1
|
Tang J, Zhao C, Lin S, Li X, Zhu B, Li Y. Controversial causal association between IGF family members and osteoporosis: a Mendelian randomization study between UK and FinnGen biobanks. Front Endocrinol (Lausanne) 2024; 14:1332803. [PMID: 38260127 PMCID: PMC10801076 DOI: 10.3389/fendo.2023.1332803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
Objectives Osteoporosis, a prevalent skeletal disorder characterized by reduced bone strength, is closely linked to the IGF system, crucial for skeletal metabolism. However, the precise nature of this relationship remains elusive. In this study, we employed Mendelian randomization (MR) to unravel the associations between genetically predicted serum IGF system member levels and osteoporosis. Methods A two-sample MR approach was employed to investigate these causal associations based on two individual datasets. Predictions of 14 serum levels of IGF system members were made using 11,036,163 relevant Single Nucleotide Polymorphisms (SNPs) within a cohort of 4,301 individuals of European descent. Genetic association estimates for osteoporosis were derived from two publicly available GWAS consortia: the Finnish consortium from the FinnGen biobank, comprising 212,778 individuals of Finnish descent (3,203 cases and 209,575 controls), and the UK consortium from the UK Biobank, including 337,159 individuals of European descent (5,266 cases and 331,893 controls). Results According to the UK dataset, IGF-1 levels were associated with a reduced risk of osteoporosis, as indicated by the weighted median method (Odds Ratio [OR] = 0.998, 95% CI = 0.997-1.000, P = 0.032). Additionally, higher levels of IGFBP-3 were linked to a decreased risk of osteoporosis using the Inverse-Variance Weighted (IVW) method (OR = 0.999, 95% CI = 0.998-1.000, P = 0.019), and CTGF levels exhibited a negative association with osteoporosis, as determined by the weighted median method (OR = 0.998, 95% CI = 0.996-0.999, P = 0.004). In the FinnGen dataset, IGF-1 and IGFBP-3 were not identified to be associated with osteoporosis. While, IGF-LR1 levels displayed a negative association with osteoporosis, according to the MR-Egger method (OR = 0.886, 95% CI = 0.795-0.987, P = 0.036), while CYR61 was linked to an increased risk of osteoporosis based on both the weighted median and IVW methods (OR = 1.154, 95% CI = 1.009-1.319, P = 0.037, and OR = 1.115, 95% CI = 1.022-1.215, P = 0.014, respectively). Conclusion This study provides compelling evidence that certain IGF family members play a role in the pathogenesis of osteoporosis between different datasets, indicating population specific causal effects between IGF family and osteoporosis. Although the results from both datasets demonstrated that IGF family involved in the pathogenesis of osteoporosis, but the responding key molecules might be various among different population. Subsequent research is warranted to evaluate the potential of these biomarkers as targets for osteoporosis prevention and treatment in specific population.
Collapse
Affiliation(s)
| | | | | | | | - Binlu Zhu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education (MOE), Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education (MOE), Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
2
|
LaManna L, Chou CH, Lei H, Barton ER, Maliga P. Chloroplast transformation for bioencapsulation and oral delivery using the immunoglobulin G fragment crystallizable (Fc) domain. Sci Rep 2023; 13:18916. [PMID: 37919321 PMCID: PMC10622566 DOI: 10.1038/s41598-023-45698-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 10/23/2023] [Indexed: 11/04/2023] Open
Abstract
Proinsulin Like Growth Factor I (prolGF-I) and myostatin (Mstn) regulate muscle regeneration and mass when intravenously delivered. We tested if chloroplast bioencapsulated forms of these proteins may serve as a non-invasive means of drug delivery through the digestive system. We created tobacco (Nicotiana tabacum) plants carrying GFP-Fc1, proIGF-I-Fc1, and Mstn-Fc1 fusion genes, in which fusion with the immunoglobulin G Fc domain improved both protein stability and absorption in the small intestine. No transplastomic plants were obtained with the Mstn-Fc1 gene, suggesting that the protein is toxic to plant cells. proIGF-I-Fc1 protein levels were too low to enable in vivo testing. However, GFP-Fc1 accumulated at a high level, enabling evaluation of chloroplast-made Fc fusion proteins for oral delivery. Tobacco leaves were lyophilized for testing in a mouse system. We report that the orally administered GFP-Fc1 fusion protein (5.45 µg/g GFP-Fc1) has been taken up by the intestinal epithelium cells, evidenced by confocal microscopy. GFP-Fc1 subsequently entered the circulation where it was detected by ELISA. Data reported here confirm that chloroplast expression and oral administration of lyophilized leaves is a potential delivery system of therapeutic proteins fused with Fc1, with the advantage that the proteins may be stored at room temperature.
Collapse
Affiliation(s)
- Lisa LaManna
- Waksman Institute of Microbiology, Rutgers University, Piscataway, NJ, 08854, USA
| | - Chih-Hsuan Chou
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, 32611, USA
| | - Hanqin Lei
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, 32611, USA
| | - Elisabeth R Barton
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, 32611, USA.
| | - Pal Maliga
- Waksman Institute of Microbiology, Rutgers University, Piscataway, NJ, 08854, USA.
- Department of Plant Biology, Rutgers University, New Brunswick, NJ, 08901, USA.
| |
Collapse
|
3
|
Wong L, McMahon LP. Crosstalk between bone and muscle in chronic kidney disease. Front Endocrinol (Lausanne) 2023; 14:1146868. [PMID: 37033253 PMCID: PMC10076741 DOI: 10.3389/fendo.2023.1146868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/14/2023] [Indexed: 04/11/2023] Open
Abstract
With increasing life expectancy, the related disorders of bone loss, metabolic dysregulation and sarcopenia have become major health threats to the elderly. Each of these conditions is prevalent in patients with chronic kidney disease (CKD), particularly in more advanced stages. Our current understanding of the bone-muscle interaction is beyond mechanical coupling, where bone and muscle have been identified as interrelated secretory organs, and regulation of both bone and muscle metabolism occurs through osteokines and myokines via autocrine, paracrine and endocrine systems. This review appraises the current knowledge regarding biochemical crosstalk between bone and muscle, and considers recent progress related to the role of osteokines and myokines in CKD, including modulatory effects of physical exercise and potential therapeutic targets to improve musculoskeletal health in CKD patients.
Collapse
Affiliation(s)
- Limy Wong
- Department of Renal Medicine, Monash University Eastern Health Clinical School, Box Hill, VIC, Australia
- Department of Renal Medicine, Eastern Health, Box Hill, VIC, Australia
- *Correspondence: Limy Wong,
| | - Lawrence P. McMahon
- Department of Renal Medicine, Monash University Eastern Health Clinical School, Box Hill, VIC, Australia
- Department of Renal Medicine, Eastern Health, Box Hill, VIC, Australia
| |
Collapse
|
4
|
Insulin-like growth factor-1 levels are associated with high comorbidity of metabolic disorders in obese subjects; a Japanese single-center, retrospective-study. Sci Rep 2022; 12:20130. [PMID: 36418379 PMCID: PMC9684525 DOI: 10.1038/s41598-022-23521-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 11/01/2022] [Indexed: 11/24/2022] Open
Abstract
Insulin like growth factor-1 (IGF-1) plays important roles in metabolic functions, especially in adulthood. Additionally, obese subjects are reportedly predisposed to having low absolute IGF-1 levels. However, the prevalence and clinical characteristics of obese subjects with low IGF-1 levels are unknown. We examined 64 obese subjects with a body mass index (BMI) ≥ 35 kg/m2, with no history of endocrinological disorders, receiving inpatient care. IGF-1 levels were interpreted based on the IGF-1 standard deviation score (SDS) clinically used and standardized by age and sex (low IGF-1 group; ≤ - 2.0 SDS and standard IGF-1 group; - 2.0 < and < + 2.0 SDS). Notably, 26.6% of the subjects had low IGF-1. Body fat mass and percentage, but not BMI, were significantly higher in the low than in the standard IGF-1 group. Furthermore, natural log-transformed high-sensitivity C-reactive protein, and the frequencies of dyslipidemia and hyperuricemia were higher in the low IGF-1 group. Moreover, among the subjects without diabetes, fasting glucose levels were significantly higher in the low IGF-1 group. Stepwise variable selection procedure revealed body fat percentage to be a parameter most strongly associated with low IGF-1. Thus, low IGF-1 levels may be an important marker of adiposity-associated metabolic disorders in obese patients.
Collapse
|
5
|
LIM CHANGHYUN, NUNES EVERSONA, CURRIER BRADS, MCLEOD JONATHANC, THOMAS AARONCQ, PHILLIPS STUARTM. An Evidence-Based Narrative Review of Mechanisms of Resistance Exercise-Induced Human Skeletal Muscle Hypertrophy. Med Sci Sports Exerc 2022; 54:1546-1559. [PMID: 35389932 PMCID: PMC9390238 DOI: 10.1249/mss.0000000000002929] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Skeletal muscle plays a critical role in physical function and metabolic health. Muscle is a highly adaptable tissue that responds to resistance exercise (RE; loading) by hypertrophying, or during muscle disuse, RE mitigates muscle loss. Resistance exercise training (RET)-induced skeletal muscle hypertrophy is a product of external (e.g., RE programming, diet, some supplements) and internal variables (e.g., mechanotransduction, ribosomes, gene expression, satellite cells activity). RE is undeniably the most potent nonpharmacological external variable to stimulate the activation/suppression of internal variables linked to muscular hypertrophy or countering disuse-induced muscle loss. Here, we posit that despite considerable research on the impact of external variables on RET and hypertrophy, internal variables (i.e., inherent skeletal muscle biology) are dominant in regulating the extent of hypertrophy in response to external stimuli. Thus, identifying the key internal skeletal muscle-derived variables that mediate the translation of external RE variables will be pivotal to determining the most effective strategies for skeletal muscle hypertrophy in healthy persons. Such work will aid in enhancing function in clinical populations, slowing functional decline, and promoting physical mobility. We provide up-to-date, evidence-based perspectives of the mechanisms regulating RET-induced skeletal muscle hypertrophy.
Collapse
Affiliation(s)
- CHANGHYUN LIM
- Department of Kinesiology, McMaster University, Hamilton, Ontario, CANADA
| | - EVERSON A. NUNES
- Department of Kinesiology, McMaster University, Hamilton, Ontario, CANADA
- Department of Physiological Science, Federal University of Santa Catarina, Florianópolis, Santa-Catarina, BRAZIL
| | - BRAD S. CURRIER
- Department of Kinesiology, McMaster University, Hamilton, Ontario, CANADA
| | - JONATHAN C. MCLEOD
- Department of Kinesiology, McMaster University, Hamilton, Ontario, CANADA
| | - AARON C. Q. THOMAS
- Department of Kinesiology, McMaster University, Hamilton, Ontario, CANADA
| | - STUART M. PHILLIPS
- Department of Kinesiology, McMaster University, Hamilton, Ontario, CANADA
| |
Collapse
|
6
|
Wong L, Duque G, McMahon LP. Sarcopenia and Frailty: Challenges in Mainstream Nephrology Practice. Kidney Int Rep 2021; 6:2554-2564. [PMID: 34622096 PMCID: PMC8484128 DOI: 10.1016/j.ekir.2021.05.039] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 05/20/2021] [Accepted: 05/31/2021] [Indexed: 01/06/2023] Open
Abstract
Sarcopenia and frailty are prevalent in the chronic kidney disease (CKD) population. Sarcopenia is characterised by the loss of muscle mass and function, while frailty is defined as a multi-system impairment associated with increased vulnerability to stressors. There is substantial overlap between the 2 conditions, particularly with regards to physical aspects: low grip strength, gait speed and low muscle mass. Both sarcopenia and frailty have been associated with a wide range of adverse health outcomes. Although there is no recommended pharmacological treatment as yet, it is widely accepted that exercise training and nutritional supplementation are the key interventions to maintain skeletal muscle mass and strength. This review aims to present a comprehensive overview of sarcopenia and frailty in patients with CKD.
Collapse
Affiliation(s)
- Limy Wong
- Eastern Health Integrated Renal Service, Box Hill Hospital, Victoria, Australia.,Department of Renal Medicine, Monash University Eastern Health Clinical School, Victoria, Australia
| | - Gustavo Duque
- Australian Institute for Musculoskeletal Science (AIMSS), The University of Melbourne and Western Health, St. Albans, Victoria, Australia.,Department of Medicine-Western Health, Melbourne Medical School, The University of Melbourne, St. Albans, Victoria, Australia
| | - Lawrence P McMahon
- Eastern Health Integrated Renal Service, Box Hill Hospital, Victoria, Australia.,Department of Renal Medicine, Monash University Eastern Health Clinical School, Victoria, Australia
| |
Collapse
|
7
|
Jarmusch S, Baber L, Bidlingmaier M, Ferrari U, Hofmeister F, Hintze S, Mehaffey S, Meinke P, Neuerburg C, Schoser B, Tanganelli F, Drey M. Influence of IGF-I serum concentration on muscular regeneration capacity in patients with sarcopenia. BMC Musculoskelet Disord 2021; 22:807. [PMID: 34544407 PMCID: PMC8454138 DOI: 10.1186/s12891-021-04699-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 09/10/2021] [Indexed: 02/01/2023] Open
Abstract
Background Previous research has described a neuroprotective effect of IGF-I, supporting neuronal survival, axon growth and proliferation of muscle cells. Therefore, the association between IGF-I concentration, muscle histology and electrophysiological markers in a cohort of patients with sarcopenia dares investigation. Methods Measurement of serum concentrations of IGF-I and binding partners, electromyographic measurements with the MUNIX (Motor Unit Number Index) method and muscle biopsies were performed in 31 patients with acute hip fracture older age 60 years. Molecular markers for denervation (neural cell adhesion molecule NCAM) and proliferation markers (Ki67) were assessed by immunofluorescence staining of muscle biopsy tissue. Skeletal muscle mass by bioelectrical impedance analysis and hand-grip strength were measured to assess sarcopenia status according to EWGSOP2 criteria. Results Thirty-one patients (20 women) with a mean age of 80.6 ± 7.4 years were included. Concentrations of IGF-I and its binding partners were significantly associated with sarcopenia (ß = − 0.360; p = 0.047) and MUNIX (ß = 0.512; p = 0.005). Further, expression of NCAM (ß = 0.380; p = 0.039) and Ki67 (ß = 0.424; p = 0.022) showed significant associations to IGF-I concentrations. Conclusions The findings suggest a pathogenetic role of IGF-I in sarcopenia based on muscle denervation. Supplementary Information The online version contains supplementary material available at 10.1186/s12891-021-04699-3.
Collapse
Affiliation(s)
- Stefanie Jarmusch
- Department of Medicine IV, Geriatrics, University Hospital of LMU Munich, Munich, Germany
| | - Lisa Baber
- Department of Medicine IV, Geriatrics, University Hospital of LMU Munich, Munich, Germany
| | - Martin Bidlingmaier
- Department of Medicine IV, Endocrinological Laboratory, University Hospital of LMU Munich, Munich, Germany
| | - Uta Ferrari
- Department of Medicine IV, Geriatrics, University Hospital of LMU Munich, Munich, Germany
| | - Fabian Hofmeister
- Department of Medicine IV, Geriatrics, University Hospital of LMU Munich, Munich, Germany
| | - Stefan Hintze
- Friedrich-Baur-Institute, Department of Neurology, University Hospital of LMU Munich, Munich, Germany
| | - Stefan Mehaffey
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), University Hospital of LMU Munich, Munich, Germany
| | - Peter Meinke
- Friedrich-Baur-Institute, Department of Neurology, University Hospital of LMU Munich, Munich, Germany
| | - Carl Neuerburg
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), University Hospital of LMU Munich, Munich, Germany
| | - Benedikt Schoser
- Friedrich-Baur-Institute, Department of Neurology, University Hospital of LMU Munich, Munich, Germany
| | - Fabiana Tanganelli
- Department of Medicine IV, Geriatrics, University Hospital of LMU Munich, Munich, Germany
| | - Michael Drey
- Department of Medicine IV, Geriatrics, University Hospital of LMU Munich, Munich, Germany.
| |
Collapse
|
8
|
Yuan S, Wan ZH, Cheng SL, Michaëlsson K, Larsson SC. Insulin-like Growth Factor-1, Bone Mineral Density, and Fracture: A Mendelian Randomization Study. J Clin Endocrinol Metab 2021; 106:e1552-e1558. [PMID: 33462619 PMCID: PMC7993594 DOI: 10.1210/clinem/dgaa963] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Indexed: 12/14/2022]
Abstract
CONTEXT The associations of circulating insulin-like growth factor-1 (IGF-1) levels with bone mineral density and fracture risk are inconclusive in observational studies. OBJECTIVE We conducted a mendelian randomization study to assess the associations of serum IGF-1 levels with estimated bone mineral density (eBMD) and fracture. METHODS Genetic instruments for IGF-1 were selected at the genome-wide significance level (P < 5 × 10-8) from a genome-wide association study including 358 072 individuals of European ancestry. Summary-level data for eBMD (426 824 individuals) and fracture (53 184 fracture cases and 373 611 noncases) were obtained from the UK Biobank study. Univariable and multivariable mendelian randomization analyses methods were used to estimate the associations of IGF-1 with eBMD and fracture. The main outcome measure included the change of eBMD and odds ratio of fracture per genetically predicted 1-SD increase of serum IGF-1 levels. RESULTS For 1-SD increase in IGF-1, the change of eBMD levels was 0.04 g/cm2 (95% CI, 0.01-0.07; P = .011) and the odds ratio of fracture was 0.94 (95% CI, 0.91-0.98; P = .003). The associations persisted with similar magnitude after adjustment for height. The association was consistent for fracture but not for eBMD after excluding genetic instruments that might directly influence these outcomes. The association between IGF-1 and fracture was somewhat attenuated after adjustment for eBMD (odds ratio 0.96; 95% CI, 0.92-0.99; P = .012). CONCLUSION The present study supports a role for IGF-1 in preventing fracture, possibly and partly mediated by greater bone mineral density.
Collapse
Affiliation(s)
- Shuai Yuan
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Zi-Hao Wan
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Shi-Le Cheng
- Department of Orthopedic Surgery, Renmin Hospital, Wuhan University, Wuhan, China
| | - Karl Michaëlsson
- Unit of Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Susanna C Larsson
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Unit of Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
- Correspondence: Susanna C. Larsson, PhD, Department of Surgical Sciences, Uppsala University, Dag Hammarskjölds väg 14 B, 75185 Uppsala, Sweden.
| |
Collapse
|
9
|
Merlotti D, Cosso R, Eller-Vainicher C, Vescini F, Chiodini I, Gennari L, Falchetti A. Energy Metabolism and Ketogenic Diets: What about the Skeletal Health? A Narrative Review and a Prospective Vision for Planning Clinical Trials on this Issue. Int J Mol Sci 2021; 22:ijms22010435. [PMID: 33406758 PMCID: PMC7796307 DOI: 10.3390/ijms22010435] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/27/2020] [Accepted: 12/30/2020] [Indexed: 12/15/2022] Open
Abstract
The existence of a common mesenchymal cell progenitor shared by bone, skeletal muscle, and adipocytes cell progenitors, makes the role of the skeleton in energy metabolism no longer surprising. Thus, bone fragility could also be seen as a consequence of a “poor” quality in nutrition. Ketogenic diet was originally proven to be effective in epilepsy, and long-term follow-up studies on epileptic children undergoing a ketogenic diet reported an increased incidence of bone fractures and decreased bone mineral density. However, the causes of such negative impacts on bone health have to be better defined. In these subjects, the concomitant use of antiepileptic drugs and the reduced mobilization may partly explain the negative effects on bone health, but little is known about the effects of diet itself, and/or generic alterations in vitamin D and/or impaired growth factor production. Despite these remarks, clinical studies were adequately designed to investigate bone health are scarce and bone health related aspects are not included among the various metabolic pathologies positively influenced by ketogenic diets. Here, we provide not only a narrative review on this issue, but also practical advice to design and implement clinical studies on ketogenic nutritional regimens and bone health outcomes. Perspectives on ketogenic regimens, microbiota, microRNAs, and bone health are also included.
Collapse
Affiliation(s)
- Daniela Merlotti
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (D.M.); (L.G.)
| | - Roberta Cosso
- Istituto Auxologico Italiano “Scientific Institute for Hospitalisation and Care”, 20100 Milano, Italy; (R.C.); (I.C.)
| | - Cristina Eller-Vainicher
- Unit of Endocrinology, Fondazione IRCCS Cà Granda-Ospedale Maggiore Policlinico Milano, 20122 Milano, Italy;
| | - Fabio Vescini
- Endocrinology and Metabolism Unit, University-Hospital S. Maria della Misericordia of Udine, 33100 Udine, Italy;
| | - Iacopo Chiodini
- Istituto Auxologico Italiano “Scientific Institute for Hospitalisation and Care”, 20100 Milano, Italy; (R.C.); (I.C.)
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20122 Milano, Italy
| | - Luigi Gennari
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (D.M.); (L.G.)
| | - Alberto Falchetti
- Istituto Auxologico Italiano “Scientific Institute for Hospitalisation and Care”, 20100 Milano, Italy; (R.C.); (I.C.)
- Correspondence:
| |
Collapse
|
10
|
Endo Y, Nourmahnad A, Sinha I. Optimizing Skeletal Muscle Anabolic Response to Resistance Training in Aging. Front Physiol 2020; 11:874. [PMID: 32792984 PMCID: PMC7390896 DOI: 10.3389/fphys.2020.00874] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/29/2020] [Indexed: 12/13/2022] Open
Abstract
Loss of muscle mass and strength with aging, also termed sarcopenia, results in a loss of mobility and independence. Exercise, particularly resistance training, has proven to be beneficial in counteracting the aging-associated loss of skeletal muscle mass and function. However, the anabolic response to exercise in old age is not as robust, with blunted improvements in muscle size, strength, and function in comparison to younger individuals. This review provides an overview of several physiological changes which may contribute to age-related loss of muscle mass and decreased anabolism in response to resistance training in the elderly. Additionally, the following supplemental therapies with potential to synergize with resistance training to increase muscle mass are discussed: nutrition, creatine, anti-inflammatory drugs, testosterone, and growth hormone (GH). Although these interventions hold some promise, further research is necessary to optimize the response to exercise in elderly patients.
Collapse
Affiliation(s)
- Yori Endo
- Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Atousa Nourmahnad
- Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Indranil Sinha
- Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,Harvard Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Cambridge, MA, United States
| |
Collapse
|
11
|
Zhang WB, Aleksic S, Gao T, Weiss EF, Demetriou E, Verghese J, Holtzer R, Barzilai N, Milman S. Insulin-like Growth Factor-1 and IGF Binding Proteins Predict All-Cause Mortality and Morbidity in Older Adults. Cells 2020; 9:cells9061368. [PMID: 32492897 PMCID: PMC7349399 DOI: 10.3390/cells9061368] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 05/27/2020] [Accepted: 05/28/2020] [Indexed: 01/02/2023] Open
Abstract
While the growth hormone/insulin-like growth factor-1 (GH/IGF-1) pathway plays essential roles in growth and development, diminished signaling via this pathway in model organisms extends lifespan and health-span. In humans, circulating IGF-1 and IGF-binding proteins 3 and 1 (IGFBP-3 and 1), surrogate measures of GH/IGF-1 system activity, have not been consistently associated with morbidity and mortality. In a prospective cohort of independently-living older adults (n = 840, mean age 76.1 ± 6.8 years, 54.5% female, median follow-up 6.9 years), we evaluated the age- and sex-adjusted hazards for all-cause mortality and incident age-related diseases, including cardiovascular disease, diabetes, cancer, and multiple-domain cognitive impairment (MDCI), as predicted by baseline total serum IGF-1, IGF-1/IGFBP-3 molar ratio, IGFBP-3, and IGFBP-1 levels. All-cause mortality was positively associated with IGF-1/IGFBP-3 molar ratio (HR 1.28, 95% CI 1.05–1.57) and negatively with IGFBP-3 (HR 0.82, 95% CI 0.680–0.998). High serum IGF-1 predicted greater risk for MDCI (HR 1.56, 95% CI 1.08–2.26) and composite incident morbidity (HR 1.242, 95% CI 1.004–1.538), whereas high IGFBP-1 predicted lower risk for diabetes (HR 0.50, 95% CI 0.29–0.88). In conclusion, higher IGF-1 levels and bioavailability predicted mortality and morbidity risk, supporting the hypothesis that diminished GH/IGF-1 signaling may contribute to human longevity and health-span.
Collapse
Affiliation(s)
- William B. Zhang
- Department of Medicine, Division of Endocrinology, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (W.B.Z.); (S.A.); (T.G.); (N.B.)
| | - Sandra Aleksic
- Department of Medicine, Division of Endocrinology, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (W.B.Z.); (S.A.); (T.G.); (N.B.)
| | - Tina Gao
- Department of Medicine, Division of Endocrinology, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (W.B.Z.); (S.A.); (T.G.); (N.B.)
| | - Erica F. Weiss
- Department of Neurology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.F.W.); (J.V.); (R.H.)
| | - Eleni Demetriou
- Ferkauf Graduate School of Psychology, Yeshiva University, New York, NY 10033, USA;
| | - Joe Verghese
- Department of Neurology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.F.W.); (J.V.); (R.H.)
- Department of Medicine, Division of Geriatrics, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Roee Holtzer
- Department of Neurology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.F.W.); (J.V.); (R.H.)
- Ferkauf Graduate School of Psychology, Yeshiva University, New York, NY 10033, USA;
| | - Nir Barzilai
- Department of Medicine, Division of Endocrinology, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (W.B.Z.); (S.A.); (T.G.); (N.B.)
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Sofiya Milman
- Department of Medicine, Division of Endocrinology, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (W.B.Z.); (S.A.); (T.G.); (N.B.)
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Correspondence:
| |
Collapse
|
12
|
Kim B, Elzinga SE, Henn RE, McGinley LM, Feldman EL. The effects of insulin and insulin-like growth factor I on amyloid precursor protein phosphorylation in in vitro and in vivo models of Alzheimer's disease. Neurobiol Dis 2019; 132:104541. [PMID: 31349033 DOI: 10.1016/j.nbd.2019.104541] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 07/15/2019] [Accepted: 07/22/2019] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is a growing problem worldwide, and there are currently no effective treatments for this devastating disease. The neurotrophic growth factors insulin and insulin-like growth factor-I (IGF-I) are currently being investigated as potential therapeutic approaches for AD in preclinical and clinical studies. However, given that the metabolic syndrome (MetS) and diabetes are risk factors for AD, it is unknown how associated insulin resistance (IR) in the brain may impact the effectiveness of these therapies for AD. In this report, we therefore investigated the mechanisms underlying the effects of insulin and IGF-I on AD-associated pathology in the context of IR, with particular emphasis on phosphorylation of amyloid precursor protein (APP), a key step in promoting amyloid plaque formation in AD. Both insulin and IGF-I decreased APP phosphorylation in cultured primary cortical neurons, supporting their therapeutic use in AD. Induction of IR blocked the beneficial effect of insulin and reduced the effect of IGF-I on APP dephosphorylation. These effects were mediated by the phosphatidylinositol 3-kinase (PI3-K)/protein kinase B (Akt) pathway, as inhibition of this pathway during IR restored the effect of IGF-I on APP dephosphorylation. Finally, we explored the translational relevance of these results in vivo by demonstrating that high fat diet fed mice, a robust model of IR and MetS, exhibited the expected increased brain APP phosphorylation. Overall, these data suggest that the beneficial therapeutic effect of insulin and IGF-I on APP phosphorylation is negatively impacted by IR, and suggest that insulin and IGF-I alone may not be appropriate therapies for AD patients with IR, MetS, or diabetes.
Collapse
Affiliation(s)
- Bhumsoo Kim
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, United States of America
| | - Sarah E Elzinga
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, United States of America
| | - Rosemary E Henn
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, United States of America
| | - Lisa M McGinley
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, United States of America
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, United States of America.
| |
Collapse
|
13
|
Lewitt MS, Boyd GW. The Role of Insulin-Like Growth Factors and Insulin-Like Growth Factor-Binding Proteins in the Nervous System. BIOCHEMISTRY INSIGHTS 2019; 12:1178626419842176. [PMID: 31024217 PMCID: PMC6472167 DOI: 10.1177/1178626419842176] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 03/15/2019] [Indexed: 01/23/2023]
Abstract
The insulin-like growth factors (IGF-I and IGF-II) and their receptors are widely expressed in nervous tissue from early embryonic life. They also cross the blood brain barriers by active transport, and their regulation as endocrine factors therefore differs from other tissues. In brain, IGFs have paracrine and autocrine actions that are modulated by IGF-binding proteins and interact with other growth factor signalling pathways. The IGF system has roles in nervous system development and maintenance. There is substantial evidence for a specific role for this system in some neurodegenerative diseases, and neuroprotective actions make this system an attractive target for new therapeutic approaches. In developing new therapies, interaction with IGF-binding proteins and other growth factor signalling pathways should be considered. This evidence is reviewed, gaps in knowledge are highlighted, and recommendations are made for future research.
Collapse
Affiliation(s)
- Moira S Lewitt
- School of Health & Life Sciences, University of the West of Scotland, Paisley, UK
| | - Gary W Boyd
- School of Health & Life Sciences, University of the West of Scotland, Paisley, UK
| |
Collapse
|
14
|
Gubbi S, Quipildor GF, Barzilai N, Huffman DM, Milman S. 40 YEARS of IGF1: IGF1: the Jekyll and Hyde of the aging brain. J Mol Endocrinol 2018; 61:T171-T185. [PMID: 29739805 PMCID: PMC5988994 DOI: 10.1530/jme-18-0093] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 05/08/2018] [Indexed: 12/30/2022]
Abstract
The insulin-like growth factor 1 (IGF1) signaling pathway has emerged as a major regulator of the aging process, from rodents to humans. However, given the pleiotropic actions of IGF1, its role in the aging brain remains complex and controversial. While IGF1 is clearly essential for normal development of the central nervous system, conflicting evidence has emerged from preclinical and human studies regarding its relationship to cognitive function, as well as cerebrovascular and neurodegenerative disorders. This review delves into the current state of the evidence examining the role of IGF1 in the aging brain, encompassing preclinical and clinical studies. A broad examination of the data indicates that IGF1 may indeed play opposing roles in the aging brain, depending on the underlying pathology and context. Some evidence suggests that in the setting of neurodegenerative diseases that manifest with abnormal protein deposition in the brain, such as Alzheimer's disease, reducing IGF1 signaling may serve a protective role by slowing disease progression and augmenting clearance of pathologic proteins to maintain cellular homeostasis. In contrast, inducing IGF1 deficiency has also been implicated in dysregulated function of cognition and the neurovascular system, suggesting that some IGF1 signaling may be necessary for normal brain function. Furthermore, states of acute neuronal injury, which necessitate growth, repair and survival signals to persevere, typically demonstrate salutary effects of IGF1 in that context. Appreciating the dual, at times opposing 'Dr Jekyll' and 'Mr Hyde' characteristics of IGF1 in the aging brain, will bring us closer to understanding its impact and devising more targeted IGF1-related interventions.
Collapse
Affiliation(s)
- Sriram Gubbi
- Institute for Aging ResearchAlbert Einstein College of Medicine, Bronx, New York, USA
- Department of Internal MedicineJacobi Medical Center, Bronx, New York, USA
| | - Gabriela Farias Quipildor
- Institute for Aging ResearchAlbert Einstein College of Medicine, Bronx, New York, USA
- Department of Molecular PharmacologyAlbert Einstein College of Medicine, Bronx, New York, USA
- Division of EndocrinologyDepartment of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Nir Barzilai
- Institute for Aging ResearchAlbert Einstein College of Medicine, Bronx, New York, USA
- Division of EndocrinologyDepartment of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
- Division of GeriatricsDepartment of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of GeneticsAlbert Einstein College of Medicine, Bronx, New York, USA
| | - Derek M Huffman
- Institute for Aging ResearchAlbert Einstein College of Medicine, Bronx, New York, USA
- Department of Molecular PharmacologyAlbert Einstein College of Medicine, Bronx, New York, USA
- Division of EndocrinologyDepartment of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Sofiya Milman
- Institute for Aging ResearchAlbert Einstein College of Medicine, Bronx, New York, USA
- Division of EndocrinologyDepartment of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
- Division of GeriatricsDepartment of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
15
|
Ajdžanovic VZ, Trifunovic S, Miljic D, Šošic-Jurjevic B, Filipovic B, Miler M, Ristic N, Manojlovic-Stojanoski M, Miloševic V. Somatopause, weaknesses of the therapeutic approaches and the cautious optimism based on experimental ageing studies with soy isoflavones. EXCLI JOURNAL 2018; 17:279-301. [PMID: 29743865 PMCID: PMC5938552 DOI: 10.17179/excli2017-956] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 03/10/2018] [Indexed: 12/15/2022]
Abstract
The pathological phenomenon of somatopause, noticeable in hypogonadal ageing subjects, is based on the growth hormone (GH) production and secretion decrease along with the fall in GH binding protein and insulin-like growth factor 1 (IGF-1) levels, causing different musculoskeletal, metabolic and mental issues. From the perspective of safety and efficacy, GH treatment is considered to be highly controversial, while some other therapeutic approaches (application of IGF-1, GH secretagogues, gonadal steroids, cholinesterase-inhibitors or various combinations) exhibit more or less pronounced weaknesses in this respect. Soy isoflavones, phytochemicals that have already demonstrated the health benefits in treated elderly, at least experimentally reveal their potential for the somatopausal symptoms remediation. Namely, genistein enhanced GHRH-stimulated cAMP accumulation and GH release in rat anterior pituitary cells; refreshed and stimulated the somatotropic system (hypothalamic nuclei and pituitary GH cells) function in a rat model of the mild andropause, and stimulated the GH output in ovariectomized ewes as well as the amplitude of GH pulses in the rams. Daidzein, on the other hand, increased body mass, trabecular bone mass and decreased bone turnover in the animal model of severe andropause, while both isoflavones demonstrated blood cholesterol-lowering effect in the same model. These data, which necessarily need to be preclinically and clinically filtered, hint some cautious optimism and call for further innovative designing of balanced soy isoflavone-based therapeutics.
Collapse
Affiliation(s)
- Vladimir Z Ajdžanovic
- Department of Cytology, Institute for Biological Research "Siniša Stankovic", University of Belgrade, Belgrade, Serbia
| | - Svetlana Trifunovic
- Department of Cytology, Institute for Biological Research "Siniša Stankovic", University of Belgrade, Belgrade, Serbia
| | - Dragana Miljic
- Clinic for Endocrinology, Diabetes and Diseases of Metabolism, Clinical Center of Serbia, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Branka Šošic-Jurjevic
- Department of Cytology, Institute for Biological Research "Siniša Stankovic", University of Belgrade, Belgrade, Serbia
| | - Branko Filipovic
- Department of Cytology, Institute for Biological Research "Siniša Stankovic", University of Belgrade, Belgrade, Serbia
| | - Marko Miler
- Department of Cytology, Institute for Biological Research "Siniša Stankovic", University of Belgrade, Belgrade, Serbia
| | - Nataša Ristic
- Department of Cytology, Institute for Biological Research "Siniša Stankovic", University of Belgrade, Belgrade, Serbia
| | - Milica Manojlovic-Stojanoski
- Department of Cytology, Institute for Biological Research "Siniša Stankovic", University of Belgrade, Belgrade, Serbia
| | - Verica Miloševic
- Department of Cytology, Institute for Biological Research "Siniša Stankovic", University of Belgrade, Belgrade, Serbia
| |
Collapse
|
16
|
Anderson LJ, Tamayose JM, Garcia JM. Use of growth hormone, IGF-I, and insulin for anabolic purpose: Pharmacological basis, methods of detection, and adverse effects. Mol Cell Endocrinol 2018; 464:65-74. [PMID: 28606865 PMCID: PMC5723243 DOI: 10.1016/j.mce.2017.06.010] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 06/02/2017] [Accepted: 06/08/2017] [Indexed: 12/24/2022]
Abstract
Hormones with anabolic properties such as growth hormone (GH), insulin-like growth factor-1 (IGF-I), and insulin are commonly abused among professional and recreational athletes to enhance physical ability. Performance enhancing drugs (PEDs) such as these are also commonly used by recreational athletes to improve body aesthetics. The perception of increased muscle mass due to supraphysiologic hormone supplementation, or doping, is widespread among PED users despite a paucity of evidence-based data in humans. Even still, athletes will continue to abuse PEDs in hopes of replicating anecdotal results. It is important to educate the general public and potential treating physicians of the risks of PED use, including the dangers of polypharmacy and substance dependence. It will also be important for the research community to address the common challenges associated with studying PED use such as the ethical considerations of PED administration, the general reticence of the PED-using community to volunteer information, and the constant need to improve or create new detection methods as athletes continually attempt to circumvent current methods. This review highlights the anabolic mechanisms and suggestive data implicating GH, IGF-I, and insulin for use as PEDs, the specific detection methods with cutoff ranges that may be utilized to diagnose abuse of each substance, and their respective side effects.
Collapse
Affiliation(s)
- Lindsey J Anderson
- Geriatric Research, Education and Clinical Center (GRECC), VA Puget Sound Health Care System, Seattle, WA, United States
| | - Jamie M Tamayose
- Geriatric Research, Education and Clinical Center (GRECC), VA Puget Sound Health Care System, Seattle, WA, United States
| | - Jose M Garcia
- Geriatric Research, Education and Clinical Center (GRECC), VA Puget Sound Health Care System, Seattle, WA, United States; Department of Medicine, Division of Gerontology & Geriatric Medicine, University of Washington School of Medicine, Seattle, WA, United States.
| |
Collapse
|
17
|
Perice L, Barzilai N, Verghese J, Weiss EF, Holtzer R, Cohen P, Milman S. Lower circulating insulin-like growth factor-I is associated with better cognition in females with exceptional longevity without compromise to muscle mass and function. Aging (Albany NY) 2017; 8:2414-2424. [PMID: 27744417 PMCID: PMC5115897 DOI: 10.18632/aging.101063] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Accepted: 09/28/2016] [Indexed: 12/21/2022]
Abstract
Mutations that reduce somatotropic signaling result in improved lifespan and health-span in model organisms and humans. However, whether reduced circulating insulin-like growth factor-I (IGF-I) level is detrimental to cognitive and muscle function in older adults remains understudied. A cross-sectional analysis was performed in Ashkenazi Jews with exceptional longevity (age ≥95 years). Cognition was assessed using the Mini-Mental State Examination and muscle function with the chair rise test, grip-strength, and gait speed. Muscle mass was estimated using the skeletal muscle index. Serum IGF-I was measured with liquid chromatography mass spectrometry. In gender stratified age-adjusted logistic regression analysis, females with IGF-I levels in the first tertile had lower odds of being cognitively impaired compared to females with IGF-I levels within the upper two tertiles, OR (95% CI) 0.39 (0.19-0.82). The result remained significant after adjustment for multiple parameters. No significant association was identified in males between IGF-I and cognition. No relationship was found between IGF-I tertiles and muscle function and muscle mass in females or males. Lower circulating IGF-I is associated with better cognitive function in females with exceptional longevity, with no detriment to skeletal muscle mass and function.
Collapse
Affiliation(s)
- Leland Perice
- Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Nir Barzilai
- Department of Medicine, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA.,Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Joe Verghese
- Department of Medicine, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA.,Department of Neurology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Erica F Weiss
- Department of Neurology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Roee Holtzer
- Department of Neurology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.,Ferkauf Graduate School of Psychology, Yeshiva University, Bronx, NY 10461
| | - Pinchas Cohen
- Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Sofiya Milman
- Department of Medicine, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
18
|
Heer M, Baecker N, Frings-Meuthen P, Graf S, Zwart SR, Biolo G, Smith SM. Effects of high-protein intake on bone turnover in long-term bed rest in women. Appl Physiol Nutr Metab 2017; 42:537-546. [DOI: 10.1139/apnm-2016-0292] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Bed rest (BR) causes bone loss, even in otherwise healthy subjects. Several studies suggest that ambulatory subjects may benefit from high-protein intake to stimulate protein synthesis and to maintain muscle mass. However, increasing protein intake above the recommended daily intake without adequate calcium and potassium intake may increase bone resorption. We hypothesized that a regimen of high-protein intake (HiPROT), applied in an isocaloric manner during BR, with calcium and potassium intake meeting recommended values, would prevent any effect of BR on bone turnover. After a 20-day ambulatory adaptation to a controlled environment, 16 women participated in a 60-day, 6° head-down-tilt (HDT) BR and were assigned randomly to 1 of 2 groups. Control (CON) subjects (n = 8) received 1 g/(kg body mass·day)−1 dietary protein. HiPROT subjects (n = 8) received 1.45 g protein/(kg body mass·day)−1 plus an additional 0.72 g branched-chain amino acids per day during BR. All subjects received an individually tailored diet (before HDTBR: 1888 ± 98 kcal/day; during HDTBR: 1604 ± 125 kcal/day; after HDTBR: 1900 ± 262 kcal/day), with the CON group’s diet being higher in fat and carbohydrate intake. High-protein intake exacerbated the BR-induced increase in bone resorption marker C-telopeptide (>30%) (p < 0.001) by the end of BR. Bone formation markers were unaffected by BR and high-protein intake. We conclude that high-protein intake in BR might increase bone loss. Further long-duration studies are mandatory to show how the positive effect of protein on muscle mass can be maintained without the risk of reducing bone mineral density.
Collapse
Affiliation(s)
- Martina Heer
- Department of Nutrition and Food Sciences, Nutritional Physiology, University of Bonn, 53115 Bonn, Germany
| | - Natalie Baecker
- Department of Nutrition and Food Sciences, Nutritional Physiology, University of Bonn, 53115 Bonn, Germany
| | - Petra Frings-Meuthen
- German Aerospace Center (DLR), Institute of Aerospace Medicine, 51147 Cologne, Germany
| | - Sonja Graf
- Department of Nutrition and Food Sciences, Nutritional Physiology, University of Bonn, 53115 Bonn, Germany
| | - Sara R. Zwart
- University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Gianni Biolo
- Department of Clinical, Technological and Morphological Sciences, Division of Internal Medicine, University of Trieste, 34127 Trieste, Italy
| | - Scott M. Smith
- Human Health and Performance Directorate, NASA Lyndon B. Johnson Space Center, Houston, TX 77058, USA
| |
Collapse
|
19
|
Wrigley S, Arafa D, Tropea D. Insulin-Like Growth Factor 1: At the Crossroads of Brain Development and Aging. Front Cell Neurosci 2017; 11:14. [PMID: 28203146 PMCID: PMC5285390 DOI: 10.3389/fncel.2017.00014] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 01/16/2017] [Indexed: 12/15/2022] Open
Abstract
Insulin-like growth factor 1 (IGF1) is a polypeptide hormone structurally similar to insulin. It is central to the somatotropic axis, acting downstream of growth hormone (GH). It activates both the mitogen-activated protein (MAP) kinase and PI3K signaling pathways, acting in almost every tissue in the body to promote tissue growth and maturation through upregulation of anabolic processes. Overall GH and IGF1 signaling falls with age, suggesting that it is this reduced IGF1 activity that leads to age-related changes in organisms. However, mutations that reduce IGF1-signaling activity can dramatically extend the lifespan of organisms. Therefore, the role of IGF1 in the overall aging process is unclear. This review article will focus on the role of IGF1 in brain development and aging. The evidence points towards a role for IGF1 in neurodevelopment both prenatally and in the early post-natal period, and in plasticity and remodeling throughout life. This review article will then discuss the hallmarks of aging and cognitive decline associated with falls in IGF1 levels towards the end of life. Finally, the role of IGF1 will be discussed within the context of both neuropsychiatric disorders caused by impaired development of the nervous system, and neurodegenerative disorders associated with aging. IGF1 and its derivatives are shown to improve the symptoms of certain neuropsychiatric disorders caused by deranged neurodevelopment and these effects have been correlated with changes in the underlying biology in both in vitro and in vivo studies. On the other hand, studies looking at IGF1 in neurodegenerative diseases have been conflicting, supporting both a role for increased and decreased IGF1 signaling in the underlying pathogenesis of these diseases.
Collapse
Affiliation(s)
- Sarah Wrigley
- School of Medicine, Trinity College Dublin Dublin, Ireland
| | - Donia Arafa
- School of Medicine, Trinity College Dublin Dublin, Ireland
| | - Daniela Tropea
- Neuropsychiatric Genetics, Trinity Translational Medicine Institute St. James HospitalDublin, Ireland; Institute of Neuroscience, Trinity College DublinDublin, Ireland
| |
Collapse
|
20
|
Wyss-Coray T. Ageing, neurodegeneration and brain rejuvenation. Nature 2016; 539:180-186. [PMID: 27830812 PMCID: PMC5172605 DOI: 10.1038/nature20411] [Citation(s) in RCA: 680] [Impact Index Per Article: 85.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 09/02/2016] [Indexed: 02/08/2023]
Abstract
Although systemic diseases take the biggest toll on human health and well-being, increasingly, a failing brain is the arbiter of a death preceded by a gradual loss of the essence of being. Ageing, which is fundamental to neurodegeneration and dementia, affects every organ in the body and seems to be encoded partly in a blood-based signature. Indeed, factors in the circulation have been shown to modulate ageing and to rejuvenate numerous organs, including the brain. The discovery of such factors, the identification of their origins and a deeper understanding of their functions is ushering in a new era in ageing and dementia research.
Collapse
Affiliation(s)
- Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Palo Alto, California 94304, USA
- Center for Tissue Regeneration, Repair and Restoration, VA Palo Alto Health Care System, Palo Alto, California 94304, USA
| |
Collapse
|
21
|
Lindsey RC, Mohan S. Skeletal effects of growth hormone and insulin-like growth factor-I therapy. Mol Cell Endocrinol 2016; 432:44-55. [PMID: 26408965 PMCID: PMC4808510 DOI: 10.1016/j.mce.2015.09.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 09/21/2015] [Accepted: 09/22/2015] [Indexed: 10/23/2022]
Abstract
The growth hormone/insulin-like growth factor (GH/IGF) axis is critically important for the regulation of bone formation, and deficiencies in this system have been shown to contribute to the development of osteoporosis and other diseases of low bone mass. The GH/IGF axis is regulated by a complex set of hormonal and local factors which can act to regulate this system at the level of the ligands, receptors, IGF binding proteins (IGFBPs), or IGFBP proteases. A combination of in vitro studies, transgenic animal models, and clinical human investigations has provided ample evidence of the importance of the endocrine and local actions of both GH and IGF-I, the two major components of the GH/IGF axis, in skeletal growth and maintenance. GH- and IGF-based therapies provide a useful avenue of approach for the prevention and treatment of diseases such as osteoporosis.
Collapse
Affiliation(s)
- Richard C Lindsey
- Musculoskeletal Disease Center, Loma Linda VA Healthcare System, Loma Linda, CA 92357, USA; Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; Department of Biochemistry, Loma Linda University, Loma Linda, CA 92354, USA
| | - Subburaman Mohan
- Musculoskeletal Disease Center, Loma Linda VA Healthcare System, Loma Linda, CA 92357, USA; Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; Department of Biochemistry, Loma Linda University, Loma Linda, CA 92354, USA.
| |
Collapse
|
22
|
Milman S, Huffman DM, Barzilai N. The Somatotropic Axis in Human Aging: Framework for the Current State of Knowledge and Future Research. Cell Metab 2016; 23:980-989. [PMID: 27304500 PMCID: PMC4919980 DOI: 10.1016/j.cmet.2016.05.014] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 05/24/2016] [Accepted: 05/26/2016] [Indexed: 12/19/2022]
Abstract
Mutations resulting in reduced signaling of the growth hormone/insulin-like growth factor-1 (GH/IGF-1) axis are associated with increased life- and healthspan across model organisms. Similar findings have been noted in human cohorts with functional mutations in the somatotropic axis, suggesting that this pathway may also be relevant to human aging and protection from age-related diseases. While epidemiological data indicate that low circulating IGF-1 level may protect aging populations from cancer, results remain inconclusive regarding most other diseases. We propose that studies in humans and animals need to consider differences in sex, pathway function, organs, and time-specific effects of GH/IGF-1 signaling in order to better define the role of the somatotropic axis in aging. Agents that modulate signaling of the GH/IGF-1 pathway are available for human use, but before they can be implemented in clinical studies that target aging and age-related diseases, researchers need to address the challenges discussed in this Review.
Collapse
Affiliation(s)
- Sofiya Milman
- Department of Medicine, Division of Endocrinology, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Medicine, Division of Geriatrics, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Derek M Huffman
- Department of Medicine, Division of Endocrinology, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Nir Barzilai
- Department of Medicine, Division of Endocrinology, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Medicine, Division of Geriatrics, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
23
|
Philippou A, Barton ER. Optimizing IGF-I for skeletal muscle therapeutics. Growth Horm IGF Res 2014; 24:157-163. [PMID: 25002025 PMCID: PMC4665094 DOI: 10.1016/j.ghir.2014.06.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 06/09/2014] [Indexed: 12/13/2022]
Abstract
It is virtually undisputed that IGF-I promotes cell growth and survival. However, the presence of several IGF-I isoforms, vast numbers of intracellular signaling components, and multiple receptors results in a complex and highly regulated system by which IGF-I actions are mediated. IGF-I has long been recognized as one of the critical factors for coordinating muscle growth, enhancing muscle repair, and increasing muscle mass and strength. How to optimize this panoply of pathways to drive anabolic processes in muscle as opposed to aberrant growth in other tissues is an area that deserves focus. This review will address how advances in the bioavailability, potency, and tissue response of IGF-I can provide new potential directions for skeletal muscle therapeutics.
Collapse
Affiliation(s)
- Anastassios Philippou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Elisabeth R Barton
- Department of Anatomy and Cell Biology, School of Dental Medicine, and Pennsylvania Muscle Institute, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
24
|
Abstract
Frailty is a common and important geriatric syndrome characterized by age-associated declines in physiologic reserve and function across multiorgan systems, leading to increased vulnerability for adverse health outcomes. Two major frailty models have been described in the literature. The frailty phenotype defines frailty as a distinct clinical syndrome meeting three or more of five phenotypic criteria: weakness, slowness, low level of physical activity, self-reported exhaustion, and unintentional weight loss. The frailty index defines frailty as cumulative deficits identified in a comprehensive geriatric assessment. Significant progress has recently been made in understanding the pathogenesis of frailty. Chronic inflammation is likely a key pathophysiologic process that contributes to the frailty syndrome directly and indirectly through other intermediate physiologic systems, such as the musculoskeletal, endocrine, and hematologic systems. The complex multifactorial etiologies of frailty also include obesity and specific diseases. Major clinical applications include risk assessment and stratification. This can be applied to the elderly population in the community and in a variety of care settings. Frailty may also be useful for risk assessment in surgical patients and those with cardiovascular diseases, cancer, or human immunodeficiency virus infection, as well as for assessment of vaccine effectiveness in older adults. Currently, exercise and comprehensive geriatric interdisciplinary assessment and treatment are key interventions for frailty. As understanding of the biologic basis and complexity of frailty further improves, more effective and targeted interventional strategies and innovative geriatric-care models will likely be developed.
Collapse
Affiliation(s)
- Xujiao Chen
- Department of Geriatrics, Zhejiang Hospital, Hangzhou, People's Republic of China
| | - Genxiang Mao
- Department of Geriatrics, Zhejiang Hospital, Hangzhou, People's Republic of China
| | - Sean X Leng
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
25
|
Locatelli V, Bianchi VE. Effect of GH/IGF-1 on Bone Metabolism and Osteoporsosis. Int J Endocrinol 2014; 2014:235060. [PMID: 25147565 PMCID: PMC4132406 DOI: 10.1155/2014/235060] [Citation(s) in RCA: 179] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 06/17/2014] [Accepted: 06/18/2014] [Indexed: 01/25/2023] Open
Abstract
Background. Growth hormone (GH) and insulin-like growth factor (IGF-1) are fundamental in skeletal growth during puberty and bone health throughout life. GH increases tissue formation by acting directly and indirectly on target cells; IGF-1 is a critical mediator of bone growth. Clinical studies reporting the use of GH and IGF-1 in osteoporosis and fracture healing are outlined. Methods. A Pubmed search revealed 39 clinical studies reporting the effects of GH and IGF-1 administration on bone metabolism in osteopenic and osteoporotic human subjects and on bone healing in operated patients with normal GH secretion. Eighteen clinical studies considered the effect with GH treatment, fourteen studies reported the clinical effects with IGF-1 administration, and seven related to the GH/IGF-1 effect on bone healing. Results. Both GH and IGF-1 administration significantly increased bone resorption and bone formation in the most studies. GH/IGF-1 administration in patients with hip or tibial fractures resulted in increased bone healing, rapid clinical improvements. Some conflicting results were evidenced. Conclusions. GH and IGF-1 therapy has a significant anabolic effect. GH administration for the treatment of osteoporosis and bone fractures may greatly improve clinical outcome. GH interacts with sex steroids in the anabolic process. GH resistance process is considered.
Collapse
Affiliation(s)
- Vittorio Locatelli
- Department of Health Sciences, School of Medicine, University of Milano Bicocca, Milan, Italy
| | - Vittorio E. Bianchi
- Endocrinology Department, Area Vasta N. 1, Cagli, Italy
- *Vittorio E. Bianchi:
| |
Collapse
|
26
|
Crane JL, Cao X. Function of matrix IGF-1 in coupling bone resorption and formation. J Mol Med (Berl) 2013; 92:107-15. [PMID: 24068256 DOI: 10.1007/s00109-013-1084-3] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Revised: 08/16/2013] [Accepted: 09/01/2013] [Indexed: 12/13/2022]
Abstract
Balancing bone resorption and formation is the quintessential component for the prevention of osteoporosis. Signals that determine the recruitment, replication, differentiation, function, and apoptosis of osteoblasts and osteoclasts direct bone remodeling and determine whether bone tissue is gained, lost, or balanced. Therefore, understanding the signaling pathways involved in the coupling process will help develop further targets for osteoporosis therapy, by blocking bone resorption or enhancing bone formation in a space- and time-dependent manner. Insulin-like growth factor type 1 (IGF-1) has long been known to play a role in bone strength. It is one of the most abundant substances in the bone matrix, circulates systemically and is secreted locally, and has a direct relationship with bone mineral density. Recent data has helped further our understanding of the direct role of IGF-1 signaling in coupling bone remodeling which will be discussed in this review. The bone marrow microenvironment plays a critical role in the fate of mesenchymal stem cells and hematopoietic stem cells and thus how IGF-1 interacts with other factors in the microenvironment are equally important. While previous clinical trials with IGF-1 administration have been unsuccessful at enhancing bone formation, advances in basic science studies have provided insight into further mechanisms that should be considered for future trials. Additional basic science studies dissecting the regulation and the function of matrix IGF-1 in modeling and remodeling will continue to provide further insight for future directions for anabolic therapies for osteoporosis.
Collapse
Affiliation(s)
- Janet L Crane
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Ross Building, Room 229, 720 Rutland Ave, Baltimore, MD, 21205, USA,
| | | |
Collapse
|
27
|
Friedman SD, Baker LD, Borson S, Jensen JE, Barsness SM, Craft S, Merriam GR, Otto RK, Novotny EJ, Vitiello MV. Growth hormone-releasing hormone effects on brain γ-aminobutyric acid levels in mild cognitive impairment and healthy aging. JAMA Neurol 2013; 70:883-90. [PMID: 23689947 DOI: 10.1001/jamaneurol.2013.1425] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
IMPORTANCE Growth hormone-releasing hormone (GHRH) has been previously shown to have cognition-enhancing effects. The role of neurotransmitter changes, measured by proton magnetic resonance spectroscopy, may inform the mechanisms for this response. OBJECTIVE To examine the neurochemical effects of GHRH in a subset of participants from the parent trial. DESIGN Randomized, double-blind, placebo-controlled substudy of a larger trial. SETTING Clinical research unit at the University of Washington School of Medicine. PARTICIPANTS Thirty adults (17 with mild cognitive impairment [MCI]), ranging in age from 55 to 87 years, were enrolled and successfully completed the study. INTERVENTIONS Participants self-administered daily subcutaneous injections of tesamorelin (Theratechnologies Inc), a stabilized analogue of human GHRH (1 mg/d), or placebo 30 minutes before bedtime for 20 weeks. At baseline and weeks 10 and 20, participants underwent brain magnetic resonance imaging and spectroscopy protocols and cognitive testing and provided blood samples after fasting. Participants also underwent glucose tolerance tests before and after intervention. MAIN OUTCOMES AND MEASURES Brain levels of glutamate, inhibitory transmitters γ-aminobutyric acid (GABA) and N-acetylaspartylglutamate (NAAG), and myo-inositol (MI), an osmolyte linked to Alzheimer disease in humans, were measured in three 2 × 2 × 2-cm3 left-sided brain regions (dorsolateral frontal, posterior cingulate, and posterior parietal). Glutamate, GABA, and MI levels were expressed as ratios to creatine plus phosphocreatine, and NAAG was expressed as a ratio to N-acetylaspartate. RESULTS After 20 weeks of GHRH administration, GABA levels were increased in all brain regions (P < .04), NAAG levels were increased (P = .03) in the dorsolateral frontal cortex, and MI levels were decreased in the posterior cingulate (P = .002). These effects were similar in adults with MCI and older adults with normal cognitive function. No changes in the brain levels of glutamate were observed. In the posterior cingulate, treatment-related changes in serum insulin-like growth factor 1 were positively correlated with changes in GABA (r = 0.47; P = .001) and tended to be negatively correlated with MI (r = -0.34; P = .06). Consistent with the results of the parent trial, a favorable treatment effect on cognition was observed in substudy participants (P = .03). No significant associations were observed between treatment-related changes in neurochemical and cognitive outcomes. Glucose homeostasis in the periphery was not reliably affected by GHRH administration and did not account for treatment neurochemical effects. CONCLUSIONS Twenty weeks of GHRH administration increased GABA levels in all 3 brain regions, increased NAAG levels in the frontal cortex, and decreased MI levels in the posterior cingulate. To our knowledge, this is the first evidence that 20 weeks of somatotropic supplementation modulates inhibitory neurotransmitter and brain metabolite levels in a clinical trial, and it provides preliminary support for one possible mechanism to explain favorable GHRH effects on cognition in adults with MCI and in healthy older adults. TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT00257712.
Collapse
Affiliation(s)
- Seth D Friedman
- Department of Radiology, Seattle Children's Hospital, Seattle, Washington 98105, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Baker LD, Barsness SM, Borson S, Merriam GR, Friedman SD, Craft S, Vitiello MV. Effects of growth hormone–releasing hormone on cognitive function in adults with mild cognitive impairment and healthy older adults: results of a controlled trial. ACTA ACUST UNITED AC 2013; 69:1420-9. [PMID: 22869065 DOI: 10.1001/archneurol.2012.1970] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Growth hormone–releasing hormone(GHRH), growth hormone, and insulin like growth factor 1 have potent effects on brain function, their levels decrease with advancing age, and they likely play a role in the pathogenesis of Alzheimer disease. Previously, we reported favorable cognitive effects of short-term GHRH administration in healthy older adults and provided preliminary evidence to suggest a similar benefit in adults with mild cognitive impairment (MCI). OBJECTIVE To examine the effects of GHRH on cognitive function in healthy older adults and in adults with MCI. DESIGN Randomized,double-blind,placebo-controlled trial. SETTING Clinical Research Center, University of Washington School of Medicine in Seattle. PARTICIPANTS A total of 152 adults (66 with MCI) ranging in age from 55 to 87 years (mean age, 68 years); 137 adults (76 healthy participants and 61 participants with MCI) successfully completed the study. INTERVENTION Participants self-administered daily subcutaneous injections of tesamorelin (Theratechnologies Inc),a stabilized analog of human GHRH (1 mg/d), or placebo 30 minutes before bedtime for 20 weeks. At baseline, at weeks 10 and 20 of treatment, and after a 10-week washout(week 30), blood samples were collected, and parallel versions of a cognitive battery were administered. Before and after the 20-week intervention, participants completed an oral glucose tolerance test and a dual-energy x-ray absorptiometry scan to measure body composition. MAIN OUTCOME MEASURES Primary cognitive outcomes were analyzed using analysis of variance and included 3 composites reflecting executive function, verbal memory, and visual memory. Executive function was assessed with Stroop Color-Word Interference,Task Switching, the Self-Ordered Pointing Test, and Word Fluency, verbal memory was assessed with Story Recall and the Hopkins Verbal Learning Test,and visual memory was assessed with the Visual-Spatial Learning Test and Delayed Match-to-Sample. RESULTS The intent-to-treat analysis indicated a favorable effect of GHRH on cognition (P=.03), which was comparable in adults with MCI and healthy older adults.The completer analysis showed a similar pattern, with a more robust GHRH effect (P=.002). Subsequent analyses indicated a positive GHRH effect on executive function (P=.005) and a trend showing a similar treatment-related benefit in verbal memory(P=.08). Treatment with GHRH increased insulin like growth factor 1 levels by 117 %(P.001), which remained within the physiological range, and reduced percent body fat by 7.4%(P.001). Treatment with GHRH increased fasting insulin levels within the normal range by 35%in adults with MCI (P.001) but not in healthy adults. Adverse events were mild and were reported by 68%of GHRH treated adults and 36% of those who received placebo. CONCLUSIONS Twenty weeks of GHRH administration had favorable effects on cognition in both adults with MCI and healthy older adults. Longer-duration treatment trials are needed to further examine the therapeutic potential of GHRH administration on brain health during normal aging and “pathological aging.” TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT00257712
Collapse
Affiliation(s)
- Laura D Baker
- Department of Psychiatry and Behavioral Science, University of Washington School of Medicine, USA.
| | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
Frailty is the most problematic expression of population ageing. It is a state of vulnerability to poor resolution of homoeostasis after a stressor event and is a consequence of cumulative decline in many physiological systems during a lifetime. This cumulative decline depletes homoeostatic reserves until minor stressor events trigger disproportionate changes in health status. In landmark studies, investigators have developed valid models of frailty and these models have allowed epidemiological investigations that show the association between frailty and adverse health outcomes. We need to develop more efficient methods to detect frailty and measure its severity in routine clinical practice, especially methods that are useful for primary care. Such progress would greatly inform the appropriate selection of elderly people for invasive procedures or drug treatments and would be the basis for a shift in the care of frail elderly people towards more appropriate goal-directed care.
Collapse
Affiliation(s)
- Andrew Clegg
- Academic Unit of Elderly Care and Rehabilitation, University of Leeds, Leeds, UK.
| | | | | | | | | |
Collapse
|
30
|
Deak F, Sonntag WE. Aging, synaptic dysfunction, and insulin-like growth factor (IGF)-1. J Gerontol A Biol Sci Med Sci 2012; 67:611-25. [PMID: 22503992 PMCID: PMC3348499 DOI: 10.1093/gerona/gls118] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Accepted: 03/16/2012] [Indexed: 01/05/2023] Open
Abstract
Insulin-like growth factor (IGF)-1 is an important neurotrophic hormone. Deficiency of this hormone has been reported to influence the genesis of cognitive impairment and dementia in the elderly patients. Nevertheless, there are studies indicating that cognitive function can be maintained into old age even in the absence of circulating IGF-1 and studies that link IGF-1 to an acceleration of neurological diseases. Although IGF-1 has a complex role in brain function, synaptic effects appear to be central to the IGF-1-induced improvement in learning and memory. In this review, synaptic mechanisms of learning and memory and the effects of IGF-1 on synaptic communication are discussed. The emerging data indicate that synaptic function decreases with age and that IGF-1 contributes to information processing in the brain. Further studies that detail the specific actions of this important neurotrophic hormone will likely lead to therapies that result in improved cognitive function for the elderly patients.
Collapse
Affiliation(s)
- Ferenc Deak
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
| | - William E. Sonntag
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, Oklahoma City, Oklahoma
| |
Collapse
|
31
|
McGonnell IM, Grigoriadis AE, Lam EWF, Price JS, Sunters A. A specific role for phosphoinositide 3-kinase and AKT in osteoblasts? Front Endocrinol (Lausanne) 2012; 3:88. [PMID: 22833734 PMCID: PMC3400941 DOI: 10.3389/fendo.2012.00088] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Accepted: 06/29/2012] [Indexed: 12/25/2022] Open
Abstract
The phosphoinositide 3-kinase and AKT (protein kinase B) signaling pathway (PI3K/AKT) plays a central role in the control of cell survival, growth, and proliferation throughout the body. With regard to bone, and particularly in osteoblasts, there is an increasing amount of evidence that the many signaling molecules exert some of their bone-specific effects in part via selectively activating some of the generic effects of the PI3K/AKT pathway in osteoblasts. There is further data demonstrating that PI3K/AKT has the capacity to specifically cross-talk with other signaling pathways and transcriptional networks controlling bone cells' development in order to fine-tune the osteoblast phenotype. There is also evidence that perturbations in the PI3K/AKT pathway may well be responsible for certain bone pathologies. In this review, we discuss some of these findings and suggest that the PI3K/AKT pathway is a central nexus in the extensive network of extracellular signaling pathways that control the osteoblast.
Collapse
Affiliation(s)
- Imelda M. McGonnell
- Department of Veterinary Basic Sciences, The Royal Veterinary College,London, UK
| | - Agamemnon E. Grigoriadis
- Department of Craniofacial Development and Stem Cell Biology, King’s College London, Guy’s Hospital,London, UK
| | - Eric W.-F. Lam
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital,London, UK
| | - Joanna S. Price
- School of Veterinary Sciences, University of Bristol,Bristol, UK
| | - Andrew Sunters
- Department of Veterinary Basic Sciences, The Royal Veterinary College,London, UK
- *Correspondence: Andrew Sunters, Department of Veterinary Basic Sciences, The Royal Veterinary College, Royal College Street, Camden, London NW1 0TU, UK. e-mail:
| |
Collapse
|
32
|
Brar KS. Prevalent and Emerging Therapies for Osteoporosis. Med J Armed Forces India 2010; 66:249-54. [PMID: 27408312 PMCID: PMC4921246 DOI: 10.1016/s0377-1237(10)80050-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2008] [Accepted: 05/14/2010] [Indexed: 10/18/2022] Open
Abstract
Osteoporosis and fractures associated with it constitute a real and serious socio-medical problem, which only recently has come to the forefront of social consciousness. With increasing number of exservicemen and their dependents, osteoporosis management has become very important in our setup. Currently available pharmacological therapies for prevention of fragility fractures are limited in scope, efficacy and acceptability to patients. Oral bisphosphonates are the standard treatment for osteoporosis which are associated with significant gastrointestinal side effects and thus poor patient compliance. Newer regimens, including intravenous (IV) formulations of bisphosphonates, have successfully come in vogue with greater patient compliance and equal or better benefits. The real need in osteoporosis treatment is for additional anabolic drugs. The only currently approved anabolic agent for treating osteoporosis is teriparatide (recombinant human parathyroid hormone 1-34), which stimulates new bone formation. Considerable efforts are being made to develop new, more effective treatment for osteoporosis. These novel drugs under trial include those primarily inhibiting osteoclastic bone resorption (like bisphosphonates) such as inhibitors of receptor activator of nuclear factor-kappa B ligand (RANKL) signalling, cathepsin K inhibitors, c-Src kinase inhibitors, integrin inhibitors, chloride channel inhibitors and the drugs with osteo-anabolic actions such as orally active parathyroid hormone (PTH) analogues, calcium sensing receptor antagonists, PTH-related peptide analogues and agents that induce osteoblast anabolism via pathways involving key, recently identified, molecular targets (wnt low-density lipoprotein receptor-related protein-5 signalling; sclerostin antibodies).
Collapse
Affiliation(s)
- KS Brar
- Classified Specialist (Medicine & Endocrinology), Command Hospital (EC), Kolkata-27
| |
Collapse
|
33
|
Al-Delaimy WK, von Muhlen D, Barrett-Connor E. Insulinlike growth factor-1, insulinlike growth factor binding protein-1, and cognitive function in older men and women. J Am Geriatr Soc 2009; 57:1441-6. [PMID: 19515112 DOI: 10.1111/j.1532-5415.2009.02343.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVES To assess the association between insulinlike growth factor (IGF)-1 and IGF binding protein (IGFBP)-1 with three cognitive function tests in a healthy elderly population. DESIGN Cross-sectional analyses from the Rancho Bernardo Cohort Study. SETTING Southern California community of Rancho Bernardo. PARTICIPANTS Men (n=636) and women (n=899) from the Rancho Bernardo study (median age 74) were assessed between 1988 and 1992 for cognitive function using the Mini-Mental State Examination (MMSE), Verbal Fluency (VF) test, and Trail-Making Test Part B (Trails B). Blood samples were obtained at the same time for IGF-1 and IGFBP-1 levels. The association between biomarkers and cognitive function tests was assessed by dichotomizing tests at the clinically relevant cutoff using logistic regression and according to tertiles and continuous IGF-1 and IGFBP-1 levels using multivariate linear regression analyses. RESULTS The mean MMSE, VF and Trails B scores indicated better cognitive function with higher IGF-1 tertile. Multivariate analyses showed that VF and MMSE were each significantly associated with IGF-1 in a dose-response manner for men (P for trend=.001), but no cognitive function tests were related to IGF-1 in women. For men, the highest IGFBP-1 tertile was inversely and significantly different from the lowest tertile for the MMSE test only (P for trend=.02). CONCLUSION IGF-1 was independently and positively related to MMSE and VF in men, and IGFBP-1 was inversely associated with MMSE in men. Sex differences in the association should be further investigated.
Collapse
Affiliation(s)
- Wael K Al-Delaimy
- Department of Family and Preventive Medicine, School of Medicine, University of California at San Diego, La Jolla, California 92093, USA.
| | | | | |
Collapse
|
34
|
Abstract
Growth hormone (GH) is widely used as a performance-enhancing drug. One of its best-characterized effects is increasing levels of circulating insulin-like growth factor I (IGF-I), which is primarily of hepatic origin. It also induces synthesis of IGF-I in most non-hepatic tissues. The effects of GH in promoting postnatal body growth are IGF-I dependent, but IGF-I-independent functions are beginning to be elucidated. Although benefits of GH administration have been reported for those who suffer from GH deficiency, there is currently very little evidence to support an anabolic role for supraphysiological levels of systemic GH or IGF-I in skeletal muscle of healthy individuals. There may be other performance-enhancing effects of GH. In contrast, the hypertrophic effects of muscle-specific IGF-I infusion are well documented in animal models and muscle cell culture systems. Studies examining the molecular responses to hypertrophic stimuli in animals and humans frequently cite upregulation of IGF-I messenger RNA or immunoreactivity. The circulatory/systemic (endocrine) and local (autocrine/paracrine) effects of GH and IGF-I may have distinct effects on muscle mass regulation.
Collapse
|