1
|
Hawazie A, Druce M. Breast Cancer Risk and Management in the Endocrine Clinic: A Comprehensive Review. Clin Endocrinol (Oxf) 2025. [PMID: 39905814 DOI: 10.1111/cen.15209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 12/30/2024] [Accepted: 01/19/2025] [Indexed: 02/06/2025]
Abstract
OBJECTIVE This review seeks to provide endocrine clinicians with a comprehensive analysis of breast cancer risk, diagnostic modalities and management strategies in women with endocrine disorders, with particular emphasis on the influence of metabolic factors such as diabetes and obesity, and the role of Menopausal Hormone Therapy (MHT). DESIGN The review examines a spectrum of endocrine disorders commonly encountered in clinical practice, including Multiple Endocrine Neoplasia Types 1 (MEN1), 2 (MEN2) and 4 (MEN4), Von Hippel-Lindau syndrome (VHL), Pheochromocytoma and Paraganglioma (PPGL), Acromegaly, Hyperprolactinaemia, Polycystic Ovary Syndrome (PCOS), Congenital Adrenal Hyperplasia (CAH), Turner Syndrome, alongside metabolic conditions such as diabetes and obesity and the effects of MHT. The review critically appraises each disorder's association with breast cancer risk, screening implications and therapeutic management. PATIENTS This analysis focuses on women with the aforementioned endocrine and metabolic disorders, assessing their specific breast cancer risk profiles, informed by the latest clinical evidence and molecular insights. MEASUREMENTS The review comprehensively evaluates current evidence-based approaches to screening, diagnostic accuracy and treatment in this patient cohort. Emphasis is placed on the metabolic derangements, hormonal influences and genetic predispositions that modulate breast cancer risk, providing disorder-specific recommendations for individualised care. RESULTS The findings indicate a significantly elevated breast cancer risk in patients with MEN1, necessitating early initiation of MRI screening by age 40. In MEN2, emerging evidence suggests that combining RET inhibitors with endocrine therapy may yield clinical benefits, although further research is needed to validate this approach. The breast cancer risk associated with MEN4 and VHL syndromes, while documented, remains less well-characterised, requiring further investigation. Diabetes and obesity are confirmed as major modifiable risk factors, particularly in postmenopausal women, where hyperinsulinemia and metabolic dysfunction contribute to increased incidence and poorer outcomes, notably in triple-negative breast cancer (TNBC). The role of MHT, particularly combined oestrogen-progestogen therapy, is strongly associated with increased breast cancer risk, particularly for hormone receptor-positive malignancies, necessitating cautious use and personalised treatment planning. In contrast, oestrogen-only MHT appears to confer a reduced risk in women post-hysterectomy. For patients with PCOS, CAH and Turner Syndrome, while definitive evidence of elevated breast cancer risk is lacking, individualised screening strategies and careful hormone therapy management remain essential due to the complex interplay of hormonal and metabolic factors. CONCLUSIONS The review highlights the need for personalised breast cancer screening and management protocols in women with endocrine and metabolic disorders. For high-risk groups such as MEN1 patients, early initiation of MRI screening is warranted. In women with diabetes and obesity, targeted interventions addressing hyperinsulinemia and metabolic dysfunction are critical to mitigating their increased cancer risk. The association between MHT and breast cancer underscores the importance of individualised risk stratification in hormone therapy administration, particularly in women with predisposing genetic or endocrine conditions. Enhanced surveillance tailored to the unique risk profiles of endocrine disorder patients will facilitate early detection and improve clinical outcomes. However, further large-scale studies are necessary to refine these associations and develop robust, evidence-based guidelines.
Collapse
Affiliation(s)
- Arie Hawazie
- Centre for Endocrinology, Queen Mary University, London, UK
| | - Maralyn Druce
- Centre for Endocrinology, Queen Mary University, London, UK
| |
Collapse
|
2
|
Chen D, Ma Y, Li J, Wen L, Zhang G, Huang C, Yao X. Causality between insulin use and malignant tumors of the digestive system: a two-sample mendelian randomized study. BMC Cancer 2025; 25:31. [PMID: 39773128 PMCID: PMC11708065 DOI: 10.1186/s12885-025-13452-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 01/03/2025] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Existing cohort studies show no association between insulin use and cancers of the digestive system, while numerous meta-analyses suggest that insulin use increases the risk of digestive system tumours. This study uses two-sample Mendelian randomization (MR) to further investigate the causal relationship between the two. METHODS We selected single nucleotide polymorphisms (SNPs) strongly associated with insulin use as instrumental variables and used aggregated statistics on digestive system neoplasms as the outcome event. The primary method of analysis was inverse variance weighting (IVW), supplemented by weighted median, MR-Egger regression, weighted mode and simple mode methods. The reliability of the study was assessed by heterogeneity testing, pleiotropy analysis and sensitivity analysis. RESULT A total of 8 SNPs associated with insulin use were included as instrumental variables. Random-effects IVW analysis showed an association between insulin use and increased risk of colorectal cancer (OR = 1.1037, 95%CI = 1.0183-1.1962, P = 0.016). No statistically significant association was found between insulin use and the development of other digestive system tumours. The results were unaffected by pleiotropy and heterogeneity, and the reliability of the findings was confirmed by sensitivity analysis. CONCLUSION Our Mendelian randomization study suggests an association between insulin use and an increased risk of CRC, with no clear association observed for other digestive system tumours. However, further MR studies with larger sample sizes from genome-wide association study (GWAS) data are needed to verify this association.
Collapse
Affiliation(s)
- DengZhuo Chen
- Gannan Medical University, Ganzhou, China
- Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - YongLi Ma
- Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, China
| | - JingHui Li
- Gannan Medical University, Ganzhou, China
- Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Liang Wen
- Gannan Medical University, Ganzhou, China
- Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - GuoSheng Zhang
- Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, China
| | - ChengZhi Huang
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| | - XueQing Yao
- Gannan Medical University, Ganzhou, China.
- Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, China.
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| |
Collapse
|
3
|
Leitner BP, Siebel S, Akingbesote ND, Zhang X, Perry RJ. Insulin and cancer: a tangled web. Biochem J 2022; 479:583-607. [PMID: 35244142 PMCID: PMC9022985 DOI: 10.1042/bcj20210134] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 02/13/2022] [Accepted: 02/15/2022] [Indexed: 12/13/2022]
Abstract
For a century, since the pioneering work of Otto Warburg, the interwoven relationship between metabolism and cancer has been appreciated. More recently, with obesity rates rising in the U.S. and worldwide, epidemiologic evidence has supported a link between obesity and cancer. A substantial body of work seeks to mechanistically unpack the association between obesity, altered metabolism, and cancer. Without question, these relationships are multifactorial and cannot be distilled to a single obesity- and metabolism-altering hormone, substrate, or factor. However, it is important to understand the hormone-specific associations between metabolism and cancer. Here, we review the links between obesity, metabolic dysregulation, insulin, and cancer, with an emphasis on current investigational metabolic adjuncts to standard-of-care cancer treatment.
Collapse
Affiliation(s)
- Brooks P. Leitner
- Departments of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, U.S.A
- Departments of Internal Medicine, Yale School of Medicine, New Haven, CT, U.S.A
| | - Stephan Siebel
- Departments of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, U.S.A
- Departments of Internal Medicine, Yale School of Medicine, New Haven, CT, U.S.A
- Departments of Pediatrics, Yale School of Medicine, New Haven, CT, U.S.A
| | - Ngozi D. Akingbesote
- Departments of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, U.S.A
- Departments of Internal Medicine, Yale School of Medicine, New Haven, CT, U.S.A
| | - Xinyi Zhang
- Departments of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, U.S.A
- Departments of Internal Medicine, Yale School of Medicine, New Haven, CT, U.S.A
| | - Rachel J. Perry
- Departments of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, U.S.A
- Departments of Internal Medicine, Yale School of Medicine, New Haven, CT, U.S.A
| |
Collapse
|
4
|
Bolli GB, Porcellati F, Lucidi P, Fanelli CG, Owens DR. One-hundred year evolution of prandial insulin preparations: From animal pancreas extracts to rapid-acting analogs. Metabolism 2022; 126:154935. [PMID: 34762931 DOI: 10.1016/j.metabol.2021.154935] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/26/2021] [Accepted: 11/04/2021] [Indexed: 11/23/2022]
Abstract
The first insulin preparation injected in humans in 1922 was short-acting, extracted from animal pancreas, contaminated by impurities. Ever since the insulin extracted from animal pancreas has been continuously purified, until an unlimited synthesis of regular human insulin (RHI) became possible in the '80s using the recombinant-DNA (rDNA) technique. The rDNA technique then led to the designer insulins (analogs) in the early '90s. Rapid-acting insulin analogs were developed to accelerate the slow subcutaneous (sc) absorption of RHI, thus lowering the 2-h post-prandial plasma glucose (PP-PG) and risk for late hypoglycemia as comparing with RHI. The first rapid-acting analog was lispro (in 1996), soon followed by aspart and glulisine. Rapid-acting analogs are more convenient than RHI: they improve early PP-PG, and 24-h PG and A1C as long as basal insulin is also optimized; they lower the risk of late PP hypoglycemia and they allow a shorter time-interval between injection and meal. Today rapid-acting analogs are the gold standard prandial insulins. Recently, even faster analogs have become available (faster aspart, ultra-rapid lispro) or are being studied (Biochaperone lispro), making additional gains in lowering PP-PG. Rapid-acting analogs are recommended in all those with type 1 and type 2 diabetes who need prandial insulin replacement.
Collapse
Affiliation(s)
- Geremia B Bolli
- Section of Endocrinology and Metabolism, Department of Medicine and Surgery, Perugia University School of Medicine, Perugia, Italy.
| | - Francesca Porcellati
- Section of Endocrinology and Metabolism, Department of Medicine and Surgery, Perugia University School of Medicine, Perugia, Italy
| | - Paola Lucidi
- Section of Endocrinology and Metabolism, Department of Medicine and Surgery, Perugia University School of Medicine, Perugia, Italy
| | - Carmine G Fanelli
- Section of Endocrinology and Metabolism, Department of Medicine and Surgery, Perugia University School of Medicine, Perugia, Italy
| | - David R Owens
- Diabetes Research Unit Cymru, University of Swansea Medical School, Singleton Park, Swansea SA2 8PP, Wales, United Kingdom
| |
Collapse
|
5
|
Vella V, Giuliano M, La Ferlita A, Pellegrino M, Gaudenzi G, Alaimo S, Massimino M, Pulvirenti A, Dicitore A, Vigneri P, Vitale G, Malaguarnera R, Morrione A, Sims AH, Ferro A, Maggiolini M, Lappano R, De Francesco EM, Belfiore A. Novel Mechanisms of Tumor Promotion by the Insulin Receptor Isoform A in Triple-Negative Breast Cancer Cells. Cells 2021; 10:3145. [PMID: 34831367 PMCID: PMC8621444 DOI: 10.3390/cells10113145] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 10/16/2021] [Accepted: 11/10/2021] [Indexed: 02/07/2023] Open
Abstract
The insulin receptor isoform A (IR-A) plays an increasingly recognized role in fetal growth and tumor biology in response to circulating insulin and/or locally produced IGF2. This role seems not to be shared by the IR isoform B (IR-B). We aimed to dissect the specific impact of IR isoforms in modulating insulin signaling in triple negative breast cancer (TNBC) cells. We generated murine 4T1 TNBC cells deleted from the endogenous insulin receptor (INSR) gene and expressing comparable levels of either human IR-A or IR-B. We then measured IR isoform-specific in vitro and in vivo biological effects and transcriptome in response to insulin. Overall, the IR-A was more potent than the IR-B in mediating cell migration, invasion, and in vivo tumor growth. Transcriptome analysis showed that approximately 89% of insulin-stimulated transcripts depended solely on the expression of the specific isoform. Notably, in cells overexpressing IR-A, insulin strongly induced genes involved in tumor progression and immune evasion including chemokines and genes related to innate immunity. Conversely, in IR-B overexpressing cells, insulin predominantly induced the expression of genes primarily involved in the regulation of metabolic pathways and, to a lesser extent, tumor growth and angiogenesis.
Collapse
Affiliation(s)
- Veronica Vella
- Endocrinology Unit, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania, Italy; (V.V.); (M.G.); (E.M.D.F.)
| | - Marika Giuliano
- Endocrinology Unit, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania, Italy; (V.V.); (M.G.); (E.M.D.F.)
| | - Alessandro La Ferlita
- Bioinformatics Unit, Department of Clinical and Experimental Medicine, University of Catania, 95131 Catania, Italy; (A.L.F.); (S.A.); (A.P.); (A.F.)
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH 43210, USA
| | - Michele Pellegrino
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (M.P.); (M.M.); (R.L.)
| | - Germano Gaudenzi
- Laboratory of Geriatric and Oncologic Neuroendocrinology Research, Istituto Auxologico Italiano, IRCCS, 20095 Cusano Milanino, Italy; (G.G.); (A.D.); (G.V.)
| | - Salvatore Alaimo
- Bioinformatics Unit, Department of Clinical and Experimental Medicine, University of Catania, 95131 Catania, Italy; (A.L.F.); (S.A.); (A.P.); (A.F.)
| | - Michele Massimino
- Oncology Unit, Department of Clinical and Experimental Medicine, University of Catania, 95124 Catania, Italy; (M.M.); (P.V.)
| | - Alfredo Pulvirenti
- Bioinformatics Unit, Department of Clinical and Experimental Medicine, University of Catania, 95131 Catania, Italy; (A.L.F.); (S.A.); (A.P.); (A.F.)
| | - Alessandra Dicitore
- Laboratory of Geriatric and Oncologic Neuroendocrinology Research, Istituto Auxologico Italiano, IRCCS, 20095 Cusano Milanino, Italy; (G.G.); (A.D.); (G.V.)
| | - Paolo Vigneri
- Oncology Unit, Department of Clinical and Experimental Medicine, University of Catania, 95124 Catania, Italy; (M.M.); (P.V.)
| | - Giovanni Vitale
- Laboratory of Geriatric and Oncologic Neuroendocrinology Research, Istituto Auxologico Italiano, IRCCS, 20095 Cusano Milanino, Italy; (G.G.); (A.D.); (G.V.)
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20122 Milan, Italy
| | | | - Andrea Morrione
- Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA;
| | - Andrew H. Sims
- MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Scotland EH4 2XR, UK;
| | - Alfredo Ferro
- Bioinformatics Unit, Department of Clinical and Experimental Medicine, University of Catania, 95131 Catania, Italy; (A.L.F.); (S.A.); (A.P.); (A.F.)
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (M.P.); (M.M.); (R.L.)
| | - Rosamaria Lappano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (M.P.); (M.M.); (R.L.)
| | - Ernestina Marianna De Francesco
- Endocrinology Unit, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania, Italy; (V.V.); (M.G.); (E.M.D.F.)
| | - Antonino Belfiore
- Endocrinology Unit, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania, Italy; (V.V.); (M.G.); (E.M.D.F.)
| |
Collapse
|
6
|
Dhayalan B, Chatterjee D, Chen YS, Weiss MA. Structural Lessons From the Mutant Proinsulin Syndrome. Front Endocrinol (Lausanne) 2021; 12:754693. [PMID: 34659132 PMCID: PMC8514764 DOI: 10.3389/fendo.2021.754693] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 09/13/2021] [Indexed: 12/30/2022] Open
Abstract
Insight into folding mechanisms of proinsulin has been provided by analysis of dominant diabetes-associated mutations in the human insulin gene (INS). Such mutations cause pancreatic β-cell dysfunction due to toxic misfolding of a mutant proinsulin and impairment in trans of wild-type insulin secretion. Anticipated by the "Akita" mouse (a classical model of monogenic diabetes mellitus; DM), this syndrome illustrates the paradigm endoreticulum (ER) stress leading to intracellular proteotoxicity. Diverse clinical mutations directly or indirectly perturb native disulfide pairing leading to protein misfolding and aberrant aggregation. Although most introduce or remove a cysteine (Cys; leading in either case to an unpaired thiol group), non-Cys-related mutations identify key determinants of folding efficiency. Studies of such mutations suggest that the hormone's evolution has been constrained not only by structure-function relationships, but also by the susceptibility of its single-chain precursor to impaired foldability. An intriguing hypothesis posits that INS overexpression in response to peripheral insulin resistance likewise leads to chronic ER stress and β-cell dysfunction in the natural history of non-syndromic Type 2 DM. Cryptic contributions of conserved residues to folding efficiency, as uncovered by rare genetic variants, define molecular links between biophysical principles and the emerging paradigm of Darwinian medicine: Biosynthesis of proinsulin at the edge of non-foldability provides a key determinant of "diabesity" as a pandemic disease of civilization.
Collapse
Affiliation(s)
| | | | | | - Michael A. Weiss
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
7
|
Zhang AM, Wellberg EA, Kopp JL, Johnson JD. Hyperinsulinemia in Obesity, Inflammation, and Cancer. Diabetes Metab J 2021; 45:285-311. [PMID: 33775061 PMCID: PMC8164941 DOI: 10.4093/dmj.2020.0250] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/23/2020] [Indexed: 12/13/2022] Open
Abstract
The relative insufficiency of insulin secretion and/or insulin action causes diabetes. However, obesity and type 2 diabetes mellitus can be associated with an absolute increase in circulating insulin, a state known as hyperinsulinemia. Studies are beginning to elucidate the cause-effect relationships between hyperinsulinemia and numerous consequences of metabolic dysfunctions. Here, we review recent evidence demonstrating that hyperinsulinemia may play a role in inflammation, aging and development of cancers. In this review, we will focus on the consequences and mechanisms of excess insulin production and action, placing recent findings that have challenged dogma in the context of the existing body of literature. Where relevant, we elaborate on the role of specific signal transduction components in the actions of insulin and consequences of chronic hyperinsulinemia. By discussing the involvement of hyperinsulinemia in various metabolic and other chronic diseases, we may identify more effective therapeutics or lifestyle interventions for preventing or treating obesity, diabetes and cancer. We also seek to identify pertinent questions that are ripe for future investigation.
Collapse
Affiliation(s)
- Anni M.Y. Zhang
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Elizabeth A. Wellberg
- Department of Pathology, University of Oklahoma Health Sciences Center, Stephenson Cancer Center, Harold Hamm Diabetes Center, Oklahoma City, OK, USA
| | - Janel L. Kopp
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - James D. Johnson
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
8
|
Dhayalan B, Chatterjee D, Chen YS, Weiss MA. Diabetes mellitus due to toxic misfolding of proinsulin variants. Mol Metab 2021:101229. [PMID: 33823319 DOI: 10.1016/j.molmet.2021.101229] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 03/10/2021] [Accepted: 03/29/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Dominant mutations in the human insulin gene (INS) lead to pancreatic β-cell dysfunction and diabetes mellitus (DM) due to toxic misfolding of a mutant proinsulin. Analogous to a classical mouse model of monogenic DM ("Akita"), this syndrome highlights the susceptibility of β-cells to endoreticulum (ER) stress due to protein misfolding and aberrant aggregation. SCOPE OF REVIEW Diverse clinical mutations directly or indirectly perturb native disulfide pairing. Whereas most introduce or remove a cysteine (Cys; leading in either case to an unpaired thiol group), non-Cys-related mutations identify key determinants of folding efficiency. Studies of such mutations suggest that the hormone's evolution has been constrained not only by structure-function relationships but also by the susceptibility of its single-chain precursor to impaired foldability. An intriguing hypothesis posits that INS overexpression in response to peripheral insulin resistance likewise leads to chronic ER stress and β-cell dysfunction in the natural history of nonsyndromic Type 2 DM. MAJOR CONCLUSIONS Cryptic contributions of conserved residues to folding efficiency, as uncovered by rare genetic variants, define molecular links between biophysical principles and the emerging paradigm of Darwinian medicine: Biosynthesis of proinsulin at the edge of nonfoldability provides a key determinant of "diabesity" as a pandemic disease of civilization.
Collapse
Affiliation(s)
- Balamurugan Dhayalan
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Deepak Chatterjee
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yen-Shan Chen
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Michael A Weiss
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
9
|
Holowatyj AN, Viskochil R, Ose D, Tingey B, Haaland B, Wilson D, Larson M, Feltz S, Lewis MA, Colman H, Ulrich CM. Diabetes, Body Fatness, and Insulin Prescription Among Adolescents and Young Adults with Cancer. J Adolesc Young Adult Oncol 2021; 10:217-225. [PMID: 32749900 PMCID: PMC8064923 DOI: 10.1089/jayao.2020.0071] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Purpose: Rates of obesity and obesity-related health consequences, including type 2 diabetes (T2D) and cancer, continue to rise. While cancer patients are at an increased risk of developing T2D, the prevalence of T2D and insulin prescription among young patients with cancer remains unknown. Methods: Using the Total Cancer Care Study cohort at Huntsman Cancer Institute (Salt Lake City, UT), we identified individuals age 18-39 years at cancer diagnosis between 2009 and 2019. Multivariable logistic regression was used to investigate associations between body mass index (BMI) with insulin prescription within 1 year of cancer diagnosis. Results: In total, 344 adolescents and young adults (AYAs) were diagnosed with primary invasive cancer. Within this cohort, 19 patients (5.5%) were ever diagnosed with T2D, 48 AYAs ever received an insulin prescription (14.0%), and 197 were overweight or obese (BMI: 25+ kg/m2) at cancer diagnosis. Each kg/m2 unit increase in BMI was associated with 6% increased odds of first insulin prescription within 1 year of cancer diagnosis among AYAs, even after adjustment for age, sex, smoking history, marital status, glucocorticoid prescription, and cancer treatments (odds ratio = 1.06, 95% confidence interval 1.02-1.11; p = 0.005). Conclusion: One in every 18 AYAs with cancer ever had T2D, 1 in 7 AYA patients with cancer ever received an insulin prescription, and higher BMI was associated with increased risk of insulin prescription within a year of cancer diagnosis among AYAs. Understanding the incidence of T2D and insulin prescription/use is critical for short-term and long-term clinical management of AYAs with cancer.
Collapse
Affiliation(s)
- Andreana N. Holowatyj
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt-Ingram Cancer Center, Nashville, Tennessee, USA
- Huntsman Cancer Institute, Salt Lake City, Utah, USA
- Department of Population Health Sciences, University of Utah, Salt Lake City, Utah, USA
| | - Richard Viskochil
- Huntsman Cancer Institute, Salt Lake City, Utah, USA
- Department of Population Health Sciences, University of Utah, Salt Lake City, Utah, USA
| | - Dominik Ose
- Huntsman Cancer Institute, Salt Lake City, Utah, USA
- Department of Family and Preventive Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Benjamin Tingey
- Department of Family and Preventive Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Benjamin Haaland
- Huntsman Cancer Institute, Salt Lake City, Utah, USA
- Department of Population Health Sciences, University of Utah, Salt Lake City, Utah, USA
| | - Dalton Wilson
- Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | | | - Sara Feltz
- Data Science Services, University of Utah, Salt Lake City, Utah, USA
| | - Mark A. Lewis
- Department of Internal Medicine, Intermountain Healthcare, Murray, Utah, USA
| | - Howard Colman
- Huntsman Cancer Institute, Salt Lake City, Utah, USA
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
- Department of Neurosurgery, University of Utah, Salt Lake City, Utah, USA
| | - Cornelia M. Ulrich
- Huntsman Cancer Institute, Salt Lake City, Utah, USA
- Department of Population Health Sciences, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
10
|
Scully T, Ettela A, LeRoith D, Gallagher EJ. Obesity, Type 2 Diabetes, and Cancer Risk. Front Oncol 2021; 10:615375. [PMID: 33604295 PMCID: PMC7884814 DOI: 10.3389/fonc.2020.615375] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 12/09/2020] [Indexed: 12/12/2022] Open
Abstract
Obesity and type 2 diabetes have both been associated with increased cancer risk and are becoming increasingly prevalent. Metabolic abnormalities such as insulin resistance and dyslipidemia are associated with both obesity and type 2 diabetes and have been implicated in the obesity-cancer relationship. Multiple mechanisms have been proposed to link obesity and diabetes with cancer progression, including an increase in insulin/IGF-1 signaling, lipid and glucose uptake and metabolism, alterations in the profile of cytokines, chemokines, and adipokines, as well as changes in the adipose tissue directly adjacent to the cancer sites. This review aims to summarize and provide an update on the epidemiological and mechanistic evidence linking obesity and type 2 diabetes with cancer, focusing on the roles of insulin, lipids, and adipose tissue.
Collapse
Affiliation(s)
- Tiffany Scully
- Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Abora Ettela
- Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Derek LeRoith
- Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
- Tisch Cancer Institute at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Emily Jane Gallagher
- Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
- Tisch Cancer Institute at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| |
Collapse
|
11
|
Das M, Ellies LG, Kumar D, Sauceda C, Oberg A, Gross E, Mandt T, Newton IG, Kaur M, Sears DD, Webster NJG. Time-restricted feeding normalizes hyperinsulinemia to inhibit breast cancer in obese postmenopausal mouse models. Nat Commun 2021; 12:565. [PMID: 33495474 PMCID: PMC7835248 DOI: 10.1038/s41467-020-20743-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 12/04/2020] [Indexed: 01/30/2023] Open
Abstract
Accumulating evidence indicates that obesity with its associated metabolic dysregulation, including hyperinsulinemia and aberrant circadian rhythms, increases the risk for a variety of cancers including postmenopausal breast cancer. Caloric restriction can ameliorate the harmful metabolic effects of obesity and inhibit cancer progression but is difficult to implement and maintain outside of the clinic. In this study, we aim to test a time-restricted feeding (TRF) approach on mouse models of obesity-driven postmenopausal breast cancer. We show that TRF abrogates the obesity-enhanced mammary tumor growth in two orthotopic models in the absence of calorie restriction or weight loss. TRF also reduces breast cancer metastasis to the lung. Furthermore, TRF delays tumor initiation in a transgenic model of mammary tumorigenesis prior to the onset of obesity. Notably, TRF increases whole-body insulin sensitivity, reduces hyperinsulinemia, restores diurnal gene expression rhythms in the tumor, and attenuates tumor growth and insulin signaling. Importantly, inhibition of insulin secretion with diazoxide mimics TRF whereas artificial elevation of insulin through insulin pumps implantation reverses the effect of TRF, suggesting that TRF acts through modulating hyperinsulinemia. Our data suggest that TRF is likely to be effective in breast cancer prevention and therapy.
Collapse
Affiliation(s)
- Manasi Das
- VA San Diego Healthcare System, San Diego, CA, USA
- Department of Medicine, Division of Endocrinology and Metabolism, University of California San Diego, La Jolla, CA, USA
| | - Lesley G Ellies
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Deepak Kumar
- VA San Diego Healthcare System, San Diego, CA, USA
- Department of Medicine, Division of Endocrinology and Metabolism, University of California San Diego, La Jolla, CA, USA
| | - Consuelo Sauceda
- VA San Diego Healthcare System, San Diego, CA, USA
- Department of Medicine, Division of Endocrinology and Metabolism, University of California San Diego, La Jolla, CA, USA
| | - Alexis Oberg
- VA San Diego Healthcare System, San Diego, CA, USA
| | - Emilie Gross
- VA San Diego Healthcare System, San Diego, CA, USA
- Department of Medicine, Division of Endocrinology and Metabolism, University of California San Diego, La Jolla, CA, USA
| | - Tyler Mandt
- Department of Radiology, University of California, San Diego, La Jolla, CA, USA
| | - Isabel G Newton
- Department of Radiology, University of California, San Diego, La Jolla, CA, USA
| | - Mehak Kaur
- Department of Medicine, Division of Endocrinology and Metabolism, University of California San Diego, La Jolla, CA, USA
| | - Dorothy D Sears
- Department of Medicine, Division of Endocrinology and Metabolism, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
- Department of Family Medicine and Public Health, Division of Preventive Medicine, University of California San Diego, La Jolla, CA, USA
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
| | - Nicholas J G Webster
- VA San Diego Healthcare System, San Diego, CA, USA.
- Department of Medicine, Division of Endocrinology and Metabolism, University of California San Diego, La Jolla, CA, USA.
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
12
|
Disrupting Insulin and IGF Receptor Function in Cancer. Int J Mol Sci 2021; 22:ijms22020555. [PMID: 33429867 PMCID: PMC7827299 DOI: 10.3390/ijms22020555] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 12/29/2020] [Accepted: 01/06/2021] [Indexed: 02/07/2023] Open
Abstract
The insulin and insulin-like growth factor (IGF) system plays an important role in regulating normal cell proliferation and survival. However, the IGF system is also implicated in many malignancies, including breast cancer. Preclinical studies indicate several IGF blocking approaches, such as monoclonal antibodies and tyrosine kinase inhibitors, have promising therapeutic potential for treating diseases. Uniformly, phase III clinical trials have not shown the benefit of blocking IGF signaling compared to standard of care arms. Clinical and laboratory data argue that targeting Type I IGF receptor (IGF1R) alone may be insufficient to disrupt this pathway as the insulin receptor (IR) may also be a relevant cancer target. Here, we review the well-studied role of the IGF system in regulating malignancies, the limitations on the current strategies of blocking the IGF system in cancer, and the potential future directions for targeting the IGF system.
Collapse
|
13
|
Jensen VFH, Brinck PR, Nowak J, Thorup I, Sjögren I, Mølck AM. Evaluation of Cell Proliferation in Rat Mammary Glands Is Not Predictive of the Carcinogenic Potential of Insulin In Vivo. Int J Toxicol 2020; 39:560-576. [PMID: 32723118 DOI: 10.1177/1091581820941776] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
For nonclinical safety-assessment of insulin analogues in vivo, mitogenic effects are compared to that of human insulin. Besides histopathologic evaluation, this usually includes assessment of cell proliferation (CP) in mammary glands. Insulin analogue X10 is recommended as positive control, due to its known carcinogenic effect in rat mammary glands. Here, we discuss the mitogenic effect of insulin in vivo and use of X10 as positive control. We present results from 4 nonclinical rat studies evaluating effects of repeated dosing with insulin detemir (≤26 weeks) or degludec (52 weeks) in mammary glands. Studies included human insulin-dosed groups as comparators, CP, and histopathologic evaluation. One study included an X10-dosed group (26 weeks), another ≤3 weeks of dosing with X10 or human insulin evaluating effects of these comparators. Neither human insulin, insulin detemir, degludec, nor X10 induced mammary tumors or increased CP in the studies. The CP marker proliferating cell nuclear antigen varied within/between studies and was not correlated with the remaining markers or CP fluctuations during estrous cycle, whereas the other CP markers, Ki-67 and 5-bromo-2'-deoxyuridine (BrdU), correlated with estrous cycle changes and each other. In conclusion, we propose that the mitogenic effect of insulin in rat mammary glands is weak in vivo. Cell proliferation evaluation in nonclinical safety assessment studies is not predictive of the carcinogenic potential of insulin, thus, the value of including this end point is debatable. Moreover, X10 is not recommended as positive control, due to lack of proliferative effects. Typical CP markers vary greatly in quality, BrdU seemingly most reliable.
Collapse
Affiliation(s)
| | - Peter R Brinck
- Department of Safety Sciences & Imaging, 1450Novo Nordisk A/S, Maaloev, Denmark
| | - Jette Nowak
- Department of Safety Sciences & Imaging, 1450Novo Nordisk A/S, Maaloev, Denmark
| | - Inger Thorup
- Department of Safety Sciences & Imaging, 1450Novo Nordisk A/S, Maaloev, Denmark
| | - Ingrid Sjögren
- Department of Safety Sciences & Imaging, 1450Novo Nordisk A/S, Maaloev, Denmark
| | - Anne-Marie Mølck
- Department of Safety Sciences & Imaging, 1450Novo Nordisk A/S, Maaloev, Denmark
| |
Collapse
|
14
|
Hvid H, Glendorf T, Brandt J, Slaaby R, Lützen A, Kristensen K, Hansen BF. Increased insulin receptor binding and increased IGF-1 receptor binding are linked with increased growth of L6hIR cell xenografts in vivo. Sci Rep 2020; 10:7247. [PMID: 32350367 PMCID: PMC7190841 DOI: 10.1038/s41598-020-64318-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 04/15/2020] [Indexed: 11/09/2022] Open
Abstract
Insulin analogue X10 has a higher mitogenic potency than native human insulin in vitro and supra-pharmacological doses of insulin X10 increased the incidence of mammary tumours in rats. Compared to native human insulin, insulin X10 has increased binding affinity to the insulin receptor and the IGF-1 receptor, but it is not known whether either or both characteristics are important for stimulation of cell proliferation in vivo. The aim of this study was to explore how increased binding affinity to the insulin receptor or the IGF-1 receptor contributes to stimulation of cell proliferation in vivo. A mouse xenograft model was established with rat L6 myoblast cells transfected with the human insulin receptor (L6hIR cells) and effects of supra-pharmacological doses of native human insulin, insulin X10 or novel insulin analogues with increased binding affinity to either the insulin receptor or the IGF-1 receptor were examined. Treatment with insulin X10 and insulin analogues with increased binding affinity to either the insulin receptor or the IGF-1 receptor increased growth of L6hIR cell xenografts significantly compared to native human insulin. Thus, increased binding affinity to the insulin receptor and the IGF-1 receptor are each independently linked to increased growth of L6hIR cell xenografts in vivo.
Collapse
Affiliation(s)
- Henning Hvid
- Global Drug Discovery, Novo Nordisk A/S, Copenhagen, Denmark.
| | - Tine Glendorf
- Global Drug Discovery, Novo Nordisk A/S, Copenhagen, Denmark
| | - Jakob Brandt
- Global Research Technologies, Novo Nordisk A/S, Copenhagen, Denmark
| | - Rita Slaaby
- Global Drug Discovery, Novo Nordisk A/S, Copenhagen, Denmark
| | - Anne Lützen
- Global Drug Discovery, Novo Nordisk A/S, Copenhagen, Denmark
| | - Kim Kristensen
- Global Drug Discovery, Novo Nordisk A/S, Copenhagen, Denmark
| | - Bo F Hansen
- Global Drug Discovery, Novo Nordisk A/S, Copenhagen, Denmark
| |
Collapse
|
15
|
Nasiri AR, Rodrigues MR, Li Z, Leitner BP, Perry RJ. SGLT2 inhibition slows tumor growth in mice by reversing hyperinsulinemia. Cancer Metab 2019; 7:10. [PMID: 31867105 PMCID: PMC6907191 DOI: 10.1186/s40170-019-0203-1] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 11/20/2019] [Indexed: 02/07/2023] Open
Abstract
Background Obesity confers an increased risk and accelerates the progression of multiple tumor types in rodents and humans, including both breast and colon cancer. Because sustained weight loss is rarely achieved, therapeutic approaches to slow or prevent obesity-associated cancer development have been limited, and mechanistic insights as to the obesity-cancer connection have been lacking. Methods E0771 breast tumors and MC38 colon tumors were treated in vivo in mice and in vitro with two mechanistically different insulin-lowering agents, a controlled-release mitochondrial protonophore (CRMP) and sodium-glucose cotransporter-2 (SGLT2) inhibitors, and tumor growth and glucose metabolism were assessed. Groups were compared by ANOVA with Bonferroni’s multiple comparisons test. Results Dapagliflozin slows tumor growth in two mouse models (E0771 breast cancer and MC38 colon adenocarcinoma) of obesity-associated cancers in vivo, and a mechanistically different insulin-lowering agent, CRMP, also slowed breast tumor growth through its effect to reverse hyperinsulinemia. In both models and with both agents, tumor glucose uptake and oxidation were not constitutively high, but were hormone-responsive. Restoration of hyperinsulinemia by subcutaneous insulin infusion abrogated the effects of both dapagliflozin and CRMP to slow tumor growth. Conclusions Taken together, these data demonstrate that hyperinsulinemia per se promotes both breast and colon cancer progression in obese mice, and highlight SGLT2 inhibitors as a clinically available means of slowing obesity-associated tumor growth due to their glucose- and insulin-lowering effects.
Collapse
Affiliation(s)
- Ali R Nasiri
- 1Department of Internal Medicine, School of Medicine, Yale University, PO Box 208020, TAC S269, New Haven, CT 06520 USA
| | - Marcos R Rodrigues
- 1Department of Internal Medicine, School of Medicine, Yale University, PO Box 208020, TAC S269, New Haven, CT 06520 USA.,3Department of Surgery, State University of Ponta Grossa, Ponta Grossa, Brazil
| | - Zongyu Li
- 1Department of Internal Medicine, School of Medicine, Yale University, PO Box 208020, TAC S269, New Haven, CT 06520 USA.,2Department of Cellular & Molecular Physiology, School of Medicine Yale University, PO Box 208020, TAC S269, New Haven, CT 06520 USA
| | - Brooks P Leitner
- 1Department of Internal Medicine, School of Medicine, Yale University, PO Box 208020, TAC S269, New Haven, CT 06520 USA.,2Department of Cellular & Molecular Physiology, School of Medicine Yale University, PO Box 208020, TAC S269, New Haven, CT 06520 USA
| | - Rachel J Perry
- 1Department of Internal Medicine, School of Medicine, Yale University, PO Box 208020, TAC S269, New Haven, CT 06520 USA.,2Department of Cellular & Molecular Physiology, School of Medicine Yale University, PO Box 208020, TAC S269, New Haven, CT 06520 USA
| |
Collapse
|
16
|
Vella V, Nicolosi ML, Giuliano M, Morrione A, Malaguarnera R, Belfiore A. Insulin Receptor Isoform A Modulates Metabolic Reprogramming of Breast Cancer Cells in Response to IGF2 and Insulin Stimulation. Cells 2019; 8:cells8091017. [PMID: 31480557 PMCID: PMC6770491 DOI: 10.3390/cells8091017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 08/27/2019] [Accepted: 08/29/2019] [Indexed: 12/15/2022] Open
Abstract
Previously published work has demonstrated that overexpression of the insulin receptor isoform A (IR-A) might play a role in cancer progression and metastasis. The IR has a predominant metabolic role in physiology, but the potential role of IR-A in cancer metabolic reprogramming is unknown. We aimed to characterize the metabolic impact of IR-A and its ligand insulin like growth factor 2 (IGF2) in human breast cancer (BC) cells. To establish autocrine IGF2 action, we generated human BC cells MCF7 overexpressing the human IGF2, while we focused on the metabolic effect of IR-A by stably infecting IGF1R-ablated MCF7 (MCF7IGF1R-ve) cells with a human IR-A cDNA. We then evaluated the expression of key metabolism related molecules and measured real-time extracellular acidification rates and oxygen consumption rates using the Seahorse technology. MCF7/IGF2 cells showed increased proliferation and invasion associated with aerobic glycolysis and mitochondrial biogenesis and activity. In MCF7IGF1R-ve/IR-A cells insulin and IGF2 stimulated similar metabolic changes and were equipotent in eliciting proliferative responses, while IGF2 more potently induced invasion. The combined treatment with the glycolysis inhibitor 2-deoxyglucose (2DG) and the mitochondrial inhibitor metformin blocked cell invasion and colony formation with additive effects. Overall, these results indicate that IGF2 and IR-A overexpression may contribute to BC metabolic reprogramming.
Collapse
Affiliation(s)
- Veronica Vella
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, Catania 95122, Italy
| | - Maria Luisa Nicolosi
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, Catania 95122, Italy
| | - Marika Giuliano
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, Catania 95122, Italy
| | - Andrea Morrione
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Roberta Malaguarnera
- School of Human and Social Sciences, "Kore" University of Enna, Enna 94100, Italy
| | - Antonino Belfiore
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, Catania 95122, Italy.
| |
Collapse
|
17
|
Sheng X, Yao K, Shao A, Tu S, Zhang X, Chen T, Yao D. The Role of Insulin Glargine and Human Insulin in the Regulation of Thyroid Proliferation Through Mitogenic Signaling. Front Endocrinol (Lausanne) 2019; 10:594. [PMID: 31555212 PMCID: PMC6723759 DOI: 10.3389/fendo.2019.00594] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 08/13/2019] [Indexed: 12/29/2022] Open
Abstract
Our aim was to investigate whether human insulin (HI) or insulin glargine treatment could promote the proliferation of thyroid cells and determine the association between type 2 diabetes and thyroid disease. Rats were treated with different doses of HI and insulin glargine. Plasma glucose and the phosphorylation levels of the insulin receptor (IR), insulin-like growth factor 1 receptor (IGF-1R), protein kinase B (Akt), and extracellular signal-regulated kinase 1/2 (ERK1/2) were measured. A total of 105 rats were randomly assigned to three groups as follows: control group, HI group, and glargine group. Both drugs promoted the phosphorylation of IR, Akt, and ERK1/2 in a dose-dependent manner (p < 0.05), and the effect of glargine persisted for longer period. Treatment with ultra-therapeutic doses of HI or glargine (p < 0.05) increased the expression of Ki-67 in thyroid cells. The results demonstrated that therapeutic doses of glargine have a longer-lasting hypoglycemic control than HI. Based on the results, HI or glargine did not stimulate thyroid cell proliferation at therapeutic doses, but high doses did.
Collapse
Affiliation(s)
- Xiaoli Sheng
- Department of Obstetrics, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Kannan Yao
- The Second Central Laboratory, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Sheng Tu
- Department of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xinxia Zhang
- Department of Geriatrics, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ting Chen
- Department of Ultrasonography, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Dingguo Yao
- Department of Endocrinology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
18
|
Senapati P, Kato H, Lee M, Leung A, Thai C, Sanchez A, Gallagher EJ, LeRoith D, Seewaldt VL, Ann DK, Schones DE. Hyperinsulinemia promotes aberrant histone acetylation in triple-negative breast cancer. Epigenetics Chromatin 2019; 12:44. [PMID: 31315653 PMCID: PMC6636093 DOI: 10.1186/s13072-019-0290-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 07/07/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Hyperinsulinemia, the presence of excess insulin relative to glucose in the blood, is considered to be a poor prognostic indicator for patients with triple-negative breast cancer (TNBC). mTOR, a downstream effector of insulin, enhances mitochondrial biogenesis and activity, thereby increasing acetyl-CoA precursors. Increased acetyl-CoA can, in turn, be utilized by nuclear acetyltransferases for histone acetylation, a critical feature of genome regulation. While signaling pathways downstream of insulin have been established for sometime, the effect of insulin on chromatin remains unclear. We hypothesized that hyperinsulinemia-induced metabolic changes lead to genome-wide changes in histone acetylation in TNBC. RESULTS MDA-MB-231 cells were xenografted into hyperinsulinemic and wild-type mice. Tumors in the hyperinsulinemic mice displayed elevated levels of histone acetylation compared to tumors in normal insulin conditions. We show that insulin treatment in vitro leads to global increase in chromatin-associated histone acetylation, in particular at H3K9, through the PI3K/AKT/mTOR pathway. Genome-wide analyses revealed that most promoter regions have an increase in histone acetylation upon insulin treatment. In addition, insulin induces higher levels of reactive oxygen species and DNA damage foci in cells. CONCLUSIONS These results demonstrate the impact of hyperinsulinemia on altered gene regulation through chromatin and the importance of targeting hyperinsulinemia-induced processes that lead to chromatin dysfunction in TNBC.
Collapse
Affiliation(s)
- Parijat Senapati
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of Hope, Duarte, CA 91010 USA
| | - Hiroyuki Kato
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of Hope, Duarte, CA 91010 USA
| | - Michael Lee
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of Hope, Duarte, CA 91010 USA
- Irell & Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA USA
| | - Amy Leung
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of Hope, Duarte, CA 91010 USA
| | - Christine Thai
- Department of Population Sciences, Beckman Research Institute, City of Hope, Duarte, CA 91010 USA
| | - Angelica Sanchez
- Department of Population Sciences, Beckman Research Institute, City of Hope, Duarte, CA 91010 USA
| | - Emily J. Gallagher
- Division of Endocrinology, Diabetes and Bone Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Derek LeRoith
- Division of Endocrinology, Diabetes and Bone Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Victoria L. Seewaldt
- Irell & Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA USA
- Department of Population Sciences, Beckman Research Institute, City of Hope, Duarte, CA 91010 USA
| | - David K. Ann
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of Hope, Duarte, CA 91010 USA
- Irell & Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA USA
| | - Dustin E. Schones
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of Hope, Duarte, CA 91010 USA
- Irell & Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA USA
| |
Collapse
|
19
|
Hvid H, Jørgensen MS, Blume N, Slaaby R, Lützen A, Hansen BF. Activation of insulin receptors and IGF-1 receptors in COLO-205 colon cancer xenografts by insulin and insulin analogue X10 does not enhance growth under normo- or hypoglycaemic conditions. Diabetologia 2018; 61:2447-2457. [PMID: 30003309 DOI: 10.1007/s00125-018-4684-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 06/04/2018] [Indexed: 12/29/2022]
Abstract
AIMS/HYPOTHESIS Recent studies with normal rats and mouse allograft models have reported that insulin and insulin analogues do not activate the IGF-1 receptor in vivo, and that this characteristic therefore cannot be responsible for the increased incidence of mammary tumours observed for the insulin analogue X10 in chronic toxicity studies with Sprague Dawley rats. This is in clear contrast to reports of insulin and insulin analogues in vitro. Clarification of this is important for understanding the mechanisms behind possible growth-promoting effects of insulin analogues, and will have implications for the development of novel insulin analogues. METHODS We established a xenograft model in BALB/c nude mice with the human colon cancer cell line COLO-205, which expresses human insulin and IGF-1 receptors, and explored the acute and chronic effects of treatment with supra-pharmacological doses of human insulin, insulin analogue X10 and human IGF-1. With a novel antibody, acute IGF-1 receptor activation was also examined in various tissues from normal rats treated with human insulin, insulin analogue X10 or human IGF-1. Finally, the effects of pharmacologically relevant doses of human insulin and insulin analogue X10 on receptor activation and growth of COLO-205 xenograft were explored in BALB/c nude mice with alloxan-induced hyperglycaemia. RESULTS In normal rats and in BALB/c nude mice bearing a COLO-205 cell xenograft, treatment with supra-pharmacological doses of human insulin, insulin analogue X10 or human IGF-1 resulted in activation of insulin receptors as well as IGF-1 receptors. Treatment of diabetic nude mice with pharmacologically relevant doses of human insulin or insulin analogue X10, which decreased blood glucose from hyperglycaemic levels to the normoglycaemic range, did not increase IGF-1 receptor activation. Furthermore, repeated treatment with supra-pharmacological as well as pharmacological doses of human insulin or insulin analogue X10 did not influence the growth of COLO-205 xenografts. CONCLUSIONS/INTERPRETATION This study demonstrates that activation of IGF-1 receptors in cancer cells by insulin and insulin analogues cannot be considered as a purely in vitro phenomenon. It does occur in vivo in animal models, although only after treatment with supra-pharmacological doses. Furthermore, treatment with insulin or insulin analogue X10 did not influence the growth of COLO-205 xenografts under normo- or hypoglycaemic conditions. Further studies are needed before a conclusion can be reached on whether IGF-1 receptor activation by insulin analogues correlates with increased growth in vivo.
Collapse
Affiliation(s)
- Henning Hvid
- Insulin Research, Novo Nordisk A/S, Novo Nordisk Park, DK-2760, Maaloev, Denmark.
| | - Mikkel S Jørgensen
- Insulin Research, Novo Nordisk A/S, Novo Nordisk Park, DK-2760, Maaloev, Denmark
| | - Niels Blume
- Insulin Research, Novo Nordisk A/S, Novo Nordisk Park, DK-2760, Maaloev, Denmark
| | - Rita Slaaby
- Insulin Research, Novo Nordisk A/S, Novo Nordisk Park, DK-2760, Maaloev, Denmark
| | - Anne Lützen
- Insulin Research, Novo Nordisk A/S, Novo Nordisk Park, DK-2760, Maaloev, Denmark
| | - Bo F Hansen
- Insulin Research, Novo Nordisk A/S, Novo Nordisk Park, DK-2760, Maaloev, Denmark
| |
Collapse
|
20
|
Kang C, LeRoith D, Gallagher EJ. Diabetes, Obesity, and Breast Cancer. Endocrinology 2018; 159:3801-3812. [PMID: 30215698 PMCID: PMC6202853 DOI: 10.1210/en.2018-00574] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 09/05/2018] [Indexed: 12/13/2022]
Abstract
The rates of obesity and diabetes are increasing worldwide, whereas the age of onset for both obesity and diabetes are decreasing steadily. Obesity and diabetes are associated with multiple factors that contribute to the increased risk of a number of different cancers, including breast cancer. These factors are hyperinsulinemia, elevated IGFs, hyperglycemia, dyslipidemia, adipokines, inflammatory cytokines, and the gut microbiome. In this review, we discuss the current understanding of the complex signaling pathways underlying these multiple factors involved in the obesity/diabetes-breast cancer link, with a focus particularly on the roles of the insulin/IGF system and dyslipidemia in preclinical breast cancer models. We review some of the therapeutic strategies to target these metabolic derangements in cancer. Future research directions and potential therapeutic strategies are also discussed.
Collapse
Affiliation(s)
- Chifei Kang
- Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Derek LeRoith
- Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Emily J Gallagher
- Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
21
|
Cignarelli A, Genchi VA, Caruso I, Natalicchio A, Perrini S, Laviola L, Giorgino F. Diabetes and cancer: Pathophysiological fundamentals of a 'dangerous affair'. Diabetes Res Clin Pract 2018; 143:378-388. [PMID: 29679627 DOI: 10.1016/j.diabres.2018.04.002] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 03/27/2018] [Accepted: 04/03/2018] [Indexed: 02/06/2023]
Abstract
Diabetes and cancer are worldwide chronic diseases with a major impact on the quality and expectancy of life. Metabolic abnormalities observed during the onset and progression of diabetes may have a critical role on the initiation and progression of carcinogenesis. To date, there are no conclusive data on the mechanisms underlying the relationship between diabetes and any type of human cancer. However, recent evidence suggests that both hyperglycemia and hyperinsulinemia in diabetes could elicit cell damage responses, such as glucotoxicity, lipotoxicity and oxidative stress, which participate in the cell transformation process raising the risk of cancer development. In addition, clinical trials have revealed that several anti-diabetes therapies may potentially affect the risk of cancer though largely undefined mechanisms. In this review, we highlight epidemiological and pathophysiological aspects of diabetes, which may influence cancer initiation and progression.
Collapse
Affiliation(s)
- Angelo Cignarelli
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Valentina Annamaria Genchi
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Irene Caruso
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Annalisa Natalicchio
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Sebastio Perrini
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Luigi Laviola
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Francesco Giorgino
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy.
| |
Collapse
|
22
|
Juárez-Vázquez CI, Gurrola-Díaz CM, Vargas-Guerrero B, Domínguez-Rosales JA, Rodriguez-Ortiz JF, Barros-Núñez P, Flores-Martínez SE, Sánchez-Corona J, Rosales-Reynoso MA. Insulin glargine affects the expression of Igf-1r, Insr, and Igf-1 genes in colon and liver of diabetic rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2018; 21:489-494. [PMID: 29922429 PMCID: PMC6000212 DOI: 10.22038/ijbms.2018.24867.6185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Objective(s): The mitogenic effect of the analogous insulin glargine is currently under debate since several clinical studies have raised the possibility that insulin glargine treatment has a carcinogenic potential in different tissues. This study aimed to evaluate the Igf-1r, Insr, and Igf-1 gene expression in colon and liver of streptozotocin-induced diabetic rats in response to insulin glargine, neutral protamine Hagedorn (NPH) insulin, and metformin treatments. Materials and Methods: Male Wistar rats were induced during one week with streptozotocin to develop Type 2 Diabetes (T2D) and then randomly distributed into four groups. T2D rats included in the first group received insulin glargine, the second group received NPH insulin, the third group received metformin; finally, untreated T2D rats were included as the control group. All groups were treated for seven days; after the treatment, tissue samples of liver and colon were obtained. Quantitative PCR (qPCR) was performed to analyze the Igf-1r, Insr and Igf-1 gene expression in each tissue sample. Results: The liver tissue showed overexpression of the Insr and Igf-1r genes (P>0.001) in rats treated with insulin glargine in comparison with the control group. Similar results were observed for the Insr gene (P>0.011) in colonic tissue of rats treated with insulin glargine. Conclusion: These observations demonstrate that insulin glargine promote an excess of insulin and IGF-1 receptors in STZ-induced diabetic rats, which could overstimulate the mitogenic signaling pathways.
Collapse
Affiliation(s)
- Clara I Juárez-Vázquez
- División de Medicina Molecular, Centro de Investigación Biomédica de Occidente. Instituto Mexicano del Seguro Social. Guadalajara, Jalisco, México
| | - Carmen M Gurrola-Díaz
- Instituto de Enfermedades Crónico Degenerativas, Departamento de Biología Molecular y Genómica, C.U.C.S, Universidad de Guadalajara. Guadalajara, Jalisco, México
| | - Belinda Vargas-Guerrero
- Instituto de Enfermedades Crónico Degenerativas, Departamento de Biología Molecular y Genómica, C.U.C.S, Universidad de Guadalajara. Guadalajara, Jalisco, México
| | - José A Domínguez-Rosales
- Instituto de Enfermedades Crónico Degenerativas, Departamento de Biología Molecular y Genómica, C.U.C.S, Universidad de Guadalajara. Guadalajara, Jalisco, México
| | - Jessica F Rodriguez-Ortiz
- División de Genética, Centro de Investigación Biomédica de Occidente. Instituto Mexicano del Seguro Social. Guadalajara, Jalisco, México
| | - Patricio Barros-Núñez
- División de Genética, Centro de Investigación Biomédica de Occidente. Instituto Mexicano del Seguro Social. Guadalajara, Jalisco, México
| | - Silvia E Flores-Martínez
- División de Medicina Molecular, Centro de Investigación Biomédica de Occidente. Instituto Mexicano del Seguro Social. Guadalajara, Jalisco, México
| | - José Sánchez-Corona
- División de Medicina Molecular, Centro de Investigación Biomédica de Occidente. Instituto Mexicano del Seguro Social. Guadalajara, Jalisco, México
| | - Mónica A Rosales-Reynoso
- División de Medicina Molecular, Centro de Investigación Biomédica de Occidente. Instituto Mexicano del Seguro Social. Guadalajara, Jalisco, México
| |
Collapse
|
23
|
Vella V, Malaguarnera R, Nicolosi ML, Palladino C, Spoleti C, Massimino M, Vigneri P, Purrello M, Ragusa M, Morrione A, Belfiore A. Discoidin domain receptor 1 modulates insulin receptor signaling and biological responses in breast cancer cells. Oncotarget 2018; 8:43248-43270. [PMID: 28591735 PMCID: PMC5522143 DOI: 10.18632/oncotarget.18020] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 04/14/2017] [Indexed: 12/21/2022] Open
Abstract
The fetal isoform A of the insulin receptor (IR-A) is frequently overexpressed in a variety of malignancies including breast cancer. IR overexpression has a recognized role in cancer progression and resistance to anticancer therapies. In particular, IR-A has a peculiar mitogenic potential and is activated not only by insulin but also by IGF-2. Previously, we identified discoidin domain receptor 1 (DDR1) as a new IR-A interacting protein. DDR1, a non-integrin collagen tyrosine kinase receptor, is overexpressed in several malignancies and plays a role in cancer progression and metastasis. We now evaluated whether DDR1 is able to exert a role in breast cancer biology by functionally cross-talking with IR. In MCF-7 human breast cancer cells, IR and DDR1 co-immunoprecipitated and co-localized after insulin or IGF-2 stimulation. In a panel of breast cancer cells, DDR1 knockdown by specific siRNAs markedly inhibited IR downstream signaling as well as proliferation, migration and colony formation in response to insulin and IGF-2. These effects were accompanied by reduction of IR protein and mRNA expression, which involved both transcriptional and post-transcriptional effects. DDR1 overexpression elicited opposite effects. Bioinformatics analysis of public domain databases showed that IR and DDR1 co-expression significantly correlates with several clinically relevant histopathological and molecular features of human breast carcinomas. These findings demonstrate that, in human breast cancer cells, DDR1 regulates IR expression and ligand dependent biological actions. This novel functional crosstalk is likely clinically relevant and may become a new molecular target in breast cancer.
Collapse
Affiliation(s)
- Veronica Vella
- School of Motor Sciences, Faculty of Human and Social Sciences, Kore University of Enna, Enna, Italy
| | - Roberta Malaguarnera
- Endocrinology, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Maria Luisa Nicolosi
- Endocrinology, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Chiara Palladino
- Endocrinology, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Cristina Spoleti
- Endocrinology, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Michele Massimino
- Department of Clinical and Experimental Medicine, Faculty of Medicine, University of Catania, Catania, Italy
| | - Paolo Vigneri
- Department of Clinical and Experimental Medicine, Faculty of Medicine, University of Catania, Catania, Italy
| | - Michele Purrello
- Department of Biomedical and Biotechnological Sciences, Unit of BioMolecular, Genome, and Complex System BioMedicine, University of Catania, Catania, Italy
| | - Marco Ragusa
- Department of Biomedical and Biotechnological Sciences, Unit of BioMolecular, Genome, and Complex System BioMedicine, University of Catania, Catania, Italy
| | - Andrea Morrione
- Department of Urology and Biology of Prostate Cancer Program, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Antonino Belfiore
- Endocrinology, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| |
Collapse
|
24
|
Glidden MD, Aldabbagh K, Phillips NB, Carr K, Chen YS, Whittaker J, Phillips M, Wickramasinghe NP, Rege N, Swain M, Peng Y, Yang Y, Lawrence MC, Yee VC, Ismail-Beigi F, Weiss MA. An ultra-stable single-chain insulin analog resists thermal inactivation and exhibits biological signaling duration equivalent to the native protein. J Biol Chem 2017; 293:47-68. [PMID: 29114035 DOI: 10.1074/jbc.m117.808626] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 10/12/2017] [Indexed: 12/12/2022] Open
Abstract
Thermal degradation of insulin complicates its delivery and use. Previous efforts to engineer ultra-stable analogs were confounded by prolonged cellular signaling in vivo, of unclear safety and complicating mealtime therapy. We therefore sought an ultra-stable analog whose potency and duration of action on intravenous bolus injection in diabetic rats are indistinguishable from wild-type (WT) insulin. Here, we describe the structure, function, and stability of such an analog, a 57-residue single-chain insulin (SCI) with multiple acidic substitutions. Cell-based studies revealed native-like signaling properties with negligible mitogenic activity. Its crystal structure, determined as a novel zinc-free hexamer at 2.8 Å, revealed a native insulin fold with incomplete or absent electron density in the C domain; complementary NMR studies are described in the accompanying article. The stability of the analog (ΔGU 5.0(±0.1) kcal/mol at 25 °C) was greater than that of WT insulin (3.3(±0.1) kcal/mol). On gentle agitation, the SCI retained full activity for >140 days at 45 °C and >48 h at 75 °C. These findings indicate that marked resistance to thermal inactivation in vitro is compatible with native duration of activity in vivo Further, whereas WT insulin forms large and heterogeneous aggregates above the standard 0.6 mm pharmaceutical strength, perturbing the pharmacokinetic properties of concentrated formulations, dynamic light scattering, and size-exclusion chromatography revealed only limited SCI self-assembly and aggregation in the concentration range 1-7 mm Such a combination of favorable biophysical and biological properties suggests that SCIs could provide a global therapeutic platform without a cold chain.
Collapse
Affiliation(s)
- Michael D Glidden
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106; Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio 44106
| | - Khadijah Aldabbagh
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Nelson B Phillips
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Kelley Carr
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Yen-Shan Chen
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Jonathan Whittaker
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Manijeh Phillips
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | | | - Nischay Rege
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Mamuni Swain
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Yi Peng
- Department of Nutrition, Case Western Reserve University, Cleveland, Ohio 44106
| | - Yanwu Yang
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Michael C Lawrence
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Vivien C Yee
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Faramarz Ismail-Beigi
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106; Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio 44106; Department of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Michael A Weiss
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106; Department of Medicine, Case Western Reserve University, Cleveland, Ohio 44106; Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106.
| |
Collapse
|
25
|
Belfiore A, Malaguarnera R, Vella V, Lawrence MC, Sciacca L, Frasca F, Morrione A, Vigneri R. Insulin Receptor Isoforms in Physiology and Disease: An Updated View. Endocr Rev 2017; 38:379-431. [PMID: 28973479 PMCID: PMC5629070 DOI: 10.1210/er.2017-00073] [Citation(s) in RCA: 257] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 06/13/2017] [Indexed: 02/08/2023]
Abstract
The insulin receptor (IR) gene undergoes differential splicing that generates two IR isoforms, IR-A and IR-B. The physiological roles of IR isoforms are incompletely understood and appear to be determined by their different binding affinities for insulin-like growth factors (IGFs), particularly for IGF-2. Predominant roles of IR-A in prenatal growth and development and of IR-B in metabolic regulation are well established. However, emerging evidence indicates that the differential expression of IR isoforms may also help explain the diversification of insulin and IGF signaling and actions in various organs and tissues by involving not only different ligand-binding affinities but also different membrane partitioning and trafficking and possibly different abilities to interact with a variety of molecular partners. Of note, dysregulation of the IR-A/IR-B ratio is associated with insulin resistance, aging, and increased proliferative activity of normal and neoplastic tissues and appears to sustain detrimental effects. This review discusses novel information that has generated remarkable progress in our understanding of the physiology of IR isoforms and their role in disease. We also focus on novel IR ligands and modulators that should now be considered as an important strategy for better and safer treatment of diabetes and cancer and possibly other IR-related diseases.
Collapse
Affiliation(s)
- Antonino Belfiore
- Endocrinology, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy
| | - Roberta Malaguarnera
- Endocrinology, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy
| | - Veronica Vella
- School of Human and Social Sciences, University Kore of Enna, via della Cooperazione, 94100 Enna, Italy
| | - Michael C. Lawrence
- Structural Biology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Laura Sciacca
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania, Italy
| | - Francesco Frasca
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania, Italy
| | - Andrea Morrione
- Department of Urology and Biology of Prostate Cancer Program, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107
| | - Riccardo Vigneri
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania, Italy
| |
Collapse
|
26
|
Tuccori M, Convertino I, Galiulo MT, Marino A, Capogrosso-Sansone A, Blandizzi C. Diabetes drugs and the incidence of solid cancers: a survey of the current evidence. Expert Opin Drug Saf 2017; 16:1133-1148. [PMID: 28748718 DOI: 10.1080/14740338.2017.1361401] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The evaluation of the relationship between the use of antidiabetic drug and the occurrence of cancer is extremely challenging, both from the clinical and pharmacoepidemiological standpoint. This narrative review described the current evidence supporting a relationship between the use of antidiabetic drugs and the incidence of solid cancers. Areas covered: Data from pharmacoepidemiological studies on cancer incidence were presented for the main antidiabetic drugs and drug classes, including human insulin and insulin analogues, metformin, sulfonylureas, glinides, alpha-glucosidase inhibitors, thiazolidinediones, incretin mimetics, and sodium glucose co-transporter 2 inhibitors. The relationship between the use of antidiabetics and the incidence of solid cancer was described in strata by any cancer and by organ-specific cancer and by drug and by drug classes. Information supporting biological evidence and putative mechanisms were also provided. Expert opinion: The history of exploration of the relationship between antidiabetic drugs and the risk of solid cancers has showed several issues. Unrecognized biases and misinterpretations of study results have had important consequences that delayed the identification of actual risk and benefits of the use of antidiabetic drugs associated with cancer occurrence or progression. The lesson learned from the past should address the future research in this area, since in the majority of cases findings are controversial and confirmatory studies are warranted.
Collapse
Affiliation(s)
- Marco Tuccori
- a Unit of Adverse Drug Reaction Monitoring , University Hospital of Pisa , Pisa , Italy
| | - Irma Convertino
- b Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| | - Maria Teresa Galiulo
- b Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| | - Alessandra Marino
- b Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| | | | - Corrado Blandizzi
- a Unit of Adverse Drug Reaction Monitoring , University Hospital of Pisa , Pisa , Italy.,b Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| |
Collapse
|
27
|
Mishra S, Nyomba BG. Prohibitin - At the crossroads of obesity-linked diabetes and cancer. Exp Biol Med (Maywood) 2017; 242:1170-1177. [PMID: 28399645 DOI: 10.1177/1535370217703976] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The promoter of a gene that is selectively expressed in just a few cell types provides unique opportunities to study: (1) the pleiotropic function of a protein in two different cell types including the cell compartment specific function, and (2) the crosstalk between two cell/tissue types at the systemic level. This is not possible with a ubiquitous or a highly specific gene promoter. The adipocyte protein-2 ( aP2) is one such gene. It is primarily expressed in adipocytes, but also selectively in monocytic macrophages and dendritic cells, among various immune cell types. Thus, the adipocyte protein-2 gene promoter provides an opportunity to simultaneously manipulate adipose and immune functions in a transgenic animal. Prohibitin (PHB) is a pleiotropic protein that has roles in both adipocytes and immune cells. Adipocyte specific functions of prohibitin are mediated through its mitochondrial function, whereas its immune functions are mediated in a phosphorylation-dependent manner. We capitalized on this attribute of prohibitin to explore the crosstalk between adipose and immune functions, and to discern mitochondrial and plasma membrane-associated cell signaling functions of prohibitin, by expressing wild type prohibitin (Mito-Ob) and a phospho-mutant form of prohibitin (m-Mito-Ob) from the protein-2 gene promoter, individually. Both transgenic mice develop obesity in a sex-neutral manner, but develop obesity-related metabolic dysregulation in a male sex-specific manner. Subsequently, the male Mito-Ob mice spontaneously developed type 2 diabetes and liver cancer, whereas the male m-Mito-Ob mice developed lymph node tumors or autoimmune diabetes in a context-dependent manner. This review provides a point of view on the role of prohibitin in mediating sex differences in adipose and immune functions at the systemic level. We discuss the unique attributes of prohibitin and provide a new paradigm in adipose-immune crosstalk mediated through a pleiotropic protein. Impact statement Prohibitin (PHB) is ubiquitously expressed and plays a role in adipocyte-immune cell cross-talk. Both male and female transgenic mice expressing wild-type PHB in adipose tissue and in macrophages are obese, but only males develop diabetes and liver cancer. When the mice express PHB mutated on tyrosine-114 in adipocytes and macrophages, both males and females are still obese, but none develops liver cancer; instead, males develop lymph node tumors. Adipocyte specific functions of PHB are mediated through its mitochondrial function, whereas its immune functions are mediated in a phosphorylation-dependent manner. Thus, PHB appears to be an important molecule linking obesity, diabetes, and cancer. In addition, this link appears to be affected by sex steroids. Therefore, targeting PHB may lead to a better understanding of the pathogenesis of obesity, diabetes and cancer.
Collapse
Affiliation(s)
- Suresh Mishra
- 1 Department of Internal Medicine, University of Manitoba, Winnipeg R3E3P4, Canada.,2 Department of Physiology & Pathophysiology, University of Manitoba, Winnipeg R3E3P4, Canada
| | - Bl Grégoire Nyomba
- 1 Department of Internal Medicine, University of Manitoba, Winnipeg R3E3P4, Canada
| |
Collapse
|
28
|
Ter Braak B, Siezen CL, Lee JS, Rao P, Voorhoeve C, Ruppin E, van der Laan JW, van de Water B. Insulin-like growth factor 1 receptor activation promotes mammary gland tumor development by increasing glycolysis and promoting biomass production. Breast Cancer Res 2017; 19:14. [PMID: 28173837 PMCID: PMC5297135 DOI: 10.1186/s13058-017-0802-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 01/05/2017] [Indexed: 01/13/2023] Open
Abstract
Background The insulin-like growth factor 1 (IGF1) signaling axis plays a major role in tumorigenesis. In a previous experiment, we chronically treated mice with several agonists of the IGF1 receptor (IGF1R). We found that chronic treatment with insulin analogues with high affinity towards the IGF1R (IGF1 and X10) decreased the mammary gland tumor latency time in a p53R270H/+WAPCre mouse model. Frequent injections with insulin analogues that only mildly activated the IGF1R in vivo (glargine and insulin) did not significantly decrease the tumor latency time in this mouse model. Methods Here, we performed next-generation RNA sequencing (40 million, 100 bp reads) on 50 mammary gland tumors to unravel the underlying mechanisms of IGF1R-promoted tumorigenesis. Mutational profiling of the individual tumors was performed to screen for treatment-specific mutations. The transcriptomic data were used to construct a support vector machine (SVM) classifier so that the phenotypic characteristics of tumors exposed to the different insulin analogue treatments could be predicted. For translational purposes, we ran the same classifiers on transcriptomic (micro-array) data of insulin analogue-exposed human breast cancer cell lines. Genome-scale metabolic modeling was performed with iMAT. Results We found that chronic X10 and IGF1 treatment resulted in tumors with an increased and sustained proliferative and invasive transcriptomic profile. Furthermore, a Warburg-like effect with increased glycolysis was observed in tumors of the X10/IGF1 groups and, to a lesser extent, also in glargine-induced tumors. A metabolic flux analysis revealed that this enhanced glycolysis programming in X10/IGF1 tumors was associated with increased biomass production programs. Although none of the treatments induced genetic instability or enhanced mutagenesis, mutations in Ezh2 and Hras were enriched in X10/IGF1 treatment tumors. Conclusions Overall, these data suggest that the decreased mammary gland tumor latency time caused by chronic IGF1R activation is related to modulation of tumor progression rather than increased tumor initiation. Electronic supplementary material The online version of this article (doi:10.1186/s13058-017-0802-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bas Ter Braak
- Division of Toxicology, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333, CC, Leiden, The Netherlands
| | - Christine L Siezen
- Medicines Evaluation Board (MEB), Graadt van Roggenweg 500, 3531, AH, Utrecht, The Netherlands
| | - Joo S Lee
- The Blavatnik School of Computer Science, Tel-Aviv University, Tel Aviv, 69978, Israel
| | - Pooja Rao
- ServiceXS, Plesmanlaan 1 /D, 2333, BZ, Leiden, The Netherlands
| | - Charlotte Voorhoeve
- Division of Toxicology, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333, CC, Leiden, The Netherlands
| | - Eytan Ruppin
- The Blavatnik School of Computer Science, Tel-Aviv University, Tel Aviv, 69978, Israel
| | - Jan Willem van der Laan
- Division of Toxicology, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333, CC, Leiden, The Netherlands. .,Medicines Evaluation Board (MEB), Graadt van Roggenweg 500, 3531, AH, Utrecht, The Netherlands. .,Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721, MA, Bilthoven, The Netherlands.
| | - Bob van de Water
- Division of Toxicology, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333, CC, Leiden, The Netherlands.
| |
Collapse
|
29
|
Li P, Zhou H, Di G, Liu J, Liu Y, Wang Z, Sun Y, Duan H, Sun J. Mesenchymal stem cell-conditioned medium promotes MDA-MB-231 cell migration and inhibits A549 cell migration by regulating insulin receptor and human epidermal growth factor receptor 3 phosphorylation. Oncol Lett 2017; 13:1581-1586. [PMID: 28454294 PMCID: PMC5403177 DOI: 10.3892/ol.2017.5641] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 10/18/2016] [Indexed: 01/10/2023] Open
Abstract
Various in vitro and in vivo studies have linked mesenchymal stem cells (MSCs) with cancer, but little is known about the effect of MSCs on tumor progression. The present study aimed to analyze the role of the MSCs from different tissues, consisting of human bone marrow, adipose and the umbilical cord tissues, and the heterogeneity of tumors in tumor progression. By collecting the culture supernatants of MSCs as MSC-conditioned media (CMs), the present study found that MSC-CM produces no significant effect on the proliferation of MDA-MB-231 and A549 tumor cells. The migration of MDA-MB-231 cells was enhanced upon incubation with MSC-CM, while that of A549 cells was inhibited. Furthermore, the phosphorylation of insulin receptors (IRs) was upregulated in MSC-CM-treated MDA-MB-231 cells, while in MSC-CM-treated A549 cells, the phosphorylation of human epidermal growth factor receptor 3 (Her3) was downregulated. Taken together, the findings suggest that the phosphorylation of IR and Her3 may contribute to the discrepant effects of MSC-CM on the migration of the 2 cell lines.
Collapse
Affiliation(s)
- Pengfei Li
- Department of Oncology, Liaoning Medical College, Jinzhou, Liaoning 121000, P.R. China.,Department of Hematology and Oncology, The First Affiliated Hospital of General Hospital of Chinese People's Liberation Army, Beijing 100039, P.R. China.,Department of Hematology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Hongwei Zhou
- Department of Hematology and Oncology, The First Affiliated Hospital of General Hospital of Chinese People's Liberation Army, Beijing 100039, P.R. China
| | - Guohu Di
- Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
| | - Jin Liu
- Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
| | - Yang Liu
- Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
| | - Zhihong Wang
- Department of Hematology and Oncology, The First Affiliated Hospital of General Hospital of Chinese People's Liberation Army, Beijing 100039, P.R. China
| | - Yinxuan Sun
- The School of Management, South China University of Technology, Guangzhou, Guangdong 510000, P.R. China
| | - Haifeng Duan
- Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
| | - Junzhong Sun
- Department of Oncology, Liaoning Medical College, Jinzhou, Liaoning 121000, P.R. China.,Department of Hematology and Oncology, The First Affiliated Hospital of General Hospital of Chinese People's Liberation Army, Beijing 100039, P.R. China
| |
Collapse
|
30
|
Pan F, Hong LQ. Insulin promotes proliferation and migration of breast cancer cells through the extracellular regulated kinase pathway. Asian Pac J Cancer Prev 2017; 15:6349-52. [PMID: 25124623 DOI: 10.7314/apjcp.2014.15.15.6349] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The present study was undertaken to determine the roles of insulin in the growth of transplanted breast cancer in nude mice, and the proliferation and migration of MCF-7 human breast cancer cells and assess its influence on downstream signaling pathways. In a xenograft mouse model with injection of MCF-7 human breast cancer cells, tumor size was measured every other day. The insulin level and insulin receptor (IR) were increased in the breast cancer patient tissues. Insulin injected subcutaneously around the tumor site in mice caused increase in the size and weight of tumor masses, and promoted proliferation and migration of MCF-7 cells. The effects of insulin on the increase in the proliferation and migration of MCF-7 human breast cancer cells were abolished by pretreatment with the extracellular regulated kinase (ERK) inhibitor PD98059. Insulin increased the phosphorylation of ERK in the MCF-7 cells. These results indicate that insulin promotes the growth of breast cancer in nude mice, and increases the proliferation and migration of MCF-7 human breast cancer cells via the ERK pathway.
Collapse
Affiliation(s)
- Feng Pan
- Department of Laboratory Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China E-mail :
| | | |
Collapse
|
31
|
Mu L, Zhu N, Zhang J, Xing F, Li D, Wang X. Type 2 diabetes, insulin treatment and prognosis of breast cancer. Diabetes Metab Res Rev 2017; 33. [PMID: 27184049 DOI: 10.1002/dmrr.2823] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 04/21/2016] [Accepted: 05/09/2016] [Indexed: 12/29/2022]
Abstract
BACKGROUND The aim of our study was to investigate whether pre-existing type 2 diabetes and insulin therapy have an impact on the prognosis of breast cancer patients. METHODS We performed a retrospective analysis of 462 type 2 diabetic breast cancer patients and 1644 non-diabetic breast cancer patients treated in our institute from January 2005 to August 2010. Patients were divided by diabetes status and insulin use. The clinicopathological characteristics and clinical outcomes of patients within 5 years following breast cancer diagnosed were analysed. RESULTS Diabetic patients tended to have higher body mass index and higher histological grade tumours. Five-year disease-free survival and overall survival were reduced in diabetic patients (P < 0.001), and diabetes was an independent predictor for an increased risk of breast cancer relapse and death within 5 years (P < 0.001). Insulin treatment was associated with reduced 5-year disease-free survival and overall survival (P < 0.05); the risk of 5-year relapse and breast cancer mortality in the insulin group increased compared to that of non-insulin group after adjusting for age, tumour size, histological grade, oestrogen receptor, progesterone receptor, chemotherapy and hormone therapy (P < 0.05). After adjusting for age and other factors, the risk of breast cancer relapse was also increased in the insulin subgroup, while the risk of breast cancer mortality did not increase statistically. CONCLUSIONS Type 2 diabetes and insulin treatment might be independently associated with poorer prognosis of breast cancer. However, caution is needed when interpreting our results, and further investigations are needed. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Lan Mu
- First Department of Breast Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Nannan Zhu
- 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital, Tianjin Medical University, Tianjin, China
| | - Jiao Zhang
- First Department of Breast Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Fang Xing
- First Department of Breast Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Daiqing Li
- 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital, Tianjin Medical University, Tianjin, China
| | - Xin Wang
- First Department of Breast Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| |
Collapse
|
32
|
Malaguarnera R, Vella V, Nicolosi ML, Belfiore A. Insulin Resistance: Any Role in the Changing Epidemiology of Thyroid Cancer? Front Endocrinol (Lausanne) 2017; 8:314. [PMID: 29184536 PMCID: PMC5694441 DOI: 10.3389/fendo.2017.00314] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 10/30/2017] [Indexed: 12/13/2022] Open
Abstract
In the past few decades, the incidence of thyroid cancer (TC), namely of its papillary hystotype (PTC), has shown a steady increase worldwide, which has been attributed at least in part to the increasing diagnosis of early stage tumors. However, some evidence suggests that environmental and lifestyle factors can also play a role. Among the potential risk factors involved in the changing epidemiology of TC, particular attention has been drawn to insulin-resistance and related metabolic disorders, such as obesity, type 2 diabetes, and metabolic syndrome, which have been also rapidly increasing worldwide due to widespread dietary and lifestyle changes. In accordance with this possibility, various epidemiological studies have indeed gathered substantial evidence that insulin resistance-related metabolic disorders might be associated with an increased TC risk either through hyperinsulinemia or by affecting other TC risk factors including iodine deficiency, elevated thyroid stimulating hormone, estrogen-dependent signaling, chronic autoimmune thyroiditis, and others. This review summarizes the current literature evaluating the relationship between metabolic disorders characterized by insulin resistance and the risk for TC as well as the possible underlying mechanisms. The potential implications of such association in TC prevention and therapy are discussed.
Collapse
Affiliation(s)
- Roberta Malaguarnera
- Endocrinology, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Veronica Vella
- School of Human and Social Sciences, “Kore” University of Enna, Enna, Italy
- *Correspondence: Veronica Vella, ; Antonino Belfiore,
| | - Maria Luisa Nicolosi
- Endocrinology, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Antonino Belfiore
- Endocrinology, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
- *Correspondence: Veronica Vella, ; Antonino Belfiore,
| |
Collapse
|
33
|
Gallagher EJ, Zelenko Z, Tobin-Hess A, Werner U, Tennagels N, LeRoith D. Non-metabolisable insulin glargine does not promote breast cancer growth in a mouse model of type 2 diabetes. Diabetologia 2016; 59:2018-25. [PMID: 27241182 PMCID: PMC4970885 DOI: 10.1007/s00125-016-4000-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 05/03/2016] [Indexed: 12/29/2022]
Abstract
AIMS/HYPOTHESIS Previous epidemiological studies have reported a potential link between insulin analogues and breast cancer; however, a prospective randomised controlled trial showed neutral effects of insulin glargine on cancer risk. Insulin glargine is metabolised in vivo to an M1 metabolite. A question remains whether a subset of individuals with slower rates of glargine metabolism or who are on high doses could, theoretically, have an increased risk of cancer progression if a tumour is already present. In this study, we aimed to determine whether a non-metabolisable form of insulin glargine induced murine breast cancer growth. METHODS A mouse model of type 2 diabetes (MKR) was used for these studies. MKR mice were injected with two murine mammary cancer cell lines: Mvt-1 cells (derived from MMTV-c-Myc/Vegf tumours) and Met1 cells (derived from MMTV-polyoma virus middle T antigen tumours). Mice were treated with 25 U/kg per day of the long-acting insulin analogues, insulin glargine, insulin detemir, insulin degludec or non-metabolisable glargine, or vehicle. RESULTS No difference in tumour growth was seen in terms of tumour size after insulin glargine, detemir, degludec or vehicle injections. Non-metabolisable glargine did not increase tumour growth compared with insulin glargine or vehicle. Insulin glargine and non-metabolisable glargine led to insulin receptor phosphorylation in vivo rather than IGF-1 receptor phosphorylation. CONCLUSIONS/INTERPRETATION These results demonstrate that in a mouse model of type 2 diabetes, at high concentrations, basal insulin analogues and a non-metabolisable glargine analogue do not promote the progression of breast tumours.
Collapse
Affiliation(s)
- Emily J Gallagher
- Division of Endocrinology, Diabetes and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, Box 1055, New York, NY, 10029, USA.
| | - Zara Zelenko
- Division of Endocrinology, Diabetes and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, Box 1055, New York, NY, 10029, USA
| | - Aviva Tobin-Hess
- Division of Endocrinology, Diabetes and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, Box 1055, New York, NY, 10029, USA
| | - Ulrich Werner
- Diabetes Research & Translational Medicine, Insulin Biology, Sanofi-Aventis Deutschland GmbH, Frankfurt am Main, Germany
| | - Norbert Tennagels
- Diabetes Research & Translational Medicine, Insulin Biology, Sanofi-Aventis Deutschland GmbH, Frankfurt am Main, Germany
| | - Derek LeRoith
- Division of Endocrinology, Diabetes and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, Box 1055, New York, NY, 10029, USA
| |
Collapse
|
34
|
Ferroni P, Riondino S, Laudisi A, Portarena I, Formica V, Alessandroni J, D'Alessandro R, Orlandi A, Costarelli L, Cavaliere F, Guadagni F, Roselli M. Pretreatment Insulin Levels as a Prognostic Factor for Breast Cancer Progression. Oncologist 2016; 21:1041-9. [PMID: 27388232 DOI: 10.1634/theoncologist.2015-0462] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 02/09/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Based on the hypothesis that impaired glucose metabolism might be associated with survival outcomes independently of overt diabetes, we sought to investigate the prognostic value of routinely used glycemic parameters in a prospective study of breast cancer (BC) patients. PATIENTS AND METHODS Fasting blood glucose, insulin and HbA1c levels, and insulin resistance (assessed by the Homeostasis Model Assessment [HOMA] index) at diagnosis were evaluated in 286 nondiabetic BC patients (249 with primary cancer, 37 with metastatic) with respect to those parameters' possible associations with clinicopathological features and survival outcomes. As a control group, 143 healthy women matched in a 2:1 ratio for age, blood lipid levels, and body mass index were also investigated. RESULTS Fasting blood glucose level (mean ± SD: 99 ± 26 vs. 85 ± 15 mg/dL), insulin level (median: 10.0 vs. 6.8 μIU/mL), and HOMA index (median: 2.2 vs. 1.4), but not HbA1c level, were significantly elevated in BC patients compared with control subjects. Receiver operating characteristics analysis showed comparable areas for blood glucose and insulin levels, and HOMA index (ranging from 0.668 to 0.671). Using a cutoff level of 13 μIU/mL, insulin had the best specificity (92%) and sensitivity (41%), was significantly associated with disease stage, and acted as a negative prognostic marker of progression-free survival (hazard ratio: 2.17; 95% confidence interval: 1.13-4.20) independently of menopausal status, disease stage, hormone receptor status, and human epidermal growth factor receptor 2 and Ki67 expression. CONCLUSION These results suggest that insulin determination might provide prognostic information in BC and support the hypothesis that lifestyle and/or pharmacological interventions targeting glucose metabolism could be considered to improve survival outcome of selected BC patients. IMPLICATIONS FOR PRACTICE Pretreatment insulin levels may represent a biomarker of adverse prognosis in nondiabetic women with breast cancer, independently of other well-established prognostic factors (i.e., stage, hormone receptors, HER2/neu, and Ki67). This finding has important implications, because it provides the rationale for lifestyle or insulin-targeting pharmacologic interventions as a means of improving breast cancer outcomes not only in early stages, but also in advanced-stage breast cancer patients with aggressive tumor phenotypes (HER2-negative hormone-resistant, or triple-negative breast cancer), in which treatments are still challenging. The possibility of using insulin as a biomarker to guide insulin-targeted interventions also should be taken into account.
Collapse
Affiliation(s)
| | - Silvia Riondino
- Interinstitutional Multidisciplinary Biobank, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Pisana, Rome, Italy Department of Systems Medicine, Medical Oncology, Tor Vergata Clinical Center, Tor Vergata University of Rome, Rome, Italy
| | - Anastasia Laudisi
- Department of Systems Medicine, Medical Oncology, Tor Vergata Clinical Center, Tor Vergata University of Rome, Rome, Italy
| | - Ilaria Portarena
- Department of Systems Medicine, Medical Oncology, Tor Vergata Clinical Center, Tor Vergata University of Rome, Rome, Italy
| | - Vincenzo Formica
- Department of Systems Medicine, Medical Oncology, Tor Vergata Clinical Center, Tor Vergata University of Rome, Rome, Italy
| | - Jhessica Alessandroni
- Interinstitutional Multidisciplinary Biobank, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Pisana, Rome, Italy
| | - Roberta D'Alessandro
- Interinstitutional Multidisciplinary Biobank, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Pisana, Rome, Italy
| | - Augusto Orlandi
- Anatomic Pathology, Department of Biomedicine and Prevention, Tor Vergata University of Rome, Rome, Italy
| | | | | | - Fiorella Guadagni
- San Raffaele Roma Open University, Rome, Italy Interinstitutional Multidisciplinary Biobank, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Pisana, Rome, Italy
| | - Mario Roselli
- Department of Systems Medicine, Medical Oncology, Tor Vergata Clinical Center, Tor Vergata University of Rome, Rome, Italy
| |
Collapse
|
35
|
Flannery CA, Saleh FL, Choe GH, Selen DJ, Kodaman PH, Kliman HJ, Wood TL, Taylor HS. Differential Expression of IR-A, IR-B and IGF-1R in Endometrial Physiology and Distinct Signature in Adenocarcinoma. J Clin Endocrinol Metab 2016; 101:2883-91. [PMID: 27088794 PMCID: PMC4929835 DOI: 10.1210/jc.2016-1795] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 04/13/2016] [Indexed: 01/07/2023]
Abstract
CONTEXT Type 2 diabetes and obesity are risk factors for endometrial hyperplasia and cancer, suggesting that hyperinsulinemia contributes to pathogenesis. Insulin action through insulin receptor (IR) splice variants IR-A and IR-B regulates cellular mitogenesis and metabolism, respectively. OBJECTIVE We hypothesized that IR-A and IR-B are differentially regulated in normal endometrium, according to mitogenic and metabolic requirements through the menstrual cycle, as well as in endometrial hyperplasia and cancer. DESIGN IR-A, IR-B, and IGF-1 receptor (IGF-1R) mRNA was quantified in endometrium, endometrial epithelial and stromal cells, and in vitro after hormone stimulation. SETTING Academic center. PATIENTS Endometrium was collected from women with regular cycles (n = 71), complex hyperplasia (n = 5), or endometrioid adenocarcinoma (n = 11). INTERVENTION(S) In vitro sex-steroid treatment. MAIN OUTCOME MEASURE(S) IR-A and IR-B expression Results: IR-A increased dramatically during the early proliferative phase, 20-fold more than IR-B. In early secretory phase, IR-B and IGF-1R expression increased, reaching maximal expression, whereas IR-A decreased. In adenocarcinoma, IR-B and IGF-1R expression was 5- to 6-fold higher than normal endometrium, whereas IR-A expression was similar to IR-B. Receptor expression was unrelated to body mass index. CONCLUSION IR-A was elevated during the normal proliferative phase, and in endometrial hyperplasia and adenocarcinoma. The dramatic early rise of IR-A in normal endometrium indicates IR-A is the predominant isoform responsible for initial estrogen-independent endometrial proliferation as well as that of cancer. IR-B is elevated during the normal secretory phase when glucose uptake and glycogen synthesis support embryo development. Differing from other cancers, IR-B expression equals mitogenic IR-A in endometrial adenocarcinoma. Differential IR isoform expression suggests a distinct role for each in endometrial physiology and cancer.
Collapse
MESH Headings
- Adenocarcinoma/genetics
- Adenocarcinoma/metabolism
- Adenocarcinoma/pathology
- Adult
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Carcinoma, Endometrioid/genetics
- Carcinoma, Endometrioid/metabolism
- Carcinoma, Endometrioid/pathology
- Cells, Cultured
- Endometrial Hyperplasia/genetics
- Endometrial Hyperplasia/metabolism
- Endometrial Hyperplasia/pathology
- Endometrial Neoplasms/genetics
- Endometrial Neoplasms/metabolism
- Endometrial Neoplasms/pathology
- Endometrium/metabolism
- Endometrium/physiology
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Humans
- Menstrual Cycle/genetics
- Menstrual Cycle/metabolism
- Middle Aged
- Protein Subunits/genetics
- Protein Subunits/metabolism
- Receptor, IGF Type 1/genetics
- Receptor, IGF Type 1/metabolism
- Receptor, Insulin/genetics
- Receptor, Insulin/metabolism
- Transcriptome
Collapse
Affiliation(s)
- Clare A Flannery
- Obstetrics, Gynecology, and Reproductive Sciences (C.A.F., F.L.S., G.H.C., D.J.S., P.H.K., H.J.K., H.S.T.), Yale School of Medicine, New Haven, Connecticut 06520; Internal Medicine (C.A.F.), Yale School of Medicine, New Haven, Connecticut 06520; and Pharmacology, Physiology and Neuroscience and Cancer Center (T.L.W.), NJ Medical School, Rutgers University, Newark, New Jersey 07101
| | - Farrah L Saleh
- Obstetrics, Gynecology, and Reproductive Sciences (C.A.F., F.L.S., G.H.C., D.J.S., P.H.K., H.J.K., H.S.T.), Yale School of Medicine, New Haven, Connecticut 06520; Internal Medicine (C.A.F.), Yale School of Medicine, New Haven, Connecticut 06520; and Pharmacology, Physiology and Neuroscience and Cancer Center (T.L.W.), NJ Medical School, Rutgers University, Newark, New Jersey 07101
| | - Gina H Choe
- Obstetrics, Gynecology, and Reproductive Sciences (C.A.F., F.L.S., G.H.C., D.J.S., P.H.K., H.J.K., H.S.T.), Yale School of Medicine, New Haven, Connecticut 06520; Internal Medicine (C.A.F.), Yale School of Medicine, New Haven, Connecticut 06520; and Pharmacology, Physiology and Neuroscience and Cancer Center (T.L.W.), NJ Medical School, Rutgers University, Newark, New Jersey 07101
| | - Daryl J Selen
- Obstetrics, Gynecology, and Reproductive Sciences (C.A.F., F.L.S., G.H.C., D.J.S., P.H.K., H.J.K., H.S.T.), Yale School of Medicine, New Haven, Connecticut 06520; Internal Medicine (C.A.F.), Yale School of Medicine, New Haven, Connecticut 06520; and Pharmacology, Physiology and Neuroscience and Cancer Center (T.L.W.), NJ Medical School, Rutgers University, Newark, New Jersey 07101
| | - Pinar H Kodaman
- Obstetrics, Gynecology, and Reproductive Sciences (C.A.F., F.L.S., G.H.C., D.J.S., P.H.K., H.J.K., H.S.T.), Yale School of Medicine, New Haven, Connecticut 06520; Internal Medicine (C.A.F.), Yale School of Medicine, New Haven, Connecticut 06520; and Pharmacology, Physiology and Neuroscience and Cancer Center (T.L.W.), NJ Medical School, Rutgers University, Newark, New Jersey 07101
| | - Harvey J Kliman
- Obstetrics, Gynecology, and Reproductive Sciences (C.A.F., F.L.S., G.H.C., D.J.S., P.H.K., H.J.K., H.S.T.), Yale School of Medicine, New Haven, Connecticut 06520; Internal Medicine (C.A.F.), Yale School of Medicine, New Haven, Connecticut 06520; and Pharmacology, Physiology and Neuroscience and Cancer Center (T.L.W.), NJ Medical School, Rutgers University, Newark, New Jersey 07101
| | - Teresa L Wood
- Obstetrics, Gynecology, and Reproductive Sciences (C.A.F., F.L.S., G.H.C., D.J.S., P.H.K., H.J.K., H.S.T.), Yale School of Medicine, New Haven, Connecticut 06520; Internal Medicine (C.A.F.), Yale School of Medicine, New Haven, Connecticut 06520; and Pharmacology, Physiology and Neuroscience and Cancer Center (T.L.W.), NJ Medical School, Rutgers University, Newark, New Jersey 07101
| | - Hugh S Taylor
- Obstetrics, Gynecology, and Reproductive Sciences (C.A.F., F.L.S., G.H.C., D.J.S., P.H.K., H.J.K., H.S.T.), Yale School of Medicine, New Haven, Connecticut 06520; Internal Medicine (C.A.F.), Yale School of Medicine, New Haven, Connecticut 06520; and Pharmacology, Physiology and Neuroscience and Cancer Center (T.L.W.), NJ Medical School, Rutgers University, Newark, New Jersey 07101
| |
Collapse
|
36
|
Klement RJ, Fink MK. Dietary and pharmacological modification of the insulin/IGF-1 system: exploiting the full repertoire against cancer. Oncogenesis 2016; 5:e193. [PMID: 26878387 PMCID: PMC5154349 DOI: 10.1038/oncsis.2016.2] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 11/10/2015] [Accepted: 11/16/2015] [Indexed: 12/19/2022] Open
Abstract
As more and more links between cancer and metabolism are discovered, new approaches to treat cancer using these mechanisms are considered. Dietary restriction of either calories or macronutrients has shown great potential in animal studies to both reduce the incidence and growth of cancer, and to act synergistically with other treatment strategies. These studies have also shown that dietary restriction simultaneously targets many of the molecular pathways that are targeted individually by anticancer drugs. The insulin/insulin-like growth factor-1 (IGF-1) system has thereby emerged as a key regulator of cancer growth pathways. Although lowering of insulin levels with diet or drugs such as metformin and diazoxide seems generally beneficial, some practitioners also utilize strategic elevations of insulin levels in combination with chemotherapeutic drugs. This indicates a broad spectrum of possibilities for modulating the insulin/IGF-1 system in cancer treatment. With a specific focus on dietary restriction, insulin administration and the insulin-lowering drug diazoxide, such modifications of the insulin/IGF-1 system are the topic of this review. Although preclinical data are promising, we point out that insulin regulation and the metabolic response to a certain diet often differ between mice and humans. Thus, the need for collecting more human data has to be emphasized.
Collapse
Affiliation(s)
- R J Klement
- Department of Radiation Oncology, Leopoldina Hospital Schweinfurt, Schweinfurt, Germany
| | - M K Fink
- Onkologische Praxis, Fürth, Germany
| |
Collapse
|
37
|
Yan D, Han W, Dong Z, Liu Q, Jin Z, Chu D, Tian Y, Zhang J, Song D, Wang D, Zhu X. Homology modeling and docking studies of ENPP4: a BCG activated tumoricidal macrophage protein. Lipids Health Dis 2016; 15:19. [PMID: 26823374 PMCID: PMC4730737 DOI: 10.1186/s12944-016-0189-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 01/21/2016] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND The 3D structure and functions of ENPP4, a protein expressed on the surface of Bacillus Calmette-Guerin (BCG)-activated macrophages, are unknown. In this study, we analyzed the 3D structure of ENPP4 and determined its tumoricidal effects on MCA207 cells. RESULTS Homology modeling showed that Arg305, Tyr341, Asn291, and Asn295 are important residues in substrate, adenosine triphosphate (ATP), binding. A molecular dynamics study was also carried out to study the stability of ENPP4 (including zinc atoms) as well as its ligand-enzyme complex. BCG increased ENPP4 expression in macrophages, and specific blocking of ENPP4 in BCG-activated macrophages (BAMs) significantly reduced their cytotoxicity against MCA207 cells. CONCLUSIONS These results indicate that zinc remains inside the ENPP4 protein, a BCG activated tumoricidal macrophage protein, throughout the simulation. Important information for the design of new inhibitors was obtained.
Collapse
Affiliation(s)
- Dongmei Yan
- Department of Immunology, College of basic Medical sciences, Jilin University, Xinmin Street 126#, Changchun City, Jilin Province, China.
| | - Weiwei Han
- Key Laboratory for Molecular Enzymology and Engineering, Ministry of Education, Jilin University, Qianjin Street 2699#, Changchun City, Jilin Province, China.
| | - Zehua Dong
- Intensive Care Unit, The Affiliated Hospital of Qingdao University, Jiangsu Road 16#, Qingdao, China.
| | - Qihui Liu
- Department of Immunology, College of basic Medical sciences, Jilin University, Xinmin Street 126#, Changchun City, Jilin Province, China.
| | - Zheng Jin
- Department of Immunology, College of basic Medical sciences, Jilin University, Xinmin Street 126#, Changchun City, Jilin Province, China.
| | - Dong Chu
- Department of Immunology, College of basic Medical sciences, Jilin University, Xinmin Street 126#, Changchun City, Jilin Province, China.
| | - Yuan Tian
- Department of Immunology, College of basic Medical sciences, Jilin University, Xinmin Street 126#, Changchun City, Jilin Province, China.
| | - Jinpei Zhang
- Department of Immunology, College of basic Medical sciences, Jilin University, Xinmin Street 126#, Changchun City, Jilin Province, China.
| | - Dandan Song
- Department of Immunology, College of basic Medical sciences, Jilin University, Xinmin Street 126#, Changchun City, Jilin Province, China.
| | - Dunhuang Wang
- Department of Immunology, College of basic Medical sciences, Jilin University, Xinmin Street 126#, Changchun City, Jilin Province, China.
| | - Xun Zhu
- Department of Immunology, College of basic Medical sciences, Jilin University, Xinmin Street 126#, Changchun City, Jilin Province, China.
| |
Collapse
|
38
|
Ande SR, Nguyen KH, Padilla-Meier GP, Nyomba BLG, Mishra S. Expression of a mutant prohibitin from the aP2 gene promoter leads to obesity-linked tumor development in insulin resistance-dependent manner. Oncogene 2016; 35:4459-70. [PMID: 26751773 DOI: 10.1038/onc.2015.501] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 10/19/2015] [Accepted: 11/14/2015] [Indexed: 12/26/2022]
Abstract
A critical unmet need for the study of obesity-linked cancer is the lack of preclinical models that spontaneously develop obesity and cancer sequentially. Prohibitin (PHB) is a pleiotropic protein that has a role in adipose and immune functions. We capitalized on this attribute of PHB to develop a mouse model for obesity-linked tumor. We achieved this by expressing Y114F-PHB (m-PHB) from the aP2 gene promoter for simultaneous manipulation of adipogenic and immune signaling functions. The m-PHB mice develop obesity in a sex-neutral manner, but only male mice develop impaired glucose homeostasis and hyperinsulinemia similar to transgenic mice expressing PHB. Interestingly, only male m-PHB mice develop histiocytosis with lymphadenopathy, suggesting that metabolic dysregulation or m-PHB alone is not sufficient for the tumor development and that both are required for tumorigenesis. Moreover, ovariectomy in female m-PHB mice resulted in impaired glucose homeostasis, hyperinsulinemia and consequently tumor development similar to male m-PHB mice. These changes were not observed in sham-operated control m-Mito-Ob mice, further confirming the role of obesity-related metabolic dysregulation in tumor development in m-PHB mice. Our data provide a proof-of-concept that obesity-associated hyperinsulinemia promotes tumor development by facilitating dormant mutant to manifest and reveals a sex-dimorphic role of PHB in adipose-immune interaction or immunometabolism. Targeting PHB may provide a unique opportunity for the modulation of immunometabolism in obesity, cancer and in immune diseases.
Collapse
Affiliation(s)
- S R Ande
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - K H Nguyen
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - G P Padilla-Meier
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - B L G Nyomba
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - S Mishra
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada.,Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
39
|
Obesity and cancer, a case for insulin signaling. Cell Death Dis 2015; 6:e2037. [PMID: 26720346 PMCID: PMC4720912 DOI: 10.1038/cddis.2015.381] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 11/24/2015] [Accepted: 11/26/2015] [Indexed: 02/08/2023]
Abstract
Obesity is a worldwide epidemic, with the number of overweight and obese individuals climbing from just over 500 million in 2008 to 1.9 billion in 2014. Type 2 diabetes (T2D), cardiovascular disease and non-alcoholic fatty liver disease have long been associated with the obese state, whereas cancer is quickly emerging as another pathological consequence of this disease. Globally, at least 2.8 million people die each year from being overweight or obese. It is estimated that by 2020 being overweight or obese will surpass the health burden of tobacco consumption. Increase in the body mass index (BMI) in overweight (BMI>25 kg/m2) and obese (BMI>30 kg/m2) individuals is a result of adipose tissue (AT) expansion, which can lead to fat comprising >50% of the body weight in the morbidly obese. Extensive research over the last several years has painted a very complex picture of AT biology. One clear link between AT expansion and etiology of diseases like T2D and cancer is the development of insulin resistance (IR) and hyperinsulinemia. This review focuses on defining the link between obesity, IR and cancer.
Collapse
|
40
|
Abstract
Preclinical studies in the 1980s defined a role for IGF signaling in the development and sustainability of the malignant process. Subsequently, antibody, tyrosine kinase, and ligand inhibitors of the IGF receptor were manufactured. In the past decade, numerous clinical trials have tested the efficacy of IGF receptor inhibitors in the treatment of advanced tumors. Early-phase trials in heavily pretreated populations showed promise with complete or partial responses in a few patients and stable disease in many more. Unfortunately, the results of the early-phase trials did not pan out to later-phase trials. The lack of use of biomarkers to define subsets of patients that may benefit from IGF receptor blockade and compensatory signaling via other growth factor receptors such as the insulin, GH, and epidermal growth factor receptors may have played a role in the lack of efficacy of IGF receptor inhibition in phase III trials. Although these trials failed to show benefit, the trials have revealed previously unknown knowledge regarding the complex nature of IGF signaling. The knowledge obtained from these trials will be useful in designing future trials studying inhibitors of growth factor signaling.
Collapse
Affiliation(s)
- Heather Beckwith
- Departments of Medicine (H.B., D.Y.) and Pharmacology (D.Y.) and Masonic Cancer Center (D.Y.), University of Minnesota, Minneapolis, Minnesota 55455
| | - Douglas Yee
- Departments of Medicine (H.B., D.Y.) and Pharmacology (D.Y.) and Masonic Cancer Center (D.Y.), University of Minnesota, Minneapolis, Minnesota 55455
| |
Collapse
|
41
|
Gallagher EJ, LeRoith D. Obesity and Diabetes: The Increased Risk of Cancer and Cancer-Related Mortality. Physiol Rev 2015; 95:727-48. [PMID: 26084689 DOI: 10.1152/physrev.00030.2014] [Citation(s) in RCA: 520] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Obesity and type 2 diabetes are becoming increasingly prevalent worldwide, and both are associated with an increased incidence and mortality from many cancers. The metabolic abnormalities associated with type 2 diabetes develop many years before the onset of diabetes and, therefore, may be contributing to cancer risk before individuals are aware that they are at risk. Multiple factors potentially contribute to the progression of cancer in obesity and type 2 diabetes, including hyperinsulinemia and insulin-like growth factor I, hyperglycemia, dyslipidemia, adipokines and cytokines, and the gut microbiome. These metabolic changes may contribute directly or indirectly to cancer progression. Intentional weight loss may protect against cancer development, and therapies for diabetes may prove to be effective adjuvant agents in reducing cancer progression. In this review we discuss the current epidemiology, basic science, and clinical data that link obesity, diabetes, and cancer and how treating obesity and type 2 diabetes could also reduce cancer risk and improve outcomes.
Collapse
Affiliation(s)
| | - Derek LeRoith
- Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
42
|
Bronsveld HK, ter Braak B, Karlstad Ø, Vestergaard P, Starup-Linde J, Bazelier MT, De Bruin ML, de Boer A, Siezen CLE, van de Water B, van der Laan JW, Schmidt MK. Treatment with insulin (analogues) and breast cancer risk in diabetics; a systematic review and meta-analysis of in vitro, animal and human evidence. Breast Cancer Res 2015; 17:100. [PMID: 26242987 PMCID: PMC4531810 DOI: 10.1186/s13058-015-0611-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 07/07/2015] [Indexed: 12/29/2022] Open
Abstract
INTRODUCTION Several studies have suggested that anti-diabetic insulin analogue treatment might increase cancer risk. The aim of this study was to review the postulated association between insulin and insulin analogue treatment and breast cancer development, and plausible mechanisms. METHOD A systematic literature search was performed on breast cell-line, animal and human studies using the key words 'insulin analogue' and 'breast neoplasia' in MEDLINE at PubMed, EMBASE, and ISI Web of Science databases. A quantitative and qualitative review was performed on the epidemiological data; due to a limited number of reported estimates, a meta-analysis was performed for glargine only. A comprehensive overview was composed for in vitro and animal studies. Protein and gene expression was analysed for the cell lines most frequently used in the included in vitro studies. RESULTS In total 16 in vitro, 5 animal, 2 in vivo human and 29 epidemiological papers were included. Insulin AspB10 showed mitogenic properties in vitro and in animal studies. Glargine was the only clinically available insulin analogue for which an increased proliferative potential was found in breast cancer cell lines. However, the pooled analysis of 13 epidemiological studies did not show evidence for an association between insulin glargine treatment and an increased breast cancer risk (HR 1.04; 95 % CI 0.91-1.17; p=0.49) versus no glargine in patients with diabetes mellitus. It has to be taken into account that the number of animal studies was limited, and epidemiological studies were underpowered and suffered from methodological limitations. CONCLUSION There is no compelling evidence that any clinically available insulin analogue (Aspart, Determir, Glargine, Glulisine or Lispro), nor human insulin increases breast cancer risk. Overall, the data suggests that insulin treatment is not involved in breast tumour initiation, but might induce breast tumour progression by up regulating mitogenic signalling pathways.
Collapse
Affiliation(s)
- Heleen K Bronsveld
- Division of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
- Division of Pharmacoepidemiology & Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.
| | - Bas ter Braak
- Division of Toxicology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands.
| | - Øystein Karlstad
- Department of Pharmacoepidemiology, Norwegian Institute of Public Health, Oslo, Norway.
| | - Peter Vestergaard
- Departments of Clinical Medicine and Endocrinology, Aalborg University, Aalborg, Denmark.
| | - Jakob Starup-Linde
- Departments of Clinical Medicine and Endocrinology, Aalborg University, Aalborg, Denmark.
- Department of Endocrinology and Internal Medicine (MEA), Aarhus University Hospital THG, Aarhus, Denmark.
| | - Marloes T Bazelier
- Division of Pharmacoepidemiology & Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.
| | - Marie L De Bruin
- Division of Pharmacoepidemiology & Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.
| | - Anthonius de Boer
- Division of Pharmacoepidemiology & Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.
| | | | - Bob van de Water
- Division of Toxicology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands.
| | - Jan Willem van der Laan
- Division of Toxicology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands.
- Medicines Evaluation Board (MEB), Utrecht, The Netherlands.
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands.
| | - Marjanka K Schmidt
- Division of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
| |
Collapse
|
43
|
Nienhuis H, Gaykema S, Timmer-Bosscha H, Jalving M, Brouwers A, Lub-de Hooge M, van der Vegt B, Overmoyer B, de Vries E, Schröder C. Targeting breast cancer through its microenvironment: Current status of preclinical and clinical research in finding relevant targets. Pharmacol Ther 2015; 147:63-79. [DOI: 10.1016/j.pharmthera.2014.11.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 10/27/2014] [Indexed: 12/31/2022]
|
44
|
ter Braak B, Siezen C, Speksnijder EN, Koedoot E, van Steeg H, Salvatori DCF, van de Water B, van der Laan JW. Mammary gland tumor promotion by chronic administration of IGF1 and the insulin analogue AspB10 in the p53R270H/⁺WAPCre mouse model. Breast Cancer Res 2015; 17:14. [PMID: 25848982 PMCID: PMC4349771 DOI: 10.1186/s13058-015-0518-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 01/12/2015] [Indexed: 12/29/2022] Open
Abstract
INTRODUCTION Insulin analogues are structurally modified molecules with altered pharmaco-kinetic and -dynamic properties compared to regular human insulin used by diabetic patients. While these compounds are tested for undesired mitogenic effects, an epidemiological discussion is ongoing regarding an association between insulin analogue therapy and increased cancer incidence, including breast cancer. Standard in vivo rodent carcinogenesis assays do not pick up this possible increased carcinogenic potential. METHODS Here we studied the role of insulin analogues in breast cancer development. For this we used the human relevant mammary gland specific p53R270H/⁺WAPCre mouse model. Animals received life long repeated treatment with four different insulin (-like) molecules: normal insulin, insulin glargine, insulin X10 (AspB10) or insulin-like growth factor 1 (IGF1). RESULTS Insulin-like molecules with strong mitogenic signaling, insulin X10 and IGF1, significantly decreased the time for tumor development. Yet, insulin glargine and normal insulin, did not significantly decrease the latency time for (mammary gland) tumor development. The majority of tumors had an epithelial to mesenchymal transition phenotype (EMT), irrespective of treatment condition. Enhanced extracellular signaling related kinase (Erk) or serine/threonine kinase (Akt) mitogenic signaling was in particular present in tumors from the insulin X10 and IGF1 treatment groups. CONCLUSIONS These data indicate that insulin-like molecules with enhanced mitogenic signaling increase the risk of breast cancer development. Moreover, the use of a tissue specific cancer model, like the p53R270H/⁺WAPCre mouse model, is relevant to assess the intrinsic pro-carcinogenic potential of mitogenic and non-mitogenic biologicals such as insulin analogues.
Collapse
|
45
|
Salisbury TB, Tomblin JK. Insulin/Insulin-like growth factors in cancer: new roles for the aryl hydrocarbon receptor, tumor resistance mechanisms, and new blocking strategies. Front Endocrinol (Lausanne) 2015; 6:12. [PMID: 25699021 PMCID: PMC4313785 DOI: 10.3389/fendo.2015.00012] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 01/19/2015] [Indexed: 12/29/2022] Open
Abstract
The insulin-like growth factor 1 receptor (IGF1R) and the insulin receptor (IR) are receptor tyrosine kinases that are expressed in cancer cells. The results of different studies indicate that tumor proliferation and survival is dependent on the IGF1R and IR, and that their inhibition leads to reductions in proliferation and increases in cell death. Molecular targeting therapies that have been used in solid tumors include anti-IGF1R antibodies, anti-IGF1/IGF2 antibodies, and small molecule inhibitors that suppress IGF1R and IR kinase activity. New advances in the molecular basis of anti-IGF1R blocking antibodies reveal they are biased agonists and promote the binding of IGF1 to integrin β3 receptors in some cancer cells. Our recent reports indicate that pharmacological aryl hydrocarbon receptor (AHR) ligands inhibit breast cancer cell responses to IGFs, suggesting that targeting AHR may have benefit in cancers whose proliferation and survival are dependent on insulin/IGF signaling. Novel aspects of IGF1R/IR in cancer, such as biased agonism, integrin β3 signaling, AHR, and new therapeutic targeting strategies will be discussed.
Collapse
Affiliation(s)
- Travis B. Salisbury
- Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
- *Correspondence: Travis B. Salisbury, Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV 25755, USA e-mail:
| | - Justin K. Tomblin
- Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
| |
Collapse
|
46
|
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
|
47
|
Abstract
AIMS This review is aimed at highlighting the potential mitogenic/tumour growth-promoting or antimitogenic/tumour growth-inhibiting effects of the main antihyperglycaemic drug classes. METHODS We review and discuss the most current studies evaluating the association between antidiabetic medications used in clinical practice and malignancies as described so far. RESULTS Metformin seems to be the only antidiabetic drug to exert protective effects both on monotherapy and also when combined with other oral antidiabetic drugs or insulins in several site-specific cancers. In contrast, several other drug classes may increase cancer risk. Some reason for concern remains regarding sulphonylureas and also the incretin-based therapies regarding pancreas and thyroid cancers and the sodium glucose cotransporter-2 inhibitors as well as pioglitazone regarding bladder cancer. The majority of meta-analyses suggest that there is no evidence for a causal relationship between insulin glargine and elevated cancer risk, although the studies have been controversially discussed. For α-glucosidase inhibitors and glinides, neutral or only few data upon cancer risk exist. CONCLUSION Although the molecular mechanisms are not fully understood, a potential risk of mitogenicity and tumour growth promotion cannot be excluded in case of several antidiabetic drug classes. However, more large-scale, randomized, well-designed clinical studies with especially long follow-up time periods are needed to get reliable answers to these safety issues.
Collapse
Affiliation(s)
- Stefan Z Lutz
- Division of Endocrinology, Diabetology, Vascular Disease, Nephrology and Clinical Chemistry, Department of Internal Medicine, University of Tübingen, Tübingen, Germany German Centre for Diabetes Research (DZD), Tübingen, Germany
| | - Harald Staiger
- Division of Endocrinology, Diabetology, Vascular Disease, Nephrology and Clinical Chemistry, Department of Internal Medicine, University of Tübingen, Tübingen, Germany German Centre for Diabetes Research (DZD), Tübingen, Germany Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany
| | - Andreas Fritsche
- Division of Endocrinology, Diabetology, Vascular Disease, Nephrology and Clinical Chemistry, Department of Internal Medicine, University of Tübingen, Tübingen, Germany German Centre for Diabetes Research (DZD), Tübingen, Germany Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany Division of Nutritional and Preventive Medicine, Department of Internal Medicine, University of Tübingen, Tübingen, Germany
| | - Hans-Ulrich Häring
- Division of Endocrinology, Diabetology, Vascular Disease, Nephrology and Clinical Chemistry, Department of Internal Medicine, University of Tübingen, Tübingen, Germany German Centre for Diabetes Research (DZD), Tübingen, Germany Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany
| |
Collapse
|
48
|
Werner U, Korn M, Schmidt R, Wendrich TM, Tennagels N. Metabolic effect and receptor signalling profile of a non-metabolisable insulin glargine analogue. Arch Physiol Biochem 2014; 120:158-65. [PMID: 25144413 PMCID: PMC4219851 DOI: 10.3109/13813455.2014.950589] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
CONTEXT Insulin glargine (GLA) is rapidly metabolized in vivo to metabolite M1, which has in vitro metabolic and mitogenic profiles comparable with human insulin (HI). OBJECTIVE To investigate the pharmacologic and signalling profiles of a non-metabolizable analogue (A21Gly,DiD-Arg) insulin (D-GLA). METHODS Rats were injected s.c. with 1, 12.5 or 200 U/kg of GLA or D-GLA; blood glucose and phosphorylation status of the insulin receptor (IR), Akt and IGF-1 receptor (IGF1R) in tissue samples were investigated after 1 h. Plasma samples were analysed for insulin by LC-MS/MS. RESULTS Blood glucose lowering was prolonged with D-GLA. D-GLA comprised ≥98% of insulin after D-GLA injection; M1 comprised 76-92% after GLA injection. IR and Akt phosphorylation were comparable with GLA and D-GLA. Neither analogue stimulated IGF1R phosphorylation. CONCLUSIONS Suprapharmacological doses of D-GLA did not activate IGF1R in vivo. Mitogenic effects of insulin and insulin analogues might be solely based on IR growth-promoting activity.
Collapse
|
49
|
LaPorta E, Welsh J. Modeling vitamin D actions in triple negative/basal-like breast cancer. J Steroid Biochem Mol Biol 2014; 144 Pt A:65-73. [PMID: 24239860 PMCID: PMC4021002 DOI: 10.1016/j.jsbmb.2013.10.022] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 10/21/2013] [Accepted: 10/23/2013] [Indexed: 10/26/2022]
Abstract
Breast cancer is a heterogeneous disease with six molecularly defined subtypes, the most aggressive of which are triple negative breast cancers that lack expression of estrogen receptor (ER) and progesterone receptor (PR) and do not exhibit amplification of the growth factor receptor HER2. Triple negative breast cancers often exhibit basal-like gene signatures and are enriched for CD44+ cancer stem cells. In this report we have characterized the molecular actions of the VDR in a model of triple negative breast cancer. Estrogen independent, invasive mammary tumor cell lines established from wild-type (WT) and VDR knockout (VDRKO) mice were used to demonstrate that VDR is necessary for 1,25-dihydroxyvitamin D3 (1,25D) mediated anti-cancer actions in vitro and to identify novel targets of this receptor. Western blotting confirmed differential VDR expression and demonstrated the lack of ER, PR and Her2 in these cell lines. Re-introduction of human VDR (hVDR) into VDRKO cells restored the anti-proliferative actions of 1,25D. Genomic profiling demonstrated that 1,25D failed to alter gene expression in KO240 cells whereas major changes were observed in WT145 cells and in KO clones stably expressing hVDR (KO(hVDR) cells). With a 2-fold cutoff, 117 transcripts in WT145 cells and 197 transcripts in the KO(hVDR) clones were significantly altered by 1,25D. Thirty-five genes were found to be commonly regulated by 1,25D in all VDR-positive cell lines. Of these, we identified a cohort of four genes (Plau, Hbegf, Postn, Has2) that are known to drive breast cancer invasion and metastasis whose expression was markedly down regulated by 1,25D. These data support a model whereby 1,25D coordinately suppresses multiple proteins that are required for survival of triple-negative/basal-like breast cancer cells. Since studies have demonstrated a high prevalence of vitamin D deficiency in women with basal-like breast cancer, correction of vitamin D deficiency in these women represents a reasonable, but as yet untested, strategy to delay recurrence and extend survival. This article is part of a Special Issue entitled '16th Vitamin D Workshop'.
Collapse
Affiliation(s)
- Erika LaPorta
- Cancer Research Center, University at Albany, USA; Department of Biomedical Sciences, University at Albany, USA
| | - JoEllen Welsh
- Cancer Research Center, University at Albany, USA; Department of Environmental Health Sciences, University at Albany, USA.
| |
Collapse
|
50
|
Santoro MA, Andres SF, Galanko JA, Sandler RS, Keku TO, Lund PK. Reduced insulin-like growth factor I receptor and altered insulin receptor isoform mRNAs in normal mucosa predict colorectal adenoma risk. Cancer Epidemiol Biomarkers Prev 2014; 23:2093-100. [PMID: 25017244 DOI: 10.1158/1055-9965.epi-14-0177] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Hyperinsulinemia resulting from obesity and insulin resistance is associated with increased risk of many cancers, but the biology underlying this risk is unclear. We hypothesized that increased mRNA levels of the insulin-like growth factor I receptor (IGFIR) versus the insulin receptor (IR) or elevated ratio of IR-A:IR-B isoforms in normal rectal mucosa would predict adenoma risk, particularly in individuals with high body mass index (BMI) or plasma insulin. METHODS Biopsies from normal rectal mucosa were obtained from consenting patients undergoing routine colonoscopy at University of North Carolina Hospitals (Chapel Hill, NC). Subjects with colorectal adenomas were classified as cases (n = 100) and were matched to adenoma-free controls (n = 98) based on age, sex, and BMI. IGFIR and IR mRNA levels were assessed by qRT-PCR, and IR-A:IR-B mRNA ratios by standard PCR. Plasma insulin and crypt apoptosis were measured by ELISA and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL), respectively. Logistic regression models examined relationships between receptor mRNAs, BMI, plasma insulin, and adenoma risk. RESULTS Unexpectedly, cases were significantly more likely to have lower IGFIR mRNA levels than controls. No overall differences in total IR mRNA or IR-A:IR-B ratios were observed between cases and controls. Interestingly, in patients with high plasma insulin, increased IR-A:IR-B ratio was associated with increased likelihood of having adenomas. CONCLUSIONS Our work shows novel findings that reduced IGFIR mRNA and, during high plasma insulin, increased IR-A:IR-B ratios in normal rectal mucosa are associated with colorectal adenoma risk. IMPACT Our work provides evidence supporting a link between IGFIR and IR isoform expression levels and colorectal adenoma risk.
Collapse
Affiliation(s)
- M Agostina Santoro
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Sarah F Andres
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Joseph A Galanko
- Department of Medicine and Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Robert S Sandler
- Department of Medicine and Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Temitope O Keku
- Department of Medicine and Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - P Kay Lund
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
| |
Collapse
|