1
|
Winias S, Sarasati A, Wicaksono S, Ayuningtyas NF, Ernawati DS, Radithia D. BDNF and Krox20 as Indicators of Platelet-rich Plasma-Induced Nerve Regeneration in a Neuropathic Orofacial Pain Model. Eur J Dent 2024; 18:131-137. [PMID: 37311554 PMCID: PMC10959617 DOI: 10.1055/s-0043-1761194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023] Open
Abstract
OBJECTIVE Various growth factors contained in PRP can increase angiogenesis and cell proliferation, which plays an essential role in the process of neuroregeneration and peripheral nerve injury recovery. This study analyzed PRP effects in the neuro-regeneration of axonotmesis through brain-derived neurotrophic factor (BDNF) and Krox20 expressions. MATERIALS AND METHODS Freeze-dried allogeneic platelet-rich plasma (PRP) were prepared from allogeneic sources. Forty-two Rattus norvegicus were divided into three groups: negative control group, positive control group (crushing infraorbital nerve) and treatment group (crushing infraorbital nerve without PRP injection). Each group was observed for fourteen and twenty-one days after injury. Infraorbital nerve tissue is isolated for indirect immunohistochemistry examination with BDNF and Krox20 antibodies. Data analysis was performed using One-Way ANOVA and Mann-Whitney tests with significant value as p < 0.05. RESULTS The PRP group showed BDNF expression significantly higher than control positive groups, both observation days (p = 0.00). A higher Korx20 expression showed by the PRP group after 21 days than in the control positive groups (p = 0.002). CONCLUSION PRP can potentially improve neuroregeneration of axonotmesis through increased BDNF and Krox20 expression on the twenty-one days after injury.
Collapse
Affiliation(s)
- Saka Winias
- Department of Oral Medicine, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Andari Sarasati
- Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Satutya Wicaksono
- Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | | | - Diah Savitri Ernawati
- Department of Oral Medicine, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Desiana Radithia
- Department of Oral Medicine, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| |
Collapse
|
2
|
Tientcheu JPD, Ngueguim FT, Gounoue RK, Mbock MA, Ngapout R, Kandeda AK, Dimo T. The extract of Sclerocarya birrea, Nauclea latifolia, and Piper longum mixture ameliorates diabetes-associated cognitive dysfunction. Metab Brain Dis 2023; 38:2773-2796. [PMID: 37821784 DOI: 10.1007/s11011-023-01291-7] [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: 05/23/2023] [Accepted: 09/06/2023] [Indexed: 10/13/2023]
Abstract
Diabetes-associated cognitive dysfunction is linked to chronic hyperglycemia, oxidative stress, inflammation, cholinergic dysfunction, and neuronal degeneration. We investigated the antidiabetic and neuroprotective activity of a mixture of Sclerocarya birrea, Nauclea latifolia, and Piper longum (SNP) in type 2 diabetic (T2D) rat model-induced memory impairment. Fructose (10%) and streptozotocin (35 mg/kg) were used to induce T2D in male Wistar rats. Diabetic animals received distilled water, metformin (200 mg/kg), or SNP mixture (75, 150, or 300 mg/kg). HPLC-MS profiling of the mixture was performed. Behavioral testing was conducted using the Y-maze, NORT, and Morris water mazes to assess learning and memory. Biochemical markers were evaluated, including carbohydrate metabolism, oxidative/nitrative stress, pro-inflammatory markers, and acetylcholinesterase activity. Histopathological examination of the pancreas and hippocampus was also performed. Fructose/STZ administration resulted in T2D, impaired short- and long-term memory, significantly increased oxidative/nitrative stress, pro-inflammatory cytokine levels, acetylcholinesterase activity (AChE), hippocampal neuronal loss and degeneration in CA1 and CA3 subfields, and neuronal vacuolation in DG. SNP mixture at 150 and 300 mg/kg significantly improved blood glucose and memory function in diabetic rats. The mixture reduced oxidative/nitrative stress and increased endogenous antioxidant levels. It also reduced serum IL-1β, INF-γ and TNF-α levels and ameliorated AChE activity. Histologically, SNP protected hippocampus neurons against T2D-induced neuronal necrosis and degeneration. We conclude that the aqueous extract of SNP mixture has antidiabetic and neuroprotective activities thanks to active metabolites identified in the plant mixture, which consequently normalized blood glucose, protected hippocampus neurons, and improved memory function in diabetic rats.
Collapse
Affiliation(s)
| | - Florence Tsofack Ngueguim
- Laboratory of Animal Physiology, Faculty of Science, University of Yaoundé 1, P.O. Box 812, Yaoundé, Cameroon.
| | - Racéline Kamkumo Gounoue
- Laboratory of Animal Physiology, Faculty of Science, University of Yaoundé 1, P.O. Box 812, Yaoundé, Cameroon
| | - Michel Arnaud Mbock
- Department of Biochemistry, Laboratory of Biochemistry, Faculty of Science, University of Douala, PO Box 24 157, Douala, Cameroon
| | - Rodrigue Ngapout
- Laboratory of Animal Physiology, Faculty of Science, University of Yaoundé 1, P.O. Box 812, Yaoundé, Cameroon
| | - Antoine Kavaye Kandeda
- Laboratory of Animal Physiology, Faculty of Science, University of Yaoundé 1, P.O. Box 812, Yaoundé, Cameroon
| | - Théophile Dimo
- Laboratory of Animal Physiology, Faculty of Science, University of Yaoundé 1, P.O. Box 812, Yaoundé, Cameroon
| |
Collapse
|
3
|
Greco M, Munir A, Musarò D, Coppola C, Maffia M. Restoring autophagic function: a case for type 2 diabetes mellitus drug repurposing in Parkinson's disease. Front Neurosci 2023; 17:1244022. [PMID: 38027497 PMCID: PMC10654753 DOI: 10.3389/fnins.2023.1244022] [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] [Received: 06/22/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Parkinson's disease (PD) is a predominantly idiopathic pathological condition characterized by protein aggregation phenomena, whose main component is alpha-synuclein. Although the main risk factor is ageing, numerous evidence points to the role of type 2 diabetes mellitus (T2DM) as an etiological factor. Systemic alterations classically associated with T2DM like insulin resistance and hyperglycemia modify biological processes such as autophagy and mitochondrial homeostasis. High glucose levels also compromise protein stability through the formation of advanced glycation end products, promoting protein aggregation processes. The ability of antidiabetic drugs to act on pathways impaired in both T2DM and PD suggests that they may represent a useful tool to counteract the neurodegeneration process. Several clinical studies now in advanced stages are looking for confirmation in this regard.
Collapse
Affiliation(s)
- Marco Greco
- Department of Biological and Environmental Science and Technology, University of Salento, Lecce, Italy
| | - Anas Munir
- Department of Biological and Environmental Science and Technology, University of Salento, Lecce, Italy
- Department of Mathematics and Physics “E. De Giorgi”, University of Salento, Lecce, Italy
| | - Debora Musarò
- Department of Biological and Environmental Science and Technology, University of Salento, Lecce, Italy
| | - Chiara Coppola
- Department of Biological and Environmental Science and Technology, University of Salento, Lecce, Italy
- Department of Mathematics and Physics “E. De Giorgi”, University of Salento, Lecce, Italy
| | - Michele Maffia
- Department of Biological and Environmental Science and Technology, University of Salento, Lecce, Italy
| |
Collapse
|
4
|
Zochodne DW. Growth factors and molecular-driven plasticity in neurological systems. HANDBOOK OF CLINICAL NEUROLOGY 2023; 196:569-598. [PMID: 37620091 DOI: 10.1016/b978-0-323-98817-9.00017-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/26/2023]
Abstract
It has been almost 70 years since the discovery of nerve growth factor (NGF), a period of a dramatic evolution in our understanding of dynamic growth, regeneration, and rewiring of the nervous system. In 1953, the extraordinary finding that a protein found in mouse submandibular glands generated a halo of outgrowing axons has now redefined our concept of the nervous system connectome. Central and peripheral neurons and their axons or dendrites are no longer considered fixed or static "wiring." Exploiting this molecular-driven plasticity as a therapeutic approach has arrived in the clinic with a slate of new trials and ideas. Neural growth factors (GFs), soluble proteins that alter the behavior of neurons, have expanded in numbers and our understanding of the complexity of their signaling and interactions with other proteins has intensified. However, beyond these "extrinsic" determinants of neuron growth and function are the downstream pathways that impact neurons, ripe for translational development and potentially more important than individual growth factors that may trigger them. Persistent and ongoing nuances in clinical trial design in some of the most intractable and irreversible neurological conditions give hope for connecting new biological ideas with clinical benefits. This review is a targeted update on neural GFs, their signals, and new therapeutic ideas, selected from an expansive literature.
Collapse
Affiliation(s)
- Douglas W Zochodne
- Division of Neurology, Department of Medicine and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
5
|
Poitras T, Zochodne DW. Unleashing Intrinsic Growth Pathways in Regenerating Peripheral Neurons. Int J Mol Sci 2022; 23:13566. [PMID: 36362354 PMCID: PMC9654452 DOI: 10.3390/ijms232113566] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/24/2022] [Accepted: 10/28/2022] [Indexed: 10/17/2023] Open
Abstract
Common mechanisms of peripheral axon regeneration are recruited following diverse forms of damage to peripheral nerve axons. Whether the injury is traumatic or disease related neuropathy, reconnection of axons to their targets is required to restore function. Supporting peripheral axon regrowth, while not yet available in clinics, might be accomplished from several directions focusing on one or more of the complex stages of regrowth. Direct axon support, with follow on participation of supporting Schwann cells is one approach, emphasized in this review. However alternative approaches might include direct support of Schwann cells that instruct axons to regrow, manipulation of the inflammatory milieu to prevent ongoing bystander axon damage, or use of inflammatory cytokines as growth factors. Axons may be supported by a growing list of growth factors, extending well beyond the classical neurotrophin family. The understanding of growth factor roles continues to expand but their impact experimentally and in humans has faced serious limitations. The downstream signaling pathways that impact neuron growth have been exploited less frequently in regeneration models and rarely in human work, despite their promise and potency. Here we review the major regenerative signaling cascades that are known to influence adult peripheral axon regeneration. Within these pathways there are major checkpoints or roadblocks that normally check unwanted growth, but are an impediment to robust growth after injury. Several molecular roadblocks, overlapping with tumour suppressor systems in oncology, operate at the level of the perikarya. They have impacts on overall neuron plasticity and growth. A second approach targets proteins that largely operate at growth cones. Addressing both sites might offer synergistic benefits to regrowing neurons. This review emphasizes intrinsic aspects of adult peripheral axon regeneration, emphasizing several molecular barriers to regrowth that have been studied in our laboratory.
Collapse
Affiliation(s)
| | - Douglas W. Zochodne
- Neuroscience and Mental Health Institute, Division of Neurology, Department of Medicine, University of Alberta, Edmonton, AB T6G 2G3, Canada
| |
Collapse
|
6
|
Chau DDL, Li W, Chan WWR, Sun JKL, Zhai Y, Chow HM, Lau KF. Insulin stimulates atypical protein kinase C-mediated phosphorylation of the neuronal adaptor FE65 to potentiate neurite outgrowth by activating ARF6-Rac1 signaling. FASEB J 2022; 36:e22594. [PMID: 36250347 DOI: 10.1096/fj.202200757r] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/21/2022] [Accepted: 09/26/2022] [Indexed: 11/11/2022]
Abstract
Neurite outgrowth is a fundamental process in neurons that produces extensions and, consequently, neural connectivity. Neurite damage and atrophy are observed in various brain injuries and disorders. Understanding the intrinsic pathways of neurite outgrowth is essential for developing strategies to stimulate neurite regeneration. Insulin is a pivotal hormone in the regulation of glucose homeostasis. There is increasing evidence for the neurotrophic functions of insulin, including the induction of neurite outgrowth. However, the associated mechanism remains elusive. Here, we demonstrate that insulin potentiates neurite outgrowth mediated by the small GTPases ADP-ribosylation factor 6 (ARF6) and Ras-related C3 botulinum toxin substrate 1 (Rac1) through the neuronal adaptor FE65. Moreover, insulin enhances atypical protein kinase Cι/λ (PKCι/λ) activation and FE65 phosphorylation at serine 459 (S459) in neurons and mouse brains. In vitro and cellular assays show that PKCι/λ phosphorylated FE65 at S459. Consistently, insulin potentiates FE65 S459 phosphorylation only in the presence of PKCι/λ. Phosphomimetic studies show that an FE65 S459E mutant potently activates ARF6, Rac1, and neurite outgrowth. Notably, this phosphomimetic mutation enhances the FE65-ARF6 interaction, a process that promotes ARF6-Rac1-mediated neurite outgrowth. Likewise, insulin treatment and PKCι/λ overexpression potentiate the FE65-ARF6 interaction. Conversely, PKCι/λ knockdown suppresses the stimulatory effect of FE65 on ARF6-Rac1-mediated neurite outgrowth. The effect of insulin on neurite outgrowth is also markedly attenuated in PKCι/λ knockdown neurons, in the presence and absence of FE65. Our findings reveal a novel mechanism linking insulin with ARF6-Rac1-dependent neurite extension through the PKCι/λ-mediated phosphorylation of FE65.
Collapse
Affiliation(s)
- Dennis Dik-Long Chau
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Wen Li
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, China.,Research Laboratory for Biomedical Optics and Molecular Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Wai Wa Ray Chan
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Jacquelyne Ka-Li Sun
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Yuqi Zhai
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Hei-Man Chow
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Kwok-Fai Lau
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
7
|
Yang JJ. Brain insulin resistance and the therapeutic value of insulin and insulin-sensitizing drugs in Alzheimer's disease neuropathology. Acta Neurol Belg 2022; 122:1135-1142. [PMID: 35482277 DOI: 10.1007/s13760-022-01907-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 02/17/2022] [Indexed: 02/06/2023]
Abstract
The incidence of Alzheimer's disease (AD) is significantly higher in people with diabetes. Insulin and insulin receptor (IR) signaling intermediates are expressed in the brain. Insulin exerts multiple function in the brain. The role of compromised IR signaling in AD pathogenesis and the therapeutic value of insulin attract broad attention. This review summarizes the collective insulin action in the brain related to key factors of AD pathogenesis, updates the key features of insulin resistance in the AD brain and assesses the therapeutic potential of insulin and insulin-sensitizing drugs. Insulin stimulates neural growth and survival, suppresses amyloidogenic processing of the amyloid precursor protein (AβPP) and inhibits the Tau phosphorylation kinase, glycogen synthase kinase 3β. Central nervous IR signaling regulates systemic metabolism and increases glucose availability to neurons. The expression of IR and its downstream effectors is reduced in AD brain tissues. Insulin and insulin-sensitizing drugs can improve cognitive function in AD patients and AD animal models. Systemic insulin delivery is less effective than intranasal insulin treatment. The penetrance of insulin-sensitizing drugs to the blood brain barrier is problematic and new brain-prone drugs need be developed. Insulin resistance manifested by the degradation and the altered phosphorylation of IR intermediates precedes overt AD syndrome. Type 3 diabetes as a pure form of brain insulin resistance without systemic insulin resistance is proposed as a causal factor in AD. Further research is needed for the identification of critical factors leading to impaired IR signaling and the development of new molecules to stimulate brain IR signaling.
Collapse
Affiliation(s)
- James J Yang
- Marriotts Ridge High School, 12100 Woodford Dr, Marriottsville, MD, 21104, USA.
- , 3060 Seneca Chief Trail, Ellicott City, MD, 21042, USA.
| |
Collapse
|
8
|
Morgunova GV, Shilovsky GA, Khokhlov AN. Effect of Caloric Restriction on Aging: Fixing the Problems of Nutrient Sensing in Postmitotic Cells? BIOCHEMISTRY. BIOKHIMIIA 2021; 86:1352-1367. [PMID: 34903158 DOI: 10.1134/s0006297921100151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The review discusses the role of metabolic disorders (in particular, insulin resistance) in the development of age-related diseases and normal aging with special emphasis on the changes in postmitotic cells of higher organisms. Caloric restriction helps to prevent such metabolic disorders, which could probably explain its ability to prolong the lifespan of laboratory animals. Maintaining metabolic homeostasis is especially important for the highly differentiated long-lived body cells, whose lifespan is comparable to the lifespan of the organism itself. Normal functioning of these cells can be ensured only upon correct functioning of the cytoplasm clean-up system and availability of all required nutrients and energy sources. One of the central problems in gerontology is the age-related disruption of glucose metabolism leading to obesity, diabetes, metabolic syndrome, and other related pathologies. Along with the adipose tissue, skeletal muscles are the main consumers of insulin; hence the physical activity of muscles, which supports their energy metabolism, delays the onset of insulin resistance. Insulin resistance disrupts the metabolism of cardiomyocytes, so that they fail to utilize the nutrients to perform their functions even being surrounded by a nutrient-rich environment, which contributes to the development of age-related cardiovascular diseases. Metabolic pathologies also alter the nutrient sensitivity of neurons, thus disrupting the action of insulin in the central nervous system. In addition, there is evidence that neurons can develop insulin resistance as well. It has been suggested that affecting nutritional sensors (e.g., AMPK) in postmitotic cells might improve the state of the entire multicellular organism, slow down its aging, and increase the lifespan.
Collapse
Affiliation(s)
- Galina V Morgunova
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia.
| | - Gregory A Shilovsky
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | | |
Collapse
|
9
|
Liu Y, Peng Y, Jin J, Chen Y, Chen C, Chen Z, Huang H, Xu L. Insulin resistance is independently associated with cardiovascular autonomic neuropathy in type 2 diabetes. J Diabetes Investig 2021; 12:1651-1662. [PMID: 33460512 PMCID: PMC8409868 DOI: 10.1111/jdi.13507] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/26/2020] [Accepted: 01/13/2021] [Indexed: 12/24/2022] Open
Abstract
AIMS/INTRODUCTION Diabetic cardiovascular autonomic neuropathy (DCAN) seriously threatens the prognosis and quality of life of patients with type 2 diabetes mellitus, associated with increased mortality. The present study aimed to investigate the relevant risk factors of DCAN. MATERIALS AND METHODS The present study enrolled a total of 109 patients with type 2 diabetes mellitus. DCAN was defined as a score of at least 2 points in Ewing tests. The updated homeostasis model assessment of insulin resistance (HOMA2-IR) based on fasting C-peptide was calculated to reflect insulin resistance. Logistic regression analysis, interaction and stratified analyses were used to investigate the relationship between HOMA2-IR or other indicators and DCAN. Receiver operating characteristic analysis was carried out to estimate the discriminative value of the variables independently associated with DCAN and to determine the optimal cut-off point of these models to screen DCAN. RESULTS The HOMA2-IR levels were significantly higher in patients with DCAN, and tended to be worsened with the progression of the DCAN. Logistic regression analysis showed an independent association between HOMA2-IR (odds ratio 39.30, 95% confidence interval 7.17-215.47) and DCAN. HOMA2-IR (area under the curve 0.878, 95% confidence interval 0.810-0.946; cut-off value 1.735) individually predicted DCAN significantly higher than the other independent risk factors individually used, whereas models combining HOMA2-IR and other risk factors did not significantly boost the diagnostic power. CONCLUSIONS Insulin resistance is independently associated with DCAN. HOMA2-IR presents to be a highly accurate and parsimonious indicator for DCAN screening. Patients with HOMA2-IR >1.735 are at a high risk of DCAN; thus, priority diagnostic tests should be carried out for these patients for timely integrated intervention.
Collapse
Affiliation(s)
- Yingshan Liu
- Department of EndocrinologyShenzhen HospitalSouthern Medical UniversityShenzhenChina
- The Third School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
| | - Yu Peng
- Department of NeurologyNanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Jing Jin
- Department of EndocrinologyShenzhen HospitalSouthern Medical UniversityShenzhenChina
- The Third School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
| | - Yanshan Chen
- Department of EndocrinologyShenzhen HospitalSouthern Medical UniversityShenzhenChina
- The Third School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
| | - Chuna Chen
- Department of EndocrinologyShenzhen HospitalSouthern Medical UniversityShenzhenChina
- The Third School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
| | - Zhenguo Chen
- Department of EndocrinologyShenzhen HospitalSouthern Medical UniversityShenzhenChina
- The Third School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
| | - Haishan Huang
- Department of EndocrinologyShenzhen HospitalSouthern Medical UniversityShenzhenChina
- The Third School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
| | - Lingling Xu
- Department of EndocrinologyShenzhen HospitalSouthern Medical UniversityShenzhenChina
- The Third School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
10
|
Kobayashi M, Zochodne DW. Diabetic polyneuropathy: Bridging the translational gap. J Peripher Nerv Syst 2021; 25:66-75. [PMID: 32573914 DOI: 10.1111/jns.12392] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/14/2020] [Accepted: 05/15/2020] [Indexed: 12/22/2022]
Abstract
Clinical trials for diabetic polyneuropathy (DPN) have failed to identify therapeutic impacts that have arrested or reversed the disorder, despite a long history. This review considers DPN in the context of a unique neurodegenerative disorder that targets peripheral neurons and their companion glial cells. The approach is to examine what cells, cell substructures, and pathways are implicated in causing DPN and how they might be addressed therapeutically. These include axonopathy, neuronopathy, hyperglycemia, polyol flux, advanced glycation endproduct (AGE)-receptor AGE signaling, growth factor disruption, abnormal insulin signaling, and abnormalities of other intrinsic neuron pathways. Mitochondrial dysfunction and lipid toxicity are largely delegated to the companion review in this issue by Stino and Feldman. Finally, the linkage between axon plasticity of cutaneous nerves, peripheral neuroregenerative pathways, and diabetes are discussed.
Collapse
Affiliation(s)
- Masaki Kobayashi
- Department of Neurology, Nissan Tamagawa Hospital, Tokyo, Japan.,Department of Neurology and Neurological Science, Tokyo Medical and Dental University, Tokyo, Japan
| | - Douglas W Zochodne
- Division of Neurology and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
11
|
Saiki T, Nakamura N, Miyabe M, Ito M, Minato T, Sango K, Matsubara T, Naruse K. The Effects of Insulin on Immortalized Rat Schwann Cells, IFRS1. Int J Mol Sci 2021; 22:ijms22115505. [PMID: 34071138 PMCID: PMC8197103 DOI: 10.3390/ijms22115505] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 01/02/2023] Open
Abstract
Schwann cells play an important role in peripheral nerve function, and their dysfunction has been implicated in the pathogenesis of diabetic neuropathy and other demyelinating diseases. The physiological functions of insulin in Schwann cells remain unclear and therefore define the aim of this study. By using immortalized adult Fischer rat Schwann cells (IFRS1), we investigated the mechanism of the stimulating effects of insulin on the cell proliferation and expression of myelin proteins (myelin protein zero (MPZ) and myelin basic protein (MBP). The application of insulin to IFRS1 cells increased the proliferative activity and induced phosphorylation of Akt and ERK, but not P38-MAPK. The proliferative potential of insulin-stimulated IFRS1 was significantly suppressed by the addition of LY294002, a PI3 kinase inhibitor. The insulin-stimulated increase in MPZ expression was significantly suppressed by the addition of PD98059, a MEK inhibitor. Furthermore, insulin-increased MBP expression was significantly suppressed by the addition of LY294002. These findings suggest that both PI3-K/Akt and ERK/MEK pathways are involved in insulin-induced cell growth and upregulation of MPZ and MBP in IFRS1 Schwann cells.
Collapse
Affiliation(s)
- Tomokazu Saiki
- Department of Pharmacy, Aichi Gakuin University Dental Hospital, Nagoya 464-8651, Japan;
| | - Nobuhisa Nakamura
- Department of Internal Medicine, School of Dentistry, Aichi Gakuin University, Nagoya 464-8651, Japan; (M.M.); (M.I.); (T.M.); (K.N.)
- Correspondence: ; Tel.: +81-52-759-2111; Fax: +81-52-759-2168
| | - Megumi Miyabe
- Department of Internal Medicine, School of Dentistry, Aichi Gakuin University, Nagoya 464-8651, Japan; (M.M.); (M.I.); (T.M.); (K.N.)
| | - Mizuho Ito
- Department of Internal Medicine, School of Dentistry, Aichi Gakuin University, Nagoya 464-8651, Japan; (M.M.); (M.I.); (T.M.); (K.N.)
| | - Tomomi Minato
- Department of Clinical Laboratory, Aichi Gakuin University Dental Hospital, Nagoya 464-8651, Japan;
| | - Kazunori Sango
- Diabetic Neuropathy Project, Department of Diseases and Infection, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan;
| | - Tatsuaki Matsubara
- Department of Internal Medicine, School of Dentistry, Aichi Gakuin University, Nagoya 464-8651, Japan; (M.M.); (M.I.); (T.M.); (K.N.)
| | - Keiko Naruse
- Department of Internal Medicine, School of Dentistry, Aichi Gakuin University, Nagoya 464-8651, Japan; (M.M.); (M.I.); (T.M.); (K.N.)
| |
Collapse
|
12
|
Komirishetty P, Zubkow K, Areti A, Ong H, Zochodne DW. Delayed manipulation of regeneration within injured peripheral axons. Neurobiol Dis 2021; 155:105383. [PMID: 33945876 DOI: 10.1016/j.nbd.2021.105383] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/20/2021] [Accepted: 04/29/2021] [Indexed: 10/21/2022] Open
Abstract
While several new translational strategies to enhance regrowth of peripheral axons have been identified, combined approaches with different targets are rare. Moreover, few have been studied after a significant delay when growth programs are already well established and regeneration-related protein expression has waned. Here we study two approaches, Rb1 (Retinoblastoma 1) knockdown that targets overall neuron plasticity, and near nerve insulin acting as a growth factor. Both are validated to boost regrowth only at the outset of regeneration. We show that local delivery of Rb1 siRNA alone, with electroporation to an area of prior sciatic nerve injury generated knockdown of Rb1 mRNA in ipsilateral lumbar dorsal root ganglia. While mice treated with Rb1-targeted siRNA, compared with scrambled control siRNA, starting 2 weeks after the onset of regeneration, had only limited behavioural or electrophysiological benefits, they had enhanced reinnervation of epidermal axons. We next confirmed that intrinsic Rb1 knockdown combined with exogenous insulin had dramatic synergistic impacts on the growth patterns of adult sensory neurons studied in vitro, prompting analysis of a combined approach in vivo. Using an identical delayed post-injury protocol, we noted that added insulin not only augmented epidermal reinnervation rendered by Rb1 knockdown alone but also improved indices of mechanical sensation and motor axon recovery. The findings illustrate that peripheral neurons that are well into attempted regrowth retain their responsiveness to both intrinsic and exogenous approaches that improve their recovery. We also identify a novel local approach to manipulate gene expression and outcome in regrowing axons.
Collapse
Affiliation(s)
- P Komirishetty
- Peripheral Nerve Research Laboratory, Division of Neurology, Department of Medicine and the Neuroscience and Mental Health Institute, University of Alberta, Canada
| | - K Zubkow
- Peripheral Nerve Research Laboratory, Division of Neurology, Department of Medicine and the Neuroscience and Mental Health Institute, University of Alberta, Canada
| | - A Areti
- Peripheral Nerve Research Laboratory, Division of Neurology, Department of Medicine and the Neuroscience and Mental Health Institute, University of Alberta, Canada
| | - H Ong
- Peripheral Nerve Research Laboratory, Division of Neurology, Department of Medicine and the Neuroscience and Mental Health Institute, University of Alberta, Canada
| | - D W Zochodne
- Peripheral Nerve Research Laboratory, Division of Neurology, Department of Medicine and the Neuroscience and Mental Health Institute, University of Alberta, Canada.
| |
Collapse
|
13
|
Abstract
Neuropathy is a common complication of long-term diabetes that impairs quality of life by producing pain, sensory loss and limb amputation. The presence of neuropathy in both insulin-deficient (type 1) and insulin resistant (type 2) diabetes along with the slowing of progression of neuropathy by improved glycemic control in type 1 diabetes has caused the majority of preclinical and clinical investigations to focus on hyperglycemia as the initiating pathogenic lesion. Studies in animal models of diabetes have identified multiple plausible mechanisms of glucotoxicity to the nervous system including post-translational modification of proteins by glucose and increased glucose metabolism by aldose reductase, glycolysis and other catabolic pathways. However, it is becoming increasingly apparent that factors not necessarily downstream of hyperglycemia can also contribute to the incidence, progression and severity of neuropathy and neuropathic pain. For example, peripheral nerve contains insulin receptors that transduce the neurotrophic and neurosupportive properties of insulin, independent of systemic glucose regulation, while the detection of neuropathy and neuropathic pain in patients with metabolic syndrome and failure of improved glycemic control to protect against neuropathy in cohorts of type 2 diabetic patients has placed a focus on the pathogenic role of dyslipidemia. This review provides an overview of current understanding of potential initiating lesions for diabetic neuropathy and the multiple downstream mechanisms identified in cell and animal models of diabetes that may contribute to the pathogenesis of diabetic neuropathy and neuropathic pain.
Collapse
|
14
|
Bautista J, Chandrasekhar A, Komirishetty PK, Duraikannu A, Zochodne DW. Regenerative plasticity of intact human skin axons. J Neurol Sci 2020; 417:117058. [PMID: 32755738 DOI: 10.1016/j.jns.2020.117058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/01/2020] [Accepted: 07/22/2020] [Indexed: 01/29/2023]
Abstract
The evaluation of human epidermal innervation and its impact by disease has largely focused on rigorous immunohistochemical counts of PGP 9.5 labelled axons. In this brief and preliminary report, we expand the repertoire of epidermal axon markers to include those with an influence on their regenerative plasticity. We studied human lower limb punch skin samples with tandem analyses of their mRNA content using qRT-PCR. Normal human subjects (n = 11) and two patients with newly diagnosed CIDP were sampled with the latter undergoing serial tandem biopsies before and after 3 months of immunotherapy. Controls expressed regeneration proteins within dermal and epidermal axons: GAP43 (growth associated protein 43), Shh (sonic hedgehog) and SCG (superior cervical ganglion-10; stathmin 2). Moreover, this expression accompanied intraepidermal nerve fiber density (IENF) within normal established values. CIDP patients had lower IENF but also expressed GAP43, Shh, and SCG. Tandem qRT-PCR identified confirmed the presence not only of these plasticity markers but of additional regeneration related mRNAs. CIDP patients had marked elevation of several mRNAs, with improvement following treatment. The findings support the concept of dynamic skin axon plasticity in humans is relevant toward consideration of newer therapeutic approaches.
Collapse
Affiliation(s)
- J Bautista
- Division of Neurology, Department of Medicine, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - A Chandrasekhar
- Division of Neurology, Department of Medicine, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - P K Komirishetty
- Division of Neurology, Department of Medicine, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - A Duraikannu
- Division of Neurology, Department of Medicine, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - D W Zochodne
- Division of Neurology, Department of Medicine, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
15
|
Helman A, Cangelosi AL, Davis JC, Pham Q, Rothman A, Faust AL, Straubhaar JR, Sabatini DM, Melton DA. A Nutrient-Sensing Transition at Birth Triggers Glucose-Responsive Insulin Secretion. Cell Metab 2020; 31:1004-1016.e5. [PMID: 32375022 PMCID: PMC7480404 DOI: 10.1016/j.cmet.2020.04.004] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 01/14/2020] [Accepted: 03/31/2020] [Indexed: 12/31/2022]
Abstract
A drastic transition at birth, from constant maternal nutrient supply in utero to intermittent postnatal feeding, requires changes in the metabolic system of the neonate. Despite their central role in metabolic homeostasis, little is known about how pancreatic β cells adjust to the new nutritional challenge. Here, we find that after birth β cell function shifts from amino acid- to glucose-stimulated insulin secretion in correlation with the change in the nutritional environment. This adaptation is mediated by a transition in nutrient sensitivity of the mTORC1 pathway, which leads to intermittent mTORC1 activity. Disrupting nutrient sensitivity of mTORC1 in mature β cells reverts insulin secretion to a functionally immature state. Finally, manipulating nutrient sensitivity of mTORC1 in stem cell-derived β cells in vitro strongly enhances their glucose-responsive insulin secretion. These results reveal a mechanism by which nutrients regulate β cell function, thereby enabling a metabolic adaptation for the newborn.
Collapse
Affiliation(s)
- Aharon Helman
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA.
| | - Andrew L Cangelosi
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jeffrey C Davis
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Quan Pham
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Arielle Rothman
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Aubrey L Faust
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Juerg R Straubhaar
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - David M Sabatini
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Douglas A Melton
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Howard Hughes Medical Institute, Cambridge, MA 02139, USA.
| |
Collapse
|
16
|
Abstract
PURPOSE OF REVIEW The current review addresses one of the most common neurological disorders, diabetic polyneuropathy (DPN). DPN is debilitating, irreversible and dwarfs the prevalence of most other chronic disorders of the nervous system. Its complications include foot ulceration, amputation, falling and intractable neuropathic pain. Moreover, tight control of hyperglycemia reduces the incidence of DPN in type 1 diabetes mellitus but its role in type 2 diabetes mellitus is less clear. RECENT FINDINGS New therapeutic options to reverse the development of DPN or its associated pain have been proposed but none have significantly changed the clinical approach. The cause of DPN remains controversial traditionally focused on the impact of metabolic abnormalities, polyol flux, microvascular changes, mitochondria, oxidative stress, lipid biology and others. In particular, there has been less attention toward how this chronic disorder alters peripheral neurobiology. It is now clear that in chronic models of diabetes mellitus there exists a unique form of neurodegeneration with a range of protein, mRNA and microRNA alterations to consider. How to reconcile these molecular and structural alterations with metabolic mechanisms is a challenge. In sensory neurons alone, a primary target of DPN, both central perikaryal cytoplasmic and nuclear changes and altered distal sensory axon terminal plasticity may be involved. SUMMARY In this review, the current therapeutic status of DPN is described with greater emphasis on some new but selected thoughts on its neurobiology. New mechanistic understanding will be essential to developing precision therapeutics for DPN.
Collapse
|
17
|
Lázár BA, Jancsó G, Sántha P. Modulation of Sensory Nerve Function by Insulin: Possible Relevance to Pain, Inflammation and Axon Growth. Int J Mol Sci 2020; 21:E2507. [PMID: 32260335 PMCID: PMC7177741 DOI: 10.3390/ijms21072507] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 03/31/2020] [Accepted: 04/02/2020] [Indexed: 12/16/2022] Open
Abstract
Insulin, besides its pivotal role in energy metabolism, may also modulate neuronal processes through acting on insulin receptors (InsRs) expressed by neurons of both the central and the peripheral nervous system. Recently, the distribution and functional significance of InsRs localized on a subset of multifunctional primary sensory neurons (PSNs) have been revealed. Systematic investigations into the cellular electrophysiology, neurochemistry and morphological traits of InsR-expressing PSNs indicated complex functional interactions among specific ion channels, proteins and neuropeptides localized in these neurons. Quantitative immunohistochemical studies have revealed disparate localization of the InsRs in somatic and visceral PSNs with a dominance of InsR-positive neurons innervating visceral organs. These findings suggested that visceral spinal PSNs involved in nociceptive and inflammatory processes are more prone to the modulatory effects of insulin than somatic PSNs. Co-localization of the InsR and transient receptor potential vanilloid 1 (TRPV1) receptor with vasoactive neuropeptides calcitonin gene-related peptide and substance P bears of crucial importance in the pathogenesis of inflammatory pathologies affecting visceral organs, such as the pancreas and the urinary bladder. Recent studies have also revealed significant novel aspects of the neurotrophic propensities of insulin with respect to axonal growth, development and regeneration.
Collapse
Affiliation(s)
| | - Gábor Jancsó
- Department of Physiology, University of Szeged, H-6720 Szeged, Hungary; (G.J.); (P.S.)
| | - Péter Sántha
- Department of Physiology, University of Szeged, H-6720 Szeged, Hungary; (G.J.); (P.S.)
| |
Collapse
|
18
|
Sanjari Moghaddam H, Ghazi Sherbaf F, Aarabi MH. Brain microstructural abnormalities in type 2 diabetes mellitus: A systematic review of diffusion tensor imaging studies. Front Neuroendocrinol 2019; 55:100782. [PMID: 31401292 DOI: 10.1016/j.yfrne.2019.100782] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 07/27/2019] [Accepted: 08/07/2019] [Indexed: 12/13/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is associated with deficits in the structure and function of the brain. Diffusion tensor imaging (DTI) is a highly sensitive method for characterizing cerebral tissue microstructure. Using PRISMA guidelines, we identified 29 studies which have demonstrated widespread brain microstructural impairment and topological network disorganization in patients with T2DM. Most consistently reported structures with microstructural abnormalities were frontal, temporal, and parietal lobes in the lobar cluster; corpus callosum, cingulum, uncinate fasciculus, corona radiata, and internal and external capsules in the white matter cluster; thalamus in the subcortical cluster; and cerebellum. Microstructural abnormalities were correlated with pathological derangements in the endocrine profile as well as deficits in cognitive performance in the domains of memory, information-processing speed, executive function, and attention. Altogether, the findings suggest that the detrimental effects of T2DM on cognitive functions might be due to microstructural disruptions in the central neural structures.
Collapse
Affiliation(s)
| | - Farzaneh Ghazi Sherbaf
- Neuroradiology Division, Tehran University of Medical Sciences, School of Medicine, Tehran, Iran
| | - Mohammad Hadi Aarabi
- Neuroradiology Division, Tehran University of Medical Sciences, School of Medicine, Tehran, Iran.
| |
Collapse
|
19
|
Kallinikou D, Soldatou A, Tsentidis C, Louraki M, Kanaka-Gantenbein C, Kanavakis E, Karavanaki K. Diabetic neuropathy in children and adolescents with type 1 diabetes mellitus: Diagnosis, pathogenesis, and associated genetic markers. Diabetes Metab Res Rev 2019; 35:e3178. [PMID: 31083769 DOI: 10.1002/dmrr.3178] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 04/10/2019] [Accepted: 05/07/2019] [Indexed: 01/09/2023]
Abstract
Diabetic neuropathy (DN) is a common long-term complication of type 1 (T1D) and type 2 (T2D) diabetes mellitus, with significant morbidity and mortality. DN is defined as impaired function of the autonomic and/or peripheral nervous system, often subclinical, particularly in children and adolescents with T1D. Nerve conduction studies (NCS) and skin biopsies are considered gold-standard methods in the assessment of DN. Multiple environmental and genetic factors are involved in the pathogenesis of DN. Specifically, the role of metabolic control and glycemic variability is of paramount importance. A number of recently identified genes, including the AKR1B1, VEGF, MTHFR, APOE, and ACE genes, contribute significantly in the pathogenesis of DN. These genes may serve as biomarkers to predict future DN development or treatment response. In addition, they may serve as the basis for the development of new medications or gene therapy. In this review, the diagnostic evaluation, pathogenesis, and associated genetic markers of DN in children and adolescents with T1D are presented and discussed.
Collapse
Affiliation(s)
- Dimitra Kallinikou
- Diabetes and Metabolism Clinic, 2nd Department of Pediatrics, National and Kapodistrian University of Athens-Faculty of Medicine, "P.&A. Kyriakou" Children's Hospital, Athens, Greece
| | - Alexandra Soldatou
- Diabetes and Metabolism Clinic, 2nd Department of Pediatrics, National and Kapodistrian University of Athens-Faculty of Medicine, "P.&A. Kyriakou" Children's Hospital, Athens, Greece
| | - Charalambos Tsentidis
- Diabetes and Metabolism Clinic, 2nd Department of Pediatrics, National and Kapodistrian University of Athens-Faculty of Medicine, "P.&A. Kyriakou" Children's Hospital, Athens, Greece
| | - Maria Louraki
- Diabetes and Metabolism Clinic, 2nd Department of Pediatrics, National and Kapodistrian University of Athens-Faculty of Medicine, "P.&A. Kyriakou" Children's Hospital, Athens, Greece
| | - Christina Kanaka-Gantenbein
- Diabetes Center, Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens-Faculty of Medicine, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Emmanouil Kanavakis
- Diabetes and Metabolism Clinic, 2nd Department of Pediatrics, National and Kapodistrian University of Athens-Faculty of Medicine, "P.&A. Kyriakou" Children's Hospital, Athens, Greece
- Department of Medical Genetics, Choremeio Research Laboratory, National and Kapodistrian University of Athens, Athens, Greece
| | - Kyriaki Karavanaki
- Diabetes and Metabolism Clinic, 2nd Department of Pediatrics, National and Kapodistrian University of Athens-Faculty of Medicine, "P.&A. Kyriakou" Children's Hospital, Athens, Greece
| |
Collapse
|
20
|
Hamer JA, Testani D, Mansur RB, Lee Y, Subramaniapillai M, McIntyre RS. Brain insulin resistance: A treatment target for cognitive impairment and anhedonia in depression. Exp Neurol 2019; 315:1-8. [DOI: 10.1016/j.expneurol.2019.01.016] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 01/22/2019] [Accepted: 01/24/2019] [Indexed: 12/12/2022]
|
21
|
Duraikannu A, Krishnan A, Chandrasekhar A, Zochodne DW. Beyond Trophic Factors: Exploiting the Intrinsic Regenerative Properties of Adult Neurons. Front Cell Neurosci 2019; 13:128. [PMID: 31024258 PMCID: PMC6460947 DOI: 10.3389/fncel.2019.00128] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/14/2019] [Indexed: 01/19/2023] Open
Abstract
Injuries and diseases of the peripheral nervous system (PNS) are common but frequently irreversible. It is often but mistakenly assumed that peripheral neuron regeneration is robust without a need to be improved or supported. However, axonal lesions, especially those involving proximal nerves rarely recover fully and injuries generally are complicated by slow and incomplete regeneration. Strategies to enhance the intrinsic growth properties of reluctant adult neurons offer an alternative approach to consider during regeneration. Since axons rarely regrow without an intimately partnered Schwann cell (SC), approaches to enhance SC plasticity carry along benefits to their axon partners. Direct targeting of molecules that inhibit growth cone plasticity can inform important regenerative strategies. A newer approach, a focus of our laboratory, exploits tumor suppressor molecules that normally dampen unconstrained growth. However several are also prominently expressed in stable adult neurons. During regeneration their ongoing expression “brakes” growth, whereas their inhibition and knockdown may enhance regrowth. Examples have included phosphatase and tensin homolog deleted on chromosome ten (PTEN), a tumor suppressor that inhibits PI3K/pAkt signaling, Rb1, the protein involved in retinoblastoma development, and adenomatous polyposis coli (APC), a tumor suppressor that inhibits β-Catenin transcriptional signaling and its translocation to the nucleus. The identification of several new targets to manipulate the plasticity of regenerating adult peripheral neurons is exciting. How they fit with canonical regeneration strategies and their feasibility require additional work. Newer forms of nonviral siRNA delivery may be approaches for molecular manipulation to improve regeneration.
Collapse
Affiliation(s)
- Arul Duraikannu
- Division of Neurology, Department of Medicine, and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Anand Krishnan
- Division of Neurology, Department of Medicine, and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Ambika Chandrasekhar
- Division of Neurology, Department of Medicine, and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Douglas W Zochodne
- Division of Neurology, Department of Medicine, and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
22
|
Rhea EM, Salameh TS, Banks WA. Routes for the delivery of insulin to the central nervous system: A comparative review. Exp Neurol 2018; 313:10-15. [PMID: 30500332 DOI: 10.1016/j.expneurol.2018.11.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 11/18/2018] [Accepted: 11/24/2018] [Indexed: 12/15/2022]
Abstract
Central nervous system (CNS) insulin resistance is a condition in which the cells within the CNS do not respond to insulin appropriately and is often linked to aberrant CNS insulin levels. CNS insulin is primarily derived from the periphery. Aberrant CNS insulin levels can arise due to various factors including i) decreased endogenous insulin transport into the brain, across the blood-brain barrier (BBB), ii) reduced CNS sequestration of insulin, and iii) increased CNS degradation. While the sole route of endogenous insulin transport into the brain is via the BBB, there are multiple therapeutic routes of administration that have been investigated to deliver exogenous insulin to the CNS. These alternative administrative routes can be utilized to increase the amount of CNS insulin and aid in overcoming CNS insulin resistance. This review focuses on the intravenous, intracerebroventricular, intranasal, ocular, and intrathecal routes of administration and compares the impact of insulin delivery.
Collapse
Affiliation(s)
- Elizabeth M Rhea
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA; Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Therese S Salameh
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA; Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
| | - William A Banks
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA; Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
23
|
Kobayashi M, Zochodne DW. Diabetic neuropathy and the sensory neuron: New aspects of pathogenesis and their treatment implications. J Diabetes Investig 2018; 9:1239-1254. [PMID: 29533535 PMCID: PMC6215951 DOI: 10.1111/jdi.12833] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 02/20/2018] [Accepted: 03/03/2018] [Indexed: 12/17/2022] Open
Abstract
Diabetic polyneuropathy (DPN) continues to be generally considered as a "microvascular" complication of diabetes mellitus alongside nephropathy and retinopathy. The microvascular hypothesis, however, might be tempered by the concept that diabetes directly targets dorsal root ganglion sensory neurons. This neuron-specific concept, supported by accumulating evidence, might account for important features of DPN, such as its early sensory neuron degeneration. Diabetic sensory neurons develop neuronal atrophy alongside a series of messenger ribonucleic acid (RNA) changes related to declines in structural proteins, increases in heat shock protein, increases in the receptor for advanced glycation end-products, declines in growth factor signaling and other changes. Insulin is recognized as a potent neurotrophic factor, and insulin ligation enhances neurite outgrowth through activation of the phosphoinositide 3-kinase-protein kinase B pathway within sensory neurons and attenuates phenotypic features of experimental DPN. Several interventions, including glucagon-like peptide-1 agonism, and phosphatase and tensin homolog inhibition to activate growth signals in sensory neurons, or heat shock protein overexpression, prevent or reverse neuropathic abnormalities in experimental DPN. Diabetic sensory neurons show a unique pattern of microRNA alterations, a key element of messenger RNA silencing. For example, let-7i is widely expressed in sensory neurons, supports their growth and is depleted in experimental DPN; its replenishment improves features of DPN models. Finally, impairment of pre-messenger RNA splicing in diabetic sensory neurons including abnormal nuclear RNA metabolism and structure with loss of survival motor neuron protein, a neuron survival molecule, and overexpression of CWC22, a splicing factor, offer further novel insights. The present review addresses these new aspects of DPN sensory neurodegeneration.
Collapse
Affiliation(s)
- Masaki Kobayashi
- Department of Neurology and Neurological ScienceGraduate School of MedicineTokyo Medical and Dental UniversityTokyoJapan
- Department of NeurologyYokufukai Geriatric HospitalTokyoJapan
| | - Douglas W Zochodne
- Division of Neurology and Department of MedicineNeuroscience and Mental Health InstituteFaculty of Medicine and DentistryUniversity of AlbertaEdmontonAlbertaCanada
| |
Collapse
|
24
|
Lázár BA, Jancsó G, Pálvölgyi L, Dobos I, Nagy I, Sántha P. Insulin Confers Differing Effects on Neurite Outgrowth in Separate Populations of Cultured Dorsal Root Ganglion Neurons: The Role of the Insulin Receptor. Front Neurosci 2018; 12:732. [PMID: 30364236 PMCID: PMC6191510 DOI: 10.3389/fnins.2018.00732] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 09/24/2018] [Indexed: 01/04/2023] Open
Abstract
Apart from its pivotal role in the regulation of carbohydrate metabolism, insulin exerts important neurotrophic and neuromodulator effects on dorsal root ganglion (DRG) neurons. The neurite outgrowth-promoting effect is one of the salient features of insulin's action on cultured DRG neurons. Although it has been established that a significant population of DRG neurons express the insulin receptor (InsR), the significance of InsR expression and the chemical phenotype of DRG neurons in relation to the neurite outgrowth-promoting effect of insulin has not been studied. Therefore, in this study by using immunohistochemical and quantitative stereological methods we evaluated the effect of insulin on neurite outgrowth of DRG neurons of different chemical phenotypes which express or lack the InsR. Insulin, at a concentration of 10 nM, significantly increased total neurite length, the length of the longest neurite and the number of branch points of cultured DRG neurons as compared to neurons cultured in control medium or in the presence of 1 μM insulin. In both the control and the insulin exposed cultures, ∼43% of neurons displayed InsR-immunoreactivity. The proportions of transient receptor potential vanilloid type 1 receptor (TRPV1)-immunoreactive (IR), calcitonin gene-related peptide (CGRP)-IR and Bandeiraea simplicifolia isolectin B4 (IB4)-binding neurons amounted to ∼61%, ∼57%, and ∼31% of DRG neurons IR for the InsR. Of the IB4-positive population only neurons expressing the InsR were responsive to insulin. In contrast, TRPV1-IR nociceptive and CGRP-IR peptidergic neurons showed increased tendency for neurite outgrowth which was further enhanced by insulin. However, the responsiveness of DRG neurons expressing the InsR was superior to populations of DRG neurons which lack this receptor. The findings also revealed that besides the expression of the InsR, inherent properties of peptidergic, but not non-peptidergic nociceptive neurons may also significantly contribute to the mechanisms of neurite outgrowth of DRG neurons. These observations suggest distinct regenerative propensity for differing populations of DRG neurons which is significantly affected through insulin receptor signaling.
Collapse
Affiliation(s)
- Bence András Lázár
- Department of Psychiatry, Faculty of Medicine, University of Szeged, Szeged, Hungary.,Department of Physiology, University of Szeged, Szeged, Hungary
| | - Gábor Jancsó
- Department of Physiology, University of Szeged, Szeged, Hungary
| | - Laura Pálvölgyi
- Department of Physiology, University of Szeged, Szeged, Hungary
| | - Ildikó Dobos
- Department of Physiology, University of Szeged, Szeged, Hungary
| | - István Nagy
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Péter Sántha
- Department of Physiology, University of Szeged, Szeged, Hungary
| |
Collapse
|
25
|
Nieuwenhuis B, Haenzi B, Andrews MR, Verhaagen J, Fawcett JW. Integrins promote axonal regeneration after injury of the nervous system. Biol Rev Camb Philos Soc 2018; 93:1339-1362. [PMID: 29446228 PMCID: PMC6055631 DOI: 10.1111/brv.12398] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 12/23/2017] [Accepted: 01/11/2018] [Indexed: 12/13/2022]
Abstract
Integrins are cell surface receptors that form the link between extracellular matrix molecules of the cell environment and internal cell signalling and the cytoskeleton. They are involved in several processes, e.g. adhesion and migration during development and repair. This review focuses on the role of integrins in axonal regeneration. Integrins participate in spontaneous axonal regeneration in the peripheral nervous system through binding to various ligands that either inhibit or enhance their activation and signalling. Integrin biology is more complex in the central nervous system. Integrins receptors are transported into growing axons during development, but selective polarised transport of integrins limits the regenerative response in adult neurons. Manipulation of integrins and related molecules to control their activation state and localisation within axons is a promising route towards stimulating effective regeneration in the central nervous system.
Collapse
Affiliation(s)
- Bart Nieuwenhuis
- John van Geest Centre for Brain Repair, Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0PYU.K.
- Laboratory for Regeneration of Sensorimotor SystemsNetherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW)1105 BAAmsterdamThe Netherlands
| | - Barbara Haenzi
- John van Geest Centre for Brain Repair, Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0PYU.K.
| | | | - Joost Verhaagen
- Laboratory for Regeneration of Sensorimotor SystemsNetherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW)1105 BAAmsterdamThe Netherlands
- Centre for Neurogenomics and Cognitive Research, Amsterdam NeuroscienceVrije Universiteit Amsterdam1081 HVAmsterdamThe Netherlands
| | - James W. Fawcett
- John van Geest Centre for Brain Repair, Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0PYU.K.
- Centre of Reconstructive NeuroscienceInstitute of Experimental Medicine142 20Prague 4Czech Republic
| |
Collapse
|
26
|
Etchegoyen M, Nobile MH, Baez F, Posesorski B, González J, Lago N, Milei J, Otero-Losada M. Metabolic Syndrome and Neuroprotection. Front Neurosci 2018; 12:196. [PMID: 29731703 PMCID: PMC5919958 DOI: 10.3389/fnins.2018.00196] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 03/12/2018] [Indexed: 12/17/2022] Open
Abstract
Introduction: Over the years the prevalence of metabolic syndrome (MetS) has drastically increased in developing countries as a major byproduct of industrialization. Many factors, such as the consumption of high-calorie diets and a sedentary lifestyle, bolster the spread of this disorder. Undoubtedly, the massive and still increasing incidence of MetS places this epidemic as an important public health issue. Hereon we revisit another outlook of MetS beyond its classical association with cardiovascular disease (CVD) and Diabetes Mellitus Type 2 (DM2), for MetS also poses a risk factor for the nervous tissue and threatens neuronal function. First, we revise a few essential concepts of MetS pathophysiology. Second, we explore some neuroprotective approaches in MetS pertaining brain hypoxia. The articles chosen for this review range from the years 1989 until 2017; the selection criteria was based on those providing data and exploratory information on MetS as well as those that studied innovative therapeutic approaches. Pathophysiology: The characteristically impaired metabolic pathways of MetS lead to hyperglycemia, insulin resistance (IR), inflammation, and hypoxia, all closely associated with an overall pro-oxidative status. Oxidative stress is well-known to cause the wreckage of cellular structures and tissue architecture. Alteration of the redox homeostasis and oxidative stress alter the macromolecular array of DNA, lipids, and proteins, in turn disrupting the biochemical pathways necessary for normal cell function. Neuroprotection: Different neuroprotective strategies are discussed involving lifestyle changes, medication aimed to mitigate MetS cardinal symptoms, and treatments targeted toward reducing oxidative stress. It is well-known that the routine practice of physical exercise, aerobic activity in particular, and a complete and well-balanced nutrition are key factors to prevent MetS. Nevertheless, pharmacological control of MetS as a whole and pertaining hypertension, dyslipidemia, and endothelial injury contribute to neuronal health improvement. Conclusion: The development of MetS has risen as a risk factor for neurological disorders. The therapeutic strategies include multidisciplinary approaches directed to address different pathological pathways all in concert.
Collapse
Affiliation(s)
- Melisa Etchegoyen
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Mariana H Nobile
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Francisco Baez
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Barbara Posesorski
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Julian González
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Néstor Lago
- Institute of Cardiovascular Pathophysiology, School of Medicine, University of Buenos Aires, UBA-CONICET, Buenos Aires, Argentina
| | - José Milei
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Matilde Otero-Losada
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
27
|
Nedelcovych MT, Gadiano AJ, Wu Y, Manning AA, Thomas AG, Khuder SS, Yoo SW, Xu J, McArthur JC, Haughey NJ, Volsky DJ, Rais R, Slusher BS. Pharmacokinetics of Intranasal versus Subcutaneous Insulin in the Mouse. ACS Chem Neurosci 2018; 9:809-816. [PMID: 29257872 PMCID: PMC5906198 DOI: 10.1021/acschemneuro.7b00434] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Insulin delivery to the brain has emerged as an important therapeutic target for cognitive disorders associated with abnormal brain energy metabolism. Although insulin is transported across the blood-brain barrier, peripheral routes of administration are problematic due to systemic effects of insulin on blood glucose. Intranasal (IN) administration is being investigated as an alternative route. We conducted a head-to-head comparison of subcutaneous (SC) and IN insulin, assessing plasma and brain pharmacokinetics and blood glucose levels in the mouse. SC insulin (2.4 IU) achieved therapeutically relevant concentrations in the brain (AUCbrain = 2537 h·μIU/mL) but dramatically increased plasma insulin (AUCplasma = 520 351 h·*μIU/mL), resulting in severe hypoglycemia and in some cases death. IN administration of the same dose resulted in similar insulin levels in the brain (AUCbrain = 3442 h·μIU/mL) but substantially lower plasma concentrations (AUCplasma = 354 h·μIU/mL), amounting to a ∼ 2000-fold increase in the AUCbrain:plasma ratio relative to SC. IN dosing also had no significant effect on blood glucose. When administered daily for 9 days, IN insulin increased brain glucose and energy metabolite concentrations (e.g., adenosine triphosphate and phosphocreatine) without causing overt toxicity, suggesting that IN insulin may be a safe therapeutic option for cognitively impaired patients.
Collapse
Affiliation(s)
- Michael T. Nedelcovych
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Alexandra J. Gadiano
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Ying Wu
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Arena A. Manning
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Ajit G. Thomas
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Saja S. Khuder
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Department of Neuroimmunology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Seung-Wan Yoo
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Jiadi Xu
- Department of Radiology and Radiological Science, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Justin C. McArthur
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Norman J. Haughey
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - David J. Volsky
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Rana Rais
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Barbara S. Slusher
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| |
Collapse
|
28
|
Kobayashi M, Chandrasekhar A, Cheng C, Martinez JA, Ng H, de la Hoz C, Zochodne DW. Diabetic polyneuropathy, sensory neurons, nuclear structure and spliceosome alterations: a role for CWC22. Dis Model Mech 2017; 10:215-224. [PMID: 28250049 PMCID: PMC5374325 DOI: 10.1242/dmm.028225] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 12/23/2016] [Indexed: 12/31/2022] Open
Abstract
Unique deficits in the function of adult sensory neurons as part of their early neurodegeneration might account for progressive polyneuropathy during chronic diabetes mellitus. Here, we provide structural and functional evidence for aberrant pre-mRNA splicing in a chronic type 1 model of experimental diabetic polyneuropathy (DPN). Cajal bodies (CBs), unique nuclear substructures involved in RNA splicing, increased in number in diabetic sensory neurons, but their expected colocalization with survival motor neuron (SMN) proteins was reduced - a mislocalization described in motor neurons of spinal muscular atrophy. Small nuclear ribonucleoprotein particles (snRNPs), also participants in the spliceosome, had abnormal multiple nuclear foci unassociated with CBs, and their associated snRNAs were reduced. CWC22, a key spliceosome protein, was aberrantly upregulated in diabetic dorsal root ganglia (DRG), and impaired neuronal function. CWC22 attenuated sensory neuron plasticity, with knockdown in vitro enhancing their neurite outgrowth. Further, axonal delivery of CWC22 siRNA unilaterally to locally knock down the aberrant protein in diabetic nerves improved aspects of sensory function in diabetic mice. Collectively, our findings identify subtle but significant alterations in spliceosome structure and function, including dysregulated CBs and CWC22 overexpression, in diabetic sensory neurons that offer new ideas regarding diabetic sensory neurodegeneration in polyneuropathy.
Collapse
Affiliation(s)
- Masaki Kobayashi
- Division of Neurology and Department of Medicine, Faculty of Medicine and Dentistry, and the Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada, T6G 2G3
| | - Ambika Chandrasekhar
- Division of Neurology and Department of Medicine, Faculty of Medicine and Dentistry, and the Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada, T6G 2G3
| | - Chu Cheng
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, Faculty of Medicine, University of Calgary, Canada, T2N 4N1
| | - Jose A Martinez
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, Faculty of Medicine, University of Calgary, Canada, T2N 4N1
| | - Hilarie Ng
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, Faculty of Medicine, University of Calgary, Canada, T2N 4N1
| | - Cristiane de la Hoz
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, Faculty of Medicine, University of Calgary, Canada, T2N 4N1
| | - Douglas W Zochodne
- Division of Neurology and Department of Medicine, Faculty of Medicine and Dentistry, and the Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada, T6G 2G3 .,Hotchkiss Brain Institute and Department of Clinical Neurosciences, Faculty of Medicine, University of Calgary, Canada, T2N 4N1
| |
Collapse
|
29
|
Bendella H, Rink S, Grosheva M, Sarikcioglu L, Gordon T, Angelov DN. Putative roles of soluble trophic factors in facial nerve regeneration, target reinnervation, and recovery of vibrissal whisking. Exp Neurol 2017; 300:100-110. [PMID: 29104116 DOI: 10.1016/j.expneurol.2017.10.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 10/25/2017] [Accepted: 10/30/2017] [Indexed: 12/15/2022]
Abstract
It is well-known that, after nerve transection and surgical repair, misdirected regrowth of regenerating motor axons may occur in three ways. The first way is that the axons enter into endoneurial tubes that they did not previously occupy, regenerate through incorrect fascicles and reinnervate muscles that they did not formerly supply. Consequently the activation of these muscles results in inappropriate movements. The second way is that, in contrast with the precise target-directed pathfinding by elongating motor nerves during embryonic development, several axons rather than a single axon grow out from each transected nerve fiber. The third way of misdirection occurs by the intramuscular terminal branching (sprouting) of each regenerating axon to culminate in some polyinnervation of neuromuscular junctions, i.e. reinnervation of junctions by more than a single axon. Presently, "fascicular" or "topographic specificity" cannot be achieved and hence target-directed nerve regeneration is, as yet, unattainable. Nonetheless, motor and sensory reinnervation of appropriate endoneurial tubes does occur and can be promoted by brief nerve electrical stimulation. This review considers the expression of neurotrophic factors in the neuromuscular system and how this expression can promote functional recovery, with emphasis on the whisking of vibrissae on the rat face in relationship to the expression of the factors. Evidence is reviewed for a role of neurotrophic factors as short-range diffusible sprouting stimuli in promoting complete functional recovery of vibrissal whisking in blind Sprague Dawley (SD)/RCS rats but not in SD rats with normal vision, after facial nerve transection and surgical repair. Briefly, a complicated time course of growth factor expression in the nerves and denervated muscles include (1) an early increase in FGF2 and IGF2, (2) reduced NGF between 2 and 14days after nerve transection and surgical repair, (3) a late rise in BDNF and (4) reduced IGF1 protein in the denervated muscles at 28days. These findings suggest that recovery of motor function after peripheral nerve injury is due, at least in part, to a complex regulation of nerve injury-associated neurotrophic factors and cytokines at the neuromuscular junctions of denervated muscles. In particular, the increase of FGF2 and concomittant decrease of NGF during the first week after facial nerve-nerve anastomosis in SD/RCS blind rats may prevent intramuscular axon sprouting and, in turn, reduce poly-innervation of the neuromuscular junction.
Collapse
Affiliation(s)
- Habib Bendella
- Department of Neurosurgery, University of Witten/Herdecke, Cologne Merheim Medical Center (CMMC), Cologne, Germany
| | - Svenja Rink
- Department of Prosthetic Dentistry, School of Dental and Oral Medicine, University of Cologne, Germany
| | - Maria Grosheva
- Department of Oto-Rhino-Laryngology, University of Cologne, Germany
| | | | - Tessa Gordon
- Department of Surgery, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | | |
Collapse
|
30
|
Increased cutaneous miR-let-7d expression correlates with small nerve fiber pathology in patients with fibromyalgia syndrome. Pain 2017; 157:2493-2503. [PMID: 27429177 DOI: 10.1097/j.pain.0000000000000668] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Fibromyalgia syndrome (FMS) is a chronic widespread pain condition probably comprising subgroups with different underlying pathomechanisms. There is increasing evidence for small nerve fiber impairment in subgroups of patients with FMS. MicroRNAs (miRNAs) regulate molecular factors determining nerve de- and re-generation. We investigated whether systemic and cutaneous miRNA expression in patients with FMS is related to small nerve fiber pathology. We confirmed previous findings of disturbed small fiber function and reduced intraepidermal nerve fiber density in subgroups of patients with FMS. We found 51 aberrantly expressed miRNAs in white blood cells of patients with FMS, of which miR-let-7d correlated with reduced small nerve fiber density in patients with FMS. Furthermore, we demonstrated miR-let-7d and its downstream target insulin-like growth factor-1 receptor as being aberrantly expressed in skin of patients with FMS with small nerve fiber impairment. Our study gives further evidence of small nerve fiber pathology in FMS subgroups and provides a missing link in the pathomechanism that may lead to small fiber loss in subgroups of patients with FMS.
Collapse
|
31
|
Association of Insulin and Cholesterol Levels With Peripheral Nervous System Function in Overweight Adults: A 3-Year Follow-up. J Clin Neurophysiol 2017; 34:492-496. [PMID: 29023304 DOI: 10.1097/wnp.0000000000000425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
PURPOSE The purpose of this prospective 3-year follow-up was to investigate the association of glucose, insulin, and cholesterol levels with peripheral nervous system function in overweight and obese subjects. METHODS Forty nondiabetic overweight and obese adults were enrolled, of whom 29 completed the follow-up. Peripheral nervous system function was measured and defined by conduction studies of the peroneal motor nerve and the radial, sural, and medial plantar sensory nerves. Serum insulin and glucose levels were determined with an oral glucose tolerance test, and cholesterol levels were measured. The measurements were performed at baseline and after 3 years. RESULTS The change in serum insulin level at 120 minutes after an oral glucose tolerance test was positively associated with changes in peroneal nerve conduction velocities and F-wave mean, sural nerve conduction and medial plantar nerve conduction velocities. Action potential amplitudes decreased consistently and significantly in all sensory nerves. CONCLUSIONS The change in serum insulin level at 120 minutes appears to be positively associated with changes in nerve conduction velocities more than 3 years but not with nerve action potential amplitudes. Significant decreases in the action potential amplitudes of all sensory nerves suggest that such changes might be the earliest detectable sign of damage to the peripheral nervous system in overweight and obese people without type 2 diabetes.
Collapse
|
32
|
Hippocampal insulin resistance and altered food decision-making as players on obesity risk. Neurosci Biobehav Rev 2017; 77:165-176. [DOI: 10.1016/j.neubiorev.2017.03.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 03/17/2017] [Accepted: 03/19/2017] [Indexed: 12/17/2022]
|
33
|
Grote CW, Wright DE. A Role for Insulin in Diabetic Neuropathy. Front Neurosci 2016; 10:581. [PMID: 28066166 PMCID: PMC5179551 DOI: 10.3389/fnins.2016.00581] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 12/06/2016] [Indexed: 12/13/2022] Open
Abstract
The peripheral nervous system is one of several organ systems that are profoundly affected in diabetes. The longstanding view is that insulin does not have a major role in modulating neuronal function in both central and peripheral nervous systems is now being challenged. In the setting of insulin deficiency or excess insulin, it is logical to propose that insulin dysregulation can contribute to neuropathic changes in sensory neurons. This is particularly important as sensory nerve damage associated with prediabetes, type 1 and type 2 diabetes is so prevalent. Here, we discuss the current experimental literature related to insulin's role as a potential neurotrophic factor in peripheral nerve function, as well as the possibility that insulin deficiency plays a role in diabetic neuropathy. In addition, we discuss how sensory neurons in the peripheral nervous system respond to insulin similar to other insulin-sensitive tissues. Moreover, studies now suggest that sensory neurons can also become insulin resistant like other tissues. Collectively, emerging studies are revealing that insulin signaling pathways are active contributors to sensory nerve modulation, and this review highlights this novel activity and should provide new insight into insulin's role in both peripheral and central nervous system diseases.
Collapse
Affiliation(s)
- Caleb W Grote
- Department of Anatomy and Cell Biology, University of Kansas Medical Center Kansas City, KS, USA
| | - Douglas E Wright
- Department of Anatomy and Cell Biology, University of Kansas Medical Center Kansas City, KS, USA
| |
Collapse
|
34
|
Jolivalt CG, Frizzi KE, Guernsey L, Marquez A, Ochoa J, Rodriguez M, Calcutt NA. Peripheral Neuropathy in Mouse Models of Diabetes. ACTA ACUST UNITED AC 2016; 6:223-255. [PMID: 27584552 DOI: 10.1002/cpmo.11] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Peripheral neuropathy is a frequent complication of chronic diabetes that most commonly presents as a distal degenerative polyneuropathy with sensory loss. Around 20% to 30% of such patients may also experience neuropathic pain. The underlying pathogenic mechanisms are uncertain, and therapeutic options are limited. Rodent models of diabetes have been used for more than 40 years to study neuropathy and evaluate potential therapies. For much of this period, streptozotocin-diabetic rats were the model of choice. The emergence of new technologies that allow relatively cheap and routine manipulations of the mouse genome has prompted increased use of mouse models of diabetes to study neuropathy. In this article, we describe the commonly used mouse models of type 1 and type 2 diabetes, and provide protocols to phenotype the structural, functional, and behavioral indices of peripheral neuropathy, with a particular emphasis on assays pertinent to the human condition. © 2016 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Corinne G Jolivalt
- Department of Pathology, University of California San Diego, La Jolla, California
| | - Katie E Frizzi
- Department of Pathology, University of California San Diego, La Jolla, California
| | - Lucie Guernsey
- Department of Pathology, University of California San Diego, La Jolla, California
| | - Alex Marquez
- Department of Pathology, University of California San Diego, La Jolla, California
| | - Joseline Ochoa
- Department of Pathology, University of California San Diego, La Jolla, California
| | - Maria Rodriguez
- Department of Pathology, University of California San Diego, La Jolla, California
| | - Nigel A Calcutt
- Department of Pathology, University of California San Diego, La Jolla, California
| |
Collapse
|
35
|
Abstract
Diabetic polyneuropathy (DPN) is a common but intractable degenerative disorder of peripheral neurons. DPN first results in retraction and loss of sensory terminals in target organs such as the skin, whereas the perikarya (cell bodies) of neurons are relatively preserved. This is important because it implies that regrowth of distal terminals, rather than neuron replacement or rescue, may be useful clinically. Although a number of neuronal molecular abnormalities have been examined in experimental DPN, several are prominent: loss of structural proteins, neuropeptides, and neurotrophic receptors; upregulation of "stress" and "repair" proteins; elevated nitric oxide synthesis; increased AGE-RAGE signaling, NF-κB and PKC; altered neuron survival pathways; changes of pain-related ion channel investment. There is also a role for abnormalities of direct signaling of neurons by insulin, an important trophic factor for neurons that express its receptors. While evidence implicating each of these pathways has emerged, how they link together and result in neuronal degeneration remains unclear. However, several offer interesting new avenues for more definitive therapy of this condition.
Collapse
Affiliation(s)
- Douglas W Zochodne
- Division of Neurology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
36
|
Zochodne DW. Sensory Neurodegeneration in Diabetes: Beyond Glucotoxicity. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2016; 127:151-80. [PMID: 27133149 DOI: 10.1016/bs.irn.2016.03.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Diabetic polyneuropathy in humans is of gradual, sometimes insidious onset, and is more likely to occur if glucose control is poor. Arguments that the disorder arises chiefly from glucose toxicity however ignore the greater complexity of a unique neurodegenerative disorder. For example, sensory neurons regularly thrive in media with levels of glucose at or exceeding those of poorly controlled diabetic persons. Also, all of the linkages between hyperglycemia and neuropathy develop in the setting of altered insulin availability or sensitivity. Insulin itself is recognized as a potent growth, or trophic factor for adult sensory neurons. Low doses of insulin, insufficient to alter blood glucose levels, reverse features of diabetic neurodegeneration in animal models. Insulin resistance, as occurs in diabetic adipose tissue, liver, and muscle, also develops in sensory neurons, offering a mechanism for neurodegeneration in the setting of normal or elevated insulin levels. Other interventions that "shore up" sensory neurons prevent features of diabetic polyneuropathy from developing despite persistent hyperglycemia. More recently evidence has emerged that a series of subtle molecular changes in sensory neurons can be linked to neurodegeneration including epigenetic changes in the control of gene expression. Understanding the new complexity of sensory neuron degeneration may give rise to therapeutic strategies that have a higher chance of success in the clinical trial arena.
Collapse
Affiliation(s)
- D W Zochodne
- Neuroscience and Mental Health Institute and Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
37
|
Grosheva M, Nohroudi K, Schwarz A, Rink S, Bendella H, Sarikcioglu L, Klimaschewski L, Gordon T, Angelov DN. Comparison of trophic factors' expression between paralyzed and recovering muscles after facial nerve injury. A quantitative analysis in time course. Exp Neurol 2016; 279:137-148. [PMID: 26940083 DOI: 10.1016/j.expneurol.2016.02.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 02/07/2016] [Accepted: 02/26/2016] [Indexed: 01/08/2023]
Abstract
After peripheral nerve injury, recovery of motor performance negatively correlates with the poly-innervation of neuromuscular junctions (NMJ) due to excessive sprouting of the terminal Schwann cells. Denervated muscles produce short-range diffusible sprouting stimuli, of which some are neurotrophic factors. Based on recent data that vibrissal whisking is restored perfectly during facial nerve regeneration in blind rats from the Sprague Dawley (SD)/RCS strain, we compared the expression of brain derived neurotrophic factor (BDNF), fibroblast growth factor-2 (FGF2), insulin growth factors 1 and 2 (IGF1, IGF2) and nerve growth factor (NGF) between SD/RCS and SD-rats with normal vision but poor recovery of whisking function after facial nerve injury. To establish which trophic factors might be responsible for proper NMJ-reinnervation, the transected facial nerve was surgically repaired (facial-facial anastomosis, FFA) for subsequent analysis of mRNA and proteins expressed in the levator labii superioris muscle. A complicated time course of expression included (1) a late rise in BDNF protein that followed earlier elevated gene expression, (2) an early increase in FGF2 and IGF2 protein after 2 days with sustained gene expression, (3) reduced IGF1 protein at 28 days coincident with decline of raised mRNA levels to baseline, and (4) reduced NGF protein between 2 and 14 days with maintained gene expression found in blind rats but not the rats with normal vision. These findings suggest that recovery of motor function after peripheral nerve injury is due, at least in part, to a complex regulation of lesion-associated neurotrophic factors and cytokines in denervated muscles. The increase of FGF-2 protein and concomittant decrease of NGF (with no significant changes in BDNF or IGF levels) during the first week following FFA in SD/RCS blind rats possibly prevents the distal branching of regenerating axons resulting in reduced poly-innervation of motor endplates.
Collapse
Affiliation(s)
- Maria Grosheva
- Department of Oto-Rhino-Laryngology, University of Cologne, Germany
| | | | - Alisa Schwarz
- Department of Anatomy I, University of Cologne, Germany
| | - Svenja Rink
- Department of Anatomy I, University of Cologne, Germany
| | - Habib Bendella
- Department of Neurosurgery, Hospital Merheim, University of Witten-Herdecke, Cologne, Germany
| | | | - Lars Klimaschewski
- Division of Neuroanatomy Innsbruck Medical University, 6020 Innsbruck, Austria
| | - Tessa Gordon
- Department of Surgery,The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | | |
Collapse
|
38
|
Yoon BI, Han KD, Lee KW, Kwon HS, Kim SW, Sohn DW, Cho YH, Ha US. Insulin Resistance Is Associated with Prevalence of Physician-Diagnosed Urinary Incontinence in Postmenopausal Non-Diabetic Adult Women: Data from the Fourth Korea National Health and Nutrition Examination Survey. PLoS One 2015; 10:e0141720. [PMID: 26529410 PMCID: PMC4631470 DOI: 10.1371/journal.pone.0141720] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 10/11/2015] [Indexed: 12/04/2022] Open
Abstract
Objective To investigate the association between insulin resistance (IR) and urinary incontinence in Korean adult women by analyzing the data from the Korea National Health and Nutrition Examination Survey IV (KNHANES) 2007–2009 Methods A nationally representative sample of 5318 non-diabetic Korean women ≥19-years-of-age (3043 premenopausal and 2275 postmenopausal women) was included from KNHANES 2008–2010. IR was measured using the homeostasis model assessment of IR (HOMA-IR). Participants in the highest and lowest quartile of HOMA-IR were defined as insulin-resistant and insulin-sensitive respectively. Women who have current physician-diagnosed urinary incontinence were classified as having urinary incontinence. Results Incontinence was found in 9.18% of the total population, 8.51% of the premenopausal population, and 10.86% of the postmenopausal population. The prevalence of incontinence increased with age, reaching a peak at 60-69-years-of-age. The prevalence of urinary incontinence increased significantly with higher HOMA-IR quartiles in pre- and post-menopausal women (p for linear association = 0.0458 and 0.0009 respectively). Among post-menopausal women, those in the highest quartile of HOMA-IR were significantly more likely to have urinary incontinence compared to those in the lowest quartile [adjusted odds ratio, 1.72; 95% confidence interval, 1.07–2.77]. However premenopausal population exhibited no association between incontinence and HOMA-IR quartiles Conclusion Our results suggest that the prevalence of incontinence increased across HOMA-IR in non-diabetic adult women, and especially, IR might be a risk factor for incontinence in postmenopausal non-diabetic women.
Collapse
Affiliation(s)
- Byung Il Yoon
- Department of Urology, The Catholic Kwandong University of Korea, International St Mary’s hospital, Incheon, Korea
| | - Kyung-Do Han
- Department of Biostatistics, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Kyu Won Lee
- Department of Urology, College of Medicine, The Catholic University of Korea, St Paul’s hospital, Seoul, Korea
| | - Hyuk Sang Kwon
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Yeoido St Mary’s hospital, Seoul, Korea
| | - Sun Wook Kim
- Department of Urology, College of Medicine, The Catholic University of Korea, Yeoido St Mary’s hospital, Seoul, Korea
| | - Dong Wan Sohn
- Department of Urology, College of Medicine, The Catholic University of Korea, Yeoido St Mary’s hospital, Seoul, Korea
| | - Yong-Hyun Cho
- Department of Urology, College of Medicine, The Catholic University of Korea, Yeoido St Mary’s hospital, Seoul, Korea
| | - U-Syn Ha
- Department of Urology, College of Medicine, The Catholic University of Korea, Yeoido St Mary’s hospital, Seoul, Korea
- * E-mail:
| |
Collapse
|
39
|
Fukui K, Ferris HA, Kahn CR. Effect of cholesterol reduction on receptor signaling in neurons. J Biol Chem 2015; 290:26383-92. [PMID: 26370080 DOI: 10.1074/jbc.m115.664367] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Indexed: 11/06/2022] Open
Abstract
Diabetes mellitus is associated with a variety of complications, including alterations in the central nervous system (CNS). We have recently shown that diabetes results in a reduction of cholesterol synthesis in the brain due to decreased insulin stimulation of SREBP2-mediated cholesterol synthesis in neuronal and glial cells. In the present study, we explored the effects of the decrease in cholesterol on neuronal cell function using GT1-7 hypothalamic cells subjected to cholesterol depletion in vitro using three independent methods: 1) exposure to methyl-β-cyclodextrin, 2) treatment with the HMG-CoA reductase inhibitor simvastatin, and 3) shRNA-mediated knockdown of SREBP2. All three methods produced 20-31% reductions in cellular cholesterol content, similar to the decrease in cholesterol synthesis observed in diabetes. All cholesterol-depleted neuron-derived cells, independent of the method of reduction, exhibited decreased phosphorylation/activation of IRS-1 and AKT following stimulation by insulin, insulin-like growth factor-1, or the neurotrophins (NGF and BDNF). ERK phosphorylation/activation was also decreased after methyl-β-cyclodextrin and statin treatment but increased in cells following SREBP2 knockdown. In addition, apoptosis in the presence of amyloid-β was increased. Reduction in cellular cholesterol also resulted in increased basal autophagy and impairment of induction of autophagy by glucose deprivation. Together, these data indicate that a reduction in neuron-derived cholesterol content, similar to that observed in diabetic brain, creates a state of insulin and growth factor resistance that could contribute to CNS-related complications of diabetes, including increased risk of neurodegenerative diseases, such as Alzheimer disease.
Collapse
Affiliation(s)
- Kenji Fukui
- From the Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts 02215
| | - Heather A Ferris
- From the Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts 02215
| | - C Ronald Kahn
- From the Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts 02215
| |
Collapse
|
40
|
Evidence for Epigenetic Regulation of Gene Expression and Function in Chronic Experimental Diabetic Neuropathy. J Neuropathol Exp Neurol 2015; 74:804-17. [DOI: 10.1097/nen.0000000000000219] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
41
|
Zochodne DW. Diabetes and the plasticity of sensory neurons. Neurosci Lett 2015; 596:60-5. [DOI: 10.1016/j.neulet.2014.11.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 11/11/2014] [Accepted: 11/13/2014] [Indexed: 12/13/2022]
|
42
|
Han L, Ji L, Chang J, Wen J, Zhao W, Shi H, Zhou L, Li Y, Hu R, Hu J, Lu B. Peripheral neuropathy is associated with insulin resistance independent of metabolic syndrome. Diabetol Metab Syndr 2015; 7:14. [PMID: 25774226 PMCID: PMC4359792 DOI: 10.1186/s13098-015-0010-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 02/13/2015] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND To determine the association of insulin resistance, metabolic syndrome (MetS) with peripheral neuropathy (PN). METHODS This cross-sectional study consisted of 2035 subjects in Shanghai who were classified as with MetS and without MetS. The new International Diabetes Federation (IDF) criterion was used to define MetS. HOMA-IR was applied to evaluate insulin resistance. All subjects underwent complete foot examination. PN was assessed according to the neuropathy symptom and neuropathy disability scores. Binary logistic regression was performed to analyze the contributions of insulin resistance, features of MetS to PN. RESULTS (1) The percentage of PN was 4.0% in our study. Patients with MetS (47.7%) had a higher percentage of PN (5.5% vs. 2.6%, respectively, P = 0.001). With the components of MetS increased (non-MetS, three, four, five), a linear increase in the proportion of peripheral neuropathy was observed (2.6%, 4.8%, 5.6% and 7.2%; respectively, P for trend = 0.001). (2) In patients with PN, the average age of patients was significantly older than the corresponding non-PN patients. Waist circumference, fasting blood glucose, HbA1c, proportion of treatment for diabetes and hypertension were significantly higher in PN group compared with non-PN group in MetS patients. (3) The frequency of dysglycemia was the highest in PN patients both with and without MetS (96.2% and 82.1%, P = 0.084). (4) After adjusting for gender and smoking history, the PN was associated with MetS [odds ratio (OR) 2.0; 95% confidence interval (CI) 1.2, 3.2; P = 0.006], and age (OR 1.1; 95% CI 1.1, 1.1; P < 0.001). When HOMA-IR was added to this binary logistic regression, the association of PN with MetS disappeared (P = 0.110), but the PN was still associated with HOMA-IR (OR 1.2; 95% CI 1.1, 1.4, P < 0.001). CONCLUSIONS In metabolic syndrome, insulin resistance might play an important role in the development of peripheral neuropathy.
Collapse
Affiliation(s)
- Ling Han
- />Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, No, 12 Wulumuqi Middle Road, Jing’an District, Shanghai 200040 China
- />Department of Endocrinology, the Second Affiliated Hospital of Soochow University, Jiangsu, 215004 China
| | - Lijin Ji
- />Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, No, 12 Wulumuqi Middle Road, Jing’an District, Shanghai 200040 China
| | - Jing Chang
- />Department of the Third Internal Medicine, Affiliated hospital of Shandong Academy of Medical Sciences, No. 38 Wuyingshan Road, Shandong, 250031 China
| | - Jian Wen
- />Department of Endocrinology, the Second Affiliated Hospital of Soochow University, Jiangsu, 215004 China
| | - Wenting Zhao
- />Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, No, 12 Wulumuqi Middle Road, Jing’an District, Shanghai 200040 China
| | - Hongli Shi
- />Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, No, 12 Wulumuqi Middle Road, Jing’an District, Shanghai 200040 China
| | - Linuo Zhou
- />Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, No, 12 Wulumuqi Middle Road, Jing’an District, Shanghai 200040 China
| | - Yiming Li
- />Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, No, 12 Wulumuqi Middle Road, Jing’an District, Shanghai 200040 China
| | - Renming Hu
- />Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, No, 12 Wulumuqi Middle Road, Jing’an District, Shanghai 200040 China
| | - Ji Hu
- />Department of Endocrinology, the Second Affiliated Hospital of Soochow University, Jiangsu, 215004 China
| | - Bin Lu
- />Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, No, 12 Wulumuqi Middle Road, Jing’an District, Shanghai 200040 China
| |
Collapse
|
43
|
Chronic Pain Syndromes, Mechanisms, and Current Treatments. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 131:565-611. [DOI: 10.1016/bs.pmbts.2015.01.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
44
|
Nunes AR, Alves MG, Moreira PI, Oliveira PF, Silva BM. Can Tea Consumption be a Safe and Effective Therapy Against Diabetes Mellitus-Induced Neurodegeneration? Curr Neuropharmacol 2014; 12:475-89. [PMID: 25977676 PMCID: PMC4428023 DOI: 10.2174/1570159x13666141204220539] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 10/10/2014] [Accepted: 12/03/2014] [Indexed: 12/20/2022] Open
Abstract
Diabetes mellitus (DM) is a metabolic disease that is rapidly increasing and has become a major public health problem. Type 2 DM (T2DM) is the most common type, accounting for up to 90-95% of the new diagnosed DM cases. The brain is very susceptible to glucose fluctuations and hyperglycemia-induced oxidative stress (OS). It is well known that DM and the risk of developing neurodegenerative diseases are associated. Tea, Camellia sinensis L., is one of the most consumed beverages. It contains several phytochemicals, such as polyphenols, methylxanthines (mainly caffeine) and L-theanine that are often reported to be responsible for tea's health benefits, including in brain. Tea phytochemicals have been reported to be responsible for tea's significant antidiabetic and neuroprotective properties and antioxidant potential. Epidemiological studies have shown that regular consumption of tea has positive effects on DM-caused complications and protects the brain against oxidative damage, contributing to an improvement of the cognitive function. Among the several reported benefits of tea consumption, those related with neurodegenerative diseases are of great interest. Herein, we discuss the potential beneficial effects of tea consumption and tea phytochemicals on DM and how their action can counteract the severe brain damage induced by this disease.
Collapse
Affiliation(s)
- Ana R. Nunes
- CICS – UBI – Health Sciences Research Centre, University of Beira Interior, 6201-506 Covilhã, Portugal
| | - Marco G. Alves
- CICS – UBI – Health Sciences Research Centre, University of Beira Interior, 6201-506 Covilhã, Portugal
| | - Paula I. Moreira
- CNC – Center for Neuroscience and Cell Biology, University of Coimbra and Laboratory of Physiology, Faculty of Medicine, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Pedro F. Oliveira
- CICS – UBI – Health Sciences Research Centre, University of Beira Interior, 6201-506 Covilhã, Portugal
| | - Branca M. Silva
- CICS – UBI – Health Sciences Research Centre, University of Beira Interior, 6201-506 Covilhã, Portugal
| |
Collapse
|
45
|
Chen DK, Frizzi KE, Guernsey LS, Ladt K, Mizisin AP, Calcutt NA. Repeated monitoring of corneal nerves by confocal microscopy as an index of peripheral neuropathy in type-1 diabetic rodents and the effects of topical insulin. J Peripher Nerv Syst 2014; 18:306-15. [PMID: 24147903 DOI: 10.1111/jns5.12044] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 08/07/2013] [Accepted: 10/11/2013] [Indexed: 02/06/2023]
Abstract
We developed a reliable imaging and quantitative analysis method for in vivo corneal confocal microscopy (CCM) in rodents and used it to determine whether models of type 1 diabetes replicate the depletion of corneal nerves reported in diabetic patients. Quantification was reproducible between observers and stable across repeated time points in two rat strains. Longitudinal studies were performed in normal and streptozotocin (STZ)-diabetic rats, with innervation of plantar paw skin quantified using standard histological methods after 40 weeks of diabetes. Diabetic rats showed an initial increase, then a gradual reduction in occupancy of nerves in the sub-basal plexus so that values were significantly lower at week 40 (68 ± 6%) than age-matched controls (80 ± 2%). No significant loss of stromal or intra-epidermal nerves was detected. In a separate study, insulin was applied daily to the eye of control and STZ-diabetic mice and this treatment prevented depletion of nerves of the sub-basal plexus. Longitudinal studies are viable in rodents using CCM and depletion of distal corneal nerves precedes detectable loss of epidermal nerves in the foot, suggesting that diabetic neuropathy is not length dependent. Loss of insulin-derived neurotrophic support may contribute to the pathogenesis of corneal nerve depletion in type 1 diabetes.
Collapse
Affiliation(s)
- Debbie K Chen
- Department of Pathology, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | | | | | | | | | | |
Collapse
|
46
|
Diabetes and the brain: oxidative stress, inflammation, and autophagy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:102158. [PMID: 25215171 PMCID: PMC4158559 DOI: 10.1155/2014/102158] [Citation(s) in RCA: 319] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 07/30/2014] [Accepted: 07/30/2014] [Indexed: 12/19/2022]
Abstract
Diabetes mellitus is a common metabolic disorder associated with chronic complications including a state of mild to moderate cognitive impairment, in particular psychomotor slowing and reduced mental flexibility, not attributable to other causes, and shares many symptoms that are best described as accelerated brain ageing. A common theory for aging and for the pathogenesis of this cerebral dysfunctioning in diabetes relates cell death to oxidative stress in strong association to inflammation, and in fact nuclear factor κB (NFκB), a master regulator of inflammation and also a sensor of oxidative stress, has a strategic position at the crossroad between oxidative stress and inflammation. Moreover, metabolic inflammation is, in turn, related to the induction of various intracellular stresses such as mitochondrial oxidative stress, endoplasmic reticulum (ER) stress, and autophagy defect. In parallel, blockade of autophagy can relate to proinflammatory signaling via oxidative stress pathway and NFκB-mediated inflammation.
Collapse
|
47
|
O'Brien PD, Hinder LM, Sakowski SA, Feldman EL. ER stress in diabetic peripheral neuropathy: A new therapeutic target. Antioxid Redox Signal 2014; 21:621-33. [PMID: 24382087 DOI: 10.1089/ars.2013.5807] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
SIGNIFICANCE Diabetes and other diseases that comprise the metabolic syndrome have reached epidemic proportions. Diabetic peripheral neuropathy (DPN) is the most prevalent complication of diabetes, affecting ~50% of diabetic patients. Characterized by chronic pain or loss of sensation, recurrent foot ulcerations, and risk for amputation, DPN is associated with significant morbidity and mortality. Mechanisms underlying DPN pathogenesis are complex and not well understood, and no effective treatments are available. Thus, an improved understanding of DPN pathogenesis is critical for the development of successful therapeutic options. RECENT ADVANCES Recent research implicates endoplasmic reticulum (ER) stress as a novel mechanism in the onset and progression of DPN. ER stress activates the unfolded protein response (UPR), a well-orchestrated signaling cascade responsible for relieving stress and restoring normal ER function. CRITICAL ISSUES During times of extreme or chronic stress, such as that associated with diabetes, the UPR may be insufficient to alleviate ER stress, resulting in apoptosis. Here, we discuss the potential role of ER stress in DPN, as well as evidence demonstrating how ER stress intersects with pathways involved in DPN development and progression. An improved understanding of how ER stress contributes to peripheral nerve dysfunction in diabetes will provide important insight into DPN pathogenesis. FUTURE DIRECTIONS Future studies aimed at gaining the necessary insight into ER stress in DPN pathogenesis will ultimately facilitate the development of novel therapies.
Collapse
|
48
|
Grote CW, Ryals JM, Wright DE. In vivo peripheral nervous system insulin signaling. J Peripher Nerv Syst 2014; 18:209-19. [PMID: 24028189 DOI: 10.1111/jns5.12033] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 06/07/2013] [Accepted: 07/24/2013] [Indexed: 01/06/2023]
Abstract
Alterations in peripheral nervous system (PNS) insulin support may contribute to diabetic neuropathy (DN); yet, PNS insulin signaling is not fully defined. Here, we investigated in vivo insulin signaling in the PNS and compared the insulin responsiveness to that of muscle, liver, and adipose. Non-diabetic mice were administered increasing doses of insulin to define a dose-response relationship between insulin and Akt activation in the dorsal root ganglion (DRG) and sciatic nerve. Resulting EC50 doses were used to characterize the PNS insulin signaling time course and make comparisons between insulin signaling in the PNS and other peripheral tissues (i.e., muscle, liver, and adipose). The results demonstrate that the PNS is responsive to insulin and that differences in insulin signaling pathway activation exist between PNS compartments. At a therapeutically relevant dose, Akt was activated in the muscle, liver, and adipose at 30 min, correlating with the changes in blood glucose levels. Interestingly, the sciatic nerve showed a similar signaling profile as insulin-sensitive tissues; however, there was not a comparable activation in the DRG or spinal cord. These results present new evidence regarding PNS insulin signaling pathways in vivo and provide a baseline for studies investigating the contribution of disrupted PNS insulin signaling to DN pathogenesis.
Collapse
Affiliation(s)
- Caleb W Grote
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | | | | |
Collapse
|
49
|
Blázquez E, Velázquez E, Hurtado-Carneiro V, Ruiz-Albusac JM. Insulin in the brain: its pathophysiological implications for States related with central insulin resistance, type 2 diabetes and Alzheimer's disease. Front Endocrinol (Lausanne) 2014; 5:161. [PMID: 25346723 PMCID: PMC4191295 DOI: 10.3389/fendo.2014.00161] [Citation(s) in RCA: 329] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 09/21/2014] [Indexed: 12/21/2022] Open
Abstract
Although the brain has been considered an insulin-insensitive organ, recent reports on the location of insulin and its receptors in the brain have introduced new ways of considering this hormone responsible for several functions. The origin of insulin in the brain has been explained from peripheral or central sources, or both. Regardless of whether insulin is of peripheral origin or produced in the brain, this hormone may act through its own receptors present in the brain. The molecular events through which insulin functions in the brain are the same as those operating in the periphery. However, certain insulin actions are different in the central nervous system, such as hormone-induced glucose uptake due to a low insulin-sensitive GLUT-4 activity, and because of the predominant presence of GLUT-1 and GLUT-3. In addition, insulin in the brain contributes to the control of nutrient homeostasis, reproduction, cognition, and memory, as well as to neurotrophic, neuromodulatory, and neuroprotective effects. Alterations of these functional activities may contribute to the manifestation of several clinical entities, such as central insulin resistance, type 2 diabetes mellitus (T2DM), and Alzheimer's disease (AD). A close association between T2DM and AD has been reported, to the extent that AD is twice more frequent in diabetic patients, and some authors have proposed the name "type 3 diabetes" for this association. There are links between AD and T2DM through mitochondrial alterations and oxidative stress, altered energy and glucose metabolism, cholesterol modifications, dysfunctional protein O-GlcNAcylation, formation of amyloid plaques, altered Aβ metabolism, and tau hyperphosphorylation. Advances in the knowledge of preclinical AD and T2DM may be a major stimulus for the development of treatment for preventing the pathogenic events of these disorders, mainly those focused on reducing brain insulin resistance, which is seems to be a common ground for both pathological entities.
Collapse
Affiliation(s)
- Enrique Blázquez
- Departamento de Bioquímica y Biología Molecular III, Facultad de Medicina, Universidad Complutense, Madrid, Spain
- The Center for Biomedical Research in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdiSSC), Madrid, Spain
- *Correspondence: Enrique Blázquez, Departamento de Bioquímica y Biología Molecular III, Facultad de Medicina, Universidad Complutense de Madrid, Plaza Ramón y Cajal s/n, Madrid 28040, Spain e-mail:
| | - Esther Velázquez
- Departamento de Bioquímica y Biología Molecular III, Facultad de Medicina, Universidad Complutense, Madrid, Spain
- The Center for Biomedical Research in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdiSSC), Madrid, Spain
| | - Verónica Hurtado-Carneiro
- Departamento de Bioquímica y Biología Molecular III, Facultad de Medicina, Universidad Complutense, Madrid, Spain
- The Center for Biomedical Research in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdiSSC), Madrid, Spain
| | - Juan Miguel Ruiz-Albusac
- Departamento de Bioquímica y Biología Molecular III, Facultad de Medicina, Universidad Complutense, Madrid, Spain
- The Center for Biomedical Research in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdiSSC), Madrid, Spain
| |
Collapse
|
50
|
Abstract
This review provides an overview of selected aspects of peripheral nerve regeneration and potential avenues to explore therapeutically. The overall coordinated and orchestrated pattern of recovery from peripheral nerve injury has a beauty of execution and progress that rivals all other forms of neurobiology. It involves changes at the level of the perikaryon, coordination with important peripheral glial partners, the Schwann cells, a controlled inflammatory response, and growth that overcomes surprising intrinsic roadblocks. Both regenerative axon growth and collateral sprouting encompass fascinating aspects of this story. Better understanding of peripheral nerve regeneration may also lead to enhanced central nervous system recovery.
Collapse
Affiliation(s)
- Douglas W Zochodne
- Department of Clinical Neurosciences and the Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|