic of China
| | - Fengju Song
- Department of Epidemiology and Biostatistics, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, People's Republic of China
| | - Kexin Chen
- Department of Epidemiology and Biostatistics, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, People's Republic of China
| | - Wei Zhang
- Cancer Genomics and Precision Medicine, Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, USA
| | - Jilong Yang
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, People's Republic of China. .,National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, People's Republic of China.
| |
Collapse
|
38
|
Marinho CG, Mermejo LM, Salvatori R, Assirati JA, Oliveira CRP, Santos EG, Leal ÂCGB, Barros-Oliveira CS, Damascena NP, Lima CA, Farias CT, Moreira AC, Aguiar-Oliveira MH. Occurrence of neoplasms in individuals with congenital, severe GH deficiency from the Itabaianinha kindred. Growth Horm IGF Res 2018; 41:71-74. [PMID: 29571594 DOI: 10.1016/j.ghir.2018.03.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 03/10/2018] [Accepted: 03/11/2018] [Indexed: 11/18/2022]
Abstract
Growth hormone (GH) and the insulin-like growth factor I (IGF-I) have cell proliferative and differentiation properties. Whether these hormones have a role in mutagenesis is unknown. Nevertheless, severe IGF-I deficiency seems to confer protection against the development of neoplasms. Here, we report five cases of adult patients with severe and congenital isolated GH deficiency (IGHD) due to the c.57+1G>A mutation in the GHRH receptor gene, who developed tumors. Four GH-naïve subjects presented skin tumors: a 42-year-old man with a fibroepithelial polyp, a 53-year-old woman and two men (59 and 56 years old) with epidermoid skin cancers. One of these died from it after three surgeries and radiotherapy. The fifth patient was a 25-year-old woman, who had intermittently received GH replacement therapy (GHRT) from age 11 to 18, who developed an ependymoma extending from the fourth ventricle to the end of the thoracic spine. She underwent three surgical procedures, without obvious evidence of tumor recurrence during the six years follow up. These observations suggest that severe IGHD does not protect completely from development of tumors.
Collapse
Affiliation(s)
- Cindi G Marinho
- Division of Endocrinology, Federal University of Sergipe, 49060-100 Aracaju, Sergipe, Brazil
| | - Lívia M Mermejo
- Division of Endocrinology, Faculty of Medicine of Ribeirao Preto of the University of São Paulo (USP), 14049-900 Ribeirao Preto, São Paulo, Brazil
| | - Roberto Salvatori
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, The Johns Hopkins University School of Medicine Baltimore, MD 21287, USA
| | - João A Assirati
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine of Ribeirao Preto of the University of São Paulo (USP), 14049-900 Ribeirao Preto, São Paulo, Brazil
| | - Carla R P Oliveira
- Division of Endocrinology, Federal University of Sergipe, 49060-100 Aracaju, Sergipe, Brazil
| | - Elenilde G Santos
- Division of Endocrinology, Federal University of Sergipe, 49060-100 Aracaju, Sergipe, Brazil
| | - Ângela C G B Leal
- Division of Endocrinology, Federal University of Sergipe, 49060-100 Aracaju, Sergipe, Brazil
| | | | - Nayra P Damascena
- Division of Endocrinology, Federal University of Sergipe, 49060-100 Aracaju, Sergipe, Brazil
| | - Carlos A Lima
- Division of Surgery, Federal University of Sergipe, 49060-100 Aracaju, Sergipe, Brazil
| | - Catarine T Farias
- Division of Endocrinology, Federal University of Sergipe, 49060-100 Aracaju, Sergipe, Brazil
| | - Ayrton C Moreira
- Division of Endocrinology, Faculty of Medicine of Ribeirao Preto of the University of São Paulo (USP), 14049-900 Ribeirao Preto, São Paulo, Brazil
| | | |
Collapse
|
39
|
Basu R, Qian Y, Kopchick JJ. MECHANISMS IN ENDOCRINOLOGY: Lessons from growth hormone receptor gene-disrupted mice: are there benefits of endocrine defects? Eur J Endocrinol 2018; 178:R155-R181. [PMID: 29459441 DOI: 10.1530/eje-18-0018] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 02/19/2018] [Indexed: 12/12/2022]
Abstract
Growth hormone (GH) is produced primarily by anterior pituitary somatotroph cells. Numerous acute human (h) GH treatment and long-term follow-up studies and extensive use of animal models of GH action have shaped the body of GH research over the past 70 years. Work on the GH receptor (R)-knockout (GHRKO) mice and results of studies on GH-resistant Laron Syndrome (LS) patients have helped define many physiological actions of GH including those dealing with metabolism, obesity, cancer, diabetes, cognition and aging/longevity. In this review, we have discussed several issues dealing with these biological effects of GH and attempt to answer the question of whether decreased GH action may be beneficial.
Collapse
Affiliation(s)
- Reetobrata Basu
- Edison Biotechnology Institute, Ohio University, Athens, Ohio, USA
| | - Yanrong Qian
- Edison Biotechnology Institute, Ohio University, Athens, Ohio, USA
| | - John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, Ohio, USA
- Ohio University Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA
| |
Collapse
|
40
|
Autocrine hGH stimulates oncogenicity, epithelial-mesenchymal transition and cancer stem cell-like behavior in human colorectal carcinoma. Oncotarget 2017; 8:103900-103918. [PMID: 29262609 PMCID: PMC5732775 DOI: 10.18632/oncotarget.21812] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 09/08/2017] [Indexed: 11/25/2022] Open
Abstract
Tumor derived human growth hormone (hGH) has been implicated in cancer development and progression. However, the specific functional role of autocrine/paracrine hGH in colorectal cancer (CRC) remains largely to be determined. Herein, we demonstrated a crucial oncogenic role of autocrine hGH in CRC progression. Elevated hGH expression was detected in CRC compared to normal colorectal tissue, and hGH expression in CRC was positively associated with tumor size and lymph node metastasis. Forced expression of hGH stimulated cell proliferation, survival, oncogenicity and epithelial to mesenchymal transition (EMT) of CRC cells, and promoted xenograft growth and local invasion in vivo. Autocrine hGH expression in CRC cells stimulated the activation of the ERK1/2 pathway, which in turn resulted in increased transcription of the mesenchymal marker FIBRONECTIN 1 and transcriptional repression of the epithelial marker E-CADHERIN. The autocrine hGH-stimulated increase in CRC cell proliferation, cell survival and EMT was abrogated upon ERK1/2 inhibition. Furthermore, autocrine hGH-stimulated CRC cell migration and invasion was dependent on the ERK1/2-mediated increase in FIBRONECTIN 1 expression and decrease in E-CADHERIN expression. Forced expression of hGH also enhanced CSC-like behavior of CRC cells, as characterized by increased colonosphere formation, ALDH-positive population and CSC marker expression. Autocrine hGH-enhanced cancer stem cell (CSC)-like behavior in CRC cells was also observed to be E-CADHERIN-dependent. Thus, autocrine hGH plays a critical role in CRC progression, and inhibition of hGH could be a promising targeted therapeutic approach to limit disease progression in metastatic CRC patients.
Collapse
|
41
|
Brittain AL, Basu R, Qian Y, Kopchick JJ. Growth Hormone and the Epithelial-to-Mesenchymal Transition. J Clin Endocrinol Metab 2017; 102:3662-3673. [PMID: 28938477 DOI: 10.1210/jc.2017-01000] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 07/31/2017] [Indexed: 02/07/2023]
Abstract
CONTEXT Previous studies have implicated growth hormone (GH) in the progression of several cancers, including breast, colorectal, and pancreatic. A mechanism by which GH may play this role in cancer is through the induction of the epithelial-to-mesenchymal transition (EMT). During the EMT process, epithelial cells lose their defining phenotypes, causing loss of cellular adhesion and increased cell migration. This review aims to carefully summarize the previous two decades of research that points to GH as an initiator of EMT, in both cancerous and noncancerous tissues. EVIDENCE ACQUISITION Sources were collected using PubMed and Google Scholar search engines by using specific GH- and/or EMT-related terms. Identified manuscripts were selected for further analysis based on presentation of GH-induced molecular markers of the EMT process in vivo or in vitro. EVIDENCE SYNTHESIS Cellular mechanisms involved in GH-induced EMT are the focus of this review, both in cancerous and noncancerous epithelial cells. CONCLUSIONS Our findings suggest that a myriad of molecular mechanisms are induced by GH that cause EMT and may point to potential therapeutic use of GH antagonists or any downregulator of GH action in EMT-related disease.
Collapse
Affiliation(s)
- Alison L Brittain
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701
- Ohio University Heritage College of Osteopathic Medicine, Athens, Ohio 45701
| | - Reetobrata Basu
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701
| | - Yanrong Qian
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701
| | - John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701
- Ohio University Heritage College of Osteopathic Medicine, Athens, Ohio 45701
| |
Collapse
|
42
|
Abstract
The growth hormone (GH) and insulin-like growth factor-1 (IGF1) axis is the key regulator of longitudinal growth, promoting postnatal bone and muscle growth. The available data suggest that GH expression by tumour cells is associated with the aetiology and progression of various cancers such as endometrial, breast, liver, prostate, and colon cancer. Accordingly there has been increased interest in targeting GH-mediated signal transduction in a therapeutic setting. Because GH has endocrine, autocrine, and paracrine actions, therapeutic strategies will need to take into account systemic and local functions. Activation of related hormone receptors and crosstalk with other signalling pathways are also key considerations.
Collapse
Affiliation(s)
- Jo K Perry
- Liggins Institute, University of Auckland, 1023 Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, 1023 Auckland, New Zealand
| | - Zheng-Sheng Wu
- Department of Pathology, Anhui Medical University, Hefei, Anhui, PR China
| | - Hichem C Mertani
- Centre de Recherche en Cancérologie de Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1052-Centre National de la Recherche Scientifique (CNRS) 5286, Centre Léon Bérard, Université Claude Bernard Lyon I, Université de Lyon, Lyon, France
| | - Tao Zhu
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, PR China
| | - Peter E Lobie
- Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, 117456 Singapore; Tsinghua Berkeley Shenzhen Institute, Tsinghua University Graduate School, Shenzhen, PR China.
| |
Collapse
|
43
|
Basu R, Baumgaertel N, Wu S, Kopchick JJ. Growth Hormone Receptor Knockdown Sensitizes Human Melanoma Cells to Chemotherapy by Attenuating Expression of ABC Drug Efflux Pumps. HORMONES & CANCER 2017; 8:143-156. [PMID: 28293855 PMCID: PMC10355985 DOI: 10.1007/s12672-017-0292-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Accepted: 03/02/2017] [Indexed: 12/16/2022]
Abstract
Melanoma remains one of the most therapy-resistant forms of human cancer despite recent introductions of highly efficacious targeted therapies. The intrinsic therapy resistance of human melanoma is largely due to abundant expression of a repertoire of xenobiotic efflux pumps of the ATP-binding cassette (ABC) transporter family. Here, we report that GH action is a key mediator of chemotherapeutic resistance in human melanoma cells. We investigated multiple ABC efflux pumps (ABCB1, ABCB5, ABCB8, ABCC1, ABCC2, ABCG1, and ABCG2) reportedly associated with melanoma drug resistance in different human melanoma cells and tested the efficacy of five different anti-cancer compounds (cisplatin, doxorubicin, oridonin, paclitaxel, vemurafenib) with decreased GH action. We found that GH treatment of human melanoma cells upregulates expression of multiple ABC transporters and increases the EC50 of melanoma drug vemurafenib. Also, vemurafenib-resistant melanoma cells had upregulated levels of GH receptor (GHR) expression as well as ABC efflux pumps. GHR knockdown (KD) using siRNA in human melanoma cells treated with sub-EC50 doses of anti-tumor compounds resulted in significantly increased drug retention, decreased cell proliferation and increased drug efficacy, compared to mock-transfected controls. Our set of findings identify an unknown mechanism of GH regulation in mediating melanoma drug resistance and validates GHR as a unique therapeutic target for sensitizing highly therapy-resistant human melanoma cells to lower doses of anti-cancer drugs.
Collapse
Affiliation(s)
- Reetobrata Basu
- Edison Biotechnology Institute, Konneker Research Laboratory 206, Ohio University, Athens, OH, 45701, USA
- Molecular and Cell Biology Program, Ohio University, Athens, OH, USA
| | - Nicholas Baumgaertel
- Edison Biotechnology Institute, Konneker Research Laboratory 206, Ohio University, Athens, OH, 45701, USA
- Department of Biological Sciences, Ohio University, Athens, OH, USA
| | - Shiyong Wu
- Edison Biotechnology Institute, Konneker Research Laboratory 206, Ohio University, Athens, OH, 45701, USA
- Molecular and Cell Biology Program, Ohio University, Athens, OH, USA
| | - John J Kopchick
- Edison Biotechnology Institute, Konneker Research Laboratory 206, Ohio University, Athens, OH, 45701, USA.
- Molecular and Cell Biology Program, Ohio University, Athens, OH, USA.
- Heritage College of Osteopathic Medicine, Athens, OH, USA.
| |
Collapse
|