1
|
Karamanolis NN, Kounatidis D, Vallianou NG, Alexandropoulos K, Kovlakidi E, Kaparou P, Karampela I, Stratigou T, Dalamaga M. Paraneoplastic hypoglycemia: An overview for optimal clinical guidance. Metabol Open 2024; 23:100305. [PMID: 39185033 PMCID: PMC11342107 DOI: 10.1016/j.metop.2024.100305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/15/2024] [Revised: 07/28/2024] [Accepted: 07/29/2024] [Indexed: 08/27/2024] Open
Abstract
Paraneoplastic hypoglycemia, also known as non-islet cell tumor hypoglycemia (NICTH), is a rare but critical condition occurring in patients with different types of malignancy. This condition is commonly linked to tumors producing insulin-like growth (IGF) factors, particularly IGF-2 and its precursors, which disrupt glucose homeostasis and lead to excessive glucose consumption. The diagnosis typically involves documenting symptomatic hypoglycemia and ruling out other potential causes. Essential diagnostic tools include imaging studies and laboratory tests, specifically measuring IGF-2 levels and the IGF-2:IGF-1 ratio. Treatment strategies for NICTH are multifaceted and may include surgical resection of the tumor if feasible, pharmacological interventions such as corticosteroids to suppress IGF-2 production, or supportive measures to manage acute hypoglycemic episodes. Novel therapeutic approaches targeting IGF-2, such as monoclonal antibodies or siRNA, are also being explored and hold promise for future treatment options. This review aims to enhance understanding of paraneoplastic hypoglycemia, focusing on its pathogenesis and diagnosis, to guide optimal medical treatment.
Collapse
Affiliation(s)
- Nikolaos Nektarios Karamanolis
- 2th Department of Internal Medicine, Medical School, National & Kapodistrian University of Athens, Hippokratio General Hospital, 11527, Athens, Greece
| | - Dimitris Kounatidis
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, 11527, Athens, Greece
| | - Natalia G. Vallianou
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126, Athens, Greece
| | - Konstantinos Alexandropoulos
- 2th Department of Internal Medicine, Medical School, National & Kapodistrian University of Athens, Hippokratio General Hospital, 11527, Athens, Greece
| | - Eleni Kovlakidi
- 2th Department of Internal Medicine, Medical School, National & Kapodistrian University of Athens, Hippokratio General Hospital, 11527, Athens, Greece
| | - Pinelopi Kaparou
- 2th Department of Internal Medicine, Medical School, National & Kapodistrian University of Athens, Hippokratio General Hospital, 11527, Athens, Greece
| | - Irene Karampela
- Second Department of Critical Care, Attikon General University Hospital, Medical School, National and Kapodistrian University of Athens, 12462, Athens, Greece
| | - Theodora Stratigou
- Department of Endocrinology and Metabolism, Evangelismos General Hospital, 10676, Athens, Greece
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece
| |
Collapse
|
2
|
Erdogan MA, Nesil P, Altuntas I, Sirin C, Uyanikgil Y, Erbas O. Amelioration of propionic acid-induced autism spectrum disorder in rats through dapagliflozin: The role of IGF-1/IGFBP-3 and the Nrf2 antioxidant pathway. Neuroscience 2024; 554:16-25. [PMID: 39004410 DOI: 10.1016/j.neuroscience.2024.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/16/2024] [Revised: 07/07/2024] [Accepted: 07/09/2024] [Indexed: 07/16/2024]
Abstract
The biological effects of dapagliflozin, a sodium-glucose cotransporter-2 (SGLT2) inhibitor, reveal its antioxidant and anti-inflammatory properties, suggesting therapeutic benefits beyond glycemic control. This study explores the neuroprotective effects of dapagliflozin in a rat model of autism spectrum disorder (ASD) induced by propionic acid (PPA), characterized by social interaction deficits, communication challenges, repetitive behaviors, cognitive impairments, and oxidative stress. Our research aims to find effective treatments for ASD, a condition with limited therapeutic options and significant impacts on individuals and families. PPA induces ASD-like symptoms in rodents, mimicking biochemical and behavioral features of human ASD. This study explores dapagliflozin's potential to mitigate these symptoms, providing insights into novel therapeutic avenues. The findings demonstrate that dapagliflozin enhances the activation of the nuclear factor erythroid 2-related factor 2 (Nrf2) antioxidant pathway and increases levels of neurotrophic and growth factors such as brain-derived neurotrophic factor (BDNF), insulin-like growth factor-1 (IGF-1), and insulin-like growth factor-binding protein-3 (IGFBP-3). Additionally, dapagliflozin reduces pro-inflammatory cytokines, including tumor necrosis factor-alpha (TNF-α) and interleukin-17 (IL-17), and decreases the oxidative stress marker malondialdehyde (MDA). Dapagliflozin's antioxidant properties support cognitive functions by modulating apoptotic mechanisms and enhancing antioxidant capacity. These combined effects contribute to reducing learning and memory impairments in PPA-induced ASD, highlighting dapagliflozin's potential as an adjunctive therapy for oxidative stress and inflammation-related cognitive decline in ASD. This study underscores the importance of exploring new therapeutic strategies targeting molecular pathways involved in the pathophysiology of ASD, potentially improving the quality of life for individuals affected by this disorder.
Collapse
Affiliation(s)
- Mumin Alper Erdogan
- Department of Physiology, Izmir Katip Celebi University, Faculty of Medicine, Izmir, Turkey.
| | - Pemra Nesil
- Istanbul University, Faculty of Medicine, Istanbul, Turkey
| | | | - Cansın Sirin
- Department of Histology and Embryology, Ege University, Faculty of Medicine, Izmir, Turkey
| | - Yigit Uyanikgil
- Department of Histology and Embryology, Ege University, Faculty of Medicine, Izmir, Turkey
| | - Oytun Erbas
- Demiroglu Bilim University, Department of Physiology, İstanbul, Turkey
| |
Collapse
|
3
|
Janssen JAMJL. The Impact of Westernization on the Insulin/IGF-I Signaling Pathway and the Metabolic Syndrome: It Is Time for Change. Int J Mol Sci 2023; 24:ijms24054551. [PMID: 36901984 PMCID: PMC10003782 DOI: 10.3390/ijms24054551] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/27/2023] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023] Open
Abstract
The metabolic syndrome is a cluster of overlapping conditions resulting in an increased incidence of type 2 diabetes, cardiovascular disease, and cancer. In the last few decades, prevalence of the metabolic syndrome in the Western world has reached epidemic proportions and this is likely due to alterations in diet and the environment as well as decreased physical activity. This review discusses how the Western diet and lifestyle (Westernization) has played an important etiological role in the pathogenesis of the metabolic syndrome and its consequences by exerting negative effects on activity of the insulin-insulin-like growth factor-I (insulin-IGF-I) system. It is further proposed that interventions that normalize/reduce activity of the insulin-IGF-I system may play a key role in the prevention and treatment of the metabolic syndrome. For successful prevention, limitation, and treatment of the metabolic syndrome, the focus should be primarily on changing our diets and lifestyle in accordance with our genetic make-up, formed in adaptation to Paleolithic diets and lifestyles during a period of several million years of human evolution. Translating this insight into clinical practice, however, requires not only individual changes in our food and lifestyle, starting in pediatric populations at a very young age, but also requires fundamental changes in our current health systems and food industry. Change is needed: primary prevention of the metabolic syndrome should be made a political priority. New strategies and policies should be developed to stimulate and implement behaviors encouraging the sustainable use of healthy diets and lifestyles to prevent the metabolic syndrome before it develops.
Collapse
Affiliation(s)
- Joseph A M J L Janssen
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
4
|
Wang Z, Tan D, Wang F, Guo S, Liu J, Cuthbertson AGS, Qiu B, Sang W. Insulin peptides and their receptors regulate ovarian development and oviposition behavior in Diaphorina citri. INSECT SCIENCE 2023; 30:95-108. [PMID: 35510515 PMCID: PMC10084437 DOI: 10.1111/1744-7917.13048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Academic Contribution Register] [Received: 11/11/2021] [Revised: 03/19/2022] [Accepted: 04/02/2022] [Indexed: 06/14/2023]
Abstract
Diaphorina citri is an important vector of Citrus Huanglongbing (HLB) disease. After feeding on young host plant shoots, the population of D. citri can increase significantly. Females also only lay eggs on young shoots. However, there are few studies on the mechanism of this phenomenon. Exogenous nutrient signals can affect the insulin signaling system of D. citri after feeding on young shoots. In this study, the expression of upstream factors DcILP1, DcILP2, and DcIR in the insulin signaling system of D. citri was upregulated after feeding on young shoots. After being silenced by RNA interference technology, the results showed that the number of oviposited eggs of D. citri was significantly decreased and the ovarian development was inhibited with severe vacuolation. In addition, detection using quantitative reverse transcription-polymerase chain reaction showed that the upstream regulatory gene DcRheb of the target of rapamycin (TOR) pathway and the downstream reproduction-related DcVg gene were also significantly downregulated. These results suggest that feeding upon young shoots may upregulate the expression levels of upstream factors DcILP1, DcILP2, and DcIR in the insulin signaling system. The signal will be through upregulating the expression of DcRheb, an upstream gene of the TOR signaling pathway. This in turn influences yolk metabolism, which eventually causes the ovaries of female D. citri to mature and therefore initiate oviposition behavior.
Collapse
Affiliation(s)
- Ziye Wang
- Key Laboratory of Bio‐Pesticide Innovation and Application of Guangdong ProvinceSouth China Agricultural UniversityGuangzhouChina
- Guangdong Laboratory for Lingnan Modern AgricultureGuangzhouChina
| | - Delong Tan
- Institute of Facility AgricultureGuangdong Academy of Agricultural SciencesGuangzhouChina
| | - Feifeng Wang
- Key Laboratory of Bio‐Pesticide Innovation and Application of Guangdong ProvinceSouth China Agricultural UniversityGuangzhouChina
- Guangdong Laboratory for Lingnan Modern AgricultureGuangzhouChina
| | - Shuhao Guo
- Key Laboratory of Bio‐Pesticide Innovation and Application of Guangdong ProvinceSouth China Agricultural UniversityGuangzhouChina
| | - Jinhua Liu
- Natural Medicine Institute of Zhejiang YangShengTang Co.LTDHangzhouChina
| | | | - Baoli Qiu
- Key Laboratory of Bio‐Pesticide Innovation and Application of Guangdong ProvinceSouth China Agricultural UniversityGuangzhouChina
- Guangdong Laboratory for Lingnan Modern AgricultureGuangzhouChina
| | - Wen Sang
- Key Laboratory of Bio‐Pesticide Innovation and Application of Guangdong ProvinceSouth China Agricultural UniversityGuangzhouChina
- Guangdong Laboratory for Lingnan Modern AgricultureGuangzhouChina
| |
Collapse
|
5
|
Cook TW, Wilstermann AM, Mitchell JT, Arnold NE, Rajasekaran S, Bupp CP, Prokop JW. Understanding Insulin in the Age of Precision Medicine and Big Data: Under-Explored Nature of Genomics. Biomolecules 2023; 13:257. [PMID: 36830626 PMCID: PMC9953665 DOI: 10.3390/biom13020257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/06/2022] [Revised: 01/20/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023] Open
Abstract
Insulin is amongst the human genome's most well-studied genes/proteins due to its connection to metabolic health. Within this article, we review literature and data to build a knowledge base of Insulin (INS) genetics that influence transcription, transcript processing, translation, hormone maturation, secretion, receptor binding, and metabolism while highlighting the future needs of insulin research. The INS gene region has 2076 unique variants from population genetics. Several variants are found near the transcriptional start site, enhancers, and following the INS transcripts that might influence the readthrough fusion transcript INS-IGF2. This INS-IGF2 transcript splice site was confirmed within hundreds of pancreatic RNAseq samples, lacks drift based on human genome sequencing, and has possible elevated expression due to viral regulation within the liver. Moreover, a rare, poorly characterized African population-enriched variant of INS-IGF2 results in a loss of the stop codon. INS transcript UTR variants rs689 and rs3842753, associated with type 1 diabetes, are found in many pancreatic RNAseq datasets with an elevation of the 3'UTR alternatively spliced INS transcript. Finally, by combining literature, evolutionary profiling, and structural biology, we map rare missense variants that influence preproinsulin translation, proinsulin processing, dimer/hexamer secretory storage, receptor activation, and C-peptide detection for quasi-insulin blood measurements.
Collapse
Affiliation(s)
- Taylor W. Cook
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA
| | | | - Jackson T. Mitchell
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA
| | - Nicholas E. Arnold
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA
| | - Surender Rajasekaran
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
- Office of Research, Corewell Health, Grand Rapids, MI 49503, USA
| | - Caleb P. Bupp
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
- Division of Medical Genetics, Corewell Health, Grand Rapids, MI 49503, USA
| | - Jeremy W. Prokop
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA
- Office of Research, Corewell Health, Grand Rapids, MI 49503, USA
| |
Collapse
|
6
|
Krentz AJ. Classic endocrine disorders: implications for cardiovascular disease. CARDIOVASCULAR ENDOCRINOLOGY AND METABOLISM 2023:233-270. [DOI: 10.1016/b978-0-323-99991-5.00014-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 01/03/2025]
|
7
|
Park JG, Iwata H, Tue NM, Kunisue T, Kim EY. Effects of 1,3,7-tribromodibenzo-p-dioxin, a natural dioxin on chicken embryos: Comparison with effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 237:113538. [PMID: 35483143 DOI: 10.1016/j.ecoenv.2022.113538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 12/30/2021] [Revised: 04/15/2022] [Accepted: 04/16/2022] [Indexed: 06/14/2023]
Abstract
Several naturally occurring dioxins, including 1,3,7-tribromodibenzo-p-dioxin (1,3,7-TriBDD), synthesized by red algae, have been detected in the marine environment. As 1,3,7-TriBDD is accumulated in mussels and fish, predators, such as marine birds, are exposed to this congener, similar to anthropogenic dioxins (including 2,3,7,8-tetrachlorodibenzo-p-dioxin TCDD). However, little is known about the impact of 1,3,7-TriBDD exposure on the bird health. To understand the effects of 1,3,7-TriBDD on birds, the phenotypic effects and hepatic transcriptome were investigated in chicken (Gallus gallus) embryos treated with 27 μM (2.9 ng/g egg) and 137 μM (14.4 ng/g egg) 1,3,7-TriBDD. The blood glucose levels in the 1,3,7-TriBDD-treated groups were lower than those in the control group. The transcriptome analysis of 6520 sequences in the 27 and 137 μM 1,3,7-TriBDD-treated groups identified 733 and 596 differentially expressed genes (DEGs). Cytochrome P450 1A4 and 1A5 were also identified as DEGs, suggesting that the aryl hydrocarbon receptor is activated by this congener. Pathway and network analyses with DEGs suggested that 1,3,7-TriBDD may induce carcinogenic effects and metabolic alterations. These results were similar to the effects on TCDD-treated embryos. Nevertheless, the overall transcriptome results suggested that compared with TCDD, 1,3,7-TriBDD has a unique impact on insulin- and peroxisome-signaling pathways in chicken embryos. Differences in altered transcriptome profiles between 1,3,7-TriBDD- and TCDD-treated embryos may lead to different phenotypic effects: less severe effects of 1,3,7-TriBDD and more fatal effects of TCDD. Collectively, these findings warrant the further assessment of the hazard and risk of 1,3,7-TriBDD on marine animals, considering increased exposure due to climate change.
Collapse
Affiliation(s)
- Jae Gon Park
- Department of Biology, Kyung Hee University, Hoegi-Dong, Dongdaemun-Gu, Seoul 130-701, Republic of Korea
| | - Hisato Iwata
- Center for Marine Environmental Studies (CMES), Ehime University, Matsuyama 790-8577, Japan
| | - Nguyen Minh Tue
- Center for Marine Environmental Studies (CMES), Ehime University, Matsuyama 790-8577, Japan
| | - Tatsuya Kunisue
- Center for Marine Environmental Studies (CMES), Ehime University, Matsuyama 790-8577, Japan
| | - Eun-Young Kim
- Department of Biology, Kyung Hee University, Hoegi-Dong, Dongdaemun-Gu, Seoul 130-701, Republic of Korea; Center for Marine Environmental Studies (CMES), Ehime University, Matsuyama 790-8577, Japan.
| |
Collapse
|
8
|
Henniger MT, Wells JE, Hales KE, Lindholm-Perry AK, Freetly HC, Kuehn LA, Schneider LG, McLean KJ, Campagna SR, Christopher CJ, Myer PR. Effects of a Moderate or Aggressive Implant Strategy on the Rumen Microbiome and Metabolome in Steers. FRONTIERS IN ANIMAL SCIENCE 2022. [DOI: 10.3389/fanim.2022.889817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/13/2022] Open
Abstract
The effects of growth-promoting implants have been well-defined for their ability to impact growth performance in beef cattle. Production-relevant microbes and microbiomes in the rumen have also been associated with growth traits. However, the role of implants on the rumen microbiome has not been determined. The objective of this study was to determine if different doses of implant hormones cause gain-associated ruminal microbial community changes. To assess this, a completely randomized design was used and 336 fall-born steers 450 to 470 days of age from the germplasm evaluation population at the US Meat Animal Research Center (Clay Center, NE) were divided into two treatment groups: 1) a moderate implant strategy (n = 167) of Revalor-IS (80 mg trenbolone acetate and 16 mg estradiol) followed by Revalor-S (120 mg trenbolone acetate and 24 mg estradiol) or 2) an aggressive implant strategy (n = 169) of Revalor-IS followed by Revalor-200 (200 mg trenbolone acetate and 20 mg estradiol). Steers were fed the same diet (57.0% dry-rolled corn, 30% wet distiller’s grains with solubles, 8.0% alfalfa hay, 4.25% vitamin and mineral supplement, and 0.75% urea, on a DM basis). On d 85 after implants administration, rumen contents were collected via orogastric tubing. Samples were sequenced to target and identify bacteria, archaea, and protozoa. Untargeted metabolomics was performed on rumen content using ultra high performance liquid chromatography high resolution mass spectrometry. Production data between implant strategies was analyzed using a mixed model ANOVA (SASv9.4, Cary, NC) followed by separation of least squares means. Microbial diversity between strategies did not differ for archaea or protozoa (P > 0.05). Average daily gain was different (P = 0.01; 1.72 vs 1.66 ± 0.02 kg, aggressive vs moderate, respectively); however, large microbial community shifts were not associated with implant strategy. Two metabolites, N-acetyllysine and N-acetylornithine, were found in greater abundance in the moderate implant strategy (P ≤ 0.04). Understanding associations between the rumen microbiome and implant strategies may allow improvement of growth efficiency in beef cattle.
Collapse
|
9
|
Tin Tin S, Key TJ, Reeves GK. Alcohol Intake and Endogenous Hormones in Pre- and Postmenopausal Women: Findings from the UK Biobank. Cancer Epidemiol Biomarkers Prev 2021; 30:2294-2301. [PMID: 34607837 PMCID: PMC9398104 DOI: 10.1158/1055-9965.epi-21-0789] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/25/2021] [Revised: 08/19/2021] [Accepted: 09/27/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Alcohol intake may influence breast cancer risk in women through hormonal changes, but the evidence to date is inconclusive. We investigated cross-sectional associations between habitual alcohol intake and serum concentrations of testosterone, sex hormone binding globulin (SHBG), insulin-like growth factor-1 (IGF-1), and estradiol (premenopausal women only) in UK Biobank. METHODS We included 30,557 premenopausal and 134,029 postmenopausal women aged between 40 and 69 years when recruited between 2006 and 2010. At their initial assessment visit, habitual alcohol intake was assessed using a touchscreen questionnaire, and serum hormone concentrations were assayed. Multivariable linear regression analysis was performed. RESULTS Per 10 g/day increment in alcohol intake, testosterone concentration was 3.9% [95% confidence intervals (CI): 3.3%-4.5%] higher in premenopausal women and 2.3% (1.8%-2.7%) higher in postmenopausal women (P heterogeneity < 0.0001); SHBG concentration was 0.7% (0.2%-1.1%) higher in premenopausal women and 2.4% (2.2%-2.6%) lower in postmenopausal women (P heterogeneity < 0.0001); and IGF-1 concentration was 1.9% (1.7%-2.1%) lower in premenopausal women and 0.8% (0.6%-0.9%) lower in postmenopausal women (P heterogeneity < 0.0001). In premenopausal women, there was no significant overall association of alcohol with estradiol but a positive association was observed in the early and mid-luteal phases: 1.9% (95% CI: 0.2%-3.6%) and 2.4% (95% CI: 0.7%-4.2%) higher, respectively. CONCLUSIONS This study confirms significant but modest associations between alcohol intake and hormones, with evidence of heterogeneity by menopausal status. IMPACT The findings facilitate better understanding of whether alcohol intake influences hormone concentrations, but further work is necessary to fully understand the mechanisms linking alcohol with cancer risk.
Collapse
Affiliation(s)
- Sandar Tin Tin
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK.
| | - Timothy J Key
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Gillian K Reeves
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| |
Collapse
|
10
|
Janssen JAMJL. Hyperinsulinemia and Its Pivotal Role in Aging, Obesity, Type 2 Diabetes, Cardiovascular Disease and Cancer. Int J Mol Sci 2021; 22:ijms22157797. [PMID: 34360563 PMCID: PMC8345990 DOI: 10.3390/ijms22157797] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/16/2021] [Revised: 07/11/2021] [Accepted: 07/13/2021] [Indexed: 01/10/2023] Open
Abstract
For many years, the dogma has been that insulin resistance precedes the development of hyperinsulinemia. However, recent data suggest a reverse order and place hyperinsulinemia mechanistically upstream of insulin resistance. Genetic background, consumption of the “modern” Western diet and over-nutrition may increase insulin secretion, decrease insulin pulses and/or reduce hepatic insulin clearance, thereby causing hyperinsulinemia. Hyperinsulinemia disturbs the balance of the insulin–GH–IGF axis and shifts the insulin : GH ratio towards insulin and away from GH. This insulin–GH shift promotes energy storage and lipid synthesis and hinders lipid breakdown, resulting in obesity due to higher fat accumulation and lower energy expenditure. Hyperinsulinemia is an important etiological factor in the development of metabolic syndrome, type 2 diabetes, cardiovascular disease, cancer and premature mortality. It has been further hypothesized that nutritionally driven insulin exposure controls the rate of mammalian aging. Interventions that normalize/reduce plasma insulin concentrations might play a key role in the prevention and treatment of age-related decline, obesity, type 2 diabetes, cardiovascular disease and cancer. Caloric restriction, increasing hepatic insulin clearance and maximizing insulin sensitivity are at present the three main strategies available for managing hyperinsulinemia. This may slow down age-related physiological decline and prevent age-related diseases. Drugs that reduce insulin (hyper) secretion, normalize pulsatile insulin secretion and/or increase hepatic insulin clearance may also have the potential to prevent or delay the progression of hyperinsulinemia-mediated diseases. Future research should focus on new strategies to minimize hyperinsulinemia at an early stage, aiming at successfully preventing and treating hyperinsulinemia-mediated diseases.
Collapse
Affiliation(s)
- Joseph A M J L Janssen
- Department of internal Medicine, Division of Endocrinology, Erasmus Medical Center, 40, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
11
|
Liao S, Post S, Lehmann P, Veenstra JA, Tatar M, Nässel DR. Regulatory Roles of Drosophila Insulin-Like Peptide 1 (DILP1) in Metabolism Differ in Pupal and Adult Stages. Front Endocrinol (Lausanne) 2020; 11:180. [PMID: 32373064 PMCID: PMC7186318 DOI: 10.3389/fendo.2020.00180] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 01/27/2020] [Accepted: 03/13/2020] [Indexed: 01/12/2023] Open
Abstract
The insulin/IGF-signaling pathway is central in control of nutrient-dependent growth during development, and in adult physiology and longevity. Eight insulin-like peptides (DILP1-8) have been identified in Drosophila, and several of these are known to regulate growth, metabolism, reproduction, stress responses, and lifespan. However, the functional role of DILP1 is far from understood. Previous work has shown that dilp1/DILP1 is transiently expressed mainly during the pupal stage and the first days of adult life. Here, we study the role of dilp1 in the pupa, as well as in the first week of adult life, and make some comparisons to dilp6 that displays a similar pupal expression profile, but is expressed in fat body rather than brain neurosecretory cells. We show that mutation of dilp1 diminishes organismal weight during pupal development, whereas overexpression increases it, similar to dilp6 manipulations. No growth effects of dilp1 or dilp6 manipulations were detected during larval development. We next show that dilp1 and dilp6 increase metabolic rate in the late pupa and promote lipids as the primary source of catabolic energy. Effects of dilp1 manipulations can also be seen in the adult fly. In newly eclosed female flies, survival during starvation is strongly diminished in dilp1 mutants, but not in dilp2 and dilp1/dilp2 mutants, whereas in older flies, only the double mutants display reduced starvation resistance. Starvation resistance is not affected in male dilp1 mutant flies, suggesting a sex dimorphism in dilp1 function. Overexpression of dilp1 also decreases survival during starvation in female flies and increases egg laying and decreases egg to pupal viability. In conclusion, dilp1 and dilp6 overexpression promotes metabolism and growth of adult tissues during the pupal stage, likely by utilization of stored lipids. Some of the effects of the dilp1 manipulations may carry over from the pupa to affect physiology in young adults, but our data also suggest that dilp1 signaling is important in metabolism and stress resistance in the adult stage.
Collapse
Affiliation(s)
- Sifang Liao
- Department of Zoology, Stockholm University, Stockholm, Sweden
| | - Stephanie Post
- Department of Ecology and Evolutionary Biology, Brown University, Providence, RI, United States
| | - Philipp Lehmann
- Department of Zoology, Stockholm University, Stockholm, Sweden
| | - Jan A. Veenstra
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (CNRS UMR5287), University of Bordeaux, Pessac, France
| | - Marc Tatar
- Department of Ecology and Evolutionary Biology, Brown University, Providence, RI, United States
| | - Dick R. Nässel
- Department of Zoology, Stockholm University, Stockholm, Sweden
- *Correspondence: Dick R. Nässel
| |
Collapse
|
12
|
Zhang G, Li J, Zhang J, Liang X, Zhang X, Wang T, Yin S. Integrated Analysis of Transcriptomic, miRNA and Proteomic Changes of a Novel Hybrid Yellow Catfish Uncovers Key Roles for miRNAs in Heterosis. Mol Cell Proteomics 2019; 18:1437-1453. [PMID: 31092672 PMCID: PMC6601203 DOI: 10.1074/mcp.ra118.001297] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/26/2018] [Revised: 04/09/2019] [Indexed: 01/14/2023] Open
Abstract
Heterosis is a complex biological phenomenon in which hybridization produces offspring that exhibit superior phenotypic characteristics compared with the parents. Heterosis is widely utilized in agriculture, for example in fish farming; however, its underlying molecular basis remains elusive. To gain a comprehensive and unbiased molecular understanding of fish heterosis, we analyzed the mRNA, miRNA, and proteomes of the livers of three catfish species, Pelteobagrus fulvidraco, P. vachelli, and their hybrid, the hybrid yellow catfish "Huangyou-1" (P. fulvidraco ♀ × P. vachelli ♂). Using next-generation sequencing and mass spectrometry, we show that the nonadditive, homoeolog expression bias and expression level dominance pattern were readily identified at the transcriptional, post-transcriptional, or protein levels, providing the evidence for the widespread presence of dominant models during hybridization. A number of predicted miRNA-mRNA-protein pairs were found and validated by qRT-PCR and PRM assays. Furthermore, several diverse key pathways were identified, including immune defense, metabolism, digestion and absorption, and cell proliferation and development, suggesting the vital mechanisms involved in the generation of the heterosis phenotype in progenies. We propose that the high parental expression of genes/proteins (growth, nutrition, feeding, and disease resistance) coupled with low parental miRNAs of the offspring, are inherited from the mother or father, thus indicating that the offspring were enriched with the advantages of the father or mother. We provide new and important information about the molecular mechanisms of heterosis, which represents a significant step toward a more complete elucidation of this phenomenon.
Collapse
Affiliation(s)
- Guosong Zhang
- From the ‡College of Marine Science and Engineering, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China;; §Key Laboratory for Physiology Biochemistry and Application, School of Agriculture and Bioengineering, Heze University, Heze, Shandong 274015, China
| | - Jie Li
- From the ‡College of Marine Science and Engineering, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China;; ¶Co-Innovation Center for Marine Bio-Industry Technology, Lian Yungang, 222005, China
| | - Jiajia Zhang
- From the ‡College of Marine Science and Engineering, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China;; ¶Co-Innovation Center for Marine Bio-Industry Technology, Lian Yungang, 222005, China
| | - Xia Liang
- §Key Laboratory for Physiology Biochemistry and Application, School of Agriculture and Bioengineering, Heze University, Heze, Shandong 274015, China
| | - Xinyu Zhang
- From the ‡College of Marine Science and Engineering, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China;; ¶Co-Innovation Center for Marine Bio-Industry Technology, Lian Yungang, 222005, China
| | - Tao Wang
- From the ‡College of Marine Science and Engineering, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China;; ¶Co-Innovation Center for Marine Bio-Industry Technology, Lian Yungang, 222005, China
| | - Shaowu Yin
- From the ‡College of Marine Science and Engineering, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China;; ¶Co-Innovation Center for Marine Bio-Industry Technology, Lian Yungang, 222005, China.
| |
Collapse
|
13
|
Jannin A, Espiard S, Benomar K, Do Cao C, Mycinski B, Porte H, D’Herbomez M, Penel N, Vantyghem MC. Non-islet-cell tumour hypoglycaemia (NICTH): About a series of 6 cases. ANNALES D'ENDOCRINOLOGIE 2019; 80:21-25. [DOI: 10.1016/j.ando.2018.01.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 08/24/2017] [Revised: 11/21/2017] [Accepted: 01/04/2018] [Indexed: 12/30/2022]
|
14
|
Garla V, Sonani H, Palabindala V, Gomez-Sanchez C, Subauste J, Lien LF. Non-islet Cell Hypoglycemia: Case Series and Review of the Literature. Front Endocrinol (Lausanne) 2019; 10:316. [PMID: 31156561 PMCID: PMC6529841 DOI: 10.3389/fendo.2019.00316] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 01/08/2019] [Accepted: 05/01/2019] [Indexed: 12/30/2022] Open
Abstract
Non-islet cell hypoglycemia (NICH) is hypoglycemia due to the overproduction of insulin-like growth factor-2 (IGF-2) and its precursors which can activate the insulin receptor. Typically, large mesenchymal and epithelial tumors can cause NICH. Diagnosis is confirmed by finding an elevated IGF-2/IGF-1 ratio. The mainstay of treatment is surgical excision. Glucocorticoids may be used in cases where surgery is not possible. We present two cases of NICH with different outcomes. A 33-year-old male patient admitted with altered mental. He was found walking naked outside his house. Laboratory assessment revealed severe hypoglycemia. Further evaluation showed low levels of insulin, C-peptide, and beta-hydroxybutyrate along with an elevated IGF-2/IGF-1 ratio confirming the diagnosis of NICH. Computed tomography (CT) of the abdomen showed a massive tumor of the liver consistent with hepatocellular carcinoma. Since the patient refused surgery, he was started on prednisone however the hypoglycemia persisted. A 54-year-old female patient with a history of type 2 diabetes mellitus (DM) admitted with recent onset hypoglycemia. Despite stopping her insulin, she continued to have hypoglycemia necessitating the administration of high concentrations of intravenous dextrose. Further evaluation showed low levels of insulin, C-peptide, and beta-hydroxybutyrate along with an elevated IGF-2/IGF-1 ratio consistent with the diagnosis of NICH. CT abdomen showed a 24 cm tumor near the uterus. The pathology was consistent with a gastrointestinal stromal tumor (GIST). After surgical excision of the tumor, the hypoglycemia resolved.
Collapse
Affiliation(s)
- Vishnu Garla
- Department of Internal Medicine, University of Mississippi Medical Center, Jackson, MS, United States
- *Correspondence: Vishnu Garla
| | - Hardik Sonani
- Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Venkatraman Palabindala
- Department of Internal Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| | - Celso Gomez-Sanchez
- Department of Internal Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| | - Jose Subauste
- Department of Internal Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| | - Lillian Francis Lien
- Department of Internal Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| |
Collapse
|
15
|
Censi S, Mian C, Betterle C. Insulin autoimmune syndrome: from diagnosis to clinical management. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:335. [PMID: 30306074 DOI: 10.21037/atm.2018.07.32] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 12/21/2022]
Abstract
Autoimmune forms of hypoglycemia are a rare cause of low blood sugar levels among Caucasians, and often go misdiagnosed, exposing patients to lengthy series of pointless, potentially harmful and expensive tests. There are two types of autoimmune hypoglycemia. One is insulin autoimmune syndrome (IAS), which is characterized by hyperinsulinemic hypoglycemia, elevated insulin autoantibody (IAA) titers, no prior exposure to exogenous insulin, and no of pathological abnormalities of the pancreatic islets. This condition is also known as "Hirata's disease". The other is type B insulin resistance syndrome (TBIRS), a rare autoimmune disorder resulting in a broad array of abnormalities in glucose homeostasis-from hypoglycemia to extremely insulin-resistant hyperglycemia-caused by the presence of insulin receptor autoantibodies (IRAbs). This review focuses on these two syndromes, describing their epidemiology, possible genetic background, clinical presentation, pathophysiology, diagnosis and treatment.
Collapse
Affiliation(s)
- Simona Censi
- Endocrinology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Caterina Mian
- Endocrinology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Corrado Betterle
- Endocrinology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| |
Collapse
|
16
|
Ma X, Yang C, Zhang J, Wang J, Li W, Xu C, Rong P, Ye B, Wu M, Jiang J, Yi S, Wang W. Culturing with modified EGM2 medium enhances porcine neonatal islet-like cell clusters resistance to apoptosis in islet xenotransplantation. Xenotransplantation 2017; 25. [PMID: 29131417 DOI: 10.1111/xen.12358] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/06/2017] [Revised: 09/12/2017] [Accepted: 09/12/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND Neonatal pig islet-like cell clusters (NICC) are an attractive source of insulin-producing tissue for potential transplantation treatment of type 1 diabetic patients. However, a considerable loss of NICC after their transplantation due to apoptosis resulted from islet isolation and instant blood-mediated inflammatory reaction remains to be overcome. METHODS EGM2 medium depleted with hydrocortisone and supplemented with 50 mmol/L isobutylmethylxanthine, 10 mmol/L nicotinamide, and 10 mmol/L glucose was used to culture NICC at day 1, the day after isolation and changed every other day. NICC cultured with EGM2 or control Ham's F-10 medium were collected at day 7 of culture for the following assays. The viability of NICC was evaluated by AO/EB staining and FACS. Static assay and oxygen consumption rate analysis were performed to assess the function of NICC. Insulin and glucagon gene expression were measured by real-time PCR. Tubing loops model and TUNEL assay were performed to confirm the apoptosis-resistant ability of NICC cultured with modified EGM2 medium. Serum starvation and hypoxia treatment were used to test the tolerant capability of NICC in the microenvironment of hypoxia/nutrient deficiency in vitro. The molecules involved in apoptosis pathways in NICC were analyzed by Western blotting. RESULTS Compared with Ham's F-10 medium, culturing NICC with EGM2 medium led to increased number and viability of NICC with higher stimulation index, upregulated gene expression of both insulin and glucagon, and enhanced mitochondria function. Furthermore, fewer modified EGM2 medium cultured NICC were found under apoptosis when evaluated in an in vitro tubing loop model of IBMIR. Moreover, EGM2 medium cultured NICC demonstrated much less apoptotic cells under either serum starvation or hypoxia condition than their Ham's F-10 medium cultured counterparts. The enhanced capability of EGM2 cultured NICC to resist apoptosis was associated with their elevated protein levels of anti-apoptotic Bcl-2 family member Mcl-1. CONCLUSION Culturing NICC with EGM2 provides a simple and effective approach not only to increase NICC yield, viability, and maturation but also to enhance their resistance to apoptosis to preserve the initial graft mass for successful islet xenotransplantation.
Collapse
Affiliation(s)
- Xiaoqian Ma
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.,Engineering and Technology Research Center for Xenotransplantation of Human Province, Changsha, Hunan, China
| | - Cejun Yang
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.,Engineering and Technology Research Center for Xenotransplantation of Human Province, Changsha, Hunan, China
| | - Juan Zhang
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.,Engineering and Technology Research Center for Xenotransplantation of Human Province, Changsha, Hunan, China
| | - Jia Wang
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.,Engineering and Technology Research Center for Xenotransplantation of Human Province, Changsha, Hunan, China
| | - Wei Li
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.,Engineering and Technology Research Center for Xenotransplantation of Human Province, Changsha, Hunan, China
| | - Chang Xu
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.,Engineering and Technology Research Center for Xenotransplantation of Human Province, Changsha, Hunan, China
| | - Pengfei Rong
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.,Engineering and Technology Research Center for Xenotransplantation of Human Province, Changsha, Hunan, China
| | - Bin Ye
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.,Engineering and Technology Research Center for Xenotransplantation of Human Province, Changsha, Hunan, China
| | - Minghua Wu
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jianhui Jiang
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, China
| | - Shounan Yi
- Center for Transplant and Renal Research, Westmead Institute for Medical Research, University of Sydney, Westmead, NSW, Australia
| | - Wei Wang
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.,Engineering and Technology Research Center for Xenotransplantation of Human Province, Changsha, Hunan, China
| |
Collapse
|
17
|
Fujinaga D, Kohmura Y, Okamoto N, Kataoka H, Mizoguchi A. Insulin-like growth factor (IGF)-like peptide and 20-hydroxyecdysone regulate the growth and development of the male genital disk through different mechanisms in the silkmoth, Bombyx mori. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2017; 87:35-44. [PMID: 28610907 DOI: 10.1016/j.ibmb.2017.06.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 02/28/2017] [Revised: 06/07/2017] [Accepted: 06/08/2017] [Indexed: 06/07/2023]
Abstract
It is well established that ecdysteroids play pivotal roles in the regulation of insect molting and metamorphosis. However, the mechanisms by which ecdysteroids regulate the growth and development of adult organs after pupation are poorly understood. Recently, we have identified insulin-like growth factor (IGF)-like peptides (IGFLPs), which are secreted after pupation under the control of 20-hydroxyecdysone (20E). In the silkmoth, Bombyx mori, massive amounts of Bombyx-IGFLP (BIGFLP) are present in the hemolymph during pupal-adult development, suggesting its importance in the regulation of adult tissue growth. Thus, we hypothesized that the growth and development of adult tissues including imaginal disks are regulated by the combined effects of BIGFLP and 20E. In this study, we investigated the growth-promoting effects of BIGFLP and 20E using the male genital disks of B. mori cultured ex vivo, and further analyzed the cell signaling pathways mediating hormone actions. We demonstrate that 20E induces the elongation of genital disks, that both hormones stimulate protein synthesis in an additive manner, and that BIGFLP and 20E exert their effects through the insulin/IGF signaling pathway and mitogen-activated protein kinase pathway, respectively. These results show that the growth and development of the genital disk are coordinately regulated by both BIGFLP and 20E.
Collapse
Affiliation(s)
- Daiki Fujinaga
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba 277-8562, Japan
| | - Yusuke Kohmura
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan
| | - Naoki Okamoto
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan
| | - Hiroshi Kataoka
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba 277-8562, Japan.
| | - Akira Mizoguchi
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan.
| |
Collapse
|
18
|
Nässel DR, Vanden Broeck J. Insulin/IGF signaling in Drosophila and other insects: factors that regulate production, release and post-release action of the insulin-like peptides. Cell Mol Life Sci 2016; 73:271-90. [PMID: 26472340 PMCID: PMC11108470 DOI: 10.1007/s00018-015-2063-3] [Citation(s) in RCA: 214] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/02/2015] [Revised: 10/01/2015] [Accepted: 10/05/2015] [Indexed: 01/02/2023]
Abstract
Insulin, insulin-like growth factors (IGFs) and insulin-like peptides (ILPs) are important regulators of metabolism, growth, reproduction and lifespan, and mechanisms of insulin/IGF signaling (IIS) have been well conserved over evolution. In insects, between one and 38 ILPs have been identified in each species. Relatively few insect species have been investigated in depth with respect to ILP functions, and therefore we focus mainly on the well-studied fruitfly Drosophila melanogaster. In Drosophila eight ILPs (DILP1-8), but only two receptors (dInR and Lgr3) are known. DILP2, 3 and 5 are produced by a set of neurosecretory cells (IPCs) in the brain and their biosynthesis and release are controlled by a number of mechanisms differing between larvae and adults. Adult IPCs display cell-autonomous sensing of circulating glucose, coupled to evolutionarily conserved mechanisms for DILP release. The glucose-mediated DILP secretion is modulated by neurotransmitters and neuropeptides, as well as by factors released from the intestine and adipocytes. Larval IPCs, however, are indirectly regulated by glucose-sensing endocrine cells producing adipokinetic hormone, or by circulating factors from the intestine and fat body. Furthermore, IIS is situated within a complex physiological regulatory network that also encompasses the lipophilic hormones, 20-hydroxyecdysone and juvenile hormone. After release from IPCs, the ILP action can be modulated by circulating proteins that act either as protective carriers (binding proteins), or competitive inhibitors. Some of these proteins appear to have additional functions that are independent of ILPs. Taken together, the signaling with multiple ILPs is under complex control, ensuring tightly regulated IIS in the organism.
Collapse
Affiliation(s)
- Dick R Nässel
- Department of Zoology, Stockholm University, 10691, Stockholm, Sweden.
| | - Jozef Vanden Broeck
- Molecular Developmental Physiology and Signal Transduction, Department of Animal Physiology and Neurobiology, Zoological Institute, K.U. Leuven, Louvain, Belgium
| |
Collapse
|
19
|
Function of Membrane-Associated Proteoglycans in the Regulation of Satellite Cell Growth. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 900:61-95. [DOI: 10.1007/978-3-319-27511-6_4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 12/31/2022]
|
20
|
Smith TJ, Janssen JAMJL. Building the Case for Insulin-Like Growth Factor Receptor-I Involvement in Thyroid-Associated Ophthalmopathy. Front Endocrinol (Lausanne) 2016; 7:167. [PMID: 28096798 PMCID: PMC5206614 DOI: 10.3389/fendo.2016.00167] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 11/15/2016] [Accepted: 12/13/2016] [Indexed: 11/18/2022] Open
Abstract
The pathogenesis of orbital Graves' disease (GD), a process known as thyroid-associated ophthalmopathy (TAO), remains incompletely understood. The thyrotropin receptor (TSHR) represents the central autoantigen involved in GD and has been proposed as the thyroid antigen shared with the orbit that could explain the infiltration of immune cells into tissues surrounding the eye. Another cell surface protein, insulin-like growth factor-I receptor (IGF-IR), has recently been proposed as a second antigen that participates in TAO by virtue of its interactions with anti-IGF-IR antibodies generated in GD, its apparent physical and functional complex formation with TSHR, and its necessary involvement in TSHR post-receptor signaling. The proposal that IGF-IR is involved in TAO has provoked substantial debate. Furthermore, several studies from different laboratory groups, each using different experimental models, have yielded conflicting results. In this article, we attempt to summarize the biological characteristics of IGF-IR and TSHR. We also review the evidence supporting and refuting the postulate that IGF-IR is a self-antigen in GD and that it plays a potentially important role in TAO. The putative involvement of IGF-IR in disease pathogenesis carries substantial clinical implications. Specifically, blocking this receptor with monoclonal antibodies can dramatically attenuate the induction by TSH and pathogenic antibodies generated in GD of proinflammatory genes in cultured orbital fibroblasts and fibrocytes. These cell types appear critical to the development of TAO. These observations have led to the conduct of a now-completed multicenter therapeutic trial of a fully human monoclonal anti-IGF-IR blocking antibody in moderate to severe, active TAO.
Collapse
Affiliation(s)
- Terry J. Smith
- Department of Ophthalmology and Visual Sciences, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan Medical School, Ann Arbor, MI, USA
- *Correspondence: Terry J. Smith,
| | - Joseph A. M. J. L. Janssen
- Department of Internal Medicine, Erasmus Medical Center, Division of Endocrinology, Rotterdam, Netherlands
| |
Collapse
|
21
|
Tay CKJ, Teoh HL, Su S. A common problem in the elderly with an uncommon cause: hypoglycaemia secondary to the Doege-Potter syndrome. BMJ Case Rep 2015; 2015:bcr-2014-207995. [PMID: 25636632 DOI: 10.1136/bcr-2014-207995] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/04/2022] Open
Abstract
Falls due to hypoglycaemia in the elderly is usually a complication of diabetic treatment. In the absence of diabetes, hypoglycaemia may be due to insulin or insulin-like producing tumours. The Doege-Potter syndrome is a rare paraneoplastic syndrome, characterised by non-islet cell tumour hypoglycaemia (NICTH) secondary to a solitary fibrous tumour that secretes insulin-like growth factor (IGF) 2. Definitive treatment of hypoglycaemia due to NICTH is by tumour resection. Our patient was a 78-year-old woman admitted after a fall with a facial injury and a history of significant weight loss. Her blood sugar was persistently low despite intravenous dextrose infusion. CT of the thorax revealed a large heterogeneous mass measuring 11.6×16.3×15.6 cm in the right hemithorax. A biopsy of the mass was reported as a solitary fibrous tumour. Biochemical investigations revealed low insulin, C-peptide, IGF-1 and a high IGF-2:IGF-1 ratio, consistent with NICTH. The patient underwent tumour resection and the hypoglycaemia normalised completely immediately after surgery.
Collapse
Affiliation(s)
| | - Hui Lin Teoh
- Department of Geriatric Medicine, Khoo Teck Puat Hospital, Singapore
| | - Su Su
- Department of Geriatric Medicine, Khoo Teck Puat Hospital, Singapore
| |
Collapse
|
22
|
Abstract
In view of the lifelong exposure and large patient populations involved, insulin analogs with an increased mitogenic effect in comparison to human insulin may potentially constitute a major health problem, since these analogs may possibly induce the growth of pre-existing neoplasms. At present, the available data suggest that insulin analogs are safe. In line with these findings, we observed that serum of diabetic patients treated with insulin analogs, compared to that of diabetic patients treated with human insulin, did not induce an increased phosphorylation of tyrosine residues of the insulin-like growth factor-I receptor (IGF-IR). However, the classical model of the IGF-IR signaling may be insufficient to explain (all) mitogenic effects of insulin analogs since also non-canonical signaling pathways of the IGF-IR may play a major role in this respect. Although phosphorylation of tyrosine residues of the IGF-IR is generally considered to be the initial activation step within the intracellular IGF-IR signaling pathway, it has been found that cells undergo a signaling switch under hyperglycemic conditions. After this switch, a completely different mechanism is utilized to activate the mitogenic (mitogen-activated protein kinase) pathways of the IGF-IR that is independent from tyrosine phosphorylation of the IGF-IR. At present it is unknown whether activation of this alternative intracellular pathway of the IGF-IR occurs during hyperglycemia in vivo and whether it is stronger in patients treated with (some) insulin analogs than in patients treated with human insulin. In addition, it is unknown whether the insulin receptors (IRs) also undergo a signaling switch during hyperglycemia. This should be investigated in future studies. Finally, relative overexpression of IR isoform A (IR-A) in (pre) cancer tissues may play a key role in the development and progression of human cancers during treatment with insulin (analogs). Further studies are required to unravel whether the IR-A is involved in the development of cancers and whether, in this respect (some) insulin analogs differ from human insulin.
Collapse
Affiliation(s)
- Joseph A. M. J. L. Janssen
- Division of Endocrinology, Department of Internal Medicine, Erasmus MC, Rotterdam, Netherlands
- *Correspondence: Joseph A. M. J. L. Janssen, Division of Endocrinology, Department of Internal Medicine, Erasmus MC, Room D-443, ‘s-Gravendijkwal 230, Rotterdam 3015 CE, Netherlands e-mail:
| | - Aimee J. Varewijck
- Division of Endocrinology, Department of Internal Medicine, Erasmus MC, Rotterdam, Netherlands
| |
Collapse
|
23
|
Licht CMM, van Turenhout LC, Deijen JB, Koppes LLJ, van Mechelen W, Twisk JWR, Drent ML. The Association between IGF-1 Polymorphisms, IGF-1 Serum Levels, and Cognitive Functions in Healthy Adults: The Amsterdam Growth and Health Longitudinal Study. Int J Endocrinol 2014; 2014:181327. [PMID: 25114679 PMCID: PMC4120488 DOI: 10.1155/2014/181327] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 04/06/2014] [Revised: 06/19/2014] [Accepted: 06/20/2014] [Indexed: 01/24/2023] Open
Abstract
Several studies have demonstrated an association between polymorphisms in the insulin-like growth factor-1 (IGF-1) gene and IGF-1 serum levels. IGF-1 levels have been associated with cognitive functioning in older persons and growth hormone deficient patients. The present study investigates whether IGF-1 polymorphisms, IGF-1 levels, and cognition are interconnected in healthy adults. Data of 277 participants (mean age: 42.4 years) of the Amsterdam Growth and Health Longitudinal Study on IGF-1 promoter polymorphisms, IGF-1 serum level, spatial working memory (SWM), paired associate learning (PAL), and IQ tests were analyzed. (M)ANOVAs were applied to confirm the associations between IGF-1 polymorphisms and IGF-1 levels and between IGF-1 levels and cognition. Three groups were distinguished based on specific IGF-1 polymorphism alleles: a homozygote 192 bp/192 bp genotype, a heterozygote 192 bp/x genotype, and a noncarrier x/x genotype. Although different IGF-1 levels were found for the three genotypes, performance on all cognitive tasks and IQ measures was similar. Despite the associations between IGF-1 polymorphisms and IGF-1 levels, no association was found between cognition and IGF-1 levels. It seems that IGF-1 does not play a role in the cognitive performance of healthy middle-aged adults. Possible, IGF-1 fulfills a more developmental and protective role in cognition which becomes apparent during childhood, old-age, or disease.
Collapse
Affiliation(s)
- Carmilla M. M. Licht
- Department of Clinical Neuropsychology, VU University, Van der Boechorststraat 1, 1081 BT Amsterdam, The Netherlands
- Neuroscience Campus Amsterdam, VU University, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
- Department of Epidemiology and Biostatistics, The EMGO Institute for Health and Care Research, VU University Medical Center, Van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| | - Lise C. van Turenhout
- Department of Internal Medicine, Endocrine Section, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Jan Berend Deijen
- Department of Clinical Neuropsychology, VU University, Van der Boechorststraat 1, 1081 BT Amsterdam, The Netherlands
- Neuroscience Campus Amsterdam, VU University, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
- *Jan Berend Deijen:
| | - Lando L. J. Koppes
- Work and Employment Division, Netherlands Organization for Applied Scientific Research (TNO), Polarisavenue 151, 2132 JJ Hoofddorp, The Netherlands
| | - Willem van Mechelen
- Department of Public and Occupational Health, EMGO Institute for Health and Care Research, VU University Medical Center, Van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
- Body@Work, Research Center on Physical Activity, Work and Health, TNO-VU University Medical Center, Van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| | - Jos W. R. Twisk
- Department of Epidemiology and Biostatistics, The EMGO Institute for Health and Care Research, VU University Medical Center, Van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
- Department of Health Sciences, Faculty of Earth and Life Sciences, VU University, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - Madeleine L. Drent
- Department of Clinical Neuropsychology, VU University, Van der Boechorststraat 1, 1081 BT Amsterdam, The Netherlands
- Neuroscience Campus Amsterdam, VU University, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
- Department of Internal Medicine, Endocrine Section, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
24
|
Dynkevich Y, Rother KI, Whitford I, Qureshi S, Galiveeti S, Szulc AL, Danoff A, Breen TL, Kaviani N, Shanik MH, Leroith D, Vigneri R, Koch CA, Roth J. Tumors, IGF-2, and hypoglycemia: insights from the clinic, the laboratory, and the historical archive. Endocr Rev 2013; 34:798-826. [PMID: 23671155 DOI: 10.1210/er.2012-1033] [Citation(s) in RCA: 149] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 12/31/2022]
Abstract
Tumors of mesenchymal and epithelial origin produce IGF-2, which activates pathways in the tumors. In a minority of patients, the tumors (hepatomas, fibromas, and fibrosarcomas are the most common among many) release into the circulation enough IGF-2-related peptides to mimic the fasting hypoglycemia characteristic of patients with insulin-producing islet-cell tumors. Rarely, markedly elevated IGF-2 levels produce somatic changes suggestive of acromegaly. Typically, the elevated IGF-2 levels are associated with suppressed plasma levels of insulin, IGF-1, and GH. Complicating the pathophysiology are the IGF binding proteins (IGFBPs) that can bind IGF-2 and IGF-1, modifying hormone metabolism and action. IGFBP concentrations are often altered in the presence of these tumors. At the cellular level, the 3 hormone-related ligands, IGF-2, IGF-1, and insulin, all bind to 4 (or more) types of IGF-1 receptor (IGF-1R) and insulin receptor (IR). Each receptor has its own characteristic affinity for each ligand, a tyrosine kinase, and overlapping profiles of action in the target cells. The IGF-2R, in addition to binding mannose-6-phosphate-containing proteins, provides an IGF-2 degradation pathway. Recent evidence suggests IGF-2R involvement also in signal transduction. Surgery, the treatment of choice, can produce a cure. For patients not cured by surgery, multiple therapies exist, for the tumor and for hypoglycemia. Potential future therapeutic approaches are sketched. From 1910 to 1930, hypoglycemia, insulin, insulinomas, and non-islet-cell tumors were recognized. The latter third of the century witnessed the emergence of the immunoassay for insulin; the IGFs, their binding proteins, and assays to measure them; and receptors for the insulin-related peptides as well as the intracellular pathways beyond the receptor. In closing, we replace non-islet-cell tumor hypoglycemia, an outdated and misleading label, with IGF-2-oma, self-explanatory and consistent with names of other hormone-secreting tumors.
Collapse
Affiliation(s)
- Yevgeniya Dynkevich
- MD, FACP, Investigator, Feinstein Institute for Medical Research, Laboratory of Diabetes and Diabetes-Related Research, 350 Community Drive, Manhasset, NY 11030.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Narayanan RP, Gittins M, Siddals KW, Oliver RL, Hudson JE, White A, Durrington P, Davies RR, Rutter MK, Gibson JM. Atorvastatin administration is associated with dose-related changes in IGF bioavailability. Eur J Endocrinol 2013; 168:543-8. [PMID: 23333902 DOI: 10.1530/eje-12-0844] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 01/10/2023]
Abstract
OBJECTIVE IGF levels, their binding proteins (IGFBPs) and high-dose statin therapy have been linked to the development of diabetes. We aimed to identify whether atorvastatin caused dose-related changes in IGF proteins. DESIGN AND METHODS We measured IGF1, IGF2, IGFBP1 and IGFBP3 concentrations at baseline, 6 and 12 months in Protection Against Nephropathy in Diabetes with Atorvastatin trial participants with type 2 diabetes randomised to 10 mg (n=59) vs 80 mg (n=60) of atorvastatin (n=119; mean (S.D.): age 64 (10) years; 83% male; HbA1c 61 (10) mmol/mol; blood pressure 131/73 mmHg). RESULTS Atorvastatin was associated with overall reductions in circulating IGF1, IGF2 and IGFBP3 concentrations (P<0.05 for all changes). The adjusted mean (95% CI) between-group differences that indicate dose-related changes in IGF proteins were not significant for IGF1: -3 (-21 to 14) ng/ml; IGF2: -23 (-65 to 18) ng/ml and IGFBP3: -0.34 (-0.71 to 0.03) μg/ml, negative values indicating numerically greater lowering with high dose. The IGFBP1 concentration did not change with atorvastatin therapy overall but the adjusted mean (95% CI) between-group difference indicating a dose-related change in log IGFBP1 was highly significant -0.41 (-0.69 to 0.13, P=0.004). CONCLUSION IGF1, IGF2 and IGFBP3 concentrations decreased following atorvastatin therapy. A differential effect of low- vs high-dose atorvastatin on IGFBP1 concentrations was observed with likely implications for IGF bioavailability. The dose-related differential impact of atorvastatin treatment on concentration of IGF proteins merits investigation as a mechanism to explain the worsening of glucose tolerance with statin therapy.
Collapse
Affiliation(s)
- Ram P Narayanan
- Salford Royal Hospital NHS Foundation Trust, The University of Manchester, Salford, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Diamanti-Kandarakis E, Dunaif A. Insulin resistance and the polycystic ovary syndrome revisited: an update on mechanisms and implications. Endocr Rev 2012; 33:981-1030. [PMID: 23065822 PMCID: PMC5393155 DOI: 10.1210/er.2011-1034] [Citation(s) in RCA: 1103] [Impact Index Per Article: 84.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 02/06/2023]
Abstract
Polycystic ovary syndrome (PCOS) is now recognized as an important metabolic as well as reproductive disorder conferring substantially increased risk for type 2 diabetes. Affected women have marked insulin resistance, independent of obesity. This article summarizes the state of the science since we last reviewed the field in the Endocrine Reviews in 1997. There is general agreement that obese women with PCOS are insulin resistant, but some groups of lean affected women may have normal insulin sensitivity. There is a post-binding defect in receptor signaling likely due to increased receptor and insulin receptor substrate-1 serine phosphorylation that selectively affects metabolic but not mitogenic pathways in classic insulin target tissues and in the ovary. Constitutive activation of serine kinases in the MAPK-ERK pathway may contribute to resistance to insulin's metabolic actions in skeletal muscle. Insulin functions as a co-gonadotropin through its cognate receptor to modulate ovarian steroidogenesis. Genetic disruption of insulin signaling in the brain has indicated that this pathway is important for ovulation and body weight regulation. These insights have been directly translated into a novel therapy for PCOS with insulin-sensitizing drugs. Furthermore, androgens contribute to insulin resistance in PCOS. PCOS may also have developmental origins due to androgen exposure at critical periods or to intrauterine growth restriction. PCOS is a complex genetic disease, and first-degree relatives have reproductive and metabolic phenotypes. Several PCOS genetic susceptibility loci have been mapped and replicated. Some of the same susceptibility genes contribute to disease risk in Chinese and European PCOS populations, suggesting that PCOS is an ancient trait.
Collapse
|
27
|
Velleman SG, Shin J, Li X, Song Y. Review: The skeletal muscle extracellular matrix: Possible roles in the regulation of muscle development and growth. CANADIAN JOURNAL OF ANIMAL SCIENCE 2012. [DOI: 10.4141/cjas2011-098] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 12/21/2022]
Abstract
Velleman, S. G., Shin, J., Li, X. and Song, Y. 2012. Review: The skeletal muscle extracellular matrix: Possible roles in the regulation of muscle development and growth. Can. J. Anim. Sci. 92: 1–10. Skeletal muscle fibers are surrounded by an extrinsic extracellular matrix environment. The extracellular matrix is composed of collagens, proteoglycans, glycoproteins, growth factors, and cytokines. How the extracellular matrix influences skeletal muscle development and growth is an area that is not completely understood at this time. Studies on myogenesis have largely been directed toward the cellular components and overlooked that muscle cells secrete a complex extracellular matrix network. The extracellular matrix modulates muscle development by acting as a substrate for muscle cell migration, growth factor regulation, signal transduction of information from the extracellular matrix to the intrinsic cellular environment, and provides a cellular structural architecture framework necessary for tissue function. This paper reviews extracellular matrix regulation of muscle growth with a focus on secreted proteoglycans, cell surface proteoglycans, growth factors and cytokines, and the dynamic nature of the skeletal muscle extracellular matrix, because of its impact on the regulation of muscle cell proliferation and differentiation during myogenesis.
Collapse
Affiliation(s)
- Sandra G. Velleman
- Ohio Agricultural Research and Development Center/The Ohio State University, Department of Animal Sciences, Wooster, OH 44691, USA
| | - Jonghyun Shin
- Ohio Agricultural Research and Development Center/The Ohio State University, Department of Animal Sciences, Wooster, OH 44691, USA
| | - Xuehui Li
- University of Florida, Department of Anatomy and Cell Biology, Gainesville, FL 32610, USA
| | - Yan Song
- Ohio Agricultural Research and Development Center/The Ohio State University, Department of Animal Sciences, Wooster, OH 44691, USA
| |
Collapse
|
28
|
Loganathan G, Dawra RK, Pugazhenthi S, Guo Z, Soltani SM, Wiseman A, Sanders MA, papas KK, Kumaravel V, Saluja AK, Sutherland DE, Hering BJ, Balamurugan AN. Insulin degradation by acinar cell proteases creates a dysfunctional environment for human islets before/after transplantation: benefits of α-1 antitrypsin treatment. Transplantation 2011; 92:1222-30. [PMID: 22089666 PMCID: PMC3587768 DOI: 10.1097/tp.0b013e318237585c] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Pancreatic acinar cells are commonly cotransplanted along with islets during auto- and allotransplantations. The aims of this study were to identify how acinar cell proteases cause human islet cell loss before and after transplantation of impure islet preparations and to prevent islet loss and improve function with supplementation of α-1 antitrypsin (A1AT). METHODS Acinar cell protease activity, insulin levels, and percent islet loss were measured after culture of pure and impure clinical islet preparations. The effect of proteases on ultrastructure of islets and β-cell insulin granules were examined by transmission electron microscopy. The number of insulin granules and insulin-labeled immunogold particles were counted. The in vivo effect of proteases on islet function was studied by transplanting acinar cells adjacent to islet grafts in diabetic mice. The effects of A1AT culture supplementation on protease activity, insulin levels, and islet function were assessed in pure and impure islets. RESULTS Islet loss after culture was significantly higher in impure relative to pure preparations (30% vs. 14%, P<0.04). Lower islet purity was associated with increased protease activity and decreased insulin levels in culture supernatants. Reduced β-cell insulin granules and insulin degradation by proteases were confirmed by transmission electron microscopy. Transplantations in mice showed delayed islet graft function when acinar cells were transplanted adjacent to the islets under the kidney capsule. Supplementation of A1AT to impure islet cultures maintained islet cell mass, restored insulin levels, and preserved islet functional integrity. CONCLUSION Culture of impure human islet fractions in the presence of A1AT prevents insulin degradation and improves islet recovery.
Collapse
Affiliation(s)
| | - Rajinder K. Dawra
- Department of Surgery, Basic and Translational Research, University of Minnesota, Minneapolis, MN.
| | | | - Zhiguang Guo
- Sanford Project, Sanford Health/University of South Dakota, Sioux Falls, SD.
| | - Sajjad M. Soltani
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN.
| | | | | | - Klearchos K. papas
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN.
| | - V. Kumaravel
- Alpha Hospital and Research Center, Institute of Diabetes and Endocrinology, Madurai-9, Tamilnadu, India.
| | - Ashok K. Saluja
- Department of Surgery, Basic and Translational Research, University of Minnesota, Minneapolis, MN.
| | - David E.R. Sutherland
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN.
| | - Bernhard J. Hering
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN.
| | - A. N. Balamurugan
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN.
- Corresponding author: Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN, 55455, USA. Phone: 651-253-0656, Fax: 612-626-5855
| |
Collapse
|
29
|
|
30
|
Daoud J, Rosenberg L, Tabrizian M. Pancreatic Islet Culture and Preservation Strategies: Advances, Challenges, and Future Outlook. Cell Transplant 2010; 19:1523-35. [DOI: 10.3727/096368910x515872] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/07/2023] Open
Abstract
Postisolation islet survival is a critical step for achieving successful and efficient islet transplantation. This involves the optimization of islet culture in order to prolong survival and functionality in vitro. Many studies have focused on different strategies to culture pancreatic islets in vitro through manipulation of culture media, surface modified substrates, and the use of various techniques such as encapsulation, embedding, scaffold, and bioreactor culture strategies. This review aims to present and discuss the different methodologies employed to optimize pancreatic islet culture in vitro as well as address their respective advantages and drawbacks.
Collapse
Affiliation(s)
- Jamal Daoud
- Department of Biomedical Engineering, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Lawrence Rosenberg
- Department of Surgery, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Maryam Tabrizian
- Department of Biomedical Engineering, Faculty of Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
31
|
Brothers KJ, Wu S, DiVall SA, Messmer MR, Kahn CR, Miller RS, Radovick S, Wondisford FE, Wolfe A. Rescue of obesity-induced infertility in female mice due to a pituitary-specific knockout of the insulin receptor. Cell Metab 2010; 12:295-305. [PMID: 20816095 PMCID: PMC2935812 DOI: 10.1016/j.cmet.2010.06.010] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 11/12/2009] [Revised: 03/17/2010] [Accepted: 06/01/2010] [Indexed: 12/21/2022]
Abstract
Obesity is associated with insulin resistance in metabolic tissues such as adipose, liver, and muscle, but it is unclear whether nonclassical target tissues, such as those of the reproductive axis, are also insulin resistant. To determine if the reproductive axis maintains insulin sensitivity in obesity in vivo, murine models of diet-induced obesity (DIO) with and without intact insulin signaling in pituitary gonadotrophs were created. Diet-induced obese wild-type female mice (WT DIO) were infertile and experienced a robust increase in luteinizing hormone (LH) after gonadotropin-releasing hormone (GnRH) or insulin stimulation. By contrast, both lean and obese mice with a pituitary-specific knockout of the insulin receptor (PitIRKO) exhibited reproductive competency, indicating that insulin signaling in the pituitary is required for the reproductive impairment seen in DIO and that the gonadotroph maintains insulin sensitivity in a setting of peripheral insulin resistance.
Collapse
Affiliation(s)
- Kathryn J Brothers
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
ABSTRACTSomatomedin C and other hormones, as well as blood metabolites, were measured during the dry period and during lactation in dairy cows, given different amounts of energy and protein, to study metabolic and endocrine adaptations. Somatomedin C, specifically measured by radioimmunoassay after separation from its binding protein, did not exhibit typical diurnal variations, in contrast to somatotropin and insulin, which increased particularly after concentrate intake. Somatomedin C markedly decreased at parturition and reached lowest values around the peak of lactation, while levels of somatotropin, nonesterified fatty acids and ketone bodies were high and those of glucose, insulin, thyroxine and triiodothyronine were low. Thereafter somatomedin C values slowly increased up to the 12th week of lactation and remained elevated. Low energy and protein balances were characterized by particularly low somatomedin C concentrations. An additional protein deficit at peak lactation, when cows were already provided with low amounts of energy, did not further decrease somatomedin C levels. However, when high amounts of energy were given in the form of starch or crystalline fat, somatomedin C increased. Overall, there was a positive correlation of somatomedin C primarily with energy, but also with protein balances and a negative correlation with milk yield. Conversely, somatotropin increased markedly after parturition and was positively correlated with milk production and negatively with protein and energy balances. Thus, somatomedin C levels were paradoxically low in the presence of high circulating somatotropin. Insulin most closely paralleled somatomedin C levels. Therefore the anabolic state of metabolism at the end of pregnancy was characterized by high somatomedin C and insulin and relatively low somatotropin, whereas the catabolic state of early lactation was characterized by high somatotropin, low somatomedin C, insulin and thyroid hormones.
Collapse
|
33
|
Kelly CJ, Stenton SR, Lashen H. Insulin-like growth factor binding protein-1 in PCOS: a systematic review and meta-analysis. Hum Reprod Update 2010; 17:4-16. [PMID: 20634211 DOI: 10.1093/humupd/dmq027] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Despite extensive research, the pathogenesis of polycystic ovary syndrome (PCOS) remains unclear. Putatively, an elevated circulating concentration of insulin inhibits the production of insulin-like growth factor binding protein-1 (IGFBP-1), thus increasing the level of free IGF-I in serum and stimulating ovarian androgen production. Decreased IGFBP-1 has been reported in PCOS and in obesity; however, there are inconsistencies in the evidence. This systematic review and meta-analysis aimed to determine whether IGFBP-1 is decreased in PCOS when controlling for the influence of BMI. METHODS Articles published between 1988 and 2008 were searched using MEDLINE, PubMed, SCOPUS and Web of Knowledge. Unpublished literature, trials in progress, and recent reviews were also searched. Original articles were selected by two investigators. To be included, the study must have compared serum IGFBP-1 in two populations: either PCOS versus controls, or an overweight subgroup versus the normal weight subgroup in either population. From 617 identified articles, 12 were included in the meta-analysis. Data were abstracted by two reviewers independently and standardized for errors. RESULTS The population difference is presented as the Weighted Mean Difference (95% CI). PCOS subjects had a significantly lower serum concentrations of IGFBP-1 compared with controls [P< 0.00001; -36.6 (-52.0, -21.2) µg/l]. Overweight PCOS subjects also had lower IGFBP-1 levels compared with normal weight PCOS subjects [P < 0.006; -30.6 (-52.3, -8.8) µg/l]. No significant difference was found between overweight PCOS patients and overweight controls [P = 0.23; -5.1 (-13.5, 3.2) µg/l] or between normal weight PCOS patients and normal weight controls [P = 0.50; -3.8 (-14.9, 7.3) µg/l]. Overweight controls had significantly lower IGFBP-1 concentrations than normal weight controls [P = 0.03; -18.0 (-34.4, - 1.5) µg/l]. CONCLUSION These data indicate that a decreased serum level of IGFBP-1 is unlikely to be a mechanism for ovarian hyperandrogenism in PCOS. BMI may be the major determinant of serum IGFBP-1.
Collapse
Affiliation(s)
- C J Kelly
- Department of Reproductive and Developmental Medicine, University of Sheffield, New Jessop Wing, Sheffield S10 2SF, UK
| | | | | |
Collapse
|
34
|
Okamoto N, Yamanaka N, Yagi Y, Nishida Y, Kataoka H, O'Connor MB, Mizoguchi A. A fat body-derived IGF-like peptide regulates postfeeding growth in Drosophila. Dev Cell 2010; 17:885-91. [PMID: 20059957 DOI: 10.1016/j.devcel.2009.10.008] [Citation(s) in RCA: 194] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/22/2009] [Revised: 09/14/2009] [Accepted: 10/21/2009] [Indexed: 01/13/2023]
Abstract
Members of the insulin family of peptides have conserved roles in the regulation of growth and metabolism in a wide variety of metazoans. Here we show that Drosophila insulin-like peptide 6 (DILP6), which is structurally similar to vertebrate insulin-like growth factor (IGF), is predominantly expressed in the fat body, a functional equivalent of the vertebrate liver and adipocytes. This expression occurs during the postfeeding stage under the direct regulation of ecdysteroid. We further reveal that dilp6 mutants show growth defects during the postfeeding stage, which results in reduced adult body size through a decrease in cell number. This phenotype is rescued by fat body-specific expression of dilp6. These data indicate that DILP6 is a functional, as well as a structural, counterpart of vertebrate IGFs. Our data provide in vivo evidence for a role of ILPs in determining adult body size through the regulation of postfeeding growth.
Collapse
Affiliation(s)
- Naoki Okamoto
- Division of Biological Science, Graduate School of Science, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8602, Japan
| | | | | | | | | | | | | |
Collapse
|
35
|
Okamoto N, Yamanaka N, Satake H, Saegusa H, Kataoka H, Mizoguchi A. An ecdysteroid-inducible insulin-like growth factor-like peptide regulates adult development of the silkmoth Bombyx mori. FEBS J 2009; 276:1221-32. [PMID: 19175674 DOI: 10.1111/j.1742-4658.2008.06859.x] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/29/2022]
Abstract
Insulin-like growth factors (IGFs) play essential roles in fetal and postnatal growth and development of mammals. They are secreted by a wide variety of tissues, with the liver being the major source of circulating IGFs, and regulate cell growth, differentiation and survival. IGFs share some biological activities with insulin but are secreted in distinct physiological and developmental contexts, having specific functions. Although recent analyses of invertebrate genomes have revealed the presence of multiple insulin family peptide genes in each genome, little is known about functional diversification of the gene products. Here we show that a novel insulin family peptide of the silkmoth Bombyx mori, which was purified and sequenced from the hemolymph, is more like IGFs than like insulin, in contrast to bombyxins, which are previously identified insulin-like peptides in B. mori. Expression analysis reveals that this IGF-like peptide is predominantly produced by the fat body, a functional equivalent of the vertebrate liver and adipocytes, and is massively released during pupa-adult development. Studies using in vitro tissue culture systems show that secretion of the peptide is stimulated by ecdysteroid and that the secreted peptide promotes the growth of adult-specific tissues. These observations suggest that this peptide is a Bombyx counterpart of vertebrate IGFs and that functionally IGF-like peptides may be more ubiquitous in the animal kingdom than previously thought. Our results also suggest that the known effects of ecdysteroid on insect adult development may be in part mediated by IGF-like peptides.
Collapse
Affiliation(s)
- Naoki Okamoto
- Division of Biological Science, Graduate School of Science, Nagoya University, Chikusa-ku, Nagoya, Japan
| | | | | | | | | | | |
Collapse
|
36
|
THORNDYKE MC. Molecular diversity and conformity of neurohormonal peptides: clues to an adaptive role in evolution. Biol J Linn Soc Lond 2008. [DOI: 10.1111/j.1095-8312.1988.tb01962.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/30/2022]
|
37
|
Gutiérrez A, Nieto J, Pozo F, Stern S, Schoofs L. Effect of insulin/IGF-I like peptides on glucose metabolism in the white shrimp Penaeus vannamei. Gen Comp Endocrinol 2007; 153:170-5. [PMID: 17574553 DOI: 10.1016/j.ygcen.2007.04.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 09/15/2006] [Revised: 03/02/2007] [Accepted: 04/19/2007] [Indexed: 11/20/2022]
Abstract
The insulin-like hormone superfamily encompasses insulin, relaxin, and insulin-like growth factors I (IGF1) and II (IGF2). Insulin hormones regulate cell growth, metabolism, and tissue-specific functions. The presence of insulin has been demonstrated in various invertebrates, and their function as growth promoting or controlling factors has been established in molluscs and insects. In crustaceans, the presence of insulin/insulin-like growth factor (IGF)-like peptides has also been suggested and functional studies have been associated with metabolic control. The general aim of the current study was to elucidate the functional significance of insulin-like peptides in the white shrimp Penaeus vannamei. Because the primary structure of Penaeus insulin is yet unknown, we examined the effect of mammalian insulin/IGF-I on glucose metabolism in P. vannamei. Juvenile shrimps were injected with a single dose of recombinant human (rh) IGF-I or bovine insulin in intermolt stage. Glucose/glycogen levels in shrimp hemolymph and tissues (muscle, hepatopancreas and gills) were determined over a 5h period by means of an enzymatic analysis. We showed that an injection of rhIGF-I induced a significant (P<0.01) increase in glucose levels in hemolymph, 1h after injection and followed by a decrease (P<0.05) 5h post-injection. In the hepatopancreas, an increase (P<0.05) in the glycogen content was observed 3h after insulin treatment. Finally, a significant elevation (P<0.01) of glycogen content in the gills throughout the entire sampling period was detected. Our study suggests the presence of endogenous Penaeus insulin(s) that, just like its vertebrate counterparts, is likely to be involved in the regulation of carbohydrate metabolism in crustaceans.
Collapse
Affiliation(s)
- Ana Gutiérrez
- Laboratory for Developmental Physiology, Genomics and Proteomics, Katholieke Universiteit Leuven, Naamsestraat 59, B-3000 Leuven, Belgium.
| | | | | | | | | |
Collapse
|
38
|
Takahashi S, Kajikawa M, Umezawa T, Takahashi SI, Kato H, Miura Y, Nam TJ, Noguchi T, Naito H. Effect of dietary proteins on the plasma immunoreactive insulin-like growth factor-1/somatomedin C concentration in the rat. Br J Nutr 2007. [DOI: 10.1079/bjn19900139] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/11/2022]
|
39
|
Murdoch TB, McGhee-Wilson D, Shapiro AMJ, Lakey JRT. Methods of human islet culture for transplantation. Cell Transplant 2005; 13:605-618. [PMID: 15648731 DOI: 10.3727/000000004783983602] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/07/2023] Open
Abstract
The ability to maintain isolated human islet preparations in tissue culture has recently been adopted by most islet transplant centers, and improves the safety as well as the practicality of islet transplantation. Maintaining islet viability and recovery, however, remains challenging in a clinical setting, due to stringent conditions required for culture. Islet culture is further complicated by the fact that islets do not form a monolayer. This review aims to clarify media, supplementation, and conditions that have been shown to be relevant to human islets, as well as to offer avenues of future research. Factors examined that may influence islet survival include base medium, glucose concentration, vitamin, inorganic ion, lipid, hormone, growth factor, amino acid, and binding protein composition and concentration, as well as culture temperature and seeding density. In addition, this article reviews novel techniques, such as coculture and matrices, that have been employed in an attempt to improve islet survival and functional viability.
Collapse
Affiliation(s)
- T B Murdoch
- Clinical Islet Transplant Program, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | |
Collapse
|
40
|
Abstract
Recent animal studies have demonstrated evidence of the involvement of insulin and insulin-like growth factor (IGF)-I signalling in the control of ageing and longevity. Disruption of insulin/IGF-I signalling pathways significantly extends lifespan in several animal models. Similarities among these signalling pathways in animals and humans raise the possibility that modifications in the IGF-I signalling system could also extend lifespan in humans. However, in contrast to the findings in animal studies, reduced IGF-I activity in humans is not associated with longevity. In humans, low IGF-I activity is even associated with an increased risk of developing cardiovascular disease and diabetes. High IGF-I activity in humans is associated with an increased risk of developing cancer. In addition, genetic predisposition and lifestyle play a major role in determining age-associated disease. For each individual there is probably a specific optimal 'setpoint' for the insulin/growth hormone/IGF-I axis which co-determines survival.
Collapse
Affiliation(s)
- J A M J L Janssen
- Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands.
| | | |
Collapse
|
41
|
Naumann A, Dennis JE, Aigner J, Coticchia J, Arnold J, Berghaus A, Kastenbauer ER, Caplan AI. Tissue engineering of autologous cartilage grafts in three-dimensional in vitro macroaggregate culture system. ACTA ACUST UNITED AC 2005; 10:1695-706. [PMID: 15684678 DOI: 10.1089/ten.2004.10.1695] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/13/2022]
Abstract
In the field of tissue engineering, techniques have been described to generate cartilage tissue with isolated chondrocytes and bioresorbable or nonbioresorbable biomaterials serving as three-dimensional cell carriers. In spite of successful cartilage engineering, problems of uneven degradation of biomaterial, and unforeseeable cell-biomaterial interactions remain. This study represents a novel technique to engineer cartilage by an in vitro macroaggregate culture system without the use of biomaterials. Human nasoseptal or auricular chondrocytes were enzymatically isolated and amplified in conventional monolayer culture before the cells were seeded into a cell culture insert with a track-etched membrane and cultured in vitro for 3 weeks. The new cartilage formed within the in vitro macroaggregates was analyzed by histology (toluidine blue, von Kossa-safranin O staining), and immunohistochemistry (collagen types I, II, V, VI, and X and elastin). The total glycosaminoglycan (GAG) content of native and engineered auricular as well as nasal cartilage was assayed colorimetrically in a safranin O assay. The biomechanical properties of engineered cartilage were determined by biphasic indentation assay. After 3 weeks of in vitro culture, nasoseptal and auricular chondrocytes synthesized new cartilage with the typical appearance of hyaline nasal cartilage and elastic auricular cartilage. Immunohistochemical staining of cartilage samples showed a characteristic pattern of staining for collagen antibodies that varied in location and intensity. In all samples, intense staining for cartilage-specific collagen types I, II, and X was observed. By the use of von Kossa-safranin O staining a few positive patches-a possible sign of beginning mineralization within the engineered cartilages-were detected. The unique pattern for nasoseptal cartilage is intense staining for type V collagen, whereas auricular cartilage is only weakly positive for collagen types V and VI. Engineered nasal and auricular macroaggregates were negative for anti-elastin antibody (interterritorially). The measurement of total GAG content demonstrated higher GAG content for reformed nasoseptal cartilage compared with elastic auricular cartilage. However, the total GAG content of engineered macroaggregates was lower than that of native cartilage. In spite of the mechanical stability of the auricular macroaggregates, there was no equilibrium of indentation. The histomorphological and immunohistochemical results demonstrate successful cartilage engineering without the use of biomaterials, and identify characteristics unique to hyaline as well as elastic cartilage. The GAG content of engineered cartilage was lower than in native cartilage and the biomechanical properties were not determinable by indentation assay. This study illustrates a novel in vitro macroaggregate culture system as a promising technique for tissue engineering of cartilage grafts. Further long-term in vitro and in vivo studies must be done before this method can be applied to reconstructive surgery of the nose or auricle.
Collapse
Affiliation(s)
- Andreas Naumann
- Department of Otorhinolaryngology, Head and Neck Surgery, Ludwig Maximilian University, Munich, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Janssen JAMJL, van der Lely AJ, Lamberts SWJ. Circulating free insulin-like growth-factor-I (IGF-I) levels should also be measured to estimate the IGF-I bioactivity. J Endocrinol Invest 2003; 26:588-94. [PMID: 12952376 DOI: 10.1007/bf03345225] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 10/25/2022]
Abstract
Free IGF-I by analogy with sex and adrenal steroids and thyroid hormones, may be the major biologically active hormonal form of IGF-I. Because of methodological difficulties in measuring the free IGF-I the measurement of total IGF-I in blood is often used to assess the activity of the endocrine GH-IGF-I axis in clinical studies. However, there is currently no reliable standard reference method for circulating total IGF-I against which individual samples can be calibrated. In addition, in many of the common methods used to measure circulating total IGF-I levels, remaining insulin-like growth factor, binding proteins (IGFBPs) or binding protein fragments after sample extraction, may still interfere and produce falsely increased or decreased circulating total IGF-I levels. This latter phenomenon occurs especially under pathologic conditions. In addition, it has also been suggested that altered post-sampling integrity of IGF-I in vitro might contribute to the reported inconsistencies in circulating total IGF-I levels in literature. Although at the moment there is also no "golden standard" for the measurement of circulating free IGF-I levels, we discuss some studies in this paper that in our opinion, have demonstrated conclusively that circulating free IGF-I levels in several conditions reflect the IGF-I bioactivity better than circulating total IGF-I levels. Therefore, when evaluating the IGF-I bioactivity in health and disease, we recommend measuring also circulating free IGF-I.
Collapse
Affiliation(s)
- J A M J L Janssen
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands.
| | | | | |
Collapse
|
43
|
Senthil D, Choudhury GG, Abboud HE, Sonenberg N, Kasinath BS. Regulation of protein synthesis by IGF-I in proximal tubular epithelial cells. Am J Physiol Renal Physiol 2002; 283:F1226-36. [PMID: 12388420 DOI: 10.1152/ajprenal.00109.2002] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/22/2022] Open
Abstract
Protein synthesis is required for renal hypertrophy, and proximal tubular epithelial cells are an important cell type involved in this process. We examined IGF-I regulation of protein synthesis in murine proximal tubular epithelial (MCT) cells. We focused on initial events in protein translation and the signaling events involved. Translation of capped mRNAs is under the control of eukaryotic initiation factor 4E (eIF4E). In the resting cell, eIF4E is normally kept in an inactive state by binding to 4E-BP1, its binding protein. Phosphorylation of 4E-BP1 results in dissociation of the eIF4E-4E-BP1 complex allowing eIF4E to initiate peptide synthesis. IGF-I stimulated protein synthesis, augmented phosphorylation of 4E-BP1 and promoted the dissociation of eIF4E from 4E-BP1. IGF-I stimulated the activities of phosphatidylinositol (PI) 3-kinase, Akt, and ERK1/2-type MAPK in MCT cells. IGF-I-induced phosphorylation of 4E-BP1, dissociation of the 4E-BP1-eIF4E complex, and increase in protein synthesis required activation of both PI 3-kinase and ERK pathways. Furthermore, ERK activation by IGF-I was also PI 3-kinase dependent. Transfection with the Thr37,46-->Ala37,46 mutant of 4E-BP1 showed that phosphorylation of Thr37,46 residues was required for IGF-I induction of protein synthesis in MCT cells. Our observations reveal the importance of initial events in protein translation in IGF-I-induced protein synthesis in MCT cells and identify the regulatory signaling pathways involved.
Collapse
Affiliation(s)
- Duraisamy Senthil
- Division of Nephrology, Department of Medicine, University of Texas Health Science Center, South Texas Veterans Health Care System, Geriatrics Research and Education Center, San Antonio, Texas 78229-3900, USA
| | | | | | | | | |
Collapse
|
44
|
Claeys I, Simonet G, Poels J, Van Loy T, Vercammen L, De Loof A, Vanden Broeck J. Insulin-related peptides and their conserved signal transduction pathway. Peptides 2002; 23:807-16. [PMID: 11897402 DOI: 10.1016/s0196-9781(01)00666-0] [Citation(s) in RCA: 137] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 02/06/2023]
Abstract
The 'insulin superfamily' is an ancient category of small, structurally related proteins, such as insulin, insulin-like growth factors (IGF) and relaxin. Insulin-like signaling molecules have also been described in different invertebrates, including nematodes, mollusks, and insects. They initiate an evolutionary conserved signal transduction mechanism by binding to a heterotetrameric, membrane-spanning receptor tyrosine kinase. Recent physiological and genetic studies have revealed that, in different metazoans, the insulin signaling pathway plays a pivotal role in the regulation of a variety of interrelated, fundamental processes, such as metabolism, growth, reproduction and aging.
Collapse
Affiliation(s)
- Ilse Claeys
- Laboratory for Developmental Physiology and Molecular Biology, K.U., Leuven, Belgium.
| | | | | | | | | | | | | |
Collapse
|
45
|
Thorne RG, Frey WH. Delivery of neurotrophic factors to the central nervous system: pharmacokinetic considerations. Clin Pharmacokinet 2002; 40:907-46. [PMID: 11735609 DOI: 10.2165/00003088-200140120-00003] [Citation(s) in RCA: 348] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/21/2022]
Abstract
Neurotrophic factors are proteins with considerable potential in the treatment of central nervous system (CNS) diseases and traumatic injuries. However, a significant challenge to their clinical use is the difficulty associated with delivering these proteins to the CNS. Neurotrophic factors are hydrophilic, typically basic, monomeric or dimeric proteins, mostly in the size range of 5 to 30 kDa. Neurotrophic factors potently support the development, growth and survival of neurons, eliciting biological effects at concentrations in the nanomolar to femtomolar range. They are not orally bioavailable and the blood-brain and blood-cerebrospinal fluid barriers severely limit their ability to enter into and act on sites in the CNS following parenteral systemic routes of administration. Most neurotrophic factors have short in vivo half-lives and poor pharmacokinetic profiles. Their access to the CNS is restricted by rapid enzymatic inactivation, multiple clearance processes, potential immunogenicity and sequestration by binding proteins and other components of the blood and peripheral tissues. The development of targeted drug delivery strategies for neurotrophic factors will probably determine their clinical effectiveness for CNS conditions. Achieving significant CNS target site concentrations while limiting systemic exposure and distribution to peripheral sites of action will lessen unwanted pleiotropic effects and toxicity. Local introduction of neurotrophic factors into the CNS intraparenchymally by direct injection/infusion or by implantation of delivery vectors such as polymer matrices or genetically modified cells yields the highest degree of targeting, but is limited by diffusion restrictions and invasiveness. Delivery of neurotrophic factors into the cerebrospinal fluid (CSF) following intracerebroventricular or intrathecal administration is less invasive and allows access to a much wider area of the CNS through CSF circulation pathways. However, diffusional and cellular barriers to penetration into surrounding CNS tissue and significant clearance of CSF into the venous and lymphatic circulation are also limiting. Unconventional delivery strategies such as intranasal administration may offer some degree of CNS targeting with minimal invasiveness. This review presents a summary of the neurotrophic factors and their indications for CNS disorders, their physicochemical characteristics and the different approaches that have been attempted or suggested for their delivery to the CNS. Future directions for further research such as the potential for CNS disease treatment utilising combinations of neurotrophic factors, displacement strategies, small molecule mimetics, chimaeric molecules and gene therapy are also discussed.
Collapse
Affiliation(s)
- R G Thorne
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota, USA
| | | |
Collapse
|
46
|
Affiliation(s)
- Denis A Magoffin
- Department of Obstetrics and Gynecology, CSMC Burns and Allen Research Institute, UCLA School of Medicine, Los Angeles, California 90048-0750, USA.
| |
Collapse
|
47
|
Bayram F, Unluhizarci K, Kelestimur F. Potential Utility of Insulin Sensitizers in the Treatment of Patients with Polycystic Ovary Syndrome. ACTA ACUST UNITED AC 2002; 1:45-53. [PMID: 15765620 DOI: 10.2165/00024677-200201010-00005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/02/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a form of functional ovarian hyperandrogenism. It is one of the most common endocrinologic disorders in women of reproductive age. Women with PCOS may present with menstrual irregularities, chronic anovulation, infertility, obesity and hyperandrogenism. PCOS is associated with insulin resistance and hyperinsulinemia in most, if not all, patients. Besides its gynecological manifestations, PCOS is also associated with increased metabolic and cardiovascular risks. These risks are strongly linked to insulin resistance, and during recent years a novel therapeutic approach using insulin-sensitizing agents has been extensively investigated. In most studies conducted to date, metformin and the thiazolidinedione agent troglitazone have resulted in improved insulin sensitivity, resumption of regular menses and decreased serum androgen levels. Insulin-sensitizing agents may be a useful adjuvant therapy for women with PCOS.
Collapse
Affiliation(s)
- Fahri Bayram
- Department of Endocrinology, Erciyes University Medical School, Kayseri, Turkey.
| | | | | |
Collapse
|
48
|
Abstract
Over the past 20 years, it has been clearly documented that the polycystic ovary syndrome (PCOS) has major metabolic sequelae related to insulin resistance and that insulin resistance plays an important role in the pathogenesis of the reproductive disturbances of the disorder. Family studies have indicated a genetic susceptibility to PCOS. Polycystic ovaries and hyperandrogenemia are present in approximately 50% of sisters of affected women. Increased androgen secretion and insulin resistance persist in cultured theca cells and skin fibroblasts, respectively, from women with PCOS; this finding suggests that these are intrinsic, presumably genetic, defects. Insulin resistance and elevated low-density lipoprotein (LDL) levels also cluster in the sisters of women with PCOS, consistent with genetic traits. Moreover, the brothers of women with PCOS have insulin resistance and elevated dehydroepiandrosterone sulfate (DHEAS) levels, which supports a genetic basis for these findings. Family-based studies of linkage and association have implicated several genes in the pathogenesis of PCOS. The strongest evidence to date points to a gene in the region of the insulin receptor. Insulin-sensitizing therapy mitigates the reproductive disturbances of PCOS.
Collapse
Affiliation(s)
- A Dunaif
- Division of Women's Health, Brigham and Women's Hospital, 75 Francis Street, Boston, Massachusetts 02115, USA.
| | | |
Collapse
|
49
|
A Lynch S, Elton CW, Melinda Carver F, Pennington SN. Alcohol-Induced Modulation of the Insulin-Like Growth Factor System in Early Chick Embryo Cranial Tissue. Alcohol Clin Exp Res 2001. [DOI: 10.1111/j.1530-0277.2001.tb02276.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/28/2022]
|
50
|
Abstract
There has been a debate for many years on whether muscular training is beneficial or harmful for patients with myopathic disorders and the role of exercise training in the management of these patients is still controversial. Much of this confusion is because of the lack of well-designed controlled training studies on this heterogenic group of disorders. Because effective therapies are still lacking, the patients have to rely on symptomatic treatment in which continuous physiotherapy plays an important role. There is thus still a need for studies evaluating the short- and long-term effects of muscular training in different types of myopathic disorders. We need to elucidate whether muscular training can increase strength and resistance to fatigue, but most importantly, we need to clarify whether training can improve specific functional abilities of the patient with myopathy. Future studies should give us specific information on what type of training, endurance or strength training, is to be preferred for different myopathies. The effect of strength training in one type of muscle disorder is not directly applicable to another, but is largely dependent on the underlying biological defect. From the studies published so far, high-resistance strength training at submaximal and possibly also at near-maximal levels seem beneficial, at least in the short perspective for slowly progressive myopathic disorders. However, the long-term effects of such training have not been systematically studied. In rapidly progressive myopathies, which are caused by deficient structural proteins such as in Duchenne's muscular dystrophy, the use of high-resistance training is far more controversial and questionable. If exercise regimens are to be used, they should preferably commence in the early stages of the disease, at which time there is still a substantial amount of trainable muscle fibres.
Collapse
Affiliation(s)
- T Ansved
- Department of Clinical Neuroscience, Division of Neurology and Clinical Neurophysiology, Karolinska Hospital, Sweden
| |
Collapse
|