1
|
Arjunan A, Sah DK, Woo M, Song J. Identification of the molecular mechanism of insulin-like growth factor-1 (IGF-1): a promising therapeutic target for neurodegenerative diseases associated with metabolic syndrome. Cell Biosci 2023; 13:16. [PMID: 36691085 PMCID: PMC9872444 DOI: 10.1186/s13578-023-00966-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 01/17/2023] [Indexed: 01/24/2023] Open
Abstract
Neurodegenerative disorders are accompanied by neuronal degeneration and glial dysfunction, resulting in cognitive, psychomotor, and behavioral impairment. Multiple factors including genetic, environmental, metabolic, and oxidant overload contribute to disease progression. Recent evidences suggest that metabolic syndrome is linked to various neurodegenerative diseases. Metabolic syndrome (MetS) is known to be accompanied by symptoms such as hyperglycemia, abdominal obesity, hypertriglyceridemia, and hypertension. Despite advances in knowledge about the pathogenesis of neurodegenerative disorders, effective treatments to combat neurodegenerative disorders caused by MetS have not been developed to date. Insulin growth factor-1 (IGF-1) deficiency has been associated with MetS-related pathologies both in-vivo and in-vitro. IGF-1 is essential for embryonic and adult neurogenesis, neuronal plasticity, neurotropism, angiogenesis, metabolic function, and protein clearance in the brain. Here, we review the evidence for the potential therapeutic effects of IGF-1 in the neurodegeneration related to metabolic syndrome. We elucidate how IGF-1 may be involved in molecular signaling defects that occurs in MetS-related neurodegenerative disorders and highlight the importance of IGF-1 as a potential therapeutic target in MetS-related neurological diseases.
Collapse
Affiliation(s)
- Archana Arjunan
- grid.14005.300000 0001 0356 9399Department of Anatomy, Chonnam National University Medical School, Hwasun, Jeollanam-Do 58128 Republic of Korea
| | - Dhiraj Kumar Sah
- grid.14005.300000 0001 0356 9399Department of Biochemistry, Chonnam National University Medical School, Hwasun, 58128 Republic of Korea ,grid.14005.300000 0001 0356 9399BioMedical Sciences Graduate Program (BMSGP), Chonnam National University, 264 Seoyangro, Hwasun, 58128 Republic of Korea
| | - Minna Woo
- grid.17063.330000 0001 2157 2938Division of Endocrinology and Metabolism, University Health Network and and Banting and Best Diabetes Centre, University of Toronto, Toronto, ON Canada
| | - Juhyun Song
- grid.14005.300000 0001 0356 9399Department of Anatomy, Chonnam National University Medical School, Hwasun, Jeollanam-Do 58128 Republic of Korea ,grid.14005.300000 0001 0356 9399BioMedical Sciences Graduate Program (BMSGP), Chonnam National University, 264 Seoyangro, Hwasun, 58128 Republic of Korea
| |
Collapse
|
2
|
Bieniussa L, Kahraman B, Skornicka J, Schulte A, Voelker J, Jablonka S, Hagen R, Rak K. Pegylated Insulin-Like Growth Factor 1 attenuates Hair Cell Loss and promotes Presynaptic Maintenance of Medial Olivocochlear Cholinergic Fibers in the Cochlea of the Progressive Motor Neuropathy Mouse. Front Neurol 2022; 13:885026. [PMID: 35720065 PMCID: PMC9203726 DOI: 10.3389/fneur.2022.885026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 04/26/2022] [Indexed: 11/13/2022] Open
Abstract
The progressive motor neuropathy (PMN) mouse is a model of an inherited motor neuropathy disease with progressive neurodegeneration. Axon degeneration associates with homozygous mutations of the TBCE gene encoding the tubulin chaperone E protein. TBCE is responsible for the correct dimerization of alpha and beta-tubulin. Strikingly, the PMN mouse also develops a progressive hearing loss after normal hearing onset, characterized by degeneration of the auditory nerve and outer hair cell (OHC) loss. However, the development of this neuronal and cochlear pathology is not fully understood yet. Previous studies with pegylated insulin-like growth factor 1 (peg-IGF-1) treatment in this mouse model have been shown to expand lifespan, weight, muscle strength, and motor coordination. Accordingly, peg-IGF-1 was evaluated for an otoprotective effect. We investigated the effect of peg-IGF-1 on the auditory system by treatment starting at postnatal day 15 (p15). Histological analysis revealed positive effects on OHC synapses of medial olivocochlear (MOC) neuronal fibers and a short-term attenuation of OHC loss. Peg-IGF-1 was able to conditionally restore the disorganization of OHC synapses and maintain the provision of cholinergic acetyltransferase in presynapses. To assess auditory function, frequency-specific auditory brainstem responses and distortion product otoacoustic emissions were recorded in animals on p21 and p28. However, despite the positive effect on MOC fibers and OHC, no restoration of hearing could be achieved. The present work demonstrates that the synaptic pathology of efferent MOC fibers in PMN mice represents a particular form of “efferent auditory neuropathy.” Peg-IGF-1 showed an otoprotective effect by preventing the degeneration of OHCs and efferent synapses. However, enhanced efforts are needed to optimize the treatment to obtain detectable improvements in hearing performances.
Collapse
Affiliation(s)
- Linda Bieniussa
- Department of Oto-Rhino-Laryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, University Hospital Würzburg, Würzburg, Germany
| | - Baran Kahraman
- Department of Oto-Rhino-Laryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, University Hospital Würzburg, Würzburg, Germany
| | - Johannes Skornicka
- Department of Oto-Rhino-Laryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, University Hospital Würzburg, Würzburg, Germany
| | - Annemarie Schulte
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| | - Johannes Voelker
- Department of Oto-Rhino-Laryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, University Hospital Würzburg, Würzburg, Germany
| | - Sibylle Jablonka
- Institute of Clinical Neurobiology, University of Würzburg, Würzburg, Germany
| | - Rudolf Hagen
- Department of Oto-Rhino-Laryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, University Hospital Würzburg, Würzburg, Germany
| | - Kristen Rak
- Department of Oto-Rhino-Laryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, University Hospital Würzburg, Würzburg, Germany
- *Correspondence: Kristen Rak
| |
Collapse
|
3
|
Pham VM, Thakor N. Insulin enhances neurite extension and myelination of diabetic neuropathy neurons. Korean J Pain 2022; 35:160-172. [PMID: 35354679 PMCID: PMC8977202 DOI: 10.3344/kjp.2022.35.2.160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/03/2022] [Accepted: 03/09/2022] [Indexed: 11/24/2022] Open
Abstract
Background The authors established an in vitro model of diabetic neuropathy based on the culture system of primary neurons and Schwann cells (SCs) to mimic similar symptoms observed in in vivo models of this complication, such as impaired neurite extension and impaired myelination. The model was then utilized to investigate the effects of insulin on enhancing neurite extension and myelination of diabetic neurons. Methods SCs and primary neurons were cultured under conditions mimicking hyperglycemia prepared by adding glucose to the basal culture medium. In a single culture, the proliferation and maturation of SCs and the neurite extension of neurons were evaluated. In a co-culture, the percentage of myelination of diabetic neurons was investigated. Insulin at different concentrations was supplemented to culture media to examine its effects on neurite extension and myelination. Results The cells showed similar symptoms observed in in vivo models of this complication. In a single culture, hyperglycemia attenuated the proliferation and maturation of SCs, induced apoptosis, and impaired neurite extension of both sensory and motor neurons. In a co-culture of SCs and neurons, the percentage of myelinated neurites in the hyperglycemia-treated group was significantly lower than that in the control group. This impaired neurite extension and myelination was reversed by the introduction of insulin to the hyperglycemic culture media. Conclusions Insulin may be a potential candidate for improving diabetic neuropathy. Insulin can function as a neurotrophic factor to support both neurons and SCs. Further research is needed to discover the potential of insulin in improving diabetic neuropathy.
Collapse
Affiliation(s)
- Vuong M Pham
- Singapore Institute for Neurotechnology, National University of Singapore, Singapore.,Department of Biotechnology, Ho Chi Minh City University of Food Industry, Ho Chi Minh City, Vietnam
| | - Nitish Thakor
- Singapore Institute for Neurotechnology, National University of Singapore, Singapore.,Department of Biomedical Engineering, National University of Singapore, Singapore.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
4
|
Aghanoori MR, Agarwal P, Gauvin E, Nagalingam RS, Bonomo R, Yathindranath V, Smith DR, Hai Y, Lee S, Jolivalt CG, Calcutt NA, Jones MJ, Czubryt MP, Miller DW, Dolinsky VW, Mansuy-Aubert V, Fernyhough P. CEBPβ regulation of endogenous IGF-1 in adult sensory neurons can be mobilized to overcome diabetes-induced deficits in bioenergetics and axonal outgrowth. Cell Mol Life Sci 2022; 79:193. [PMID: 35298717 PMCID: PMC8930798 DOI: 10.1007/s00018-022-04201-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 01/28/2022] [Accepted: 02/08/2022] [Indexed: 11/26/2022]
Abstract
Aberrant insulin-like growth factor 1 (IGF-1) signaling has been proposed as a contributing factor to the development of neurodegenerative disorders including diabetic neuropathy, and delivery of exogenous IGF-1 has been explored as a treatment for Alzheimer's disease and amyotrophic lateral sclerosis. However, the role of autocrine/paracrine IGF-1 in neuroprotection has not been well established. We therefore used in vitro cell culture systems and animal models of diabetic neuropathy to characterize endogenous IGF-1 in sensory neurons and determine the factors regulating IGF-1 expression and/or affecting neuronal health. Single-cell RNA sequencing (scRNA-Seq) and in situ hybridization analyses revealed high expression of endogenous IGF-1 in non-peptidergic neurons and satellite glial cells (SGCs) of dorsal root ganglia (DRG). Brain cortex and DRG had higher IGF-1 gene expression than sciatic nerve. Bidirectional transport of IGF-1 along sensory nerves was observed. Despite no difference in IGF-1 receptor levels, IGF-1 gene expression was significantly (P < 0.05) reduced in liver and DRG from streptozotocin (STZ)-induced type 1 diabetic rats, Zucker diabetic fatty (ZDF) rats, mice on a high-fat/ high-sugar diet and db/db type 2 diabetic mice. Hyperglycemia suppressed IGF-1 gene expression in cultured DRG neurons and this was reversed by exogenous IGF-1 or the aldose reductase inhibitor sorbinil. Transcription factors, such as NFAT1 and CEBPβ, were also less enriched at the IGF-1 promoter in DRG from diabetic rats vs control rats. CEBPβ overexpression promoted neurite outgrowth and mitochondrial respiration, both of which were blunted by knocking down or blocking IGF-1. Suppression of endogenous IGF-1 in diabetes may contribute to neuropathy and its upregulation at the transcriptional level by CEBPβ can be a promising therapeutic approach.
Collapse
MESH Headings
- Aging/metabolism
- Animals
- Antibodies, Neutralizing/pharmacology
- Axons/drug effects
- Axons/metabolism
- Axons/pathology
- Base Sequence
- CCAAT-Enhancer-Binding Protein-beta/genetics
- CCAAT-Enhancer-Binding Protein-beta/metabolism
- Cell Respiration/drug effects
- Cells, Cultured
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/pathology
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/pathology
- Energy Metabolism/drug effects
- Ganglia, Spinal/drug effects
- Ganglia, Spinal/metabolism
- Gene Expression Regulation/drug effects
- Glycolysis/drug effects
- HEK293 Cells
- Humans
- Insulin-Like Growth Factor I/genetics
- Insulin-Like Growth Factor I/metabolism
- Liver/metabolism
- Male
- Mitochondria/drug effects
- Mitochondria/metabolism
- NFATC Transcription Factors/metabolism
- Neuronal Outgrowth/drug effects
- Polymers/metabolism
- Promoter Regions, Genetic/genetics
- Protein Transport/drug effects
- Rats, Sprague-Dawley
- Sensory Receptor Cells/metabolism
- Sensory Receptor Cells/pathology
- Signal Transduction/drug effects
- Rats
Collapse
Affiliation(s)
- Mohamad-Reza Aghanoori
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada.
- Dept of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB, Canada.
- Dept of Medical Genetics, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4N2, Canada.
| | - Prasoon Agarwal
- Dept of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB, Canada
- Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, Canada
- School of Electrical Engineering and Computer Science, KTH Royal Institute of Technology, 10044, Stockholm, Sweden
| | - Evan Gauvin
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
| | - Raghu S Nagalingam
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
| | - Raiza Bonomo
- Cellular and Molecular Department, Stritch School of Medicine, Loyola University Chicago, Chicago, USA
| | - Vinith Yathindranath
- Kleysen Institute for Advanced Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Darrell R Smith
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
| | - Yan Hai
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Samantha Lee
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | | | | | - Meaghan J Jones
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Michael P Czubryt
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
| | - Donald W Miller
- Kleysen Institute for Advanced Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Vernon W Dolinsky
- Dept of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB, Canada
- Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, Canada
| | - Virginie Mansuy-Aubert
- Cellular and Molecular Department, Stritch School of Medicine, Loyola University Chicago, Chicago, USA
| | - Paul Fernyhough
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
- Dept of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
5
|
Kimura T, Yamada H, Teraoka M, Joko T, Iwata S, Tabata Y, Wakisaka H, Hato N. Intratympanic Insulin-like Growth Factor-1 Administration Via the Otic Bulla in a Severe Facial Paralysis Model. Otol Neurotol 2021; 42:e1376-e1381. [PMID: 34224549 DOI: 10.1097/mao.0000000000003263] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
HYPOTHESIS We investigated the treatment effect of intratympanic insulin-like growth factor-1 (IGF-1) on severe facial paralysis in guinea pigs. BACKGROUND The use of regenerative medicine involving growth factors has been reported in the treatment of peripheral nerve diseases. IGF-1 plays a crucial role in nerve regeneration. METHODS We performed the following procedures on guinea pigs. In the normal group (n = 7), no procedure was performed. In the saline (n = 7) and IGF-1 (n = 7) groups, facial paralysis was induced by freezing of the facial canal. Subsequently, in the saline and IGF-1 groups, a gelatin hydrogel impregnated with 100 μL saline and 400 μg/100 μL IGF-1, respectively, was placed in the facial canal. Facial nerve functions were evaluated using three test batteries: facial movement observation, electrophysiological testing, and histological assessment. RESULTS At 10 weeks postoperatively, the facial movement scores for the IGF-1 group were improved compared to those in the saline group. The conductive velocity was significantly faster in the IGF-1 group than in the saline group. There was a significant between-group difference in the nerve fiber number and myelin thickness. CONCLUSION Intratympanic IGF-1 administration improved facial nerve regeneration. This novel method could provide prompt ambulatory regenerative treatment and reduce the incidence of poor recovery in patients with severe facial paralysis.
Collapse
Affiliation(s)
- Takuya Kimura
- Department of Otorhinolaryngology Head and Neck Surgery, Ehime University
| | - Hiroyuki Yamada
- Department of Otorhinolaryngology Head and Neck Surgery, Ehime University
| | - Masato Teraoka
- Department of Otorhinolaryngology Head and Neck Surgery, Ehime University
| | - Tomonori Joko
- Department of Otorhinolaryngology Head and Neck Surgery, Ehime University
| | - Shinji Iwata
- Department of Otorhinolaryngology Head and Neck Surgery, Ehime University
| | - Yasuhiro Tabata
- Laboratory of Biomaterials, Department of Regeneration Science and Engineering Institute for Frontier Life and Medical Sciences, Kyoto University
| | - Hiroyuki Wakisaka
- Laboratory of Head and Neck Surgery, Ehime Prefectural University of Health Sciences, Ehime, Japan
| | - Naohito Hato
- Department of Otorhinolaryngology Head and Neck Surgery, Ehime University
| |
Collapse
|
6
|
Lopez J, Quan A, Budihardjo J, Xiang S, Wang H, Kiron Koshy, Cashman C, Lee WPA, Hoke A, Tuffaha S, Brandacher G. Growth Hormone Improves Nerve Regeneration, Muscle Re-innervation, and Functional Outcomes After Chronic Denervation Injury. Sci Rep 2019; 9:3117. [PMID: 30816300 PMCID: PMC6395714 DOI: 10.1038/s41598-019-39738-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 01/17/2019] [Indexed: 01/08/2023] Open
Abstract
This study investigates the efficacy of systemic growth hormone (GH) therapy in ameliorating the deleterious effects of chronic denervation (CD) injury on nerve regeneration and resulting motor function. Using a forelimb CD model, 4 groups of Lewis rats were examined (n = 8 per group): Group-1 (negative control) 8 weeks of median nerve CD followed by ulnar-to-median nerve transfer; Group-2 (experimental) 8 weeks of median nerve CD followed by ulnar-to-median nerve transfer and highly purified lyophilized pituitary porcine GH treatment (0.6 mg/day); Group-3 (positive control) immediate ulnar-to-median nerve transfer without CD; Group-4 (baseline) naïve controls. All animals underwent weekly grip strength testing and were sacrificed 14 weeks following nerve transfer for histomorphometric analysis of median nerve regeneration, flexor digitorum superficialis atrophy, and neuromuscular junction reinnervation. In comparison to untreated controls, GH-treated animals demonstrated enhanced median nerve regeneration as measured by axon density (p < 0.005), axon diameter (p < 0.0001), and myelin thickness (p < 0.0001); improved muscle re-innervation (27.9% vs 38.0% NMJs re-innervated; p < 0.02); reduced muscle atrophy (1146 ± 93.19 µm2 vs 865.2 ± 48.33 µm2; p < 0.02); and greater recovery of motor function (grip strength: p < 0.001). These findings support the hypothesis that GH-therapy enhances axonal regeneration and maintains chronically-denervated muscle to thereby promote motor re-innervation and functional recovery.
Collapse
Affiliation(s)
- Joseph Lopez
- Department of Plastic & Reconstructive Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Amy Quan
- Department of Plastic & Reconstructive Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Joshua Budihardjo
- Department of Plastic & Reconstructive Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sinan Xiang
- Department of Plastic & Reconstructive Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Howard Wang
- Department of Plastic & Reconstructive Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kiron Koshy
- Department of Plastic & Reconstructive Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - W P Andrew Lee
- Department of Plastic & Reconstructive Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ahmet Hoke
- Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| | - Sami Tuffaha
- Department of Plastic & Reconstructive Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Gerald Brandacher
- Department of Plastic & Reconstructive Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
7
|
Aghanoori MR, Smith DR, Shariati-Ievari S, Ajisebutu A, Nguyen A, Desmond F, Jesus CHA, Zhou X, Calcutt NA, Aliani M, Fernyhough P. Insulin-like growth factor-1 activates AMPK to augment mitochondrial function and correct neuronal metabolism in sensory neurons in type 1 diabetes. Mol Metab 2019; 20:149-165. [PMID: 30545741 PMCID: PMC6358538 DOI: 10.1016/j.molmet.2018.11.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 11/20/2018] [Accepted: 11/23/2018] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE Diabetic sensorimotor polyneuropathy (DSPN) affects approximately half of diabetic patients leading to significant morbidity. There is impaired neurotrophic growth factor signaling, AMP-activated protein kinase (AMPK) activity and mitochondrial function in dorsal root ganglia (DRG) of animal models of type 1 and type 2 diabetes. We hypothesized that sub-optimal insulin-like growth factor 1 (IGF-1) signaling in diabetes drives loss of AMPK activity and mitochondrial function, both contributing to development of DSPN. METHODS Age-matched control Sprague-Dawley rats and streptozotocin (STZ)-induced type 1 diabetic rats with/without IGF-1 therapy were used for in vivo studies. For in vitro studies, DRG neurons from control and STZ-diabetic rats were cultured and treated with/without IGF-1 in the presence or absence of inhibitors or siRNAs. RESULTS Dysregulation of mRNAs for IGF-1, AMPKα2, ATP5a1 (subunit of ATPase), and PGC-1β occurred in DRG of diabetic vs. control rats. IGF-1 up-regulated mRNA levels of these genes in cultured DRGs from control or diabetic rats. IGF-1 treatment of DRG cultures significantly (P < 0.05) increased phosphorylation of Akt, P70S6K, AMPK and acetyl-CoA carboxylase (ACC). Mitochondrial gene expression and oxygen consumption rate (spare respiratory capacity), ATP production, mtDNA/nDNA ratio and neurite outgrowth were augmented (P < 0.05). AMPK inhibitor, Compound C, or AMPKα1-specific siRNA suppressed IGF-1 elevation of mitochondrial function, mtDNA and neurite outgrowth. Diabetic rats treated with IGF-1 exhibited reversal of thermal hypoalgesia and, in a separate study, reversed the deficit in corneal nerve profiles. In diabetic rats, IGF-1 elevated the levels of AMPK and P70S6K phosphorylation, raised Complex IV-MTCO1 and Complex V-ATP5a protein expression, and restored the enzyme activities of Complex IV and I in the DRG. IGF-1 prevented TCA metabolite build-up in nerve. CONCLUSIONS In DRG neuron cultures IGF-1 signals via AMPK to elevate mitochondrial function and drive axonal outgrowth. We propose that this signaling axis mediates IGF-1-dependent protection from distal dying-back of fibers in diabetic neuropathy.
Collapse
Affiliation(s)
- Mohamad-Reza Aghanoori
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada; Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB, Canada
| | - Darrell R Smith
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
| | - Shiva Shariati-Ievari
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Canada
| | - Andrew Ajisebutu
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
| | - Annee Nguyen
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Fiona Desmond
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Carlos H A Jesus
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Xiajun Zhou
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Nigel A Calcutt
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Michel Aliani
- Department of Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada; Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Canada; Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada
| | - Paul Fernyhough
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada; Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
8
|
Wang H, Zhang H, Cao F, Lu J, Tang J, Li H, Zhang Y, Feng B, Tang Z. Protection of insulin‑like growth factor 1 on experimental peripheral neuropathy in diabetic mice. Mol Med Rep 2018; 18:4577-4586. [PMID: 30221656 DOI: 10.3892/mmr.2018.9435] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Accepted: 07/19/2018] [Indexed: 11/05/2022] Open
Abstract
The present study investigated whether insulin‑like growth factor‑1 (IGF‑1) exerts a protective effect against neuropathy in diabetic mice and its potential underlying mechanisms. Mice were divided into four groups: Db/m (control), db/db (diabetes), IGF‑1‑treated db/db and IGF‑1‑picropodophyllin (PPP)‑treated db/db. Behavioral studies were conducted using the hot plate and von Frey methods at 6 weeks of age prior to treatment. The motor nerve conduction velocity (NCV) of the sciatic nerve was measured using a neurophysiological method at 8 weeks of age. The alterations in the expression levels of IGF‑1 receptor (IGF‑1R), c‑Jun N‑terminal kinase (JNK), extracellular signal‑regulated kinase (ERK), p38 and effect of IGF‑1 on the sciatic nerve morphology were observed by western blotting and electron microscopy. Compared with the control group, the diabetes group developed hypoalgesia after 12 weeks, and neurological lesions improved following an intraperitoneal injection of recombinant (r)IGF‑1. The sciatic NCV in the diabetes group was significantly lower compared with the control group. The sciatic NCV improved following rIGF‑1 intervention; however, was impaired following administration of the IGF‑1 receptor antagonist, PPP. The myelin sheath in the sciatic nerve of the diabetes group was significantly more impaired compared with the control group. The myelin sheath in the sciatic nerves of the rIGF‑1‑treated group was significantly improved compared with the diabetes group; whereas, they were significantly impaired following administration of the IGF‑1R inhibitor. In addition, the expression of IGF‑1R, phosphorylated (p)‑JNK and p‑ERK of sciatic nerves in the db/db mice was significantly increased following treatment with IGF‑1. The expression levels of these proteins were significantly lower in the IGF‑1‑PPP group compared with the IGF‑1 group; however, no significant difference was observed in the expression levels of p‑p38 following treatment with IGF‑1. The results of the present study demonstrated that IGF‑1 may improve neuropathy in diabetic mice. This IGF‑1‑induced neurotrophic effect may be associated with the increased phosphorylation levels of JNK and ERK, not p38; however, it was attenuated by administration of an IGF‑1R antagonist.
Collapse
Affiliation(s)
- Hua Wang
- Department of Endocrinology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Hao Zhang
- Department of Endocrinology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Fuming Cao
- Department of Endocrinology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Jiaping Lu
- Department of Endocrinology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Jin Tang
- Department of Endocrinology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Huizhi Li
- Department of Endocrinology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Yiyun Zhang
- Department of Endocrinology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Bo Feng
- Department of Endocrinology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Zhaosheng Tang
- Department of Endocrinology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| |
Collapse
|
9
|
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
|
10
|
Rauskolb S, Dombert B, Sendtner M. Insulin-like growth factor 1 in diabetic neuropathy and amyotrophic lateral sclerosis. Neurobiol Dis 2017; 97:103-113. [DOI: 10.1016/j.nbd.2016.04.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 03/29/2016] [Accepted: 04/29/2016] [Indexed: 12/12/2022] Open
|
11
|
Cui X, Chopp M, Zacharek A, Cui C, Yan T, Ning R, Chen J. D-4F Decreases White Matter Damage After Stroke in Mice. Stroke 2015; 47:214-20. [PMID: 26604250 DOI: 10.1161/strokeaha.115.011046] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 10/27/2015] [Indexed: 01/10/2023]
Abstract
BACKGROUND AND PURPOSE Stroke-induced neuroinflammation and white matter damage are associated with neurological deficits. Whether D-4F, an apolipoprotein A-I mimetic peptide, treatment of stroke decreases neuroinflammation and white matter damage and improves functional outcome has not been investigated. METHODS Adult male C57BL/6 mice were subjected to permanent middle cerebral artery occlusion (MCAo) and were orally administered saline as a vehicle control and different doses of D-4F (2, 4, 8, 16, or 32 mg/kg) starting at 2 h after MCAo and daily until euthanized at 7 days after MCAo. D-4F treatment did not alter the blood levels of high-density lipoprotein, total cholesterol, triglyceride, blood-brain barrier leakage, and infarction volume compared with control group. RESULTS D-4F (16 mg/kg) treatment of stroke significantly improved functional outcome, increased the white matter density and the number of oligodendrocyte progenitor cells in the ischemic boundary zone of the ipsilateral striatum, and increased myelin basic protein, insulin-like growth factor-1 (IGF1), but decreased inflammatory factor Toll-like receptor-4 and tumor necrosis factor-α expression in the ischemic brain 7 days after MCAo (P<0.05, n=11/group). The neurite/axonal outgrowth in primary cultured neurons was significantly increased when treated with D-4F (100 ng/mL) and IGF1 (100 ng/mL) compared with the nontreatment control. Inhibition of IGF1 significantly attenuated D-4F or IGF1 treatment-induced axonal outgrowth. D-4F-treatment did not increase oligodendrocyte-progenitor cell proliferation but decreased oligodendrocyte-progenitor cell death. CONCLUSIONS D-4F treatment initiated 2 h after MCAo decreases neuroinflammation and white matter damage and improves functional outcome after stroke. D-4F-induced increase in IGF1 may contribute to D-4F-induced neurite/axonal outgrowth after stroke.
Collapse
Affiliation(s)
- Xu Cui
- From the Department of Neurology, Henry Ford Hospital, Detroit, MI (X.C., M.C., A.Z., C.C., T.Y., R.N., J.C.); and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Michael Chopp
- From the Department of Neurology, Henry Ford Hospital, Detroit, MI (X.C., M.C., A.Z., C.C., T.Y., R.N., J.C.); and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Alex Zacharek
- From the Department of Neurology, Henry Ford Hospital, Detroit, MI (X.C., M.C., A.Z., C.C., T.Y., R.N., J.C.); and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Chengcheng Cui
- From the Department of Neurology, Henry Ford Hospital, Detroit, MI (X.C., M.C., A.Z., C.C., T.Y., R.N., J.C.); and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Tao Yan
- From the Department of Neurology, Henry Ford Hospital, Detroit, MI (X.C., M.C., A.Z., C.C., T.Y., R.N., J.C.); and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Ruizhuo Ning
- From the Department of Neurology, Henry Ford Hospital, Detroit, MI (X.C., M.C., A.Z., C.C., T.Y., R.N., J.C.); and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Jieli Chen
- From the Department of Neurology, Henry Ford Hospital, Detroit, MI (X.C., M.C., A.Z., C.C., T.Y., R.N., J.C.); and Department of Physics, Oakland University, Rochester, MI (M.C.).
| |
Collapse
|
12
|
Influence of insulin-like growth factor I on nerve regeneration using allografts: a sciatic nerve model. J Craniofac Surg 2015; 25:1510-4. [PMID: 25006924 DOI: 10.1097/scs.0000000000000783] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Effects of insulin-like growth factor I (IGF I) on peripheral nerve regeneration was studied using allografts in a rat sciatic nerve model. Thirty male white Wistar rats were divided into 3 experimental groups (n = 10) randomly: normal control group (NC), allograft group (ALLO), and IGF I-treated group (ALLO/IGF). In the NC group, the left sciatic nerve was exposed through a gluteal muscle incision and, after homeostasis, the muscle was sutured. In the ALLO group, the left sciatic nerve was exposed through a gluteal muscle incision and transected proximal to the tibioperoneal bifurcation where a 10-mm segment was excised. The same procedure was performed in the ALLO/IGF group. The harvested nerves of the rats of the ALLO group were served as allograft for the ALLO/IGF group and vice versa. The NC and ALLO groups received 10 μL of sterile phosphate buffered saline intraperitoneally once a day for 1 week, and the ALLO/IGF group received 10 μL of IGF I (100 ng/kg per day) intraperitoneally once a day for 1 week. Behavioral testing, sciatic nerve functional study and the gastrocnemius muscle mass showed earlier regeneration of axons in the ALLO/IGF group than in the ALLO group (P < 0.05). Administration of IGF I could accelerate functional recovery after nerve allografting in the sciatic nerve and may have clinical implications for the surgical management of patients after facial nerve transection.
Collapse
|
13
|
Abstract
Akt signalling has emerged as one of the major pathways involved in myelination, implicated in the regulation of several steps during the development of myelinating Schwann cells and oligodendrocytes. One of the main pathways intimately linked with Akt is mTOR [mammalian (or mechanistic) target of rapamycin] signalling. Recent evidence suggests that many processes attributed to the Akt pathway in myelination depend, at least partly, on mTOR signalling. In the present mini-review, we summarize the major aspects of Akt/mTOR signalling and myelination, and how they appear to be linked. We focus on the PNS (peripheral nervous system), but also cover the key points of CNS (central nervous system) myelination, pointing out differences and similarities between the PNS and the CNS.
Collapse
|
14
|
Effect of local administration of insulin-like growth factor I combined with inside-out artery graft on peripheral nerve regeneration. Injury 2013; 44:1295-301. [PMID: 23747124 DOI: 10.1016/j.injury.2013.04.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 04/09/2013] [Accepted: 04/14/2013] [Indexed: 02/02/2023]
Abstract
The objective was to assess the effect of topically administered insulin-like growth factor (IGF I) on peripheral nerve regeneration and functional recovery. Eighty male healthy white Wistar rats were divided into four experimental groups (n=20), randomly: in transected group (TC), the left sciatic nerve was transected and stumps were fixed in the adjacent muscle. In treatment group, defect was bridged using an inside-out artery graft (IOAG/IGF) filled with 10 μL IGF I (100 ng/kg). In artery graft group (IOAG), the graft was filled with phosphate-buffered saline alone. In sham-operated group (SHAM), sciatic nerve was exposed and manipulated. Each group was subdivided into five subgroups of five animals each and regenerated nerve fibres were studied 4, 8, 12 and 16 weeks after surgery. Behavioural testing, sciatic nerve functional study, gastrocnemius muscle mass and morphometric indices confirmed faster recovery of regenerated axons in IOAG/IGF than IOAG group (P<0.05). In immunohistochemistry, location of reactions to S-100 in IOAG/IGF was clearly more positive than that in IOAG group. When loaded in an artery graft, IGF I accelerated and improved functional recovery and morphometric indices of sciatic nerve.
Collapse
|
15
|
Myelination and node of Ranvier formation on sensory neurons in a defined in vitro system. In Vitro Cell Dev Biol Anim 2013; 49:608-618. [PMID: 23949775 DOI: 10.1007/s11626-013-9647-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Accepted: 05/29/2013] [Indexed: 10/26/2022]
Abstract
One of the most important developmental modifications of the nervous system is Schwann cell myelination of axons. Schwann cells ensheath axons to create myelin segments to provide protection to the axon as well as increase the conduction of action potentials. In vitro neuronal systems provide a unique modality to study a variety of factors influencing myelination as well as diseases associated with myelin sheath degradation. This work details the development of a patterned in vitro myelinating dorsal root ganglion culture. This defined system utilized a serum-free medium in combination with a patterned substrate, utilizing the cytophobic and cytophilic molecules (poly)ethylene glycol (PEG) and N-1[3 (trimethoxysilyl) propyl] diethylenetriamine (DETA), respectively. Directional outgrowth of the neurites and subsequent myelination was controlled by surface modifications, and conformity to the pattern was measured over the duration of the experiments. The myelinated segments and nodal proteins were visualized and quantified using confocal microscopy. This tissue-engineered system provides a highly controlled, reproducible model for studying Schwann cell interactions with sensory neurons, as well as the myelination process, and its effect on neuronal plasticity and peripheral nerve regeneration. It is also compatible for use in bio-hybrid constructs to reproduce the stretch reflex arc on a chip because the media combination used is the same that we have used previously for motoneurons, muscle, and for neuromuscular junction (NMJ) formation. This work could have application for the study of demyelinating diseases such as diabetes induced peripheral neuropathy and could rapidly translate to a role in the discovery of drugs promoting enhanced peripheral nervous system (PNS) remyelination.
Collapse
|
16
|
Sakowski SA, Feldman EL. Insulin-like growth factors in the peripheral nervous system. Endocrinol Metab Clin North Am 2012; 41:375-93, vii. [PMID: 22682636 DOI: 10.1016/j.ecl.2012.04.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Insulin-like growth factors (IGFs) play an integral role in development, growth, and survival. This article details the current understanding of the effects of IGFs in the peripheral nervous system (PNS) during health and disease, and introduces how the IGF system regulates PNS development and impacts growth and survival of PNS cells. Also discussed are implications of IGF signaling in neurodegeneration and the status and prospects of IGF therapies for PNS conditions. There is substantial support for the application of IGF therapies in the treatment of PNS injury and disease.
Collapse
Affiliation(s)
- Stacey A Sakowski
- A. Alfred Taubman Medical Research Institute, University of Michigan, 109 Zina Pitcher Place, 4019 AAT-BSRB, Ann Arbor, MI 48109, USA
| | | |
Collapse
|
17
|
Chirivella L, Cano-Jaimez M, Pérez-Sánchez F, Herraez L, Carretero J, Fariñas I, Burks DJ, Kirstein M. IRS2 signalling is required for the development of a subset of sensory spinal neurons. Eur J Neurosci 2012; 35:341-52. [PMID: 22288475 DOI: 10.1111/j.1460-9568.2011.07959.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Insulin and insulin-like growth factor-I play important roles in the development and maintenance of neurons and glial cells of the nervous system. Both factors activate tyrosine kinase receptors, which signal through adapter proteins of the insulin receptor substrate (IRS) family. Although insulin and insulin-like growth factor-I receptors are expressed in dorsal root ganglia (DRG), the function of IRS-mediated signalling in these structures has not been studied. Here we address the role of IRS2-mediated signalling in murine DRG. Studies in cultured DRG neurons from different embryonic stages indicated that a subset of nerve growth factor-responsive neurons is also dependent on insulin for survival at very early time points. Consistent with this, increased apoptosis during gangliogenesis resulted in a partial loss of trkA-positive neurons in DRG of Irs2 mutant embryos. Analyses in adult Irs2(-/-) mice revealed that unmyelinated fibre afferents, which express calcitonin gene-related peptide/substance P and isolectin B4, as well as some myelinated afferents to the skin were affected by the mutation. The diminished innervation of glabrous skin in adult Irs2(-/-) mice correlated with longer paw withdrawal latencies in the hot-plate assay. Collectively, these findings indicate that IRS2 signalling is required for the proper development of spinal sensory neurons involved in the perception of pain.
Collapse
Affiliation(s)
- Laura Chirivella
- Departamento de Biología Celular and Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, Universidad de Valencia, Doctor Moliner 50, 46100 Burjassot, Spain
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Ammoun S, Hanemann CO. Emerging therapeutic targets in schwannomas and other merlin-deficient tumors. Nat Rev Neurol 2011; 7:392-9. [DOI: 10.1038/nrneurol.2011.82] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
19
|
Miura M, Sasaki M, Mizukoshi K, Shibasaki M, Izumi Y, Shimosato G, Amaya F. Peripheral sensitization caused by insulin-like growth factor 1 contributes to pain hypersensitivity after tissue injury. Pain 2011; 152:888-895. [PMID: 21296499 DOI: 10.1016/j.pain.2011.01.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2010] [Revised: 12/03/2010] [Accepted: 01/04/2011] [Indexed: 01/25/2023]
Abstract
Sensitization of primary afferent neurons is one of the most important components of pain hypersensitivity after tissue injury. Insulin-like growth factor 1 (IGF-1), involved in wound repair in injured tissue, also plays an important role in maintaining neuronal function. In the present study, we investigated the effect of tissue IGF-1 on nociceptive sensitivity of primary afferent neurons. Local administration of IGF-1 induced thermal and mechanical pain hypersensitivity in a dose-dependent manner, and was attenuated by IGF-1 receptor (IGF1R) inhibition. Tissue but not plasma IGF-1 levels, as determined by enzyme-linked immunosorbent assay, significantly increased after plantar incision. Immunohistochemistry revealed that IGF1R was predominantly expressed in neurons as well as in satellite glial cells in the dorsal root ganglion (DRG). Double-labeling immunohistochemistry showed that IGF1R expression colocalized with peripherin and TRPV1, but not with NF200 in DRG neurons. The IGF1R inhibitor successfully alleviated mechanical allodynia, heat hyperalgesia, and spontaneous pain behavior observed after plantar incision. Expression of phosphorylated Akt in DRG neurons significantly increased after plantar incision and was suppressed by IGF1R inhibition. These results demonstrate that increased tissue IGF-1 production sensitizes primary afferent neurons via the IGF1R/Akt pathway to facilitate pain hypersensitivity after tissue damage.
Collapse
Affiliation(s)
- Mayumi Miura
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan Pain Mechanism Research Group, Kyoto, Japan
| | | | | | | | | | | | | |
Collapse
|
20
|
Xiang Y, Ding N, Xing Z, Zhang W, Liu H, Li Z. Insulin-like growth factor-1 regulates neurite outgrowth and neuronal migration from organotypic cultured dorsal root ganglion. Int J Neurosci 2010; 121:101-6. [PMID: 21110707 DOI: 10.3109/00207454.2010.535935] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
ABSTRACT Insulin-like growth factor-1 (IGF-1) is a neurotrophic factor and plays an important role in promoting axonal growth from neurons. Whether IGF-1 could promote neurite outgrowth and neuronal migration of dorsal root ganglion (DRG) explants in vitro remains unknown. In the present study, organotypic rat DRG explant culture model was established. Using this unique culture system, outgrowth of neurites from the peripheral nerve attached to DRG explant and migration of neurons from DRG explant to the peripheral area were quantified in the presence (5 nmol/L, 10 nmol/L, 20 nmol/L) or absence of IGF-1. The number of nerve fiber bundles extended from DRG explant increased significantly in the presence of IGF-1 (5 nmol/L, 19.25 ± 3.11, p < .05; 10 nmol/L, 20.92 ± 2.31, p < .01; 20 nmol/L, 23.00 ± 4.09, p < .001) as compared with that in the absence of IGF-1 (16.58 ± 2.94). The number of neurons migrated from DRG explant increased significantly in the presence of IGF-1 (5 nmol/L, 104.08 ± 16.70, p < .05; 10 nmol/L, 115.25 ± 13.68, p < .001; 20 nmol/L, 138.75 ± 18.05, p < .001) as compared with that in the absence of IGF-1 (90.25 ± 8.53). These data implicated that IGF-1 could promote neurite outgrowth and neuronal migration from DRG explants in vitro.
Collapse
Affiliation(s)
- Yujuan Xiang
- Faculty of Clinical Medicine, Shandong University School of Medicine, Jinan, China
| | | | | | | | | | | |
Collapse
|
21
|
Local insulin-like growth factor I expression is essential for Purkinje neuron survival at birth. Cell Death Differ 2010; 18:48-59. [PMID: 20596079 DOI: 10.1038/cdd.2010.78] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
IGF1, an anabolic and neuroprotective factor, promotes neuronal survival by blocking apoptosis. It is released into the bloodstream by the liver, or synthesized locally by muscles and neural cells, acting in an autocrine or paracrine fashion. Intriguingly, genetic studies conducted in invertebrate and murine models also suggest that an excess of IGF1 signaling may trigger neurodegeneration. This emphasizes the importance of gaining a better understanding of the mechanisms controlling IGF1 regulation and gene transcription. In the cerebellum, Igf1 expression is activated just before birth in a subset of Purkinje cells (PCs). Mice carrying a null mutation for HLH transcription factor EBF2 feature PC apoptosis at birth. We show that Igf1 is sharply downregulated in Ebf2 null PCs starting before the onset of PC death. In vitro, EBF2 binds a conserved distal Igf1 promoter region. The pro-survival PI3K signaling pathway is strongly inhibited in mutant cerebella. Finally, Ebf2 null organotypic cultures respond to IGF1 treatment by inhibiting PC apoptosis. Consistently, wild type slices treated with an IGF1 competitor feature a sharp increase in PC death. Our findings reveal that IGF1 is required for PC survival in the neonatal cerebellum, and identify a new mechanism regulating its local production in the CNS.
Collapse
|
22
|
Resmini E, Tagliafico A, Nizzo R, Bianchi F, Minuto F, Derchi L, Martinoli C, Ferone D. Ultrasound of peripheral nerves in acromegaly: changes at 1-year follow-up. Clin Endocrinol (Oxf) 2009; 71:220-5. [PMID: 19178522 DOI: 10.1111/j.1365-2265.2008.03468.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
CONTEXT We have previously demonstrated peripheral nerve enlargement in acromegaly. OBJECTIVE The aim of this study was to use ultrasound (US) to assess any changes in the peripheral nerves of patients with acromegaly 1 year after the first evaluation. PATIENTS We prospectively examined the median and ulnar nerve cross-sectional area (CSA) in 34 non-diabetic, patients with acromegaly (18 females and 16 males; 18-79 years) and 34 age-, sex-, BMI-matched controls, using a 17-5 MHz US probe. INTERVENTION The median nerve was examined at the mid-forearm (MN-f) and at the carpal tunnel (MN-Ct) levels; the ulnar nerve at mid-forearm (UN-f) and at distal arm (UN-a). Patients were grouped according to the clinical control of the disease: 'improved'; 'always controlled'; 'always uncontrolled'; and 'worsened'. RESULTS The median nerve at mid-forearm (MN-f), the ulnar nerve at mid-forearm (UN-f) and at distal arm (UN-a) were significantly reduced after 1-year follow-up in all patients (P < 0.001, P < 0.008, P < 0.012, respectively). In the 'improved' group, there was a significant reduction of median nerve CSA examined at mid-forearm (MN-f) (P = 0.02), and distal arm ulnar nerve CSA (UN-a) (P = 0.002). In the other groups no statistically significant differences in ultrasound parameters were recorded. However, UN-a, UN-f, MN-f, MN-ct were still significantly higher in all groups compared with controls (P < 0.001). CONCLUSION These data demonstrate that median and ulnar nerves CSA are reduced after 1 year follow-up, in line with the reduction of GH/IGF-I levels. However, as the control of the disease incompletely reverts nerve enlargement, this phenomenon could be only partially reversible.
Collapse
Affiliation(s)
- Eugenia Resmini
- Department of Endocrinology and Medical Sciences (DiSEM), Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
IGF-I and -II are potent neuronal mitogens and survival factors. The actions of IGF-I and -II are mediated via the type I IGF receptor (IGF-IR) and IGF binding proteins regulate the bioavailability of the IGFs. Cell viability correlates with IGF-IR expression and intact IGF-I/IGF-IR signaling pathways, including activation of MAPK/phosphatidylinositol-3 kinase. The expression of IGF-I and -II, IGF-IR, and IGF binding proteins are developmentally regulated in the central and peripheral nervous system. IGF-I therapy demonstrates mixed therapeutic results in the treatment of peripheral nerve injury, neuropathy, and motor neuron diseases such as amyotrophic lateral sclerosis. In this review we discuss the role of IGFs during peripheral nervous system development and the IGF signaling system as the potential therapeutic target for the treatment of nerve injury and motor neuron diseases.
Collapse
Affiliation(s)
- Kelli A Sullivan
- Department of Neurology, University of Michigan, Ann Arbor, Michigan 48109-2200, USA
| | | | | |
Collapse
|
24
|
Systemic Insulin-like Growth Factor-1 Reverses Hypoalgesia and Improves Mobility in a Mouse Model of Diabetic Peripheral Neuropathy. Mol Ther 2008; 16:1400-8. [DOI: 10.1038/mt.2008.115] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
|
25
|
Anjaneyulu M, Berent-Spillson A, Inoue T, Choi J, Cherian K, Russell JW. Transforming growth factor-beta induces cellular injury in experimental diabetic neuropathy. Exp Neurol 2008; 211:469-79. [PMID: 18406405 DOI: 10.1016/j.expneurol.2008.02.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2007] [Revised: 02/08/2008] [Accepted: 02/16/2008] [Indexed: 11/27/2022]
Abstract
The mechanism/s leading to diabetic neuropathy are complex. Transforming growth factor-beta1 (TGF-beta1) has been associated with diabetic nephropathy and retinopathy but not neuropathy. In this study, changes in TGF-beta isoforms were examined in vivo and in vitro. Two groups of animals, streptozotocin diabetic with neuropathy and non-diabetic controls were examined at 4 weeks (n=10/group) and 12 weeks (n=8/group). In diabetic DRG using quantitative real-time PCR (QRT-PCR), TGF-beta1 and TGF-beta2 mRNA, but not TGF-beta3, was increased at 4 and 12 weeks. In sciatic nerve TGF-beta3 mRNA was primarily increased. Immunohistochemistry (DRG) and immunoblotting (sciatic nerve) showed similar differential protein expression. In sciatic nerve TGF-beta formed homo- and hetero-dimers, of which beta(2)/beta(3), beta(1)/beta(1), and beta(1)/beta(3) were significantly increased, while that of the TGF-beta(2)/beta(2) homodimer was decreased, in diabetic compared to non-diabetic rats. In vitro, pretreatment of embryonic DRG with TGF-beta neutralizing antibody prevents the increase in total TGF-beta protein observed with high glucose using immunoblotting. In high glucose conditions, combination with TGF-beta2>beta1 increases the percent of cleaved caspase-3 compared to high glucose alone and TGF-beta neutralizing antibody inhibits this increase. Furthermore, consistent with the findings in diabetic DRG and nerve, TGF-beta isoforms applied directly in vitro reduce neurite outgrowth, and this effect is partially reversed by TGF-beta neutralizing antibody. These findings implicate upregulation of TGF-beta in experimental diabetic peripheral neuropathy and indicate a novel mechanism of cellular injury related to elevated glucose levels. In combination, these findings indicate a potential new target for treatment of diabetic peripheral neuropathy.
Collapse
Affiliation(s)
- Muragundla Anjaneyulu
- Department of Neurology, University of Maryland, School of Medicine, 22 South Greene Street, Box 175, Baltimore, MD 21201-1595, USA
| | | | | | | | | | | |
Collapse
|
26
|
Shokouhi G, Tubbs RS, Shoja MM, Roshangar L, Mesgari M, Ghorbanihaghjo A, Ahmadi N, Sheikhzadeh F, Rad JS. The effects of aerobic exercise training on the age-related lipid peroxidation, Schwann cell apoptosis and ultrastructural changes in the sciatic nerve of rats. Life Sci 2008; 82:840-6. [PMID: 18336840 DOI: 10.1016/j.lfs.2008.01.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2007] [Revised: 12/10/2007] [Accepted: 01/25/2008] [Indexed: 12/20/2022]
Abstract
The potential role of exercise in preventing the age-related spontaneous peripheral neuropathy has not been studied. We examined the effects of long-term aerobic exercise training on lipid peroxidation, Schwann cell (SC) apoptosis and ultrastructural changes in the sciatic nerve of rats during aging. Three groups of 12-week old Wistar rats ran on a treadmill for 6, 9 and 12 months (exercise trained (ET) group, n=10 each) according to an exercise training program targeted at a speed of 22 m/min (at 7 degrees incline), 60 min/day, 6 days/week. Three corresponding groups of untrained rats were used as the controls (sedentary (SED) group). At the end of each period, sciatic nerve biopsies were performed, and processed for biochemical, immunohistochemical and ultrastructural analyses. The results showed that aging was associated with an increased level of nerve malondialdehyde (MDA, marker of lipid peroxidation) and a higher number of SC apoptosis in SED group. The SED group showed irregular nerve fibers with thin myelin sheaths and areas of myelin-axon detachment. However, the ET group had significantly diminished nerve lipid peroxidation and SC apoptosis. In the ET group, nerve fibers had a thick myelin sheath with frequent folding. These findings suggest that aerobic exercise training protects peripheral nerves by attenuating oxidative reactions, and preserving SCs and myelin sheath from pathologic changes, which occur during normal aging.
Collapse
Affiliation(s)
- Ghaffar Shokouhi
- Department of Neurosurgery, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
p38 Mitogen-activated protein kinase regulates myelination. J Mol Neurosci 2007; 35:23-33. [PMID: 17994198 DOI: 10.1007/s12031-007-9011-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2007] [Accepted: 09/15/2007] [Indexed: 12/16/2022]
Abstract
The p38 mitogen-activated protein kinase family is emerging as a crucial signaling molecule for a vast number of cellular functions including cell migration, proliferation, and differentiation. The function of p38 in myelination has only been recently addressed. Using pyridinyl imidazole-based p38 alpha/beta selective inhibitors, we have reported a critical role for this kinase in the regulation of myelination, specifically, in controlling the differentiation of Schwann cells, and oligodendrocytes, the myelinating glia of the peripheral and central nervous systems, respectively. These compounds inhibited the accumulation of myelin-cell-specific markers, including myelin-specific glycosphingolipids, myelin-associated glycoprotein, and myelin basic protein. More significantly, myelination of dorsal root ganglia neurons by oligodendrocytes was irreversibly blocked by p38 inhibitors. Our current studies are focusing on the molecular mechanisms by which p38 regulates oligodendrocyte and Schwann cell differentiation and its role in models of myelination and remyelination.
Collapse
|
28
|
Liang G, Cline GW, Macica CM. IGF-1 stimulates de novo fatty acid biosynthesis by Schwann cells during myelination. Glia 2007; 55:632-41. [PMID: 17299765 DOI: 10.1002/glia.20496] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Schwann cell (SC) differentiation to the myelinating phenotype is characterized by the elaboration of a lipid-rich membrane and the expression of myelin-specific proteins. Insulin-like growth factor-1 (IGF-1) has been identified as a growth factor that stimulates the early events of myelination in SCs that signals via the PI3K/Akt pathway. Given the role of IGF-1 in promoting myelination, we performed studies to determine if the fatty acid biosynthetic pathway was a target of IGF-1 signaling in the formation of myelin membrane in dorsal root ganglion neuron/Schwann cell (DRG/SC) cocultures. We report that the fatty acid profile of lipid extracts of cocultures treated with IGF-1 match that reported for native myelin membrane by electrospray mass spectroscopy analysis. We also demonstrate de novo fatty acid biosynthesis in response to IGF-1 treatment in DRG/SC cocultures metabolically labeled with (13)C-acetate as a carbon source for fatty acid synthesis. Consistent with this finding, Western blot analysis of lysates from both cocultures and purified SCs reveal that IGF-1 stimulates two key fatty acid synthesizing enzymes. Additionally, we show that stimulation of fatty acid synthesizing enzymes is mediated by the PI3K/Akt signaling pathway. We also show that the fatty acid synthesizing enzymes and associated signaling pathways are elevated during the period of myelin membrane formation in sciatic nerve. Collectively, these findings demonstrate that IGF-1 plays an important regulatory function during myelin membrane formation.
Collapse
Affiliation(s)
- Guoying Liang
- Division of Endocrinology, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06520-8020, USA
| | | | | |
Collapse
|
29
|
Ogata T, Yamamoto SI, Nakamura K, Tanaka S. Signaling axis in schwann cell proliferation and differentiation. Mol Neurobiol 2006; 33:51-62. [PMID: 16388110 DOI: 10.1385/mn:33:1:051] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2005] [Accepted: 06/09/2005] [Indexed: 12/18/2022]
Abstract
Recent progress in molecular biology has markedly expanded our knowledge of the molecular mechanism behind the proliferation and differentiation processes of Schwann cells, the myelin-forming cells in peripheral nervous systems. Intracellular signaling molecules participate in integrating various stimuli from cytokines and other humoral factors and control the transcriptional activities of the genes that regulate mitosis or differentiation. This article provides an overview of the roles played by the intracellular pathways regulating Schwann cell functions. In Schwann cell proliferation, cyclic adenosine monophosphate signals and mitogen-activated protein kinase pathways play pivotal roles and may also interact with each other. Regarding differentiation, myelin formation is regulated by various cytokines and extracellular matrix molecules. Specifically, platelet-derived growth factor, neuregulin, and insulin-like growth factor-I all are classified as ligands for receptor-type tyrosine kinase and activate common intracellular signaling cascades, mitogen-activated protein kinase pathways, and phosphatidylinositol-3-kinase pathways. The balance of activities between these two pathways appears crucial in regulating Schwann cell differentiation, in which phosphatidylinositol-3-kinase pathways promote myelin formation. Analysis of these signaling molecules in Schwann cells will enable us not only to understand their physiological development but also to innovate new approaches to treat disorders related to myelination.
Collapse
Affiliation(s)
- Toru Ogata
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | | | | | | |
Collapse
|
30
|
Ogata T, Iijima S, Hoshikawa S, Miura T, Yamamoto SI, Oda H, Nakamura K, Tanaka S. Opposing extracellular signal-regulated kinase and Akt pathways control Schwann cell myelination. J Neurosci 2005; 24:6724-32. [PMID: 15282275 PMCID: PMC6729716 DOI: 10.1523/jneurosci.5520-03.2004] [Citation(s) in RCA: 195] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Schwann cells are the myelinating glia of the peripheral nervous system, and their development is regulated by various growth factors, such as neuregulin, platelet-derived growth factor (PDGF), and insulin-like growth factor-I (IGF-I). However, the mechanism of intracellular signaling pathways following these ligand stimuli in Schwann cell differentiation remains elusive. Here, we demonstrate that in cultured Schwann cells, neuregulin and PDGF suppressed the expression of myelin-associated protein markers, whereas IGF-I promoted it. Although these ligands activated common downstream signaling pathways [i.e., extracellular signal-regulated kinase (Erk) and phosphatidylinositol-3-kinase (PI3K)-Akt pathways], the profiles of activation varied among ligands. To elucidate the function of these pathways and the mechanisms underlying Schwann cell differentiation, we used adenoviral vectors to selectively activate or inactivate these pathways. We found that the selective activation of Erk pathways suppressed Schwann cell differentiation, whereas that of PI3K pathways promoted it. Furthermore, lithium chloride, a modulator of glycogen synthase kinase-3beta (GSK-3beta) promoted Schwann cell differentiation, suggesting the involvement of GSK-3beta as a downstream molecule of PI3K-Akt pathways. Selective activation of PI3K pathways in Schwann cells by gene transfer also demonstrated increased myelination in in vitro Schwann cell-DRG neuron cocultures and in vivo allogenic nerve graft experiments. We conclude that signals mediated by PI3K-Akt are crucial for initiation of myelination and that the effects of growth factors are primarily dependent on the balance between Erk and PI3K-Akt activation. Our results also propose the possibility of augmenting Schwann cell functions by modulating intracellular signals in light of future cell therapies.
Collapse
Affiliation(s)
- Toru Ogata
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Rabinovsky ED, Draghia-Akli R. Insulin-like growth factor I plasmid therapy promotes in vivo angiogenesis. Mol Ther 2004; 9:46-55. [PMID: 14741777 DOI: 10.1016/j.ymthe.2003.10.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Angiogenesis, the formation of neovessels from the endothelium of preexisting vessels, is stimulated by soluble angiogenic factors. Insulin-like growth factor I (IGF-I) stimulates myogenesis and induces nerve regeneration after injury, and it has been shown to stimulate angiogenesis. However, the in vivo angiogenic effects of IGF-I in regenerating and diabetic muscle have yet to be described. Therefore, we studied the effects of human IGF-I (hIGF-I) delivered by a plasmid-mediated therapy on angiogenesis in mouse models of these two conditions. Plasmid hIGF-I was delivered to the injured tibialis muscle by direct intramuscular injection followed by electroporation. Initial experiments compared two muscle-specific hIGF-I-expressing constructs containing either a skeletal actin 3'UTR (pAV2001) or a human growth hormone (GH) 3'UTR (pAV2002). Skeletal actin 3'UTR mediates sequestration of hIGF-I in the muscle and was more active, while the GH 3'UTR mediated release of IGF-I into the circulation. Treatment of regenerating muscle with pAV2001 and sequestration of IGF-I in muscle led to increased expression of vascular endothelial growth factor (VEGF) and VEGF receptors fetal liver kinase-1 and FmS-like tyrosine kinase receptor-1, as well as platelet endothelial cell adhesion molecule-1, on endothelial cells. These results indicate that IGF-I can amplify angiogenic responses in regenerating muscle. In a mouse diabetic model, plasmid-mediated IGF-I therapy reversed diabetic microangiopathy, as shown by increased angiogenesis and arterial flow as analyzed by Doppler imaging. These studies show that plasmid IGF-I delivery and sequestration in muscle can augment angiogenesis in regenerating muscle and increase blood flow and angiogenesis in the diabetic limb.
Collapse
Affiliation(s)
- Eric D Rabinovsky
- Division of Plastic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas 77030, USA.
| | | |
Collapse
|
32
|
Fragoso G, Robertson J, Athlan E, Tam E, Almazan G, Mushynski WE. Inhibition of p38 mitogen-activated protein kinase interferes with cell shape changes and gene expression associated with Schwann cell myelination. Exp Neurol 2003; 183:34-46. [PMID: 12957486 DOI: 10.1016/s0014-4886(03)00101-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In the present study we demonstrate that p38, a member of the mitogen-activated protein kinase (MAPK) family, is essential for ascorbate- and laminin-induced myelination in Schwann cell-dorsal root ganglion neuron cocultures. The inhibitory effect of the specific p38 blockers, PD 169316 and SB 203580, on ascorbate-induced myelination was exerted during the early stages (1-2 days) of ascorbate treatment. Inhibition of p38 was further shown to prevent the alignment of Schwann cells along axons in laminin-treated cocultures. The addition of laminin to Schwann cell-dorsal root ganglion neuron cocultures stimulated phosphorylation of p38, thereby demonstrating a link between laminin-induced myelination and p38 activation. Similarly, the small heat shock protein, Hsp27, which is phosphorylated by MAPKAPK2, a downstream substrate of p38, was phosphorylated in response to the addition of laminin to the cocultures. The p38 inhibitors did not affect the proliferation or survival of Schwann cells in the cocultures as assessed by BrdU incorporation and total cell counts. However, p38 inhibition interfered with an early stage in myelination, thereby preventing ascorbate-induced increases in the levels of mRNAs encoding MBP, MAG, and P(0) and reducing laminin deposition. These results indicate that activation of p38 by a signaling pathway(s) involving laminin and appropriate integrin receptor(s) is required for the alignment of Schwann cells with axons that precedes myelination.
Collapse
Affiliation(s)
- Gabriela Fragoso
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | | | | | | | | | | |
Collapse
|
33
|
Abstract
Protein zero (P0) is an integral transmembrane glycoprotein that serves as the major protein component of peripheral nerve myelin and is a member of the immunoglobulin (IgG) gene superfamily. As a cell adhesion molecule, P0 mediates homophilic adhesive interactions between Schwann cell plasma membranes and is a key structural constituent of both the major dense line and intraperiod line of compact myelin. Both the extracellular and cytoplasmic domains contribute to these interactions and evidence indicates that the post-translational modifications of the molecule, including glycosylation, acylation and phosphorylation, play an important modulatory role in adhesion and likely in the proper trafficking of P0 from the endoplasmic reticulum to the plasma membrane as well. Structural and genetic studies indicate that mutations in P0 producing human demyelinating diseases probably do so by perturbing or preventing homophilic interactions during myelination, or by producing cellular toxicity or an unstable myelin sheath. A variety of transcription factors, growth factors and neurosteroids both directly and indirectly influence P0 gene expression during maturation of the myelinating Schwann cell. Besides its structural function in myelin, P0 may have roles in the delivery of other Schwann cell proteins to their proper location, especially at or near nodes of Ranvier, and in neuronal-glial interactions.
Collapse
Affiliation(s)
- Joseph Eichberg
- Department of Biology and Biochemistry, University of Houston, Houston, Texas 77204, USA.
| |
Collapse
|
34
|
Abstract
Recent evidence in both animal models and human sural nerve biopsies indicates an association with oxidative stress, mitochondrial (Mt) membrane depolarization (MMD), and induction of programmed cell death (PCD). In streptozotocin (STZ)-treated diabetic rats, hyperglycemia induces typical apoptotic changes as well as swelling and disruption of the Mt cristae in diabetic dorsal root ganglion neurons (DRG) and Schwann cells (SC), but these changes are only rarely observed in control neurons. In human sural nerve biopsies, from patients with diabetic sensory neuropathy, there is transmission electromicrograph evidence of swelling and disruption of the Mt and cristae compared to patients without peripheral neuropathy. In human SH-SY5Y neurons, rat sensory neurons, and SC, in vivo, there is an increase in reactive oxygen species (ROS) after exposure to 20 mM added glucose. In parallel, there is an initial Mt membrane hyperpolarization followed by depolarization (MMD). In turn, MMD is coupled with cleavage of caspases. Various strategies aimed at inhibiting the oxidative burst, or stabilizing the DeltaPsi(M), block induction of PCD. First, growth factors such as NGF can block induction of ROS and/or stabilize the DeltaPsi(M). This, in turn, is associated with inhibition of PCD. Second, reduction of ROS generation in neuronal Mt prevents neuronal PCD. Third, up-regulation of uncoupling proteins (UCPs), which stabilize the DeltaPsi(M), blocks induction of caspase cleavage. Collectively, these findings indicate that hyperglycemic conditions observed in diabetes mellitus are associated with oxidative stress-induced neuronal and SC death, and targeted therapies aimed at regulating ROS may prove effective in therapy of diabetic neuropathy.
Collapse
Affiliation(s)
- Andrea M Vincent
- Department of Neurology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | |
Collapse
|
35
|
Mathonnet M, Comte I, Lalloué F, Ayer-Le Lièvre C. Insulin-like growth factor I induced survival of axotomized olfactory neurons in the chick. Neurosci Lett 2001; 308:67-70. [PMID: 11457561 DOI: 10.1016/s0304-3940(01)01715-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Insulin-like growth factor 1 (IGF1) receptor is expressed in avian olfactory neurons and IGF1 in the bulb. To explore the function of IGF1 in olfactory system in the chick, we infused IGF1 at the lesion site 0 and 12 h after olfactory axotomy. The animals were killed 1-3 days later. TdT mediated dUTP nick end labeling method and bromodeoxyuridine incorporation allowed the evaluation of programmed cell death and mitotic activity respectively in the olfactory epithelia of IGF1 treated or untreated lesioned animals and controls. IGF1 treatment suppressed the apoptotic wave, stimulated mitosis which peaked within 24 h (instead of 48 h), to return promptly to normal, and transiently maintained the number of calmodulin related kinase II expressing neurons at normal levels. It reveals a long lasting effect of IGF1 on the survival of lesioned olfactory neurons and transient effects on maintenance of differentiation and mitosis stimulation.
Collapse
Affiliation(s)
- M Mathonnet
- UMR-CNRS 6101-Faculté de Médecine, 2 rue du Dr Marcland, 87025 Limoges, cedex, France
| | | | | | | |
Collapse
|