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
|
Yang J, Gelb MB, Tamshen K, Forsythe NL, Ko JH, Puente EG, Pelegri-O'Day E, Jamieson SMF, Perry JK, Maynard HD. Site-Selective Zwitterionic Poly(caprolactone-carboxybetaine)-Growth Hormone Receptor Antagonist Conjugate: Synthesis and Biological Evaluation. Biomacromolecules 2024; 25:7619-7629. [PMID: 39471281 DOI: 10.1021/acs.biomac.4c00828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2024]
Abstract
Zwitterionic polymers have been found to be biocompatible alternatives to poly(ethylene glycol) (PEG) for conjugation to proteins. This work reports the site-selective conjugation of poly(caprolactone-carboxybetaine) (pCLZ) to human growth hormone receptor antagonist (GHA) B2036-alkyne and investigation of safety, activity, and pharmacokinetics. Azide-end-functionalized pCLZs were synthesized and conjugated to GHA B2036-alkyne via copper-catalyzed click reaction. The resulting inhibitory bioactivity concentration responses in Ba/F3-GHR cells were compared to those of PEGylated GHA B2036. IgG and IgM antibody production was tested in mice, and no measurable antibody or cytokine production was detected for the pCLZ conjugate. Using 18F-labeled PET/CT imaging, the pCLZ conjugate showed an increase in circulation time compared to that of GHA B2036. Acute toxicity of the polymer was investigated in vivo and found to be nontoxic. Ex vivo degradation of the polymer on the conjugate was investigated. The results suggest that pCLZ-GHA is a potentially safe alternative to PEG-GHA.
Collapse
Affiliation(s)
- Jane Yang
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
- California NanoSystems Institute, University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1569, United States
| | - Madeline B Gelb
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
- California NanoSystems Institute, University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1569, United States
| | - Kyle Tamshen
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
- California NanoSystems Institute, University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1569, United States
| | - Neil L Forsythe
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
- California NanoSystems Institute, University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1569, United States
| | - Jeong Hoon Ko
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
- California NanoSystems Institute, University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1569, United States
| | - Ellie G Puente
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
- California NanoSystems Institute, University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1569, United States
| | - Emma Pelegri-O'Day
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
- California NanoSystems Institute, University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1569, United States
| | - Stephen M F Jamieson
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland 1023, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland 1023, New Zealand
| | - Jo K Perry
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland 1023, New Zealand
- Liggins Institute, University of Auckland, Auckland 1203, New Zealand
| | - Heather D Maynard
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
- California NanoSystems Institute, University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1569, United States
| |
Collapse
|
3
|
Wu G, Dong Y, Hu Q, Ma H, Xu Q, Xu K, Chen H, Yang Z, He M. HGH1 and the immune landscape: a novel prognostic marker for immune-desert tumor microenvironment identification and immunotherapy outcome prediction in human cancers. Cell Cycle 2023; 22:1969-1985. [PMID: 37811868 PMCID: PMC10761050 DOI: 10.1080/15384101.2023.2260163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 05/21/2023] [Accepted: 09/06/2023] [Indexed: 10/10/2023] Open
Abstract
HGH1 homolog, a protein-coding gene, plays a crucial role in human growth and development. However, its role in human cancer remains unclear. For the first time, this study comprehensively evaluated the potential involvement of HGH1 in cancer prognosis and immunological function. To achieve this, data from various databases, including The Cancer Genome Atlas, Genotype Tissue Expression, Cancer Cell Lineage Encyclopedia, Human Protein Atlas, cBioPortal, Tumor Immune Estimation Resource and Immune Cell Abundance Identifier, were collated, as well as from one large clinical study, three immunotherapy cohorts and in vitro experiments. This study aims to elucidate the role of HGH1 expression in cancer prognosis and immune response. Our findings revealed a significant association between increased HGH1 expression and a worse prognosis across various cancer types. Predominantly, copy number variations were identified as the most common genetic mutations. Additionally, HGH1 was observed to not only regulate cell cycle-related functions to promote cell proliferation but also influence autoimmunity-related functions within both the innate and adaptive immune systems, along with other relevant immune-related signaling pathways. Gene set enrichment analysis and gene set variation analysis were used to substantiate these findings. HGH1 overexpression contributed to an immune-deficient (immune-desert) tumor microenvironment, which was characterized by a significant expression of immune-related features such as immune-related gene and pathway expression and the number of immune-infiltrating cells. Furthermore, the correlation between HGH1 expression and tumor mutational burden in four cancers and microsatellite instability in eight cancers was observed. This suggests that HGH1 has potential as an immunotherapeutic target. Immunotherapy data analysis supports this notion, demonstrating that patients with low HGH1 expression treated with immune checkpoint inhibitors exhibit improved survival rates and a higher likelihood of responding to immunotherapy than patients with high HGH1 expression. Collectively, these findings highlight the significant role of HGH1 in human cancers, illuminating its involvement in tumorigenesis and cancer immunity. Elevated HGH1 expression was identified to be indicative of an immune-desert tumor microenvironment. Consequently, the targeting of HGH1, particularly in combination with immune checkpoint inhibitor therapy, holds promise for enhancing therapeutic outcomes in patients with cancer.
Collapse
Affiliation(s)
- Gujie Wu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Human Phenome Institute, Fudan University, Shanghai, China
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Yipeng Dong
- School of Medicine, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qin Hu
- Shanghai Medical College, Fudan University, Shanghai, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Huiyun Ma
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Qun Xu
- School of Medicine, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Kun Xu
- Department of Chemotherapy, Jiangsu Cancer Hospital, Nanjing Medical University, Nanjing, China
| | - Hongyu Chen
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zheng Yang
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Min He
- Shanghai Medical College, Fudan University, Shanghai, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
4
|
Wang Y, Kim M, Buckley C, Maynard HD, Langley RJ, Perry JK. Growth hormone receptor agonists and antagonists: From protein expression and purification to long-acting formulations. Protein Sci 2023; 32:e4727. [PMID: 37428391 PMCID: PMC10443362 DOI: 10.1002/pro.4727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/11/2023]
Abstract
Recombinant human growth hormone (rhGH) and GH receptor antagonists (GHAs) are used clinically to treat a range of disorders associated with GH deficiency or hypersecretion, respectively. However, these biotherapeutics can be difficult and expensive to manufacture with multiple challenges from recombinant protein generation through to the development of long-acting formulations required to improve the circulating half-life of the drug. In this review, we summarize methodologies and approaches used for making and purifying recombinant GH and GHA proteins, and strategies to improve pharmacokinetic and pharmacodynamic properties, including PEGylation and fusion proteins. Therapeutics that are in clinical use or are currently under development are also discussed.
Collapse
Affiliation(s)
- Yue Wang
- Liggins Institute, University of AucklandAucklandNew Zealand
- Maurice Wilkins Centre for Molecular BiodiscoveryAucklandNew Zealand
| | - Minah Kim
- Liggins Institute, University of AucklandAucklandNew Zealand
| | - Chantal Buckley
- Liggins Institute, University of AucklandAucklandNew Zealand
| | - Heather D. Maynard
- Department of Chemistry and Biochemistry and the California NanoSystems InstituteUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Ries J. Langley
- Maurice Wilkins Centre for Molecular BiodiscoveryAucklandNew Zealand
- Department of Molecular Medicine and PathologyUniversity of AucklandAucklandNew Zealand
| | - Jo K. Perry
- Liggins Institute, University of AucklandAucklandNew Zealand
- Maurice Wilkins Centre for Molecular BiodiscoveryAucklandNew Zealand
| |
Collapse
|
5
|
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
|
6
|
Unterberger CJ, McGregor SM, Kopchick JJ, Swanson SM, Marker PC. Mammary Tumor Growth and Proliferation Are Dependent on Growth Hormone in Female SV40 C3(1) T-Antigen Mice. Endocrinology 2022; 164:6767904. [PMID: 36269749 PMCID: PMC9923789 DOI: 10.1210/endocr/bqac174] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 10/14/2022] [Indexed: 01/16/2023]
Abstract
Female SV40 C3(1) T-antigen (C3(1)/TAg) transgenic mice develop mammary tumors that are molecularly similar to human basal-like breast cancers with 100% incidence at 16 weeks of age. To determine the requirement for growth hormone (GH) signaling in these tumors, genetic crosses were used to create cohorts of female mice that were homozygous for a floxed growth hormone receptor (Ghr) gene and carried one copy each of the Rosa-Cre-ERT2 transgene and the C3(1)/TAg transgene (Ghrflox/flox; Rosa-Cre-ERT2; C3(1)/TAg+/0 mice). When the largest mammary tumor reached 200 mm3, mice were treated with tamoxifen to delete Ghr or with vehicle as a control. An additional group of Ghrflox/flox; C3(1)/TAg+/0 mice were also treated with tamoxifen when the largest mammary tumor reached 200 mm3 as a control for the effects of tamoxifen. After 3 weeks, tumors in mice in which Ghr was deleted began to shrink while vehicle and tamoxifen treatment control mouse tumors continued to grow. Pathological analysis of tumors revealed similar growth patterns and varying levels of necrosis throughout all groups. A decrease in cancer cell proliferation in Ghr-/- tumors relative to controls was observed as measured by Ki67 immunohistochemistry labeling index. These data suggest that even established C3(1)/TAg mammary tumors are dependent on the GH/IGF-1 axis.
Collapse
Affiliation(s)
- Christopher J Unterberger
- School of Pharmacy, Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Stephanie M McGregor
- School of Medicine and Public Health, Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53792, USA
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - John J Kopchick
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Steven M Swanson
- School of Pharmacy, Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Paul C Marker
- Correspondence: Paul Marker, PhD, Pharmaceutical Sciences Division, University of Wisconsin—Madison, 777 Highland Ave, Madison, WI 53705, USA.
| |
Collapse
|
7
|
Kaseb AO, Haque A, Vishwamitra D, Hassan MM, Xiao L, George B, Sahu V, Mohamed YI, Carmagnani Pestana R, Lombardo JL, Avritscher R, Yao JC, Wolff RA, Rashid A, Morris JS, Amin HM. Blockade of growth hormone receptor signaling by using pegvisomant: A functional therapeutic strategy in hepatocellular carcinoma. Front Oncol 2022; 12:986305. [PMID: 36276070 PMCID: PMC9582251 DOI: 10.3389/fonc.2022.986305] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 09/07/2022] [Indexed: 11/30/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is an aggressive neoplasm with poor clinical outcome because most patients present at an advanced stage, at which point curative surgical options, such as tumor excision or liver transplantation, are not feasible. Therefore, the majority of HCC patients require systemic therapy. Nonetheless, the currently approved systemic therapies have limited effects, particularly in patients with advanced and resistant disease. Hence, there is a critical need to identify new molecular targets and effective systemic therapies to improve HCC outcome. The liver is a major target of the growth hormone receptor (GHR) signaling, and accumulating evidence suggests that GHR signaling plays an important role in HCC pathogenesis. We tested the hypothesis that GHR could represent a potential therapeutic target in this aggressive neoplasm. We measured GH levels in 767 HCC patients and 200 healthy controls, and then carried out clinicopathological correlation analyses. Moreover, specific inhibition of GHR was performed in vitro using siRNA and pegvisomant (a small peptide that blocks GHR signaling and is currently approved by the FDA to treat acromegaly) and in vivo, also using pegvisomant. GH was significantly elevated in 49.5% of HCC patients, and these patients had a more aggressive disease and poorer clinical outcome (P<0.0001). Blockade of GHR signaling with siRNA or pegvisomant induced substantial inhibitory cellular effects in vitro. In addition, pegvisomant potentiated the effects of sorafenib (P<0.01) and overcame sorafenib resistance (P<0.0001) in vivo. Mechanistically, pegvisomant decreased the phosphorylation of GHR downstream survival proteins including JAK2, STAT3, STAT5, IRS-1, AKT, ERK, and IGF-IR. In two patients with advanced-stage HCC and high GH who developed sorafenib resistance, pegvisomant caused tumor stability. Our data show that GHR signaling represents a novel “druggable” target, and pegvisomant may function as an effective systemic therapy in HCC. Our findings could also lead to testing GHR inhibition in other aggressive cancers.
Collapse
Affiliation(s)
- Ahmed O. Kaseb
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- *Correspondence: Hesham M. Amin, ; Ahmed O. Kaseb,
| | - Abedul Haque
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Deeksha Vishwamitra
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Manal M. Hassan
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Lianchun Xiao
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Bhawana George
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Vishal Sahu
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Yehia I. Mohamed
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Roberto Carmagnani Pestana
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jamie Lynne Lombardo
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Rony Avritscher
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - James C. Yao
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Robert A. Wolff
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Asif Rashid
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jeffrey S. Morris
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Hesham M. Amin
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, United States
- *Correspondence: Hesham M. Amin, ; Ahmed O. Kaseb,
| |
Collapse
|
8
|
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
|
9
|
Basu R, Qian Y, Mathes S, Terry J, Arnett N, Riddell T, Stevens A, Funk K, Bell S, Bokal Z, Batten C, Smith C, Mendez-Gibson I, Duran-Ortiz S, Lach G, Mora-Criollo PA, Kulkarni P, Davis E, Teaford E, Berryman DE, List EO, Neggers S, Kopchick JJ. Growth hormone receptor antagonism downregulates ATP-binding cassette transporters contributing to improved drug efficacy against melanoma and hepatocarcinoma in vivo. Front Oncol 2022; 12:936145. [PMID: 35865483 PMCID: PMC9296106 DOI: 10.3389/fonc.2022.936145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/14/2022] [Indexed: 01/06/2023] Open
Abstract
Knockdown of GH receptor (GHR) in melanoma cells in vitro downregulates ATP-binding cassette-containing (ABC) transporters and sensitizes them to anti-cancer drug treatments. Here we aimed to determine whether a GHR antagonist (GHRA) could control cancer growth by sensitizing tumors to therapy through downregulation of ABC transporters in vivo. We intradermally inoculated Fluc-B16-F10 mouse melanoma cells into GHA mice, transgenic for a GHR antagonist (GHRA), and observed a marked reduction in tumor size, mass and tumoral GH signaling. Moreover, constitutive GHRA production in the transgenic mice significantly improved the response to cisplatin treatment by suppressing expression of multiple ABC transporters and sensitizing the tumors to the drug. We confirmed that presence of a GHRA and not a mere absence of GH is essential for this chemo-sensitizing effect using Fluc-B16-F10 allografts in GH knockout (GHKO) mice, where tumor growth was reduced relative to that in GH-sufficient controls but did not sensitize the tumor to cisplatin. We extended our investigation to hepatocellular carcinoma (HCC) using human HCC cells in vitro and a syngeneic mouse model of HCC with Hepa1-6 allografts in GHA mice. Gene expression analyses and drug-efflux assays confirm that blocking GH significantly suppresses the levels of ABC transporters and improves the efficacy of sorafenib towards almost complete tumor clearance. Human patient data for melanoma and HCC show that GHR RNA levels correlate with ABC transporter expression. Collectively, our results validate in vivo that combination of a GHRA with currently available anti-cancer therapies can be effective in attacking cancer drug resistance.
Collapse
Affiliation(s)
- Reetobrata Basu
- Edison Biotechnology Institute, Ohio University, Athens, OH, United States
| | - Yanrong Qian
- Edison Biotechnology Institute, Ohio University, Athens, OH, United States
| | - Samuel Mathes
- Edison Biotechnology Institute, Ohio University, Athens, OH, United States
| | - Joseph Terry
- Edison Biotechnology Institute, Ohio University, Athens, OH, United States
- Department of Biological Sciences, Ohio University, Athens, OH, United States
| | - Nathan Arnett
- Edison Biotechnology Institute, Ohio University, Athens, OH, United States
- Russ College of Engineering, Ohio University, Athens, OH, United States
| | - Trent Riddell
- Edison Biotechnology Institute, Ohio University, Athens, OH, United States
- Department of Biological Sciences, Ohio University, Athens, OH, United States
| | - Austin Stevens
- Edison Biotechnology Institute, Ohio University, Athens, OH, United States
- Department of Biological Sciences, Ohio University, Athens, OH, United States
| | - Kevin Funk
- Edison Biotechnology Institute, Ohio University, Athens, OH, United States
- Department of Biological Sciences, Ohio University, Athens, OH, United States
- Molecular Cellular Biology Program, Ohio University, Athens, OH, United States
| | - Stephen Bell
- Edison Biotechnology Institute, Ohio University, Athens, OH, United States
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| | - Zac Bokal
- Edison Biotechnology Institute, Ohio University, Athens, OH, United States
| | - Courtney Batten
- Edison Biotechnology Institute, Ohio University, Athens, OH, United States
| | - Cole Smith
- Edison Biotechnology Institute, Ohio University, Athens, OH, United States
| | | | | | - Grace Lach
- Edison Biotechnology Institute, Ohio University, Athens, OH, United States
- Department of Biological Sciences, Ohio University, Athens, OH, United States
| | | | - Prateek Kulkarni
- Edison Biotechnology Institute, Ohio University, Athens, OH, United States
- Department of Biological Sciences, Ohio University, Athens, OH, United States
- Molecular Cellular Biology Program, Ohio University, Athens, OH, United States
| | - Emily Davis
- Edison Biotechnology Institute, Ohio University, Athens, OH, United States
- Department of Biological Sciences, Ohio University, Athens, OH, United States
- Molecular Cellular Biology Program, Ohio University, Athens, OH, United States
| | - Elizabeth Teaford
- Edison Biotechnology Institute, Ohio University, Athens, OH, United States
| | - Darlene E. Berryman
- Edison Biotechnology Institute, Ohio University, Athens, OH, United States
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| | - Edward O. List
- Edison Biotechnology Institute, Ohio University, Athens, OH, United States
| | - Sebastian Neggers
- Edison Biotechnology Institute, Ohio University, Athens, OH, United States
| | - John J. Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, OH, United States
- Molecular Cellular Biology Program, Ohio University, Athens, OH, United States
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
- Translational Biological Sciences Program, Ohio University, Athens, OH, United States
- *Correspondence: John J. Kopchick,
| |
Collapse
|
10
|
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
|
11
|
Unterberger CJ, Maklakova VI, Lazar M, Arneson PD, Mcilwain SJ, Tsourkas PK, Hu R, Kopchick JJ, Swanson SM, Marker PC. GH Action in Prostate Cancer Cells Promotes Proliferation, Limits Apoptosis, and Regulates Cancer-related Gene Expression. Endocrinology 2022; 163:6564019. [PMID: 35383352 PMCID: PMC8995093 DOI: 10.1210/endocr/bqac031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Indexed: 11/19/2022]
Abstract
Previous studies investigating the effects of blocking the growth hormone (GH)/insulin-like growth factor-1 (IGF-1) axis in prostate cancer found no effects of the growth hormone receptor (GHR) antagonist, pegvisomant, on the growth of grafted human prostate cancer cells in vivo. However, human GHR is not activated by mouse GH, so direct actions of GH on prostate cancer cells were not evaluated in this context. The present study addresses the species specificity of GH-GHR activity by investigating GH actions in prostate cancer cell lines derived from a mouse Pten-deletion model. In vitro cell growth was stimulated by GH and reduced by pegvisomant. These in vitro GH effects were mediated at least in part by the activation of JAK2 and STAT5. When Pten-mutant cells were grown as xenografts in mice, pegvisomant treatment dramatically reduced xenograft size, and this was accompanied by decreased proliferation and increased apoptosis. RNA sequencing of xenografts identified 1765 genes upregulated and 953 genes downregulated in response to pegvisomant, including many genes previously implicated as cancer drivers. Further evaluation of a selected subset of these genes via quantitative reverse transcription-polymerase chain reaction determined that some genes exhibited similar regulation by pegvisomant in prostate cancer cells whether treatment was in vivo or in vitro, indicating direct regulation by GH via GHR activation in prostate cancer cells, whereas other genes responded to pegvisomant only in vivo, suggesting indirect regulation by pegvisomant effects on the host endocrine environment. Similar results were observed for a prostate cancer cell line derived from the mouse transgenic adenocarcinoma of the mouse prostate (TRAMP) model.
Collapse
Affiliation(s)
- Christopher J Unterberger
- School of Pharmacy, Pharmaceutical Sciences Division, University of Wisconsin–Madison, Madison, Wisconsin 53705, USA
| | - Vilena I Maklakova
- School of Pharmacy, Pharmaceutical Sciences Division, University of Wisconsin–Madison, Madison, Wisconsin 53705, USA
| | - Michelle Lazar
- School of Pharmacy, Pharmaceutical Sciences Division, University of Wisconsin–Madison, Madison, Wisconsin 53705, USA
| | - Paige D Arneson
- School of Pharmacy, Pharmaceutical Sciences Division, University of Wisconsin–Madison, Madison, Wisconsin 53705, USA
| | - Sean J Mcilwain
- School of Medicine and Public Health, University of Wisconsin–Madison, Madison, Wisconsin 53705, USA
| | - Philippos K Tsourkas
- School of Medicine and Public Health, University of Wisconsin–Madison, Madison, Wisconsin 53705, USA
| | - Rong Hu
- School of Medicine and Public Health, Department of Pathology and Laboratory Medicine, University of Wisconsin–Madison, Madison, Wisconsin 53792, USA
| | - John J Kopchick
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio 45701, USA
| | - Steven M Swanson
- School of Pharmacy, Pharmaceutical Sciences Division, University of Wisconsin–Madison, Madison, Wisconsin 53705, USA
| | - Paul C Marker
- School of Pharmacy, Pharmaceutical Sciences Division, University of Wisconsin–Madison, Madison, Wisconsin 53705, USA
- Correspondence: Paul C. Marker, PhD, Pharmaceutical Sciences Division, University of Wisconsin–Madison, 777 Highland Ave, Madison, WI 53705, USA.
| |
Collapse
|
12
|
Meng Y, Zhou B, Pei Z, Chen Y, Chang D. The nuclear-localized GHR is involved in the cell proliferation of gastric cancer, and pegvisomant may be an important potential drug to inhibit the proliferation of gastric cancer cells. Biochem Cell Biol 2022; 100:125-135. [PMID: 35061546 DOI: 10.1139/bcb-2021-0386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Under normal physiological conditions, growth hormones (GH) play an important role in body growth and metabolism. A recent study showed that GH has important biological effects on gastric cancer (GC) both in vitro and in vivo. However, the biological properties of GH/GHR (GHR, growth hormone receptor) in GC cells have not been fully elucidated. To this end, we systemically studied the biological properties of GH in GC cells and found that GH/GHR was transported into the nuclei of GC cells. Furthermore, we investigated the functions of nuclear GHR and its potential mechanisms of action. We found that nuclear-localized GHR was closely related to the proliferation of GC cells. In addition, we systematically studied the effect of a GHR inhibitor (pegvisomant) on GC in vivo and in vitro, and the results showed that pegvisomant can not only inhibit the proliferation of GC cells but also inhibit the nuclear localization of GHR, suggesting that pegvisomant may be a dual-effect antagonist. Current research indicates that GHR may be a potential target for the treatment of GC.
Collapse
Affiliation(s)
- YuanPu Meng
- Department of Surgical Oncology, Xi'an, Shaanxi, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Bo Zhou
- Department of Gastrointestinal Oncology, the First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471000, China
| | - Zhe Pei
- Department of Gastrointestinal Oncology, the First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471000, China
| | - Ye Chen
- Department of Gastrointestinal Oncology, the First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471000, China
| | - Dongmin Chang
- Department of Surgical Oncology, Xi'an, Shaanxi, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| |
Collapse
|
13
|
Salvadori G, Mirisola MG, Longo VD. Intermittent and Periodic Fasting, Hormones, and Cancer Prevention. Cancers (Basel) 2021; 13:cancers13184587. [PMID: 34572814 PMCID: PMC8472354 DOI: 10.3390/cancers13184587] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 09/09/2021] [Indexed: 12/25/2022] Open
Abstract
The restriction of proteins, amino acids or sugars can have profound effects on the levels of hormones and factors including growth hormone, IGF-1 and insulin. In turn, these can regulate intracellular signaling pathways as well as cellular damage and aging, but also multisystem regeneration. Both intermittent (IF) and periodic fasting (PF) have been shown to have both acute and long-term effects on these hormones. Here, we review the effects of nutrients and fasting on hormones and genes established to affect aging and cancer. We describe the link between dietary interventions and genetic pathways affecting the levels of these hormones and focus on the mechanisms responsible for the cancer preventive effects. We propose that IF and PF can reduce tumor incidence both by delaying aging and preventing DNA damage and immunosenescence and also by killing damaged, pre-cancerous and cancer cells.
Collapse
Affiliation(s)
- Giulia Salvadori
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy;
- IFOM, FIRC Institute of Molecular Oncology, 20139 Milan, Italy
| | - Mario Giuseppe Mirisola
- Department of Surgical, Oncological, and Oral Sciences, University of Palermo, 90127 Palermo, Italy;
| | - Valter D. Longo
- IFOM, FIRC Institute of Molecular Oncology, 20139 Milan, Italy
- Department of Biological Sciences, Longevity Institute, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
- Correspondence:
| |
Collapse
|
14
|
Cheng Y, Li W, Gui R, Wang C, Song J, Wang Z, Wang X, Shen Y, Wang Z, Hao L. Dual Characters of GH-IGF1 Signaling Pathways in Radiotherapy and Post-radiotherapy Repair of Cancers. Front Cell Dev Biol 2021; 9:671247. [PMID: 34178997 PMCID: PMC8220142 DOI: 10.3389/fcell.2021.671247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 05/17/2021] [Indexed: 12/02/2022] Open
Abstract
Radiotherapy remains one of the most important cancer treatment modalities. In the course of radiotherapy for tumor treatment, the incidental irradiation of adjacent tissues could not be completely avoided. DNA damage is one of the main factors of cell death caused by ionizing radiation, including single-strand (SSBs) and double-strand breaks (DSBs). The growth hormone-Insulin-like growth factor 1 (GH-IGF1) axis plays numerous roles in various systems by promoting cell proliferation and inhibiting apoptosis, supporting its effects in inducing the development of multiple cancers. Meanwhile, the GH-IGF1 signaling involved in DNA damage response (DDR) and DNA damage repair determines the radio-resistance of cancer cells subjected to radiotherapy and repair of adjacent tissues damaged by radiotherapy. In the present review, we firstly summarized the studies on GH-IGF1 signaling in the development of cancers. Then we discussed the adverse effect of GH-IGF1 signaling in radiotherapy to cancer cells and the favorable impact of GH-IGF1 signaling on radiation damage repair to adjacent tissues after irradiation. This review further summarized recent advances on research into the molecular mechanism of GH-IGF1 signaling pathway in these effects, expecting to specify the dual characters of GH-IGF1 signaling pathways in radiotherapy and post-radiotherapy repair of cancers, subsequently providing theoretical basis of their roles in increasing radiation sensitivity during cancer radiotherapy and repairing damage after radiotherapy.
Collapse
Affiliation(s)
- Yunyun Cheng
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China
| | - Wanqiao Li
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China
| | - Ruirui Gui
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China
| | - Chunli Wang
- College of Animal Science, Jilin University, Changchun, China
| | - Jie Song
- College of Animal Science, Jilin University, Changchun, China
| | - Zhaoguo Wang
- College of Animal Science, Jilin University, Changchun, China
| | - Xue Wang
- The First Hospital of Jilin University, Changchun, China
| | - Yannan Shen
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China
| | - Zhicheng Wang
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China
| | - Linlin Hao
- College of Animal Science, Jilin University, Changchun, China
| |
Collapse
|
15
|
Lantvit DD, Unterberger CJ, Lazar M, Arneson PD, Longhurst CA, Swanson SM, Marker PC. Mammary Tumors Growing in the Absence of Growth Hormone Are More Sensitive to Doxorubicin Than Wild-Type Tumors. Endocrinology 2021; 162:bqab013. [PMID: 33475144 PMCID: PMC7881836 DOI: 10.1210/endocr/bqab013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Indexed: 12/18/2022]
Abstract
Previously, we reported that N-methyl-N-nitrosourea (MNU)-induced mammary tumors could be established in mutant spontaneous dwarf rats (SDRs), which lack endogenous growth hormone (GH) by supplementing with exogenous GH, and almost all such tumors regressed upon GH withdrawal. When the highly inbred SDR line was outcrossed to wild-type (WT) Sprague-Dawley rats, MNU-induced mammary tumors could still be established in resulting outbred SDRs by supplementing with exogenous GH. However, unlike tumors in inbred SDRs, 65% of mammary tumors established in outbred SDRs continued growth after GH withdrawal. We further tested whether these tumors were more sensitive to doxorubicin than their WT counterparts. To accomplish this, MNU-induced mammary tumors were established in WT rats and in SDRs supplemented with exogenous GH. Once mammary tumors reached 1 cm3 in size, exogenous GH was withdrawn from SDRs, and the subset that harbored tumors that continued or resumed growth in the absence of GH were selected for doxorubicin treatment. Doxorubicin was then administered in 6 injections over 2 weeks at 2.5 mg/kg or 1.25 mg/kg for both the WT and SDR groups. The SDR mammary tumors that had been growing in the absence of GH regressed at both doxorubicin doses while WT tumors continued to grow robustly. The regression of SDR mammary tumors treated with 1.25 mg/kg doxorubicin was accompanied by reduced proliferation and dramatically higher apoptosis relative to the WT mammary tumors treated with 1.25 mg/kg doxorubicin. These data suggest that downregulating GH signaling may decrease the doxorubicin dose necessary to effectively treat breast cancer.
Collapse
Affiliation(s)
- Daniel D Lantvit
- College of Pharmacy, Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Christopher J Unterberger
- School of Pharmacy, Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI, USA
| | - Michelle Lazar
- School of Pharmacy, Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI, USA
| | - Paige D Arneson
- School of Pharmacy, Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI, USA
| | - Colin A Longhurst
- School of Medicine and Public Health, Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - Steven M Swanson
- College of Pharmacy, Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, USA
- School of Pharmacy, Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI, USA
| | - Paul C Marker
- School of Pharmacy, Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
16
|
Chhabra Y, Lee CMM, Müller AF, Brooks AJ. GHR signalling: Receptor activation and degradation mechanisms. Mol Cell Endocrinol 2021; 520:111075. [PMID: 33181235 DOI: 10.1016/j.mce.2020.111075] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 11/01/2020] [Accepted: 11/03/2020] [Indexed: 12/21/2022]
Abstract
Growth hormone (GH) actions via initiating cell signalling through the GH receptor (GHR) are important for many physiological processes, in addition to its well-known role in regulating growth. The activation of JAK-STAT signalling by GH is well characterized, however knowledge on GH activation of SRC family kinases (SFKs) is still limited. In this review we summarise the collective knowledge on the activation, regulation, and downstream signalling of GHR. We highlight studies on GH activation of SFKs and the important outcome of this signalling pathway with a focus on the different degradation mechanisms that can regulate GHR availability since this is an area that warrants further study considering its role in tumour progression.
Collapse
Affiliation(s)
- Yash Chhabra
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, 4102, Australia; Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21231, USA
| | - Christine M M Lee
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - Alexandra Franziska Müller
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - Andrew J Brooks
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, 4102, Australia.
| |
Collapse
|
17
|
Wang Y, Langley RJ, Tamshen K, Harms J, Middleditch MJ, Maynard HD, Jamieson SMF, Perry JK. Enhanced Bioactivity of a Human GHR Antagonist Generated by Solid-Phase Site-Specific PEGylation. Biomacromolecules 2020; 22:299-308. [PMID: 33295758 DOI: 10.1021/acs.biomac.0c01105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Growth hormone (GH) has been implicated in cancer progression andis a potential target for anticancer therapy. Currently, pegvisomant is the only GH receptor (GHR) antagonist approved for clinical use. Pegvisomant is a mutated GH molecule (B2036) which is PEGylated on amine groups to extend serum half-life. However, PEGylation significantly reduces the bioactivity of the antagonist in mice. To improve bioactivity, we generated a series of B2036 conjugates with the site-specific attachment of 20, 30, or 40 kDa methoxyPEG maleimide (mPEG maleimide) by introduction of a cysteine residue at amino acid 144 (S144C). Recombinant B2036-S144C was expressed in Escherichia coli, purified, and then PEGylated using cysteine-specific conjugation chemistry. To avoid issues with dimerization due to the introduced cysteine, B2036-S144C was PEGylated while immobilized on an Ni-nitrilotriacetic (Ni-NTA) acid column, which effectively reduced disulfide-mediated dimer formation and allowed efficient conjugation to mPEG maleimide. Following PEGylation, the IC50 values for the 20, 30, and 40 kDa mPEG maleimide B2036-S144C conjugates were 66.2 ± 3.8, 106.1 ± 7.1, and 127.4 ± 3.6 nM, respectively. The circulating half-life of the 40 kDa mPEG conjugate was 58.3 h in mice. Subcutaneous administration of the 40 kDa mPEG conjugate (10 mg/kg/day) reduced serum insulin-like growth factor I (IGF-I) concentrations by 50.6%. This in vivo reduction in serum IGF-I was at a considerably lower dose compared to the higher doses required to observe comparable activity in studies with pegvisomant. In conclusion, we have generated a novel PEGylated GHR antagonist by the solid-phase site-specific attachment of mPEG maleimide at an introduced cysteine residue, which effectively reduces serum IGF-I in vivo.
Collapse
Affiliation(s)
- Yue Wang
- Liggins Institute, University of Auckland, Auckland 1023, New Zealand
| | - Ries J Langley
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland 1023, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, Auckland 1023, New Zealand
| | - Kyle Tamshen
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles 90095-1569, California, United States
| | - Julia Harms
- Liggins Institute, University of Auckland, Auckland 1023, New Zealand.,Auckland Cancer Society Research Centre, University of Auckland, Auckland 1023, New Zealand
| | - Martin J Middleditch
- School of Biological Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Heather D Maynard
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles 90095-1569, California, United States.,California NanoSystems Institute, University of California, Los Angeles, Los Angeles 90095-1569, California, United States
| | - Stephen M F Jamieson
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland 1023, New Zealand.,Auckland Cancer Society Research Centre, University of Auckland, Auckland 1023, New Zealand.,Department of Pharmacology and Clinical Pharmacology, University of Auckland, Auckland 1023, New Zealand
| | - Jo K Perry
- Liggins Institute, University of Auckland, Auckland 1023, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, Auckland 1023, New Zealand
| |
Collapse
|
18
|
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
|
19
|
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
|
20
|
Tamshen K, Wang Y, Jamieson SM, Perry JK, Maynard HD. Genetic Code Expansion Enables Site-Specific PEGylation of a Human Growth Hormone Receptor Antagonist through Click Chemistry. Bioconjug Chem 2020; 31:2179-2190. [PMID: 32786367 PMCID: PMC8291075 DOI: 10.1021/acs.bioconjchem.0c00365] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Regulation of human growth hormone (GH) signaling has important applications in the remediation of several diseases including acromegaly and cancer. Growth hormone receptor (GHR) antagonists currently provide the most effective means for suppression of GH signaling. However, these small 22 kDa recombinantly engineered GH analogues exhibit short plasma circulation times. To improve clinical viability, between four and six molecules of 5 kDa poly(ethylene glycol) (PEG) are nonspecifically conjugated to the nine amines of the GHR antagonist designated as B2036 in the FDA-approved therapeutic pegvisomant. PEGylation increases the molecular weight of B2036 and considerably extends its circulation time, but also dramatically reduces its bioactivity, contributing to high dosing requirements and increased cost. As an alternative to nonspecific PEGylation, we report the use of genetic code expansion technology to site-specifically incorporate the unnatural amino acid propargyl tyrosine (pglY) into B2036 with the goal of producing site-specific protein-polymer conjugates. Substitution of tyrosine 35 with pglY yielded a B2036 variant containing an alkyne functional group without compromising bioactivity, as verified by a cellular assay. Subsequent conjugation of 5, 10, and 20 kDa azide-containing PEGs via the copper-catalyzed click reaction yielded high purity, site-specific conjugates with >89% conjugation efficiencies. Site-specific attachment of PEG to B2036 is associated with substantially improved in vitro bioactivity values compared to pegvisomant, with an inverse relationship between polymer size and activity observed. Notably, the B2036-20 kDa PEG conjugate has a molecular weight comparable to pegvisomant, while exhibiting a 12.5 fold improvement in half-maximal inhibitory concentration in GHR-expressing Ba/F3 cells (103.3 nM vs 1289 nM). We expect that this straightforward route to achieve site-specific GHR antagonists will be useful for GH signal regulation.
Collapse
Affiliation(s)
- Kyle Tamshen
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095-1569, United States
| | - Yue Wang
- Liggins Institute, University of Auckland, Auckland 1203, New Zealand
| | - Stephen M.F. Jamieson
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland 1023, New Zealand
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Jo K. Perry
- Liggins Institute, University of Auckland, Auckland 1203, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland 1023, New Zealand
| | - Heather D. Maynard
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095-1569, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095-1569, United States
- Department of Bioengineering, University of California, Los Angeles, California 90095-1569, United States
| |
Collapse
|
21
|
Leone S, Chiavaroli A, Recinella L, Di Valerio V, Veschi S, Gasparo I, Bitto A, Ferrante C, Orlando G, Salvatori R, Brunetti L. Growth hormone-releasing hormone (GHRH) deficiency promotes inflammation-associated carcinogenesis. Pharmacol Res 2019; 152:104614. [PMID: 31874252 DOI: 10.1016/j.phrs.2019.104614] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/25/2019] [Accepted: 12/18/2019] [Indexed: 12/13/2022]
Abstract
The somatotropic axis, in addition to its well-known metabolic and endocrine effects, plays a pivotal role in modulation of inflammation. Moreover, growth hormone (GH)-releasing hormone (GHRH) has been involved in the development of various human tumors. In this work we aimed to investigate the consequences of GHRH deficiency on the development of inflammation-associated colon carcinogenesis in a mouse model of isolated GH deficiency due to generalized ablation of the GHRH gene [GHRH knock out (GHRHKO)]. Homozygous GHRHKO (-/-) male mice and wild type (C57/BL6, +/+) male mice as control group, were used. After azoxymetane (AOM)/dextran sodium sulfate (DSS) treatment -/- mice displayed higher Disease Activity Index (DAI) score, and more marked weight loss compared to +/+ animals. Additionally, -/- mice showed a significant increase in total tumors, in particular of large size predominantly localized in distal colon. In colonic tissue of AOM/DSS-treated -/- mice we found the presence of invasive adenocarcinomas, dysplasia and colitis with mucosal ulceration. Conversely, AOM/DSS-treated +/+ mice showed only presence of adenomas, without invasion of sub-mucosa. Treatment with AOM/DSS significantly increased prostaglandin (PG)E2 and 8-iso-PGF2α levels along with cyclooxygenase-2 (COX-2), tumor necrosis factor (TNF)-α, nuclear factor kappa B (NF-kB) and inducible nitric oxide synthase (iNOS) gene expression, in colon specimens. The degree of increase of all these parameters was more markedly in -/- than +/+ mice. In conclusion, generalized GHRH ablation increases colon carcinogenesis responsiveness in male mice. Whether this results from lack of GH or GHRH remains to be established.
Collapse
Affiliation(s)
- Sheila Leone
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Annalisa Chiavaroli
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Lucia Recinella
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy.
| | - Valentina Di Valerio
- Department of Medicine and Ageing Sciences, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Serena Veschi
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Irene Gasparo
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Alessandra Bitto
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Claudio Ferrante
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Giustino Orlando
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Roberto Salvatori
- Division of Endocrinology Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Luigi Brunetti
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
22
|
Lan H, Li W, Li R, Zheng X, Luo G. Endocytosis and Degradation of Pegvisomant and a Potential New Mechanism That Inhibits the Nuclear Translocation of GHR. J Clin Endocrinol Metab 2019; 104:1887-1899. [PMID: 30602026 DOI: 10.1210/jc.2018-02063] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 12/26/2018] [Indexed: 02/13/2023]
Abstract
CONTEXT Pegvisomant, a growth hormone receptor (GHR) antagonist, is a well-known drug that was designed to treat acromegaly. However, recent studies have indicated that the GHR is a "moonlighting" protein that may exhibit dual functions based on its localization in the plasma membrane and nucleus. In light of this finding, we explored whether pegvisomant is a potential "moonlighting" GHR antagonist. In addition, the mechanisms of the endocytosis, postendocytic sorting, and degradation of pegvisomant are not fully understood. OBJECTIVE This study investigated whether pegvisomant is a "moonlighting" antagonist and explored the mechanisms of the endocytosis, postendocytic sorting, and degradation of pegvisomant. METHODS Indirect immunofluorescence and Western blot coupled with pharmacological inhibitors and gene silencing (small interfering RNA) were used to explore the mechanisms of the endocytosis, postendocytic sorting, and degradation of pegvisomant. Western blot, immunohistochemistry, and indirect immunofluorescence coupled with subcellular fractionation analysis were used to determine the effect of pegvisomant on GHR's nuclear localization in vitro and in vivo. RESULTS Here, we show that the endocytosis of pegvisomant is mainly mediated though the clathrin pathway. Further study of the postendocytic sorting of pegvisomant shows that pegvisomant enters into different types of endosomes under GHR mediation. In addition, GHR is slightly downregulated by pegvisomant; further study indicates that proteasomes and lysosomes may cooperate to regulate pegvisomant/GHR degradation. Most importantly, we show that pegvisomant inhibits the nuclear localization of GHR. CONCLUSION Our study showed that pegvisomant is a "moonlighting" antagonist. In addition, we revealed the mechanisms of the endocytosis, postendocytic sorting, and degradation of pegvisomant.
Collapse
Affiliation(s)
- Hainan Lan
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Wei Li
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Ruonan Li
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Xin Zheng
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Gan Luo
- Department of Anesthesiology and Intensive Care, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| |
Collapse
|
23
|
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: 85] [Impact Index Per Article: 14.2] [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
|
24
|
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: 225] [Impact Index Per Article: 37.5] [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
|
25
|
Arumugam A, Subramani R, Nandy SB, Terreros D, Dwivedi AK, Saltzstein E, Lakshmanaswamy R. Silencing growth hormone receptor inhibits estrogen receptor negative breast cancer through ATP-binding cassette sub-family G member 2. Exp Mol Med 2019; 51:1-13. [PMID: 30617282 PMCID: PMC6323053 DOI: 10.1038/s12276-018-0197-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 09/11/2018] [Accepted: 10/02/2018] [Indexed: 12/14/2022] Open
Abstract
Growth hormone receptor (GHR) plays a vital role in breast cancer chemoresistance and metastasis but the mechanism is not fully understood. We determined if GHR could be a potential therapeutic target for estrogen receptor negative (ER-ve) breast cancer, which are highly chemoresistant and metastatic. GHR was stably knocked down in ER-ve breast cancer cells and its effect on cell proliferation, metastatic behavior, and chemosensitivity to docetaxel (DT) was assessed. Microarray analysis was performed to identify potential GHR downstream targets involved in chemoresistance. GHR and ATP-binding cassette sub-family G member 2 (ABCG2) overexpression and knockdown studies were performed to investigate the mechanism of GHR-induced chemoresistance. Patient-derived xenografts was used to study the effect of GHR and ABCG2. Immunohistochemical data was used to determine the correlation between GHR, pAKT, pmTOR, and ABCG2 expressions. GHR silencing drastically reduced the chemoresistant and metastatic behavior of ER-ve breast cancer cells and also inhibited AKT/mTOR pathway. In contrast, activation, or overexpression of GHR increased chemoresistance and metastasis by increasing the expression and promoter activity, of ABCG2. Inhibition of JAK2/STAT5 signaling repressed GHR-induced ABCG2 promoter activity and expression. Further, ABCG2 knockdown significantly increased the chemosensitivity. Finally, patient-derived xenograft studies revealed the role of GHR in chemoresistance. Overall, these findings demonstrate that targeting GHR could be a novel therapeutic approach to overcome chemoresistance and associated metastasis in aggressive ER-ve breast cancers.
Collapse
Affiliation(s)
- Arunkumar Arumugam
- Center of Emphasis in Cancer Research, Texas Tech University Health Sciences Center El Paso, El Paso, TX, 79905, USA
| | - Ramadevi Subramani
- Center of Emphasis in Cancer Research, Texas Tech University Health Sciences Center El Paso, El Paso, TX, 79905, USA
| | - Sushmita Bose Nandy
- Center of Emphasis in Cancer Research, Texas Tech University Health Sciences Center El Paso, El Paso, TX, 79905, USA
| | - Daniel Terreros
- Research Core Laboratory, Texas Tech University Health Sciences Center El Paso, El Paso, TX, 79905, USA
| | - Alok Kumar Dwivedi
- Division of Biostatistics & Epidemiology, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, 79905, USA
| | - Edward Saltzstein
- University Breast Care Center, Texas Tech University Health Sciences Center El Paso, El Paso, TX, 79905, USA
| | - Rajkumar Lakshmanaswamy
- Center of Emphasis in Cancer Research, Texas Tech University Health Sciences Center El Paso, El Paso, TX, 79905, USA. .,Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, El Paso, TX, 79905, USA.
| |
Collapse
|
26
|
Abstract
Early preclinical and population data suggested a role for the type I insulin-like growth factor receptor (IGF1R) in the regulation of breast cancer growth and survival. To target this pathway, multiple monoclonal antibodies and tyrosine kinase inhibitors were developed and tested in clinical trials. While some of the early clinical trials suggested a benefit for these drugs, none of the attempts showed improved outcomes when compared to conventional therapy. This failure of the IGF1R inhibitors was pronounced in breast cancer; multiple trials testing IGF1R inhibition in estrogen receptor-positive breast cancer were conducted, none showed benefit. This review will evaluate the rationale for IGF1R inhibition, discuss results of the clinical trials and suggest a path forward.
Collapse
Affiliation(s)
- Douglas Yee
- Masonic Cancer CenterUniversity of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
27
|
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
|
28
|
Girgert R, Emons G, Gründker C. Inhibition of growth hormone receptor by Somavert reduces expression of GPER and prevents growth stimulation of triple-negative breast cancer by 17β-estradiol. Oncol Lett 2018; 15:9559-9566. [PMID: 29805678 DOI: 10.3892/ol.2018.8521] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 03/02/2018] [Indexed: 12/12/2022] Open
Abstract
Currently, conventional chemotherapy is the only treatment option for triple-negative breast cancers (TNBC) due to a lack of a unique target. In TNBC, a high expression of the membrane bound G protein-coupled estrogen receptor (GPER), correlates with a worse outcome. There is a potential for an association between growth hormone receptor (GHR) and GPER expression. To confirm this hypothesis, GHR was inhibited in TNBC cells with Somavert, and GPER expression levels, and the effect on signal transduction and proliferation induction in TNBC cells were analyzed. Proliferation of TNBC cells was measured using an Alamar-blue assay. Expression of GPER and activation of c-src and epidermal growth factor receptor (EGFR) by 17β-estradiol was analyzed by western blotting. Induction of c-fos, cyclin D1 and aromatase expression was determined by reverse transcription-semi-quantitative polymerase chain reaction. The expression of GPER was concentration- and time-dependently reduced by Somavert down to 46±7% (P<0.01) of the control. Furthermore, 17β-estradiol significantly increased the cell number of HCC1806 cells to 128±14% (P<0.05), and that of MDA-MB-453 cells to 115±3%. This increase in cell number was reduced to 103±11% in HCC1806 cells in which GPER expression was downregulated by Somavert, and to 102±3% in MDA-MB-453 cells. In addition, 17β-estradiol increased the activation of c-src in HCC1806 cells by 1.8-fold, and Somavert reduced p-src to 63% of control. In MDA-MB-453 cells src phosphorylation increased by 7-fold upon stimulation with estradiol, but after treatment with Somavert only a 4-fold increase was observed. Phosphorylation of EGFR was increased by 2.2-fold of control in HCC1806 cells by 17β-estradiol, and by 1.4-fold in MDA-MD-453 cells. Somavert completely prevented this activation. Induction of cyclin D1 and aromatase expression by 17β-estradiol was also prevented by Somavert. Somavert reduces GPER expression in triple negative breast cancer cells. Treatment with Somavert prevents induction of genes regulating proliferation by 17β-estradiol. Inhibition of GPER expression is a promising therapeutic intervention for TNBC.
Collapse
Affiliation(s)
- Rainer Girgert
- Department of Obstetrics and Gynecology, University Medical Center Goettingen, D-37075 Goettingen, Germany
| | - Günter Emons
- Department of Obstetrics and Gynecology, University Medical Center Goettingen, D-37075 Goettingen, Germany
| | - Carsten Gründker
- Department of Obstetrics and Gynecology, University Medical Center Goettingen, D-37075 Goettingen, Germany
| |
Collapse
|
29
|
Abdelmegeed SM, Mohammed S. Canine mammary tumors as a model for human disease. Oncol Lett 2018; 15:8195-8205. [PMID: 29928319 PMCID: PMC6004712 DOI: 10.3892/ol.2018.8411] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 02/12/2018] [Indexed: 12/13/2022] Open
Abstract
Animal models for examining human breast cancer (HBC) carcinogenesis have been extensively studied and proposed. With the recent advent of immunotherapy, significant attention has been focused on the dog as a model for human cancer. Dogs develop mammary tumors and other cancer types spontaneously with an intact immune system, which exhibit a number of clinical and molecular similarities to HBC. In addition to the spontaneous tumor presentation, the clinical similarities between human and canine mammary tumors (CMT) include the age at onset, hormonal etiology and course of the diseases. Furthermore, factors that affect the disease outcome, including tumor size, stage and lymph node invasion, are similar in HBC and CMT. Similarly, the molecular characteristics of steroid receptor, epidermal growth factor, proliferation marker, metalloproteinase and cyclooxygenase expression, and the mutation of the p53 tumor suppressor gene in CMT, mimic HBC. Furthermore, ductal carcinomas in situ in human and canine mammary glands are particularly similar in their pathological, molecular and visual characteristics. These CMT characteristics and their similarities to HBC indicate that the dog could be an excellent model for the study of human disease. These similarities are discussed in detail in the present review, and are compared with the in vitro and other in vivo animal models available.
Collapse
Affiliation(s)
- Somaia M Abdelmegeed
- Department of Comparative Pathobiology, Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - Sulma Mohammed
- Department of Comparative Pathobiology, Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
30
|
Chhabra Y, Wong HY, Nikolajsen LF, Steinocher H, Papadopulos A, Tunny KA, Meunier FA, Smith AG, Kragelund BB, Brooks AJ, Waters MJ. A growth hormone receptor SNP promotes lung cancer by impairment of SOCS2-mediated degradation. Oncogene 2018; 37:489-501. [PMID: 28967904 PMCID: PMC5799715 DOI: 10.1038/onc.2017.352] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 08/13/2017] [Accepted: 08/16/2017] [Indexed: 02/07/2023]
Abstract
Both humans and mice lacking functional growth hormone (GH) receptors are known to be resistant to cancer. Further, autocrine GH has been reported to act as a cancer promoter. Here we present the first example of a variant of the GH receptor (GHR) associated with cancer promotion, in this case lung cancer. We show that the GHRP495T variant located in the receptor intracellular domain is able to prolong the GH signal in vitro using stably expressing mouse pro-B-cell and human lung cell lines. This is relevant because GH secretion is pulsatile, and extending the signal duration makes it resemble autocrine GH action. Signal duration for the activated GHR is primarily controlled by suppressor of cytokine signalling 2 (SOCS2), the substrate recognition component of the E3 protein ligase responsible for ubiquitinylation and degradation of the GHR. SOCS2 is induced by a GH pulse and we show that SOCS2 binding to the GHR is impaired by a threonine substitution at Pro 495. This results in decreased internalisation and degradation of the receptor evident in TIRF microscopy and by measurement of mature (surface) receptor expression. Mutational analysis showed that the residue at position 495 impairs SOCS2 binding only when a threonine is present, consistent with interference with the adjacent Thr494. The latter is key for SOCS2 binding, together with nearby Tyr487, which must be phosphorylated for SOCS2 binding. We also undertook nuclear magnetic resonance spectroscopy approach for structural comparison of the SOCS2 binding scaffold Ile455-Ser588, and concluded that this single substitution has altered the structure of the SOCS2 binding site. Importantly, we find that lung BEAS-2B cells expressing GHRP495T display increased expression of transcripts associated with tumour proliferation, epithelial-mesenchymal transition and metastases (TWIST1, SNAI2, EGFR, MYC and CCND1) at 2 h after a GH pulse. This is consistent with prolonged GH signalling acting to promote cancer progression in lung cancer.
Collapse
Affiliation(s)
- Y Chhabra
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - H Y Wong
- University of Queensland Centre for Clinical Research, The University of Queensland, Herston, Queensland, Australia
| | - L F Nikolajsen
- Structural Biology and NMR Laboratory (SBiNLab), Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - H Steinocher
- Structural Biology and NMR Laboratory (SBiNLab), Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - A Papadopulos
- The Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - K A Tunny
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - F A Meunier
- The Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - A G Smith
- School of Biomedical Sciences, Institute of Health and Biomedical Innovation at the Translational Research Institute, Queensland University of Technology, Woolloongabba, Queensland, Australia
| | - B B Kragelund
- Structural Biology and NMR Laboratory (SBiNLab), Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - A J Brooks
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - M J Waters
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
31
|
Kong X, Wu W, Yuan Y, Pandey V, Wu Z, Lu X, Zhang W, Chen Y, Wu M, Zhang M, Li G, Tan S, Qian P, Perry JK, Lobie PE, Zhu T. Human growth hormone and human prolactin function as autocrine/paracrine promoters of progression of hepatocellular carcinoma. Oncotarget 2017; 7:29465-79. [PMID: 27102295 PMCID: PMC5045410 DOI: 10.18632/oncotarget.8781] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 03/24/2016] [Indexed: 11/25/2022] Open
Abstract
The death rates of hepatocellular carcinoma (HCC) are extremely high due to the paucity of therapeutic options. Animal models and anecdotal clinical evidence indicate a potential role of hGH and hPRL in HCC. However, the prognostic relevance and the functional role of tumor expression of these hormones in human HCC are not defined. Herein, we analyzed the mRNA and protein expression of hGH and hPRL in histopathological samples of non-neoplastic liver and HCC by in situ hybridization, PCR and immunohistochemistry techniques. Increased mRNA and protein expression of both hormones was observed in HCC compared with non-neoplastic liver tissues. hGH expression was significantly associated with tumor size and tumor grade. No significant association was observed between the expression of hPRL and any histopathological features. Amplification of both hGH and hPRL genes in HCC was observed when compared to non-neoplastic tissue. Expression of both hGH and hPRL was associated with worse relapse-free and overall survival in HCC patients. In vitro and in vivo functional assays performed with HCC cell lines demonstrated that autocrine expression of hGH or hPRL in HCC cells increased STAT3 activation, oncogenicity and tumor growth while functional antagonism with hGH-G120R significantly reduced these parameters. Hence, tumor expression of hGH/hPRL is associated with a worse survival outcome for patients with HCC and hGH/hPRL function as autocrine/paracrine promoters of HCC progression.
Collapse
Affiliation(s)
- Xiangjun Kong
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui, China
| | - Wenyong Wu
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yan Yuan
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui, China
| | - Vijay Pandey
- Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, Singapore
| | - Zhengsheng Wu
- Department of Pathology, Anhui Medical University, Hefei, Anhui, China
| | - Xuefei Lu
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui, China
| | - Weijie Zhang
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui, China
| | - Yijun Chen
- Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, Singapore
| | - Mingming Wu
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui, China
| | - Min Zhang
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui, China
| | - Gaopeng Li
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui, China
| | - Sheng Tan
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui, China
| | - Pengxu Qian
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui, China
| | - Jo K Perry
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Peter E Lobie
- Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, Singapore.,National University Cancer Institute of Singapore, National University Health System, Singapore
| | - Tao Zhu
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui, China
| |
Collapse
|
32
|
Christopoulos PF, Corthay A, Koutsilieris M. Aiming for the Insulin-like Growth Factor-1 system in breast cancer therapeutics. Cancer Treat Rev 2017; 63:79-95. [PMID: 29253837 DOI: 10.1016/j.ctrv.2017.11.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 11/29/2017] [Accepted: 11/30/2017] [Indexed: 12/23/2022]
Abstract
Despite the major discoveries occurred in oncology the recent years, breast malignancies remain one of the most common causes of cancer-related deaths for women in developed countries. Development of HER2-targeting drugs has been considered a breakthrough in anti-cancer approaches and alluded to the potential of targeting growth factors in breast cancer (BrCa) therapeutics. More than twenty-five years have passed since the Insulin-like Growth Factor-1 (IGF-1) system was initially recognized as a potential target candidate in BrCa therapy. To date, a growing body of studies have implicated the IGF-1 signaling with the BrCa biology. Despite the promising experimental evidence, the impression from clinical trials is rather disappointing. Several reasons may account for this and the last word regarding the efficacy of this system as a target candidate in BrCa therapeutics is probably not written yet. Herein, we provide the theoretical basis, as well as, a comprehensive overview of the current literature, regarding the different strategies targeting the various components of the IGF-1/IGF-1R axis in several pathophysiological aspects of BrCa, including the tumor micro-environment and cancer stemness. In addition, we review the rationale for targeting the IGF-1 system in the different BrCa molecular subtypes and in treatment resistant breast tumors with a focus on both the molecular mechanisms and on the clinical perspectives of such approaches in specific population subgroups. We also discuss the future challenges, as well as, the development of novel molecules and strategies targeting the system and suggest potential improvements in the field.
Collapse
Affiliation(s)
- Panagiotis F Christopoulos
- Department of Experimental Physiology, Medical School, National & Kapodistrian University of Athens, Athens, Greece; Tumor Immunology Lab, Department of Pathology, Rikshospitalet, Oslo University Hospital and University of Oslo, Oslo, Norway; Department of Medical Biology, Faculty of Health Sciences, UiT the Arctic University of Norway, Tromsø, Norway.
| | - Alexandre Corthay
- Tumor Immunology Lab, Department of Pathology, Rikshospitalet, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Michael Koutsilieris
- Department of Experimental Physiology, Medical School, National & Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
33
|
Cui Y, Sun X, Jin L, Yu G, Li Q, Gao X, Ao J, Wang C. MiR-139 suppresses β-casein synthesis and proliferation in bovine mammary epithelial cells by targeting the GHR and IGF1R signaling pathways. BMC Vet Res 2017; 13:350. [PMID: 29178948 PMCID: PMC5702184 DOI: 10.1186/s12917-017-1267-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 11/09/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND MicroRNAs have important roles in many biological processes. However, the role of miR-139 in healthy mammary gland remains unclear. The objective of this study was to investigate the effects of miR-139 on lactation in dairy cows. RESULTS Here, we found that miR-139 was down-regulated in mid-lactation dairy cow mammary tissues compared with mid-pregnancy tissues. Then, we prioritized two of potential target genes of miR-139 in cow, growth hormone receptor (GHR) and type I insulin-like growth factor receptor (IGF1R) for further functional studies based on their roles in lactation processes. Dual luciferase reporter assays validated direct binding of miR-139 to the 3'- untranslated region (UTR) of GHR and IGF1R. Moreover, over-expression or silencing of miR-139 affected mRNA levels of GHR and IGF1R in cultured bovine mammary epithelial cells (BMECs). Furthermore, over-expression of miR-139 decreased protein levels of β-casein, proliferation in mammary epithelial cell, and the protein levels of IGF1R and key members of the GHR or IGF1R pathways as well, whereas silencing miR-139 produced the opposite result. Among these signal molecules, signal transducer and activator of transcription-5 (STAT5), protein kinase B (also known as AKT1), mammalian target of rapamycin (mTOR), and p70-S6 Kinase (p70S6K) are involed in β-casein synthesis, and Cyclin D1 is involved in cell proliferation. In addition, silencing GHR decreased protein levels of β-casein, IGF1R, and key members of the IGF1R pathway, whereas co-silencing miR-139 and GHR rescued the expression of GHR and reversed GHR silencing effects. CONCLUSIONS Our results demonstrate that GHR and IGF1R are target genes of miR-139 in dairy cow. MiR-139 suppresses β-casein synthesis and proliferation in BMECs by targeting the GHR and IGF1R signaling pathways.
Collapse
Affiliation(s)
- Yingjun Cui
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, Heilongjiang, 150030, People's Republic of China.,College of Life Science, Northeast Agricultural University, Harbin, Heilongjiang, 150030, People's Republic of China
| | - Xia Sun
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, Heilongjiang, 150030, People's Republic of China
| | - Lianfeng Jin
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, Heilongjiang, 150030, People's Republic of China
| | - Guangpu Yu
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, Heilongjiang, 150030, People's Republic of China
| | - Qingzhang Li
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, Heilongjiang, 150030, People's Republic of China
| | - Xuejun Gao
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, Heilongjiang, 150030, People's Republic of China
| | - Jinxia Ao
- College of Life Science, Northeast Agricultural University, Harbin, Heilongjiang, 150030, People's Republic of China
| | - Chunmei Wang
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, Heilongjiang, 150030, People's Republic of China.
| |
Collapse
|
34
|
Recouvreux MV, Wu JB, Gao AC, Zonis S, Chesnokova V, Bhowmick N, Chung LW, Melmed S. Androgen Receptor Regulation of Local Growth Hormone in Prostate Cancer Cells. Endocrinology 2017; 158:2255-2268. [PMID: 28444169 PMCID: PMC5505214 DOI: 10.1210/en.2016-1939] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 04/18/2017] [Indexed: 01/22/2023]
Abstract
Prostate cancer (PCa) growth is mainly driven by androgen receptor (AR), and tumors that initially respond to androgen deprivation therapy (ADT) or AR inhibition usually relapse into a more aggressive, castration-resistant PCa (CRPC) stage. Circulating growth hormone (GH) has a permissive role in PCa development in animal models and in human PCa xenograft growth. As GH and GH receptor (GHR) are both expressed in PCa cells, we assessed whether prostatic GH production is linked to AR activity and whether GH contributes to the castration-resistant phenotype. Using online datasets, we found that GH is highly expressed in human CRPC. We observed increased GH expression in castration-resistant C4-2 compared with castration-sensitive LNCaP cells as well as in enzalutamide (MDV3100)-resistant (MDVR) C4-2B (C4-2B MDVR) cells compared with parental C4-2B. We describe a negative regulation of locally produced GH by androgens/AR in PCa cells following treatment with AR agonists (R1881) and antagonists (enzalutamide, bicalutamide). We also show that GH enhances invasive behavior of CRPC 22Rv1 cells, as reflected by increased migration, invasion, and anchorage-independent growth, as well as expression of matrix metalloproteases. Moreover, GH induces expression of the AR splice variant 7, which correlates with antiandrogen resistance, and also induces insulinlike growth factor 1, which is implicated in PCa progression and ligand-independent AR activation. In contrast, blockade of GH action with the GHR antagonist pegvisomant reverses these effects both in vitro and in vivo. GH induction following ADT or AR inhibition may contribute to CRPC progression by bypassing androgen growth requirements.
Collapse
Affiliation(s)
| | - J. Boyang Wu
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, 90048
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, Washington, 99202
| | - Allen C. Gao
- Department of Urology, University of California at Davis, Sacramento, California, 95817
| | - Svetlana Zonis
- Pituitary Center, Cedars Sinai Medical Center, Los Angeles, California, 90048
| | - Vera Chesnokova
- Pituitary Center, Cedars Sinai Medical Center, Los Angeles, California, 90048
| | - Neil Bhowmick
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, 90048
| | - Leland W. Chung
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, 90048
| | - Shlomo Melmed
- Pituitary Center, Cedars Sinai Medical Center, Los Angeles, California, 90048
| |
Collapse
|
35
|
Subramani R, Nandy SB, Pedroza DA, Lakshmanaswamy R. Role of Growth Hormone in Breast Cancer. Endocrinology 2017; 158:1543-1555. [PMID: 28379395 DOI: 10.1210/en.2016-1928] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 03/28/2017] [Indexed: 12/19/2022]
Abstract
Breast cancer is one of the most common cancers diagnosed in women. Approximately two-thirds of all breast cancers diagnosed are classified as hormone dependent, which indicates that hormones are the key factors that drive the growth of these breast cancers. Ovarian and pituitary hormones play a major role in the growth and development of normal mammary glands and breast cancer. In particular, the effect of the ovarian hormone estrogen has received much attention in regard to breast cancer. Pituitary hormones prolactin and growth hormone have also been associated with breast cancer. Although the role of these pituitary hormones in breast cancers has been studied, it has not been investigated extensively. In this review, we attempt to compile basic information from most of the currently available literature to understand and demonstrate the significance of growth hormone in breast cancer. Based on the available literature, it is clear that growth hormone plays a significant role in the development, progression, and metastasis of breast cancer by influencing tumor angiogenesis, stemness, and chemoresistance.
Collapse
Affiliation(s)
- Ramadevi Subramani
- Center of Emphasis in Cancer Research, Department of Biomedical Sciences MSB1, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, Texas 79905
| | - Sushmita B Nandy
- Center of Emphasis in Cancer Research, Department of Biomedical Sciences MSB1, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, Texas 79905
| | - Diego A Pedroza
- Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, El Paso, Texas 79905
| | - Rajkumar Lakshmanaswamy
- Center of Emphasis in Cancer Research, Department of Biomedical Sciences MSB1, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, Texas 79905
- Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, El Paso, Texas 79905
| |
Collapse
|
36
|
Minutolo F. Immune by Heart: Unexpected Observations Inspiring Perspective Therapeutic/Preventive Strategies against Cancer. ChemMedChem 2016; 11:2560-2566. [PMID: 27922212 DOI: 10.1002/cmdc.201600483] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 10/20/2016] [Indexed: 12/18/2022]
Abstract
We often overlook ordinary events that take place right in front of our eyes. However, such events might be inspiring sources of revolutionary scientific discovery. Simple observations, such as those regarding the substantial immunity of the heart to cancer or the noninvasive behavior of plant tumors, are just the tips of icebergs hiding profound mechanistic causes that deserve deeper investigation. Several current or unprecedented approaches aimed at improving both the prevention and treatment of tumors are discussed on these bases herein. This viewpoint is not intended to give definitive answers, but rather to provide cues for discussion and motivation to pursue unexplored and accessible strategies to fight cancer.
Collapse
Affiliation(s)
- Filippo Minutolo
- Dipartimento di Farmacia, Università di Pisa, Via Bonanno 33, 56126, Pisa, Italy
| |
Collapse
|
37
|
Evans A, Jamieson SM, Liu DX, Wilson WR, Perry JK. Growth hormone receptor antagonism suppresses tumour regrowth after radiotherapy in an endometrial cancer xenograft model. Cancer Lett 2016; 379:117-23. [DOI: 10.1016/j.canlet.2016.05.031] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 05/23/2016] [Accepted: 05/25/2016] [Indexed: 11/25/2022]
|
38
|
Bak MJ, Das Gupta S, Wahler J, Suh N. Role of dietary bioactive natural products in estrogen receptor-positive breast cancer. Semin Cancer Biol 2016; 40-41:170-191. [PMID: 27016037 DOI: 10.1016/j.semcancer.2016.03.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 03/16/2016] [Accepted: 03/20/2016] [Indexed: 12/20/2022]
Abstract
Estrogen receptor (ER)-positive breast cancer, including luminal-A and -B, is the most common type of breast cancer. Extended exposure to estrogen is associated with an increased risk of breast cancer. Both ER-dependent and ER-independent mechanisms have been implicated in estrogen-mediated carcinogenesis. The ER-dependent pathway involves cell growth and proliferation triggered by the binding of estrogen to the ER. The ER-independent mechanisms depend on the metabolism of estrogen to generate genotoxic metabolites, free radicals and reactive oxygen species to induce breast cancer. A better understanding of the mechanisms that drive ER-positive breast cancer will help optimize targeted approaches to prevent or treat breast cancer. A growing emphasis is being placed on alternative medicine and dietary approaches toward the prevention and treatment of breast cancer. Many natural products and bioactive compounds found in foods have been shown to inhibit breast carcinogenesis via inhibition of estrogen induced oxidative stress as well as ER signaling. This review summarizes the role of bioactive natural products that are involved in the prevention and treatment of estrogen-related and ER-positive breast cancer.
Collapse
Affiliation(s)
- Min Ji Bak
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Soumyasri Das Gupta
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Joseph Wahler
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Nanjoo Suh
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA; Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA.
| |
Collapse
|
39
|
Beckwith H, Yee D. Insulin-like growth factors, insulin, and growth hormone signaling in breast cancer: implications for targeted therapy. Endocr Pract 2014; 20:1214-21. [PMID: 25297664 DOI: 10.4158/ep14208.ra] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE In recent decades, multiple therapeutics targeting the estrogen and human epidermal growth factor-2 (HER2) receptors have been approved for the treatment of breast cancer. METHODS This review discusses a number of growth factor pathways that have been implicated in resistance to both anti-estrogen and HER2-targeted therapies. The association between growth factors and breast cancer is well established. Over decades, numerous laboratories have studied the link between insulin-like growth factor (IGF), insulin, and growth hormone (GH) to the development and progression of breast cancer. RESULTS Although preclinical data demonstrates that blockade of these receptors inhibits breast cancer growth, progression, and drug resistance, therapies targeting the IGF, insulin, and GH receptors (GHRs) have not been successful in producing significant increases in progression-free, disease-free, or overall survival for patients with breast cancer. The failure to demonstrate a benefit of growth factor blockade in clinical trials can be attributed to redundancy in IGF, insulin, and GHR signaling pathways. All 3 receptors are able to activate oncogenic phosphoinositide-3 kinase (PI3K) and mitogen-activated protein kinase (MAPK) pathways. CONCLUSION Consequently, multitargeted blockade of growth factor receptors and their common downstream kinases will be necessary for the successful treatment of breast cancer.
Collapse
Affiliation(s)
- Heather Beckwith
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Douglas Yee
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota Department of Medicine, University of Minnesota, Minneapolis, Minnesota Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
40
|
Bielohuby M, Zarkesh-Esfahani SH, Manolopoulou J, Wirthgen E, Walpurgis K, Toghiany Khorasgani M, Aghili ZS, Wilkinson IR, Hoeflich A, Thevis M, Ross RJ, Bidlingmaier M. Validation of serum IGF-I as a biomarker to monitor the bioactivity of exogenous growth hormone agonists and antagonists in rabbits. Dis Model Mech 2014; 7:1263-73. [PMID: 25239917 PMCID: PMC4213730 DOI: 10.1242/dmm.016519] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The development of new growth hormone (GH) agonists and growth hormone antagonists (GHAs) requires animal models for pre-clinical testing. Ideally, the effects of treatment are monitored using the same pharmacodynamic marker that is later used in clinical practice. However, intact rodents are of limited value for this purpose because serum IGF-I, the most sensitive pharmacodynamic marker for the action of GH in humans, shows no response to treatment with recombinant human GH and there is little evidence for the effects of GHAs, except when administered at very high doses or when overexpressed. As an alternative, more suitable model, we explored pharmacodynamic markers of GH action in intact rabbits. We performed the first validation of an IGF-I assay for the analysis of rabbit serum and tested precision, sensitivity, linearity and recovery using an automated human IGF-I assay (IDS-iSYS). Furthermore, IGF-I was measured in rabbits of different strains, age groups and sexes, and we monitored IGF-I response to treatment with recombinant human GH or the GHA Pegvisomant. For a subset of samples, we used LC-MS/MS to measure IGF-I, and quantitative western ligand blot to analyze IGF-binding proteins (IGFBPs). Although recovery of recombinant rabbit IGF-I was only 50% in the human IGF-I assay, our results show that the sensitivity, precision (1.7–3.3% coefficient of variation) and linearity (90.4–105.6%) were excellent in rabbit samples. As expected, sex, age and genetic background were major determinants of IGF-I concentration in rabbits. IGF-I and IGFBP-2 levels increased after single and multiple injections of recombinant human GH (IGF-I: 286±22 versus 434±26 ng/ml; P<0.01) and were highly correlated (P<0.0001). Treatment with the GHA lowered IGF-I levels from the fourth injection onwards (P<0.01). In summary, we demonstrated that the IDS-iSYS IGF-I immunoassay can be used in rabbits. Similar to rodents, rabbits display variations in IGF-I depending on sex, age and genetic background. Unlike in rodents, the IGF-I response to treatment with recombinant human GH or a GHA closely mimics the pharmacodynamics seen in humans, suggesting that rabbits are a suitable new model to test human GH agonists and antagonists.
Collapse
Affiliation(s)
- Maximilian Bielohuby
- Endocrine Research Unit, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians University, 80336 Munich, Germany
| | - Sayyed Hamid Zarkesh-Esfahani
- Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan 81746-73695, Iran. Department of Immunology, Medical School, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran. The Department of Human Metabolism, The University of Sheffield, Sheffield S10 2JF, UK
| | | | - Elisa Wirthgen
- Ligandis GbR, Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Katja Walpurgis
- German Sport University Cologne, Institute of Biochemisty/Center for Preventive Doping Research, 50933 Cologne, Germany
| | | | - Zahra Sadat Aghili
- Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan 81746-73695, Iran
| | - Ian Robert Wilkinson
- The Department of Human Metabolism, The University of Sheffield, Sheffield S10 2JF, UK
| | - Andreas Hoeflich
- Leibniz Institute for Farm Animal Biology (FBN), Institute of Genome Biology, 18196 Dummerstorf, Germany
| | - Mario Thevis
- German Sport University Cologne, Institute of Biochemisty/Center for Preventive Doping Research, 50933 Cologne, Germany
| | - Richard J Ross
- The Department of Human Metabolism, The University of Sheffield, Sheffield S10 2JF, UK
| | - Martin Bidlingmaier
- Endocrine Research Unit, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians University, 80336 Munich, Germany.
| |
Collapse
|
41
|
Pivonello C, De Martino MC, Negri M, Cuomo G, Cariati F, Izzo F, Colao A, Pivonello R. The GH-IGF-SST system in hepatocellular carcinoma: biological and molecular pathogenetic mechanisms and therapeutic targets. Infect Agent Cancer 2014; 9:27. [PMID: 25225571 PMCID: PMC4164328 DOI: 10.1186/1750-9378-9-27] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 06/23/2014] [Indexed: 12/15/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common malignancy worldwide. Different signalling pathways have been identified to be implicated in the pathogenesis of HCC; among these, GH, IGF and somatostatin (SST) pathways have emerged as some of the major pathways implicated in the development of HCC. Physiologically, GH-IGF-SST system plays a crucial role in liver growth and development since GH induces IGF1 and IGF2 secretion and the expression of their receptors, involved in hepatocytes cell proliferation, differentiation and metabolism. On the other hand, somatostatin receptors (SSTRs) are exclusively present on the biliary tract. Importantly, the GH-IGF-SST system components have been indicated as regulators of hepatocarcinogenesis. Reduction of GH binding affinity to GH receptor, decreased serum IGF1 and increased serum IGF2 production, overexpression of IGF1 receptor, loss of function of IGF2 receptor and appearance of SSTRs are frequently observed in human HCC. In particular, recently, many studies have evaluated the correlation between increased levels of IGF1 receptors and liver diseases and the oncogenic role of IGF2 and its involvement in angiogenesis, migration and, consequently, in tumour progression. SST directly or indirectly influences tumour growth and development through the inhibition of cell proliferation and secretion and induction of apoptosis, even though SST role in hepatocarcinogenesis is still opened to argument. This review addresses the present evidences suggesting a role of the GH-IGF-SST system in the development and progression of HCC, and describes the therapeutic perspectives, based on the targeting of GH-IGF-SST system, which have been hypothesised and experimented in HCC.
Collapse
Affiliation(s)
- Claudia Pivonello
- Dipartimento di Medicina Clinica e Chirurgia, Università Federico II di Napoli, Via Sergio Pansini, 5, Naples 80131, Italy
| | - Maria Cristina De Martino
- Dipartimento di Medicina Clinica e Chirurgia, Università Federico II di Napoli, Via Sergio Pansini, 5, Naples 80131, Italy
| | - Mariarosaria Negri
- Dipartimento di Medicina Clinica e Chirurgia, Università Federico II di Napoli, Via Sergio Pansini, 5, Naples 80131, Italy
| | | | - Federica Cariati
- Dipartimento di Medicina Clinica e Chirurgia, Università Federico II di Napoli, Via Sergio Pansini, 5, Naples 80131, Italy
| | - Francesco Izzo
- National Cancer Institute G Pascale Foundation, Naples, Italy
| | - Annamaria Colao
- Dipartimento di Medicina Clinica e Chirurgia, Università Federico II di Napoli, Via Sergio Pansini, 5, Naples 80131, Italy
| | - Rosario Pivonello
- Dipartimento di Medicina Clinica e Chirurgia, Università Federico II di Napoli, Via Sergio Pansini, 5, Naples 80131, Italy
| |
Collapse
|
42
|
Lombardi S, Honeth G, Ginestier C, Shinomiya I, Marlow R, Buchupalli B, Gazinska P, Brown J, Catchpole S, Liu S, Barkan A, Wicha M, Purushotham A, Burchell J, Pinder S, Dontu G. Growth hormone is secreted by normal breast epithelium upon progesterone stimulation and increases proliferation of stem/progenitor cells. Stem Cell Reports 2014; 2:780-93. [PMID: 24936466 PMCID: PMC4050343 DOI: 10.1016/j.stemcr.2014.05.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 05/06/2014] [Accepted: 05/07/2014] [Indexed: 01/16/2023] Open
Abstract
Using in vitro and in vivo experimental systems and in situ analysis, we show that growth hormone (GH) is secreted locally by normal human mammary epithelial cells upon progesterone stimulation. GH increases proliferation of a subset of cells that express growth hormone receptor (GHR) and have functional properties of stem and early progenitor cells. In 72% of ductal carcinoma in situ lesions, an expansion of the cell population that expresses GHR was observed, suggesting that GH signaling may contribute to breast cancer development.
Collapse
Affiliation(s)
- Sara Lombardi
- Research Oncology, King's College London, London SE1 9RT, UK
| | | | - Christophe Ginestier
- Centre de Recherche et Cancérologie, Marseille, Inserm, CRCM, U1068, France ; Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Rebecca Marlow
- Research Oncology, King's College London, London SE1 9RT, UK
| | | | | | - John Brown
- Research Oncology, King's College London, London SE1 9RT, UK
| | | | - Suling Liu
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ariel Barkan
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Max Wicha
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Joy Burchell
- Research Oncology, King's College London, London SE1 9RT, UK
| | - Sarah Pinder
- Research Oncology, King's College London, London SE1 9RT, UK
| | - Gabriela Dontu
- Research Oncology, King's College London, London SE1 9RT, UK
| |
Collapse
|
43
|
Kopchick JJ, List EO, Kelder B, Gosney ES, Berryman DE. Evaluation of growth hormone (GH) action in mice: discovery of GH receptor antagonists and clinical indications. Mol Cell Endocrinol 2014; 386:34-45. [PMID: 24035867 PMCID: PMC3943600 DOI: 10.1016/j.mce.2013.09.004] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 08/29/2013] [Accepted: 09/03/2013] [Indexed: 11/28/2022]
Abstract
The discovery of a growth hormone receptor antagonist (GHA) was initially established via expression of mutated GH genes in transgenic mice. Following this discovery, development of the compound resulted in a drug termed pegvisomant, which has been approved for use in patients with acromegaly. Pegvisomant treatment in a dose dependent manner results in normalization of IGF-1 levels in most patients. Thus, it is a very efficacious and safe drug. Since the GH/IGF-1 axis has been implicated in the progression of several types of cancers, many have suggested the use of pegvisomant as an anti-cancer therapeutic. In this manuscript, we will review the use of mouse strains that possess elevated or depressed levels of GH action for unraveling many of GH actions. Additionally, we will describe experiments in which the GHA was discovered, review results of pegvisomant's preclinical and clinical trials, and provide data suggesting pegvisomant's therapeutic value in selected types of cancer.
Collapse
Affiliation(s)
- John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, United States; Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, United States.
| | - Edward O List
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, United States; Department of Specialty Medicine, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, United States
| | - Bruce Kelder
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, United States; Department of Pediatrics, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, United States
| | - Elahu S Gosney
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, United States
| | - Darlene E Berryman
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, United States; Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, United States; School of Applied Health Sciences and Wellness, Ohio University, Athens, OH 45701, United States
| |
Collapse
|
44
|
Zhou D, Yang J, Huang WD, Wang J, Zhang Q. siRNA-targeted inhibition of growth hormone receptor in human colon cancer SW480 cells. World J Gastroenterol 2013; 19:8108-8113. [PMID: 24307807 PMCID: PMC3848161 DOI: 10.3748/wjg.v19.i44.8108] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 10/22/2013] [Indexed: 02/06/2023] Open
Abstract
AIM: To determine the effects of RNAi-mediated inhibition of the growth hormone receptor (GHR) gene on tumors and colon cancer cells in vivo.
METHODS: Construction of a eukaryotic vector for human GHR expression, the pcDNA™6.2-GW/EmGFP-small interfering RNAs (siRNAs)-GHR plasmid, was used to inhibit GHR expression. Thirty-six BALB/c nude mice were randomly divided into groups and treated with normal saline (NS), recombinant plasmid (G2), growth hormone (GH), 5-fluorouracil (FU), G2+FU or G2+FU+GH. Each nude mouse was subcutaneously inoculated with 1×107 human colon cancer SW480 cells; the nude mice were weighed before inoculation and on the 2nd, 5th, 8th, 11th, 14th and 17th day after inoculation. All nude mice were sacrificed after 17 d. Each subcutaneous tumor was removed and studied. Tumor volume was measured on the 5th, 8th, 11th, 14th and 17th day after inoculation. The expression of GHR protein in the tumor tissue was detected by Western blotting analysis, and the differences in GHR mRNA expression in the tumor tissue were detected by real-time quantitative reverse transcription-polymerase chain reaction.
RESULTS: Compared to the control group, the weights of the inoculated nude mice on the 17th day after inoculation were: G2: 21.60 ± 0.71 g, GH: 21.64 ± 0.45 g, FU: 18.94 ± 0.47 g, FU+G2: 19.40 ± 0.60 g, G2+FU+GH: 21.04 ± 0.78 g vs NS: 20.68 ± 0.66 g, P < 0.05; the tumor volumes after the subcutaneous inoculation were: G2: 9.71 ± 3.82 mm3, FU: 11.54 ± 2.42 mm3, FU+G2: 11.42 ± 1.11 mm3, G2+FU+GH: 10.47 ± 1.02 mm3vs NS: 116.81 ± 10.61 mm3, P < 0.05. Compared to the GH group, the tumor volumes were significantly decreased in the experimental groups. The GHR protein expression (G2: 0.39 ± 0.02, FU: 0.40 ± 0.02, FU+G2: 0.38 ± 0.01, G2+FU+GH: 0.39 ± 0.01 vs NS: 0.94 ± 0.02, P < 0.05) and the GHR mRNA expression (G2: 14.12 ± 0.10, FU: 15.15 ± 0.44, FU+G2: 16.46 ± 0.27, G2+FU+GH: 15.37 ± 0.57 vs NS: 12.63 ± 0.14, P < 0.05) were significantly decreased and increased, respectively, in the experimental groups.
CONCLUSION: Inhibition of GHR in human colon cancer SW480 cells resulted in anti-tumor effects in nude mice.
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW Animals born with a deficiency in the cell surface receptor for growth hormone (GH) have a significantly reduced risk of developing cancer. Conversely, increased expression levels of GH and the GH receptor (GHR) are detectable in a variety of different human cancers. Here we discuss recent literature contributing to our understanding of the field. RECENT FINDINGS In addition to animal evidence, studies of individuals with Laron syndrome suggest that congenital GHR deficiency may also protect humans against cancer. GH expression in certain malignancies is correlated with clinicohistopathological parameters and may contribute the therapeutic resistance. Other recent studies have identified novel aspects of the GH signal transduction pathway, including receptor crosstalk and the involvement of microRNA in endocrine regulation of GH. SUMMARY Substantial evidence suggests the GH/insulin-like growth factor-1 axis initiates and promotes progression of cancer. However, important questions remain unanswered regarding the therapeutic utility of GH or GHR antagonism in cancer. Further clinical studies regarding the clinical association of GH expression with human malignancies and translational studies investigating GHR antagonism in animal models of human cancer are critical.
Collapse
Affiliation(s)
- Jo K Perry
- Liggins Institute, University of Auckland, Auckland, New Zealand.
| | | | | | | | | |
Collapse
|
46
|
Finley SD, Dhar M, Popel AS. Compartment model predicts VEGF secretion and investigates the effects of VEGF trap in tumor-bearing mice. Front Oncol 2013; 3:196. [PMID: 23908970 PMCID: PMC3727077 DOI: 10.3389/fonc.2013.00196] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 07/13/2013] [Indexed: 12/19/2022] Open
Abstract
Angiogenesis, the formation of new blood vessels from existing vasculature, is important in tumor growth and metastasis. A key regulator of angiogenesis is vascular endothelial growth factor (VEGF), which has been targeted in numerous anti-angiogenic therapies aimed at inhibiting tumor angiogenesis. Systems biology approaches, including computational modeling, are useful for understanding this complex biological process and can aid in the development of novel and effective therapeutics that target the VEGF family of proteins and receptors. We have developed a computational model of VEGF transport and kinetics in the tumor-bearing mouse, which includes three-compartments: normal tissue, blood, and tumor. The model simulates human tumor xenografts and includes human (VEGF121 and VEGF165) and mouse (VEGF120 and VEGF164) isoforms. The model incorporates molecular interactions between these VEGF isoforms and receptors (VEGFR1 and VEGFR2), as well as co-receptors (NRP1 and NRP2). We also include important soluble factors: soluble VEGFR1 (sFlt-1) and α-2-macroglobulin. The model accounts for transport via macromolecular transendothelial permeability, lymphatic flow, and plasma clearance. We have fit the model to available in vivo experimental data on the plasma concentration of free VEGF Trap and VEGF Trap bound to mouse and human VEGF in order to estimate the rates at which parenchymal cells (myocytes and tumor cells) and endothelial cells secrete VEGF. Interestingly, the predicted tumor VEGF secretion rates are significantly lower (0.007-0.023 molecules/cell/s, depending on the tumor microenvironment) than most reported in vitro measurements (0.03-2.65 molecules/cell/s). The optimized model is used to investigate the interstitial and plasma VEGF concentrations and the effect of the VEGF-neutralizing agent, VEGF Trap (aflibercept). This work complements experimental studies performed in mice and provides a framework with which to examine the effects of anti-VEGF agents, aiding in the optimization of such anti-angiogenic therapeutics as well as analysis of clinical data. The model predictions also have implications for biomarker discovery with anti-angiogenic therapies.
Collapse
Affiliation(s)
- Stacey D Finley
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine , Baltimore, MD , USA
| | | | | |
Collapse
|
47
|
Novosyadlyy R, Leroith D. Insulin-like growth factors and insulin: at the crossroad between tumor development and longevity. J Gerontol A Biol Sci Med Sci 2012; 67:640-51. [PMID: 22421704 DOI: 10.1093/gerona/gls065] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Numerous lines of evidence indicate that insulin-like growth factor signaling plays an important role in the regulation of life span and tumor development. In the present paper, the role of individual components of insulin-like growth factor signaling in aging and tumor development has been extensively analyzed. The molecular mechanisms underlying aging and tumor development are frequently overlapping. Although the link between reduced insulin-like growth factor signaling and suppressed tumor growth and development is well established, it remains unclear whether extended life span results from direct suppression of insulin-like growth factor signaling or this effect is caused by indirect mechanisms such as improved insulin sensitivity.
Collapse
Affiliation(s)
- Ruslan Novosyadlyy
- Department of Cell Biology, Imclone Systems, a wholly owned subsidiary of Eli Lilly & Co, New York, USA
| | | |
Collapse
|
48
|
Huderson BP, Velayudhan BT, Pearson RE, Ellis SE, Akers RM. Effect of exogenous somatotropin in Holstein calves on mammary gland composition and proliferation. J Dairy Sci 2011; 94:5005-16. [PMID: 21943751 DOI: 10.3168/jds.2011-4229] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Accepted: 06/17/2011] [Indexed: 11/19/2022]
Abstract
Pubertal mammary gland growth and development are hormonally regulated, but the details are poorly understood in calves. Our purpose was to evaluate the effects of exogenous growth hormone (GH) on the biochemical composition of the prepubertal mammary gland, mRNA expression of selected genes, and histological characteristics of the developing parenchyma (PAR). In this experiment, 19 calves (7 ± 4 d of age) were randomly assigned to 1 of 2 treatments: bovine somatotropin (bST, 500 mg; n = 10) or placebo (Sal; 0.9% saline; n = 9). Animals were treated every 3 wk beginning on d 23. Calves were assigned to an early (65 d; tissue harvested after 2 treatment injections) or late collection time (107 d; tissue harvested after 4 treatment injections). Calves were fed milk replacer and calf starter for 8 wk and starter and hay thereafter. Parenchyma and mammary fat pad (MFP) from one udder half were harvested for analysis of protein, lipid, and DNA. Additional tissues were preserved for histological analysis or snap-frozen for quantitative real-time PCR. Somatotropin treatment did not significantly alter the mass of PAR or MFP or the general pattern of development of epithelial structures. Significant increases were observed in protein/100 kg of body weight (BW), total protein, DNA concentration, DNA/100 kg of BW, and total DNA in 107-d calves, and a significant treatment by day interaction was observed for DNA and lipid concentrations in PAR. In MFP, a significant decrease was observed in protein/100 kg of BW in bST-treated calves and in total MFP protein in 65-d calves. A treatment by day interaction was found for total protein, DNA, and protein/100 kg of BW. In PAR, relative expression of ATPase-binding cassette 3 and growth hormone receptor were reduced by bST and both were lower in 107-d-harvest calves. Epithelial cell retention of bromodeoxyuridine (BrdU; possible indicator of stem-like cells) was greatest in 65-d bST-treated calves, and a significant time of sampling response and treatment × time interaction were observed. Expression of the proliferation marker protein Ki67 was numerically higher in bST-treated calves but the difference was nonsignificant. Retention of the BrdU label was reduced in 107-d calves. Exogenous growth hormone given to calves may affect mammary tissue composition and epithelial cell gene expression in subtle ways but exogenous supplementation with bST alone is not likely to alter overall development patterns or affect the mass of mammary parenchymal tissue. Whether such subtle changes have an effect on subsequent development or function is unknown.
Collapse
Affiliation(s)
- B P Huderson
- Department of Dairy Science, Virginia Polytechnic Institute and State University, Blacksburg 24061, USA
| | | | | | | | | |
Collapse
|
49
|
Ozkan EE. Plasma and tissue insulin-like growth factor-I receptor (IGF-IR) as a prognostic marker for prostate cancer and anti-IGF-IR agents as novel therapeutic strategy for refractory cases: a review. Mol Cell Endocrinol 2011; 344:1-24. [PMID: 21782884 DOI: 10.1016/j.mce.2011.07.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2011] [Accepted: 07/01/2011] [Indexed: 12/13/2022]
Abstract
Cancer database analysis indicates that prostate cancer is one of the most seen cancers in men meanwhile composing the leading cause of morbidity and mortality among developed countries. Current available therapies are surgery, radiotherapy and androgene ablation for prostate carcinoma. The response rate is as high nearly 90% however, most of these recur or become refractory and androgene independent (AI). Therefore recent studies intensified on molecular factors playing role on development of prostate carcinoma and novel treatment strategies targetting these factors and their receptors. Insulin-like growth factor-I (IGF-I) and its primary receptor insulin-like growth factor receptor-I (IGF-IR) are among these factors. Biologic functions and role in malign progression are primarily achieved via IGF-IR which is a type 2 tyrosine kinase receptor. IGF-IR plays an important role in mitogenesis, angiogenesis, transformation, apoptosis and cell motility. It also generates intensive proliferative signals leading to carcinogenesis in prostate tissue. So IGF-IR and its associated signalling system have provoked considerable interest over recent years as a novel therapeutic target in cancer. In this paper it is aimed to sum up the lately published literature searching the relation of IGF-IR and prostate cancer in terms of incidence, pathologic features, and prognosis. This is followed by a discussion of the different possible targets within the IGF-1R system, and drugs developed to interact at each target. A systems-based approach is then used to review the in vitro and in vivo data in the published literature of the following compounds targeting IGF-1R components using specific examples: growth hormone releasing hormone antagonists (e.g. JV-1-38), growth hormone receptor antagonists (e.g. pegvisomant), IGF-1R antibodies (e.g. CP-751,871, AVE1642/EM164, IMC-A12, SCH-717454, BIIB022, AMG 479, MK-0646/h7C10), and IGF-1R tyrosine kinase inhibitors (e.g. BMS-536942, BMS-554417, NVP-AEW541, NVP-ADW742, AG1024, potent quinolinyl-derived imidazo (1,5-a)pyrazine PQIP, picropodophyllin PPP, nordihydroguaiaretic acid Insm-18/NDGA). And the other end point is to yield an overview on the recent progress about usage of this receptor as a novel anticancer agent of targeted therapies in treatment of prostate carcinoma.
Collapse
Affiliation(s)
- Emine Elif Ozkan
- OSM Middle East Health Center, Department of Radiation Oncology, Sanliurfa 63000, Turkey.
| |
Collapse
|
50
|
Dearth RK, Kuiatse I, Wang YF, Liao L, Hilsenbeck SG, Brown PH, Xu J, Lee AV. A moderate elevation of circulating levels of IGF-I does not alter ErbB2 induced mammary tumorigenesis. BMC Cancer 2011; 11:377. [PMID: 21867536 PMCID: PMC3189189 DOI: 10.1186/1471-2407-11-377] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Accepted: 08/25/2011] [Indexed: 11/10/2022] Open
Abstract
Background Epidemiological evidence suggests that moderately elevated levels of circulating insulin-like growth factor-I (IGF-I) are associated with increased risk of breast cancer in women. How circulating IGF-I may promote breast cancer incidence is unknown, however, increased IGF-I signaling is linked to trastuzumab resistance in ErbB2 positive breast cancer. Few models have directly examined the effect of moderately high levels of circulating IGF-I on breast cancer initiation and progression. The purpose of this study was to assess the ability of circulating IGF-I to independently initiate mammary tumorigenesis and/or accelerate the progression of ErbB2 mediated mammary tumor growth. Methods We crossed heterozygous TTR-IGF-I mice with heterozygous MMTV-ErbB2 mice to generate 4 different genotypes: TTR-IGF-I/MMTV-ErbB2 (bigenic), TTR-IGF-I only, MMTV-ErbB2 only, and wild type (wt). Virgin females were palpated twice a week and harvested when tumors reached 1000 mm3. For study of normal development, blood and tissue were harvested at 4, 6 and 9 weeks of age in TTR-IGF-I and wt mice. Results TTR-IGF-I and TTR-IGF-I/ErbB2 bigenic mice showed a moderate 35% increase in circulating total IGF-I compared to ErbB2 and wt control mice. Elevation of circulating IGF-I had no effect upon pubertal mammary gland development. The transgenic increase in IGF-I alone wasn't sufficient to initiate mammary tumorigenesis. Elevated circulating IGF-I had no effect upon ErbB2-induced mammary tumorigenesis or metastasis, with median time to tumor formation being 30 wks and 33 wks in TTR-IGF-I/ErbB2 bigenic and ErbB2 mice respectively (p = 0.65). Levels of IGF-I in lysates from ErbB2/TTR-IGF-I tumors compared to ErbB2 was elevated in a similar manner to the circulating IGF-I, however, there was no effect on the rate of tumor growth (p = 0.23). There were no morphological differences in tumor type (solid adenocarcinomas) between bigenic and ErbB2 mammary glands. Conclusion Using the first transgenic animal model to elevate circulating levels of IGF-I to those comparable to women at increased risk of breast cancer, we showed that moderately high levels of systemic IGF-I have no effect on pubertal mammary gland development, initiating mammary tumorigenesis or promoting ErbB2 driven mammary carcinogenesis. Our work suggests that ErbB2-induced mammary tumorigenesis is independent of the normal variation in circulating levels of IGF-I.
Collapse
Affiliation(s)
- Robert K Dearth
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|