251
|
Suzuki S, McCarrey JR, Hermann BP. An mTORC1-dependent switch orchestrates the transition between mouse spermatogonial stem cells and clones of progenitor spermatogonia. Cell Rep 2021; 34:108752. [PMID: 33596419 PMCID: PMC7980622 DOI: 10.1016/j.celrep.2021.108752] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/06/2020] [Accepted: 01/25/2021] [Indexed: 12/22/2022] Open
Abstract
Spermatogonial stem cells (SSCs) sustain spermatogenesis by balancing self-renewal and initiation of differentiation to produce progenitor spermatogonia committed to forming sperm. To define the regulatory logic among SSCs and progenitors, we performed single-cell RNA velocity analyses and validated results in vivo. A predominant quiescent SSC population spawns a small subset of cell-cycle-activated SSCs via mitogen-activated protein kinase (MAPK)/AKT signaling. Activated SSCs form early progenitors and mTORC1 inhibition drives activated SSC accumulation consistent with blockade to progenitor formation. Mechanistically, mTORC1 inhibition suppresses transcription among spermatogonia and specifically alters expression of insulin growth factor (IGF) signaling in early progenitors. Tex14−/− testes lacking intercellular bridges do not accumulate activated SSCs following mTORC1 inhibition, indicating that steady-state mTORC1 signaling drives activated SSCs to produce progenitor clones. These results are consistent with a model of SSC self-renewal dependent on interconversion between activated and quiescent SSCs, and mTORC1-dependent initiation of differentiation from SSCs to progenitor clones. Suzuki et al. define relationships between subsets of adult mouse SSCs and progenitor spermatogonia using single-cell RNA velocity analyses and in vivo validations. Quiescent SCCs convert to cell-cycle-activated SCCs via MAPK/AKT signaling. Activated SCCs are driven to become early progenitor clones ready to initiate differentiation through mTORC1 signaling.
Collapse
Affiliation(s)
- Shinnosuke Suzuki
- Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - John R McCarrey
- Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Brian P Hermann
- Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249, USA.
| |
Collapse
|
252
|
Tatomir A, Beltrand A, Nguyen V, Boodhoo D, Mekala A, Cudrici C, Badea TC, Muresanu DF, Rus V, Rus H. RGC-32 Regulates Generation of Reactive Astrocytes in Experimental Autoimmune Encephalomyelitis. Front Immunol 2021; 11:608294. [PMID: 33569054 PMCID: PMC7868332 DOI: 10.3389/fimmu.2020.608294] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 12/08/2020] [Indexed: 12/31/2022] Open
Abstract
Astrocytes are increasingly recognized as critical contributors to multiple sclerosis pathogenesis. We have previously shown that lack of Response Gene to Complement 32 (RGC-32) alters astrocyte morphology in the spinal cord at the peak of experimental autoimmune encephalomyelitis (EAE), suggesting a role for RGC-32 in astrocyte differentiation. In this study, we analyzed the expression and distribution of astrocytes and astrocyte progenitors by immunohistochemistry in spinal cords of wild-type (WT) and RGC-32-knockout (KO) mice with EAE and of normal adult mice. Our analysis showed that during acute EAE, WT astrocytes had a reactive morphology and increased GFAP expression, whereas RGC-32 KO astrocytes had a morphology similar to that of radial glia and an increased expression of progenitor markers such as vimentin and fatty acid binding protein 7 (FABP7). In control mice, GFAP expression and astrocyte density were also significantly higher in the WT group, whereas the number of vimentin and FABP7-positive radial glia was significantly higher in the RGC-32 KO group. In vitro studies on cultured neonatal astrocytes from WT and RGC-32 KO mice showed that RGC-32 regulates a complex array of molecular networks pertaining to signal transduction, growth factor expression and secretion, and extracellular matrix (ECM) remodeling. Among the most differentially expressed factors were insulin-like growth factor 1 (IGF1), insulin-like growth factor binding proteins (IGFBPs), and connective tissue growth factor (CTGF); their expression was downregulated in RGC-32-depleted astrocytes. The nuclear translocation of STAT3, a transcription factor critical for astrogliogenesis and driving glial scar formation, was also impaired after RGC-32 silencing. Taken together, these data suggest that RGC-32 is an important regulator of astrocyte differentiation during EAE and that in the absence of RGC-32, astrocytes are unable to fully mature and become reactive astrocytes.
Collapse
MESH Headings
- Animals
- Astrocytes/metabolism
- Astrocytes/pathology
- Cell Differentiation
- Cell Movement
- Cell Proliferation
- Cells, Cultured
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Fatty Acid-Binding Protein 7/metabolism
- Female
- Glial Fibrillary Acidic Protein/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Phenotype
- Rats, Sprague-Dawley
- Signal Transduction
- Spinal Cord/metabolism
- Spinal Cord/pathology
- Vimentin/metabolism
- Mice
- Rats
Collapse
Affiliation(s)
- Alexandru Tatomir
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
- Department of Neurosciences, “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Austin Beltrand
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Vinh Nguyen
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Dallas Boodhoo
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Armugam Mekala
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Cornelia Cudrici
- Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Tudor C. Badea
- Retinal Circuit Development and Genetics Unit, Neurobiology Neurodegeneration & Repair Laboratory (N-NRL), National Eye Institute, Bethesda, MD, United States
| | - Dafin F. Muresanu
- Department of Neurosciences, “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Violeta Rus
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Horea Rus
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
- Research Service, Veterans Administration Maryland Health Care System, Baltimore, MD, United States
| |
Collapse
|
253
|
Boughanem H, Yubero-Serrano EM, López-Miranda J, Tinahones FJ, Macias-Gonzalez M. Potential Role of Insulin Growth-Factor-Binding Protein 2 as Therapeutic Target for Obesity-Related Insulin Resistance. Int J Mol Sci 2021; 22:ijms22031133. [PMID: 33498859 PMCID: PMC7865532 DOI: 10.3390/ijms22031133] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/13/2021] [Accepted: 01/21/2021] [Indexed: 12/26/2022] Open
Abstract
Evidence from observational and in vitro studies suggests that insulin growth-factor-binding protein type 2 (IGFBP2) is a promising protein in non-communicable diseases, such as obesity, insulin resistance, metabolic syndrome, or type 2 diabetes. Accordingly, great efforts have been carried out to explore the role of IGFBP2 in obesity state and insulin-related diseases, which it is typically found decreased. However, the physiological pathways have not been explored yet, and the relevance of IGFBP2 as an important pathway integrator of metabolic disorders is still unknown. Here, we review and discuss the molecular structure of IGFBP2 as the first element of regulating the expression of IGFBP2. We highlight an update of the association between low serum IGFBP2 and an increased risk of obesity, type 2 diabetes, metabolic syndrome, and low insulin sensitivity. We hypothesize mechanisms of IGFBP2 on the development of obesity and insulin resistance in an insulin-independent manner, which meant that could be evaluated as a therapeutic target. Finally, we cover the most interesting lifestyle modifications that regulate IGFBP2, since lifestyle factors (diet and/or physical activity) are associated with important variations in serum IGFBP2.
Collapse
Affiliation(s)
- Hatim Boughanem
- Department of Endocrinology and Nutrition, Institute of Biomedical Research Institute in Malaga (IBIMA), Virgen de la Victoria University Hospital, 29010 Málaga, Spain;
| | - Elena M. Yubero-Serrano
- Lipids and Atherosclerosis Unit, Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Reina Sofia University Hospital, University of Córdoba, 14004 Córdoba, Spain; (E.M.Y.-S.); (J.L.-M.)
- CIBEROBN (CIBER in Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - José López-Miranda
- Lipids and Atherosclerosis Unit, Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Reina Sofia University Hospital, University of Córdoba, 14004 Córdoba, Spain; (E.M.Y.-S.); (J.L.-M.)
- CIBEROBN (CIBER in Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Francisco J. Tinahones
- Department of Endocrinology and Nutrition, Institute of Biomedical Research Institute in Malaga (IBIMA), Virgen de la Victoria University Hospital, 29010 Málaga, Spain;
- CIBEROBN (CIBER in Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (F.J.T.); (M.M.-G.); Tel.: +34-951-036-2647 (F.J.T. & M.M.-G.); Fax: +34-951-924-651 (F.J.T. & M.M.-G.)
| | - Manuel Macias-Gonzalez
- Department of Endocrinology and Nutrition, Institute of Biomedical Research Institute in Malaga (IBIMA), Virgen de la Victoria University Hospital, 29010 Málaga, Spain;
- CIBEROBN (CIBER in Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (F.J.T.); (M.M.-G.); Tel.: +34-951-036-2647 (F.J.T. & M.M.-G.); Fax: +34-951-924-651 (F.J.T. & M.M.-G.)
| |
Collapse
|
254
|
Gharahdaghi N, Phillips BE, Szewczyk NJ, Smith K, Wilkinson DJ, Atherton PJ. Links Between Testosterone, Oestrogen, and the Growth Hormone/Insulin-Like Growth Factor Axis and Resistance Exercise Muscle Adaptations. Front Physiol 2021; 11:621226. [PMID: 33519525 PMCID: PMC7844366 DOI: 10.3389/fphys.2020.621226] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 12/18/2020] [Indexed: 12/13/2022] Open
Abstract
Maintenance of skeletal muscle mass throughout the life course is key for the regulation of health, with physical activity a critical component of this, in part, due to its influence upon key hormones such as testosterone, estrogen, growth hormone (GH), and insulin-like growth factor (IGF). Despite the importance of these hormones for the regulation of skeletal muscle mass in response to different types of exercise, their interaction with the processes controlling muscle mass remain unclear. This review presents evidence on the importance of these hormones in the regulation of skeletal muscle mass and their responses, and involvement in muscle adaptation to resistance exercise. Highlighting the key role testosterone plays as a primary anabolic hormone in muscle adaptation following exercise training, through its interaction with anabolic signaling pathways and other hormones via the androgen receptor (AR), this review also describes the potential importance of fluctuations in other hormones such as GH and IGF-1 in concert with dietary amino acid availability; and the role of estrogen, under the influence of the menstrual cycle and menopause, being especially important in adaptive exercise responses in women. Finally, the downstream mechanisms by which these hormones impact regulation of muscle protein turnover (synthesis and breakdown), and thus muscle mass are discussed. Advances in our understanding of hormones that impact protein turnover throughout life offers great relevance, not just for athletes, but also for the general and clinical populations alike.
Collapse
Affiliation(s)
| | | | | | | | - Daniel J. Wilkinson
- Medical Research Council-Versus Arthritis Centre for Musculoskeletal Ageing Research and Nottingham National Institute for Health Research Nottingham Biomedical Research Centre, School of Medicine, University of Nottingham, Derby, United Kingdom
| | - Philip J. Atherton
- Medical Research Council-Versus Arthritis Centre for Musculoskeletal Ageing Research and Nottingham National Institute for Health Research Nottingham Biomedical Research Centre, School of Medicine, University of Nottingham, Derby, United Kingdom
| |
Collapse
|
255
|
Öhman T, Gawriyski L, Miettinen S, Varjosalo M, Loukovaara S. Molecular pathogenesis of rhegmatogenous retinal detachment. Sci Rep 2021; 11:966. [PMID: 33441730 PMCID: PMC7806834 DOI: 10.1038/s41598-020-80005-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 12/16/2020] [Indexed: 02/08/2023] Open
Abstract
Rhegmatogenous retinal detachment (RRD) is an ophthalmic emergency, which usually requires prompt surgery to prevent further detachment and restore sensory function. Although several individual factors have been suggested, a systems level understanding of molecular pathomechanisms underlying this severe eye disorder is lacking. To address this gap in knowledge we performed the molecular level systems pathology analysis of the vitreous from 127 patients with RRD using state-of-the art quantitative mass spectrometry to identify the individual key proteins, as well as the biochemical pathways contributing to the development of the disease. RRD patients have specific vitreous proteome profiles compared to other diseases such as macular hole, pucker, or proliferative diabetic retinopathy eyes. Our data indicate that various mechanisms, including glycolysis, photoreceptor death, and Wnt and MAPK signaling, are activated during or after the RRD to promote retinal cell survival. In addition, platelet-mediated wound healing processes, cell adhesion molecules reorganization and apoptotic processes were detected during RRD progression or proliferative vitreoretinopathy formation. These findings improve the understanding of RRD pathogenesis, identify novel targets for treatment of this ophthalmic disease, and possibly affect the prognosis of eyes treated or operated upon due to RRD.
Collapse
Affiliation(s)
- Tiina Öhman
- Institute of Biotechnology and Helsinki Institute of Life Science, University of Helsinki, Viikinkaari 1, P.O. Box 65, 00014, Helsinki, Finland
| | - Lisa Gawriyski
- Institute of Biotechnology and Helsinki Institute of Life Science, University of Helsinki, Viikinkaari 1, P.O. Box 65, 00014, Helsinki, Finland
| | - Sini Miettinen
- Institute of Biotechnology and Helsinki Institute of Life Science, University of Helsinki, Viikinkaari 1, P.O. Box 65, 00014, Helsinki, Finland
| | - Markku Varjosalo
- Institute of Biotechnology and Helsinki Institute of Life Science, University of Helsinki, Viikinkaari 1, P.O. Box 65, 00014, Helsinki, Finland.
| | - Sirpa Loukovaara
- Department of Ophthalmology, Unit of Vitreoretinal Surgery, Helsinki University Hospital, Haartmaninkatu 4 C, 00290, Helsinki, Finland. .,Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
256
|
Suvorov A, Salemme V, McGaunn J, Poluyanoff A, Teffera M, Amir S. Unbiased approach for the identification of molecular mechanisms sensitive to chemical exposures. CHEMOSPHERE 2021; 262:128362. [PMID: 33182146 DOI: 10.1016/j.chemosphere.2020.128362] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/13/2020] [Accepted: 09/16/2020] [Indexed: 06/11/2023]
Abstract
Targeted methods that dominated toxicological research until recently did not allow for screening of all molecular changes involved in toxic response. Therefore, it is difficult to infer if all major mechanisms of toxicity have already been discovered, or if some of them are still overlooked. We used data on 591,084 unique chemical-gene interactions to identify genes and molecular pathways most sensitive to chemical exposures. The list of identified pathways did not change significantly when analyses were done on different subsets of data with non-overlapping lists of chemical compounds indicative that our dataset is saturated enough to provide unbiased results. One of the most important findings of this study is that almost every known molecular mechanism may be affected by chemical exposures. Predictably, xenobiotic metabolism pathways, and mechanisms of cellular response to stress and damage were among the most sensitive. Additionally, we identified highly sensitive molecular pathways, which are not widely recognized as major targets of toxicants, including lipid metabolism pathways, longevity regulation cascade, and cytokine-mediated signaling. These mechanisms are relevant to significant public health problems, such as aging, cancer, metabolic and autoimmune disease. Thus, public health field will benefit from future focus of toxicological research on identified sensitive mechanisms.
Collapse
Affiliation(s)
- Alexander Suvorov
- Department of Environmental Health Sciences, School of Public Health & Health Sciences, University of Massachusetts, 686 North Pleasant Street, Amherst, MA, 01003, USA.
| | - Victoria Salemme
- Department of Environmental Health Sciences, School of Public Health & Health Sciences, University of Massachusetts, 686 North Pleasant Street, Amherst, MA, 01003, USA
| | - Joseph McGaunn
- Department of Environmental Health Sciences, School of Public Health & Health Sciences, University of Massachusetts, 686 North Pleasant Street, Amherst, MA, 01003, USA
| | - Anthony Poluyanoff
- Department of Environmental Health Sciences, School of Public Health & Health Sciences, University of Massachusetts, 686 North Pleasant Street, Amherst, MA, 01003, USA
| | - Menna Teffera
- Department of Environmental Health Sciences, School of Public Health & Health Sciences, University of Massachusetts, 686 North Pleasant Street, Amherst, MA, 01003, USA
| | - Saira Amir
- Department of Environmental Health Sciences, School of Public Health & Health Sciences, University of Massachusetts, 686 North Pleasant Street, Amherst, MA, 01003, USA; Current Affiliation: Department of Biosciences, COMSATS University Islamabad, Pakistan
| |
Collapse
|
257
|
Rahman A, Hammad MM, Al Khairi I, Cherian P, Al-Sabah R, Al-Mulla F, Abu-Farha M, Abubaker J. Profiling of Insulin-Like Growth Factor Binding Proteins (IGFBPs) in Obesity and Their Association With Ox-LDL and Hs-CRP in Adolescents. Front Endocrinol (Lausanne) 2021; 12:727004. [PMID: 34394011 PMCID: PMC8355984 DOI: 10.3389/fendo.2021.727004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 07/15/2021] [Indexed: 12/17/2022] Open
Abstract
Insulin-like growth factor binding proteins (IGFBPs) are critical modulators of metabolism. In adults, IGFBPs are associated with obesity and insulin resistance. However, the association of IGFBPs with metabolic homeostasis in children and adolescents is not yet fully characterized. In this study we investigated the association of plasma IGFBPs (IGFBP-1, 3 and 7) with weight, central adiposity and cardiovascular disease markers Hs-CRP and Ox-LDL. A total of 420 adolescents (age 11-14 years) were recruited from public middle schools in Kuwait. IGFBPs were measured using bead-based multiplexing while Hs-CRP and Ox-LDL were measured using ELISA. Results showed that levels of IGFBP-1 were significantly lower in obese and overweight children when compared to normal weight children. Correlation analysis showed negative association between the level of IGFBP-1 and waist circumference to height (WC/Ht) ratio. IGFBP-1 level was also negatively associated with Hs-CRP. It was also observed that the levels of IGFBP-3 and IGFBP-7 were negatively correlated with Ox-LDL. Our data demonstrate a strong negative association of IGFBP-1 with overweight/obesity, and the inflammatory marker Hs-CRP. This was not seen with the levels of IGFBP-3 and 7. The association of IGFBP-1 with central adiposity (WC/Ht ratio) was stronger than its association with BMI-for-age z-score. Therefore we suggest that IGFBP-1 could potentially be used as a sensitive biomarker for obesity and its subsequent effects in screening and monitoring of obesity-related metabolic complications in adolescents.
Collapse
Affiliation(s)
- Abdur Rahman
- Department of Food Science and Nutrition, College of Life Sciences, Kuwait University, Kuwait City, Kuwait
| | - Maha M. Hammad
- Biochemistry and Molecular Biology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Irina Al Khairi
- Biochemistry and Molecular Biology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Preethi Cherian
- Biochemistry and Molecular Biology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Reem Al-Sabah
- Department of Community Medicine and Behavioural Sciences, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Fahd Al-Mulla
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Mohamed Abu-Farha
- Biochemistry and Molecular Biology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
- Special Services Department, Dasman Diabetes Institute, Kuwait City, Kuwait
- *Correspondence: Jehad Abubaker, ; Mohamed Abu-Farha, ;
| | - Jehad Abubaker
- Biochemistry and Molecular Biology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
- *Correspondence: Jehad Abubaker, ; Mohamed Abu-Farha, ;
| |
Collapse
|
258
|
Martín-Estal I, Castilla-Cortázar I, Castorena-Torres F. The Placenta as a Target for Alcohol During Pregnancy: The Close Relation with IGFs Signaling Pathway. Rev Physiol Biochem Pharmacol 2021; 180:119-153. [PMID: 34159446 DOI: 10.1007/112_2021_58] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Alcohol is one of the most consumed drugs in the world, even during pregnancy. Its use is a risk factor for developing adverse outcomes, e.g. fetal death, miscarriage, fetal growth restriction, and premature birth, also resulting in fetal alcohol spectrum disorders. Ethanol metabolism induces an oxidative environment that promotes the oxidation of lipids and proteins, triggers DNA damage, and advocates mitochondrial dysfunction, all of them leading to apoptosis and cellular injury. Several organs are altered due to this harmful behavior, the brain being one of the most affected. Throughout pregnancy, the human placenta is one of the most important organs for women's health and fetal development, as it secretes numerous hormones necessary for a suitable intrauterine environment. However, our understanding of the human placenta is very limited and even more restricted is the knowledge of the impact of toxic substances in its development and fetal growth. So, could ethanol consumption during this period have wounding effects in the placenta, compromising proper fetal organ development? Several studies have demonstrated that alcohol impairs various signaling cascades within G protein-coupled receptors and tyrosine kinase receptors, mainly through its action on insulin and insulin-like growth factor 1 (IGF-1) signaling pathway. This last cascade is involved in cell proliferation, migration, and differentiation and in placentation. This review tries to examine the current knowledge and gaps in our existing understanding of the ethanol effects in insulin/IGFs signaling pathway, which can explain the mechanism to elucidate the adverse actions of ethanol in the maternal-fetal interface of mammals.
Collapse
Affiliation(s)
- Irene Martín-Estal
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, NL, Mexico
| | | | | |
Collapse
|
259
|
Cleveland BM, Habara S, Oikawa J, Radler LM, Shimizu M. Compensatory Response of the Somatotropic Axis from IGFBP-2b Gene Editing in Rainbow Trout ( Oncorhynchus mykiss). Genes (Basel) 2020; 11:genes11121488. [PMID: 33322039 PMCID: PMC7763687 DOI: 10.3390/genes11121488] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 11/29/2020] [Accepted: 12/04/2020] [Indexed: 11/16/2022] Open
Abstract
Rainbow trout with gene editing-induced reductions in serum insulin-like growth factor binding protein (IGFBP)-2b exhibit similar growth performance compared to fish without IGFBP-2b gene disruption. The objective of this study is to determine how the components of the insulin-like growth factor (IGF)/IGFBP system respond to a reduction in serum IGFBP-2b abundance. Editing the IGFBP-2b genes in rainbow trout resulted in an 83% decrease in serum IGFBP-2b in mutants. This resulted in a 35% reduction in serum IGF-I, which was offset by reduced expression of hepatic igfbp-1a2 and increased muscle igfr-1a; these responses suggest that an increased IGF-I signaling capacity offset reductions in serum IGF-I. During feed deprivation, the differential expression of igfbp genes supports the attenuation of the growth inhibitory response, likely due to the further reduction in serum IGF-I that alleviated the need for an IGF-inhibitory response. Unique igfbp expression patterns occurred during refeeding, suggesting an enhanced IGF-I signaling capacity in controls. Collectively, these findings support that the role of IGFBP-2b is to regulate serum IGF-I concentrations. The compensatory regulation of IGF/IGFBP system genes indicates that adjustments in other IGFBP, both circulating and at the local level, maintain IGF-I signaling at a level appropriate for the nutritional state of the fish.
Collapse
Affiliation(s)
- Beth M. Cleveland
- National Center for Cool and Cold Water Aquaculture, United States Department of Agriculture/Agricultural Research Service, Leetown, WV 25430, USA;
- Correspondence: ; Tel.: +1304-724-8340 (ext. 2133)
| | - Shiori Habara
- Graduate School of Environmental Science, Hokkaido University, Sapporo 060-0808, Japan; (S.H.); (J.O.)
| | - Jin Oikawa
- Graduate School of Environmental Science, Hokkaido University, Sapporo 060-0808, Japan; (S.H.); (J.O.)
| | - Lisa M. Radler
- National Center for Cool and Cold Water Aquaculture, United States Department of Agriculture/Agricultural Research Service, Leetown, WV 25430, USA;
| | - Munetaka Shimizu
- Faculty of Fisheries Sciences, Hokkaido University, Hakodate 041-8611, Japan;
| |
Collapse
|
260
|
Liran M, Rahamim N, Ron D, Barak S. Growth Factors and Alcohol Use Disorder. Cold Spring Harb Perspect Med 2020; 10:cshperspect.a039271. [PMID: 31964648 DOI: 10.1101/cshperspect.a039271] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Neurotrophic growth factors were originally characterized for their support in neuronal differentiation, outgrowth, and survival during development. However, it has been acknowledged that they also play a vital role in the adult brain. Abnormalities in growth factors have been implicated in a variety of neurological and psychiatric disorders, including alcohol use disorder (AUD). This work focuses on the interaction between alcohol and growth factors. We review literature suggesting that several growth factors play a unique role in the regulation of alcohol consumption, and that breakdown in these growth factor systems is linked to the development of AUD. Specifically, we focus on the brain-derived neurotrophic factor (BDNF), glial cell line-derived neurotrophic factor (GDNF), fibroblast growth factor 2 (FGF2), and insulin growth factor 1 (IGF-1). We also review the literature on the potential role of midkine (MDK) and pleiotrophin (PTN) and their receptor, anaplastic lymphoma kinase (ALK), in AUD. We show that alcohol alters the expression of these growth factors or their receptors in brain regions previously implicated in addiction, and that manipulations on these growth factors and their downstream signaling can affect alcohol-drinking behaviors in animal models. We conclude that there is a need for translational and clinical research to assess the therapeutic potential of new pharmacotherapies targeting these systems.
Collapse
Affiliation(s)
- Mirit Liran
- Department of Neurobiology, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Nofar Rahamim
- Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Dorit Ron
- Department of Neurology, University of California, 675 Nelson Rising Lane, San Francisco, California 94143-0663, USA
| | - Segev Barak
- Department of Neurobiology, Tel Aviv University, 69978 Tel Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel.,School of Psychological Sciences, Tel Aviv University, 69978 Tel Aviv, Israel
| |
Collapse
|
261
|
Breves JP, Springer-Miller RH, Chenoweth DA, Paskavitz AL, Chang AYH, Regish AM, Einarsdottir IE, Björnsson BT, McCormick SD. Cortisol regulates insulin-like growth-factor binding protein (igfbp) gene expression in Atlantic salmon parr. Mol Cell Endocrinol 2020; 518:110989. [PMID: 32835784 DOI: 10.1016/j.mce.2020.110989] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 02/08/2023]
Abstract
The growth hormone (Gh)/insulin-like growth-factor (Igf)/Igf binding protein (Igfbp) system regulates growth and osmoregulation in salmonid fishes, but how this system interacts with other endocrine systems is largely unknown. Given the well-documented consequences of mounting a glucocorticoid stress response on growth, we hypothesized that cortisol inhibits anabolic processes by modulating the expression of hepatic igfbp mRNAs. Atlantic salmon (Salmo salar) parr were implanted intraperitoneally with cortisol implants (0, 10, and 40 μg g-1 body weight) and sampled after 3 or 14 days. Cortisol elicited a dose-dependent reduction in specific growth rate (SGR) after 14 days. While plasma Gh and Igf1 levels were unchanged, hepatic igf1 mRNA was diminished and hepatic igfbp1b1 and -1b2 were stimulated by the high cortisol dose. Plasma Igf1 was positively correlated with SGR at 14 days. Hepatic gh receptor (ghr), igfbp1a, -2a, -2b1, and -2b2 levels were not impacted by cortisol. Muscle igf2, but not igf1 or ghr, levels were stimulated at 3 days by the high cortisol dose. As both cortisol and the Gh/Igf axis promote seawater (SW) tolerance, and particular igfbps respond to SW exposure, we also assessed whether cortisol coordinates the expression of branchial igfbps and genes associated with ion transport. Cortisol stimulated branchial igfbp5b2 levels in parallel with Na+/K+-ATPase (NKA) activity and nka-α1b, Na+/K+/2Cl--cotransporter 1 (nkcc1), and cystic fibrosis transmembrane regulator 1 (cftr1) mRNA levels. The collective results indicate that cortisol modulates the growth of juvenile salmon via the regulation of hepatic igfbp1s whereas no clear links between cortisol and branchial igfbps previously shown to be salinity-responsive could be established.
Collapse
Affiliation(s)
- J P Breves
- Department of Biology, Skidmore College, 815 N. Broadway, Saratoga Springs, NY, 12866, USA.
| | - R H Springer-Miller
- Department of Biology, Skidmore College, 815 N. Broadway, Saratoga Springs, NY, 12866, USA
| | - D A Chenoweth
- Department of Biology, Skidmore College, 815 N. Broadway, Saratoga Springs, NY, 12866, USA
| | - A L Paskavitz
- Department of Biology, Skidmore College, 815 N. Broadway, Saratoga Springs, NY, 12866, USA
| | - A Y H Chang
- Department of Biology, Skidmore College, 815 N. Broadway, Saratoga Springs, NY, 12866, USA
| | - A M Regish
- U.S. Geological Survey, Leetown Science Center, Conte Anadromous Fish Research Laboratory, One Migratory Way, Turners Falls, MA, 01376, USA
| | - I E Einarsdottir
- Fish Endocrinology Laboratory, Department of Biological and Environmental Sciences, University of Gothenburg, Box 463 SE, 40530, Göteborg, Sweden
| | - B Th Björnsson
- Fish Endocrinology Laboratory, Department of Biological and Environmental Sciences, University of Gothenburg, Box 463 SE, 40530, Göteborg, Sweden
| | - S D McCormick
- U.S. Geological Survey, Leetown Science Center, Conte Anadromous Fish Research Laboratory, One Migratory Way, Turners Falls, MA, 01376, USA
| |
Collapse
|
262
|
Landi E, Karabatas L, Scaglia P, Pisciottano F, Gutiérrez M, Ramírez L, Bergadá I, Rey RA, Jasper HG, Domené HM, Plazas PV, Domené S. Expression of acid-labile subunit (ALS) in developing and adult zebrafish and its role in dorso-ventral patterning during development. Gen Comp Endocrinol 2020; 299:113591. [PMID: 32828812 DOI: 10.1016/j.ygcen.2020.113591] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/28/2020] [Accepted: 08/18/2020] [Indexed: 11/24/2022]
Abstract
Mammalian acid-labile subunit (ALS) is a serum protein that binds binary complexes between Insulin-like growth factors (IGFs) and Insulin-like growth factor-binding proteins (IGFBPs) extending their half-life and keeping them in the vasculature. Human ALS deficiency (ACLSD), due to homozygous or compound heterozygous mutations in IGFALS, leads to moderate short stature with reduced levels of IGF-I and IGFBP-3. There is only one corresponding zebrafish ortholog gene and it has not yet been studied. In this study we elucidate the role of igfals during zebrafish development. In zebrafish embryos igfals mRNA is expressed throughout development, mainly in the brain and subsequently also in the gut and swimbladder. To determine its role during development, we knocked down igfals gene product using morpholinos (MOs). Igfals morphant embryos displayed dorsalization in different degrees of severity, including a shortened trunk and loss of tail. Furthermore, co-injection of human IGFALS (hIGFALS) mRNA was able to rescue the MO-induced phenotype. Finally, overexpression of either hIGFALS or zebrafish igfals (zigfals) mRNA leads to ventralization of embryos including a reduced head and enlarged tail. These findings suggest that als plays an important role in dorso-ventral patterning during zebrafish development.
Collapse
Affiliation(s)
- Estefanía Landi
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina.
| | - Liliana Karabatas
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina.
| | - Paula Scaglia
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina.
| | - Francisco Pisciottano
- Instituto de Biología y Medicina Experimental (IBYME), Vuelta de Obligado 2490, C1428ADN Buenos Aires, Argentina.
| | - Mariana Gutiérrez
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina.
| | - Laura Ramírez
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina.
| | - Ignacio Bergadá
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina.
| | - Rodolfo A Rey
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina.
| | - Héctor Guillermo Jasper
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina.
| | - Horacio Mario Domené
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina.
| | - Paola Viviana Plazas
- Instituto de Farmacología, Facultad de Medicina, Universidad de Buenos Aires (UBA), Paraguay 2155, C1121ABG Buenos Aires, Argentina.
| | - Sabina Domené
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina.
| |
Collapse
|
263
|
Lodjak J, Verhulst S. Insulin-like growth factor 1 of wild vertebrates in a life-history context. Mol Cell Endocrinol 2020; 518:110978. [PMID: 32798584 DOI: 10.1016/j.mce.2020.110978] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 08/03/2020] [Accepted: 08/03/2020] [Indexed: 12/15/2022]
Abstract
Broad variation in intra- and interspecific life-history traits is largely shaped by resource limitation and the ensuing allocation trade-offs that animals are forced to make. Insulin-like growth factor 1 (IGF-1), a growth-hormone-dependent peptide, may be a key player in the regulation of allocation processes. In laboratory animals, the effects of IGF-1 on growth- and development (positive), reproduction (positive), and longevity (negative) are well established. We here review the evidence on these effects in wild vertebrates, where animals are more likely to face resource limitation and other challenges. We point out the similarities and dissimilarities in patterns of IGF-1 functions obtained in these two different study settings and discuss the knowledge we need to develop a comprehensive picture of the role of IGF-1 in mediating life-history variation of wild vertebrates.
Collapse
Affiliation(s)
- Jaanis Lodjak
- Department of Zoology, Institute of Ecology and Earth Sciences, University of Tartu, 46 Vanemuise Street, Tartu, 51014, Estonia; Groningen Institute for Evolutionary Life Sciences, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, Netherlands.
| | - Simon Verhulst
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, Netherlands
| |
Collapse
|
264
|
de Groot S, Röttgering B, Gelderblom H, Pijl H, Szuhai K, Kroep JR. Unraveling the Resistance of IGF-Pathway Inhibition in Ewing Sarcoma. Cancers (Basel) 2020; 12:cancers12123568. [PMID: 33260481 PMCID: PMC7759976 DOI: 10.3390/cancers12123568] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/22/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022] Open
Abstract
Simple Summary The insulin-like growth factor-1 receptor (IGF1R) is a receptor commonly overexpressed and overactivated in a variety of cancers, including Ewing sarcoma, and promotes cell growth and survival. After promising results with targeting and inhibiting the receptor in vitro, multiple different IGF1R targeting compounds have been clinically tried but showed limited efficacy. Here we discuss several possible resistance mechanisms which could explain why IGF1R targeting fails in the clinic and discuss possible ways to overcome these resistances. Abstract Insulin-like growth factor-1 receptor (IGF1R) inhibitors are effective in preclinical studies, but so far, no convincing benefit in clinical studies has been observed, except in some rare cases of sustained response in Ewing sarcoma patients. The mechanism of resistance is unknown, but several hypotheses are proposed. In this review, multiple possible mechanisms of resistance to IGF-targeted therapies are discussed, including activated insulin signaling, pituitary-driven feedback loops through growth hormone (GH) secretion and autocrine loops. Additionally, the outcomes of clinical trials of IGF1-targeted therapies are discussed, as well as strategies to overcome the possible resistance mechanisms. In conclusion, lowering the plasma insulin levels or blocking its activity could provide an additional target in cancer therapy in combination with IGF1 inhibition. Furthermore, because Ewing sarcoma cells predominantly express the insulin receptor A (IRA) and healthy tissue insulin receptor B (IRB), it may be possible to synthesize a specific IRA inhibitor.
Collapse
Affiliation(s)
- Stefanie de Groot
- Department of Medical Oncology, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands; (S.d.G.); (H.G.)
| | - Bas Röttgering
- Department of Cell and Chemical Biology, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands;
| | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands; (S.d.G.); (H.G.)
| | - Hanno Pijl
- Department of Endocrinology, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands;
| | - Karoly Szuhai
- Department of Cell and Chemical Biology, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands;
- Correspondence: (K.S.); (J.R.K.); Tel.: +31-715266922 (K.S.); +31-715263464 (J.R.K.)
| | - Judith R. Kroep
- Department of Medical Oncology, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands; (S.d.G.); (H.G.)
- Correspondence: (K.S.); (J.R.K.); Tel.: +31-715266922 (K.S.); +31-715263464 (J.R.K.)
| |
Collapse
|
265
|
Chramiec A, Teles D, Yeager K, Marturano-Kruik A, Pak J, Chen T, Hao L, Wang M, Lock R, Tavakol DN, Lee MB, Kim J, Ronaldson-Bouchard K, Vunjak-Novakovic G. Integrated human organ-on-a-chip model for predictive studies of anti-tumor drug efficacy and cardiac safety. LAB ON A CHIP 2020; 20:4357-4372. [PMID: 32955072 PMCID: PMC8092329 DOI: 10.1039/d0lc00424c] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Traditional drug screening models are often unable to faithfully recapitulate human physiology in health and disease, motivating the development of microfluidic organs-on-a-chip (OOC) platforms that can mimic many aspects of human physiology and in the process alleviate many of the discrepancies between preclinical studies and clinical trials outcomes. Linsitinib, a novel anti-cancer drug, showed promising results in pre-clinical models of Ewing Sarcoma (ES), where it suppressed tumor growth. However, a Phase II clinical trial in several European centers with patients showed relapsed and/or refractory ES. We report an integrated, open setting, imaging and sampling accessible, polysulfone-based platform, featuring minimal hydrophobic compound binding. Two bioengineered human tissues - bone ES tumor and heart muscle - were cultured either in isolation or in the integrated platform and subjected to a clinically used linsitinib dosage. The measured anti-tumor efficacy and cardiotoxicity were compared with the results observed in the clinical trial. Only the engineered tumor tissues, and not monolayers, recapitulated the bone microenvironment pathways targeted by linsitinib, and the clinically-relevant differences in drug responses between non-metastatic and metastatic ES tumors. The responses of non-metastatic ES tumor tissues and heart muscle to linsitinib were much closer to those observed in the clinical trial for tissues cultured in an integrated setting than for tissues cultured in isolation. Drug treatment of isolated tissues resulted in significant decreases in tumor viability and cardiac function. Meanwhile, drug treatment in an integrated setting showed poor tumor response and less cardiotoxicity, which matched the results of the clinical trial. Overall, the integration of engineered human tumor and cardiac tissues in the integrated platform improved the predictive accuracy for both the direct and off-target effects of linsitinib. The proposed approach could be readily extended to other drugs and tissue systems.
Collapse
Affiliation(s)
- Alan Chramiec
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Diogo Teles
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarāes, Braga, Portugal
| | - Keith Yeager
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Alessandro Marturano-Kruik
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Department of Chemistry, Materials and Chemical Engineering “G Natta”, Politecnico de Milano, Milano, Italy
| | - Joseph Pak
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Timothy Chen
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Luke Hao
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Miranda Wang
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Roberta Lock
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | | | - Marcus Busub Lee
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Jinho Kim
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ, USA
| | | | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Department of Medicine, Columbia University, New York, NY, USA
| |
Collapse
|
266
|
Mashinini MMA. Pituitary gland and growth hormone. SOUTHERN AFRICAN JOURNAL OF ANAESTHESIA AND ANALGESIA 2020. [DOI: 10.36303/sajaa.2020.26.6.s3.2553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The pituitary gland (PG) is said to be a “master” endocrine gland and through its tropic hormones influences other endocrine glands to secrete hormones that have a variety of effects on body systems. Growth hormone (GH) is a pituitary gland hormone that has direct and indirect effects produced by somatotrophs in early fetal life. It is essential for normal postnatal growth and has diverse effects across multiple physiological systems. The somatotrophic axis is made predominantly of GH, insulin-like growth factor 1 (IGF-1) and other factors regulating growth. This review summarises the relevant anatomical relationships of the PG, the basic aspect of GH physiology which includes mechanism of action (MOA), regulation and secretion and a direct and indirect action of GH through IGF-1, an important mediator of most of the peripheral action of GH.
Collapse
|
267
|
Insulin-Like Growth Factors Are Key Regulators of T Helper 17 Regulatory T Cell Balance in Autoimmunity. Immunity 2020; 52:650-667.e10. [PMID: 32294406 DOI: 10.1016/j.immuni.2020.03.013] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 12/17/2019] [Accepted: 03/19/2020] [Indexed: 12/14/2022]
Abstract
Appropriate balance of T helper 17 (Th17) and regulatory T (Treg) cells maintains immune tolerance and host defense. Disruption of Th17-Treg cell balance is implicated in a number of immune-mediated diseases, many of which display dysregulation of the insulin-like growth factor (IGF) system. Here, we show that, among effector T cell subsets, Th17 and Treg cells selectively expressed multiple components of the IGF system. Signaling through IGF receptor (IGF1R) activated the protein kinase B-mammalian target of rapamycin (AKT-mTOR) pathway, increased aerobic glycolysis, favored Th17 cell differentiation over that of Treg cells, and promoted a heightened pro-inflammatory gene expression signature. Group 3 innate lymphoid cells (ILC3s), but not ILC1s or ILC2s, were similarly responsive to IGF signaling. Mice with deficiency of IGF1R targeted to T cells failed to fully develop disease in the experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis. Thus, the IGF system represents a previously unappreciated pathway by which type 3 immunity is modulated and immune-mediated pathogenesis controlled.
Collapse
|
268
|
Hou ZS, Xin YR, Zeng C, Zhao HK, Tian Y, Li JF, Wen HS. GHRH-SST-GH-IGF axis regulates crosstalk between growth and immunity in rainbow trout (Oncorhynchus mykiss) infected with Vibrio anguillarum. FISH & SHELLFISH IMMUNOLOGY 2020; 106:887-897. [PMID: 32866610 DOI: 10.1016/j.fsi.2020.08.037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 08/15/2020] [Accepted: 08/20/2020] [Indexed: 06/11/2023]
Abstract
An energy trade-off is existed between immunological competence and growth. The axis of growth hormone releasing hormone, somatostatin, growth hormone, insulin-like growth factor (GHRH-SST-GH-IGF axis) regulates growth performances and immune competences in rainbow trout (Oncorhynchus mykiss). The salmonid-specific whole genome duplication event is known to result in duplicated copies of several key genes in GHRH-SST-GH-IGF axis. In this study, we evaluated the physiological functions of GHRH-SST-GH-IGF axis in regulating crosstalk between growth and immunity. Based on principal components analysis (PCA), we observed the overall expression profiles of GHRH-SST-GH-IGF axis were significantly altered by Vibrio anguillarum infection. Trout challenged with Vibrio anguillarum showed down-regulated igf1s subtypes and up-regulated igfbp1a1. The brain sst genes (sst1a, sst1b, sst3b and sst5) and igfpbs genes (igfbp4s and igfbp5b2) were significantly affected by V. anguillarum infection, while the igfbp4s, igfbp5s, igfbp6s and igf2bps genes showed significant changes in peripheral immune tissues in response to V. anguillarum infection. Gene enrichment analyses showed functional and signaling pathways associated with apoptosis (such as p53, HIF-1 or FoxO signaling) were activated. We further proposed a possible model that describes the IGF and IGFBPs-regulated interaction between cell growth and programmed death. Our study provided new insights into the physiological functions and potentially regulatory mechanisms of the GHRH-SST-GH-IGF axis, indicating the pleiotropic effects of GHRH-SST-GH-IGF axis in regulating crosstalk between growth and immunity in trout.
Collapse
Affiliation(s)
- Zhi-Shuai Hou
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education (KLMME), Qingdao, China.
| | - Yuan-Ru Xin
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education (KLMME), Qingdao, China
| | - Chu Zeng
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education (KLMME), Qingdao, China
| | - Hong-Kui Zhao
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education (KLMME), Qingdao, China
| | - Yuan Tian
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education (KLMME), Qingdao, China
| | - Ji-Fang Li
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education (KLMME), Qingdao, China
| | - Hai-Shen Wen
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education (KLMME), Qingdao, China.
| |
Collapse
|
269
|
Chen Z, Zhang N, Chu HY, Yu Y, Zhang ZK, Zhang G, Zhang BT. Connective Tissue Growth Factor: From Molecular Understandings to Drug Discovery. Front Cell Dev Biol 2020; 8:593269. [PMID: 33195264 PMCID: PMC7658337 DOI: 10.3389/fcell.2020.593269] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 10/09/2020] [Indexed: 01/18/2023] Open
Abstract
Connective tissue growth factor (CTGF) is a key signaling and regulatory molecule involved in different biological processes, such as cell proliferation, angiogenesis, and wound healing, as well as multiple pathologies, such as tumor development and tissue fibrosis. Although the underlying mechanisms of CTGF remain incompletely understood, a commonly accepted theory is that the interactions between different protein domains in CTGF and other various regulatory proteins and ligands contribute to its variety of functions. Here, we highlight the structure of each domain of CTGF and its biology functions in physiological conditions. We further summarized main diseases that are deeply influenced by CTGF domains and the potential targets of these diseases. Finally, we address the advantages and disadvantages of current drugs targeting CTGF and provide the perspective for the drug discovery of the next generation of CTGF inhibitors based on aptamers.
Collapse
Affiliation(s)
- Zihao Chen
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Ning Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Hang Yin Chu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Yuanyuan Yu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Zong-Kang Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Bao-Ting Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
270
|
Differential Expression of IGF1, IGFBP5, MSTN, and MYH1 Across Different Age Classes in American Quarter Horses. J Equine Vet Sci 2020; 94:103226. [PMID: 33077104 DOI: 10.1016/j.jevs.2020.103226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/05/2020] [Accepted: 08/07/2020] [Indexed: 11/22/2022]
Abstract
The objective of this study was to determine the influence of age on expression of insulin-like growth factor 1 (IGF1), insulin-like growth factor binding protein (IGFBP5), myostatin (MSTN), and myosin (MYH1) genes which are related to growth and muscle development in the American Quarter Horse. Thus, horses (n = 10) from weanling, yearling, 2-, 3-, and 10-year-old age classes were sampled and gene expression was assessed by RT-qPCR. ΔCT was calculated using the hypoxanthine-guanine phosphoribosyltransferase gene as an internal normalizer. The generalized linear model was used to determine differentially expressed genes, by pairwise comparison between ages. Among technical replicates, the coefficient of variation ranged from 1.0 to 5.2% and was lower than the variation observed between biological replicates (2.1-12.9%). IGF1 demonstrated significantly lower expression in the 3-year-old age class than in weanlings and yearlings, but the 10-year-old age class displayed a significantly higher level than 2- and 3-year-old age classes. Expression of IGFBP5 was highest in weanlings compared with all other age classes. Expression of MSTN was significantly higher in weanlings than in other age classes, whereas 10-year-old horses had an intermediate level of expression, but significantly different from yearlings, 2- and 3-year-old fillies. Finally, expression of MYH1 was lower in 2- and 10-year-old horses than in weanlings and yearlings, whereas 3-year-old fillies demonstrated an intermediate level of expression. Differential expression patterns observed in this preliminary study provide insight into the physiological changes occurring throughout the life span of horses. These patterns could also help explain the variation in performance and endurance between individuals at different developmental stages.
Collapse
|
271
|
Yamaguchi G, Habara S, Suzuki S, Ugachi Y, Kawai H, Nakajima T, Shimizu M. Effects of growth hormone and cortisol administration on plasma insulin-like growth factor binding proteins in juveniles of three subspecies of masu salmon (Oncorhynchus masou). Comp Biochem Physiol A Mol Integr Physiol 2020; 251:110821. [PMID: 33053436 DOI: 10.1016/j.cbpa.2020.110821] [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] [Received: 06/14/2020] [Revised: 10/06/2020] [Accepted: 10/07/2020] [Indexed: 11/26/2022]
Abstract
In this study, we examined the effects of porcine growth hormone (GH) and cortisol on plasma insulin-like growth factor binding proteins (IGFBPs) in juveniles of three subspecies of Oncorhynchus masou (masu, amago, and Biwa salmon). Ligand blotting using digoxigenin-labeled human IGF-I was used to detect and semi-quantify three major circulating IGFBP bands at 41, 28, and 22 kDa, corresponding to IGFBP-2b, -1a, and -1b, respectively. GH increased plasma IGFBP-2b concentration in masu and Biwa salmon but suppressed it in amago salmon. Plasma IGFBP-2b levels were increased by cortisol in the three subspecies. Cortisol induced plasma IGFBP-1a in the three subspecies, whereas GH had a suppressive effect in masu and Biwa salmon. Sham and cortisol injections increased plasma IGFBP-1b levels after 1 day in masu and amago salmon, suggesting that IGFBP-1b is induced following exposure to stressors via cortisol. Increased IGFBP-1b levels were restored to basal levels when co-injected with GH in Biwa salmon, and the same trend was seen in masu and amago salmon. However, the suppressive effect of GH disappeared 2 days after injection in the three subspecies. Despite some differences among subspecies, the findings suggest that cortisol is a primary inducer of plasma IGFBP-1b; however, GH counteracts it in the short term. Therefore, GH has the potential to modulate the degree of increase in circulating IGFBP-1b levels during acute stress.
Collapse
Affiliation(s)
- Ginnosuke Yamaguchi
- Graduate School of Environmental Science, Hokkaido University, Kita 10, Nishi 5, Kita-ku, Sapporo, Hokkaido 060-0810, Japan
| | - Shiori Habara
- Graduate School of Environmental Science, Hokkaido University, Kita 10, Nishi 5, Kita-ku, Sapporo, Hokkaido 060-0810, Japan
| | - Shotaro Suzuki
- Graduate School of Environmental Science, Hokkaido University, Kita 10, Nishi 5, Kita-ku, Sapporo, Hokkaido 060-0810, Japan
| | - Yuki Ugachi
- Graduate School of Environmental Science, Hokkaido University, Kita 10, Nishi 5, Kita-ku, Sapporo, Hokkaido 060-0810, Japan
| | - Hisashi Kawai
- Faculty of Fisheries Sciences, Hokkaido University, 3-1-1 Minato, Hakodate, Hokkaido 041-8611, Japan
| | - Takuro Nakajima
- Department of Fisheries, Shiga Prefecture, 4-1-1 Kyomachi, Otsu, Shiga 520-8577, Japan
| | - Munetaka Shimizu
- Graduate School of Environmental Science, Hokkaido University, Kita 10, Nishi 5, Kita-ku, Sapporo, Hokkaido 060-0810, Japan; Faculty of Fisheries Sciences, Hokkaido University, 3-1-1 Minato, Hakodate, Hokkaido 041-8611, Japan.
| |
Collapse
|
272
|
Compromised IGF signaling causes caspase-6 activation in Huntington disease. Exp Neurol 2020; 332:113396. [DOI: 10.1016/j.expneurol.2020.113396] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 06/16/2020] [Accepted: 06/26/2020] [Indexed: 11/19/2022]
|
273
|
Korkmaz Ağaoğlu Ö, Ağaoğlu AR, Özmen Ö, Saatci M, Schäfer-Somi S, Aslan S. Expression of the insulin-like growth factor (IGF) gene family in feline uterus during pregnancy. Biotech Histochem 2020; 96:439-449. [PMID: 32967473 DOI: 10.1080/10520295.2020.1818285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Members of the IGF gene family participate in cell differentiation and proliferation during pregnancy. We used 35 cats assigned to experimental groups (G) based on pregnancy stages: G1, pre-implantation; G2, implantation; G3, early pregnancy; G4, mid-pregnancy; G5, nonpregnant. Quantitative polymerase chain reaction (qPCR) and immunohistochemistry were used to analyze the expression of the IGF gene family. During pregnancy, expression of IGF-1 gene was significantly greater at implantation sites in the G1 and G2 groups than at placentation sites in G3 and G4 groups. IGF-2 expression was greater in the G2, G3 and G4 groups than in G1. Expression of the IGF-1R gene was significantly greater at placental sites in G3 than in G1 and G4. IGF-2R genes were expressed in all groups. Insulin-like growth factor binding proteins (IGFBPs) were expressed at intensities that depended on the stage of pregnancy; they were detected in different cell types and at different sites in the uterus. We found that members of the IGF gene family were expressed differentially in the endometrium during pregnancy. Our findings suggest that the IGF family may be a regulatory factor for pregnancy in cats.
Collapse
Affiliation(s)
- Özgecan Korkmaz Ağaoğlu
- Department of Animal Science, Faculty of Veterinary Medicine, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
| | - Ali Reha Ağaoğlu
- Department of Obstetrics and Gynecology, Faculty of Veterinary Medicine, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
| | - Özlem Özmen
- Department of Pathology, Faculty of Veterinary Medicine, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
| | - Mustafa Saatci
- Department of Animal Science, Fethiye Agriculture Faculty, Muğla Sıtkı Koçman University, Muğla, Turkey
| | - Sabine Schäfer-Somi
- Centre for Artificial Insemination and Embryo Transfer, University of Veterinary Medicine, Vienna, Austria
| | - Selim Aslan
- Department of Obstetrics and Gynecology, Veterinary Faculty, Near East University, Mersin, Turkey
| |
Collapse
|
274
|
Fermented Oyster Extract Promotes Insulin-Like Growth Factor-1-Mediated Osteogenesis and Growth Rate. Mar Drugs 2020; 18:md18090472. [PMID: 32962034 PMCID: PMC7551862 DOI: 10.3390/md18090472] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/04/2020] [Accepted: 09/16/2020] [Indexed: 12/15/2022] Open
Abstract
Fermented oyster (Crassostrea gigas) extract (FO) prevents ovariectomy-induced osteoporosis by inhibiting osteoclastogenesis and activating osteogenesis. However, the molecular mechanisms underlying FO-mediated bone formation and growth rate are unclear. In the current study, we found that FO significantly upregulated the expression of growth-promoting genes in zebrafish larvae including insulin-like growth factor 1 (zigf-1), insulin-like growth factor binding protein 3 (zigfbp-3), growth hormone-1 (zgh-1), growth hormone receptor-1 (zghr-1), growth hormone receptor alpha (zghra), glucokinase (zgck), and cholecystokinin (zccka). In addition, zebrafish larvae treated with 100 μg/mL FO increased in total body length (3.89 ± 0.13 mm) at 12 days post fertilization (dpf) compared to untreated larvae (3.69 ± 0.02 mm); this effect was comparable to that of the β-glycerophosphate-treated zebrafish larvae (4.00 ± 0.02 mm). Furthermore, FO time- and dose-dependently increased the extracellular release of IGF-1 from preosteoblast MC3T3-E1 cells, which was accompanied by high expression of IGF-1. Pharmacological inhibition of IGF-1 receptor (IGF-1R) using picropodophyllin (PPP) significantly reduced FO-mediated vertebrae formation (from 9.19 ± 0.31 to 5.53 ± 0.35) and growth performance (from 3.91 ± 0.02 to 3.69 ± 0.01 mm) in zebrafish larvae at 9 dpf. Similarly, PPP significantly decreased FO-induced calcium deposition in MC3T3-E1 cells by inhibiting GSK-3β phosphorylation at Ser9. Additionally, DOI hydrochloride, a potent stabilizer of GSK-3β, reduced FO-induced nuclear translocation of RUNX2. Transient knockdown of IGF-1Rα/β using specific silencing RNA also resulted in a significant decrease in calcium deposition and reduction in GSK-3β phosphorylation at Ser9 in MC3T3-E1 cells. Altogether, these results indicate that FO increased phosphorylated GSK-3β at Ser9 by activating the autocrine IGF-1-mediated IGF-1R signaling pathway, thereby promoting osteogenesis and growth performance. Therefore, FO is a potential nutritional supplement for bone formation and growth.
Collapse
|
275
|
Sidorkiewicz I, Jóźwik M, Niemira M, Krętowski A. Insulin Resistance and Endometrial Cancer: Emerging Role for microRNA. Cancers (Basel) 2020; 12:E2559. [PMID: 32911852 PMCID: PMC7563767 DOI: 10.3390/cancers12092559] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/03/2020] [Accepted: 09/07/2020] [Indexed: 12/21/2022] Open
Abstract
Endometrial cancer (EC) remains one of the most common cancers of the female reproductive system. Epidemiological and clinical data implicate insulin resistance (IR) and its accompanying hyperinsulinemia as key factors in the development of EC. MicroRNAs (miRNAs) are short molecules of non-coding endogenous RNA that function as post-transcriptional regulators. Accumulating evidence has shown that the miRNA expression pattern is also likely to be associated with EC risk factors. The aim of this work was the verification of the relationships between IR, EC, and miRNA, and, as based on the literature data, elucidation of miRNA's potential utility for EC prevention in IR patients. The pathways affected in IR relate to the insulin receptors, insulin-like growth factors and their receptors, insulin-like growth factor binding proteins, sex hormone-binding globulin, and estrogens. Herein, we present and discuss arguments for miRNAs as a plausible molecular link between IR and EC development. Specifically, our careful literature search indicated that dysregulation of at least 13 miRNAs has been ascribed to both conditions. We conclude that there is a reasonable possibility for miRNAs to become a predictive factor of future EC in IR patients.
Collapse
Affiliation(s)
- Iwona Sidorkiewicz
- Clinical Research Centre, Medical University of Białystok, M. Skłodowskiej-Curie 24a, 15-276 Białystok, Poland; (M.N.); (A.K.)
| | - Maciej Jóźwik
- Department of Gynecology and Gynecologic Oncology, Medical University of Białystok, M. Skłodowskiej-Curie 24a, 15-276 Białystok, Poland;
| | - Magdalena Niemira
- Clinical Research Centre, Medical University of Białystok, M. Skłodowskiej-Curie 24a, 15-276 Białystok, Poland; (M.N.); (A.K.)
| | - Adam Krętowski
- Clinical Research Centre, Medical University of Białystok, M. Skłodowskiej-Curie 24a, 15-276 Białystok, Poland; (M.N.); (A.K.)
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Białystok, M. Skłodowskiej-Curie 24a, 15-276 Białystok, Poland
| |
Collapse
|
276
|
Sharker MR, Hossen S, Nou IS, Kho KH. Characterization of Insulin-Like Growth Factor Binding Protein 7 (Igfbp7) and Its Potential Involvement in Shell Formation and Metamorphosis of Pacific Abalone, Haliotis discus hannai. Int J Mol Sci 2020; 21:ijms21186529. [PMID: 32906674 PMCID: PMC7555818 DOI: 10.3390/ijms21186529] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/01/2020] [Accepted: 09/04/2020] [Indexed: 12/20/2022] Open
Abstract
Insulin-like growth factor binding proteins (IGFBPs) are secreted proteins that play an important role in IGF regulation of growth and development of vertebrate and invertebrates. In this study, the IGFBP7 gene was cloned and characterized from mantle tissues of H. discus hannai, and designated as Hdh IGFBP7. The full-length cDNA sequence transcribed from the Hdh IGFBP7 gene was 1519-bp long with an open reading frame of 720-bp corresponding to a putative polypeptide of 239 amino acids. The molecular mass of its mature protein was approximately 23.44 KDa with an estimated isoelectric point (pI) of 5.35, and it shared significant homology with IGFBP7 gene of H. madaka. Hdh IGFBP7 has a characteristic IGFBP N-terminal domain (22–89 aa), a kazal-type serine proteinase inhibitor domain (77–128), and an immunoglobulin-like C2 domain (144–223). Furthermore, twelve cysteine residues and a signature motif of IGFBPs (XCGCCXXC) were found in its N-terminal domain. Phylogenetic analysis revealed that Hdh IGFBP7 was aligned with IGFBP7 of H. madaka. Tissue distribution analysis showed that the mRNA of Hdh IGFBP7 was expressed in all examined tissues, with the highest expression level observed in the mantle and gill tissues. The expression level of Hdh IGFBP7 mRNA was relatively higher at the juvenile stage during its metamorphosis period. In situ hybridization showed that Hdh IGFBP7 transcript was expressed in epithelial cells of the dorsal mantle pallial and mucus cells of the branchial epithelium in gill. These results provide basic information for future studies on the role of IGFBP7 in IGF regulation of shell growth, development and metamorphosis of abalone.
Collapse
Affiliation(s)
- Md. Rajib Sharker
- Department of Fisheries Science, College of Fisheries and Ocean Sciences, Chonnam National University, 50 Daehak-ro, Yeosu, Jeonnam 59626, Korea; (M.R.S.); (S.H.)
| | - Shaharior Hossen
- Department of Fisheries Science, College of Fisheries and Ocean Sciences, Chonnam National University, 50 Daehak-ro, Yeosu, Jeonnam 59626, Korea; (M.R.S.); (S.H.)
| | - Ill-Sup Nou
- Department of Horticulture, College of Life Science and Natural Resources, Sunchon National University, 255, Jungang-ro, Suncheon-Si, Jeollanam-do 57922, Korea;
| | - Kang Hee Kho
- Department of Fisheries Science, College of Fisheries and Ocean Sciences, Chonnam National University, 50 Daehak-ro, Yeosu, Jeonnam 59626, Korea; (M.R.S.); (S.H.)
- Correspondence: ; Tel.: +82-616-597-168; Fax: +82-616-597-169
| |
Collapse
|
277
|
Jenkins EC, Brown SO, Germain D. The Multi-Faced Role of PAPP-A in Post-Partum Breast Cancer: IGF-Signaling is Only the Beginning. J Mammary Gland Biol Neoplasia 2020; 25:181-189. [PMID: 32901383 DOI: 10.1007/s10911-020-09456-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/24/2020] [Accepted: 09/03/2020] [Indexed: 12/12/2022] Open
Abstract
Insulin-like growth factor (IGF) signaling and control of local bioavailability of free IGF by the IGF binding proteins (IGFBP) are important regulators of both mammary development and breast cancer. A recent genome-wide association study (GWAS) identified small nucleotide polymorphisms that reduce the expression of IGFBP-5 as a risk factor of developing breast cancer. This observation suggests that genetic alterations leading to a decreased level of IGFBP-5 may also contribute to breast cancer. In the current review, we focus on Pregnancy-Associated Plasma Protein A (PAPP-A), a protease involved in the degradation of IGFBP-5. PAPP-A is overexpressed in the majority of breast cancers but its role in cancer has only begun to be explored. More specifically, this review aims at highlighting the role of post-partum involution in the oncogenic function of PAPP-A. Notably, we summarize recent studies indicating that PAPP-A plays a role not only in the degradation of IGFBP-5 but also in the deposition of collagen and activation of the collagen receptor discoidin 2 (DDR2) during post-partum involution. Finally, considering the immunosuppressive microenvironment of post-partum involution, we also discuss the unexpected finding made in Ewing Sarcoma that PAPP-A plays a role in immune evasion. While the immunosuppressive role of PAPP-A in breast cancer remains to be determined, collectively these studies highlight the multifaced role of PAPP-A in cancer that extends well beyond its effect on IGF-signaling.
Collapse
Affiliation(s)
- Edmund Charles Jenkins
- Department of Medicine, Division of Hematology/ Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, 10029, USA
| | - Samantha O Brown
- Department of Medicine, Division of Hematology/ Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, 10029, USA
| | - Doris Germain
- Department of Medicine, Division of Hematology/ Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, 10029, USA.
| |
Collapse
|
278
|
Kazemi M, Jarrett BY, Parry SA, Thalacker-Mercer AE, Hoeger KM, Spandorfer SD, Lujan ME. Osteosarcopenia in Reproductive-Aged Women with Polycystic Ovary Syndrome: A Multicenter Case-Control Study. J Clin Endocrinol Metab 2020; 105:5866600. [PMID: 32614948 PMCID: PMC7418445 DOI: 10.1210/clinem/dgaa426] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 06/26/2020] [Indexed: 02/07/2023]
Abstract
CONTEXT Osteosarcopenia (loss of skeletal muscle and bone mass and/or function usually associated with aging) shares pathophysiological mechanisms with polycystic ovary syndrome (PCOS). However, the relationship between osteosarcopenia and PCOS remains unclear. OBJECTIVE We evaluated skeletal muscle index% (SMI% = [appendicular muscle mass/weight (kg)] × 100) and bone mineral density (BMD) in PCOS (hyperandrogenism + oligoamenorrhea), and contrasted these musculoskeletal markers against 3 reproductive phenotypes (i): HA (hyperandrogenism + eumenorrhea) (ii); OA (normoandrogenic + oligoamenorrhea) and (iii), controls (normoandrogenic + eumenorrhea). Endocrine predictors of SMI% and BMD were evaluated across the groups. DESIGN, SETTING, AND PARTICIPANTS Multicenter case-control study of 203 women (18-48 years old) in New York State. RESULTS PCOS group exhibited reduced SMI% (mean [95% confidence interval (CI)]; 26.2% [25.1,27.3] vs 28.8% [27.7,29.8]), lower-extremity SMI% (57.6% [56.7,60.0] vs 62.5% [60.3,64.6]), and BMD (1.11 [1.08,1.14] vs 1.17 [1.14,1.20] g/cm2) compared to controls. PCOS group also had decreased upper (0.72 [0.70,0.74] vs 0.77 [0.75,0.79] g/cm2) and lower (1.13 [1.10,1.16] vs 1.19 [1.16,1.22] g/cm2) limb BMD compared to HA. Matsuda index was lower in PCOS vs controls and positively associated with SMI% in all groups (all Ps ≤ 0.05). Only controls showed associations between insulin-like growth factor (IGF) 1 and upper (r = 0.84) and lower (r = 0.72) limb BMD (all Ps < 0.01). Unlike in PCOS, IGF-binding protein 2 was associated with SMI% in controls (r = 0.45) and HA (r = 0.67), and with upper limb BMD (r = 0.98) in HA (all Ps < 0.05). CONCLUSIONS Women with PCOS exhibit early signs of osteosarcopenia when compared to controls likely attributed to disrupted insulin function. Understanding the degree of musculoskeletal deterioration in PCOS is critical for implementing targeted interventions that prevent and delay osteosarcopenia in this clinical population.
Collapse
Affiliation(s)
- Maryam Kazemi
- Division of Nutritional Sciences, Human Metabolic Research Unit, Cornell University, Ithaca, NY, US
| | - Brittany Y Jarrett
- Division of Nutritional Sciences, Human Metabolic Research Unit, Cornell University, Ithaca, NY, US
| | - Stephen A Parry
- Cornell Statistical Consulting Unit, Cornell University, Ithaca, NY, US
| | - Anna E Thalacker-Mercer
- Division of Nutritional Sciences, Human Metabolic Research Unit, Cornell University, Ithaca, NY, US
| | - Kathleen M Hoeger
- Department of Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, NY, USA
| | - Steven D Spandorfer
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY, US
| | - Marla E Lujan
- Division of Nutritional Sciences, Human Metabolic Research Unit, Cornell University, Ithaca, NY, US
| |
Collapse
|
279
|
Tang D, Gao W, Yang J, Liu J, Zhao J, Ge J, Chen Q, Liu B. miR‑181d promotes cell proliferation via the IGF1/PI3K/AKT axis in glioma. Mol Med Rep 2020; 22:3804-3812. [PMID: 33000209 PMCID: PMC7533453 DOI: 10.3892/mmr.2020.11464] [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: 05/19/2019] [Accepted: 04/17/2020] [Indexed: 01/07/2023] Open
Abstract
Glioma is a malignant brain cancer that exhibits high invasive ability and poor prognosis. MicroRNA (miR)-181d has been reported to be involved in the development of glioma. Therefore, the aim of the present study was to investigate whether miR-181d affected cellular progression by influencing the insulin like growth factor (IGF1)/PI3K/AKT axis. Western blot analysis was performed to analyze the expression levels of specific proteins, and a Cell Counting Kit-8 assay was used to assess the proliferative ability of cells. Cell cycle progression and cellular apoptosis were both measured using flow cytometry. The results indicated that miR-181d promoted cellular proliferation and cell cycle progression, while suppressing cellular apoptosis via the IGF1/PI3K/AKT axis. It was demonstrated that the IGF1 and PI3K/AKT inhibitors reversed these observed functions of miR-181d. Furthermore, miR-181d enhanced the growth of glioma xenografts in vivo, promoted cell cycle progression and suppressed cellular apoptosis within glioma xenograft tissues. Therefore, this newly identified miR-181d/IGF1/PI3K/AKT axis may provide novel insights into the pathogenesis of glioma.
Collapse
Affiliation(s)
- Dong Tang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Wenhong Gao
- Department of Neurosurgery, Jingzhou Central Hospital, The Second Clinical Medical College, Yangtze University, Jingzhou, Hubei 434020, P.R. China
| | - Jian Yang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Junhui Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jian Zhao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jian Ge
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qianxue Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Baohui Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
280
|
Sharker MR, Kim SC, Hossen S, Kho KH. Characterization of Insulin-Like Growth Factor Binding Protein-5 (IGFBP-5) Gene and Its Potential Roles in Ontogenesis in the Pacific Abalone, Haliotis discus hannai. BIOLOGY 2020; 9:biology9080216. [PMID: 32784850 PMCID: PMC7465962 DOI: 10.3390/biology9080216] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/06/2020] [Accepted: 08/07/2020] [Indexed: 12/21/2022]
Abstract
Insulin-like growth factor binding protein family is known to be involved in regulating biological actions of insulin-like growth factors (IGFs). In the present study, a full-length cDNA encoding the IGFBP-5 gene was cloned and characterized from the cerebral ganglion of Haliotis discus hannai. The 921-bp full-length sequence of Hdh IGFBP-5 cDNA transcript had an open reading frame of 411 bp encoding a predicted polypeptide of 136 amino acids, sharing high sequence identities with IGFBP-5 of H. diversicolor. The deduced Hdh IGFBP-5 protein contained a putative transmembrane domain (13-35 aa) in the N-terminal region. It also possessed a signature domain of IGFBP protein family (IB domain, 45-120 aa). Six cysteine residues (Cys-47, Cys-55, Cys-73, Cys-85, Cys-98, and Cys-118) in this cloned sequence could potentially form an intrachain disulfide bond. Phylogenetic analysis indicated that the Hdh IGFBP-5 gene was robustly clustered with IGFBP-5 of H. diversicolor. Tissue distribution analysis based on qPCR assay showed that Hdh IGFBP-5 was widely expressed in all examined tissues, with significantly (p < 0.05) higher expression in the cerebral ganglion. In male and female gametogenetic cycles, Hdh IGFBP-5 mRNA was expressed at all stages, showing significantly higher level at ripening stage. The expression level of Hdh IGFBP-5 mRNA was significantly higher in the polar body stage than in other ontogenic stages. In situ hybridization revealed that Hdh IGFBP-5 mRNA was present in the neurosecretory cells of the cerebral ganglion. This is the first study describing IGFBP-5 in H. discus hannai that might be synthesized in the neural ganglia. Our results demonstrate Hdh IGFBP-5 is involved in regulating ontogenic development and reproductive regulation of H. discus hannai.
Collapse
Affiliation(s)
| | | | | | - Kang Hee Kho
- Correspondence: ; Tel.: +82-616-597-168; Fax: +82-616-597-169
| |
Collapse
|
281
|
Kamei H. Oxygen and embryonic growth: the role of insulin-like growth factor signaling. Gen Comp Endocrinol 2020; 294:113473. [PMID: 32247621 DOI: 10.1016/j.ygcen.2020.113473] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 03/05/2020] [Accepted: 03/28/2020] [Indexed: 01/03/2023]
Abstract
Oxygen is indispensable for the efficient release of chemical energy from nutrient molecules in cells. Therefore, the local oxygen tension is one of the most critical factors affecting physiological processes. In most viviparous species, many pathological conditions result in abnormal oxygen tension in the uterus, which modifies the growth and development of the fetus. Insulin-like growth factor (IGF/Igf) is one of the most important hormones for the regulation of somatic growth in animals. Changes in oxygen levels modulate the activity of the IGF/Igf signaling system, which in turn regulates the embryonic growth rate. In general, there are serious difficulties associated with monitoring and studying rodent embryos in utero. The zebrafish is a convenient experimental model to study the relationship between embryonic growth and environmental conditions. Most importantly, the fish model makes it possible to rapidly evaluate embryonic growth and development under entirely controlled environments without interfering with the mother organism. In this review, firstly an overview is given of the fluctuation of environmental oxygen, the IGF-system, and the advantages of the zebrafish model for studying embryonic growth. Then, the relationships of dynamic environmental oxygen and embryonic growth rate are outlined with a specific focus on the changes in the IGF/Igf-system in the zebrafish model. This review will shed light on the fine-tuning mechanisms of the embryonic IGF/Igf-system under different oxygen levels, including constant normoxia, hypoxia, and re-oxygenation.
Collapse
Affiliation(s)
- Hiroyasu Kamei
- Faculty of Biological Science and Technology, Institute of Science and Engineering, Kanazawa University, 11-4-1, Ossaka, Noto, Ishikawa 927-0552, Japan.
| |
Collapse
|
282
|
Dorandish S, Devos J, Clegg B, Price D, Muterspaugh R, Guthrie J, Heyl DL, Evans HG. Biochemical determinants of the IGFBP-3-hyaluronan interaction. FEBS Open Bio 2020; 10:1668-1684. [PMID: 32592613 PMCID: PMC7396449 DOI: 10.1002/2211-5463.12919] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/15/2020] [Accepted: 06/23/2020] [Indexed: 12/28/2022] Open
Abstract
IGFBP-3, the most abundant IGFBP and the main carrier of insulin-like growth factor I (IGF-I) in the circulation, can bind IGF-1 with high affinity, which attenuates IGF/IGF-IR interactions, thereby resulting in antiproliferative effects. The C-terminal domain of insulin-like growth factor-binding protein-3 (IGFBP-3) is known to contain an 18-basic amino acid motif capable of interacting with either humanin (HN) or hyaluronan (HA). We previously showed that the 18-amino acid IGFBP-3 peptide is capable of binding either HA or HN with comparable affinities to the full-length IGFBP-3 protein and that IGFBP-3 can compete with the HA receptor, CD44, for binding HA. Blocking the interaction between HA and CD44 reduced viability of A549 human lung cancer cells. In this study, we set out to better characterize IGFBP-3-HA interactions. We show that both stereochemistry and amino acid identity are important determinants of the interaction between the IGFBP-3 peptide and HA and for the peptide's ability to exert its cytotoxic effects. Binding of IGFBP-3 to either HA or HN was unaffected by glycosylation or reduction of IGFBP-3, suggesting that the basic 18-amino acid residue sequence of IGFBP-3 remains accessible for interaction with either HN or HA upon glycosylation or reduction of the full-length protein. Removing N-linked oligosaccharides from CD44 increased its ability to compete with IGFBP-3 for binding HA, while reduction of CD44 rendered the protein relatively ineffective at blocking IGFBP-3-HA interactions. We conclude that both deglycosylation and disulfide bond formation are important for CD44 to compete with IGFBP-3 for binding HA.
Collapse
Affiliation(s)
- Sadaf Dorandish
- Department of ChemistryEastern Michigan UniversityYpsilantiMIUSA
| | - Jonathan Devos
- Department of ChemistryEastern Michigan UniversityYpsilantiMIUSA
| | - Bradley Clegg
- Department of ChemistryEastern Michigan UniversityYpsilantiMIUSA
| | - Deanna Price
- Department of ChemistryEastern Michigan UniversityYpsilantiMIUSA
| | | | - Jeffrey Guthrie
- Department of ChemistryEastern Michigan UniversityYpsilantiMIUSA
| | - Deborah L. Heyl
- Department of ChemistryEastern Michigan UniversityYpsilantiMIUSA
| | - Hedeel Guy Evans
- Department of ChemistryEastern Michigan UniversityYpsilantiMIUSA
| |
Collapse
|
283
|
Yang G, Zhao W, Qin C, Yang L, Meng X, Lu R, Yan X, Cao X, Zhang Y, Nie G. Molecular identification of grass carp igfbp2 and the effect of glucose, insulin, and glucagon on igfbp2 mRNA expression. FISH PHYSIOLOGY AND BIOCHEMISTRY 2020; 46:1469-1482. [PMID: 32323051 DOI: 10.1007/s10695-020-00804-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Accepted: 03/30/2020] [Indexed: 06/11/2023]
Abstract
The GH (growth hormone)/IGFs (insulin-like growth factors) system has an important function in the regulation of growth. In this system, IGFBPs play a crucial regulatory role in IGF functions. As a member of the IGFBP family, IGFBP2 can bind to IGF and regulate IGF functions to regulate development and growth. In addition, IGFBP2 shows key regulatory functions in cell proliferation and metabolism. In this study, the igfbp2 gene was cloned from grass carp (Ctenopharyngodon idellus) liver. The ORF of grass carp igfbp2 is 834 bp long and encodes 277 amino acids. The tissue distribution results showed that igfbp2 is expressed in multiple tissues in grass carp and has a high expression level in the liver. In the OGTT, igfbp2 expression was significantly decreased in the liver and brain after 6 h of treatment with glucose. In vitro, igfbp2 expression in grass carp's primary hepatocytes was significantly suppressed by insulin after treatment for 6 and 12 h. Moreover, igfbp2 expression was markedly increased in a dose-dependent manner with glucagon incubation in grass carp's primary hepatocytes. To the best of our knowledge, this is the first report about Igfbp2 in grass carp. These results will provide a basis for the in-depth study of grass carp Igfbp2.
Collapse
Affiliation(s)
- Guokun Yang
- College of Fisheries, Henan Normal University, Xinxiang, 453007, People's Republic of China
- College of Fisheries, Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang, 453007, People's Republic of China
| | - Wenli Zhao
- College of Fisheries, Henan Normal University, Xinxiang, 453007, People's Republic of China
- College of Fisheries, Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang, 453007, People's Republic of China
| | - Chaobin Qin
- College of Fisheries, Henan Normal University, Xinxiang, 453007, People's Republic of China
- College of Fisheries, Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang, 453007, People's Republic of China
| | - Liping Yang
- College of Fisheries, Henan Normal University, Xinxiang, 453007, People's Republic of China
- College of Fisheries, Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang, 453007, People's Republic of China
| | - Xiaolin Meng
- College of Fisheries, Henan Normal University, Xinxiang, 453007, People's Republic of China
- College of Fisheries, Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang, 453007, People's Republic of China
| | - Ronghua Lu
- College of Fisheries, Henan Normal University, Xinxiang, 453007, People's Republic of China
- College of Fisheries, Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang, 453007, People's Republic of China
| | - Xiao Yan
- College of Fisheries, Henan Normal University, Xinxiang, 453007, People's Republic of China
- College of Fisheries, Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang, 453007, People's Republic of China
| | - Xianglin Cao
- College of Fisheries, Henan Normal University, Xinxiang, 453007, People's Republic of China
- College of Fisheries, Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang, 453007, People's Republic of China
| | - Yanmin Zhang
- College of Fisheries, Henan Normal University, Xinxiang, 453007, People's Republic of China
- College of Fisheries, Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang, 453007, People's Republic of China
| | - Guoxing Nie
- College of Fisheries, Henan Normal University, Xinxiang, 453007, People's Republic of China.
- College of Fisheries, Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang, 453007, People's Republic of China.
| |
Collapse
|
284
|
van Doorn J. Insulin-like growth factor-II and bioactive proteins containing a part of the E-domain of pro-insulin-like growth factor-II. Biofactors 2020; 46:563-578. [PMID: 32026557 PMCID: PMC7497164 DOI: 10.1002/biof.1623] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 01/15/2020] [Indexed: 12/13/2022]
Abstract
Insulin-like growth factor (IGF)-II is considered to function as an important fetal growth factor, which is structurally and functionally related to IGF-I and proinsulin. At least in vitro, IGF-II actions are mediated through the IGF-I receptor and to a lesser extent the insulin receptor. After birth, the function of IGF-II is less clear although in adults the serum level of IGF-II exceeds that of IGF-I several fold. The IGF-II gene is maternally imprinted, with exception of the liver and several parts of the brain, where it is expressed from both alleles. The regulation, organization, and translation of the IGF-II gene is complex, with five different putative promotors leading to a range of noncoding and coding mRNAs. The 180-amino acid pre-pro-IGF-II translation product can be divided into five domains and include a N-terminal signal peptide of 24 amino acid residues, the 67 amino acid long mature protein, and an 89 residues extension at the COOH terminus, designated as the E-domain. After removal of the signal peptide, the processing of pro-IGF-II into mature IGF-II requires various steps including glycosylation of the E-domain followed by the action of endo-proteases. Several of these processing intermediates can be found in the human circulation. There is increasing evidence that, besides IGF-II, several incompletely processed precursor forms of the protein, and even a 34-amino acid peptide (preptin) derived from the E-domain of pro-IGF-II, exhibit distinct biological activities. This review will focus on the current insights regarding the specific roles of the latter proteins in cancer, glucose homeostasis, and bone physiology. To address this topic clearly in the right context, a concise overview of the biological and biochemical properties of IGF-II and several relevant aspects of the IGF system will be provided.
Collapse
Affiliation(s)
- Jaap van Doorn
- Department of Genetics, Section Metabolic DiagnosticsUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| |
Collapse
|
285
|
Nichenametla SN, Mattocks DAL, Malloy VL. Age-at-onset-dependent effects of sulfur amino acid restriction on markers of growth and stress in male F344 rats. Aging Cell 2020; 19:e13177. [PMID: 32573078 PMCID: PMC7426777 DOI: 10.1111/acel.13177] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 03/29/2020] [Accepted: 05/18/2020] [Indexed: 12/24/2022] Open
Abstract
Trade-offs in life-history traits are clinically and mechanistically important. Sulfur amino acid restriction (SAAR) extends lifespan. But whether this benefit comes at the cost of other traits including stress resistance and growth is unclear. We investigated the effects of SAAR on growth markers (body weight, IGF1, and IGFBP3) and physiological stresses. Male-F344 rats were fed control (0.86% Met) and SAAR (0.17% Met) diets starting at 2, 10, and 20 months. Rats were injected with keyhole-limpet-hemocyanin (KLH) to measure immune responses (anti-KLH-IgM, anti-KLH-IgG, and delayed-type-hypersensitivity [DTH]). Markers of ER stress (FGF21 and adiponectin), detoxification capacity (glutathione [GSH] concentrations, GSH-S-transferase [GST], and cytochrome-P450 -reductase [CPR] activities), and low-grade inflammation (C-reactive protein [CRP]) were also determined. SAAR decreased body weight, liver weight, food intake, plasma IGF1, and IGFBP3; the effect size diminished with increasing age-at-onset. SAAR increased FGF21 and adiponectin, but stress damage markers GRP78 and Xbp1s/us were unchanged, suggesting that ER stress is hormetic. SAAR increased hepatic GST activity despite lower GSH, but CPR activity was unchanged, indicative of enhanced detoxification capacity. Other stress markers were either uncompromised (CRP, anti-KLH-IgM, and DTH) or slightly lower (anti-KLH-IgG). Increases in stress markers were similar across all ages-at-onset, except for adiponectin, which peaked at 2 months. Overall, SAAR did not compromise stress responses and resulted in maximal benefits with young-onset. In survival studies, median lifespan extension with initiation at 52 weeks was 7 weeks (p = .05); less than the 33.5-week extension observed in our previous study with 7-week initiation. Findings support SAAR translational studies and the need to optimize Met dose based on age-at-onset.
Collapse
Affiliation(s)
| | - Dwight A. L. Mattocks
- Animal Science Laboratory Orentreich Foundation for the Advancement of Science NY USA
| | - Virginia L Malloy
- Animal Science Laboratory Orentreich Foundation for the Advancement of Science NY USA
| |
Collapse
|
286
|
Jamshidi M, Mohammadi Pour S, Mahmoudian-Sani MR. Single Nucleotide Variants Associated with Colorectal Cancer Among Iranian Patients: A Narrative Review. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2020; 13:167-180. [PMID: 32581566 PMCID: PMC7280057 DOI: 10.2147/pgpm.s248349] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 05/19/2020] [Indexed: 12/31/2022]
Abstract
Colorectal cancer has been considered as one of the complicated multi-stage processes after adenoma-carcinoma sequence. Therefore, studies of the molecular dysregulation basis could present information on the recognition of the potent biomarkers and treatment targets for this disease. Even though outcomes of the patients with colorectal cancer have been improved largely with current annual screening plans, it is necessary to have reliable prognostic biomarkers because of the disease heterogeneity. There is a significant relationship between SNP in IL1RN* 2 (IL1ra), −509 C/T (TGFB1), rs11556218 T>G and rs4778889 T/C (IL16), miRNA-binding site polymorphisms in IL16, rs4464148 (SMAD7), rs6983267 (EGF), GSTT1, TACG haplotype (CTLA4), 1793G> A (MTHFR), Leu/Leu genotype of (EXO1), −137 G/C (IL18), C/T genotype (XRCC3), I3434T (XRCC7), MGMT, C3435T (MDR1), ff genotype of FokI, 677CT+TT (MTHFR), G2677T/A (MDR1) and CRC. Increased risk has been observed in VDR ApaI genotype “aa”. Finally, the protective effect has been explored in the TACA haplotype (CTLA4). According to the findings, the genetic polymorphisms in the immunity-associated genes are related to the CRC amongst the Iranian patients. Therefore, more large-scale functional investigations are necessary for confirming the results.
Collapse
Affiliation(s)
- Mohammad Jamshidi
- Department of Laboratory Sciences, School of Allied Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Somayeh Mohammadi Pour
- Department of Obstetrics and Gynecology, School of Medicine Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mohammad-Reza Mahmoudian-Sani
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
287
|
Barker RM, Holly JMP, Biernacka KM, Allen-Birt SJ, Perks CM. Mini Review: Opposing Pathologies in Cancer and Alzheimer's Disease: Does the PI3K/Akt Pathway Provide Clues? Front Endocrinol (Lausanne) 2020; 11:403. [PMID: 32655497 PMCID: PMC7324530 DOI: 10.3389/fendo.2020.00403] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 05/20/2020] [Indexed: 12/30/2022] Open
Abstract
This minireview is a brief overview examining the roles of insulin-like growth factors (IGFs) and the PI3K/Akt pathway in two apparently unconnected diseases: Alzheimer's dementia and cancer. For both, increased age is a major risk factor, and, in accord with the global rise in average life expectancy, their prevalence is also increasing. Cancer, however, involves excessive cell proliferation and metastasis, whereas Alzheimer's disease (AD) involves cell death and tissue destruction. The apparent "inverse" nature of these disease states is examined here, but also some important commonalities in terms of the PI3K/Akt pathway, glucose utilization and cell deregulation/death. The focus here is on four key molecules associated with this pathway; notably, the insulin receptor substrate 1 (IRS-1), cellular tumor antigen p53 (p53), peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 (PIN1) and low-density lipoprotein receptor-related protein-1 (LRP1), all previously identified as potential therapeutic targets for both diseases. The insulin-resistant state, commonly reported in AD brain, results in neuronal glucose deprivation, due to a dampening down of the PI3K/Akt pathway, including overactivity of the mammalian target of rapamycin 1 (mTORC1) complex, hyperphosphorylation of p53 and neuronal death. This contrasts with cancer, where there is overstimulation of the PI3K/Akt pathway and the suppression of mTORC1 and p53, enabling abundant energy and unrestrained cell proliferation. Although these disease states appear to be diametrically opposed, the same key molecules are controlling pathology and, with differential targeting of therapeutics, may yet provide a beneficial outcome for both.
Collapse
Affiliation(s)
- Rachel M. Barker
- IGFs & Metabolic Endocrinology Group, Bristol Medical School, Translational Health Sciences, Southmead Hospital, University of Bristol, Bristol, United Kingdom
| | - Jeff M. P. Holly
- IGFs & Metabolic Endocrinology Group, Bristol Medical School, Translational Health Sciences, Southmead Hospital, University of Bristol, Bristol, United Kingdom
| | - Kalina M. Biernacka
- IGFs & Metabolic Endocrinology Group, Bristol Medical School, Translational Health Sciences, Southmead Hospital, University of Bristol, Bristol, United Kingdom
| | - Shelley J. Allen-Birt
- Molecular Neurobiology Group, Bristol Medical School, Translational Health Sciences, Southmead Hospital, University of Bristol, Bristol, United Kingdom
| | - Claire M. Perks
- IGFs & Metabolic Endocrinology Group, Bristol Medical School, Translational Health Sciences, Southmead Hospital, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
288
|
Kashyap S, Zeidler JD, Chini CCS, Chini EN. Implications of the PAPP-A-IGFBP-IGF-1 pathway in the pathogenesis and treatment of polycystic kidney disease. Cell Signal 2020; 73:109698. [PMID: 32569826 DOI: 10.1016/j.cellsig.2020.109698] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 12/19/2022]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is one of the most common genetic diseases implicated in the development of end stage renal disease (ESRD). Although FDA has recently approved a drug against ADPKD, there is still a great need for development of alternative management strategies for ADPKD. Understanding the different mechanisms that lead to cystogenesis and cyst expansion in ADPKD is imperative to develop new therapies against ADPKD. Recently, we demonstrated that caloric restriction can prevent the development of cystic disease in animal models of ADPKD and through these studies identified a new role for pregnancy associated plasma protein-A (PAPP-A), a component of the insulin-like growth factors (IGF) pathway, in the pathogenesis of this disease. The PAPP-A-IGF pathway plays an important role in regulation of cell growth, differentiation, and transformation and dysregulation of this pathway has been implicated in many diseases. Several indirect studies support the involvement of IGF-1 in the pathogenesis of ADPKD. However, it was only recently that we described a direct role for a component of this pathway in pathogenesis of ADPKD, opening a new avenue for the therapeutic approaches for this cystic disease. The present literature review will critically discuss the evidence that supports the role of components of IGF pathway in the pathogenesis of ADPKD and discuss the pharmacological implications of PAPP-A-IGF axis in this disease.
Collapse
Affiliation(s)
- Sonu Kashyap
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Julianna D Zeidler
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Claudia C S Chini
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Eduardo Nunes Chini
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA.
| |
Collapse
|
289
|
Marchant C, Anderson P, Schwarz Q, Wiszniak S. Vessel-derived angiocrine IGF1 promotes Meckel's cartilage proliferation to drive jaw growth during embryogenesis. Development 2020; 147:dev.190488. [PMID: 32439763 PMCID: PMC7295590 DOI: 10.1242/dev.190488] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 04/23/2020] [Indexed: 12/18/2022]
Abstract
Craniofacial development is a complex morphogenic process that requires highly orchestrated interactions between multiple cell types. Blood vessel-derived angiocrine factors are known to promote proliferation of chondrocytes in Meckel's cartilage to drive jaw outgrowth, however the specific factors controlling this process remain unknown. Here, we use in vitro and ex vivo cell and tissue culture, as well as genetic mouse models, to identify IGF1 as a novel angiocrine factor directing Meckel's cartilage growth during embryonic development. We show that IGF1 is secreted by blood vessels and that deficient IGF1 signalling underlies mandibular hypoplasia in Wnt1-Cre; Vegfafl/fl mice that exhibit vascular and associated jaw defects. Furthermore, conditional removal of IGF1 from blood vessels causes craniofacial defects including a shortened mandible, and reduced proliferation of Meckel's cartilage chondrocytes. This demonstrates a crucial angiocrine role for IGF1 during craniofacial cartilage growth, and identifies IGF1 as a putative therapeutic for jaw and/or cartilage growth disorders.
Collapse
Affiliation(s)
- Ceilidh Marchant
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia
| | - Peter Anderson
- Australian Craniofacial Unit, Women's and Children's Hospital, North Adelaide, SA 5006, Australia
| | - Quenten Schwarz
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia
| | - Sophie Wiszniak
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia
| |
Collapse
|
290
|
Price D, Dorandish S, Williams A, Iwaniec B, Stephens A, Marshall K, Guthrie J, Heyl D, Evans HG. Humanin Blocks the Aggregation of Amyloid-β Induced by Acetylcholinesterase, an Effect Abolished in the Presence of IGFBP-3. Biochemistry 2020; 59:1981-2002. [PMID: 32383868 PMCID: PMC8193794 DOI: 10.1021/acs.biochem.0c00274] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
It is known that the humanin (HN) peptide binding to amyloid-β (Aβ) protects against its cytotoxic effects, while acetylcholinesterase (AChE) binding to Aβ increases its aggregation and cytotoxicity. HN is also known to bind the insulin-like growth factor binding protein-3 (IGFBP-3). Here, we examined the regulation of Aβ conformations by HN, AChE, and IGFBP-3 both in vitro and in the conditioned media from A549 and H1299 lung cancer cells. Our in vitro results showed the following: IGFBP-3 binds HN and blocks it from binding Aβ in the absence or presence of AChE; HN and AChE can simultaneously bind Aβ but not when in the presence of IGFBP-3; HN is unable to reduce the aggregation of Aβ in the presence of IGFBP-3; and HN abolishes the aggregation of Aβ induced by the addition of AChE in the absence of IGFBP-3. In the media, AChE and HN can simultaneously bind Aβ. While both AChE and HN are detected when using 6E10 Aβ antibodies, only AChE is detected when using the Aβ 17-24 antibody 4G8, the anti-oligomer A11, and the anti-amyloid fibril LOC antibodies. No signal was observed for IGFBP-3 with any of the anti-amyloid antibodies used. Exogenously added IGFBP-3 reduced the amount of HN found in a complex when using 6E10 antibodies and correlated with a concomitant increase in the amyloid oligomers. Immunodepletion of HN from the media of the A549 and H1299 cells increased the relative abundance of the oligomer vs the total amount of Aβ, the A11-positive prefibrillar oligomers, and to a lesser extent the LOC-positive fibrillar oligomers, and was also correlated with diminished cell viability and increased apoptosis.
Collapse
Affiliation(s)
- Deanna Price
- Department of Chemistry, Eastern Michigan University, Ypsilanti, Michigan 48197, United States
| | - Sadaf Dorandish
- Department of Chemistry, Eastern Michigan University, Ypsilanti, Michigan 48197, United States
| | - Asana Williams
- Department of Chemistry, Eastern Michigan University, Ypsilanti, Michigan 48197, United States
| | - Brandon Iwaniec
- Department of Chemistry, Eastern Michigan University, Ypsilanti, Michigan 48197, United States
| | - Alexis Stephens
- Department of Chemistry, Eastern Michigan University, Ypsilanti, Michigan 48197, United States
| | - Keyan Marshall
- Department of Chemistry, Eastern Michigan University, Ypsilanti, Michigan 48197, United States
| | - Jeffrey Guthrie
- Department of Chemistry, Eastern Michigan University, Ypsilanti, Michigan 48197, United States
| | - Deborah Heyl
- Department of Chemistry, Eastern Michigan University, Ypsilanti, Michigan 48197, United States
| | - Hedeel Guy Evans
- Department of Chemistry, Eastern Michigan University, Ypsilanti, Michigan 48197, United States
| |
Collapse
|
291
|
Zhang WB, Aleksic S, Gao T, Weiss EF, Demetriou E, Verghese J, Holtzer R, Barzilai N, Milman S. Insulin-like Growth Factor-1 and IGF Binding Proteins Predict All-Cause Mortality and Morbidity in Older Adults. Cells 2020; 9:cells9061368. [PMID: 32492897 PMCID: PMC7349399 DOI: 10.3390/cells9061368] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 05/27/2020] [Accepted: 05/28/2020] [Indexed: 01/02/2023] Open
Abstract
While the growth hormone/insulin-like growth factor-1 (GH/IGF-1) pathway plays essential roles in growth and development, diminished signaling via this pathway in model organisms extends lifespan and health-span. In humans, circulating IGF-1 and IGF-binding proteins 3 and 1 (IGFBP-3 and 1), surrogate measures of GH/IGF-1 system activity, have not been consistently associated with morbidity and mortality. In a prospective cohort of independently-living older adults (n = 840, mean age 76.1 ± 6.8 years, 54.5% female, median follow-up 6.9 years), we evaluated the age- and sex-adjusted hazards for all-cause mortality and incident age-related diseases, including cardiovascular disease, diabetes, cancer, and multiple-domain cognitive impairment (MDCI), as predicted by baseline total serum IGF-1, IGF-1/IGFBP-3 molar ratio, IGFBP-3, and IGFBP-1 levels. All-cause mortality was positively associated with IGF-1/IGFBP-3 molar ratio (HR 1.28, 95% CI 1.05–1.57) and negatively with IGFBP-3 (HR 0.82, 95% CI 0.680–0.998). High serum IGF-1 predicted greater risk for MDCI (HR 1.56, 95% CI 1.08–2.26) and composite incident morbidity (HR 1.242, 95% CI 1.004–1.538), whereas high IGFBP-1 predicted lower risk for diabetes (HR 0.50, 95% CI 0.29–0.88). In conclusion, higher IGF-1 levels and bioavailability predicted mortality and morbidity risk, supporting the hypothesis that diminished GH/IGF-1 signaling may contribute to human longevity and health-span.
Collapse
Affiliation(s)
- William B. Zhang
- Department of Medicine, Division of Endocrinology, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (W.B.Z.); (S.A.); (T.G.); (N.B.)
| | - Sandra Aleksic
- Department of Medicine, Division of Endocrinology, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (W.B.Z.); (S.A.); (T.G.); (N.B.)
| | - Tina Gao
- Department of Medicine, Division of Endocrinology, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (W.B.Z.); (S.A.); (T.G.); (N.B.)
| | - Erica F. Weiss
- Department of Neurology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.F.W.); (J.V.); (R.H.)
| | - Eleni Demetriou
- Ferkauf Graduate School of Psychology, Yeshiva University, New York, NY 10033, USA;
| | - Joe Verghese
- Department of Neurology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.F.W.); (J.V.); (R.H.)
- Department of Medicine, Division of Geriatrics, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Roee Holtzer
- Department of Neurology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.F.W.); (J.V.); (R.H.)
- Ferkauf Graduate School of Psychology, Yeshiva University, New York, NY 10033, USA;
| | - Nir Barzilai
- Department of Medicine, Division of Endocrinology, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (W.B.Z.); (S.A.); (T.G.); (N.B.)
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Sofiya Milman
- Department of Medicine, Division of Endocrinology, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (W.B.Z.); (S.A.); (T.G.); (N.B.)
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Correspondence:
| |
Collapse
|
292
|
Payne JA, Proszkowiec-Weglarz M, Ellestad LE. Delayed access to feed alters gene expression associated with hormonal signaling, cellular differentiation, and protein metabolism in muscle of newly hatch chicks. Gen Comp Endocrinol 2020; 292:113445. [PMID: 32135160 DOI: 10.1016/j.ygcen.2020.113445] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 02/18/2020] [Accepted: 02/27/2020] [Indexed: 10/24/2022]
Abstract
Birds rely solely on utilization of the yolk sac as a means of nutritional support throughout embryogenesis and early post-hatch, before first feeding occurs. Newly hatched broiler (meat-type) chickens are frequently not given immediate access to feed, and this can result in numerous alterations to developmental processes, including those that occur in muscle. The objective of this study was to characterize the gene expression profile of newly hatched chicks' breast muscle with regards to hormonal regulation of growth and metabolism and development and differentiation of muscle tissue, and determine impacts of delayed access to feed on these profiles. Within 3 h of hatch, birds were placed in battery pens and given immediate access to feed (Fed) or delayed access to feed for 48 h (Delayed Fed). Breast muscle collected from male birds at hatch, or 4 h, 1 day (D), 2D, 4D, and 8D after hatch was used for analysis of mRNA expression by reverse transcription-quantitative PCR. Under fully fed conditions, insulin-like growth factor receptor and leptin receptor mRNA expression decreased as birds aged; however, delayed access to feed resulted in prolonged upregulation of these genes so their mRNA levels were higher in Delayed Fed birds at 2D. These expression profiles suggest that delayed feed access alters sensitivity to hormones that may regulate muscle development. Myogenin, a muscle differentiation factor, showed increasing mRNA expression in Fed birds through 2D, after which expression decreased. A similar expression pattern in Delayed Fed birds was deferred until 4D. Levels of myostatin, a negative regulator of muscle growth, increased in Fed birds starting at 2D, while levels in Delayed Fed birds began to increase at 4D. In Fed birds, levels of transcripts for two genes associated with protein catabolism, F-box protein 32 and forkhead box O3, were lower at 2D, while Delayed Fed mRNA levels did not decrease until 4D. Mechanistic target of rapamycin mRNA levels decreased from 1D through 8D in both treatments, except for a transient increase in the Delayed Fed birds between 1D and 2D. These data suggest that within breast muscle, delayed feeding alters hormonal signaling, interrupts tissue differentiation, postpones onset of growth, and may lead to increased protein catabolism. Together, these processes could ultimately contribute to a reduction in proper growth and development of birds not given feed immediately after hatch, and ultimately hinder the long-term potential of muscle accretion in meat type birds.
Collapse
Affiliation(s)
- Jason A Payne
- Department of Poultry Science, University of Georgia, 110 Cedar St, Athens, GA 30601, USA.
| | - Monika Proszkowiec-Weglarz
- Animal Biosciences and Biotechnology Laboratory, United States Department of Agriculture, Agricultural Research Service, Northeast Area, 10300 Baltimore Ave, BARC-East, Bldg 200, Beltsville, MD 20705, USA.
| | - Laura E Ellestad
- Department of Poultry Science, University of Georgia, 110 Cedar St, Athens, GA 30601, USA.
| |
Collapse
|
293
|
Jin L, Shen F, Weinfeld M, Sergi C. Insulin Growth Factor Binding Protein 7 (IGFBP7)-Related Cancer and IGFBP3 and IGFBP7 Crosstalk. Front Oncol 2020; 10:727. [PMID: 32500027 PMCID: PMC7242731 DOI: 10.3389/fonc.2020.00727] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/16/2020] [Indexed: 12/17/2022] Open
Abstract
The insulin/insulin-like growth factors (IGFs) have crucial tasks in the growth, differentiation, and proliferation of healthy and pernicious cells. They are involved in coordinated complexes, including receptors, ligands, binding proteins, and proteases. However, the systems can become dysregulated in tumorigenesis. Insulin-like growth factor-binding protein 7 (IGFBP7) is a protein belonging to the IGFBP superfamily (also termed GFBP-related proteins). Numerous studies have provided evidence that IGFBP3 and IGFBP7 are involved in a variety of cancers, including hepatocellular carcinoma (HCC), breast cancer, gastroesophageal cancer, colon cancer, prostate cancer, among many others. Still, very few suggest an interaction between these two molecules. In studying several cancer types in our laboratories, we found that both proteins share some crucial signaling pathways. The objective of this review is to present a comprehensive overview of the relationship between IGFBP7 and cancer, as well as highlighting IGFBP3 crosstalk with IGFBP7 reported in recent studies.
Collapse
Affiliation(s)
- Li Jin
- Department of Laboratory Medicine, Shiyan Taihe Hospital, College of Biomedical Engineering, Hubei University of Medicine, Shiyan, China
| | - Fan Shen
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Michael Weinfeld
- Division of Experimental Oncology, Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB, Canada
| | - Consolato Sergi
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
- Department of Orthopedics, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- Key Laboratory of Fermentation Engineering, National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, China
- Stollery Children's Hospital, University Alberta Hospital, Edmonton, AB, Canada
| |
Collapse
|
294
|
Dabrosin N, Dabrosin C. Postmenopausal Dense Breasts Maintain Premenopausal Levels of GH and Insulin-like Growth Factor Binding Proteins in Vivo. J Clin Endocrinol Metab 2020; 105:5695904. [PMID: 31900484 DOI: 10.1210/clinem/dgz323] [Citation(s) in RCA: 3] [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: 10/03/2019] [Accepted: 01/01/2020] [Indexed: 12/24/2022]
Abstract
CONTEXT Dense breast tissue is associated with 4 to 6 times higher risk of breast cancer by poorly understood mechanisms. No preventive therapy for this high-risk group is available. After menopause, breast density decreases due to involution of the mammary gland. In dense breast tissue, this process is haltered by undetermined biological actions. Growth hormone (GH) and insulin-like binding proteins (IGFBPs) play major roles in normal mammary gland development, but their roles in maintaining breast density are unknown. OBJECTIVE To reveal in vivo levels of GH, IGFBPs, and other pro-tumorigenic proteins in the extracellular microenvironment in breast cancer, in normal breast tissue with various breast density in postmenopausal women, and premenopausal breasts. We also sought to determine possible correlations between these determinants. SETTING AND DESIGN Microdialysis was used to collect extracellular in vivo proteins intratumorally from breast cancers before surgery and from normal human breast tissue from premenopausal women and postmenopausal women with mammographic dense or nondense breasts. RESULTS Estrogen receptor positive breast cancers exhibited increased extracellular GH (P < .01). Dense breasts of postmenopausal women exhibited similar levels of GH as premenopausal breasts and significantly higher levels than in nondense breasts (P < .001). Similar results were found for IGFBP-1, -2, -3, and -7 (P < .01) and for IGFBP-6 (P <.05). Strong positive correlations were revealed between GH and IGFBPs and pro-tumorigenic matrix metalloproteinases, urokinase-type plasminogen activator, Interleukin 6, Interleukin 8, and vascular endothelial growth factor in normal breast tissue. CONCLUSIONS GH pathways may be targetable for cancer prevention therapeutics in postmenopausal women with dense breast tissue.
Collapse
Affiliation(s)
- Nina Dabrosin
- Department of Plastic and Breast Surgery, Aarhus University Hospital, Aarhus, Denmark
| | - Charlotta Dabrosin
- Department of Oncology and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
295
|
Mollinedo F, Gajate C. Lipid rafts as signaling hubs in cancer cell survival/death and invasion: implications in tumor progression and therapy: Thematic Review Series: Biology of Lipid Rafts. J Lipid Res 2020; 61:611-635. [PMID: 33715811 PMCID: PMC7193951 DOI: 10.1194/jlr.tr119000439] [Citation(s) in RCA: 179] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 01/17/2020] [Indexed: 12/13/2022] Open
Abstract
Cholesterol/sphingolipid-rich membrane domains, known as lipid rafts or membrane rafts, play a critical role in the compartmentalization of signaling pathways. Physical segregation of proteins in lipid rafts may modulate the accessibility of proteins to regulatory or effector molecules. Thus, lipid rafts serve as sorting platforms and hubs for signal transduction proteins. Cancer cells contain higher levels of intracellular cholesterol and lipid rafts than their normal non-tumorigenic counterparts. Many signal transduction processes involved in cancer development (insulin-like growth factor system and phosphatidylinositol 3-kinase-AKT) and metastasis [cluster of differentiation (CD)44] are dependent on or modulated by lipid rafts. Additional proteins playing an important role in several malignant cancers (e.g., transmembrane glycoprotein mucin 1) are also being detected in association with lipid rafts, suggesting a major role of lipid rafts in tumor progression. Conversely, lipid rafts also serve as scaffolds for the recruitment and clustering of Fas/CD95 death receptors and downstream signaling molecules leading to cell death-promoting raft platforms. The partition of death receptors and downstream signaling molecules in aggregated lipid rafts has led to the formation of the so-called cluster of apoptotic signaling molecule-enriched rafts, or CASMER, which leads to apoptosis amplification and can be pharmacologically modulated. These death-promoting rafts can be viewed as a linchpin from which apoptotic signals are launched. In this review, we discuss the involvement of lipid rafts in major signaling processes in cancer cells, including cell survival, cell death, and metastasis, and we consider the potential of lipid raft modulation as a promising target in cancer therapy.
Collapse
Affiliation(s)
- Faustino Mollinedo
- Laboratory of Cell Death and Cancer Therapy, Department of Molecular Biomedicine, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Cientificas (CSIC), E-28040 Madrid, Spain. mailto:
| | - Consuelo Gajate
- Laboratory of Cell Death and Cancer Therapy, Department of Molecular Biomedicine, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Cientificas (CSIC), E-28040 Madrid, Spain
| |
Collapse
|
296
|
Liu C, Li S, Noer PR, Kjaer-Sorensen K, Juhl AK, Goldstein A, Ke C, Oxvig C, Duan C. The metalloproteinase Papp-aa controls epithelial cell quiescence-proliferation transition. eLife 2020; 9:e52322. [PMID: 32293560 PMCID: PMC7185994 DOI: 10.7554/elife.52322] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 04/11/2020] [Indexed: 02/06/2023] Open
Abstract
Human patients carrying PAPP-A2 inactivating mutations have low bone mineral density. The underlying mechanisms for this reduced calcification are poorly understood. Using a zebrafish model, we report that Papp-aa regulates bone calcification by promoting Ca2+-transporting epithelial cell (ionocyte) quiescence-proliferation transition. Ionocytes, which are normally quiescent, re-enter the cell cycle under low [Ca2+] stress. Genetic deletion of Papp-aa, but not the closely related Papp-ab, abolished ionocyte proliferation and reduced calcified bone mass. Loss of Papp-aa expression or activity resulted in diminished IGF1 receptor-Akt-Tor signaling in ionocytes. Under low Ca2+ stress, Papp-aa cleaved Igfbp5a. Under normal conditions, however, Papp-aa proteinase activity was suppressed and IGFs were sequestered in the IGF/Igfbp complex. Pharmacological disruption of the IGF/Igfbp complex or adding free IGF1 activated IGF signaling and promoted ionocyte proliferation. These findings suggest that Papp-aa-mediated local Igfbp5a cleavage functions as a [Ca2+]-regulated molecular switch linking IGF signaling to bone calcification by stimulating epithelial cell quiescence-proliferation transition under low Ca2+ stress.
Collapse
Affiliation(s)
- Chengdong Liu
- Department of Molecular, Cellular, and Developmental Biology, University of MichiganAnn ArborUnited States
| | - Shuang Li
- Department of Molecular, Cellular, and Developmental Biology, University of MichiganAnn ArborUnited States
- College of Ocean and Earth Sciences, Xiamen UniversityXiamenChina
| | | | | | - Anna Karina Juhl
- Department of Molecular Biology and Genetics, Aarhus UniversityAarhusDenmark
| | - Allison Goldstein
- Department of Molecular, Cellular, and Developmental Biology, University of MichiganAnn ArborUnited States
| | - Caihuan Ke
- College of Ocean and Earth Sciences, Xiamen UniversityXiamenChina
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus UniversityAarhusDenmark
| | - Cunming Duan
- Department of Molecular, Cellular, and Developmental Biology, University of MichiganAnn ArborUnited States
| |
Collapse
|
297
|
Xie M, Zhong Y, Xue Q, Wu M, Deng X, O Santos H, Tan SC, Kord-Varkaneh H, Jiao P. Impact of dehydroepianrosterone (DHEA) supplementation on serum levels of insulin-like growth factor 1 (IGF-1): A dose-response meta-analysis of randomized controlled trials. Exp Gerontol 2020; 136:110949. [PMID: 32304719 DOI: 10.1016/j.exger.2020.110949] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/11/2020] [Accepted: 04/12/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND AIM Inconsistencies exist with regard to the influence of dehydroepiandrosterone (DHEA) supplementation on insulin-like growth factor 1 (IGF-1) levels. The inconsistencies could be attributed to several factors, such as dosage, gender, and duration of intervention, among others. To address these inconsistencies, we conducted a systematic review and meta-analysis to combine findings from randomized controlled trials (RCTs) on this topic. METHODS Electronic databases (Scopus, PubMed/Medline, Web of Science, Embase and Google Scholar) were searched for relevant literature published up to February 2020. RESULTS Twenty-four qualified trials were included in this meta-analysis. It was found that serum IGF-1 levels were significantly increased in the DHEA group compared to the control (weighted mean differences (WMD): 16.36 ng/ml, 95% CI: 8.99, 23.74; p = .000). Subgroup analysis revealed that a statistically significant increase in serum IGF-1 levels was found only in women (WMD: 23.30 ng/ml, 95% CI: 13.75, 32.87); in participants who supplemented 50 mg/d DHEA (WMD: 15.75 ng/ml, 95% CI: 7.61, 23.89); in participants undergoing DHEA intervention for >12 weeks (WMD: 17.2 ng/ml, 95% CI: 8.02, 26.22); in participants without an underlying comorbidity (WMD: 19.11 ng/ml, 95% CI: 10.69, 27.53); and in participants over the age of 60 years (WMD: 19.79 ng/ml, 95% CI: 9.86, 29.72). CONCLUSION DHEA supplementation may increase serum IGF-I levels especially in women and older subjects. However, further studies are warranted before DHEA can be recommended for clinical use.
Collapse
Affiliation(s)
- Min Xie
- Operating Room Nursing Department, Xiangya Third Hospital of Central South University, Changsha, Hunan 400013, China
| | - Yanfa Zhong
- Department of Laboratory Medicine, Dezhou Hospital of Traditional Chinese Medicine, Dezhou, Shandong 253000, China
| | - Qing Xue
- Department of Laboratory Medicine, Dezhou Hospital of Traditional Chinese Medicine, Dezhou, Shandong 253000, China
| | - Meirong Wu
- Operating Room Nursing Department, Xiangya Third Hospital of Central South University, Changsha, Hunan 400013, China
| | - Xiaoxian Deng
- Operating Room Nursing Department, Xiangya Third Hospital of Central South University, Changsha, Hunan 400013, China
| | - Heitor O Santos
- School of Medicine, Federal University of Uberlandia (UFU), Uberlandia, Minas Gerais, Brazil
| | - Shing Cheng Tan
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Hamed Kord-Varkaneh
- Student Research Committee, Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Peng Jiao
- Department of Endocrinology, Dezhou Hospital of Traditional Chinese Medicine, Dezhou, Shandong 253000, China.
| |
Collapse
|
298
|
Abstract
CONTEXT Recent advances in genetics and genomics present unique opportunities for enhancing knowledge of human physiology and disease susceptibility. An outstanding example of these new insights may be seen in the study of human height, of which it has been estimated that approximately 80% is genetically determined. Over the past decade, large-scale population analyses have led to the identification of novel variation in genes and loci individually associated with changes in adult height of as much as 2 cm. OBJECTIVE To assess these same variants in the genomes of 213 158 individuals compiled by the Genome Aggregation Database (GnomAD) consortium, representing different population groups from around the world. RESULTS The majority of these height-changing alleles are substantially less prevalent in GnomAD than found previously in other cohorts, with 4 of 5 amino acid substitution variants with the largest impact on adult height being more frequent in the European population than in other groups. CONCLUSIONS A larger-scale analysis of individuals from diverse backgrounds will be necessary to ensure a full and accurate understanding of the genetic underpinnings of human height throughout the world, and additional studies will be needed to discern the biochemical and molecular mechanisms governing the physiological processes that explain how these variant proteins might selectively impact the biology of the growth plate. Broader understanding of the genetics of height also should set the stage for more comprehensive investigation into the causes of prevalent polygenic human diseases.
Collapse
Affiliation(s)
- Peter Rotwein
- Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech Health University Health Sciences Center, El Paso, Texas
| |
Collapse
|
299
|
Abstract
The insulin-like growth factors (IGFs; IGF1/IGF2), known for their regulation of cell and organismal growth and development, are evolutionarily conserved ligands with equivalent peptides present in flies (
D. melanogaster), worms (
C. elegans) among others. Two receptor tyrosine kinases, the IGF1 receptor and the insulin receptor mediate the actions of these ligands with a family of IGF binding proteins serving as selective inhibitors of IGF1/2. This treatise reviews recent findings on IGF signaling in cancer biology and central nervous system function. This includes overexpression of IGF1 receptors in enhancing tumorigenesis, acquired resistance and contributions to metastasis in multiple cancer types. There is accumulating evidence that insulin resistance, a hallmark of type 2 diabetes, occurs in the central nervous system, independent of systemic insulin resistance and characterized by reduced insulin and IGF1 receptor signaling, and may contribute to dementias including Alzheimer’s Disease and cognitive impairment. Controversy over the role(s) of IGF signaling in cancer and whether its inhibition would be of benefit, still persist and extend to IGF1’s role in longevity and central nervous system function.
Collapse
Affiliation(s)
- Steven A Rosenzweig
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, 29425, USA
| |
Collapse
|
300
|
Poreba E, Durzynska J. Nuclear localization and actions of the insulin-like growth factor 1 (IGF-1) system components: Transcriptional regulation and DNA damage response. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2020; 784:108307. [PMID: 32430099 DOI: 10.1016/j.mrrev.2020.108307] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 12/14/2022]
Abstract
Insulin-like growth factor (IGF) system stimulates growth, proliferation, and regulates differentiation of cells in a tissue-specific manner. It is composed of two insulin-like growth factors (IGF-1 and IGF-2), six insulin-like growth factor-binding proteins (IGFBPs), and two insulin-like growth factor receptors (IGF-1R and IGF-2R). IGF actions take place mostly through the activation of the plasma membrane-bound IGF-Rs by the circulating ligands (IGFs) released from the IGFBPs that stabilize their levels in the serum. This review focuses on the IGF-1 part of the system. The IGF-1 gene, which is expressed mainly in the liver as well as in other tissues, comprises six alternatively spliced exons that code for three protein isoforms (pro-IGF-1A, pro-IGF-1B, and pro-IGF-1C), which are processed to mature IGF-1 and E-peptides. The IGF-1R undergoes autophosphorylation, resulting in a signaling cascade involving numerous cytoplasmic proteins such as AKT and MAPKs, which regulate the expression of target genes. However, a more complex picture of the axis has recently emerged with all its components being translocated to the nuclear compartment. IGF-1R takes part in the regulation of gene expression by forming transcription complexes, modifying the activity of chromatin remodeling proteins, and participating in DNA damage tolerance mechanisms. Four IGFBPs contain a nuclear localization signal (NLS), which targets them to the nucleus, where they regulate gene expression (IGFBP-2, IGFBP-3, IGFBP-5, IGFBP-6) and DNA damage repair (IGFBP-3 and IGFBP-6). Last but not least, the IGF-1B isoform has been reported to be localized in the nuclear compartment. However, no specific molecular actions have been assigned to the nuclear pro-IGF-1B or its derivative EB peptide. Therefore, further studies are needed to shed light on their nuclear activity. These recently uncovered nuclear actions of different components of the IGF-1 axis are relevant in cancer cell biology and are discussed in this review.
Collapse
Affiliation(s)
- Elzbieta Poreba
- Institute of Experimental Biology, Faculty of Biology, Adam Mickiewicz University, ul. Uniwersytetu Poznańskiego 6, 61-614, Poznań, Poland.
| | - Julia Durzynska
- Institute of Experimental Biology, Faculty of Biology, Adam Mickiewicz University, ul. Uniwersytetu Poznańskiego 6, 61-614, Poznań, Poland.
| |
Collapse
|