51
|
Abbas M, Alzarea S, Papke RL, Rahman S. Effects of α7 Nicotinic Acetylcholine Receptor Positive Allosteric Modulator on BDNF, NKCC1 and KCC2 Expression in the Hippocampus following Lipopolysaccharide-Induced Allodynia and Hyperalgesia in a Mouse Model of Inflammatory Pain. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2021; 20:366-377. [PMID: 33380307 DOI: 10.2174/1871527319666201230102616] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND & OBJECTIVES Hyperalgesia and allodynia are frequent symptoms of inflammatory pain. Neuronal excitability induced by the Brain-Derived Neurotrophic Factor (BDNF)-tyrosine receptor kinase B (TrkB) cascade has a role in the modulation of inflammatory pain. The effects of 3a,4,5,9b-tetrahydro-4-(1-naphthalenyl)-3H-cyclopentan[c]quinoline-8-sulfonamide (TQS), an α7 nicotinic Acetylcholine Receptor Positive Allosteric Modulator (nAChR PAM), on hippocampal BDNF, cation-chloride cotransporters, NKCC1 and KCC2, expression in inflammatory pain are not known. The objective of the study was to determine the effects of TQS on BDNF, NKCC1, and KCC2 expression in the hippocampus following lipopolysaccharide (LPS)-induced allodynia and hyperalgesia in a mouse model of inflammatory pain. METHODS Mice were treated with TQS followed by LPS (1 mg/kg, ip) administration. The effects of TQS on mRNA and BDNF in the hippocampus were examined using qRT-PCR and Western blot, respectively. Immunoreactivity of BDNF, NKCC1, and KCC2 in the hippocampus was measured after LPS administration using immunofluorescence assay. Allodynia and hyperalgesia were determined using von Frey filaments and hot plate, respectively. RESULTS The LPS (1 mg/kg) upregulates mRNA of BDNF and downregulates mRNA of KCC2 in the hippocampus and pretreatment of TQS (4 mg/kg) reversed the effects induced by LPS. In addition, the TQS decreased LPS-induced upregulation of BDNF and p-NKCC1 immunoreactivity in the dentate gyrus and CA1 region of the hippocampus. BDNF receptor (TrkB) antagonist, ANA12 (0.50 mg/kg), and NKCC1 inhibitor bumetanide (30 mg/kg) reduced LPS-induced allodynia and hyperalgesia. Blockade of TrkB with ANA12 (0.25 mg/kg) enhanced the effects of TQS (1 mg/kg) against LPS-induced allodynia and hyperalgesia. Similarly, bumetanide (10 mg/kg) enhanced the effects of TQS (1 mg/kg) against allodynia and hyperalgesia. CONCLUSION These results suggest that antinociceptive effects of α7 nAChR PAM are associated with downregulation of hippocampal BDNF and p-NKCC1 and upregulation of KCC2 in a mouse model of inflammatory pain.
Collapse
Affiliation(s)
- Muzaffar Abbas
- Department of Pharmaceutical Sciences, College of Pharmacy, South Dakota State University, Brookings SD 57007, United States
| | - Sami Alzarea
- Department of Pharmaceutical Sciences, College of Pharmacy, South Dakota State University, Brookings SD 57007, United States
| | - Roger L Papke
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville FL 32610, United States
| | - Shafiqur Rahman
- Department of Pharmaceutical Sciences, College of Pharmacy, South Dakota State University, Brookings SD 57007, United States
| |
Collapse
|
52
|
Tran EL, Crawford LK. Revisiting PNS Plasticity: How Uninjured Sensory Afferents Promote Neuropathic Pain. Front Cell Neurosci 2020; 14:612982. [PMID: 33362476 PMCID: PMC7759741 DOI: 10.3389/fncel.2020.612982] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 11/12/2020] [Indexed: 11/13/2022] Open
Abstract
Despite the widespread study of how injured nerves contribute to chronic pain, there are still major gaps in our understanding of pain mechanisms. This is particularly true of pain resulting from nerve injury, or neuropathic pain, wherein tactile or thermal stimuli cause painful responses that are particularly difficult to treat with existing therapies. Curiously, this stimulus-driven pain relies upon intact, uninjured sensory neurons that transmit the signals that are ultimately sensed as painful. Studies that interrogate uninjured neurons in search of cell-specific mechanisms have shown that nerve injury alters intact, uninjured neurons resulting in an activity that drives stimulus-evoked pain. This review of neuropathic pain mechanisms summarizes cell-type-specific pathology of uninjured sensory neurons and the sensory ganglia that house their cell bodies. Uninjured neurons have demonstrated a wide range of molecular and neurophysiologic changes, many of which are distinct from those detected in injured neurons. These intriguing findings include expression of pain-associated molecules, neurophysiological changes that underlie increased excitability, and evidence that intercellular signaling within sensory ganglia alters uninjured neurons. In addition to well-supported findings, this review also discusses potential mechanisms that remain poorly understood in the context of nerve injury. This review highlights key questions that will advance our understanding of the plasticity of sensory neuron subpopulations and clarify the role of uninjured neurons in developing anti-pain therapies.
Collapse
Affiliation(s)
- Emily L Tran
- Department of Pathobiological Sciences, University of Wisconsin-Madison School of Veterinary Medicine, Madison, WI, United States
| | - LaTasha K Crawford
- Department of Pathobiological Sciences, University of Wisconsin-Madison School of Veterinary Medicine, Madison, WI, United States
| |
Collapse
|
53
|
Ushakova VM, Morozova AY, Reznik AM, Kostyuk GP, Chekhonin VP. Molecular Biological Aspects of Depressive Disorders: A Modern View. Mol Biol 2020. [DOI: 10.1134/s0026893320050118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
54
|
Hogan MK, Hamilton GF, Horner PJ. Neural Stimulation and Molecular Mechanisms of Plasticity and Regeneration: A Review. Front Cell Neurosci 2020; 14:271. [PMID: 33173465 PMCID: PMC7591397 DOI: 10.3389/fncel.2020.00271] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 07/31/2020] [Indexed: 12/23/2022] Open
Abstract
Neural stimulation modulates the depolarization of neurons, thereby triggering activity-associated mechanisms of neuronal plasticity. Activity-associated mechanisms in turn play a major role in post-mitotic structure and function of adult neurons. Our understanding of the interactions between neuronal behavior, patterns of neural activity, and the surrounding environment is evolving at a rapid pace. Brain derived neurotrophic factor is a critical mediator of activity-associated plasticity, while multiple immediate early genes mediate plasticity of neurons following bouts of neural activity. New research has uncovered genetic mechanisms that govern the expression of DNA following changes in neural activity patterns, including RNAPII pause-release and activity-associated double stranded breaks. Discovery of novel mechanisms governing activity-associated plasticity of neurons hints at a layered and complex molecular control of neuronal response to depolarization. Importantly, patterns of depolarization in neurons are shown to be important mediators of genetic expression patterns and molecular responses. More research is needed to fully uncover the molecular response of different types of neurons-to-activity patterns; however, known responses might be leveraged to facilitate recovery after neural damage. Physical rehabilitation through passive or active exercise modulates neurotrophic factor expression in the brain and spinal cord and can initiate cortical plasticity commensurate with functional recovery. Rehabilitation likely relies on activity-associated mechanisms; however, it may be limited in its application. Electrical and magnetic stimulation direct specific activity patterns not accessible through passive or active exercise and work synergistically to improve standing, walking, and forelimb use after injury. Here, we review emerging concepts in the molecular mechanisms of activity-derived plasticity in order to highlight opportunities that could add value to therapeutic protocols for promoting recovery of function after trauma, disease, or age-related functional decline.
Collapse
Affiliation(s)
- Matthew K Hogan
- Department of Neurosurgery, Center for Neuroregeneration, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, United States
| | - Gillian F Hamilton
- Department of Neurosurgery, Center for Neuroregeneration, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, United States
| | - Philip J Horner
- Department of Neurosurgery, Center for Neuroregeneration, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
55
|
Christiansen L, Chen B, Lei Y, Urbin MA, Richardson MSA, Oudega M, Sandhu M, Rymer WZ, Trumbower RD, Mitchell GS, Perez MA. Acute intermittent hypoxia boosts spinal plasticity in humans with tetraplegia. Exp Neurol 2020; 335:113483. [PMID: 32987000 DOI: 10.1016/j.expneurol.2020.113483] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 08/31/2020] [Accepted: 09/22/2020] [Indexed: 12/19/2022]
Abstract
Paired corticospinal-motoneuronal stimulation (PCMS) elicits spinal synaptic plasticity in humans with chronic incomplete cervical spinal cord injury (SCI). Here, we examined whether PCMS-induced plasticity could be potentiated by acute intermittent hypoxia (AIH), a treatment also known to induce spinal synaptic plasticity in humans with chronic incomplete cervical SCI. During PCMS, we used 180 pairs of stimuli where corticospinal volleys evoked by transcranial magnetic stimulation over the hand representation of the primary motor cortex were timed to arrive at corticospinal-motoneuronal synapses of the first dorsal interosseous (FDI) muscle ~1-2 ms before the arrival of antidromic potentials elicited in motoneurons by electrical stimulation of the ulnar nerve. During AIH, participants were exposed to brief alternating episodes of hypoxic inspired gas (1 min episodes of 9% O2) and room air (1 min episodes of 20.9% O2). We examined corticospinal function by measuring motor evoked potentials (MEPs) elicited by cortical and subcortical stimulation of corticospinal axons and voluntary motor output in the FDI muscle before and after 30 min of PCMS combined with AIH (PCMS+AIH) or sham AIH (PCMS+sham-AIH). The amplitude of MEPs evoked by magnetic and electrical stimulation increased after both protocols, but most after PCMS+AIH, consistent with the hypothesis that their combined effects arise from spinal plasticity. Both protocols increased electromyographic activity in the FDI muscle to a similar extent. Thus, PCMS effects on spinal synapses of hand motoneurons can be potentiated by AIH. The possibility of different thresholds for physiological vs behavioral gains needs to be considered during combinatorial treatments.
Collapse
Affiliation(s)
- Lasse Christiansen
- University of Miami, Department of Neurological Surgery, The Miami Project to Cure Paralysis and Miami VA Medical Center, Miami, FL, 33136, United States of America; Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital, Amager and Hvidovre, Denmark
| | - Bing Chen
- University of Miami, Department of Neurological Surgery, The Miami Project to Cure Paralysis and Miami VA Medical Center, Miami, FL, 33136, United States of America; Shirley Ryan AbilityLab, Northwestern University, Chicago, IL 60611, United States of America
| | - Yuming Lei
- University of Miami, Department of Neurological Surgery, The Miami Project to Cure Paralysis and Miami VA Medical Center, Miami, FL, 33136, United States of America
| | - M A Urbin
- University of Miami, Department of Neurological Surgery, The Miami Project to Cure Paralysis and Miami VA Medical Center, Miami, FL, 33136, United States of America
| | | | - Martin Oudega
- University of Miami, Department of Neurological Surgery, The Miami Project to Cure Paralysis and Miami VA Medical Center, Miami, FL, 33136, United States of America; Shirley Ryan AbilityLab, Northwestern University, Chicago, IL 60611, United States of America; Edward Jr. Hines VA Hospital, Chicago, IL 60141, United States of America; Department of Physical Therapy and Human Movement Sciences, Northwestern University, Northwestern University, Chicago, IL 60611, United States of America; Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou, Guangdong 510095, PR China
| | - Milap Sandhu
- Shirley Ryan AbilityLab, Northwestern University, Chicago, IL 60611, United States of America
| | - W Zev Rymer
- Shirley Ryan AbilityLab, Northwestern University, Chicago, IL 60611, United States of America
| | - Randy D Trumbower
- Spaulding Rehabilitation Hospital, Cambridge Street, Cambridge, MA 02138, United States of America; Harvard Medical School, Department of Physical Medicine & Rehabilitation, Boston, MA 02115, United States of America
| | - Gordon S Mitchell
- Center for Respiratory Research and Rehabilitation, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, United States of America
| | - Monica A Perez
- University of Miami, Department of Neurological Surgery, The Miami Project to Cure Paralysis and Miami VA Medical Center, Miami, FL, 33136, United States of America; Shirley Ryan AbilityLab, Northwestern University, Chicago, IL 60611, United States of America; Edward Jr. Hines VA Hospital, Chicago, IL 60141, United States of America; Department of Physical Therapy and Human Movement Sciences, Northwestern University, Northwestern University, Chicago, IL 60611, United States of America.
| |
Collapse
|
56
|
Toparlak ÖD, Zasso J, Bridi S, Serra MD, Macchi P, Conti L, Baudet ML, Mansy SS. Artificial cells drive neural differentiation. SCIENCE ADVANCES 2020; 6:eabb4920. [PMID: 32948587 PMCID: PMC7500934 DOI: 10.1126/sciadv.abb4920] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 07/29/2020] [Indexed: 05/02/2023]
Abstract
We report the construction of artificial cells that chemically communicate with mammalian cells under physiological conditions. The artificial cells respond to the presence of a small molecule in the environment by synthesizing and releasing a potent protein signal, brain-derived neurotrophic factor. Genetically controlled artificial cells communicate with engineered human embryonic kidney cells and murine neural stem cells. The data suggest that artificial cells are a versatile chassis for the in situ synthesis and on-demand release of chemical signals that elicit desired phenotypic changes of eukaryotic cells, including neuronal differentiation. In the future, artificial cells could be engineered to go beyond the capabilities of typical smart drug delivery vehicles by synthesizing and delivering specific therapeutic molecules tailored to distinct physiological conditions.
Collapse
Affiliation(s)
- Ö Duhan Toparlak
- Department CIBIO, University of Trento, via Sommarive 9, 38123 Povo, Italy
| | - Jacopo Zasso
- Department CIBIO, University of Trento, via Sommarive 9, 38123 Povo, Italy
| | - Simone Bridi
- Department CIBIO, University of Trento, via Sommarive 9, 38123 Povo, Italy
| | - Mauro Dalla Serra
- National Research Council-Institute of Biophysics & Bruno Kessler Foundation, via alla Cascata 56/C, 38123 Trento, Italy
| | - Paolo Macchi
- Department CIBIO, University of Trento, via Sommarive 9, 38123 Povo, Italy
| | - Luciano Conti
- Department CIBIO, University of Trento, via Sommarive 9, 38123 Povo, Italy
| | - Marie-Laure Baudet
- Department CIBIO, University of Trento, via Sommarive 9, 38123 Povo, Italy
| | - Sheref S Mansy
- Department CIBIO, University of Trento, via Sommarive 9, 38123 Povo, Italy.
- Department of Chemistry, University of Alberta, 11227 Saskatchewan Drive, Edmonton, AB T6G 2G2, Canada
| |
Collapse
|
57
|
Gagner C, Tuerk C, De Beaumont L, Bernier A, Beauchamp MH. Brain-Derived Neurotrophic Factor Val66Met Polymorphism and Internalizing Behaviors after Early Mild Traumatic Brain Injury. J Neurotrauma 2020; 38:102-110. [PMID: 32605421 DOI: 10.1089/neu.2019.6936] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Pediatric traumatic brain injury (TBI) can lead to adverse emotional, social, and behavioral consequences. However, outcome is difficult to predict due to significant individual variability, likely reflecting a complex interaction between injury- and child-related variables. Among these variables are genetically determined individual differences, which can modulate TBI outcome through their influence on neuroplasticity mechanisms. In this study, we examined the effect of Val66Met, a common polymorphism of the brain-derived neurotrophic factor gene known to be involved in neuroplasticity mechanisms, on behavioral symptoms of mild TBI (mTBI) sustained in early childhood. This work is part of a prospective, longitudinal cohort study of early TBI. The current sample consisted of 145 children between ages 18 and 60 months assigned to one of three participant groups: mild TBI, orthopedic injury, or typically developing children. Participants provided a saliva sample to detect the presence of the Val66Met polymorphism, and the Child Behavior Checklist was used to document the presence of behavioral symptoms at 6- and 18-months post-injury. Contrary to our initial hypothesis, at 6 months post-injury, non-carriers of the Val66Met polymorphism in the mTBI group presented significantly more internalizing symptoms (e.g., anxiety/depression and somatic complaints) than Val66Met carriers, who were similar to orthopedically injured and typically developing children. However, at 18 months post-injury, all children with mTBI presented more internalizing symptoms, independent of genotype. The results of the study provide evidence for a protective effect of the Val66Met polymorphism on internalizing behavior symptoms 6 months after early childhood mTBI.
Collapse
Affiliation(s)
- Charlotte Gagner
- Department of Psychology, University of Montreal, Montréal, Québec, Canada.,Sainte-Justine Hospital Research Center, Montréal, Québec, Canada
| | - Carola Tuerk
- Department of Psychology, University of Montreal, Montréal, Québec, Canada
| | - Louis De Beaumont
- Hôpital du Sacré-Coeur de Montréal Research Center, Montréal, Québec, Canada.,Department of Surgery, University of Montreal, Montréal, Québec, Canada
| | - Annie Bernier
- Department of Psychology, University of Montreal, Montréal, Québec, Canada
| | - Miriam H Beauchamp
- Department of Psychology, University of Montreal, Montréal, Québec, Canada.,Sainte-Justine Hospital Research Center, Montréal, Québec, Canada
| |
Collapse
|
58
|
Yu Q, Zou L, Kong Z, Yang L. Cognitive Impact of Calorie Restriction: A Narrative Review. J Am Med Dir Assoc 2020; 21:1394-1401. [PMID: 32693996 DOI: 10.1016/j.jamda.2020.05.047] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 02/06/2023]
Abstract
The impairment of cognitive function can cause substantial emotional and financial burdens. A recent global increasing trend in cognitive impairment and associated disorders has been observed, which will continue to grow as the population ages rapidly. As a nonpharmaceutical approach, calorie restriction (CR) has received extensive research interests due to its health benefits, including maintaining cognitive function. In this narrative review, we first briefly introduce the role of cognitive function in activities of daily living and CR as a part of healthy lifestyle behaviors to protect against cognitive decline. Second, we present results from human studies demonstrating that CR might be beneficial for improving age-related cognitive decline and cognitive impairment in the clinical population such as obesity and type 2 diabetes. Third, the potential mechanisms regarding the protective effects of CR on cognition are discussed. Fourth, specific suggestions are highlighted to be considered in future human studies. Overall, although there are few data available from human studies, CR appears to be beneficial for cognitive protection for both healthy and clinical populations. Further scientific investigations are needed before a firm conclusion can be made.
Collapse
Affiliation(s)
- Qian Yu
- Exercise and Mental Health Laboratory, Shenzhen University, Shenzhen, China
| | - Liye Zou
- Exercise and Mental Health Laboratory, Shenzhen University, Shenzhen, China.
| | - Zhaowei Kong
- Faculty of Education, University of Macau, Macao, China
| | - Lin Yang
- Department of Cancer Epidemiology and Prevention Research, Alberta Health Services, Calgary, Canada; Departments of Oncology and Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Canada
| |
Collapse
|
59
|
O'Reilly ML, Tom VJ. Neuroimmune System as a Driving Force for Plasticity Following CNS Injury. Front Cell Neurosci 2020; 14:187. [PMID: 32792908 PMCID: PMC7390932 DOI: 10.3389/fncel.2020.00187] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 05/29/2020] [Indexed: 12/15/2022] Open
Abstract
Following an injury to the central nervous system (CNS), spontaneous plasticity is observed throughout the neuraxis and affects multiple key circuits. Much of this spontaneous plasticity can elicit beneficial and deleterious functional outcomes, depending on the context of plasticity and circuit affected. Injury-induced activation of the neuroimmune system has been proposed to be a major factor in driving this plasticity, as neuroimmune and inflammatory factors have been shown to influence cellular, synaptic, structural, and anatomical plasticity. Here, we will review the mechanisms through which the neuroimmune system mediates plasticity after CNS injury. Understanding the role of specific neuroimmune factors in driving adaptive and maladaptive plasticity may offer valuable therapeutic insight into how to promote adaptive plasticity and/or diminish maladaptive plasticity, respectively.
Collapse
Affiliation(s)
- Micaela L O'Reilly
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Veronica J Tom
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
60
|
Ifenprodil Reduced Expression of Activated Microglia, BDNF and DREAM Proteins in the Spinal Cord Following Formalin Injection During the Early Stage of Painful Diabetic Neuropathy in Rats. J Mol Neurosci 2020; 71:379-393. [PMID: 32671697 DOI: 10.1007/s12031-020-01661-1] [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: 03/02/2020] [Accepted: 07/08/2020] [Indexed: 10/23/2022]
Abstract
The pharmacological inhibition of glial activation is one of the new approaches for combating neuropathic pain in which the role of glia in the modulation of neuropathic pain has attracted significant interest and attention. Neuron-glial crosstalk is achieved with N-methyl-D-aspartate-2B receptor (NMDAR-2B) activation. This study aims to determine the effect of ifenprodil, a potent noncompetitive NMDAR-2B antagonist, on activated microglia, brain-derived neurotrophic factors (BDNF) and downstream regulatory element antagonist modulator (DREAM) protein expression in the spinal cord of streptozotocin-induced painful diabetic neuropathy (PDN) rats following formalin injection. In this experimentation, 48 Sprague-Dawley male rats were randomly selected and divided into four groups: (n = 12): control, PDN, and ifenprodil-treated PDN rats at 0.5 μg or 1.0 μg for 7 days. Type I diabetes mellitus was then induced by injecting streptozotocin (60 mg/kg, i.p.) into the rats which were then over a 2-week period allowed to progress into the early phase of PDN. Ifenprodil was administered in PDN rats while saline was administered intrathecally in the control group. A formalin test was conducted during the fourth week to induce inflammatory nerve injury, in which the rats were sacrificed at 72 h post-formalin injection. The lumbar enlargement region (L4-L5) of the spinal cord was dissected for immunohistochemistry and western blot analyses. The results demonstrated a significant increase in formalin-induced flinching and licking behavior with an increased spinal expression of activated microglia, BDNF and DREAM proteins. It was also shown that the ifenprodil-treated rats following both doses reduced the extent of their flinching and duration of licking in PDN in a dose-dependent manner. As such, ifenprodil successfully demonstrated inhibition against microglia activation and suppressed the expression of BDNF and DREAM proteins in the spinal cord of PDN rats. In conclusion, ifenprodil may alleviate PDN by suppressing spinal microglia activation, BDNF and DREAM proteins.
Collapse
|
61
|
Fang YC, Chan L, Liou JP, Tu YK, Lai MJ, Chen CI, Vidyanti AN, Lee HY, Hu CJ. HDAC inhibitor protects chronic cerebral hypoperfusion and oxygen-glucose deprivation injuries via H3K14 and H4K5 acetylation-mediated BDNF expression. J Cell Mol Med 2020; 24:6966-6977. [PMID: 32374084 PMCID: PMC7299713 DOI: 10.1111/jcmm.15358] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 04/16/2020] [Indexed: 12/14/2022] Open
Abstract
Vascular dementia (VaD) is the second most common cause of dementia, but the treatment is still lacking. Although many studies have reported that histone deacetylase inhibitors (HDACis) confer protective effects against ischemic and hypoxic injuries, their role in VaD is still uncertain. Previous studies shown, one HDACi protected against cognitive decline in animals with chronic cerebral hypoperfusion (CCH). However, the underlying mechanisms remain elusive. In this study, we tested several 10,11‐dihydro‐5H‐dibenzo[b,f]azepine hydroxamates, which act as HDACis in the CCH model (in vivo), and SH‐SY5Y (neuroblastoma cells) with oxygen‐glucose deprivation (OGD, in vitro). We identified a compound 13, which exhibited the best cell viability under OGD. The compound 13 could increase, in part, the protein levels of brain‐derived neurotrophic factor (BDNF). It increased acetylation status on lysine 14 residue of histone 3 (H3K14) and lysine 5 of histone 4 (H4K5). We further clarified which promoters (I, II, III, IV or IX) could be affected by histone acetylation altered by compound 13. The results of chromatin immunoprecipitation and Q‐PCR analysis indicate that an increase in H3K14 acetylation leads to an increase in the expression of BDNF promoter II, while an increase in H4K5 acetylation results in an increase in the activity of BDNF promoter II and III. Afterwards, these cause an increase in the expression of BDNF exon II, III and coding exon IX. In summary, the HDACi compound 13 may increase BDNF specific isoforms expression to rescue the ischemic and hypoxic injuries through changes of acetylation on histones.
Collapse
Affiliation(s)
- Yao-Ching Fang
- Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan
| | - Lung Chan
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Jing-Ping Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.,TMU Biomedical Commercialization Center, Taipei Medical University, Taipei, Taiwan
| | - Yong-Kwang Tu
- Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan.,Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Mei-Jung Lai
- TMU Biomedical Commercialization Center, Taipei Medical University, Taipei, Taiwan
| | - Chin-I Chen
- Department of Neurology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Amelia Nur Vidyanti
- Department of Neurology, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia.,International Master/PhD Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsueh-Yun Lee
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Chaur-Jong Hu
- Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan.,Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| |
Collapse
|
62
|
Sutherland TC, Geoffroy CG. The Influence of Neuron-Extrinsic Factors and Aging on Injury Progression and Axonal Repair in the Central Nervous System. Front Cell Dev Biol 2020; 8:190. [PMID: 32269994 PMCID: PMC7109259 DOI: 10.3389/fcell.2020.00190] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 03/06/2020] [Indexed: 12/21/2022] Open
Abstract
In the aging western population, the average age of incidence for spinal cord injury (SCI) has increased, as has the length of survival of SCI patients. This places great importance on understanding SCI in middle-aged and aging patients. Axon regeneration after injury is an area of study that has received substantial attention and made important experimental progress, however, our understanding of how aging affects this process, and any therapeutic effort to modulate repair, is incomplete. The growth and regeneration of axons is mediated by both neuron intrinsic and extrinsic factors. In this review we explore some of the key extrinsic influences on axon regeneration in the literature, focusing on inflammation and astrogliosis, other cellular responses, components of the extracellular matrix, and myelin proteins. We will describe how each element supports the contention that axonal growth after injury in the central nervous system shows an age-dependent decline, and how this may affect outcomes after a SCI.
Collapse
Affiliation(s)
- Theresa C Sutherland
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, TX, United States
| | - Cédric G Geoffroy
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, TX, United States
| |
Collapse
|
63
|
Cross-talk signaling in the trigeminal ganglion: role of neuropeptides and other mediators. J Neural Transm (Vienna) 2020; 127:431-444. [PMID: 32088764 PMCID: PMC7148261 DOI: 10.1007/s00702-020-02161-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 02/12/2020] [Indexed: 11/08/2022]
Abstract
The trigeminal ganglion with its three trigeminal nerve tracts consists mainly of clusters of sensory neurons with their peripheral and central processes. Most neurons are surrounded by satellite glial cells and the axons are wrapped by myelinating and non-myelinating Schwann cells. Trigeminal neurons express various neuropeptides, most notably, calcitonin gene-related peptide (CGRP), substance P, and pituitary adenylate cyclase-activating polypeptide (PACAP). Two types of CGRP receptors are expressed in neurons and satellite glia. A variety of other signal molecules like ATP, nitric oxide, cytokines, and neurotrophic factors are released from trigeminal ganglion neurons and signal to neighboring neurons or satellite glial cells, which can signal back to neurons with same or other mediators. This potential cross-talk of signals involves intracellular mechanisms, including gene expression, that can modulate mediators of sensory information, such as neuropeptides, receptors, and neurotrophic factors. From the ganglia cell bodies, which are outside the blood–brain barrier, the mediators are further distributed to peripheral sites and/or to the spinal trigeminal nucleus in the brainstem, where they can affect neural transmission. A major question is how the sensory neurons in the trigeminal ganglion differ from those in the dorsal root ganglion. Despite their functional overlap, there are distinct differences in their ontogeny, gene expression, signaling pathways, and responses to anti-migraine drugs. Consequently, drugs that modulate cross-talk in the trigeminal ganglion can modulate both peripheral and central sensitization, which may potentially be distinct from sensitization mediated in the dorsal root ganglion.
Collapse
|
64
|
Dux M, Rosta J, Messlinger K. TRP Channels in the Focus of Trigeminal Nociceptor Sensitization Contributing to Primary Headaches. Int J Mol Sci 2020; 21:ijms21010342. [PMID: 31948011 PMCID: PMC6981722 DOI: 10.3390/ijms21010342] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 12/31/2019] [Accepted: 01/02/2020] [Indexed: 12/12/2022] Open
Abstract
Pain in trigeminal areas is driven by nociceptive trigeminal afferents. Transduction molecules, among them the nonspecific cation channels transient receptor potential vanilloid 1 (TRPV1) and ankyrin 1 (TRPA1), which are activated by endogenous and exogenous ligands, are expressed by a significant population of trigeminal nociceptors innervating meningeal tissues. Many of these nociceptors also contain vasoactive neuropeptides such as calcitonin gene-related peptide (CGRP) and substance P. Release of neuropeptides and other functional properties are frequently examined using the cell bodies of trigeminal neurons as models of their sensory endings. Pathophysiological conditions cause phosphorylation, increased expression and trafficking of transient receptor potential (TRP) channels, neuropeptides and other mediators, which accelerate activation of nociceptive pathways. Since nociceptor activation may be a significant pathophysiological mechanism involved in both peripheral and central sensitization of the trigeminal nociceptive pathway, its contribution to the pathophysiology of primary headaches is more than likely. Metabolic disorders and medication-induced painful states are frequently associated with TRP receptor activation and may increase the risk for primary headaches.
Collapse
Affiliation(s)
- Mária Dux
- Department of Physiology, University of Szeged, Dóm tér 10, H-6720 Szeged, Hungary;
- Correspondence: ; Tel.: +36-62-545-374; Fax: +36-62-545-842
| | - Judit Rosta
- Department of Physiology, University of Szeged, Dóm tér 10, H-6720 Szeged, Hungary;
| | - Karl Messlinger
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-University Erlangen-Nürnberg, Universitätsstr. 17, D-91054 Erlangen, Germany;
| |
Collapse
|
65
|
Zhou X, Tao L, Zhao M, Wu S, Obeng E, Wang D, Zhang W. Wnt/ β-catenin signaling regulates brain-derived neurotrophic factor release from spinal microglia to mediate HIV 1 gp120-induced neuropathic pain. Mol Pain 2020; 16:1744806920922100. [PMID: 32354292 PMCID: PMC7227158 DOI: 10.1177/1744806920922100] [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] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 03/13/2020] [Accepted: 03/30/2020] [Indexed: 12/14/2022] Open
Abstract
HIV-associated neuropathic pain (HNP) is a common complication for AIDS patients. The pathological mechanism governing HNP has not been elucidated, and HNP has no effective analgesic treatment. Brain-derived neurotrophic factor (BDNF) is a member of the neurotrophic factor family related to the plasticity of the central nervous system. BDNF dysregulation is involved in many neurological diseases, including neuropathic pain. However, to the best of our knowledge, the role and mechanism of BDNF in HNP have not been elucidated. In this study, we explored this condition in an HNP mouse model induced by intrathecal injection of gp120. We found that Wnt3a and β-catenin expression levels increased in the spinal cord of HNP mice, consequently regulating the expression of BDNF and affecting hypersensitivity. In addition, the blockade of Wing-Int/β-catenin signaling, BDNF/TrkB or the BDNF/p75NTR pathway alleviated mechanical allodynia. BDNF immunoreactivity was colocalized with spinal microglial cells, which were activated in HNP mice. Inhibition of spinal microglial cell activation by minocycline relieved mechanical allodynia in HNP mice. This study helped to elucidate the role of the Wing-Int/β-catenin/BDNF signaling axis in HNP and may establish a foundation for further research investigating the Wing-Int/β-catenin/BDNF signaling axis as a target for HNP treatment.
Collapse
Affiliation(s)
- Xinxin Zhou
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Lei Tao
- School of Life Sciences, East China Normal University, Shanghai, China
| | - Mengru Zhao
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Shengjun Wu
- Clinical Laboratory of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Enoch Obeng
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Dan Wang
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Wenping Zhang
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| |
Collapse
|
66
|
Zortea M, Ramalho L, Alves RL, Alves CFDS, Braulio G, Torres ILDS, Fregni F, Caumo W. Transcranial Direct Current Stimulation to Improve the Dysfunction of Descending Pain Modulatory System Related to Opioids in Chronic Non-cancer Pain: An Integrative Review of Neurobiology and Meta-Analysis. Front Neurosci 2019; 13:1218. [PMID: 31803005 PMCID: PMC6876542 DOI: 10.3389/fnins.2019.01218] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 10/29/2019] [Indexed: 12/20/2022] Open
Abstract
Background: Opioid long-term therapy can produce tolerance, opioid-induced hyperalgesia (OIH), and it induces dysfunction in pain descending pain inhibitory system (DPIS). Objectives: This integrative review with meta-analysis aimed: (i) To discuss the potential mechanisms involved in analgesic tolerance and opioid-induced hyperalgesia (OIH). (ii) To examine how the opioid can affect the function of DPIS. (ii) To show evidence about the tDCS as an approach to treat acute and chronic pain. (iii) To discuss the effect of tDCS on DPIS and how it can counter-regulate the OIH. (iv) To draw perspectives for the future about the tDCS effects as an approach to improve the dysfunction in the DPIS in chronic non-cancer pain. Methods: Relevant published randomized clinical trials (RCT) comparing active (irrespective of the stimulation protocol) to sham tDCS for treating chronic non-cancer pain were identified, and risk of bias was assessed. We searched trials in PubMed, EMBASE and Cochrane trials databases. tDCS protocols accepted were application in areas of the primary motor cortex (M1), dorsolateral prefrontal cortex (DLPFC), or occipital area. Results: Fifty-nine studies were fully reviewed, and 24 with moderate to the high-quality methodology were included. tDCS improved chronic pain with a moderate effect size [pooled standardized mean difference; -0.66; 95% confidence interval (CI) -0.91 to -0.41]. On average, active protocols led to 27.26% less pain at the end of treatment compared to sham [95% CI; 15.89-32.90%]. Protocol varied in terms of anodal or cathodal stimulation, areas of stimulation (M1 and DLPFC the most common), number of sessions (from 5 to 20) and current intensity (from 1 to 2 mA). The time of application was 20 min in 92% of protocols. Conclusion: In comparison with sham stimulation, tDCS demonstrated a superior effect in reducing chronic pain conditions. They give perspectives that the top-down neuromodulator effects of tDCS are a promising approach to improve management in refractory chronic not-cancer related pain and to enhance dysfunctional neuronal circuitries involved in the DPIS and other pain dimensions and improve pain control with a therapeutic opioid-free. However, further studies are needed to determine individualized protocols according to a biopsychosocial perspective.
Collapse
Affiliation(s)
- Maxciel Zortea
- Post-graduation Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, Brazil.,Laboratory of Pain & Neuromodulation, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Leticia Ramalho
- Post-graduation Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, Brazil.,Laboratory of Pain & Neuromodulation, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Rael Lopes Alves
- Post-graduation Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, Brazil.,Laboratory of Pain & Neuromodulation, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Camila Fernanda da Silveira Alves
- Post-graduation Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, Brazil.,Laboratory of Pain & Neuromodulation, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Gilberto Braulio
- Laboratory of Pain & Neuromodulation, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil.,Service of Anesthesia and Perioperative Medicine, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Iraci Lucena da Silva Torres
- Department of Pharmacology, Institute of Health Sciences (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, Brazil.,Pharmacology of Pain and Neuromodulation: Pre-clinical Investigations Research Group, Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, Brazil
| | - Felipe Fregni
- Neuromodulation Center, Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA, United States
| | - Wolnei Caumo
- Post-graduation Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, Brazil.,Laboratory of Pain & Neuromodulation, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil.,Pain Treatment and Palliative Medicine Service, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| |
Collapse
|
67
|
Ma L, Yue L, Zhang Y, Wang Y, Han B, Cui S, Liu FY, Wan Y, Yi M. Spontaneous Pain Disrupts Ventral Hippocampal CA1-Infralimbic Cortex Connectivity and Modulates Pain Progression in Rats with Peripheral Inflammation. Cell Rep 2019; 29:1579-1593.e6. [DOI: 10.1016/j.celrep.2019.10.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 09/02/2019] [Accepted: 10/01/2019] [Indexed: 12/11/2022] Open
|
68
|
Acupuncture-Analgesia-Mediated Alleviation of Central Sensitization. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:6173412. [PMID: 30984277 PMCID: PMC6431485 DOI: 10.1155/2019/6173412] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/20/2018] [Accepted: 02/06/2019] [Indexed: 12/20/2022]
Abstract
Pain can trigger central amplification called central sensitization, which ultimately results in hyperalgesia and/or allodynia. Many reports have showed acupuncture has an analgesic effect. We searched the related article on PubMed database and Cochrane database to discover central sensitization pathway in acupuncture analgesia. We summarized that acupuncture enhances the descending inhibitory effect and modulates the feeling of pain, thus modifying central sensitization. The possible mechanisms underlying the analgesic effects of acupuncture include segmental inhibition and the activation of the endogenous opioid, adrenergic, 5-hydroxytryptamine, and N-methyl-D-aspartic acid, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid/kainate pathways. Moreover, acupuncture can locally reduce the levels of inflammatory mediators. In clinical settings, acupuncture can be used to treat headache, neuropathic pain, low back pain, osteoarthritis, and irritable bowel syndrome. These mechanisms of acupuncture analgesia may be involved in the alleviation of central sensitization.
Collapse
|
69
|
Salehi MS, Borhani-Haghighi A, Pandamooz S, Safari A, Dargahi L, Dianatpour M, Tanideh N. Dimethyl fumarate up-regulates expression of major neurotrophic factors in the epidermal neural crest stem cells. Tissue Cell 2019; 56:114-120. [PMID: 30736899 DOI: 10.1016/j.tice.2019.01.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 01/20/2019] [Accepted: 01/21/2019] [Indexed: 02/07/2023]
Abstract
There is an agreement that combining treatments can lead to substantial improvement, therefore the present study assessed the effects of different concentrations of dimethyl fumarate (DMF) on viability of epidermal neural crest stem cells (EPI-NCSCs). In addition, this investigation was designed to evaluate the effects of DMF on relative expression of major trophic factors mainly the ones with neurotrophic effects, expressed in EPI-NCSCs in order to enhance their therapeutic potential. To determine the appropriate concentration of DMF for EPI-NCSCs treatment, the MTT assay was employed and based on the obtained data, EPI-NCSCs treated with 10μM DMF for 6, 24, 72 or 168 h. In each time point, quantitative RT-PCR technique was used to evaluate NGF, NT-3, BDNF, GDNF and VEGF transcripts. The acquired data showed that 10μM DMF significantly increased the mRNA expression of NGF, NT-3 and BDNF, 72 h following treatment; however, DMF inhibitory effect on GDNF mRNA expression was observed in various time points. No significant changes were detected for VEGF transcript. Our findings reveled that expression of major neurotrophic factors were up-regulated by dimethyl fumarate treatment. Therefore, combining EPI-NCSCs with DMF treatment might be a valuable strategy to improve their therapeutic functions in vivo.
Collapse
Affiliation(s)
- Mohammad Saied Salehi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Sareh Pandamooz
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Anahid Safari
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Leila Dargahi
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Dianatpour
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nader Tanideh
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
70
|
Yi NX, Zhou LY, Wang XY, Song YJ, Han HH, Zhang TS, Wang YJ, Shi Q, Xu H, Liang QQ, Zhang T. MK-801 attenuates lesion expansion following acute brain injury in rats: a meta-analysis. Neural Regen Res 2019; 14:1919-1931. [PMID: 31290450 PMCID: PMC6676887 DOI: 10.4103/1673-5374.259619] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVE: To evaluate the efficacy and safety of MK-801 and its effect on lesion volume in rat models of acute brain injury. DATA SOURCES: Key terms were “stroke”, “brain diseases”, “brain injuries”, “brain hemorrhage, traumatic”, “acute brain injury”, “dizocilpine maleate”, “dizocilpine”, “MK-801”, “MK801”, “rat”, “rats”, “rattus” and “murine”. PubMed, Cochrane library, EMBASE, the China National Knowledge Infrastructure, WanFang database, the VIP Journal Integration Platform (VJIP) and SinoMed databases were searched from their inception dates to March 2018. DATA SELECTION: Studies were selected if they reported the effects of MK-801 in experimental acute brain injury. Two investigators independently conducted literature screening, data extraction, and methodological quality assessments. OUTCOME MEASURES: The primary outcomes included lesion volume and brain edema. The secondary outcomes included behavioral assessments with the Bederson neurological grading system and the water maze test 24 hours after brain injury. RESULTS: A total of 52 studies with 2530 samples were included in the systematic review. Seventeen of these studies had a high methodological quality. Overall, the lesion volume (34 studies, n = 966, MD = −58.31, 95% CI: −66.55 to −50.07; P < 0.00001) and degree of cerebral edema (5 studies, n = 75, MD = −1.21, 95% CI: −1.50 to −0.91; P < 0.00001) were significantly decreased in the MK-801 group compared with the control group. MK-801 improved spatial cognition assessed with the water maze test (2 studies, n = 60, MD = −10.88, 95% CI: −20.75 to −1.00; P = 0.03) and neurological function 24 hours after brain injury (11 studies, n = 335, MD = −1.04, 95% CI: −1.47 to −0.60; P < 0.00001). Subgroup analysis suggested an association of reduction in lesion volume with various injury models (34 studies, n = 966, MD = −58.31, 95% CI: −66.55 to −50.07; P = 0.004). Further network analysis showed that 0–1 mg/kg MK-801 may be the optimal dose for treatment in the middle cerebral artery occlusion animal model. CONCLUSION: MK-801 effectively reduces brain lesion volume and the degree of cerebral edema in rat models of experimental acute brain injury, providing a good neuroprotective effect. Additionally, MK-801 has a good safety profile, and its mechanism of action is well known. Thus, MK-801 may be suitable for future clinical trials and applications.
Collapse
Affiliation(s)
- Nan-Xing Yi
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine; Institute of Spine, Shanghai University of Traditional Chinese Medicine; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai, China
| | - Long-Yun Zhou
- Institute of Spine, Shanghai University of Traditional Chinese Medicine; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education; School of Rehabilitation Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao-Yun Wang
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yong-Jia Song
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine; Institute of Spine, Shanghai University of Traditional Chinese Medicine; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai, China
| | - Hai-Hui Han
- Institute of Spine; Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tian-Song Zhang
- Jing'an District Center Hospital, Fudan University, Shanghai, China
| | - Yong-Jun Wang
- Institute of Spine, Shanghai University of Traditional Chinese Medicine; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai, China
| | - Qi Shi
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine; Institute of Spine, Shanghai University of Traditional Chinese Medicine; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education; Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hao Xu
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine; Institute of Spine, Shanghai University of Traditional Chinese Medicine; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai, China
| | - Qian-Qian Liang
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine; Institute of Spine, Shanghai University of Traditional Chinese Medicine; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai, China
| | - Ting Zhang
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine; Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
71
|
Luo H, Xu C, Liu Z, Yang L, Hong Y, Liu G, Zhong H, Cai X, Lin X, Chen X, Wang C, Nanwen Z, Xu W. Neural differentiation of bone marrow mesenchymal stem cells with human brain‐derived neurotrophic factor gene‐modified in functionalized self‐assembling peptide hydrogel in vitro. J Cell Biochem 2018; 120:2828-2835. [PMID: 28929517 DOI: 10.1002/jcb.26408] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 09/12/2017] [Indexed: 11/10/2022]
Affiliation(s)
- Hongbin Luo
- Department of Orthopedics The First Affiliated Hospital of Fujian Medical University (FMU) Fuzhou People’s Republic of China
| | - Changsheng Xu
- Institute of Hypertension, The First Affiliated Hospital of Fujian Medical University (FMU) Fuzhou People’s Republic of China
| | - Zhiwei Liu
- School of Clinical Medical, Fujian Medical University (FMU) Fuzhou People’s Republic of China
| | - Lin Yang
- School of Clinical Medical, Fujian Medical University (FMU) Fuzhou People’s Republic of China
| | - Yunda Hong
- School of Clinical Medical, Fujian Medical University (FMU) Fuzhou People’s Republic of China
| | - Guisheng Liu
- School of Clinical Medical, Fujian Medical University (FMU) Fuzhou People’s Republic of China
| | - Huifen Zhong
- School of Clinical Medical, Fujian Medical University (FMU) Fuzhou People’s Republic of China
| | - Xinyi Cai
- School of Clinical Medical, Fujian Medical University (FMU) Fuzhou People’s Republic of China
| | - Xuping Lin
- School of Clinical Medical, Fujian Medical University (FMU) Fuzhou People’s Republic of China
| | - Xiaokun Chen
- School of Clinical Medical, Fujian Medical University (FMU) Fuzhou People’s Republic of China
| | - Changsheng Wang
- Department of Orthopedics The First Affiliated Hospital of Fujian Medical University (FMU) Fuzhou People’s Republic of China
| | - Zhang Nanwen
- Department of Pharmacology School of pharmacy, Fujian Medical University (FMU) Fuzhou People’s Republic of China
| | - Weihong Xu
- Department of Orthopedics The First Affiliated Hospital of Fujian Medical University (FMU) Fuzhou People’s Republic of China
| |
Collapse
|
72
|
Messlinger K, Russo AF. Current understanding of trigeminal ganglion structure and function in headache. Cephalalgia 2018; 39:1661-1674. [PMID: 29989427 DOI: 10.1177/0333102418786261] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION The trigeminal ganglion is unique among the somatosensory ganglia regarding its topography, structure, composition and possibly some functional properties of its cellular components. Being mainly responsible for the sensory innervation of the anterior regions of the head, it is a major target for headache research. One intriguing question is if the trigeminal ganglion is merely a transition site for sensory information from the periphery to the central nervous system, or if intracellular modulatory mechanisms and intercellular signaling are capable of controlling sensory information relevant for the pathophysiology of headaches. METHODS An online search based on PubMed was made using the keyword "trigeminal ganglion" in combination with "anatomy", "headache", "migraine", "neuropeptides", "receptors" and "signaling". From the relevant literature, further references were selected in view of their relevance for headache mechanisms. The essential information was organized based on location and cell types of the trigeminal ganglion, neuropeptides, receptors for signaling molecules, signaling mechanisms, and their possible relevance for headache generation. RESULTS The trigeminal ganglion consists of clusters of sensory neurons and their peripheral and central axon processes, which are arranged according to the three trigeminal partitions V1-V3. The neurons are surrounded by satellite glial cells, the axons by Schwann cells. In addition, macrophage-like cells can be found in the trigeminal ganglion. Neurons express various neuropeptides, among which calcitonin gene-related peptide is the most prominent in terms of its prevalence and its role in primary headaches. The classical calcitonin gene-related peptide receptors are expressed in non-calcitonin gene-related peptide neurons and satellite glial cells, although the possibility of a second calcitonin gene-related peptide receptor in calcitonin gene-related peptide neurons remains to be investigated. A variety of other signal molecules like adenosine triphosphate, nitric oxide, cytokines, and neurotrophic factors are released from trigeminal ganglion cells and may act at receptors on adjacent neurons or satellite glial cells. CONCLUSIONS The trigeminal ganglion may act as an integrative organ. The morphological and functional arrangement of trigeminal ganglion cells suggests that intercellular and possibly also autocrine signaling mechanisms interact with intracellular mechanisms, including gene expression, to modulate sensory information. Receptors and neurotrophic factors delivered to the periphery or the trigeminal brainstem can contribute to peripheral and central sensitization, as in the case of primary headaches. The trigeminal ganglion as a target of drug action outside the blood-brain barrier should therefore be taken into account.
Collapse
Affiliation(s)
- Karl Messlinger
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Andrew F Russo
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA.,Iowa VA Health Care System, Iowa City, IA, USA
| |
Collapse
|
73
|
Acute intermittent hypoxia and rehabilitative training following cervical spinal injury alters neuronal hypoxia- and plasticity-associated protein expression. PLoS One 2018; 13:e0197486. [PMID: 29775479 PMCID: PMC5959066 DOI: 10.1371/journal.pone.0197486] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 05/03/2018] [Indexed: 01/24/2023] Open
Abstract
One of the most promising approaches to improve recovery after spinal cord injury (SCI) is the augmentation of spontaneously occurring plasticity in uninjured neural pathways. Acute intermittent hypoxia (AIH, brief exposures to reduced O2 levels alternating with normal O2 levels) initiates plasticity in respiratory systems and has been shown to improve recovery in respiratory and non-respiratory spinal systems after SCI in experimental animals and humans. Although the mechanism by which AIH elicits its effects after SCI are not well understood, AIH is known to alter protein expression in spinal neurons in uninjured animals. Here, we examine hypoxia- and plasticity-related protein expression using immunofluorescence in spinal neurons in SCI rats that were treated with AIH combined with motor training, a protocol which has been demonstrated to improve recovery of forelimb function in this lesion model. Specifically, we assessed protein expression in spinal neurons from animals with incomplete cervical SCI which were exposed to AIH treatment + motor training either for 1 or 7 days. AIH treatment consisted of 10 episodes of AIH: (5 min 11% O2: 5 min 21% O2) for 7 days beginning at 4 weeks post-SCI. Both 1 or 7 days of AIH treatment + motor training resulted in significantly increased expression of the transcription factor hypoxia-inducible factor-1α (HIF-1α) relative to normoxia-treated controls, in neurons both proximal (cervical) and remote (lumbar) to the SCI. All other markers examined were significantly elevated in the 7 day AIH + motor training group only, at both cervical and lumbar levels. These markers included vascular endothelial growth factor (VEGF), brain-derived neurotrophic factor (BDNF), and phosphorylated and nonphosphorylated forms of the BDNF receptor tropomyosin-related kinase B (TrkB). In summary, AIH induces plasticity at the cellular level after SCI by altering the expression of major plasticity- and hypoxia-related proteins at spinal regions proximal and remote to the SCI. These changes occur under the same AIH protocol which resulted in recovery of limb function in this animal model. Thus AIH, which induces plasticity in spinal circuitry, could also be an effective therapy to restore motor function after nervous system injury.
Collapse
|
74
|
Limongi T, Rocchi A, Cesca F, Tan H, Miele E, Giugni A, Orlando M, Perrone Donnorso M, Perozziello G, Benfenati F, Di Fabrizio E. Delivery of Brain-Derived Neurotrophic Factor by 3D Biocompatible Polymeric Scaffolds for Neural Tissue Engineering and Neuronal Regeneration. Mol Neurobiol 2018; 55:8788-8798. [PMID: 29600349 DOI: 10.1007/s12035-018-1022-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 03/16/2018] [Indexed: 01/07/2023]
Abstract
Biopolymers are increasingly employed for neuroscience applications as scaffolds to drive and promote neural regrowth, thanks to their ability to mediate the upload and subsequent release of active molecules and drugs. Synthetic degradable polymers are characterized by different responses ranging from tunable distension or shrinkage to total dissolution, depending on the function they are designed for. In this paper we present a biocompatible microfabricated poly-ε-caprolactone (PCL) scaffold for primary neuron growth and maturation that has been optimized for the in vitro controlled release of brain-derived neurotrophic factor (BDNF). We demonstrate that the designed morphology confers to these devices an enhanced drug delivery capability with respect to monolithic unstructured supports. After incubation with BDNF, micropillared PCL devices progressively release the neurotrophin over 21 days in vitro. Moreover, the bioactivity of released BDNF is confirmed using primary neuronal cultures, where it mediates a consistent activation of BDNF signaling cascades, increased synaptic density, and neuronal survival. These results provide the proof-of-principle on the fabrication process of micropatterned PCL devices, which represent a promising therapeutic option to enhance neuronal regeneration after lesion and for neural tissue engineering and prosthetics.
Collapse
Affiliation(s)
- T Limongi
- SMILEs Lab, Physical Science and Engineering (PSE) and Biological and Environmental Science and Engineering (BESE) Divisions, King Abdullah University of Science and Technology, Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - A Rocchi
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - F Cesca
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - H Tan
- Analytical Core Lab, King Abdullah University of Science and Technology, Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - E Miele
- Nanostructures Department, Istituto Italiano di Tecnologia, Via Morego 30, 16163, Genoa, Italy.,Centre for BioImaging Sciences, Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore, 117543, Singapore
| | - A Giugni
- SMILEs Lab, Physical Science and Engineering (PSE) and Biological and Environmental Science and Engineering (BESE) Divisions, King Abdullah University of Science and Technology, Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - M Orlando
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132, Genoa, Italy.,Department of Neurophysiology, NeuroCure Excellence Cluster, Charité Universitäts Medizin, Charitéplatz 1, 10117, Berlin, Germany
| | - M Perrone Donnorso
- Nanostructures Department, Istituto Italiano di Tecnologia, Via Morego 30, 16163, Genoa, Italy
| | - G Perozziello
- Laboratory of Nanotechnology BioNEM Department of Experimental and Clinical Medicine, University "Magna Graecia" of Catanzaro, Viale Europa, Catanzaro, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132, Genoa, Italy.
| | - Enzo Di Fabrizio
- SMILEs Lab, Physical Science and Engineering (PSE) and Biological and Environmental Science and Engineering (BESE) Divisions, King Abdullah University of Science and Technology, Thuwal, 23955-6900, Kingdom of Saudi Arabia.
| |
Collapse
|
75
|
Abstract
BACKGROUND Calcitonin gene-related peptide (CGRP) has long been a focus of migraine research, since it turned out that inhibition of CGRP or CGRP receptors by antagonists or monoclonal IgG antibodies was therapeutic in frequent and chronic migraine. This contribution deals with the questions, from which sites CGRP is released, where it is drained and where it acts to cause its headache proliferating effects in the trigeminovascular system. RESULTS The available literature suggests that the bulk of CGRP is released from trigeminal afferents both in meningeal tissues and at the first synapse in the spinal trigeminal nucleus. CGRP may be drained off into three different compartments, the venous blood plasma, the cerebrospinal fluid and possibly the glymphatic system. CGRP receptors in peripheral tissues are located on arterial vessel walls, mononuclear immune cells and possibly Schwann cells; within the trigeminal ganglion they are located on neurons and glial cells; in the spinal trigeminal nucleus they can be found on central terminals of trigeminal afferents. All these structures are potential signalling sites for CGRP, where CGRP mediates arterial vasodilatation but not direct activation of trigeminal afferents. In the spinal trigeminal nucleus a facilitating effect on synaptic transmission seems likely. In the trigeminal ganglion CGRP is thought to initiate long-term changes including cross-signalling between neurons and glial cells based on gene expression. In this way, CGRP may upregulate the production of receptor proteins and pro-nociceptive molecules. CONCLUSIONS CGRP and other big molecules cannot easily pass the blood-brain barrier. These molecules may act in the trigeminal ganglion to influence the production of pronociceptive substances and receptors, which are transported along the central terminals into the spinal trigeminal nucleus. In this way peripherally acting therapeutics can have a central antinociceptive effect.
Collapse
Affiliation(s)
- Karl Messlinger
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsstr. 17, 91054, Erlangen, Germany.
| |
Collapse
|
76
|
Opposing Roles of Estradiol and Testosterone on Stress-Induced Visceral Hypersensitivity in Rats. THE JOURNAL OF PAIN 2018; 19:764-776. [PMID: 29496640 DOI: 10.1016/j.jpain.2018.02.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 01/30/2018] [Accepted: 02/14/2018] [Indexed: 12/19/2022]
Abstract
Chronic stress produces maladaptive pain responses, manifested as alterations in pain processing and exacerbation of chronic pain conditions including irritable bowel syndrome. Female predominance, especially during reproductive years, strongly suggests a role of gonadal hormones. However, gonadal hormone modulation of stress-induced pain hypersensitivity is not well understood. In the present study, we tested the hypothesis that estradiol is pronociceptive and testosterone is antinociceptive in a model of stress-induced visceral hypersensitivity (SIVH) in rats by recording the visceromotor response to colorectal distention after a 3-day forced swim (FS) stress paradigm. FS induced visceral hypersensitivity that persisted at least 2 weeks in female, but only 2 days in male rats. Ovariectomy blocked and orchiectomy facilitated SIVH. Furthermore, estradiol injection in intact male rats increased SIVH and testosterone in intact female rats attenuated SIVH. Western blot analyses indicated estradiol increased excitatory glutamate ionotropic receptor NMDA type subunit 1 expression and decreased inhibitory metabotropic glutamate receptor 2 expression after FS in male thoracolumbar spinal cord. In addition, the presence of estradiol during stress increased spinal brain-derived neurotrophic factor (BDNF) expression independent of sex. In contrast, testosterone blocked the stress-induced increase in BDNF expression in female rats. These data suggest that estradiol facilitates and testosterone attenuates SIVH by modulating spinal excitatory and inhibitory glutamatergic receptor expression. PERSPECTIVE SIVH is more robust in female rats. Estradiol facilitates whereas testosterone dampens the development of SIVH. This could partially explain the greater prevalence of certain chronic visceral pain conditions in women. An increase in spinal BDNF is concomitant with increased stress-induced pain. Pharmaceutical interventions targeting this molecule could provide promising alleviation of SIVH in women.
Collapse
|
77
|
Axelsen TM, Woldbye DP. Gene Therapy for Parkinson's Disease, An Update. JOURNAL OF PARKINSON'S DISEASE 2018; 8:195-215. [PMID: 29710735 PMCID: PMC6027861 DOI: 10.3233/jpd-181331] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 03/25/2018] [Indexed: 12/19/2022]
Abstract
The current mainstay treatment of Parkinson's disease (PD) consists of dopamine replacement therapy which, in addition to causing several side effects, does not delay disease progression. The field of gene therapy offers a potential means to improve current therapy. The present review gives an update of the present status of gene therapy for PD. Both non-disease and disease modifying transgenes have been tested for PD gene therapy in animal and human studies. Non-disease modifying treatments targeting dopamine or GABA synthesis have been successful and promising at improving PD symptomatology in randomized clinical studies, but substantial testing remains before these can be implemented in the standard clinical treatment repertoire. As for disease modifying targets that theoretically offer the possibility of slowing the progression of disease, several neurotrophic factors show encouraging results in preclinical models (e.g., neurturin, GDNF, BDNF, CDNF, VEGF-A). However, so far, clinical trials have only tested neurturin, and, unfortunately, no trial has been able to meet its primary endpoint. Future clinical trials with neurotrophic factors clearly deserve to be conducted, considering the still enticing goal of actually slowing the disease process of PD. As alternative types of gene therapy, opto- and chemogenetics might also find future use in PD treatment and novel genome-editing technology could also potentially be applied as individualized gene therapy for genetic types of PD.
Collapse
Affiliation(s)
- Tobias M. Axelsen
- Department of Neurology, Herlev University Hospital, Herlev, Denmark
| | - David P.D. Woldbye
- Department of Neuroscience, Panum Institute, Mærsk Tower, University of Copenhagen, Copenhagen N, Denmark
| |
Collapse
|
78
|
Neves CDC, Lacerda ACR, Lima LP, Lage VKS, Balthazar CH, Leite HR, Mendonça VA. Different levels of brain-derived neurotrophic factor and cortisol in healthy heavy smokers. ACTA ACUST UNITED AC 2017; 50:e6424. [PMID: 29069228 PMCID: PMC5649870 DOI: 10.1590/1414-431x20176424] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 08/25/2017] [Indexed: 11/22/2022]
Abstract
Studies suggest that brain-derived neurotrophic factor (BDNF) and the hypothalamic-pituitary-adrenal (HPA) axis modulate dopaminergic activity in response to nicotine and that the concentrations of BDNF and cortisol seem to be dependent on the amount and duration of smoking. Therefore, we investigated BDNF and cortisol levels in smokers ranked by daily cigarette consumption. Twenty-seven adult males (13 non-smokers and 14 smokers) participated in the study. The smokers were divided in two groups: light (n=7) and heavy smokers (n=7). Anthropometric parameters and age were paired between the groups, and plasma BDNF and salivary cortisol levels were measured. Saliva samples were collected on awakening, 30 min after awakening, at 10:00 and 12:00 am, 5:00 and 10:00 pm. Additionally, cotinine serum levels were measured in smokers. Heavy smokers had higher mean values of BDNF compared to the control group (P=0.01), whereas no difference was observed in light smokers. Moreover, heavy smokers presented lower cortisol levels in the last collection (10:00 pm) than the control group (P=0.02) and presented statically higher values of cotinine than the light smokers (P=0.002). In conclusion, changes in BDNF and cortisol levels (10:00 pm) appear to be dependent on heavy cigarette smoking and can be involved in activation and in the relationship between the mesolimbic system and the HPA axis.
Collapse
Affiliation(s)
- C D C Neves
- Programa Multicêntrico de Pós-Graduação em Ciências Fisiológicas, Sociedade Brasileira de Fisiologia, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil.,Laboratório de Inflamação e Metabolismo, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil
| | - A C R Lacerda
- Programa Multicêntrico de Pós-Graduação em Ciências Fisiológicas, Sociedade Brasileira de Fisiologia, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil
| | - L P Lima
- Laboratório de Inflamação e Metabolismo, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil
| | - V K S Lage
- Programa Multicêntrico de Pós-Graduação em Ciências Fisiológicas, Sociedade Brasileira de Fisiologia, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil.,Laboratório de Inflamação e Metabolismo, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil
| | - C H Balthazar
- Programa Multicêntrico de Pós-Graduação em Ciências Fisiológicas, Sociedade Brasileira de Fisiologia, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil.,Laboratório de Inflamação e Metabolismo, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil
| | - H R Leite
- Programa Multicêntrico de Pós-Graduação em Ciências Fisiológicas, Sociedade Brasileira de Fisiologia, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil.,Laboratório de Inflamação e Metabolismo, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil
| | - V A Mendonça
- Programa Multicêntrico de Pós-Graduação em Ciências Fisiológicas, Sociedade Brasileira de Fisiologia, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil.,Laboratório de Inflamação e Metabolismo, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil
| |
Collapse
|
79
|
Mu XP, Wang HB, Cheng X, Yang L, Sun XY, Qu HL, Zhao SS, Zhou ZK, Liu TT, Xiao T, Song B, Jolkkonen J, Zhao CS. Inhibition of Nkcc1 promotes axonal growth and motor recovery in ischemic rats. Neuroscience 2017; 365:83-93. [PMID: 28964752 DOI: 10.1016/j.neuroscience.2017.09.036] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 09/03/2017] [Accepted: 09/21/2017] [Indexed: 12/15/2022]
Abstract
Bumetanide is a selective inhibitor of the Na+-K+-Cl--co-transporter 1(NKCC1). We studied whether bumetanide could affect axonal growth and behavioral outcome in stroke rats. Adult male Wistar rats were randomly assigned to four groups: sham-operated rats treated with vehicle or bumetanide, and ischemic rats treated with vehicle or bumetanide. Endothelin-1 was used to induce focal cerebral ischemia. Bumetanide administration (i.c.v.) started on postoperative day 7 and continued for 3 weeks. Biotinylated dextran amine (BDA) was injected into the right imotor cortex on postoperative day 14 to trace corticospinal tract (CST) fibers sprouting into the denervated cervical spinal cord. Nogo-A, NKCC1, KCC2 and BDNF in the perilesional cortex and BDA, PSD-95 and vGlut1 in the denervated spinal cord were measured by immunohistochemistry and/or Western blot. Behavioral outcome of rats was assessed by the beam walking and cylinder tests. The total length of CST fibers sprouting into the denervated cervical spinal cord significantly increased after stroke and bumetanide further increased this sprouting. Bumetanide treatment also decreased the expressions of NKCC1 and Nogo-A, increased the expressions of KCC2 and BDNF in the perilesional cortex and enhanced the synaptic plasticity in the denervated cervical spinal cord after cerebral ischemia. The behavioral performance of ischemic rats was significantly improved by bumetanide. In conclusion, bumetanide promoted post-stroke axonal sprouting together accompanied by an improved behavioral outcome possibly through restoring and maintaining neuronal chloride homeostasis and creating a recovery-promoting microenvironment by overcoming the axonal growth inhibition encountered after cerebral ischemia in rats.
Collapse
Affiliation(s)
- X P Mu
- Department of Neurology, The First Affiliated Hospital, China Medical University, Shenyang, China; Department of Neurology, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - H B Wang
- Department of Neurology, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - X Cheng
- Department of Neurology, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - L Yang
- Department of Cardiology, The Affiliated Center Hospital, Shenyang Medical College, Shenyang, China
| | - X Y Sun
- Department of Neurology, The People's Hospital of Liaoning Province, Shenyang, China
| | - H L Qu
- Department of Neurology, The People's Hospital of Liaoning Province, Shenyang, China
| | - S S Zhao
- Department of Neurology, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Z K Zhou
- Department of Geriatrics, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - T T Liu
- Department of Neurology, The People's Hospital of Liaoning Province, Shenyang, China
| | - T Xiao
- Department of Dermatology, The First Affiliated Hospital, China Medical University, Shenyang, China; Key Laboratory of Immunodermatology, Ministry of Health, Ministry of Education, Shenyang, China
| | - B Song
- Regenerative Medicine, Cardiff Institute of Tissue Engineering and Repair, School of Dentistry, Cardiff University, Cardiff, UK
| | - J Jolkkonen
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, P. O. Box 1627, 70211 Kuopio, Finland
| | - C S Zhao
- Department of Neurology, The First Affiliated Hospital, China Medical University, Shenyang, China.
| |
Collapse
|
80
|
AMPAkines and morphine provide complementary analgesia. Behav Brain Res 2017; 334:1-5. [PMID: 28734765 DOI: 10.1016/j.bbr.2017.07.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 07/14/2017] [Accepted: 07/17/2017] [Indexed: 11/21/2022]
Abstract
Glutamate signaling in the central nervous system is known to play a key role in pain regulation. AMPAkines can enhance glutamate signaling through α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. previous studies have shown that AMPAkines are effective analgesic agents, and their site of action is likely in the brain. It is not known, however, if AMPAkines can provide complementary analgesia in combination with opioids, the most commonly used analgesics. Here, we show that the co-administration of an AMPAkine with morphine can provide additional analgesia, both in naïve rats and in rats that experience postoperative pain. Furthermore, we show that this AMPAkine can be administered directly into the prefrontal cortex to provide analgesia, and that prefrontal AMPAkine infusion, similar to systemic administration, can provide added pain relief to complement morphine analgesia.
Collapse
|
81
|
Yue L, Ma LY, Cui S, Liu FY, Yi M, Wan Y. Brain-derived neurotrophic factor in the infralimbic cortex alleviates inflammatory pain. Neurosci Lett 2017. [PMID: 28648456 DOI: 10.1016/j.neulet.2017.06.028] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In chronic pain, it has been reported that the medial prefrontal cortex (mPFC) takes important regulatory roles, and may change functionally and morphologically in result of chronic pain. Brain-derived neurotrophic factor (BDNF) is well known as a critical modulator of neuronal excitability and synaptic transmission in the central nervous system. The aim of the present study is to investigate the role of BDNF in the infralimbic cortex and the prelimbic cortex of the mPFC in complete Freund's adjuvant (CFA)-induced inflammatory pain. We found that the BDNF level decreased in the infralimbic cortex, but not in the prelimbic cortex, 3days after the CFA induction of the inflammatory pain. BDNF infusion into bilateral infralimbic cortices to activate neuronal activities could alleviate inflammatory pain and accelerate long-term recovery from pain. In conclusion, BDNF in the infralimbic cortex of the mPFC could accelerate recovery from inflammatory pain.
Collapse
Affiliation(s)
- Lupeng Yue
- Neuroscience Research Institute, Peking University, Beijing 100191, P. R. China
| | - Long-Yu Ma
- Neuroscience Research Institute, Peking University, Beijing 100191, P. R. China
| | - Shuang Cui
- Neuroscience Research Institute, Peking University, Beijing 100191, P. R. China
| | - Feng-Yu Liu
- Neuroscience Research Institute, Peking University, Beijing 100191, P. R. China
| | - Ming Yi
- Neuroscience Research Institute, Peking University, Beijing 100191, P. R. China
| | - You Wan
- Neuroscience Research Institute, Peking University, Beijing 100191, P. R. China; Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, P. R. China; Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing 100191, P. R. China.
| |
Collapse
|
82
|
The Link between Depression and Chronic Pain: Neural Mechanisms in the Brain. Neural Plast 2017; 2017:9724371. [PMID: 28706741 PMCID: PMC5494581 DOI: 10.1155/2017/9724371] [Citation(s) in RCA: 388] [Impact Index Per Article: 55.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Revised: 05/04/2017] [Accepted: 05/24/2017] [Indexed: 02/08/2023] Open
Abstract
Chronic pain, as a stress state, is one of the critical factors for determining depression, and their coexistence tends to further aggravate the severity of both disorders. Unfortunately, their association remains unclear, which creates a bottleneck problem for managing chronic pain-induced depression. In recent years, studies have found considerable overlaps between pain- and depression-induced neuroplasticity changes and neurobiological mechanism changes. Such overlaps are vital to facilitating the occurrence and development of chronic pain and chronic pain-induced depression. In this review, we summarized the role of neuroplasticity in the occurrence and development of the two disorders in question and explored individualized application strategies of analgesic drugs and antidepressants that have different pharmacological effects in the treatment of chronic pain-induced depression. Therefore, this review may provide new insights into the understanding of association between chronic pain and depression.
Collapse
|
83
|
Targeting Neurotrophins to Specific Populations of Neurons: NGF, BDNF, and NT-3 and Their Relevance for Treatment of Spinal Cord Injury. Int J Mol Sci 2017; 18:ijms18030548. [PMID: 28273811 PMCID: PMC5372564 DOI: 10.3390/ijms18030548] [Citation(s) in RCA: 211] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/23/2017] [Accepted: 02/24/2017] [Indexed: 01/01/2023] Open
Abstract
Neurotrophins are a family of proteins that regulate neuronal survival, synaptic function, and neurotransmitter release, and elicit the plasticity and growth of axons within the adult central and peripheral nervous system. Since the 1950s, these factors have been extensively studied in traumatic injury models. Here we review several members of the classical family of neurotrophins, the receptors they bind to, and their contribution to axonal regeneration and sprouting of sensory and motor pathways after spinal cord injury (SCI). We focus on nerve growth factor (NGF), brain derived neurotrophic factor (BDNF), and neurotrophin-3 (NT-3), and their effects on populations of neurons within diverse spinal tracts. Understanding the cellular targets of neurotrophins and the responsiveness of specific neuronal populations will allow for the most efficient treatment strategies in the injured spinal cord.
Collapse
|
84
|
Huang WL, Ma YX, Fan YB, Lai SM, Liu HQ, Liu J, Luo L, Li GY, Tian SM. Extract of Ginkgo biloba promotes neuronal regeneration in the hippocampus after exposure to acrylamide. Neural Regen Res 2017; 12:1287-1293. [PMID: 28966643 PMCID: PMC5607823 DOI: 10.4103/1673-5374.213548] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Previous studies have demonstrated a neuroprotective effect of extract of Ginkgo biloba against neuronal damage, but have mainly focused on antioxidation of extract of Ginkgo biloba. To date, limited studies have determined whether extrasct of Ginkgo biloba has a protective effect on neuronal damage. In the present study, acrylamide and 30, 60, and 120 mg/kg extract of Ginkgo biloba were administered for 4 weeks by gavage to establish mouse models. Our results showed that 30, 60, and 120 mg/kg extract of Ginkgo biloba effectively alleviated the abnormal gait of poisoned mice, and up-regulated protein expression levels of doublecortin (DCX), brain-derived neurotrophic factor, and growth associated protein-43 (GAP-43) in the hippocampus. Simultaneously, DCX- and GAP-43-immunoreactive cells increased. These findings suggest that extract of Ginkgo biloba can mitigate neurotoxicity induced by acrylamide, and thereby promote neuronal regeneration in the hippocampus of acrylamide-treated mice.
Collapse
Affiliation(s)
- Wei-Ling Huang
- Department of Anatomy, School of Basic Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China
| | - Yu-Xin Ma
- Department of Anatomy, School of Basic Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China
| | - Yu-Bao Fan
- Department of Anatomy, School of Basic Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China
| | - Sheng-Min Lai
- Department of Anatomy, School of Basic Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China
| | - Hong-Qing Liu
- Department of Anatomy, School of Basic Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China
| | - Jing Liu
- Department of Anatomy, School of Basic Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China
| | - Li Luo
- Department of Anatomy, School of Basic Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China
| | - Guo-Ying Li
- Department of Anatomy, School of Basic Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China
| | - Su-Min Tian
- Department of Physiology, School of Basic Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China
| |
Collapse
|