1
|
Bale LK, West SA, Gades NM, Baker DJ, Conover CA. Gene deletion of Pregnancy-associated Plasma Protein-A (PAPP-A) improves pathology and cognition in an Alzheimer's disease mouse model. Exp Neurol 2024; 382:114976. [PMID: 39349117 PMCID: PMC11502239 DOI: 10.1016/j.expneurol.2024.114976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 09/13/2024] [Accepted: 09/22/2024] [Indexed: 10/02/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease of age with no effective preventative or treatment approaches. Deeper understanding of the mechanisms underlying the accumulation of toxic β-amyloid oligopeptides and the formation of amyloid plaque in AD has the potential to identify new therapeutic targets. Prior research links the insulin-like growth factor (IGF) system to pathologic mechanisms underlying AD. Suppression of local IGF-I receptor (IGFIR) signaling in AD mice has been shown to reduce plaque formation in the brain and delay neurodegeneration and behavioral changes. However, direct inhibitors of IGFIR signaling are not a viable treatment option for AD due to the essentiality of the IGFIR in physiological growth and metabolism. We have previously demonstrated a more selective means to reduce local IGFIR signaling through inhibition of PAPP-A, a novel zinc metalloprotease that regulates local IGF-I bioavailability through cleavage of inhibitory IGF binding proteins. Here we tested if deletion of PAPP-A in a mouse model of AD provides protection against pathology and behavioral changes. We show that compared to AD mice, AD/PAPP-A KO mice had significantly less plaque burden, reduced astrocytic activation, decreased IGF-IR activity, and improved cognition. Human senile AD plaques showed specific immunostaining for PAPP-A. Thus, inhibition of PAPP-A expression or activity may represent a novel treatment strategy for AD.
Collapse
Affiliation(s)
- Laurie K Bale
- Department of Endocrinology, Mayo Clinic, Rochester, MN 55905, United States of America.
| | - Sally A West
- Department of Endocrinology, Mayo Clinic, Rochester, MN 55905, United States of America.
| | - Naomi M Gades
- Department of Comparative Medicine, Mayo Clinic, Scottsdale, AZ 85259, United States of America.
| | - Darren J Baker
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, United States of America; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, United States of America.
| | - Cheryl A Conover
- Department of Endocrinology, Mayo Clinic, Rochester, MN 55905, United States of America.
| |
Collapse
|
2
|
Shen J, Liu L, Yang Y, Zhou M, Xu S, Zhang W, Zhang C. Insulin-Like Growth Factor 1 Has the Potential to Be Used as a Diagnostic Tool and Treatment Target for Autism Spectrum Disorders. Cureus 2024; 16:e65393. [PMID: 39188438 PMCID: PMC11346671 DOI: 10.7759/cureus.65393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2024] [Indexed: 08/28/2024] Open
Abstract
Autism spectrum disorder (ASD), a heterogeneous group of neurodevelopmental disorders, is characterized by social impairment and repetitive and stereotypic behaviors. Because of the lack of approved laboratory diagnostic markers and effective therapeutic medications, it is one of the most challenging diseases. Therefore, it is urgent to explore potential diagnosis markers or therapeutic targets. Insulin-like growth factor 1 (IGF-1) is a neurotrophic growth factor that enhances brain development. IGF-1 levels in body fluids are lower in preschool children with ASD than in typically developing children, which may serve as a potential diagnostic marker. In various ASD models associated with genetic or environmental exposure, IGF-1 treatment can improve core symptoms or pathological changes, including neuronal development, neural cell survival, balance of synaptic excitation and inhibition, neuroimmunology, and oxidative stress status. In March 2023 an IGF-1 derivative was approved as the first drug for treating Rett syndrome, an ASD-related neurodevelopmental disorder, to improve fundamental symptoms such as social communication. Thus, in this review, we present accumulating evidence of altered IGF-1 levels in ASD patients and the possible mechanisms, as well as evidence that IGF-1 treatment improves the pathophysiology in various ASD models. IGF-1 has the potential to be an early diagnosis marker and an effective therapeutic for ASD.
Collapse
Affiliation(s)
- Jiamin Shen
- Department of Children Health Care, Jingmen Maternity and Child Health Care Hospital, Jingmen, CHN
| | - Lijuan Liu
- Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, CHN
| | - Yifan Yang
- Department of Children Health Care, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital) Tongji Medical College, Huazhong University of Science and Technology, Wuhan, CHN
| | - Miao Zhou
- Department of Children Health Care, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital) Tongji Medical College, Huazhong University of Science and Technology, Wuhan, CHN
| | - Shan Xu
- Department of Children Health Care, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital) Tongji Medical College, Huazhong University of Science and Technology, Wuhan, CHN
| | - Wanqing Zhang
- Department of Children Health Care, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital) Tongji Medical College, Huazhong University of Science and Technology, Wuhan, CHN
| | - Chuanjie Zhang
- Department of Children Health Care, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital) Tongji Medical College, Huazhong University of Science and Technology, Wuhan, CHN
| |
Collapse
|
3
|
Ryoo SW, Anita NZ, Perlman G, Xiong LY, Wu CY, Wood M, Rabin JS, Mitchell J, Swardfager W. Insulin-like growth factor-1 and cognition in normoglycemia, prediabetes, and type 2 diabetes mellitus. Psychoneuroendocrinology 2024; 161:106946. [PMID: 38198904 DOI: 10.1016/j.psyneuen.2023.106946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 11/23/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024]
Abstract
BACKGROUND The relationship between insulin-like growth factor-1 (IGF-1) and cognition has been studied in healthy individuals, but not extensively with regards to insulin resistance and type 2 diabetes mellitus (T2DM). In this retrospective observational study, we investigated relationships of IGF-1 with memory and executive function across people with normoglycemia, prediabetes, and T2DM. METHODS Data from the Midlife in the United States (MIDUS) study were used. Episodic memory and executive function were assessed using the Brief Test of Adult Cognition by Telephone approximately 21.42 ± 12.10 months prior to measuring IGF-1 levels from a fasting blood sample. Normoglycemia was identified as individuals without a physician diagnosis of diabetes and glycated hemoglobin (HbA1c) ≤5.6%. Prediabetes was identified as those without a physician diagnosis of diabetes and HbA1c between 5.7%-6.4%. T2DM was identified as anyone with a physician diagnosis of diabetes, or HbA1c ≥6.5%, or anyone using an oral hypoglycemic medication. The associations were assessed using linear regressions controlling for age, sex, education, body mass index, C-reactive protein, HbA1c or homeostatic model of insulin resistance, MIDUS wave, exercise, smoking status, sleep quality, alcohol intake, oral hypoglycemic use, and insulin use. RESULTS The study included 1400 participants, which consisted of 583 normoglycemic (48.4% female, mean age 51.0 ± 12.2 years), 512 prediabetes (58.4% female, mean age 57.3 ± 11.8 years), and 305 T2DM participants (53.8% female, mean age 57.6 ± 11.5 years). Peripheral IGF-1 concentrations were lower (F2,1397 = 28.29, p < 0.001) in people with prediabetes or T2DM, vs. normoglycemia. Participants with prediabetes or T2DM had lower episodic memory (F2,1397 = 9.21, p < 0.001) and executive function (F2,1397 = 20.29, p < 0.001) composite z-scores than people with normoglycemia. Higher IGF-1 concentrations were associated with better executive performance in individuals with prediabetes (β = 0.115 [0.028, 0.202], p = 0.010), but not in individuals with normoglycemia or T2DM. An interaction between IGF-1 and sex in predicting executive function was observed in the prediabetes group (β = -0.344, p = 0.042), where the relationship was weaker in females (β = 0.106 [-0.012, 0.224], p = 0.077) than males (β = 0.251 [0.123, 0.380], p < 0.001). No associations were seen between IGF-1 and memory. CONCLUSION The results suggest that peripheral IGF-1 concentrations may be related to executive function, and that the relationship may be sex-specific and dependent on diabetes status.
Collapse
Affiliation(s)
- Si Won Ryoo
- Department of Pharmacology and Toxicology- University of Toronto, Medical Sciences Building, 1 King's College Circle Room 4207, Toronto ON M5S 1A8, Canada; Hurvitz Brain Sciences Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto ON M4N 3M5, Canada; University Health Network Toronto Rehabilitation Institute - Rumsey Centre Cardiac Rehabilitation, 347 Rumsey Rd, East York ON M4G 2V6, Canada
| | - Natasha Z Anita
- Department of Pharmacology and Toxicology- University of Toronto, Medical Sciences Building, 1 King's College Circle Room 4207, Toronto ON M5S 1A8, Canada; Hurvitz Brain Sciences Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto ON M4N 3M5, Canada; University Health Network Toronto Rehabilitation Institute - Rumsey Centre Cardiac Rehabilitation, 347 Rumsey Rd, East York ON M4G 2V6, Canada
| | - George Perlman
- Research Institute of McGill University Health Centre, 1001 Decarie Blvd, Montreal QC H4A 3J1, Canada
| | - Lisa Y Xiong
- Department of Pharmacology and Toxicology- University of Toronto, Medical Sciences Building, 1 King's College Circle Room 4207, Toronto ON M5S 1A8, Canada; Hurvitz Brain Sciences Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto ON M4N 3M5, Canada
| | - Che-Yuan Wu
- Department of Pharmacology and Toxicology- University of Toronto, Medical Sciences Building, 1 King's College Circle Room 4207, Toronto ON M5S 1A8, Canada; Hurvitz Brain Sciences Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto ON M4N 3M5, Canada
| | - Madeline Wood
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto ON M4N 3M5, Canada; Rehabilitation Sciences Institute - University of Toronto, Rehabilitation Sciences Building, 500 University Avenue, Suite 160, Toronto ON M5G 1V7, Canada
| | - Jennifer S Rabin
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto ON M4N 3M5, Canada; Rehabilitation Sciences Institute - University of Toronto, Rehabilitation Sciences Building, 500 University Avenue, Suite 160, Toronto ON M5G 1V7, Canada; Division of Neurology, Department of Medicine - University of Toronto, C. David Naylor Building, 6 Queen's Park Crescent West, Third Floor, Toronto ON M5S 3H2, Canada; Harquail Centre for Neuromodulation, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto ON M4N 3M5, Canada
| | - Jane Mitchell
- Department of Pharmacology and Toxicology- University of Toronto, Medical Sciences Building, 1 King's College Circle Room 4207, Toronto ON M5S 1A8, Canada
| | - Walter Swardfager
- Department of Pharmacology and Toxicology- University of Toronto, Medical Sciences Building, 1 King's College Circle Room 4207, Toronto ON M5S 1A8, Canada; Hurvitz Brain Sciences Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto ON M4N 3M5, Canada; University Health Network Toronto Rehabilitation Institute - Rumsey Centre Cardiac Rehabilitation, 347 Rumsey Rd, East York ON M4G 2V6, Canada.
| |
Collapse
|
4
|
Dya GA, Klychnikov OI, Adasheva DA, Vladychenskaya EA, Katrukha AG, Serebryanaya DV. IGF-Binding Proteins and Their Proteolysis as a Mechanism of Regulated IGF Release in the Nervous Tissue. BIOCHEMISTRY (MOSCOW) 2023; 88:S105-S122. [PMID: 37069117 DOI: 10.1134/s0006297923140079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
Insulin-like growth factors 1 and 2 (IGF-1 and IGF-2) play a key role in the maintenance of the nervous tissue viability. IGF-1 and IGF-2 exhibit the neuroprotective effects by stimulating migration and proliferation of nervous cells, activating cellular metabolism, inducing regeneration of damaged cells, and regulating various stages of prenatal and postnatal development of the nervous system. The availability of IGFs for the cells is controlled via their interaction with the IGF-binding proteins (IGFBPs) that inhibit their activity. On the contrary, the cleavage of IGFBPs by specific proteases leads to the IGF release and activation of its cellular effects. The viability of neurons in the nervous tissue is controlled by a complex system of trophic factors secreted by auxiliary glial cells. The main source of IGF for the neurons are astrocytes. IGFs can accumulate as an extracellular free ligand near the neuronal membranes as a result of proteolytic degradation of IGFBPs by proteases secreted by astrocytes. This mechanism promotes interaction of IGFs with their genuine receptors and triggers intracellular signaling cascades. Therefore, the release of IGF by proteolytic cleavage of IGFBPs is an important mechanism of neuronal protection. This review summarizes the published data on the role of IGFs and IGFBPs as the key players in the neuroprotective regulation with a special focus on the specific proteolysis of IGFBPs as a mechanism for the regulation of IGF bioavailability and viability of neurons.
Collapse
Affiliation(s)
- German A Dya
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Oleg I Klychnikov
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Daria A Adasheva
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Elizaveta A Vladychenskaya
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Alexey G Katrukha
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Daria V Serebryanaya
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia.
| |
Collapse
|
5
|
Crosstalk between neurological, cardiovascular, and lifestyle disorders: insulin and lipoproteins in the lead role. Pharmacol Rep 2022; 74:790-817. [PMID: 36149598 DOI: 10.1007/s43440-022-00417-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/03/2022] [Accepted: 09/08/2022] [Indexed: 10/14/2022]
Abstract
Insulin resistance and impaired lipoprotein metabolism contribute to a plethora of metabolic and cardiovascular disorders. These alterations have been extensively linked with poor lifestyle choices, such as consumption of a high-fat diet, smoking, stress, and a redundant lifestyle. Moreover, these are also known to increase the co-morbidity of diseases like Type 2 diabetes mellitus and atherosclerosis. Under normal physiological conditions, insulin and lipoproteins exert a neuroprotective role in the central nervous system. However, the tripping of balance between the periphery and center may alter the normal functioning of the brain and lead to neurological disorders such as Alzheimer's disease, Parkinson's disease, stroke, depression, and multiple sclerosis. These neurological disorders are further characterized by certain behavioral and molecular changes that show consistent overlap with alteration in insulin and lipoprotein signaling pathways. Therefore, targeting these two mechanisms not only reveals a way to manage the co-morbidities associated with the circle of the metabolic, central nervous system, and cardiovascular disorders but also exclusively work as a disease-modifying therapy for neurological disorders. In this review, we summarize the role of insulin resistance and lipoproteins in the progression of various neurological conditions and discuss the therapeutic options currently in the clinical pipeline targeting these two mechanisms; in addition, challenges faced in designing these therapeutic approaches have also been touched upon briefly.
Collapse
|
6
|
Yesbek Kaymaz A, Kostel Bal S, Bora G, Talim B, Ozon A, Alikasifoglu A, Topaloglu H, Erdem Yurter H. Alterations in insulin-like growth factor system in spinal muscular atrophy. Muscle Nerve 2022; 66:631-638. [PMID: 36050898 DOI: 10.1002/mus.27715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 08/19/2022] [Accepted: 08/26/2022] [Indexed: 11/10/2022]
Abstract
INTRODUCTION/AIMS Spinal muscular atrophy (SMA) is an inherited neuromuscular disease caused by survival motor neuron (SMN) protein deficiency. Insulin-like growth factor-I (IGF-I) is a myotrophic and neurotrophic factor that has been reported to be dysregulated in in vivo SMA model systems. However, detailed analyses of the IGF-I system in SMA patients are missing. In this study, we analyzed the components of the IGF-I system in serum and archived skeletal muscle biopsies of SMA patients. METHODS Serum IGF-I, IGF binding protein (IGFBP)-3, and IGFBP-5 levels were analyzed in 11 SMA patients and 13 healthy children by immunoradiometric and enzyme-linked immunosorbent assays. The expression of IGF-I, IGF-I receptor, and IGFBP-5 proteins was investigated by immunofluorescence analysis in the archived skeletal muscle biopsies of 9 SMA patients, 6 patients with non-SMA-related neuromuscular disease and atrophic fibers in muscle biopsy, and 4 controls. RESULTS A significant decrease in IGF-I levels (mean ± SD: -1.39 ± 1.46 vs. 0.017 ± 0.83, p = 0.02) and increase in IGFBP-5 levels (mean ± SD: 2358.5 ± 1617.4 ng/mL vs. 1003.4 ± 274.3 ng/mL, p=0.03) were detected in serum samples of SMA patients compared to healthy controls. Increased expression of IGF-I, IGF-I receptor, and IGFBP-5 was detected in skeletal muscle biopsies of SMA patients and non-SMA neuromuscular diseases, indicating atrophy-specific alterations in the pathway. DISCUSSION Our findings suggested that the components of the IGF-I system are altered in SMA patients at both the systemic and tissue-specific levels.
Collapse
Affiliation(s)
- Ayse Yesbek Kaymaz
- Department of Medical Biology, Hacettepe University, Faculty of Medicine, Ankara, Turkey
| | - Sevgi Kostel Bal
- Department of Pediatrics, Neurology Unit, Hacettepe University, Faculty of Medicine, Ankara, Turkey
| | - Gamze Bora
- Department of Medical Biology, Hacettepe University, Faculty of Medicine, Ankara, Turkey
| | - Beril Talim
- Department of Pediatrics, Pediatric Pathology Unit, Hacettepe University, Faculty of Medicine, Ankara, Turkey
| | - Alev Ozon
- Department of Pediatrics, Division of Pediatric Endocrinology, Hacettepe University, Faculty of Medicine, Ankara, Turkey
| | - Ayfer Alikasifoglu
- Department of Pediatrics, Division of Pediatric Endocrinology, Hacettepe University, Faculty of Medicine, Ankara, Turkey
| | - Haluk Topaloglu
- Department of Pediatrics, Neurology Unit, Hacettepe University, Faculty of Medicine, Ankara, Turkey
| | - Hayat Erdem Yurter
- Department of Medical Biology, Hacettepe University, Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
7
|
Abeln V, Fomina E, Popova J, Braunsmann L, Koschate J, Möller F, Fedyay SO, Vassilieva GY, Schneider S, Strüder HK, Klein T. Chronic, acute and protocol-dependent effects of exercise on psycho-physiological health during long-term isolation and confinement. BMC Neurosci 2022; 23:41. [PMID: 35773633 PMCID: PMC9244384 DOI: 10.1186/s12868-022-00723-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 06/13/2022] [Indexed: 12/27/2022] Open
Abstract
Exercise could prevent physical and psychological deteriorations, especially during pandemic times of lock-down scenarios and social isolation. But to meet both, the common exercise protocols require optimization based on holistic investigations and with respect to underlying processes. This study aimed to explore individual chronic and acute effects of continuous and interval running exercise on physical and cognitive performance, mood, and affect and underlying neurophysiological factors during a terrestrial simulated space mission. Six volunteers (three females) were isolated for 120 days. Accompanying exercise training consisted of a continuous and interval running protocol in a cross-over design. Incremental stage tests on a treadmill were done frequently to test physical performance. Actigraphy was used to monitor physical activity level. Cognitive performance, mood (MoodMeter®), affect (PANAS), brain-derived neurotrophic factor (BDNF), insulin-like growth factor 1 (IGF-1), vascular-endothelial growth factor (VEGF), and saliva cortisol were investigated prior to, four times during, and after isolation, pre- and post-exercise on two separate days, respectively. As a chronic effect, physical performance increased (and IGF-1 tended) in the course of isolation and training until the end of isolation. Subjective mood and affect state, as well as cognitive performance, basal BDNF and VEGF levels, were well-preserved across the intervention. No acute effects of exercise were detected, besides slower reaction time after exercise in two out of nine cognitive tests, testing sensorimotor speed and memory of complex figures. Consistently higher basal IGF-1 concentrations and faster reaction time in the psychomotor vigilance test were found for the continuous compared to the interval running protocol. The results suggest that 120 days of isolation and confinement can be undergone without cognitive and mental deteriorations. Regular, individual aerobic running training supporting physical fitness is hypothesized to play an important role in this regard. Continuous running exercise seems to trigger higher IGF-1 levels and vigilance compared to interval running. Systematic and prolonged investigations and larger sample size are required to follow up on exercise-protocol specific differences in order to optimize the exercise intervention for long-term psycho-physiological health and well-being.
Collapse
Affiliation(s)
- V Abeln
- Institute of Movement and Neurosciences, Center for Health and Integrative Physiology in Space (CHIPS), German Sport University Cologne, Am Sportpark Muengersdorf 6, 50933, Cologne, Germany.
| | - E Fomina
- Institute of Biomedical Problems (IBMP), Russian Academy of Sciences, Khoroshevskoye shosse 76A, 123007, Moscow, Russia
| | - J Popova
- Institute of Biomedical Problems (IBMP), Russian Academy of Sciences, Khoroshevskoye shosse 76A, 123007, Moscow, Russia
| | - L Braunsmann
- Institute of Movement and Neurosciences, Center for Health and Integrative Physiology in Space (CHIPS), German Sport University Cologne, Am Sportpark Muengersdorf 6, 50933, Cologne, Germany
| | - J Koschate
- Geriatric Medicine, School of Medicine and Health Sciences, Carl Von Ossietzky University Oldenburg, Ammerlaender Heerstr. 140, 26129, Oldenburg, Germany
| | - F Möller
- Department of Exercise Physiology, German Sport University Cologne, Am Sportpark Muengersdorf 6, 50933, CologneCologne, Germany
| | - S O Fedyay
- Institute of Biomedical Problems (IBMP), Russian Academy of Sciences, Khoroshevskoye shosse 76A, 123007, Moscow, Russia
| | - G Y Vassilieva
- Institute of Biomedical Problems (IBMP), Russian Academy of Sciences, Khoroshevskoye shosse 76A, 123007, Moscow, Russia
| | - S Schneider
- Institute of Movement and Neurosciences, Center for Health and Integrative Physiology in Space (CHIPS), German Sport University Cologne, Am Sportpark Muengersdorf 6, 50933, Cologne, Germany
| | - H K Strüder
- Institute of Movement and Neurosciences, Center for Health and Integrative Physiology in Space (CHIPS), German Sport University Cologne, Am Sportpark Muengersdorf 6, 50933, Cologne, Germany
| | - T Klein
- Institute of Movement and Neurosciences, Center for Health and Integrative Physiology in Space (CHIPS), German Sport University Cologne, Am Sportpark Muengersdorf 6, 50933, Cologne, Germany
- Institute of Sport Science, University of Rostock, 18057, Rostock, Germany
| |
Collapse
|
8
|
Association of the Combined Effects between Insulin-Like Growth Factor-1 Gene Polymorphisms and Negative Life Events with Major Depressive Disorder among Chinese population in the Context of Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3253687. [PMID: 35498133 PMCID: PMC9054463 DOI: 10.1155/2022/3253687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 04/05/2022] [Indexed: 11/24/2022]
Abstract
Background Oxidative stress may be increased in a number of psychiatric disorders, including major depressive disorder (MDD). MDD has been shown to be related to insulin-like growth factor-1 (IGF-1) as well as to negative life events; exploring the interaction of IGF-1 polymorphisms and negative life events on the risk of MDD is needed. The aim of this study was to analyze the single and combined effects of IGF-1 polymorphisms (rs972936 and rs978458) and negative life events with MDD among Chinese population. Methods 420 MDD patients (according to DSM-V) and 420 age- and gender-matched control subjects were recruited in a case-control study. Negative life events were assessed using standard rating scales. IGF-1 rs972936 and rs978458 were identified by sequencing. The chi-square (χ2) tests were performed to explore the association of negative life events and IGF-1 polymorphisms with MDD. Results Our results found that the negative life events were associated with the risk of MDD (P < 0.001; OR = 3.28, 95% CI: 2.19-4.85). The genotypes of IGF-1 were associated with the risk of MDD (P < 0.001); carrying the IGF-1 rs972936 C allele (OR = 1.53, 95% CI: 1.26-1.85) and rs978458 T allele (OR = 1.92, 95% CI: 1.58-2.34) had a higher risk of MDD. The combined effects between IGF-1 rs978458 and negative life events were associated with the risk of MDD (P < 0.05; OR = 2.94, 95% CI: 1.23-7.03), but IGF-1 rs972936 was not associated (P > 0.05). Conclusions Based on the oxidative stress hypothesis, we confirm that carrying IGF-1 rs972936 C allele and rs978458 T allele have a higher risk of MDD and the combined effects between IGF-1 rs978458 and negative life events were associated with the risk of MDD among Chinese population.
Collapse
|
9
|
Popow C, Ohmann S, Plener P. Practitioner's review: medication for children and adolescents with autism spectrum disorder (ASD) and comorbid conditions. NEUROPSYCHIATRIE : KLINIK, DIAGNOSTIK, THERAPIE UND REHABILITATION : ORGAN DER GESELLSCHAFT OSTERREICHISCHER NERVENARZTE UND PSYCHIATER 2021; 35:113-134. [PMID: 34160787 PMCID: PMC8429404 DOI: 10.1007/s40211-021-00395-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 05/15/2021] [Indexed: 11/14/2022]
Abstract
Alleviating the multiple problems of children with autism spectrum disorder (ASD) and its comorbid conditions presents major challenges for the affected children, parents, and therapists. Because of a complex psychopathology, structured therapy and parent training are not always sufficient, especially for those patients with intellectual disability (ID) and multiple comorbidities. Moreover, structured therapy is not available for a large number of patients, and pharmacological support is often needed, especially in those children with additional attention deficit/hyperactivity and oppositional defiant, conduct, and sleep disorders.
Collapse
Affiliation(s)
- Christian Popow
- Dept. Child and Adolescent Psychiatry, Medical University of Vienna, Waehringer Guertel 18–20, 1090 Vienna, Austria
| | - Susanne Ohmann
- Dept. Child and Adolescent Psychiatry, Medical University of Vienna, Waehringer Guertel 18–20, 1090 Vienna, Austria
| | - Paul Plener
- Dept. Child and Adolescent Psychiatry, Medical University of Vienna, Waehringer Guertel 18–20, 1090 Vienna, Austria
| |
Collapse
|
10
|
Scherer T, Sakamoto K, Buettner C. Brain insulin signalling in metabolic homeostasis and disease. Nat Rev Endocrinol 2021; 17:468-483. [PMID: 34108679 DOI: 10.1038/s41574-021-00498-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/22/2021] [Indexed: 02/06/2023]
Abstract
Insulin signalling in the central nervous system regulates energy homeostasis by controlling metabolism in several organs and by coordinating organ crosstalk. Studies performed in rodents, non-human primates and humans over more than five decades using intracerebroventricular, direct hypothalamic or intranasal application of insulin provide evidence that brain insulin action might reduce food intake and, more importantly, regulates energy homeostasis by orchestrating nutrient partitioning. This Review discusses the metabolic pathways that are under the control of brain insulin action and explains how brain insulin resistance contributes to metabolic disease in obesity, the metabolic syndrome and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Thomas Scherer
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria.
| | - Kenichi Sakamoto
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Christoph Buettner
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA.
| |
Collapse
|
11
|
Effects of Antidepressant Treatment on Neurotrophic Factors (BDNF and IGF-1) in Patients with Major Depressive Disorder (MDD). J Clin Med 2021; 10:jcm10153377. [PMID: 34362162 PMCID: PMC8346988 DOI: 10.3390/jcm10153377] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/09/2021] [Accepted: 07/26/2021] [Indexed: 12/28/2022] Open
Abstract
Major depressive disorder (MDD) remains the subject of ongoing research as a multifactorial disease and a serious public health problem. There is a growing body of literature focusing on the role of neurotrophic factors in pathophysiology of MDD. A neurotrophic hypothesis of depression proposes that abnormalities of neurotrophins serum levels lead to neuronal atrophy and decreased neurogenesis, resulting in mood disorders. Consequently, in accordance with recent findings, antidepressant treatment modifies the serum levels of neurotrophins and thus leads to a clinical improvement of MDD. The purpose of this review is to summarize the available data on the effects of various antidepressants on serum levels of neurotrophins such as brain-derived neurotrophic factor (BDNF) and insulin-like growth factor (IGF-1). In addition, the authors discuss their role as prognostic factors for treatment response in MDD. A literature search was performed using the PubMed database. Following the inclusion and exclusion criteria, nine original articles and three meta-analyses were selected. The vast majority of studies have confirmed the effect of antidepressants on BDNF levels. Research on IGF-1 is limited and insufficient to describe the correlation between different antidepressant drugs and factor serum levels; however, four studies indicated a decrease in IGF-1 after treatment. Preliminary data suggest BDNF as a promising predictor of treatment response in MDD patients. The role of IGF-1 needs further investigation.
Collapse
|
12
|
Mosiołek A, Pięta A, Jakima S, Zborowska N, Mosiołek J, Szulc A. Effects of Antidepressant Treatment on Peripheral Biomarkers in Patients with Major Depressive Disorder (MDD). J Clin Med 2021; 10:jcm10081706. [PMID: 33920992 PMCID: PMC8071355 DOI: 10.3390/jcm10081706] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/01/2021] [Accepted: 04/13/2021] [Indexed: 12/15/2022] Open
Abstract
Major depressive disorder (MDD) is one of the most prevalent mental illness and a leading cause of disability worldwide. Despite a range of effective treatments, more than 30% of patients do not achieve remission as a result of conventional therapy. In these circumstances the identification of novel drug targets and pathogenic factors becomes essential for selecting more efficacious and personalized treatment. Increasing evidence has implicated the role of inflammation in the pathophysiology of depression, revealing potential new pathways and treatment options. Moreover, convergent evidence indicates that MDD is related to disturbed neurogenesis and suggests a possible role of neurotrophic factors in recovery of function in patients. Although the influence of antidepressants on inflammatory cytokines balance was widely reported in various studies, the exact correlation between drugs used and specific cytokines and neurotrophins serum levels often remains inconsistent. Available data suggest anti-inflammatory properties of selective serotonin reuptake inhibitors (SSRIs), selective serotonin and noradrenaline inhibitors (SNRIs), and tricyclic antidepressants (TCAs) as a possible additional mechanism of reduction of depressive symptoms. In this review, we outline emerging data regarding the influence of different antidepressant drugs on a wide array of peripheral biomarkers such as interleukin (IL)-1ß, IL-2, IL-5, IL-6, IL-8, IL-10, C-reactive protein (CRP), or interferon (IFN)-γ. Presented results indicate anti-inflammatory effect for selected drugs or lack of such effect. Research in this field is insufficient to define the role of inflammatory markers as a predictor of treatment response in MDD.
Collapse
Affiliation(s)
- Anna Mosiołek
- Department of Psychiatry, Faculty of Health Sciences, Medical University of Warsaw, Żwirki i Wigury 61 Street, 02-091 Warszawa, Poland; (A.P.); (A.S.)
- Mazovia Specialist Health Center in Pruszków, Partyzantów 2/4 Street, 05-802 Pruszków, Poland; (S.J.); (N.Z.)
- Correspondence:
| | - Aleksandra Pięta
- Department of Psychiatry, Faculty of Health Sciences, Medical University of Warsaw, Żwirki i Wigury 61 Street, 02-091 Warszawa, Poland; (A.P.); (A.S.)
- Mazovia Specialist Health Center in Pruszków, Partyzantów 2/4 Street, 05-802 Pruszków, Poland; (S.J.); (N.Z.)
| | - Sławomir Jakima
- Mazovia Specialist Health Center in Pruszków, Partyzantów 2/4 Street, 05-802 Pruszków, Poland; (S.J.); (N.Z.)
| | - Natalia Zborowska
- Mazovia Specialist Health Center in Pruszków, Partyzantów 2/4 Street, 05-802 Pruszków, Poland; (S.J.); (N.Z.)
| | - Jadwiga Mosiołek
- Faculty of Medicine, Wroclaw Medical University, Wybrzeże Ludwika Pasteura 1 Street, 50-367 Wrocław, Poland;
| | - Agata Szulc
- Department of Psychiatry, Faculty of Health Sciences, Medical University of Warsaw, Żwirki i Wigury 61 Street, 02-091 Warszawa, Poland; (A.P.); (A.S.)
- Mazovia Specialist Health Center in Pruszków, Partyzantów 2/4 Street, 05-802 Pruszków, Poland; (S.J.); (N.Z.)
| |
Collapse
|
13
|
Okamoto N, Yoshino K, Kitagawa S, Fujii R, Hamada S, Ikenouchi A, Konishi Y, Ueda N, Eto Y, Tsutsumi Y, Yoshimura R. Association Between Serum Insulin-Like Growth Factor 1 Levels and the Clinical Symptoms of Chronic Schizophrenia: Preliminary Findings. Front Psychiatry 2021; 12:653802. [PMID: 33746806 PMCID: PMC7966725 DOI: 10.3389/fpsyt.2021.653802] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 02/05/2021] [Indexed: 11/30/2022] Open
Abstract
Purpose: Insulin-like growth factor 1 (IGF-1) is a trophic mediator that is regulated by growth hormone and associated with the proliferation, development, and growth of neural cells. IGF-1 may be associated with the pathophysiology of schizophrenia, but this association remains controversial. This study aimed to investigate the relationship between serum IGF-1 levels and psychiatric symptoms in patients with chronic schizophrenia. Patients and Methods: A total of 65 patients were recruited from the University of Occupational and Environmental Health, Komine Eto Hospital, Moji Matsugae Hospital, Shin-Moji Hospital, and Tsutsumi Hospital in Kitakyushu between September 2019 and June 2020. Further, 20 healthy age- and sex-matched control participants were recruited from the Komine Eto Hospital and the University of Occupational and Environmental Health. Patients with schizophrenia were assessed using the Positive and Negative Syndrome Scale (PANSS) and the Drug-Induced Extrapyramidal Symptoms Scale. Serum levels of free plus albumin-bound IGF-1 (IGF-1) were measured by immunoradiometric assay. The measurements were performed using antibody beads for bound/free separation. Associations between serum IGF-1 levels and the PANSS scores were determined. We also examined the associations between serum IGF-1 levels and diabetes, antipsychotic drug use, and disease duration. Results: No significant difference was found in the serum IGF-1 level between patients with schizophrenia and healthy controls. Serum IGF-1 levels were significantly negatively correlated with the PANSS total score (R 2 = 0.06, p = 0.015) and PANSS general score (R 2 = 0.088, p = 0.008), but not with the PANSS positive scores and PANSS negative scores. Serum IGF-1 levels were not related to the prevalence of diabetes (p = 0.64). However, a significant correlation was observed between serum IGF-1 levels and age (B = -1.88, p < 0.0001). Serum IGF-1 levels could not distinguish patients with schizophrenia and healthy controls. Conclusion: The association between serum IGF-1 levels and psychiatric symptoms may be complicated in patients with chronic schizophrenia.
Collapse
Affiliation(s)
- Naomichi Okamoto
- Department of Psychiatry, University of Occupational and Environmental Health, Kitakyushu, Japan
| | | | - Shogo Kitagawa
- Department of Psychiatry, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Rintaro Fujii
- Department of Psychiatry, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Shinsuke Hamada
- Department of Psychiatry, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Atsuko Ikenouchi
- Department of Psychiatry, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yuki Konishi
- Department of Psychiatry, University of Occupational and Environmental Health, Kitakyushu, Japan
| | | | | | | | - Reiji Yoshimura
- Department of Psychiatry, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|
14
|
Gasco V, Cambria V, Bioletto F, Ghigo E, Grottoli S. Traumatic Brain Injury as Frequent Cause of Hypopituitarism and Growth Hormone Deficiency: Epidemiology, Diagnosis, and Treatment. Front Endocrinol (Lausanne) 2021; 12:634415. [PMID: 33790864 PMCID: PMC8005917 DOI: 10.3389/fendo.2021.634415] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 02/16/2021] [Indexed: 12/12/2022] Open
Abstract
Traumatic brain injury (TBI)-related hypopituitarism has been recognized as a clinical entity for more than a century, with the first case being reported in 1918. However, during the 20th century hypopituitarism was considered only a rare sequela of TBI. Since 2000 several studies strongly suggest that TBI-mediated pituitary hormones deficiency may be more frequent than previously thought. Growth hormone deficiency (GHD) is the most common abnormality, followed by hypogonadism, hypothyroidism, hypocortisolism, and diabetes insipidus. The pathophysiological mechanisms underlying pituitary damage in TBI patients include a primary injury that may lead to the direct trauma of the hypothalamus or pituitary gland; on the other hand, secondary injuries are mainly related to an interplay of a complex and ongoing cascade of specific molecular/biochemical events. The available data describe the importance of GHD after TBI and its influence in promoting neurocognitive and behavioral deficits. The poor outcomes that are seen with long standing GHD in post TBI patients could be improved by GH treatment, but to date literature data on the possible beneficial effects of GH replacement therapy in post-TBI GHD patients are currently scarce and fragmented. More studies are needed to further characterize this clinical syndrome with the purpose of establishing appropriate standards of care. The purpose of this review is to summarize the current state of knowledge about post-traumatic GH deficiency.
Collapse
|
15
|
Boyne P, Meyrose C, Westover J, Whitesel D, Hatter K, Reisman DS, Carl D, Khoury JC, Gerson M, Kissela B, Dunning K. Effects of Exercise Intensity on Acute Circulating Molecular Responses Poststroke. Neurorehabil Neural Repair 2020; 34:222-234. [PMID: 31976813 DOI: 10.1177/1545968319899915] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background. Exercise intensity can influence functional recovery after stroke, but the mechanisms remain poorly understood. Objective. In chronic stroke, an intensity-dependent increase in circulating brain-derived neurotrophic factor (BDNF) was previously found during vigorous exercise. Using the same serum samples, this study tested acute effects of exercise intensity on other circulating molecules related to neuroplasticity, including vascular-endothelial growth factor (VEGF), insulin-like growth factor-1 (IGF1), and cortisol, with some updated analyses involving BDNF. Methods. Using a repeated-measures design, 16 participants with chronic stroke performed 3 exercise protocols in random order: treadmill high-intensity interval training (HIT-treadmill), seated-stepper HIT (HIT-stepper), and treadmill moderate-intensity continuous exercise (MCT-treadmill). Serum molecular changes were compared between protocols. Mediation and effect modification analyses were also performed. Results. VEGF significantly increased during HIT-treadmill, IGF1 increased during both HIT protocols and cortisol nonsignificantly decreased during each protocol. VEGF response was significantly greater for HIT-treadmill versus MCT-treadmill when controlling for baseline. Blood lactate positively mediated the effect of HIT on BDNF and cortisol. Peak treadmill speed positively mediated effects on BDNF and VEGF. Participants with comfortable gait speed ≥0.4 m/s had significantly lower VEGF and higher IGF1 responses, with a lower cortisol response during MCT-treadmill. Conclusions. BDNF and VEGF are promising serum molecules to include in future studies testing intensity-dependent mechanisms of exercise on neurologic recovery. Fast training speed and anaerobic intensity appear to be critical ingredients for eliciting these molecular responses. Serum molecular response differences between gait speed subgroups provide a possible biologic basis for previously observed differences in training responsiveness.
Collapse
Affiliation(s)
| | | | | | | | - Kristal Hatter
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | | | - Daniel Carl
- University of Cincinnati, Cincinnati, OH, USA
| | - Jane C Khoury
- University of Cincinnati, Cincinnati, OH, USA.,Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | | | | | | |
Collapse
|
16
|
Arteaga Cabeza O, Mikrogeorgiou A, Kannan S, Ferriero DM. Advanced nanotherapies to promote neuroregeneration in the injured newborn brain. Adv Drug Deliv Rev 2019; 148:19-37. [PMID: 31678359 DOI: 10.1016/j.addr.2019.10.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 09/19/2019] [Accepted: 10/23/2019] [Indexed: 12/16/2022]
Abstract
Neonatal brain injury affects thousands of babies each year and may lead to long-term and permanent physical and neurological problems. Currently, therapeutic hypothermia is standard clinical care for term newborns with moderate to severe neonatal encephalopathy. Nevertheless, it is not completely protective, and additional strategies to restore and promote regeneration are urgently needed. One way to ensure recovery following injury to the immature brain is to augment endogenous regenerative pathways. However, novel strategies such as stem cell therapy, gene therapies and nanotechnology have not been adequately explored in this unique age group. In this perspective review, we describe current efforts that promote neuroprotection and potential targets that are unique to the developing brain, which can be leveraged to facilitate neuroregeneration.
Collapse
|
17
|
Turkson S, Kloster A, Hamilton PJ, Neigh GN. Neuroendocrine drivers of risk and resilience: The influence of metabolism & mitochondria. Front Neuroendocrinol 2019; 54:100770. [PMID: 31288042 PMCID: PMC6886586 DOI: 10.1016/j.yfrne.2019.100770] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/20/2019] [Accepted: 07/03/2019] [Indexed: 02/07/2023]
Abstract
The manifestation of risk versus resilience has been considered from varying perspectives including genetics, epigenetics, early life experiences, and type and intensity of the challenge with which the organism is faced. Although all of these factors are central to determining risk and resilience, the current review focuses on what may be a final common pathway: metabolism. When an organism is faced with a perturbation to the environment, whether internal or external, appropriate energy allocation is essential to resolving the divergence from equilibrium. This review examines the potential role of metabolism in the manifestation of stress-induced neural compromise. In addition, this review details the current state of knowledge on neuroendocrine factors which are poised to set the tone of the metabolic response to a systemic challenge. The goal is to provide an essential framework for understanding stress in a metabolic context and appreciation for key neuroendocrine signals.
Collapse
Affiliation(s)
- Susie Turkson
- Department of Anatomy & Neurobiology, Virginia Commonwealth University, Richmond, VA, United States
| | - Alix Kloster
- Department of Anatomy & Neurobiology, Virginia Commonwealth University, Richmond, VA, United States
| | - Peter J Hamilton
- Department of Anatomy & Neurobiology, Virginia Commonwealth University, Richmond, VA, United States
| | - Gretchen N Neigh
- Department of Anatomy & Neurobiology, Virginia Commonwealth University, Richmond, VA, United States.
| |
Collapse
|
18
|
Neuroprotective Actions of Glucagon-Like Peptide-1 (GLP-1) Analogues in Alzheimer's and Parkinson's Diseases. CNS Drugs 2019; 33:209-223. [PMID: 30511349 DOI: 10.1007/s40263-018-0593-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The current absence of effective treatments for Alzheimer's disease (AD) and Parkinson's disease (PD) reflects an incomplete knowledge of the underlying disease processes. Considerable efforts have been made to investigate the central pathological features of these diseases, giving rise to numerous attempts to develop compounds that interfere with such features. However, further characterization of the molecular targets within the interconnected AD and PD pathways is still required. Impaired brain insulin signaling has emerged as a feature that contributes to neuronal dysfunction in both AD and PD, leading to strategies aiming at restoring this pathway in the brain. Long-acting glucagon-like peptide-1 (GLP-1) analogues marketed for treatment of type 2 diabetes mellitus have been tested and have shown encouraging protective actions in experimental models of AD and PD as well as in initial clinical trials. We review studies revealing the neuroprotective actions of GLP-1 analogues in pre-clinical models of AD and PD and promising results from recent clinical trials.
Collapse
|
19
|
Liraglutide and its Neuroprotective Properties-Focus on Possible Biochemical Mechanisms in Alzheimer's Disease and Cerebral Ischemic Events. Int J Mol Sci 2019; 20:ijms20051050. [PMID: 30823403 PMCID: PMC6429395 DOI: 10.3390/ijms20051050] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 02/24/2019] [Accepted: 02/25/2019] [Indexed: 12/28/2022] Open
Abstract
Liraglutide is a GLP-1 analog (glucagon like peptide-1) used primarily in the treatment of diabetes mellitus type 2 (DM2) and obesity. The literature starts to suggest that liraglutide may reduce the effects of ischemic stroke by activating anti-apoptotic pathways, as well as limiting the harmful effects of free radicals. The GLP-1R expression has been reported in the cerebral cortex, especially occipital and frontal lobes, the hypothalamus, and the thalamus. Liraglutide reduced the area of ischemia caused by MCAO (middle cerebral artery occlusion), limited neurological deficits, decreased hyperglycemia caused by stress, and presented anti-apoptotic effects by increasing the expression of Bcl-2 and Bcl-xl proteins and reduction of Bax and Bad protein expression. The pharmaceutical managed to decrease concentrations of proapoptotic factors, such as NF-κB (Nuclear Factor-kappa β), ICAM-1 (Intercellular Adhesion Molecule 1), caspase-3, and reduced the level of TUNEL-positive cells. Liraglutide was able to reduce the level of free radicals by decreasing the level of malondialdehyde (MDA), and increasing the superoxide dismutase level (SOD), glutathione (GSH), and catalase. Liraglutide may affect the neurovascular unit causing its remodeling, which seems to be crucial for recovery after stroke. Liraglutide may stabilize atherosclerotic plaque, as well as counteract its early formation and further development. Liraglutide, through its binding to GLP-1R (glucagon like peptide-1 receptor) and consequent activation of PI3K/MAPK (Phosphoinositide 3-kinase/mitogen associated protein kinase) dependent pathways, may have a positive impact on Aβ (amyloid beta) trafficking and clearance by increasing the presence of Aβ transporters in cerebrospinal fluid. Liraglutide seems to affect tau pathology. It is possible that liraglutide may have some stem cell stimulating properties. The effects may be connected with PKA (phosphorylase kinase A) activation. This paper presents potential mechanisms of liraglutide activity in conditions connected with neuronal damage, with special emphasis on Alzheimer's disease and cerebral ischemia.
Collapse
|
20
|
The Protective and Restorative Effects of Growth Hormone and Insulin-Like Growth Factor-1 on Methadone-Induced Toxicity In Vitro. Int J Mol Sci 2018; 19:ijms19113627. [PMID: 30453639 PMCID: PMC6274959 DOI: 10.3390/ijms19113627] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 11/14/2018] [Accepted: 11/15/2018] [Indexed: 11/17/2022] Open
Abstract
Evidence to date suggests that opioids such as methadone may be associated with cognitive impairment. Growth hormone (GH) and insulin-like growth factor-1 (IGF-1) are suggested to be neuroprotective and procognitive in the brain and may therefore counteract these effects. This study aims to explore the protective and restorative effects of GH and IGF-1 in methadone-treated cell cultures. Primary cortical cell cultures were harvested from rat fetuses and grown for seven days in vitro. To examine the protective effects, methadone was co-treated with or without GH or IGF-1 for three consecutive days. To examine the restorative effects, methadone was added for the first 24 h, washed, and later treated with GH or IGF-1 for 48 h. At the end of each experiment, mitochondrial function and membrane integrity were evaluated. The results revealed that GH had protective effects in the membrane integrity assay and that both GH and IGF-1 effectively recovered mitochondrial function and membrane integrity in cells pretreated with methadone. The overall conclusion of the present study is that GH, but not IGF-1, protects primary cortical cells against methadone-induced toxicity, and that both GH and IGF-1 have a restorative effect on cells pretreated with methadone.
Collapse
|
21
|
Hung CL, Tseng JW, Chao HH, Hung TM, Wang HS. Effect of Acute Exercise Mode on Serum Brain-Derived Neurotrophic Factor (BDNF) and Task Switching Performance. J Clin Med 2018; 7:jcm7100301. [PMID: 30249981 PMCID: PMC6209934 DOI: 10.3390/jcm7100301] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 09/17/2018] [Accepted: 09/21/2018] [Indexed: 12/04/2022] Open
Abstract
Previous studies have consistently reported a positive effect of acute exercise on cognition, particularly on executive function. However, most studies have focused on aerobic and resistant forms of exercise. The purpose of this study was to compare the effect of ‘open-skill’ with ‘closed-skill’ exercise (defined in terms of the predictability of the performing environment) on brain-derived neurotrophic factor (BDNF) production and task switching performance. Twenty young adult males participated in both closed (running) and open (badminton) skill exercise sessions in a counterbalanced order on separate days. The exercise sessions consisted of 5 min of warm up exercises followed by 30 min of running or badminton. The exercise intensity was set at 60% (±5%) of the heart rate reserve level (HRR) with HR being monitored by a wireless heart rate monitor. Blood samples were taken and participation in a task-switching paradigm occurred before and after each exercise session. Results showed no differences in serum BDNF or task-switching performance at the pre-test stage, however, badminton exercise resulted in significantly higher serum BDNF levels (a proxy for levels of BDNF in the brain) and near significant smaller global switching costs relative to running. This study has provided preliminary evidence in support the relative benefits of open-skills exercises on BDNF and executive function.
Collapse
Affiliation(s)
- Chiao-Ling Hung
- Department of Athletic, National Taiwan University, Taipei 10617, Taiwan.
| | - Jun-Wei Tseng
- Department of Physical Education, National Taiwan Normal University, Taipei 10646, Taiwan.
| | - Hsiao-Han Chao
- Department of Athletic, National Taiwan University, Taipei 10617, Taiwan.
| | - Tsung-Min Hung
- Department of Physical Education, National Taiwan Normal University, Taipei 10646, Taiwan.
| | - Ho-Seng Wang
- Department of Physical Education, National Taiwan Normal University, Taipei 10646, Taiwan.
| |
Collapse
|
22
|
Santi A, Bot M, Aleman A, Penninx BWJH, Aleman IT. Circulating insulin-like growth factor I modulates mood and is a biomarker of vulnerability to stress: from mouse to man. Transl Psychiatry 2018; 8:142. [PMID: 30068974 PMCID: PMC6070549 DOI: 10.1038/s41398-018-0196-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 05/11/2018] [Accepted: 06/08/2018] [Indexed: 12/30/2022] Open
Abstract
Individual susceptibility to anxiety disorders after maladaptive responses to stress is not well understood. We now report that while exploring stress responses in mice after traumatic brain injury (TBI), a condition associated to stress susceptibility, we observed that the anxiogenic effects of either TBI or exposure to life-threatening experiences (predator) were blocked when both stressors were combined. Because TBI increases the entrance into the brain of serum insulin-like growth factor I (IGF-I), a known modulator of anxiety with a wide range of concentrations in the human population, we then determined whether circulating IGF-I is related to anxiety measures. In mice, anxiety-like responses to predator were inversely related to circulating IGF-I levels. Other indicators of mood regulation such as sensitivity to dexamethasone suppression and expression levels of blood and brain FK506 binding protein 5 (FKBP5), a co-chaperone of the glucocorticoid receptor that regulates its activity, were also associated to circulating IGF-I. Indeed, brain FKBP5 expression in mice was stimulated by IGF-I. In addition, we observed in a large human cohort (n = 2686) a significant relationship between plasma IGF-I and exposure to recent stressful life events, while FKBP5 expression in blood cells was significantly associated to plasma IGF-I levels. Collectively, these data indicate that circulating IGF-I appears to be involved in mood homeostasis across different species. Furthermore, the data in mice allow us to indicate that IGF-I may be acting at least in part by modulating FKBP5 expression.
Collapse
Affiliation(s)
- A. Santi
- 0000 0001 2177 5516grid.419043.bCajal Institute, Madrid, Spain ,0000 0000 9314 1427grid.413448.eCiberned, Madrid, Spain
| | - M. Bot
- grid.484519.5Department of Psychiatry, VU University Medical Center and GGZ inGeest, Amsterdam Public Health Research Institute, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - A. Aleman
- 0000 0000 9558 4598grid.4494.dDepartment of Neuroscience, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - B. W. J. H. Penninx
- grid.484519.5Department of Psychiatry, VU University Medical Center and GGZ inGeest, Amsterdam Public Health Research Institute, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - I. Torres Aleman
- 0000 0001 2177 5516grid.419043.bCajal Institute, Madrid, Spain ,0000 0000 9314 1427grid.413448.eCiberned, Madrid, Spain
| |
Collapse
|
23
|
miR-129 controls axonal regeneration via regulating insulin-like growth factor-1 in peripheral nerve injury. Cell Death Dis 2018; 9:720. [PMID: 29915198 PMCID: PMC6006361 DOI: 10.1038/s41419-018-0760-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 05/10/2018] [Accepted: 06/04/2018] [Indexed: 01/08/2023]
Abstract
The microenvironment of peripheral nerve regeneration consists of multiple neurotrophic factors, adhesion molecules, and extracellular matrix molecules, secreted by unique glial cells in the peripheral nerve system (PNS)-Schwann cell (SCs). Following peripheral nerve injury (PNI), local IGF-1 production is upregulated in SCs and denervated muscle during axonal sprouting and regeneration. Regulation of IGF-1/IGF-1R signaling is considered as a potentially targeted therapy of PNI. We previously identified a group of novel miRNAs in proximal nerve following rat sciatic nerve transection. The present work focused on the role of miR-129 in regulation of IGF-1 signaling after sciatic nerve injury. The temporal change profile of the miR-129 expression was negatively correlated with the IGF-1 expression in proximal nerve stump and dorsal root ganglion (DRG) following sciatic nerve transection. An increased expression of miR-129 inhibited proliferation and migration of SCs, and axonal outgrowth of DRG neurons, which was inversely promoted by silencing of the miR-129 expression. The IGF-1 was identified as one of the multiple target genes of miR-129, which exerted negative regulation of IGF-1 by translational suppression. Moreover, knockdown of IGF-1 attenuated the promoting effects of miR-129 inhibitor on proliferation and migration of SCs, and neurite outgrowth of DRG neurons. Overall, our data indicated that miR-129 own the potential to regulate the proliferation and migration of SCs by targeting IGF-1, providing further insight into the regulatory role of miRNAs in peripheral nerve regeneration. The present work not only provides new insight into miR-129 regulation of peripheral nerve regeneration by robust phenotypic modulation of neural cells, but also opens a novel therapeutic window for PNI by mediating IGF-1 production. Our results may provide further experimental basis for translation of the molecular therapy into the clinic.
Collapse
|
24
|
Systemic IGF-1 gene delivery by rAAV9 improves spontaneous autoimmune peripheral polyneuropathy (SAPP). Sci Rep 2018; 8:5408. [PMID: 29615658 PMCID: PMC5883061 DOI: 10.1038/s41598-018-23607-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 03/13/2018] [Indexed: 02/01/2023] Open
Abstract
Spontaneous autoimmune peripheral polyneuropathy (SAPP) is a mouse model of chronic inflammatory demyelinating polyradiculoneuropathy (CIDP) in non-obese diabetic (NOD) mice null for costimulatory molecule, B7-2 gene (B7-2−/−). SAPP is a chronic progressive and multifocal inflammatory and demyelinating polyneuropathy of spontaneous onset with secondary axonal degeneration. Insulin-like growth factor 1(IGF-1) is a pleiotropic factor with neuroprotective, regenerative, and anti-inflammatory effects with extensive experience in its preclinical and clinical use. Systemic delivery of recombinant adeno-associated virus serotype 9 (rAAV9) provides robust and widespread gene transfer to central and peripheral nervous systems making it suitable for gene delivery in neurological diseases. A significant proportion of patients with inflammatory neuropathies like CIDP do not respond to current clinical therapies and there is a need for new treatments. In this study, we examined the efficacy IGF-1 gene therapy by systemic delivery with rAAV9 in SAPP model. The rAAV9 construct also contained a reporter gene to monitor the surrogate expression of IGF-1. We found significant improvement in neuropathic disease after systemic delivery of rAAV9/IGF-1 gene at presymptomatic and symptomatic stages of SAPP model. These findings support that IGF-1 treatment (including gene therapy) is a viable therapeutic option in immune neuropathies such as CIDP.
Collapse
|
25
|
Towards a multi protein and mRNA expression of biological predictive and distinguish model for post stroke depression. Oncotarget 2018; 7:54329-54338. [PMID: 27527872 PMCID: PMC5342345 DOI: 10.18632/oncotarget.11105] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 07/19/2016] [Indexed: 01/19/2023] Open
Abstract
Previous studies suggest that neurotrophic factors participate in the development of stroke and depression. So we investigated the utility of these biomarkers as predictive and distinguish model for post stroke depression (PSD). 159 individuals including PSD, stroke without depression (Non-PSD), major depressive disorder (MDD) and normal control groups were recruited and examined the protein and mRNA expression levels of vascular endothelial growth factor (VEGF), vascular endothelial growth factor receptors (VEGFR2), placental growth factor (PIGF), insulin-like growth factor (IGF-1) and insulin-like growth factor receptors (IGF-1R). The chi-square test was used to evaluate categorical variable, while nonparametric test and one-way analysis of variance were applied to continuous variables of general characteristics, clinical and biological changes. In order to explore the predictive and distinguish role of these factors in PSD, discriminant analysis and receiver operating characteristic curve were calculated. The four groups had statistical differences in these neurotrophic factors (all P < 0.05) except VEGF concentration and IGF-1R mRNA (P = 0.776, P = 0.102 respectively). We identified these mRNA expression and protein analytes with general predictive performance for PSD and Non-PSD groups [area under the curve (AUC): 0.805, 95% CI, 0.704-0.907, P < 0.001]. Importantly, there is an excellent predictive performance (AUC: 0.984, 95% CI, 0.964-1.000, P < 0.001) to differentiate PSD patients from MDD patients. This was the first study to explore the changes of neurotrophic factors family in PSD patients, the results intriguingly demonstrated that the combination of protein and mRNA expression of biological factors could use as a predictive and discriminant model for PSD.
Collapse
|
26
|
Arellanes-Licea EC, Ávila-Mendoza J, Ramírez-Martínez EC, Ramos E, Uribe-González N, Arámburo C, Morales T, Luna M. Upregulation of GH, but not IGF1, in the hippocampus of the lactating dam after kainic acid injury. Endocr Connect 2018; 7:258-267. [PMID: 29321175 PMCID: PMC5812059 DOI: 10.1530/ec-17-0380] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Accepted: 01/10/2018] [Indexed: 12/21/2022]
Abstract
Lactation embodies a natural model of morphological, neurochemical, and functional brain plasticity. In this reproductive stage, the hippocampus of the female is less sensitive to excitotoxins in contrast to nulliparity. Growth hormone (GH) and insulin-like growth factor 1 (IGF1) are known to be neuroprotective in several experimental models of brain lesion. Here, activation of the GH-IGF1 pituitary-brain axis following kainic acid (7.5 mg/kg i.p. KA) lesion was studied in lactating and nulliparous rats. Serum concentrations of GH and IGF1 were uncoupled in lactation. Compared to virgin rats, the basal concentration of GH increased up to 40% but IGF1 decreased 58% in dams, and only GH increased further after KA treatment. In the hippocampus, basal expression of GH mRNA was higher (2.8-fold) in lactating rats than in virgin rats. GH mRNA expression in lactating rats increased further after KA administration in the hippocampus and in the hypothalamus, in parallel to GH protein concentration in the hippocampus of KA-treated lactating rats (43% vs lactating control), as detected by Western blot and immunofluorescence. Except for the significantly lower mRNA concentration in the liver of lactating rats, IGF1 expression was not altered by the reproductive condition or by KA treatment in the hippocampus and hypothalamus. Present results indicate upregulation of GH expression in the hippocampus after an excitotoxic lesion, suggesting paracrine/autocrine actions of GH as a factor underlying neuroprotection in the brain of the lactating dam. Since no induction of IGF1 was detected, present data suggest a direct action of GH.
Collapse
Affiliation(s)
- Elvira C Arellanes-Licea
- Neurobiología Celular y MolecularInstituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro, México
| | - José Ávila-Mendoza
- Neurobiología Celular y MolecularInstituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro, México
| | - Elizabeth C Ramírez-Martínez
- Neurobiología Celular y MolecularInstituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro, México
| | - Eugenia Ramos
- Neurobiología Celular y MolecularInstituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro, México
| | - Nancy Uribe-González
- Neurobiología Celular y MolecularInstituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro, México
| | - Carlos Arámburo
- Neurobiología Celular y MolecularInstituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro, México
| | - Teresa Morales
- Neurobiología Celular y MolecularInstituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro, México
| | - Maricela Luna
- Neurobiología Celular y MolecularInstituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro, México
| |
Collapse
|
27
|
Brain insulin resistance in type 2 diabetes and Alzheimer disease: concepts and conundrums. Nat Rev Neurol 2018; 14:168-181. [PMID: 29377010 DOI: 10.1038/nrneurol.2017.185] [Citation(s) in RCA: 885] [Impact Index Per Article: 147.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Considerable overlap has been identified in the risk factors, comorbidities and putative pathophysiological mechanisms of Alzheimer disease and related dementias (ADRDs) and type 2 diabetes mellitus (T2DM), two of the most pressing epidemics of our time. Much is known about the biology of each condition, but whether T2DM and ADRDs are parallel phenomena arising from coincidental roots in ageing or synergistic diseases linked by vicious pathophysiological cycles remains unclear. Insulin resistance is a core feature of T2DM and is emerging as a potentially important feature of ADRDs. Here, we review key observations and experimental data on insulin signalling in the brain, highlighting its actions in neurons and glia. In addition, we define the concept of 'brain insulin resistance' and review the growing, although still inconsistent, literature concerning cognitive impairment and neuropathological abnormalities in T2DM, obesity and insulin resistance. Lastly, we review evidence of intrinsic brain insulin resistance in ADRDs. By expanding our understanding of the overlapping mechanisms of these conditions, we hope to accelerate the rational development of preventive, disease-modifying and symptomatic treatments for cognitive dysfunction in T2DM and ADRDs alike.
Collapse
|
28
|
Rodriguez-Perez AI, Borrajo A, Diaz-Ruiz C, Garrido-Gil P, Labandeira-Garcia JL. Crosstalk between insulin-like growth factor-1 and angiotensin-II in dopaminergic neurons and glial cells: role in neuroinflammation and aging. Oncotarget 2017; 7:30049-67. [PMID: 27167199 PMCID: PMC5058663 DOI: 10.18632/oncotarget.9174] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 04/19/2016] [Indexed: 01/06/2023] Open
Abstract
The local renin-angiotensin system (RAS) and insulin-like growth factor 1 (IGF-1) have been involved in longevity, neurodegeneration and aging-related dopaminergic degeneration. However, it is not known whether IGF-1 and angiotensin-II (AII) activate each other. In the present study, AII, via type 1 (AT1) receptors, exacerbated neuroinflammation and dopaminergic cell death. AII, via AT1 receptors, also increased the levels of IGF-1 and IGF-1 receptors in microglial cells. IGF-1 inhibited RAS activity in dopaminergic neurons and glial cells, and also inhibited the AII-induced increase in markers of the M1 microglial phenotype. Consistent with this, IGF-1 decreased dopaminergic neuron death induced by the neurotoxin MPP+ both in the presence and in the absence of glia. Intraventricular administration of AII to young rats induced a significant increase in IGF-1 expression in the nigral region. However, aged rats showed decreased levels of IGF-1 relative to young controls, even though RAS activity is known to be enhanced in aged animals. The study findings show that IGF-1 and the local RAS interact to inhibit or activate neuroinflammation (i.e. transition from the M1 to the M2 phenotype), oxidative stress and dopaminergic degeneration. The findings also show that this mechanism is impaired in aged animals.
Collapse
Affiliation(s)
- Ana I Rodriguez-Perez
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Ana Borrajo
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Carmen Diaz-Ruiz
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Pablo Garrido-Gil
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Jose L Labandeira-Garcia
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| |
Collapse
|
29
|
García-Marchena N, Silva-Peña D, Martín-Velasco AI, Villanúa MÁ, Araos P, Pedraz M, Maza-Quiroga R, Romero-Sanchiz P, Rubio G, Castilla-Ortega E, Suárez J, Rodríguez de Fonseca F, Serrano A, Pavón FJ. Decreased plasma concentrations of BDNF and IGF-1 in abstinent patients with alcohol use disorders. PLoS One 2017; 12:e0187634. [PMID: 29108028 PMCID: PMC5673472 DOI: 10.1371/journal.pone.0187634] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 10/23/2017] [Indexed: 12/27/2022] Open
Abstract
The identification of growth factors as potential biomarkers in alcohol addiction may help to understand underlying mechanisms associated with the pathogenesis of alcohol use disorders (AUDs). Previous studies have linked growth factors to neural plasticity in neurocognitive impairment and mental disorders. In order to further clarify the impact of chronic alcohol consumption on circulating growth factors, a cross-sectional study was performed in abstinent AUD patients (alcohol group, N = 91) and healthy control subjects (control group, N = 55) to examine plasma concentrations of brain-derived neurotrophic factor (BDNF), insulin-like growth factor-1 (IGF-1) and IGF-1 binding protein-3 (IGFBP-3). The association of these plasma peptides with relevant AUD-related variables and psychiatric comorbidity was explored. The alcohol group was diagnosed with severe AUD and showed an average of 13 years of problematic use and 10 months of abstinence at the moment of participating in the study. Regarding common medical conditions associated with AUD, we observed an elevated incidence of alcohol-induced liver and pancreas diseases (18.7%) and psychiatric comorbidity (76.9%). Thus, AUD patients displayed a high prevalence of dual diagnosis (39.3%) [mainly depression (19.9%)] and comorbid substance use disorders (40.7%). Plasma BDNF and IGF-1 concentrations were significantly lower in the alcohol group than in the control group (p<0.001). Remarkably, there was a negative association between IGF-1 concentrations and age in the control group (r = -0.52, p<0.001) that was not found in the alcohol group. Concerning AUD-related variables, AUD patients with liver and pancreas diseases showed even lower concentrations of BDNF (p<0.05). In contrast, the changes in plasma concentrations of these peptides were not associated with abstinence, problematic use, AUD severity or lifetime psychiatric comorbidity. These results suggest that further research is necessary to elucidate the role of BDNF in alcohol-induced toxicity and the biological significance of the lack of correlation between age and plasma IGF-1 levels in abstinent AUD patients.
Collapse
Affiliation(s)
- Nuria García-Marchena
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain
| | - Daniel Silva-Peña
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain
| | | | - María Ángeles Villanúa
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Pedro Araos
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain
| | - María Pedraz
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain
| | - Rosa Maza-Quiroga
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain
| | - Pablo Romero-Sanchiz
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain
| | - Gabriel Rubio
- Instituto i+12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Estela Castilla-Ortega
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain
| | - Juan Suárez
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain
| | - Fernando Rodríguez de Fonseca
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain
- * E-mail: (FRF); (AS); (FJP)
| | - Antonia Serrano
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain
- * E-mail: (FRF); (AS); (FJP)
| | - Francisco Javier Pavón
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain
- * E-mail: (FRF); (AS); (FJP)
| |
Collapse
|
30
|
Picillo M, Pivonello R, Santangelo G, Pivonello C, Savastano R, Auriemma R, Amboni M, Scannapieco S, Pierro A, Colao A, Barone P, Pellecchia MT. Serum IGF-1 is associated with cognitive functions in early, drug-naïve Parkinson's disease. PLoS One 2017; 12:e0186508. [PMID: 29065116 PMCID: PMC5655531 DOI: 10.1371/journal.pone.0186508] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 10/03/2017] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Cognitive deficits are common in Parkinson's disease (PD) since the early stages and many patients eventually develop dementia. Yet, occurrence of dementia in PD is unpredictable. Evidence supports the hypothesis that insulin-like growth factor-1 (IGF-1) is involved in cognitive deficits. Our aim was to evaluate the relationship between serum IGF-1 levels and neuropsychological scores in a large cohort of drug-naïve PD patients during the earliest stages of the disease. METHODS Serum IGF-1 levels were determined in 405 early, drug-naïve PD patients and 191 healthy controls (HC) enrolled in the Parkinson's Progression Markers Initiative (PPMI). The association between serum IGF-1 levels and neuropsychological scores was evaluated with linear regression analysis. RESULTS IGF-1 levels were similar in PD and HC. In PD patients the lowest IGF-1 quartile was a predictor of lower performances at the Semantic Fluency task (β = -3.46, 95%CI: -5.87 to -1.01, p = 0.005), the Symbol Digit Modalities Score (β = -2.09, 95%CI: -4.02 to -0.15, p = 0.034), and Hopkins Verbal Learning Test Retention (β = -0.05, 95%CI: -0.09 to -0.009, p = 0.019). CONCLUSIONS Lower serum IGF-1 levels are associated to poor performances in cognitive tasks assessing executive function, attention and verbal memory in a large cohort of early PD patients. Follow-up studies are warranted to assess if IGF-1 is related to the development of dementia in PD.
Collapse
Affiliation(s)
- Marina Picillo
- Center for Neurodegenerative Diseases (CEMAND), Department of Medicine and Surgery, Neuroscience Section, University of Salerno, Salerno, Italy
| | - Rosario Pivonello
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Gabriella Santangelo
- Neuropsychology Laboratory, Department of Psychology, Second University of Naples, Caserta, Italy
| | - Claudia Pivonello
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Riccardo Savastano
- Center for Neurodegenerative Diseases (CEMAND), Department of Medicine and Surgery, Neuroscience Section, University of Salerno, Salerno, Italy
| | - Renata Auriemma
- IOS and Coleman Medicina Futura Medical Center, Naples, Italy
| | - Marianna Amboni
- Center for Neurodegenerative Diseases (CEMAND), Department of Medicine and Surgery, Neuroscience Section, University of Salerno, Salerno, Italy
- IDC Hermitage-Capodimonte, Naples, Italy
| | - Sara Scannapieco
- Center for Neurodegenerative Diseases (CEMAND), Department of Medicine and Surgery, Neuroscience Section, University of Salerno, Salerno, Italy
| | - Angela Pierro
- Center for Neurodegenerative Diseases (CEMAND), Department of Medicine and Surgery, Neuroscience Section, University of Salerno, Salerno, Italy
| | - Annamaria Colao
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Paolo Barone
- Center for Neurodegenerative Diseases (CEMAND), Department of Medicine and Surgery, Neuroscience Section, University of Salerno, Salerno, Italy
| | - Maria Teresa Pellecchia
- Center for Neurodegenerative Diseases (CEMAND), Department of Medicine and Surgery, Neuroscience Section, University of Salerno, Salerno, Italy
- * E-mail:
| |
Collapse
|
31
|
Tien LT, Lee YJ, Pang Y, Lu S, Lee JW, Tseng CH, Bhatt AJ, Savich RD, Fan LW. Neuroprotective Effects of Intranasal IGF-1 against Neonatal Lipopolysaccharide-Induced Neurobehavioral Deficits and Neuronal Inflammation in the Substantia Nigra and Locus Coeruleus of Juvenile Rats. Dev Neurosci 2017; 39:443-459. [PMID: 28787734 PMCID: PMC5799046 DOI: 10.1159/000477898] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 05/30/2017] [Indexed: 01/29/2023] Open
Abstract
Neonatal lipopolysaccharide (LPS) exposure-induced brain inflammation resulted in motor dysfunction and brain dopaminergic neuronal injury, and increased the risks of neurodegenerative disorders in adult rats. Our previous studies showed that intranasal administration of insulin-like growth factor-1 (IGF-1) protects against LPS-induced white matter injury in the developing rat brain. To further examine whether IGF-1 protects against LPS-induced brain neuronal injury and neurobehavioral dysfunction, recombinant human IGF-1 (rhIGF-1) at a dose of 50 µg/pup was administered intranasally 1 h following intracerebral injection of LPS (1 mg/kg) in postnatal day 5 (P5) Sprague-Dawley rat pups. Neurobehavioral tests were carried out from P7 to P21, and brain neuronal injury was examined at P21. Our results showed that LPS exposure resulted in disturbances of motor behaviors in juvenile rats. Moreover, LPS exposure caused injury to central catecholaminergic neurons, as indicated by a reduction of tyrosine hydroxylase (TH) immunoreactivity in the substantia nigra (SN), ventral tegmental area (VTA) and olfactory bulb (OB), and brain noradrenergic neurons, as indicated by a reduction of TH immunoreactivity in the locus coeruleus (LC) of the P21 rat brain. The LPS-induced reduction of TH+ cells was observed at a greater degree in the SN and LC of the P21 rat brain. Intranasal rhIGF-1 treatment attenuated LPS-induced central catecholaminergic neuronal injury and motor behavioral disturbances, including locomotion, beam walking test and gait analysis. Intranasal rhIGF-1 administration also attenuated LPS-induced elevation of IL-1β levels and numbers of activated microglia, and cyclooxygenase-2+ cells, which were double labeled with TH+ cells in the SN, VTA, OB and LC of the P21 rat brain. These results suggest that IGF-1 may provide protection against neonatal LPS exposure-induced central catecholaminergic neuronal injury and motor behavioral disturbances, and that the protective effects are associated with the inhibition of microglia activation and the reduction of neuronal oxidative stress by the suppression of the neuronal cyclooxygenase-2 expression.
Collapse
Affiliation(s)
- Lu-Tai Tien
- School of Medicine, Fu Jen Catholic University, Xinzhuang Dist, New Taipei City 24205, Taiwan
| | - Yih-Jing Lee
- School of Medicine, Fu Jen Catholic University, Xinzhuang Dist, New Taipei City 24205, Taiwan
| | - Yi Pang
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Silu Lu
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Jonathan W Lee
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Chih-Hsueh Tseng
- School of Medicine, Fu Jen Catholic University, Xinzhuang Dist, New Taipei City 24205, Taiwan
| | - Abhay J Bhatt
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Renate D Savich
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Lir-Wan Fan
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
32
|
Abstract
Both Alzheimer's disease (AD) and type 2 diabetes mellitus (DM) are two common
forms of disease worldwide and many studies indicate that people with diabetes,
especially DM, are at higher risk of developing AD. AD is characterized by
progressive cognitive decline and accumulation of β-amyloid (Aβ)
forming senile plaques. DM is a metabolic disorder characterized by
hyperglycemia in the context of insulin resistance and relative lack of insulin.
Both diseases also share common characteristics such as loss of cognitive
function and inflammation. Inflammation resulting from Aβ further induces
production of Aβ1-42 peptides. Inflammation due to
overnutrition induces insulin resistance and consequently DM. Memory deficit and
a decrease in GLUT4 and hippocampal insulin signaling have been observed in
animal models of insulin resistance. The objective of this review was to show
the shared characteristics of AD and DM.
Collapse
Affiliation(s)
- Aparecida Marcelino de Nazareth
- Physiotherapist, Specialist in Neurofunctional Physical Therapy, Master of Neurosciences from the (UFSC), SC, Brazil, and PhD in Sciences (Pharmacology and Medicinal Chemistry) from the Federal University of Rio de Janeiro (UFRJ), RJ, Brazil
| |
Collapse
|
33
|
Huffman J, Hoffmann C, Taylor GT. Integrating insulin-like growth factor 1 and sex hormones into neuroprotection: Implications for diabetes. World J Diabetes 2017; 8:45-55. [PMID: 28265342 PMCID: PMC5320748 DOI: 10.4239/wjd.v8.i2.45] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 09/24/2016] [Accepted: 11/22/2016] [Indexed: 02/05/2023] Open
Abstract
Brain integrity and cognitive aptitude are often impaired in patients with diabetes mellitus, presumably a result of the metabolic complications inherent to the disease. However, an increasing body of evidence has demonstrated the central role of insulin-like growth factor 1 (IGF1) and its relation to sex hormones in many neuroprotective processes. Both male and female patients with diabetes display abnormal IGF1 and sex-hormone levels but the comparison of these fluctuations is seldom a topic of interest. It is interesting to note that both IGF1 and sex hormones have the ability to regulate phosphoinositide 3-kinase-Akt and mitogen-activated protein kinases-extracellular signal-related kinase signaling cascades in animal and cell culture models of neuroprotection. Additionally, there is considerable evidence demonstrating the neuroprotective coupling of IGF1 and estrogen. Androgens have also been implicated in many neuroprotective processes that operate on similar signaling cascades as the estrogen-IGF1 relation. Yet, androgens have not been directly linked to the brain IGF1 system and neuroprotection. Despite the sex-specific variations in brain integrity and hormone levels observed in diabetic patients, the IGF1-sex hormone relation in neuroprotection has yet to be fully substantiated in experimental models of diabetes. Taken together, there is a clear need for the comprehensive analysis of sex differences on brain integrity of diabetic patients and the relationship between IGF1 and sex hormones that may influence brain-health outcomes. As such, this review will briefly outline the basic relation of diabetes and IGF1 and its role in neuroprotection. We will also consider the findings on sex hormones and diabetes as a basis for separately analyzing males and females to identify possible hormone-induced brain abnormalities. Finally, we will introduce the neuroprotective interplay of IGF1 and estrogen and how androgen-derived neuroprotection operates through similar signaling cascades. Future research on both neuroprotection and diabetes should include androgens into the interplay of IGF1 and sex hormones.
Collapse
|
34
|
Levada OA, Troyan AS. Insulin-like growth factor-1: a possible marker for emotional and cognitive disturbances, and treatment effectiveness in major depressive disorder. Ann Gen Psychiatry 2017; 16:38. [PMID: 29093741 PMCID: PMC5659027 DOI: 10.1186/s12991-017-0161-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 10/20/2017] [Indexed: 02/08/2023] Open
Abstract
Depression and cognitive dysfunction share a common neuropathological platform. Abnormal neural plasticity in the frontolimbic circuits has been linked to changes in the expression of neurotrophic factors, including IGF-1. These changes may result in clinical abnormalities observed over the course of major depressive disorder (MDD), including cognitive dysfunction. The present review aimed to summarize evidence regarding abnormalities of peripheral IGF-1 in MDD patients and assess a marker and predictive role of the neurotrophin for emotional and cognitive disturbances, and treatment effectiveness. A literature search of the PubMed database was conducted for studies, in which peripheral IGF-1 levels were evaluated. Our analysis revealed four main findings: (1) IGF-1 levels in MDD patients mismatch across the studies, which may arise from various factors, e.g., age, gender, the course of the disease, presence of cognitive impairment, ongoing therapy, or general health conditions; (2) the initial peripheral IGF-1 levels may predict the occurrence of depression in future; (3) peripheral IGF-1 levels may reflect cognitive dysfunction, although the data is limited; (4) it is difficult to evaluate the influence of treatment on IGF-1 levels as there is discrepancy of this growth factor among the studies at baseline, although most of them showed a decrease in IGF-1 levels after treatment.
Collapse
Affiliation(s)
- Oleg A Levada
- State Institution "Zaporizhzhia Medical Academy of Postgraduate Education Ministry of Health of Ukraine", 20 Winter boulevard, Zaporizhzhia, 69096 Ukraine
| | - Alexandra S Troyan
- State Institution "Zaporizhzhia Medical Academy of Postgraduate Education Ministry of Health of Ukraine", 20 Winter boulevard, Zaporizhzhia, 69096 Ukraine
| |
Collapse
|
35
|
Salem L, Saleh N, Désaméricq G, Youssov K, Dolbeau G, Cleret L, Bourhis ML, Azulay JP, Krystkowiak P, Verny C, Morin F, Moutereau S, Bachoud-Lévi AC, Maison P. Insulin-Like Growth Factor-1 but Not Insulin Predicts Cognitive Decline in Huntington's Disease. PLoS One 2016; 11:e0162890. [PMID: 27627435 PMCID: PMC5023180 DOI: 10.1371/journal.pone.0162890] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 08/30/2016] [Indexed: 01/05/2023] Open
Abstract
Background Huntington's disease (HD) is one of several neurodegenerative disorders that have been associated with metabolic alterations. Changes in Insulin Growth Factor 1 (IGF-1) and/or insulin input to the brain may underlie or contribute to the progress of neurodegenerative processes. Here, we investigated the association over time between changes in plasma levels of IGF-1 and insulin and the cognitive decline in HD patients. Methods We conducted a multicentric cohort study in 156 patients with genetically documented HD aged from 22 to 80 years. Among them, 146 patients were assessed at least twice with a follow-up of 3.5 ± 1.8 years. We assessed their cognitive decline using the Unified Huntington’s Disease Rating Scale, and their IGF-1 and insulin plasmatic levels, at baseline and once a year during the follow-up. Associations were evaluated using a mixed-effect linear model. Results In the cross-sectional analysis at baseline, higher levels of IGF-1 and insulin were associated with lower cognitive scores and thus with a higher degree of cognitive impairment. In the longitudinal analysis, the decrease of all cognitive scores, except the Stroop interference, was associated with the IGF-1 level over time but not of insulin. Conclusions IGF-1 levels, unlike insulin, predict the decline of cognitive function in HD.
Collapse
Affiliation(s)
- Linda Salem
- Université Paris Est, Faculté de médecine, Créteil, France
- Inserm, U955, Equipe 01, Neuropsychologie interventionnelle, Créteil, France
- Ecole Normale Supérieure, Département d'études Cognitives, Paris, France
- AP-HP, Hôpital H. Mondor- A. Chenevier, Centre de référence maladie de Huntington, Neurologie cognitive, Créteil, France
| | - Nadine Saleh
- Université Paris Est, Faculté de médecine, Créteil, France
- Inserm, U955, Equipe 01, Neuropsychologie interventionnelle, Créteil, France
- Ecole Normale Supérieure, Département d'études Cognitives, Paris, France
- AP-HP, Hôpital H. Mondor- A. Chenevier, Centre de référence maladie de Huntington, Neurologie cognitive, Créteil, France
| | - Gaelle Désaméricq
- Université Paris Est, Faculté de médecine, Créteil, France
- Inserm, U955, Equipe 01, Neuropsychologie interventionnelle, Créteil, France
- Ecole Normale Supérieure, Département d'études Cognitives, Paris, France
- AP-HP, Hôpital H. Mondor- A. Chenevier, Centre de référence maladie de Huntington, Neurologie cognitive, Créteil, France
| | - Katia Youssov
- Université Paris Est, Faculté de médecine, Créteil, France
- Inserm, U955, Equipe 01, Neuropsychologie interventionnelle, Créteil, France
- Ecole Normale Supérieure, Département d'études Cognitives, Paris, France
- AP-HP, Hôpital H. Mondor- A. Chenevier, Centre de référence maladie de Huntington, Neurologie cognitive, Créteil, France
| | - Guillaume Dolbeau
- Université Paris Est, Faculté de médecine, Créteil, France
- Inserm, U955, Equipe 01, Neuropsychologie interventionnelle, Créteil, France
- AP-HP, Hôpital H. Mondor- A. Chenevier, Unité de recherche clinique, Créteil, France
| | - Laurent Cleret
- Université Paris Est, Faculté de médecine, Créteil, France
- Inserm, U955, Equipe 01, Neuropsychologie interventionnelle, Créteil, France
- Ecole Normale Supérieure, Département d'études Cognitives, Paris, France
- AP-HP, Hôpital H. Mondor- A. Chenevier, Centre de référence maladie de Huntington, Neurologie cognitive, Créteil, France
| | - Marie-Laure Bourhis
- AP-HP, Hôpital H. Mondor- A. Chenevier, Unité de recherche clinique, Créteil, France
| | - Jean-Philippe Azulay
- Hôpital de la Timone, Service de Neurologie et pathologie du mouvement, Marseille, France
| | | | - Christophe Verny
- CHU of Angers, Centre de référence des maladies neurogénétiques, service de neurologie, Angers, France
| | - Françoise Morin
- AP-HP-GHU NORD, Hôpital Avicenne, Etablissement Français du sang, Bobigny, France
| | - Stéphane Moutereau
- AP-HP, Hôpital H. Mondor- A. Chenevier, Département de Biochimie-Pharmaco-Toxicologie, Créteil, France
| | | | - Anne-Catherine Bachoud-Lévi
- Université Paris Est, Faculté de médecine, Créteil, France
- Inserm, U955, Equipe 01, Neuropsychologie interventionnelle, Créteil, France
- Ecole Normale Supérieure, Département d'études Cognitives, Paris, France
- AP-HP, Hôpital H. Mondor- A. Chenevier, Centre de référence maladie de Huntington, Neurologie cognitive, Créteil, France
- * E-mail:
| | - Patrick Maison
- Université Paris Est, Faculté de médecine, Créteil, France
- Inserm, U955, Equipe 01, Neuropsychologie interventionnelle, Créteil, France
- Ecole Normale Supérieure, Département d'études Cognitives, Paris, France
- AP-HP, Hôpital H. Mondor- A. Chenevier, Centre de référence maladie de Huntington, Neurologie cognitive, Créteil, France
| |
Collapse
|
36
|
Anti-Inflammation of Natural Components from Medicinal Plants at Low Concentrations in Brain via Inhibiting Neutrophil Infiltration after Stroke. Mediators Inflamm 2016; 2016:9537901. [PMID: 27688603 PMCID: PMC5027307 DOI: 10.1155/2016/9537901] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 05/27/2016] [Accepted: 06/14/2016] [Indexed: 12/17/2022] Open
Abstract
Inflammation after stroke consists of activation of microglia/astrocytes in situ and infiltration of blood-borne leukocytes, resulting in brain damage and neurological deficits. Mounting data demonstrated that most natural components from medicinal plants had anti-inflammatory effects after ischemic stroke through inhibiting activation of resident microglia/astrocytes within ischemic area. However, it is speculated that this classical activity cannot account for the anti-inflammatory function of these natural components in the cerebral parenchyma, where they are detected at very low concentrations due to their poor membrane permeability and slight leakage of BBB. Could these drugs exert anti-inflammatory effects peripherally without being delivered across the BBB? Factually, ameliorating blood-borne neutrophil recruitment in peripheral circulatory system has been proved to reduce ischemic damage and improve outcomes. Thus, it is concluded that if drugs could achieve effective concentrations in the cerebral parenchyma, they can function via crippling resident microglia/astrocytes activation and inhibiting neutrophil infiltration, whereas the latter will be dominating when these drugs localize in the brain at a low concentration. In this review, the availability of some natural components crossing the BBB in stroke will be discussed, and how these drugs lead to improvements in stroke through inhibition of neutrophil rolling, adhesion, and transmigration will be illustrated.
Collapse
|
37
|
Hellström A, Ley D, Hansen-Pupp I, Hallberg B, Ramenghi LA, Löfqvist C, Smith LEH, Hård AL. Role of Insulinlike Growth Factor 1 in Fetal Development and in the Early Postnatal Life of Premature Infants. Am J Perinatol 2016; 33:1067-71. [PMID: 27603537 PMCID: PMC5779855 DOI: 10.1055/s-0036-1586109] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The neonatal period of very preterm infants is often characterized by a difficult adjustment to extrauterine life, with an inadequate nutrient supply and insufficient levels of growth factors, resulting in poor growth and a high morbidity rate. Long-term multisystem complications include cognitive, behavioral, and motor dysfunction as a result of brain damage as well as visual and hearing deficits and metabolic disorders that persist into adulthood. Insulinlike growth factor 1 (IGF-1) is a major regulator of fetal growth and development of most organs especially the central nervous system including the retina. Glucose metabolism in the developing brain is controlled by IGF-1 which also stimulates differentiation and prevents apoptosis. Serum concentrations of IGF-1 decrease to very low levels after very preterm birth and remain low for most of the perinatal development. Strong correlations have been found between low neonatal serum concentrations of IGF-1 and poor brain and retinal growth as well as poor general growth with multiorgan morbidities, such as intraventricular hemorrhage, retinopathy of prematurity, bronchopulmonary dysplasia, and necrotizing enterocolitis. Experimental and clinical studies indicate that early supplementation with IGF-1 can improve growth in catabolic states and reduce brain injury after hypoxic/ischemic events. A multicenter phase II study is currently underway to determine whether intravenous replacement of human recombinant IGF-1 up to normal intrauterine serum concentrations can improve growth and development and reduce prematurity-associated morbidities.
Collapse
Affiliation(s)
- Ann Hellström
- Department of Ophthalmology, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - David Ley
- Department of Pediatrics, Institute of Clinical Sciences, Lund University and Skane University Hospital, Lund, Sweden
| | - Ingrid Hansen-Pupp
- Department of Pediatrics, Institute of Clinical Sciences, Lund University and Skane University Hospital, Lund, Sweden
| | - Boubou Hallberg
- Department of Neonatology, Karolinska Institute and University Hospital, Stockholm, Sweden
| | - Luca A. Ramenghi
- Genova Neonatal Intensive Care Unit, Instituto Pediatrico Giannina Gaslini, Genova, Italy
| | - Chatarina Löfqvist
- Department of Ophthalmology, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Lois E. H. Smith
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Anna-Lena Hård
- Department of Ophthalmology, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
38
|
Keeney JTR, Ibrahimi S, Zhao L. Human ApoE Isoforms Differentially Modulate Glucose and Amyloid Metabolic Pathways in Female Brain: Evidence of the Mechanism of Neuroprotection by ApoE2 and Implications for Alzheimer's Disease Prevention and Early Intervention. J Alzheimers Dis 2016; 48:411-24. [PMID: 26402005 DOI: 10.3233/jad-150348] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Three major genetic isoforms of apolipoprotein E (ApoE), ApoE2, ApoE3, and ApoE4, exist in humans and lead to differences in susceptibility to Alzheimer's disease (AD). This study investigated the impact of human ApoE isoforms on brain metabolic pathways involved in glucose utilization and amyloid-β (Aβ) degradation, two major areas that are significantly perturbed in preclinical AD. Hippocampal RNA samples from middle-aged female mice with targeted human ApoE2, ApoE3, and ApoE4 gene replacement were comparatively analyzed with a qRT-PCR custom array for the expression of 85 genes involved in insulin/insulin-like growth factor (Igf) signaling. Consistent with its protective role against AD, ApoE2 brain exhibited the most metabolically robust profile among the three ApoE genotypes. When compared to ApoE2 brain, both ApoE3 and ApoE4 brains exhibited markedly reduced levels of Igf1, insulin receptor substrates (Irs), and facilitated glucose transporter 4 (Glut4), indicating reduced glucose uptake. Additionally, ApoE4 brain exhibited significantly decreased Pparg and insulin-degrading enzyme (Ide), indicating further compromised glucose metabolism and Aβ dysregulation associated with ApoE4. Protein analysis showed significantly decreased Igf1, Irs, and Glut4 in ApoE3 brain, and Igf1, Irs, Glut4, Pparg, and Ide in ApoE4 brain compared to ApoE2 brain. These data provide the first documented evidence that human ApoE isoforms differentially affect brain insulin/Igf signaling and downstream glucose and amyloid metabolic pathways, illustrating a potential mechanism for their differential risk in AD. A therapeutic strategy that enhances brain insulin/Igf1 signaling activity to a more robust ApoE2-like phenotype favoring both energy production and amyloid homeostasis holds promise for AD prevention and early intervention.
Collapse
Affiliation(s)
| | - Shaher Ibrahimi
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, USA
| | - Liqin Zhao
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, USA.,Neuroscience Graduate Program, University of Kansas, Lawrence, KS, USA
| |
Collapse
|
39
|
Zhao L, Mao Z, Woody SK, Brinton RD. Sex differences in metabolic aging of the brain: insights into female susceptibility to Alzheimer's disease. Neurobiol Aging 2016; 42:69-79. [PMID: 27143423 DOI: 10.1016/j.neurobiolaging.2016.02.011] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 02/08/2016] [Accepted: 02/10/2016] [Indexed: 10/22/2022]
Abstract
Despite recent advances in the understanding of clinical aspects of sex differences in Alzheimer's disease (AD), the underlying mechanisms, for instance, how sex modifies AD risk and why the female brain is more susceptible to AD, are not clear. The purpose of this study is to elucidate sex disparities in brain aging profiles focusing on 2 major areas-energy and amyloid metabolism-that are most significantly affected in preclinical development of AD. Total RNA isolated from hippocampal tissues of both female and male 129/C57BL/6 mice at ages of 6, 9, 12, or 15 months were comparatively analyzed by custom-designed Taqman low-density arrays for quantitative real-time polymerase chain reaction detection of a total of 182 genes involved in a broad spectrum of biological processes modulating energy production and amyloid homeostasis. Gene expression profiles revealed substantial differences in the trajectory of aging changes between female and male brains. In female brains, 44.2% of genes were significantly changed from 6 months to 9 months and two-thirds showed downregulation. In contrast, in male brains, only 5.4% of genes were significantly altered at this age transition. Subsequent changes in female brains were at a much smaller magnitude, including 10.9% from 9 months to 12 months and 6.1% from 12 months to 15 months. In male brains, most changes occurred from 12 months to 15 months and the majority were upregulated. Furthermore, gene network analysis revealed that clusterin appeared to serve as a link between the overall decreased bioenergetic metabolism and increased amyloid dyshomeostasis associated with the earliest transition in female brains. Together, results from this study indicate that: (1) female and male brains follow profoundly dissimilar trajectories as they age; (2) female brains undergo age-related changes much earlier than male brains; (3) early changes in female brains signal the onset of a hypometabolic phenotype at risk for AD. These findings provide a mechanistic rationale for female susceptibility to AD and suggest a potential window of opportunity for AD prevention and risk reduction in women.
Collapse
Affiliation(s)
- Liqin Zhao
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, USA; Neuroscience Graduate Program, University of Kansas, Lawrence, KS, USA; Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA.
| | - Zisu Mao
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Sarah K Woody
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, USA
| | - Roberta D Brinton
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA; Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
40
|
Pang Y, Lin S, Wright C, Shen J, Carter K, Bhatt A, Fan LW. Intranasal insulin protects against substantia nigra dopaminergic neuronal loss and alleviates motor deficits induced by 6-OHDA in rats. Neuroscience 2016; 318:157-65. [PMID: 26777890 DOI: 10.1016/j.neuroscience.2016.01.020] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 12/22/2015] [Accepted: 01/08/2016] [Indexed: 01/04/2023]
Abstract
Protection of substantia nigra (SN) dopaminergic (DA) neurons by neurotrophic factors (NTFs) is one of the promising strategies in Parkinson's disease (PD) therapy. A major clinical challenge for NTF-based therapy is that NTFs need to be delivered into the brain via invasive means, which often shows limited delivery efficiency. The nose to brain pathway is a non-invasive brain drug delivery approach developed in recent years. Of particular interest is the finding that intranasal insulin improves cognitive functions in Alzheimer's patients. In vitro, insulin has been shown to protect neurons against various insults. Therefore, the current study was designed to test whether intranasal insulin could afford neuroprotection in the 6-hydroxydopamine (6-OHDA)-based rat PD model. 6-OHDA was injected into the right side of striatum to induce a progressive DA neuronal lesion in the ipsilateral SN pars compact (SNc). Recombinant human insulin was applied intranasally to rats starting from 24h post lesion, once per day, for 2 weeks. A battery of motor behavioral tests was conducted on day 8 and 15. The number of DA neurons in the SNc was estimated by stereological counting. Our results showed that 6-OHDA injection led to significant motor deficits and 53% of DA neuron loss in the ipsilateral side of injection. Treatment with insulin significantly ameliorated 6-OHDA-induced motor impairments, as shown by improved locomotor activity, tapered/ledged beam-walking performance, vibrissa-elicited forelimb-placing, initial steps, as well as methamphetamine-induced rotational behavior. Consistent with behavioral improvements, insulin treatment provided a potent protection of DA neurons in the SNc against 6-OHDA neurotoxicity, as shown by a 74.8% increase in tyrosine hydroxylase (TH)-positive neurons compared to the vehicle group. Intranasal insulin treatment did not affect body weight and blood glucose levels. In conclusion, our study showed that intranasal insulin provided strong neuroprotection in the 6-OHDA rat PD model, suggesting that insulin signaling may be a novel therapeutic target in broad neurodegenerative disorders.
Collapse
Affiliation(s)
- Y Pang
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, United States.
| | - S Lin
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, United States
| | - C Wright
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, United States
| | - J Shen
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, United States
| | - K Carter
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, United States
| | - A Bhatt
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, United States
| | - L-W Fan
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, United States
| |
Collapse
|
41
|
Tu KY, Wu MK, Chen YW, Lin PY, Wang HY, Wu CK, Tseng PT. Significantly Higher Peripheral Insulin-Like Growth Factor-1 Levels in Patients With Major Depressive Disorder or Bipolar Disorder Than in Healthy Controls: A Meta-Analysis and Review Under Guideline of PRISMA. Medicine (Baltimore) 2016; 95:e2411. [PMID: 26825882 PMCID: PMC5291552 DOI: 10.1097/md.0000000000002411] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
An increasing amount of research has focused on insulin-like growth factor-1 (IGF-1) because of multiple neurotrophic effects, including neurogenesis, remyelination, and synaptogenesis. In addition, IGF-1 can mediate an antidepressant effect in patients with major affective disorder, and its levels in the cerebrospinal fluid have been found to vary with antidepressant treatment. Furthermore, it has been proven to crossover the blood-brain barrier, with a reciprocal feedback loop being the central effect. However, recent studies have reported inconclusive findings about the role of IGF-1 in major affective disorder. The aim of the current study was to conduct a thorough meta-analysis of changes in peripheral IGF-1 levels in patients with major depressive disorder (MDD) or bipolar disorder (BD). We conducted a thorough literature search and compared peripheral IGF-1 levels in patients with MDD or BD and in healthy controls, and investigated clinical variables through meta-regression. Electronic research was conducted through platform of PubMed. We used inclusion criteria as clinical trials discussing comparisons of peripheral IGF-1 protein levels in patients with MDD or BD and those in healthy controls. We analyzed the cases from 9 studies with the random-effect model. The main finding was that peripheral IGF-1 levels in the patients were significantly higher than in the healthy controls (P < 0.001), with a significant inverse association with duration of illness (P = 0.03). In meta-analysis comparing peripheral IGF-1 levels in patients with BD or MDD before and after treatment, there was no significant change in peripheral IGF-1 levels after treatment (P = 0.092). The small numbers of studies and lack of correlation data with growth hormone in current studies are the main limitations of this meta-analysis. Our results indicated that peripheral IGF-1 levels may not be an indicator of disease severity, but may be a disease trait marker or an indicator of cognition. However, further investigations on the correlation between cognitive function and peripheral IGF-1 levels are needed to explore the role of IGF-1 in the pathophysiology of MDD and BD.
Collapse
Affiliation(s)
- Kun-Yu Tu
- From the Department of Psychiatry, Tsyr-Huey Mental Hospital, Kaohsiung Jen-Ai's Home, Taiwan (K-YT, H-YW, C-KW, P-TT); Department of Neurology, E-Da Hospital, Kaohsiung, Taiwan (Y-WC); Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan (M-KW, P-YL); and Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan (P-YL)
| | | | | | | | | | | | | |
Collapse
|
42
|
Mysoet J, Dupont E, Bastide B, Canu MH. Role of IGF-1 in cortical plasticity and functional deficit induced by sensorimotor restriction. Behav Brain Res 2015; 290:117-23. [PMID: 25958232 DOI: 10.1016/j.bbr.2015.04.055] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 04/27/2015] [Accepted: 04/28/2015] [Indexed: 12/15/2022]
Abstract
In the adult rat, sensorimotor restriction by hindlimb unloading (HU) is known to induce impairments in motor behavior as well as a disorganization of somatosensory cortex (shrinkage of the cortical representation of the hindpaw, enlargement of the cutaneous receptive fields, decreased cutaneous sensibility threshold). Recently, our team has demonstrated that IGF-1 level was decreased in the somatosensory cortex of rats submitted to a 14-day period of HU. To determine whether IGF-1 is involved in these plastic mechanisms, a chronic cortical infusion of this substance was performed by means of osmotic minipump. When administered in control rats, IGF-1 affects the size of receptive fields and the cutaneous threshold, but has no effect on the somatotopic map. In addition, when injected during the whole HU period, IGF-1 is interestingly implied in cortical changes due to hypoactivity: the shrinkage of somatotopic representation of hindlimb is prevented, whereas the enlargement of receptive fields is reduced. IGF-1 has no effect on the increase in neuronal response to peripheral stimulation. We also explored the functional consequences of IGF-1 level restoration on tactile sensory discrimination. In HU rats, the percentage of paw withdrawal after a light tactile stimulation was decreased, whereas it was similar to control level in HU-IGF-1 rats. Taken together, the data clearly indicate that IGF-1 plays a key-role in cortical plastic mechanisms and in behavioral alterations induced by a decrease in sensorimotor activity.
Collapse
Affiliation(s)
- Julien Mysoet
- Laboratoire « Activité Physique, Muscle et Santé », EA 4488, IFR 114, Université Lille 1, Sciences et Technologies, F-59650 Villeneuve d'Ascq, France; Unité de Recherche Pluridisciplinaire Sport, Santé, Société (URePSSS), Université Lille Nord de France, F-59000 Lille, France.
| | - Erwan Dupont
- Laboratoire « Activité Physique, Muscle et Santé », EA 4488, IFR 114, Université Lille 1, Sciences et Technologies, F-59650 Villeneuve d'Ascq, France; Unité de Recherche Pluridisciplinaire Sport, Santé, Société (URePSSS), Université Lille Nord de France, F-59000 Lille, France.
| | - Bruno Bastide
- Laboratoire « Activité Physique, Muscle et Santé », EA 4488, IFR 114, Université Lille 1, Sciences et Technologies, F-59650 Villeneuve d'Ascq, France; Unité de Recherche Pluridisciplinaire Sport, Santé, Société (URePSSS), Université Lille Nord de France, F-59000 Lille, France.
| | - Marie-Hélène Canu
- Laboratoire « Activité Physique, Muscle et Santé », EA 4488, IFR 114, Université Lille 1, Sciences et Technologies, F-59650 Villeneuve d'Ascq, France; Unité de Recherche Pluridisciplinaire Sport, Santé, Société (URePSSS), Université Lille Nord de France, F-59000 Lille, France.
| |
Collapse
|
43
|
Altered theta oscillations and aberrant cortical excitatory activity in the 5XFAD model of Alzheimer's disease. Neural Plast 2015; 2015:781731. [PMID: 25922768 PMCID: PMC4398951 DOI: 10.1155/2015/781731] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 03/18/2015] [Indexed: 11/18/2022] Open
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disorder characterized by impairment of memory function. The 5XFAD mouse model was analyzed and compared with wild-type (WT) controls for aberrant cortical excitability and hippocampal theta oscillations by using simultaneous video-electroencephalogram (EEG) monitoring. Seizure staging revealed that 5XFAD mice exhibited cortical hyperexcitability whereas controls did not. In addition, 5XFAD mice displayed a significant increase in hippocampal theta activity from the light to dark phase during nonmotor activity. We also observed a reduction in mean theta frequency in 5XFAD mice compared to controls that was again most prominent during nonmotor activity. Transcriptome analysis of hippocampal probes and subsequent qPCR validation revealed an upregulation of Plcd4 that might be indicative of enhanced muscarinic signalling. Our results suggest that 5XFAD mice exhibit altered cortical excitability, hippocampal dysrhythmicity, and potential changes in muscarinic signaling.
Collapse
|
44
|
Pedraz M, Martín-Velasco AI, García-Marchena N, Araos P, Serrano A, Romero-Sanchiz P, Suárez J, Castilla-Ortega E, Barrios V, Campos-Cloute R, Ruiz JJ, Torrens M, Chowen JA, Argente J, de la Torre R, Santín LJ, Villanúa MÁ, Rodríguez de Fonseca F, Pavón FJ. Plasma concentrations of BDNF and IGF-1 in abstinent cocaine users with high prevalence of substance use disorders: relationship to psychiatric comorbidity. PLoS One 2015; 10:e0118610. [PMID: 25734326 PMCID: PMC4348520 DOI: 10.1371/journal.pone.0118610] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 01/19/2015] [Indexed: 11/29/2022] Open
Abstract
Recent studies have identified biomarkers related to the severity and pathogenesis of cocaine addiction and common comorbid psychiatric disorders. Monitoring these plasma mediators may improve the stratification of cocaine users seeking treatment. Because the neurotrophic factors are involved in neural plasticity, neurogenesis and neuronal survival, we have determined plasma concentrations of brain-derived neurotrophic factor (BDNF), insulin-like growth factor 1 (IGF-1) and IGF-1 binding protein 3 (IGFBP-3) in a cross-sectional study with abstinent cocaine users who sought outpatient treatment for cocaine (n = 100) and age/body mass matched controls (n = 85). Participants were assessed with the diagnostic interview 'Psychiatric Research Interview for Substance and Mental Disorders'. Plasma concentrations of these peptides were not different in cocaine users and controls. They were not associated with length of abstinence, duration of cocaine use or cocaine symptom severity. The pathological use of cocaine did not influence the association of IGF-1 with age observed in healthy subjects, but the correlation between IGF-1 and IGFBP-3 was not significantly detected. Correlation analyses were performed between these peptides and other molecules sensitive to addiction: BDNF concentrations were not associated with inflammatory mediators, lipid derivatives or IGF-1 in cocaine users, but correlated with chemokines (fractalkine/CX3CL1 and SDF-1/CXCL12) and N-acyl-ethanolamines (N-palmitoyl-, N-oleoyl-, N-arachidonoyl-, N-linoleoyl- and N-dihomo-γ-linolenoyl-ethanolamine) in controls; IGF-1 concentrations only showed association with IGFBP-3 concentrations in controls; and IGFBP-3 was only correlated with N-stearoyl-ethanolamine concentrations in cocaine users. Multiple substance use disorders and life-time comorbid psychopathologies were common in abstinent cocaine users. Interestingly, plasma BDNF concentrations were exclusively found to be decreased in users diagnosed with both primary and cocaine-induced disorders for mood and anxiety disorders. In summary, BDNF, IGF-1 and IGFBP-3 were not affected by a history of pathological use of cocaine supported by the absence of associations with other molecules sensitive to cocaine addiction. However, BDNF was affected by comorbid mood disorders. Further research is necessary to elucidate the role of BDNF and IGF-1 in the transition to cocaine addiction and associated psychiatric comorbidity.
Collapse
Affiliation(s)
- María Pedraz
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga-Universidad de Málaga, Málaga, Spain
| | | | - Nuria García-Marchena
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga-Universidad de Málaga, Málaga, Spain
| | - Pedro Araos
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga-Universidad de Málaga, Málaga, Spain
| | - Antonia Serrano
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga-Universidad de Málaga, Málaga, Spain
| | - Pablo Romero-Sanchiz
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga-Universidad de Málaga, Málaga, Spain
| | - Juan Suárez
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga-Universidad de Málaga, Málaga, Spain
| | - Estela Castilla-Ortega
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga-Universidad de Málaga, Málaga, Spain
| | - Vicente Barrios
- Department of Pediatrics & Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Rafael Campos-Cloute
- Centro de Tratamiento Ambulatorio Mijas Costa-Diputación de Málaga, Mijas, Spain
| | - Juan Jesús Ruiz
- Centro Provincial de Drogodependencia-Diputación de Málaga, Málaga, Spain
| | - Marta Torrens
- Neurosciences Program, Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), Barcelona, Spain
- Institut de Neuropsiquiatria i Addiccions (INAD) del Parc de Salut MAR, Barcelona, Spain
- Department of Psychiatry. Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Julie Ann Chowen
- Department of Pediatrics & Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Argente
- Department of Pediatrics & Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Rafael de la Torre
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Neurosciences Program, Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), Barcelona, Spain
- Facultat de Ciencies de la Salut i de la Vida, Universitat Pompeu Fabra (CEXS-UPF), Barcelona, Spain
| | - Luis Javier Santín
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Facultad de Psicología, Universidad de Málaga, Málaga, Spain
| | - María Ángeles Villanúa
- Department of Physiology, Faculty of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Fernando Rodríguez de Fonseca
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga-Universidad de Málaga, Málaga, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Francisco Javier Pavón
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga-Universidad de Málaga, Málaga, Spain
| |
Collapse
|
45
|
Fu Q, McKnight RA, Callaway CW, Yu X, Lane RH, Majnik AV. Intrauterine growth restriction disrupts developmental epigenetics around distal growth hormone response elements on the rat hepatic IGF‐1 gene. FASEB J 2014; 29:1176-84. [DOI: 10.1096/fj.14-258442] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 11/03/2014] [Indexed: 11/11/2022]
Affiliation(s)
- Qi Fu
- Division of NeonatologyDepartment of PediatricsMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Robert A. McKnight
- Division of NeonatologyDepartment of PediatricsUniversity of UtahSalt Lake CityUtahUSA
| | | | - Xing Yu
- Division of NeonatologyDepartment of PediatricsUniversity of UtahSalt Lake CityUtahUSA
| | - Robert H. Lane
- Division of NeonatologyDepartment of PediatricsMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Amber V. Majnik
- Division of NeonatologyDepartment of PediatricsMedical College of WisconsinMilwaukeeWisconsinUSA
| |
Collapse
|
46
|
Kim W, Noh H, Lee Y, Jeon J, Shanmugavadivu A, McPhie DL, Kim KS, Cohen BM, Seo H, Sonntag KC. MiR-126 Regulates Growth Factor Activities and Vulnerability to Toxic Insult in Neurons. Mol Neurobiol 2014; 53:95-108. [PMID: 25407931 DOI: 10.1007/s12035-014-8989-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 11/04/2014] [Indexed: 01/19/2023]
Abstract
Dysfunction of growth factor (GF) activities contributes to the decline and death of neurons during aging and in neurodegenerative diseases. In addition, neurons become more resistant to GF signaling with age. Micro (mi)RNAs are posttranscriptional regulators of gene expression that may be crucial to age- and disease-related changes in GF functions. MiR-126 is involved in regulating insulin/IGF-1/phosphatidylinositol-3-kinase (PI3K)/AKT and extracellular signal-regulated kinase (ERK) signaling, and we recently demonstrated a functional role of miR-126 in dopamine neuronal cell survival in models of Parkinson's disease (PD)-associated toxicity. Here, we show that elevated levels of miR-126 increase neuronal vulnerability to ubiquitous toxicity mediated by staurosporine (STS) or Alzheimer's disease (AD)-associated amyloid beta 1-42 peptides (Aβ1-42). The neuroprotective factors IGF-1, nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), and soluble amyloid precursor protein α (sAPPα) could diminish but not abrogate the toxic effects of miR-126. In miR-126 overexpressing neurons derived from Tg6799 familial AD model mice, we observed an increase in Aβ1-42 toxicity, but surprisingly, both Aβ1-42 and miR-126 promoted neurite sprouting. Pathway analysis revealed that miR-126 overexpression downregulated elements in the GF/PI3K/AKT and ERK signaling cascades, including AKT, GSK-3β, ERK, their phosphorylation, and the miR-126 targets IRS-1 and PIK3R2. Finally, inhibition of miR-126 was neuroprotective against both STS and Aβ1-42 toxicity. Our data provide evidence for a novel mechanism of regulating GF/PI3K signaling in neurons by miR-126 and suggest that miR-126 may be an important mechanistic link between metabolic dysfunction and neurotoxicity in general, during aging, and in the pathogenesis of specific neurological disorders, including PD and AD.
Collapse
Affiliation(s)
- Woori Kim
- Department of Psychiatry, McLean Hospital, Harvard Medical School, MRC 223, 115 Mill Street, Belmont, MA, 02478, USA
| | - Haneul Noh
- Department of Molecular and Life Sciences, Hanyang University, Ansan, South Korea
| | - Yenarae Lee
- Department of Psychiatry, McLean Hospital, Harvard Medical School, MRC 223, 115 Mill Street, Belmont, MA, 02478, USA
| | - Jeha Jeon
- Department of Molecular and Life Sciences, Hanyang University, Ansan, South Korea
| | - Arthi Shanmugavadivu
- Department of Psychiatry, McLean Hospital, Harvard Medical School, MRC 223, 115 Mill Street, Belmont, MA, 02478, USA
| | - Donna L McPhie
- Department of Psychiatry, McLean Hospital, Harvard Medical School, MRC 223, 115 Mill Street, Belmont, MA, 02478, USA
| | - Kwang-Soo Kim
- Department of Psychiatry, McLean Hospital, Harvard Medical School, MRC 223, 115 Mill Street, Belmont, MA, 02478, USA
| | - Bruce M Cohen
- Department of Psychiatry, McLean Hospital, Harvard Medical School, MRC 223, 115 Mill Street, Belmont, MA, 02478, USA
| | - Hyemyung Seo
- Department of Molecular and Life Sciences, Hanyang University, Ansan, South Korea
| | - Kai C Sonntag
- Department of Psychiatry, McLean Hospital, Harvard Medical School, MRC 223, 115 Mill Street, Belmont, MA, 02478, USA.
| |
Collapse
|
47
|
Yan H, Mitschelen M, Toth P, Ashpole NM, Farley JA, Hodges EL, Warrington JP, Han S, Fung KM, Csiszar A, Ungvari Z, Sonntag WE. Endothelin-1-induced focal cerebral ischemia in the growth hormone/IGF-1 deficient Lewis Dwarf rat. J Gerontol A Biol Sci Med Sci 2014; 69:1353-62. [PMID: 25098324 DOI: 10.1093/gerona/glu118] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Aging is a major risk factor for cerebrovascular disease. Growth hormone (GH) and its anabolic mediator, insulin-like growth factor (IGF)-1, decrease with advancing age and this decline has been shown to promote vascular dysfunction. In addition, lower GH/IGF-1 levels are associated with higher stroke mortality in humans. These results suggest that decreased GH/IGF-1 level is an important factor in increased risk of cerebrovascular diseases. This study was designed to assess whether GH/IGF-1-deficiency influences the outcome of cerebral ischemia. We found that endothelin-1-induced middle cerebral artery occlusion resulted in a modest but nonsignificant decrease in cerebral infarct size in GH/IGF-1 deficient dw/dw rats compared with control heterozygous littermates and dw/dw rats with early-life GH treatment. Expression of endothelin receptors and endothelin-1-induced constriction of the middle cerebral arteries were similar in the three experimental groups. Interestingly, dw/dw rats exhibited reduced brain edema and less astrocytic infiltration compared with their heterozygous littermates and this effect was reversed by GH-treatment. Because reactive astrocytes are critical for the regulation of poststroke inflammatory processes, maintenance of the blood-brain barrier and neural repair, further studies are warranted to determine the long-term functional consequences of decreased astrocytic activation in GH/IGF-1 deficient animals after cerebral ischemia.
Collapse
Affiliation(s)
- Han Yan
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City
| | - Matthew Mitschelen
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City
| | - Peter Toth
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City
| | - Nicole M Ashpole
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City
| | - Julie A Farley
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City
| | - Erik L Hodges
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City
| | - Junie P Warrington
- Present address: Department of Physiology & Biophysics, University of Mississippi Medical Center, Jackson, MS 39216
| | - Song Han
- Present address: Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | - Kar-Ming Fung
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City
| | - Anna Csiszar
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City
| | - Zoltan Ungvari
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City
| | - William E Sonntag
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City.
| |
Collapse
|
48
|
Kim W, Lee Y, McKenna ND, Yi M, Simunovic F, Wang Y, Kong B, Rooney RJ, Seo H, Stephens RM, Sonntag KC. miR-126 contributes to Parkinson's disease by dysregulating the insulin-like growth factor/phosphoinositide 3-kinase signaling. Neurobiol Aging 2014; 35:1712-21. [PMID: 24559646 PMCID: PMC3991567 DOI: 10.1016/j.neurobiolaging.2014.01.021] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 01/11/2014] [Accepted: 01/19/2014] [Indexed: 01/13/2023]
Abstract
Dopamine (DA) neurons in sporadic Parkinson's disease (PD) display dysregulated gene expression networks and signaling pathways that are implicated in PD pathogenesis. Micro (mi)RNAs are regulators of gene expression, which could be involved in neurodegenerative diseases. We determined the miRNA profiles in laser microdissected DA neurons from postmortem sporadic PD patients' brains and age-matched controls. DA neurons had a distinctive miRNA signature and a set of miRNAs was dysregulated in PD. Bioinformatics analysis provided evidence for correlations of miRNAs with signaling pathways relevant to PD, including an association of miR-126 with insulin/IGF-1/PI3K signaling. In DA neuronal cell systems, enhanced expression of miR-126 impaired IGF-1 signaling and increased vulnerability to the neurotoxin 6-OHDA by downregulating factors in IGF-1/PI3K signaling, including its targets p85β, IRS-1, and SPRED1. Blocking of miR-126 function increased IGF-1 trophism and neuroprotection to 6-OHDA. Our data imply that elevated levels of miR-126 may play a functional role in DA neurons and in PD pathogenesis by downregulating IGF-1/PI3K/AKT signaling and that its inhibition could be a mechanism of neuroprotection.
Collapse
Affiliation(s)
- Woori Kim
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, USA; Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Yenarae Lee
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Noah D McKenna
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Ming Yi
- Bioinformatics Support Group, Advanced Biomedical Computing Center, NCI-Frederick, Frederick, MD, USA
| | - Filip Simunovic
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | | | | | | | - Hyemyung Seo
- Division of Molecular & Life Sciences, College of Science & Technology, Hanyang University, Seoul, Korea
| | - Robert M Stephens
- Bioinformatics Support Group, Advanced Biomedical Computing Center, NCI-Frederick, Frederick, MD, USA
| | - Kai C Sonntag
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, USA.
| |
Collapse
|
49
|
Westwood AJ, Beiser A, Decarli C, Harris TB, Chen TC, He XM, Roubenoff R, Pikula A, Au R, Braverman LE, Wolf PA, Vasan RS, Seshadri S. Insulin-like growth factor-1 and risk of Alzheimer dementia and brain atrophy. Neurology 2014; 82:1613-9. [PMID: 24706014 PMCID: PMC4013812 DOI: 10.1212/wnl.0000000000000382] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 01/29/2014] [Indexed: 02/03/2023] Open
Abstract
OBJECTIVE To relate serum insulin-like growth factor-1 (IGF-1) to risk of Alzheimer disease (AD) dementia and to brain volumes in a dementia-free community sample spanning middle and older ages. METHODS Dementia-free Framingham participants from generation 1 (n = 789, age 79 ± 4 years, 64% women) and generation 2 (n = 2,793, age 61 ± 9 years, 55% women; total = 3,582, age 65 ± 11 years, 57% women) had serum IGF-1 measured in 1990-1994 and 1998-2001, respectively, and were followed prospectively for incident dementia and AD dementia. Brain MRI was obtained in stroke- and dementia-free survivors of both generations 1 (n = 186) and 2 (n = 1,867) during 1999-2005. Baseline IGF-1 was related to risk of incident dementia using Cox models and to total brain and hippocampal volumes using linear regression in multivariable models adjusted for age, sex, APOE ε4, plasma homocysteine, waist-hip ratio, and physical activity. RESULTS Mean IGF-1 levels were 144 ± 60 μg/L in generation 1 and 114 ± 37 μg/L in generation 2. We observed 279 cases of incident dementia (230 AD dementia) over a mean follow-up of 7.4 ± 3.1 years. Persons with IGF-1 in the lowest quartile had a 51% greater risk of AD dementia (hazard ratio = 1.51, 95% confidence interval: 1.14-2.00; p = 0.004). Among persons without dementia, higher IGF-1 levels were associated with greater total brain volumes (β/SD increment in IGF-1 was 0.55 ± 0.24, p = 0.025; and 0.26 ± 0.06, p < 0.001, for generations 1 and 2, respectively). CONCLUSION Lower serum levels of IGF-1 are associated with an increased risk of developing AD dementia and higher levels with greater brain volumes even among middle-aged community-dwelling participants free of stroke and dementia. Higher levels of IGF-1 may protect against subclinical and clinical neurodegeneration.
Collapse
Affiliation(s)
- Andrew J Westwood
- From the Department of Neurology (A.J.W., A.B., A.P., R.A., P.A.W., S.S.) and Sections of Preventative Medicine and Cardiology, Department of Medicine (R.S.V.), Boston University School of Medicine; Department of Biostatistics (A.B.), Boston University School of Public Health, Boston; Framingham Heart Study (A.B., A.P., R.A., P.A.W., R.S.V., S.S.), Framingham, MA; University of California at Davis (C.D.), Sacramento, CA; National Institute on Aging (T.B.H.), Bethesda, MD; Boston University Medical Center (T.C.C., X.-m.H., L.E.B.), Section of Endocrinology, Diabetes, and Nutrition, Boston; Novartis Institutes for Biomedical Research (R.R.), Cambridge, MA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Cyclic glycine-proline regulates IGF-1 homeostasis by altering the binding of IGFBP-3 to IGF-1. Sci Rep 2014; 4:4388. [PMID: 24633053 PMCID: PMC3955921 DOI: 10.1038/srep04388] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 02/28/2014] [Indexed: 02/02/2023] Open
Abstract
The homeostasis of insulin-like growth factor-1 (IGF-1) is essential for metabolism, development and survival. Insufficient IGF-1 is associated with poor recovery from wounds whereas excessive IGF-1 contributes to growth of tumours. We have shown that cyclic glycine-proline (cGP), a metabolite of IGF-1, can normalise IGF-1 function by showing its efficacy in improving the recovery from ischemic brain injury in rats and inhibiting the growth of lymphomic tumours in mice. Further investigation in cell culture suggested that cGP promoted the activity of IGF-1 when it was insufficient, but inhibited the activity of IGF-1 when it was excessive. Mathematical modelling revealed that the efficacy of cGP was a modulated IGF-1 effect via changing the binding of IGF-1 to its binding proteins, which dynamically regulates the balance between bioavailable and non-bioavailable IGF-1. Our data reveal a novel mechanism of auto-regulation of IGF-1, which has physiological and pathophysiological consequences and potential pharmacological utility.
Collapse
|