1
|
Frara N, Jawawdeh K, Giaddui D, Tamas IP, Gares RP, McGonagle ER, Hilliard BA, Kolpakov MA, Bright-Rowe L, Braverman AS, Brown JM, Ruggieri MR, Barbe MF. Enhanced BDNF and ROS in Mucosa of Lower Motor Neuron Lesioned Dog Bladder Following Somatic Motor Nerve Transfer. Cells 2025; 14:406. [PMID: 40136655 PMCID: PMC11941061 DOI: 10.3390/cells14060406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/03/2025] [Accepted: 03/07/2025] [Indexed: 03/27/2025] Open
Abstract
Neurotrophic factors and reactive oxygen species (ROS) modulate neuronal plasticity. In a model of a lower motor neuron lesioned bladder, somatic nerve transfer was used as a reinnervation strategy. Levels of neurotrophins, ROS, and TNF-α in bladder mucosa and muscle layers collected from three groups of adult female dogs: (1) Decentralized, via bilateral transection of coccygeal and sacral spinal roots, lumbar 7 dorsal roots, and hypogastric nerves, then 6-21 mo recovery; (2) reinnervated (ObNT-Reinn), after similar decentralization for 12 mo, then bilateral obturator-to-vesical nerve transfer and 8-12 mo recovery; and (3) Controls. In mucosa, BDNF and ROS levels were highest in ObNT-Reinn bladders, GDNF and TNF-α levels were restored to Control levels in ObNT-Reinn bladders (lowest in Decentralized). NT-3 and ARTN were lower in ObNT-Reinn and Decentralized bladders versus Controls. In muscle, ROS was lower in ObNT-Reinn muscle versus Controls. BDNF mucosa levels correlated with bladder axonal density and detrusor layer thickness; and GDNF mucosal correlated with bladder contraction after vesical or transferred obturator nerve electrical stimulation, as did BDNF and GDNF muscle levels. The increased BDNF and GDNF in bladders that underwent somatic nerve transfer with subsequent recovery suggest that BDNF and GDNF may help promote the reestablishment of bladder innervation.
Collapse
Affiliation(s)
- Nagat Frara
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (I.P.T.); (E.R.M.); (B.A.H.); (M.A.K.); (L.B.-R.); (A.S.B.)
| | - Kais Jawawdeh
- Center for Translational Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19040, USA; (K.J.); (D.G.); (R.P.G.); (M.R.R.S.)
| | - Dania Giaddui
- Center for Translational Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19040, USA; (K.J.); (D.G.); (R.P.G.); (M.R.R.S.)
| | - Istvan P. Tamas
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (I.P.T.); (E.R.M.); (B.A.H.); (M.A.K.); (L.B.-R.); (A.S.B.)
| | - Ryan P. Gares
- Center for Translational Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19040, USA; (K.J.); (D.G.); (R.P.G.); (M.R.R.S.)
| | - Elizabeth R. McGonagle
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (I.P.T.); (E.R.M.); (B.A.H.); (M.A.K.); (L.B.-R.); (A.S.B.)
| | - Brendan A. Hilliard
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (I.P.T.); (E.R.M.); (B.A.H.); (M.A.K.); (L.B.-R.); (A.S.B.)
| | - Mikhail A. Kolpakov
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (I.P.T.); (E.R.M.); (B.A.H.); (M.A.K.); (L.B.-R.); (A.S.B.)
| | - Lewis Bright-Rowe
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (I.P.T.); (E.R.M.); (B.A.H.); (M.A.K.); (L.B.-R.); (A.S.B.)
| | - Alan S. Braverman
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (I.P.T.); (E.R.M.); (B.A.H.); (M.A.K.); (L.B.-R.); (A.S.B.)
| | - Justin M. Brown
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA;
| | - Michael R. Ruggieri
- Center for Translational Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19040, USA; (K.J.); (D.G.); (R.P.G.); (M.R.R.S.)
| | - Mary F. Barbe
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (I.P.T.); (E.R.M.); (B.A.H.); (M.A.K.); (L.B.-R.); (A.S.B.)
| |
Collapse
|
2
|
Issa S, Fayoud H, Shaimardanova A, Sufianov A, Sufianova G, Solovyeva V, Rizvanov A. Growth Factors and Their Application in the Therapy of Hereditary Neurodegenerative Diseases. Biomedicines 2024; 12:1906. [PMID: 39200370 PMCID: PMC11351319 DOI: 10.3390/biomedicines12081906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 08/11/2024] [Accepted: 08/14/2024] [Indexed: 09/02/2024] Open
Abstract
Hereditary neurodegenerative diseases (hNDDs) such as Alzheimer's, Parkinson's, Huntington's disease, and others are primarily characterized by their progressive nature, severely compromising both the cognitive and motor abilities of patients. The underlying genetic component in hNDDs contributes to disease risk, creating a complex genetic landscape. Considering the fact that growth factors play crucial roles in regulating cellular processes, such as proliferation, differentiation, and survival, they could have therapeutic potential for hNDDs, provided appropriate dosing and safe delivery approaches are ensured. This article presents a detailed overview of growth factors, and explores their therapeutic potential in treating hNDDs, emphasizing their roles in neuronal survival, growth, and synaptic plasticity. However, challenges such as proper dosing, delivery methods, and patient variability can hinder their clinical application.
Collapse
Affiliation(s)
- Shaza Issa
- Department of Genetics and Biotechnology, St. Petersburg State University, 199034 St. Petersburg, Russia; (S.I.); (H.F.)
| | - Haidar Fayoud
- Department of Genetics and Biotechnology, St. Petersburg State University, 199034 St. Petersburg, Russia; (S.I.); (H.F.)
| | - Alisa Shaimardanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (A.S.); (V.S.)
| | - Albert Sufianov
- Department of Neurosurgery, Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119991 Moscow, Russia;
- The Research and Educational Institute of Neurosurgery, Peoples’ Friendship University of Russia (RUDN), 117198 Moscow, Russia
| | - Galina Sufianova
- Department of Pharmacology, Tyumen State Medical University, 625023 Tyumen, Russia;
| | - Valeriya Solovyeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (A.S.); (V.S.)
| | - Albert Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (A.S.); (V.S.)
- Division of Medical and Biological Sciences, Tatarstan Academy of Sciences, 420111 Kazan, Russia
| |
Collapse
|
3
|
Kakoty V, Sarathlal KC, Kaur P, Wadhwa P, Vishwas S, Khan FR, Alhazmi AYM, Almasoudi HH, Gupta G, Chellappan DK, Paudel KR, Kumar D, Dua K, Singh SK. Unraveling the role of glial cell line-derived neurotrophic factor in the treatment of Parkinson's disease. Neurol Sci 2024; 45:1409-1418. [PMID: 38082050 DOI: 10.1007/s10072-023-07253-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 12/02/2023] [Indexed: 03/16/2024]
Abstract
Parkinson's disease is the second most common neurodegenerative condition with its prevalence projected to 8.9 million individuals globally in the year 2019. Parkinson's disease affects both motor and certain non-motor functions of an individual. Numerous research has focused on the neuroprotective effect of the glial cell line-derived neurotrophic factor (GDNF) in Parkinson's disease. Discovered in 1993, GDNF is a neurotrophic factor identified from the glial cells which was found to have selective effects on promoting survival and regeneration of certain populations of neurons including the dopaminergic nigrostriatal pathway. Given this property, recent studies have focused on the exogenous administration of GDNF for relieving Parkinson's disease-related symptoms both at a pre-clinical and a clinical level. This review will focus on enumerating the molecular connection between Parkinson's disease and GDNF and shed light on all the available drug delivery approaches to facilitate the selective delivery of GDNF into the brain paving the way as a potential therapeutic candidate for Parkinson's disease in the future.
Collapse
Affiliation(s)
- Violina Kakoty
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, Punjab, India
| | - K C Sarathlal
- Department of Non-Communicable Disease, Translational Health Science and Technology Institute, Faridabad, India
| | - Palwinder Kaur
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, Punjab, India
| | - Pankaj Wadhwa
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, Punjab, India
| | - Sukriti Vishwas
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, Punjab, India
| | - Farhan R Khan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Al-Quwayiyah, Shaqra University, Riyadh, Saudi Arabia
| | | | - Hassan Hussain Almasoudi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran, 61441, Saudi Arabia
| | - Gaurav Gupta
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- School of Pharmacy, Graphic Era Hill University, Dehradun, 248007, India
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | | | - Keshav Raj Paudel
- Centre for Inflammation, Faculty of Science, School of Life Sciences, Centenary Institute and University of Technology Sydney, Sydney, NSW, 2050, Australia
| | - Dileep Kumar
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Kamal Dua
- School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, Punjab, India.
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| |
Collapse
|
4
|
Hu HL, Khatri L, Santacruz M, Church E, Moore C, Huang TT, Chao MV. Confronting the loss of trophic support. Front Mol Neurosci 2023; 16:1179209. [PMID: 37456526 PMCID: PMC10338843 DOI: 10.3389/fnmol.2023.1179209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/23/2023] [Indexed: 07/18/2023] Open
Abstract
Classic experiments with peripheral sympathetic neurons established an absolute dependence upon NGF for survival. A forgotten problem is how these neurons become resistant to deprivation of trophic factors. The question is whether and how neurons can survive in the absence of trophic support. However, the mechanism is not understood how neurons switch their phenotype to lose their dependence on trophic factors, such as NGF and BDNF. Here, we approach the problem by considering the requirements for trophic support of peripheral sympathetic neurons and hippocampal neurons from the central nervous system. We developed cellular assays to assess trophic factor dependency for sympathetic and hippocampal neurons and identified factors that rescue neurons in the absence of trophic support. They include enhanced expression of a subunit of the NGF receptor (Neurotrophin Receptor Homolog, NRH) in sympathetic neurons and an increase of the expression of the glucocorticoid receptor in hippocampal neurons. The results are significant since levels and activity of trophic factors are responsible for many neuropsychiatric conditions. Resistance of neurons to trophic factor deprivation may be relevant to the underlying basis of longevity, as well as an important element in preventing neurodegeneration.
Collapse
Affiliation(s)
- Hui-Lan Hu
- Department of Biochemistry and Molecular Pharmacology, New York University Langone School of Medicine, New York, NY, United States
| | - Latika Khatri
- Skirball Institute for Biomolecular Medicine, Neuroscience Institute, New York University Langone Medical Center, New York, NY, United States
| | - Marilyn Santacruz
- Department of Neuroscience, Pomona College, Claremont, CA, United States
| | - Emily Church
- Department of Neuroscience, Pomona College, Claremont, CA, United States
| | - Christopher Moore
- Skirball Institute for Biomolecular Medicine, Neuroscience Institute, New York University Langone Medical Center, New York, NY, United States
| | - Tony T. Huang
- Department of Biochemistry and Molecular Pharmacology, New York University Langone School of Medicine, New York, NY, United States
| | - Moses V. Chao
- Skirball Institute for Biomolecular Medicine, Neuroscience Institute, New York University Langone Medical Center, New York, NY, United States
- Department of Cell Biology, New York Langone Medical Center, New York, NY, United States
- Department of Psychiatry, New York Langone Medical Center, New York, NY, United States
- Department of Neuroscience and Physiology, New York Langone Medical Center, New York, NY, United States
| |
Collapse
|
5
|
Wong FSY, Tsang KK, Chan BP, Lo ACY. Both non-coated and polyelectrolytically-coated intraocular collagen-alginate composite gels enhanced photoreceptor survival in retinal degeneration. Biomaterials 2023; 293:121948. [PMID: 36516686 DOI: 10.1016/j.biomaterials.2022.121948] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 11/09/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022]
Abstract
Treatments of vision-threatening retinal diseases are often hampered by drug delivery difficulties. Polyelectrolytically-coated alginate encapsulated-cell therapy (ECT) systems have shown therapeutic efficacy through prolonged in vivo drug delivery but still face various biocompatibility, viability, drug delivery and mechanical stability issues in clinical trials. Here, novel, injectable alginate-poly-l-lysine (AP)-coated composite alginate-collagen (CAC) ECT gels were developed for sustained ocular drug delivery, and their long-term performance was compared with non-coated CAC ECT gels. All optimised AP-coated gels (AP1- and AP5.5-CAC ECT: 2 mg/ml collagen, 1.5% high molecular weight alginate, 50,000 cells/gel, with 0.01% or 0.05% poly-l-lysine coating for 5 min, followed by 0.15% alginate coating) and non-coated gels showed effective cell proliferation control, cell viability support and continuous delivery of bioactive glial cell-derived neurotrophic factor (GDNF) with no significant gel degradation in vitro and in rat vitreous. Most importantly, intravitreally injected gels demonstrated therapeutic efficacy in Royal College of Surgeons rats with retinal degeneration, resulting in reduced photoreceptor apoptosis and retinal function loss. At 6 months post-implantation, no host-tissue attachment or ingrowth was detected on the retrieved gels. Non-coated gels were mechanically more stable than AP5.5-coated ones under the current cell loading. This study demonstrated that both coated and non-coated ECT gels can serve as well-controlled, sustained drug delivery platforms for treating posterior eye diseases without immunosuppression.
Collapse
Affiliation(s)
- Francisca Siu Yin Wong
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Ken Kin Tsang
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Barbara Pui Chan
- Tissue Engineering Laboratory, Department of Mechanical Engineering, Faculty of Engineering, The University of Hong Kong, Hong Kong, China
| | - Amy Cheuk Yin Lo
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
6
|
Satish L, Santra S, Tsurkan MV, Werner C, Jana M, Sahoo H. Conformational changes of GDNF-derived peptide induced by heparin, heparan sulfate, and sulfated hyaluronic acid - Analysis by circular dichroism spectroscopy and molecular dynamics simulation. Int J Biol Macromol 2021; 182:2144-2150. [PMID: 34087306 DOI: 10.1016/j.ijbiomac.2021.05.194] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/21/2021] [Accepted: 05/28/2021] [Indexed: 01/15/2023]
Abstract
Glial-cell-line-derived neurotrophic factor (GDNF) is a protein that has therapeutic potential in the treatment of Parkinson's disease and other neurodegenerative diseases. The activity of GDNF is highly dependent on the interaction with sulfated glycans which bind at the N-terminus consisting of 19 residues. Herein, we studied the influence of different glycosaminoglycan (i.e., glycan; GAG) molecules on the conformation of a GDNF-derived peptide (GAG binding motif, sixteen amino acid residues at the N-terminus) using both experimental and theoretical studies. The GAG molecules employed in this study are heparin, heparan sulfate, hyaluronic acid, and sulfated hyaluronic acid. Circular dichroism spectroscopy was employed to detect conformational changes induced by the GAG molecules; molecular dynamics simulation studies were performed to support the experimental results. Our results revealed that the sulfated GAG molecules bind strongly with GDNF peptide and induce alpha-helical structure in the peptide to some extent.
Collapse
Affiliation(s)
- Lakkoji Satish
- Biophysical and Protein Chemistry Laboratory, Department of Chemistry, National Institute of Technology Rourkela, Odisha 769008, India; School of Chemical Sciences, National Institute of Science Education and Research, HBNI, Bhubaneswar, Odisha 752050, India
| | - Santanu Santra
- Molecular Simulation Laboratory, Department of Chemistry, National Institute of Technology Rourkela, Odisha 769008, India
| | - Mikhail V Tsurkan
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials, 01069 Dresden, Germany; Center for Regenerative Therapies Dresden, Technische Universität Dresden, 01307 Dresden, Germany
| | - Carsten Werner
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials, 01069 Dresden, Germany; Center for Regenerative Therapies Dresden, Technische Universität Dresden, 01307 Dresden, Germany
| | - Madhurima Jana
- Molecular Simulation Laboratory, Department of Chemistry, National Institute of Technology Rourkela, Odisha 769008, India
| | - Harekrushna Sahoo
- Biophysical and Protein Chemistry Laboratory, Department of Chemistry, National Institute of Technology Rourkela, Odisha 769008, India; Center for Nanomaterials, National Institute of Technology Rourkela, Odisha 769008, India.
| |
Collapse
|
7
|
Jeong JE, Jeon S, Han JS, Cho EY, Hong KS, Park SN, Kim JJ. The Mediating Effect of Psychological Distress on the Association between BDNF, 5-HTTLPR, and Tinnitus Severity. Psychiatry Investig 2021; 18:187-195. [PMID: 33685039 PMCID: PMC8016684 DOI: 10.30773/pi.2020.0295] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 11/13/2020] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE To investigate the association between genetic polymorphisms of brain-derived neurotrophic factor (BDNF) or serotonin transporter gene-linked polymorphic region (5-HTTLPR) and tinnitus, and the mediating effects of psychological distress on this association. METHODS Eighty-six patients experiencing tinnitus and 252 controls were recruited. The Tinnitus Handicap Inventory was used to assess the severity of tinnitus and the Beck Depression Inventory-II (BDI-II), Beck Anxiety Inventory-II (BAI-II), and the Korean version of the Brief Encounter Psychosocial Instrument (BEPSI-K) were used to assess psychological distress. We compared the association of BDNF rs6265 (Val66Met) and 5-HTTLPR variants in the two groups. The mediating effects of BDI-II, BAI-II, and BEPSI-K were examined using multiple regression analysis and validated by the Sobel test and bootstrapping. RESULTS No significant differences were found between the groups regarding BDNF Val66Met and 5-HTTLPR, but the 5-HTTLPR variants trended toward association. Depressive symptoms appeared to act as a mediator on the relationship within the 5-HTTLPR s/s genotype and the severity of tinnitus. CONCLUSION Our findings provide a speculative idea on the association between the serotonergic system and tinnitus and suggest that depressive symptoms act as a mediator in tinnitus. Therefore, screening for depressive symptoms in patients with tinnitus is essential and intervention for depressive symptoms may help alleviate the severity of tinnitus.
Collapse
Affiliation(s)
- Jo-Eun Jeong
- Department of Psychiatry, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sekye Jeon
- Department of Psychiatry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jae Sang Han
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eun Young Cho
- Center of Clinical Research, Samsung Biomedical Research Institute, Seoul, Republic of Korea
| | - Kyung Sue Hong
- Department of Psychiatry, Sungkyunkwan University School of Medicine, Samsung Seoul Hospital, Seoul, Republic of Korea
| | - Shi Nae Park
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jung Jin Kim
- Department of Psychiatry, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
8
|
Ferrini F, Salio C, Boggio EM, Merighi A. Interplay of BDNF and GDNF in the Mature Spinal Somatosensory System and Its Potential Therapeutic Relevance. Curr Neuropharmacol 2021; 19:1225-1245. [PMID: 33200712 PMCID: PMC8719296 DOI: 10.2174/1570159x18666201116143422] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 09/17/2020] [Accepted: 10/05/2020] [Indexed: 11/22/2022] Open
Abstract
The growth factors BDNF and GDNF are gaining more and more attention as modulators of synaptic transmission in the mature central nervous system (CNS). The two molecules undergo a regulated secretion in neurons and may be anterogradely transported to terminals where they can positively or negatively modulate fast synaptic transmission. There is today a wide consensus on the role of BDNF as a pro-nociceptive modulator, as the neurotrophin has an important part in the initiation and maintenance of inflammatory, chronic, and/or neuropathic pain at the peripheral and central level. At the spinal level, BDNF intervenes in the regulation of chloride equilibrium potential, decreases the excitatory synaptic drive to inhibitory neurons, with complex changes in GABAergic/glycinergic synaptic transmission, and increases excitatory transmission in the superficial dorsal horn. Differently from BDNF, the role of GDNF still remains to be unraveled in full. This review resumes the current literature on the interplay between BDNF and GDNF in the regulation of nociceptive neurotransmission in the superficial dorsal horn of the spinal cord. We will first discuss the circuitries involved in such a regulation, as well as the reciprocal interactions between the two factors in nociceptive pathways. The development of small molecules specifically targeting BDNF, GDNF and/or downstream effectors is opening new perspectives for investigating these neurotrophic factors as modulators of nociceptive transmission and chronic pain. Therefore, we will finally consider the molecules of (potential) pharmacological relevance for tackling normal and pathological pain.
Collapse
Affiliation(s)
- Francesco Ferrini
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
- Department of Psychiatry & Neuroscience, Université Laval, Québec, Canada
| | - Chiara Salio
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
| | - Elena M. Boggio
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
| | - Adalberto Merighi
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
- National Institute of Neuroscience, Grugliasco, Italy
| |
Collapse
|
9
|
von Loga IS, Batchelor V, Driscoll C, Burleigh A, Chia SLL, Stott B, Miotla-Zarebska J, Riley D, Dell'Accio F, Vincent TL. Does Pain at an Earlier Stage of Chondropathy Protect Female Mice Against Structural Progression After Surgically Induced Osteoarthritis? Arthritis Rheumatol 2020; 72:2083-2093. [PMID: 32602242 DOI: 10.1002/art.41421] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 06/11/2020] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Female C57BL/6 mice exhibit less severe chondropathy than male mice. This study was undertaken to test the robustness of this observation and explore underlying mechanisms. METHODS Osteoarthritis was induced in male and female C57BL/6 or DBA/1 mice (n = 6-15 per group) by destabilization of the medial meniscus (DMM) or partial meniscectomy (PMX). Some mice were ovariectomized (OVX) (n = 30). In vivo repair after focal cartilage defect or joint immobilization (sciatic neurectomy) following DMM was assessed. Histologic analysis, evaluation of gene expression in whole knees, and behavioral analysis using Laboratory Animal Behavior Observation Registration and Analysis System (LABORAS) and Linton incapacitance testing (n = 7-10 mice per group) were performed. RESULTS Female mice displayed less severe chondropathy (20-75% reduction) across both strains and after both surgeries. Activity levels after PMX were similar for male and female mice. Some repair-associated genes were increased in female mouse joints after surgery, but no repair differences were evident in vivo. Despite reduced chondropathy, female mice developed pain-like behavior at the same time as male mice. At the time of established pain-like behavior (10 weeks after PMX), pain-associated genes were significantly up-regulated in female mice, including Gdnf (mean ± SEM fold change 2.54 ± 0.30), Nrtn (6.71 ± 1.24), Ntf3 (1.92 ± 0.27), and Ntf5 (2.89 ± 0.48) (P < 0.01, P < 0.01, P < 0.05, and P < 0.001, respectively, versus male mice). Inflammatory genes were not regulated in painful joints in mice of either sex. CONCLUSION We confirm strong structural joint protection in female mice that is not due to activity or intrinsic repair differences. Female mice develop pain at the same time as males, but induce a distinct set of neurotrophins. We speculate that heightened pain sensitivity in female mice protects the joint by preventing overuse.
Collapse
Affiliation(s)
| | - Vicky Batchelor
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Clare Driscoll
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Annika Burleigh
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Shi-Lu L Chia
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Bryony Stott
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | | | - David Riley
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | | | - Tonia L Vincent
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| |
Collapse
|
10
|
Habib AM, Nagi K, Thillaiappan NB, Sukumaran V, Akhtar S. Vitamin D and Its Potential Interplay With Pain Signaling Pathways. Front Immunol 2020; 11:820. [PMID: 32547536 PMCID: PMC7270292 DOI: 10.3389/fimmu.2020.00820] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 04/09/2020] [Indexed: 12/12/2022] Open
Abstract
About 50 million of the U.S. adult population suffer from chronic pain. It is a complex disease in its own right for which currently available analgesics have been deemed woefully inadequate since ~20% of the sufferers derive no benefit. Vitamin D, known for its role in calcium homeostasis and bone metabolism, is thought to be of clinical benefit in treating chronic pain without the side-effects of currently available analgesics. A strong correlation between hypovitaminosis D and incidence of bone pain is known. However, the potential underlying mechanisms by which vitamin D might exert its analgesic effects are poorly understood. In this review, we discuss pathways involved in pain sensing and processing primarily at the level of dorsal root ganglion (DRG) neurons and the potential interplay between vitamin D, its receptor (VDR) and known specific pain signaling pathways including nerve growth factor (NGF), glial-derived neurotrophic factor (GDNF), epidermal growth factor receptor (EGFR), and opioid receptors. We also discuss how vitamin D/VDR might influence immune cells and pain sensitization as well as review the increasingly important topic of vitamin D toxicity. Further in vitro and in vivo experimental studies will be required to study these potential interactions specifically in pain models. Such studies could highlight the potential usefulness of vitamin D either alone or in combination with existing analgesics to better treat chronic pain.
Collapse
Affiliation(s)
| | | | | | | | - Saghir Akhtar
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
11
|
Three-Dimensional Model of Dorsal Root Ganglion Explant as a Method of Studying Neurotrophic Factors in Regenerative Medicine. Biomedicines 2020; 8:biomedicines8030049. [PMID: 32138155 PMCID: PMC7175199 DOI: 10.3390/biomedicines8030049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 02/21/2020] [Accepted: 02/28/2020] [Indexed: 12/12/2022] Open
Abstract
Neurotrophic factors play a key role in the development, differentiation, and survival of neurons and nerve regeneration. In the present study, we evaluated the effect of certain neurotrophic factors (NGF, BDNF, and GDNF) on axon growth and migration of Nestin-green fluorescent protein (GFP)-positive cells using a 3D model of dorsal root ganglion (DRG) explant culture in Matrigel. Our method generally represents a convenient model for assessing the effects of soluble factors and therapeutic agents on axon growth and nerve regeneration in R&D studies. By analyzing the DRG explants in ex vivo culture for 21 days, one can evaluate the parameters of neurite outgrowth and the rate of cell migration from the DRG explants into the Matrigel. For the current study, we used Nestin-GFP-expressing mice in which neural precursors express Nestin and the green fluorescent protein (GFP) under the same promoter. We revealed that GDNF significantly (two fold) stimulated axon outgrowth (p < 0.05), but not BDNF or NGF. It is well-known that axon growth can be stimulated by activated glial cells that fulfill a trophic function for regenerating nerves. For this reason, we evaluated the number of Nestin-GFP-positive cells that migrated from the DRG into the Matrigel in our 3D ex vivo explant model. We found that NGF and GDNF, but not BDNF, stimulated the migration of Nestin-GFP cells compared to the control (p < 0.05). On the basis of the aforementioned finding, we concluded that GDNF had the greatest stimulating potential for axon regeneration, as it stimulated not only the axon outgrowth, but also glial cell migration. Although NGF significantly stimulated glial cell migration, its effect on axon growth was insufficient for axon regeneration.
Collapse
|
12
|
Azma T, Nishioka A, Ogawa S, Nagasaka H, Matsumoto N. Enhanced expression of gene coding for β-endorphin in human monocytic cells exposed to pulsed radio frequency electric fields through thermal and non-thermal effects. J Pain Res 2018; 11:2887-2896. [PMID: 30532582 PMCID: PMC6247966 DOI: 10.2147/jpr.s171974] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Background The enhanced expression of endogenous opioid peptides, including β-endorphin, has been implicated in the mechanism of action of pulsed radio frequency (PRF) application in pain modulation. Because thermal effects cannot be separated from the physical property of PRF application to biological tissues, we evaluated whether temperatures higher than that of the normal body temperature (37°C) modulate mRNA expression for the precursor of β-endorphin, proopiomelanocortin (POMC) in human monocytic cells THP-1. We also attempted to examine whether mechanisms other than thermal effects also modulate such gene expression. Methods and results The mRNA for POMC in THP-1 cells increased by a 15-minutes incubation at 42°C, 45°C, or 70°C without PRF application as compared with that in cells incubated at 37°C. On the other hand, gene expression for POMC in cells incubated at 20°C as well as at 37°C with PRF application for 15 minutes increased as compared to that in cells incubated at 37°C without PRF application. Continuous radio frequency at 70°C but not PRF provoked apoptotic cell death at 1–2 hour, and necrotic cell death at 24 hours after the RF application. Conclusion A simple experimental system using human monocytic cells in culture demonstrated that a 15 minute elevation of temperature above 37°C enhanced gene expression for POMC in THP-1 cells, while a 15 minute application of PRF to these cells incubated at 37°C or lower, also enhanced gene expression, indicating that temperature-independent mechanisms as well as thermal effects may be involved in such gene expression.
Collapse
Affiliation(s)
- Toshiharu Azma
- Department of Anesthesiology and Pain Medicine, Kohnodai Hospital, National Center for Global Health and Medicine, Ichikawa, Chiba 272-8516, Japan, .,Department of Anesthesiology, Saitama Medical University Hospital, Moroyama-cho, Iruma-gun, Saitama 350-0495, Japan,
| | - Akira Nishioka
- Department of Anesthesiology and Pain Medicine, Kohnodai Hospital, National Center for Global Health and Medicine, Ichikawa, Chiba 272-8516, Japan,
| | - Saori Ogawa
- Department of Dental Anesthesiology, Matsumoto Dental University, Shiojiri, Nagano 399-0781, Japan
| | - Hiroshi Nagasaka
- Department of Anesthesiology, Saitama Medical University Hospital, Moroyama-cho, Iruma-gun, Saitama 350-0495, Japan,
| | - Nobuyuki Matsumoto
- Department of Anesthesiology, Saitama Medical University Hospital, Moroyama-cho, Iruma-gun, Saitama 350-0495, Japan,
| |
Collapse
|
13
|
Akil O, Blits B, Lustig LR, Leake PA. Virally Mediated Overexpression of Glial-Derived Neurotrophic Factor Elicits Age- and Dose-Dependent Neuronal Toxicity and Hearing Loss. Hum Gene Ther 2018; 30:88-105. [PMID: 30183384 DOI: 10.1089/hum.2018.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Contemporary cochlear implants (CI) are generally very effective for remediation of severe to profound sensorineural hearing loss, but outcomes are still highly variable. Auditory nerve survival is likely one of the major factors underlying this variability. Neurotrophin therapy therefore has been proposed for CI recipients, with the goal of improving outcomes by promoting improved survival of cochlear spiral ganglion neurons (SGN) and/or residual hair cells. Previous studies have shown that glial-derived neurotrophic factor (GDNF), brain-derived neurotrophic factor, and neurotrophin-3 can rescue SGNs following insult. The current study was designed to determine whether adeno-associated virus vector serotype 5 (AAV-5) encoding either green fluorescent protein or GDNF can transduce cells in the mouse cochlea to express useful levels of neurotrophin and to approximate the optimum therapeutic dose(s) for transducing hair cells and SGN. The findings demonstrate that AAV-5 is a potentially useful gene therapy vector for the cochlea, resulting in extremely high levels of transgene expression in the cochlear inner hair cells and SGN. However, overexpression of human GDNF in newborn mice caused severe neurological symptoms and hearing loss, likely due to Purkinje cell loss and cochlear nucleus pathology. Thus, extremely high levels of transgene protein expression should be avoided, particularly for proteins that have neurological function in neonatal subjects.
Collapse
Affiliation(s)
- Omar Akil
- 1 Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, San Francisco, California
| | - Bas Blits
- 2 Department of Research and Development, UniQure Biopharma B.V., Amsterdam, The Netherlands
| | - Lawrence R Lustig
- 3 Department of Otolaryngology-Head and Neck Surgery, Columbia University Medical Center, New York, New York
| | - Patricia A Leake
- 1 Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, San Francisco, California
| |
Collapse
|
14
|
|
15
|
De Cicco V, Tramonti Fantozzi MP, Cataldo E, Barresi M, Bruschini L, Faraguna U, Manzoni D. Trigeminal, Visceral and Vestibular Inputs May Improve Cognitive Functions by Acting through the Locus Coeruleus and the Ascending Reticular Activating System: A New Hypothesis. Front Neuroanat 2018; 11:130. [PMID: 29358907 PMCID: PMC5766640 DOI: 10.3389/fnana.2017.00130] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 12/15/2017] [Indexed: 12/25/2022] Open
Abstract
It is known that sensory signals sustain the background discharge of the ascending reticular activating system (ARAS) which includes the noradrenergic locus coeruleus (LC) neurons and controls the level of attention and alertness. Moreover, LC neurons influence brain metabolic activity, gene expression and brain inflammatory processes. As a consequence of the sensory control of ARAS/LC, stimulation of a sensory channel may potential influence neuronal activity and trophic state all over the brain, supporting cognitive functions and exerting a neuroprotective action. On the other hand, an imbalance of the same input on the two sides may lead to an asymmetric hemispheric excitability, leading to an impairment in cognitive functions. Among the inputs that may drive LC neurons and ARAS, those arising from the trigeminal region, from visceral organs and, possibly, from the vestibular system seem to be particularly relevant in regulating their activity. The trigeminal, visceral and vestibular control of ARAS/LC activity may explain why these input signals: (1) affect sensorimotor and cognitive functions which are not directly related to their specific informational content; and (2) are effective in relieving the symptoms of some brain pathologies, thus prompting peripheral activation of these input systems as a complementary approach for the treatment of cognitive impairments and neurodegenerative disorders.
Collapse
Affiliation(s)
- Vincenzo De Cicco
- Laboratory of Sensorimotor Integration, Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | - Maria P Tramonti Fantozzi
- Laboratory of Sensorimotor Integration, Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | | | - Massimo Barresi
- Institut des Maladie Neurodégénératives, University of Bordeaux, Bordeaux, France
| | - Luca Bruschini
- Department of Surgical, Medical, Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Ugo Faraguna
- Laboratory of Sensorimotor Integration, Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy.,Department of Developmental Neuroscience, IRCCS Fondazione Stella Maris, Pisa, Italy
| | - Diego Manzoni
- Laboratory of Sensorimotor Integration, Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| |
Collapse
|
16
|
Ota H, Katanosaka K, Murase S, Furuyashiki T, Narumiya S, Mizumura K. EP2 receptor plays pivotal roles in generating mechanical hyperalgesia after lengthening contractions. Scand J Med Sci Sports 2017; 28:826-833. [DOI: 10.1111/sms.12954] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2017] [Indexed: 11/26/2022]
Affiliation(s)
- H. Ota
- Department of Neuroscience II; Graduate School of Medicine; Nagoya University; Nagoya Japan
- Department of Judo Therapy; Faculty of Medical Technology; Teikyo University; Utsunomiya Japan
- Department of Physical Therapy; College of Life and Health Sciences; Chubu University; Kasugai Japan
| | - K. Katanosaka
- Department of Neuroscience II; Graduate School of Medicine; Nagoya University; Nagoya Japan
- Department of Biomedical Sciences; College of Life and Health Sciences; Chubu University; Kasugai Japan
| | - S. Murase
- Department of Neuroscience II; Graduate School of Medicine; Nagoya University; Nagoya Japan
- Department of Physical Therapy; College of Life and Health Sciences; Chubu University; Kasugai Japan
| | - T. Furuyashiki
- Department of Pharmacology; Graduate School of Medicine; Kyoto University; Kyoto Japan
| | - S. Narumiya
- Department of Pharmacology; Graduate School of Medicine; Kyoto University; Kyoto Japan
| | - K. Mizumura
- Department of Neuroscience II; Graduate School of Medicine; Nagoya University; Nagoya Japan
- Department of Physical Therapy; College of Life and Health Sciences; Chubu University; Kasugai Japan
| |
Collapse
|
17
|
Abstract
Neural stem cells (NSCs) have been proposed as a promising cellular source for the treatment of diseases in nervous systems. NSCs can self-renew and generate major cell types of the mammalian central nervous system throughout adulthood. NSCs exist not only in the embryo, but also in the adult brain neurogenic region: the subventricular zone (SVZ) of the lateral ventricle. Embryonic stem (ES) cells acquire NSC identity with a default mechanism. Under the regulations of leukemia inhibitory factor (LIF) and fibroblast growth factors, the NSCs then become neural progenitors. Neurotrophic and differentiation factors that regulate gene expression for controlling neural cell fate and function determine the differentiation of neural progenitors in the developing mammalian brain. For clinical application of NSCs in neurodegenerative disorders and damaged neurons, there are several critical problems that remain to be resolved: 1) how to obtain enough NSCs from reliable sources for autologous transplantation; 2) how to regulate neural plasticity of different adult stem cells; 3) how to control differentiation of NSCs in the adult nervous system. In order to understand the mechanisms that control NSC differentiation and behavior, we review the ontogeny of NSCs and other stem cell plasticity of neuronal differentiation. The role of NSCs and their regulation by neurotrophic factors in CNS development are also reviewed.
Collapse
Affiliation(s)
- Yi-Chao Hsu
- Stem Cell Research Center, National Health Research Institutes, Jhunan, Taiwan
| | - Don-Ching Lee
- Stem Cell Research Center, National Health Research Institutes, Jhunan, Taiwan
| | - Ing-Ming Chiu
- Stem Cell Research Center, National Health Research Institutes, Jhunan, Taiwan
- Department of Internal Medicine, Ohio State University, Columbus, OH 43210, USA
- Institute of Medical Technology, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
18
|
Plassais J, Lagoutte L, Correard S, Paradis M, Guaguère E, Hédan B, Pommier A, Botherel N, Cadiergues MC, Pilorge P, Silversides D, Bizot M, Samuels M, Arnan C, Johnson R, Hitte C, Salbert G, Méreau A, Quignon P, Derrien T, André C. A Point Mutation in a lincRNA Upstream of GDNF Is Associated to a Canine Insensitivity to Pain: A Spontaneous Model for Human Sensory Neuropathies. PLoS Genet 2016; 12:e1006482. [PMID: 28033318 PMCID: PMC5198995 DOI: 10.1371/journal.pgen.1006482] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 11/15/2016] [Indexed: 01/06/2023] Open
Abstract
Human Hereditary Sensory Autonomic Neuropathies (HSANs) are characterized by insensitivity to pain, sometimes combined with self-mutilation. Strikingly, several sporting dog breeds are particularly affected by such neuropathies. Clinical signs appear in young puppies and consist of acral analgesia, with or without sudden intense licking, biting and severe self-mutilation of the feet, whereas proprioception, motor abilities and spinal reflexes remain intact. Through a Genome Wide Association Study (GWAS) with 24 affected and 30 unaffected sporting dogs using the Canine HD 170K SNP array (Illumina), we identified a 1.8 Mb homozygous locus on canine chromosome 4 (adj. p-val = 2.5x10-6). Targeted high-throughput sequencing of this locus in 4 affected and 4 unaffected dogs identified 478 variants. Only one variant perfectly segregated with the expected recessive inheritance in 300 sporting dogs of known clinical status, while it was never present in 900 unaffected dogs from 130 other breeds. This variant, located 90 kb upstream of the GDNF gene, a highly relevant neurotrophic factor candidate gene, lies in a long intergenic non-coding RNAs (lincRNA), GDNF-AS. Using human comparative genomic analysis, we observed that the canine variant maps onto an enhancer element. Quantitative RT-PCR of dorsal root ganglia RNAs of affected dogs showed a significant decrease of both GDNF mRNA and GDNF-AS expression levels (respectively 60% and 80%), as compared to unaffected dogs. We thus performed gel shift assays (EMSA) that reveal that the canine variant significantly alters the binding of regulatory elements. Altogether, these results allowed the identification in dogs of GDNF as a relevant candidate for human HSAN and insensitivity to pain, but also shed light on the regulation of GDNF transcription. Finally, such results allow proposing these sporting dog breeds as natural models for clinical trials with a double benefit for human and veterinary medicine. In this study, we present a canine neuropathy characterized by insensitivity to pain in the feet, sometimes combined with self-mutilation described in four sporting breeds. This particular phenotype has the clinical hallmarks of human Hereditary Sensory Autonomic Neuropathies (HSAN). As we hypothesized that a monogenic recessive disorder was shared between these breeds, we performed a Genome Wide Association Study (GWAS) to search for the genetic causes and found one homozygous chromosomal region in affected dogs. High-throughput sequencing of this region allowed the identification of a point mutation upstream to the GDNF gene and located in the last exon of a long non-coding RNA, GDNF-AS. We confirmed the perfect association of this variant with the disease using more than 900 unaffected dogs that do not present with this mutation. Functional analyses (qRT-PCR, EMSA) confirmed that the mutation alters the binding of regulatory complex, leading to a significant decrease of both GDNF and GDNF-AS mRNA expression levels. This work in canine spontaneous forms of human neuropathies allowed the identification of a novel gene GDNF and its regulation mechanism, not yet described in human HSAN, opening the field of clinical trials to benefit both canine and human medicine.
Collapse
Affiliation(s)
- Jocelyn Plassais
- CNRS, UMR 6290, Institut de Génétique et Développement de Rennes, Rennes, France
- Université Rennes 1, UEB, Biosit, Faculté de Médecine, Rennes, France
- * E-mail: (CA); (JP)
| | - Laetitia Lagoutte
- CNRS, UMR 6290, Institut de Génétique et Développement de Rennes, Rennes, France
- Université Rennes 1, UEB, Biosit, Faculté de Médecine, Rennes, France
| | - Solenne Correard
- CNRS, UMR 6290, Institut de Génétique et Développement de Rennes, Rennes, France
- Université Rennes 1, UEB, Biosit, Faculté de Médecine, Rennes, France
| | - Manon Paradis
- Department of Clinical Sciences, Faculté de Médecine Vétérinaire, University of Montreal, Montreal, Québec, Canada
| | | | - Benoit Hédan
- CNRS, UMR 6290, Institut de Génétique et Développement de Rennes, Rennes, France
- Université Rennes 1, UEB, Biosit, Faculté de Médecine, Rennes, France
| | - Alix Pommier
- CNRS, UMR 6290, Institut de Génétique et Développement de Rennes, Rennes, France
- Université Rennes 1, UEB, Biosit, Faculté de Médecine, Rennes, France
| | - Nadine Botherel
- CNRS, UMR 6290, Institut de Génétique et Développement de Rennes, Rennes, France
- Université Rennes 1, UEB, Biosit, Faculté de Médecine, Rennes, France
| | | | | | - David Silversides
- Department of Clinical Sciences, Faculté de Médecine Vétérinaire, University of Montreal, Montreal, Québec, Canada
| | - Maud Bizot
- CNRS, UMR 6290, Institut de Génétique et Développement de Rennes, Rennes, France
- Université Rennes 1, UEB, Biosit, Faculté de Médecine, Rennes, France
| | - Mark Samuels
- Department of Biochemistry and Molecular Medicine, CHU Sainte-Justine, University of Montreal, Montreal, Québec, Canada
| | - Carme Arnan
- Centre for Genomic Regulation (CRG), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Institut Hospital del Mar d’Investigations Mèdiques (IMIM), Barcelona, Spain
| | - Rory Johnson
- Centre for Genomic Regulation (CRG), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Institut Hospital del Mar d’Investigations Mèdiques (IMIM), Barcelona, Spain
| | - Christophe Hitte
- CNRS, UMR 6290, Institut de Génétique et Développement de Rennes, Rennes, France
- Université Rennes 1, UEB, Biosit, Faculté de Médecine, Rennes, France
| | - Gilles Salbert
- CNRS, UMR 6290, Institut de Génétique et Développement de Rennes, Rennes, France
- Université Rennes 1, UEB, Biosit, Faculté de Médecine, Rennes, France
| | - Agnès Méreau
- CNRS, UMR 6290, Institut de Génétique et Développement de Rennes, Rennes, France
- Université Rennes 1, UEB, Biosit, Faculté de Médecine, Rennes, France
| | - Pascale Quignon
- CNRS, UMR 6290, Institut de Génétique et Développement de Rennes, Rennes, France
- Université Rennes 1, UEB, Biosit, Faculté de Médecine, Rennes, France
| | - Thomas Derrien
- CNRS, UMR 6290, Institut de Génétique et Développement de Rennes, Rennes, France
- Université Rennes 1, UEB, Biosit, Faculté de Médecine, Rennes, France
| | - Catherine André
- CNRS, UMR 6290, Institut de Génétique et Développement de Rennes, Rennes, France
- Université Rennes 1, UEB, Biosit, Faculté de Médecine, Rennes, France
- * E-mail: (CA); (JP)
| |
Collapse
|
19
|
Wong FSY, Wong CCH, Chan BP, Lo ACY. Sustained Delivery of Bioactive GDNF from Collagen and Alginate-Based Cell-Encapsulating Gel Promoted Photoreceptor Survival in an Inherited Retinal Degeneration Model. PLoS One 2016; 11:e0159342. [PMID: 27441692 PMCID: PMC4956057 DOI: 10.1371/journal.pone.0159342] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 06/30/2016] [Indexed: 11/29/2022] Open
Abstract
Encapsulated-cell therapy (ECT) is an attractive approach for continuously delivering freshly synthesized therapeutics to treat sight-threatening posterior eye diseases, circumventing repeated invasive intravitreal injections and improving local drug availability clinically. Composite collagen-alginate (CAC) scaffold contains an interpenetrating network that integrates the physical and biological merits of its constituents, including biocompatibility, mild gelling properties and availability. However, CAC ECT properties and performance in the eye are not well-understood. Previously, we reported a cultured 3D CAC system that supported the growth of GDNF-secreting HEK293 cells with sustainable GDNF delivery. Here, the system was further developed into an intravitreally injectable gel with 1x104 or 2x105 cells encapsulated in 2mg/ml type I collagen and 1% alginate. Gels with lower alginate concentration yielded higher initial cell viability but faster spheroid formation while increasing initial cell density encouraged cell growth. Continuous GDNF delivery was detected in culture and in healthy rat eyes for at least 14 days. The gels were well-tolerated with no host tissue attachment and contained living cell colonies. Most importantly, gel-implanted in dystrophic Royal College of Surgeons rat eyes for 28 days retained photoreceptors while those containing higher initial cell number yielded better photoreceptor survival. CAC ECT gels offers flexible system design and is a potential treatment option for posterior eye diseases.
Collapse
Affiliation(s)
- Francisca S. Y. Wong
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Calvin C. H. Wong
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Barbara P. Chan
- Tissue Engineering Laboratory, Department of Mechanical Engineering, Faculty of Engineering, The University of Hong Kong, Hong Kong, China
| | - Amy C. Y. Lo
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Research Centre of Heart, Brain, Hormone and Healthy Aging, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- * E-mail:
| |
Collapse
|
20
|
Bolon B, Jing S, Asuncion F, Scully S, Pisegna M, Van GY, Hu Z, Yu YB, Min H, Wild K, Rosenfeld RD, Tarpley J, Carnahan J, Duryea D, Hill D, Kaufman S, Yan XQ, Juan T, Christensen K, McCabe J, Simonet WS. The Candidate Neuroprotective Agent Artemin Induces Autonomic Neural Dysplasia without Preventing Peripheral Nerve Dysfunction. Toxicol Pathol 2016; 32:275-94. [PMID: 15204970 DOI: 10.1080/01926230490431475] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Artemin (ART) signals through the GFR α—3/RET receptor complex to support sympathetic neuron development. Here we show that ART also influences autonomic elements in adrenal medulla and enteric and pelvic ganglia. Transgenic mice over-expressing Art throughout development exhibited systemic autonomic neural lesions including fusion of adrenal medullae with adjacent paraganglia, adrenal medullary dysplasia, and marked enlargement of sympathetic (superior cervical and sympathetic chain ganglia) and parasympathetic (enteric, pelvic) ganglia. Changes began by gestational day 12.5 and formed progressively larger masses during adulthood. Art supplementation in wild type adult mice by administering recombinant protein or an Art-bearing retroviral vector resulted in hyperplasia or neuronal metaplasia at the adrenal corticomedullary junction. Expression data revealed that Gfr α—3 is expressed during development in the adrenal medulla, sensory and autonomic ganglia and their projections, while Art is found in contiguous mesenchymal domains (especially skeleton) and in certain nerves. Intrathecal Art therapy did not reduce hypalgesia in rats following nerve ligation. These data (1) confirm that ART acts as a differentiation factor for autonomic (chiefly sympathoadrenal but also parasympathetic) neurons, (2) suggest a role for ART overexpression in the genesis of pheochromocytomas and paragangliomas, and (3) indicate that ART is not a suitable therapy for peripheral neuropathy.
Collapse
Affiliation(s)
- Brad Bolon
- Department of Pathology, Amgen Inc., Thousand Oaks, California 91320-1799, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Bouilloux F, Thireau J, Ventéo S, Farah C, Karam S, Dauvilliers Y, Valmier J, Copeland NG, Jenkins NA, Richard S, Marmigère F. Loss of the transcription factor Meis1 prevents sympathetic neurons target-field innervation and increases susceptibility to sudden cardiac death. eLife 2016; 5. [PMID: 26857994 PMCID: PMC4760953 DOI: 10.7554/elife.11627] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 12/28/2015] [Indexed: 12/13/2022] Open
Abstract
Although cardio-vascular incidents and sudden cardiac death (SCD) are among the leading causes of premature death in the general population, the origins remain unidentified in many cases. Genome-wide association studies have identified Meis1 as a risk factor for SCD. We report that Meis1 inactivation in the mouse neural crest leads to an altered sympatho-vagal regulation of cardiac rhythmicity in adults characterized by a chronotropic incompetence and cardiac conduction defects, thus increasing the susceptibility to SCD. We demonstrated that Meis1 is a major regulator of sympathetic target-field innervation and that Meis1 deficient sympathetic neurons die by apoptosis from early embryonic stages to perinatal stages. In addition, we showed that Meis1 regulates the transcription of key molecules necessary for the endosomal machinery. Accordingly, the traffic of Rab5+ endosomes is severely altered in Meis1-inactivated sympathetic neurons. These results suggest that Meis1 interacts with various trophic factors signaling pathways during postmitotic neurons differentiation. DOI:http://dx.doi.org/10.7554/eLife.11627.001 Nerve cells called sympathetic neurons can control the activity of almost all of our organs without any conscious thought on our part. For example, these nerve cells are responsible for accelerating the heart rate during exercise. In a developing embryo, there are initially more of these neurons than are needed, and only those that develop correctly and form a connection with a target cell will survive. This is because the target cells provide the growing neurons with vital molecules called neurotrophins, which are trafficked back along the nerve fiber and into the main body of the nerve cell to ensure its survival. However, it is largely unknown which proteins or genes are also involved in this developmental process. Now, Bouilloux, Thireau et al. show that if a gene called Meis1 is inactivated in mice, the sympathetic neurons start to develop and grow nerve fibers, but then fail to establish connections with their target cells and finally die. The Meis1 gene encodes a transcription factor, which is a protein that regulates gene activity. Therefore, Bouilloux, Thireau et al. looked for the genes that are regulated by this transcription factor in sympathetic neurons. This search uncovered several genes that are involved in the packaging and trafficking of molecules within cells. Other experiments then revealed that the trafficking of molecules back along the nerve fiber was altered in mutant neurons in which the Meis1 gene had been inactivated. Furthermore, Meis1 mutant mice had problems with their heart rate, especially during exercise, and an increased risk of dying from a sudden cardiac arrest. These findings reveal a transcription factor that helps to establish a connection between a neuron and its target, and that activates a pattern of gene expression that works alongside the neurotrophin-based signals. Since all neurons undergo similar processes during development, future work could ask if comparable patterns of gene expression exist in other types of neurons, and if problems with such processes contribute to some neurodegenerative diseases. DOI:http://dx.doi.org/10.7554/eLife.11627.002
Collapse
Affiliation(s)
- Fabrice Bouilloux
- Institute for Neurosciences of Montpellier, Institut national de la santé et de la recherche médicale, Montpellier, France
| | - Jérôme Thireau
- Physiologie et Médecine Expérimentale du cœur et des Muscles, INSERM U1046, CNRS UMR 9214, University of Montpellier, Montpellier, France
| | - Stéphanie Ventéo
- Institute for Neurosciences of Montpellier, Institut national de la santé et de la recherche médicale, Montpellier, France
| | - Charlotte Farah
- Physiologie et Médecine Expérimentale du cœur et des Muscles, INSERM U1046, CNRS UMR 9214, University of Montpellier, Montpellier, France
| | - Sarah Karam
- Physiologie et Médecine Expérimentale du cœur et des Muscles, INSERM U1046, CNRS UMR 9214, University of Montpellier, Montpellier, France
| | - Yves Dauvilliers
- Sleep Unit, Department of Neurology, Gui-de-Chauliac hospital, Montpellier, France
| | - Jean Valmier
- Institute for Neurosciences of Montpellier, Institut national de la santé et de la recherche médicale, Montpellier, France
| | - Neal G Copeland
- Cancer Research Program, The Methodist Hospital Research Institute, Houston, United States
| | - Nancy A Jenkins
- Cancer Research Program, The Methodist Hospital Research Institute, Houston, United States
| | - Sylvain Richard
- Physiologie et Médecine Expérimentale du cœur et des Muscles, INSERM U1046, CNRS UMR 9214, University of Montpellier, Montpellier, France
| | - Frédéric Marmigère
- Institute for Neurosciences of Montpellier, Institut national de la santé et de la recherche médicale, Montpellier, France
| |
Collapse
|
22
|
BDNF and GDNF expression in discrete populations of nociceptors. Ann Anat 2015; 207:55-61. [PMID: 26706106 DOI: 10.1016/j.aanat.2015.12.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 11/30/2015] [Accepted: 12/01/2015] [Indexed: 12/25/2022]
Abstract
The brain derived neurotrophic factor (BDNF) and the glial cell line-derived neurotrophic factor (GDNF) are growth factors that promote the survival and differentiation of sensory neurons and intervene in the control of nociceptive neurotransmission. Both are synthesized by dorsal root ganglion (DRG) neurons and are anterogradely transported to the central terminals of the spinal cord dorsal horn. To better investigate the specific expression pattern of BDNF and GDNF in nociceptors, we studied their localization in relationship to other established nociceptive markers in the mouse DRGs. Our results can be summarized as follows: (1) BDNF and GDNF are expressed in distinct populations of small-to medium-sized DRG neurons, with BDNF three times more frequently expressed than GDNF (186.4±1.7 BDNF-immunoreactive (IR) cells/DRG vs 57.7±0.3 GDNF-IR cells/DRG; n=3 mice); (2) A subset of BDNF-expressing neurons and a subset of GDNF-expressing neurons are of the peptidergic type; (3) BDNF-IR neurons are a subpopulation of calcitonin gene-related peptide (CGRP)-IR neurons (41.3±0.4%), also positive for substance P (SP) (42.3±0.1%), but not for somatostatin (SST); (4) GDNF-IR neurons are a subpopulation of CGRP-IR neurons (95.8±0.1%), also positive for SST (67.9±2.1%), but not SP; (5) Neither BDNF nor GDNF colocalized with the non-peptidergic marker IB4. Our results show the existence of two subpopulations of peptidergic nociceptors characterized by the presence of CGRP, one expressing BDNF (plus SP), the other expressing GDNF (plus SST), suggesting a different role for these two neurotrophic factors in the discrimination of specific painful stimuli modalities.
Collapse
|
23
|
Mohtaram NK, Ko J, Agbay A, Rattray D, Neill PO, Rajwani A, Vasandani R, Thu HL, Jun MBG, Willerth SM. Development of a glial cell-derived neurotrophic factor-releasing artificial dura for neural tissue engineering applications. J Mater Chem B 2015; 3:7974-7985. [DOI: 10.1039/c5tb00871a] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Develop a scaffold consisting of aligned, drug releasing nanofiber to serve as a replacement for damaged dura mater.
Collapse
Affiliation(s)
- N. K. Mohtaram
- Department of Mechanical Engineering
- University of Victoria
- Victoria
- Canada
| | - J. Ko
- Department of Mechanical Engineering
- University of Victoria
- Victoria
- Canada
| | - A. Agbay
- Division of Medical Sciences
- University of Victoria
- Victoria
- Canada
| | - D. Rattray
- Department of Biochemistry and Microbiology
- University of Victoria
- Victoria
- Canada
| | - P. O. Neill
- Department of Biochemistry and Microbiology
- University of Victoria
- Victoria
- Canada
| | - A. Rajwani
- Department of Biomedical Engineering
- University of Victoria
- Victoria
- Canada
| | - R. Vasandani
- Department of Mechanical Engineering
- University of Victoria
- Victoria
- Canada
| | - H. L. Thu
- Department of Biomedical Engineering
- International University-Vietnam National University
- Vietnam
| | - M. B. G. Jun
- Department of Mechanical Engineering
- University of Victoria
- Victoria
- Canada
| | - S. M. Willerth
- Department of Mechanical Engineering
- University of Victoria
- Victoria
- Canada
- Division of Medical Sciences
| |
Collapse
|
24
|
Functional effect of local administration of glial derived neurotrophic factor combined with inside-out artery graft on sciatic nerve regeneration in rat. Int J Surg 2014; 12:457-63. [DOI: 10.1016/j.ijsu.2014.03.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Revised: 01/29/2014] [Accepted: 03/28/2014] [Indexed: 11/19/2022]
|
25
|
Immunohistochemical localization of brain-derived neurotrophic factor and glial cell line-derived neurotrophic factor in the superior olivary complex of mice after radiofrequency exposure. Neurosci Lett 2014; 564:78-82. [DOI: 10.1016/j.neulet.2014.02.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 01/22/2014] [Accepted: 02/06/2014] [Indexed: 11/22/2022]
|
26
|
Nishimune H, Stanford JA, Mori Y. Role of exercise in maintaining the integrity of the neuromuscular junction. Muscle Nerve 2013; 49:315-24. [PMID: 24122772 DOI: 10.1002/mus.24095] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2013] [Indexed: 01/16/2023]
Abstract
Physical activity plays an important role in preventing chronic disease in adults and the elderly. Exercise has beneficial effects on the nervous system, including at the neuromuscular junction (NMJ). Exercise causes hypertrophy of NMJs and improves recovery from peripheral nerve injuries, whereas decreased physical activity causes degenerative changes in NMJs. Recent studies have begun to elucidate molecular mechanisms underlying the beneficial effects of exercise. These mechanisms involve Bassoon, neuregulin-1, peroxisome proliferator-activated receptor gamma coactivator 1α, insulin-like growth factor-1, glial cell line-derived neurotrophic factor, neurotrophin 4, Homer, and nuclear factor of activated T cells c1. For example, NMJ denervation and active zone decreases have been observed in aged NMJs, but these age-dependent degenerative changes can be ameliorated by exercise. In this review we assess the effects of exercise on the maintenance and regeneration of NMJs and highlight recent insights into the molecular mechanisms underlying these exercise effects.
Collapse
Affiliation(s)
- Hiroshi Nishimune
- Department of Anatomy and Cell Biology, University of Kansas School of Medicine, 3901 Rainbow Boulevard, MS 3051, HLSIC Room 2073, Kansas City, Kansas, 66160, USA
| | | | | |
Collapse
|
27
|
Combination of Chondroitinase ABC, Glial Cell Line–Derived Neurotrophic Factor and Nogo A Antibody Delayed-Release Microspheres Promotes the Functional Recovery of Spinal Cord Injury. J Craniofac Surg 2013; 24:2153-7. [DOI: 10.1097/01.scs.0000436700.65891.3b] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
28
|
Murase S, Kato K, Taguchi T, Mizumura K. Glial cell line-derived neurotrophic factor sensitized the mechanical response of muscular thin-fibre afferents in rats. Eur J Pain 2013; 18:629-38. [PMID: 24174387 DOI: 10.1002/j.1532-2149.2013.00411.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2013] [Indexed: 11/07/2022]
Abstract
BACKGROUND The role of glial cell line-derived neurotrophic factor (GDNF) in pain and muscular nociceptor activities is not well understood. We examined pain-related behaviour and mechanical response of muscular thin-fibre afferents after intramuscular injection of GDNF in rats. METHODS GDNF and antagonist to transient receptor potential V1 or acid-sensing ion channels were injected into rat gastrocnemius muscle and muscular mechanical hyperalgesia was assessed with a Randall-Selitto analgesiometer. Activities of single C- (conduction velocity < 2.0 m/s) and Aδ-fibres (conduction velocity 2.0-12.0 m/s) were recorded from extensor digitorum longus muscle-nerve preparations in vitro. The changes in the responses to mechanical stimuli before and after GDNF injection were recorded. RESULTS Mechanical hyperalgesia was observed from 1 h to 1 day after GDNF (0.03 μM, 20 μL) injection. The decreased withdrawal threshold was temporarily reversed after intramuscular injection of amiloride (50 mM, 20 μL), but not capsazepine (50 μM, 20 μL). In single-fibre recordings, both phosphate buffered saline (PBS) and GDNF failed to induce any significant discharges. GDNF significantly enhanced the mechanical response when compared with the PBS group, but only in Aδ-fibre afferents. C-fibres were not affected. Significantly lowered threshold and increased response magnitude to mechanical stimuli were observed 30 or 60-120 min after injection. These times are compatible with the timing of the onset of the hyperalgesic effect of GDNF. CONCLUSIONS These results suggest that GDNF increased the response of muscular Aδ-fibre afferents to mechanical stimuli, resulting in muscular mechanical hyperalgesia.
Collapse
Affiliation(s)
- S Murase
- Department of Physical Therapy, College of Life and Health Sciences, Chubu University, Kasugai, Japan; Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Japan
| | | | | | | |
Collapse
|
29
|
Sympathetic innervation induced in engrafted engineered cardiomyocyte sheets by glial cell line derived neurotrophic factor in vivo. BIOMED RESEARCH INTERNATIONAL 2013; 2013:532720. [PMID: 24066291 PMCID: PMC3771253 DOI: 10.1155/2013/532720] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 07/18/2013] [Indexed: 11/17/2022]
Abstract
The aim of myocardial tissue engineering is to repair or regenerate damaged myocardium with engineered cardiac tissue. However, this strategy has been hampered by lack of functional integration of grafts with native myocardium. Autonomic innervation may be crucial for grafts to function properly with host myocardium. In this study, we explored the feasibility of in vivo induction of autonomic innervation to engineered myocardial tissue using genetic modulation by adenovirus encoding glial cell line derived neurotrophic factor (GDNF). GFP-transgene (control group) or GDNF overexpressing (GDNF group) engineered cardiomyocyte sheets were transplanted on cryoinjured hearts in rats. Nerve fibers in the grafts were examined by immunohistochemistry at 1, 2, and 4 weeks postoperatively. Growth associated protein-43 positive growing nerves and tyrosine hydroxylase positive sympathetic nerves were first detected in the grafts at 2 weeks postoperatively in control group and 1 week in GDNF group. The densities of growing nerve and sympathetic nerve in grafts were significantly increased in GDNF group. No choline acetyltransferase immunopositive parasympathetic nerves were observed in grafts. In conclusion, sympathetic innervation could be effectively induced into engrafted engineered cardiomyocyte sheets using GDNF.
Collapse
|
30
|
Ota H, Katanosaka K, Murase S, Kashio M, Tominaga M, Mizumura K. TRPV1 and TRPV4 play pivotal roles in delayed onset muscle soreness. PLoS One 2013; 8:e65751. [PMID: 23799042 PMCID: PMC3684597 DOI: 10.1371/journal.pone.0065751] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 04/29/2013] [Indexed: 11/18/2022] Open
Abstract
Unaccustomed strenuous exercise that includes lengthening contraction (LC) often causes tenderness and movement related pain after some delay (delayed-onset muscle soreness, DOMS). We previously demonstrated that nerve growth factor (NGF) and glial cell line-derived neurotrophic factor (GDNF) are up-regulated in exercised muscle through up-regulation of cyclooxygenase (COX)-2, and they sensitized nociceptors resulting in mechanical hyperalgesia. There is also a study showing that transient receptor potential (TRP) ion channels are involved in DOMS. Here we examined whether and how TRPV1 and/or TRPV4 are involved in DOMS. We firstly evaluated a method to measure the mechanical withdrawal threshold of the deep tissues in wild-type (WT) mice with a modified Randall-Selitto apparatus. WT, TRPV1−/− and TRPV4−/− mice were then subjected to LC. Another group of mice received injection of murine NGF-2.5S or GDNF to the lateral gastrocnemius (LGC) muscle. Before and after these treatments the mechanical withdrawal threshold of LGC was evaluated. The change in expression of NGF, GDNF and COX-2 mRNA in the muscle was examined using real-time RT-PCR. In WT mice, mechanical hyperalgesia was observed 6–24 h after LC and 1–24 h after NGF and GDNF injection. LC induced mechanical hyperalgesia neither in TRPV1−/− nor in TRPV4−/− mice. NGF injection induced mechanical hyperalgesia in WT and TRPV4−/− mice but not in TRPV1−/− mice. GDNF injection induced mechanical hyperalgesia in WT but neither in TRPV1−/− nor in TRPV4−/− mice. Expression of NGF and COX-2 mRNA was significantly increased 3 h after LC in all genotypes. However, GDNF mRNA did not increase in TRPV4−/− mice. These results suggest that TRPV1 contributes to DOMS downstream (possibly at nociceptors) of NGF and GDNF, while TRPV4 is located downstream of GDNF and possibly also in the process of GDNF up-regulation.
Collapse
Affiliation(s)
- Hiroki Ota
- Department of Neural Regulation, Graduate School of Medicine, Nagoya University, Nagoya, Japan
- Department of Physical Therapy, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| | - Kimiaki Katanosaka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Shiori Murase
- Department of Physical Therapy, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| | - Makiko Kashio
- Division of Cell Signaling, Okazaki Institute for Integrative Bioscience, National Institute for Physiological Sciences, Okazaki, Japan
| | - Makoto Tominaga
- Division of Cell Signaling, Okazaki Institute for Integrative Bioscience, National Institute for Physiological Sciences, Okazaki, Japan
| | - Kazue Mizumura
- Department of Physical Therapy, College of Life and Health Sciences, Chubu University, Kasugai, Japan
- * E-mail:
| |
Collapse
|
31
|
Abstract
Our aim is to provide a summary of the field of salivary gland development and regeneration from the perspective of what is known about the function of nerves during these processes. The primary function of adult salivary glands is to produce and secrete saliva. Neuronal control of adult salivary gland function has been a focus of research ever since Pavlov’s seminal experiments on salivation in dogs. Less is known about salivary gland innervation during development and how the developing nerves influence gland organogenesis and regeneration. Here, we will review what is known about the communication between the autonomic nervous system and the epithelium of the salivary glands during organogenesis. An important emerging theme is the instructive role of the nervous system on the epithelial stem/progenitor cells during development as well as regeneration after damage. We will provide a brief overview of the neuroanatomy of the salivary glands and discuss recent literature that begins to integrate neurobiology with epithelial organogenesis, which may provide paradigms for exploring these interactions in other organ systems.
Collapse
Affiliation(s)
- João N Ferreira
- Matrix and Morphogenesis Section; Laboratory of Cell and Developmental Biology; National Institute of Dental and Craniofacial Research; National Institutes of Health; Bethesda MD USA; Instructor/Lecturer at Faculty of Dentistry; Mahidol University; Rajthevi, Thailand
| | | |
Collapse
|
32
|
Allen SJ, Watson JJ, Shoemark DK, Barua NU, Patel NK. GDNF, NGF and BDNF as therapeutic options for neurodegeneration. Pharmacol Ther 2013; 138:155-75. [PMID: 23348013 DOI: 10.1016/j.pharmthera.2013.01.004] [Citation(s) in RCA: 595] [Impact Index Per Article: 49.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 01/07/2013] [Indexed: 12/16/2022]
Abstract
Glial cell-derived neurotrophic factor (GDNF), and the neurotrophin nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) are important for the survival, maintenance and regeneration of specific neuronal populations in the adult brain. Depletion of these neurotrophic factors has been linked with disease pathology and symptoms, and replacement strategies are considered as potential therapeutics for neurodegenerative diseases such as Parkinson's, Alzheimer's and Huntington's diseases. GDNF administration has recently been shown to be an effective treatment for Parkinson's disease, with clinical trials currently in progress. Trials with NGF for Alzheimer's disease are ongoing, with some degree of success. Preclinical results using BDNF also show much promise, although there are accompanying difficulties. Ultimately, the administration of a therapy involving proteins in the brain has inherent problems. Because of the blood-brain-barrier, the protein must be infused directly, produced by viral constructs, secreted from implanted protein-secreting cells or actively transported across the brain. An alternative to this is the use of a small molecule agonist, a modulator or enhancer targeting the associated receptors. We evaluate these neurotrophic factors as potential short or long-term treatments, weighing up preclinical and clinical results with the possible effects on the underlying neurodegenerative process.
Collapse
|
33
|
Murase S, Terazawa E, Hirate K, Yamanaka H, Kanda H, Noguchi K, Ota H, Queme F, Taguchi T, Mizumura K. Upregulated glial cell line-derived neurotrophic factor through cyclooxygenase-2 activation in the muscle is required for mechanical hyperalgesia after exercise in rats. J Physiol 2013; 591:3035-48. [PMID: 23587883 DOI: 10.1113/jphysiol.2012.249235] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Unaccustomed strenuous exercise that includes lengthening contraction (LC) often causes delayed onset muscle soreness (DOMS), characterised as muscular mechanical hyperalgesia. Previously we reported that a bradykinin-like substance released from the muscle during exercise plays a pivotal role in triggering the process of muscular mechanical hyperalgesia by upregulating nerve growth factor (NGF) in exercised muscle of rats. We show here that cyclooxygenase (COX)-2 and glial cell line-derived neurotrophic factor (GDNF) are also involved in DOMS. COX-2 inhibitors but not COX-1 inhibitors given orally before LC completely suppressed the development of DOMS, but when given 2 days after LC they failed to reverse the mechanical hyperalgesia. COX-2 mRNA and protein in exercised muscle increased six- to 13-fold in mRNA and 1.7-2-fold in protein 0-12 h after LC. COX-2 inhibitors did not suppress NGF upregulation after LC. Instead, we found GDNF mRNA was upregulated seven- to eight-fold in the exercised muscle 12 h-1 day after LC and blocked by pretreatment of COX-2 inhibitors. In situ hybridisation studies revealed that both COX-2 and GDNF mRNA signals increased at the periphery of skeletal muscle cells 12 h after LC. The accumulation of COX-2 mRNA signals was also observed in small blood vessels. Intramuscular injection of anti-GDNF antibody 2 days after LC partly reversed DOMS. Based on these findings, we conclude that GDNF upregulation through COX-2 activation is essential to mechanical hyperalgesia after exercise.
Collapse
Affiliation(s)
- Shiori Murase
- Department of Physical Therapy, College of Life and Health Sciences, Chubu University, Kasugai 487-8501, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Huang Y, Zheng S. Effect of exogenous glial cell-derived neurotrophic factor on development of the enteric nervous system in the rectal end of fetal rats with anorectal malformations. Pediatr Surg Int 2013; 29:375-9. [PMID: 23334620 DOI: 10.1007/s00383-012-3251-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
OBJECTIVE The objective of this study is to analyze the effect of exogenous glial cell-derived neurotrophic factor (GDNF) in the development of the enteric nervous system (ENS) in the rectal end of fetal rats. MATERIALS AND METHODS Thirty pregnant Sprague-Dawley rats were categorized randomly into three groups: ethylene thiourea (ETU), ETU + GDNF, and control. On day 10 of gestation, ETU was injected via a gastric tube in the ETU group and ETU + GDNF group. On day 11 of gestation, GDNF was administered through the tail vein in the ETU + GDNF group. On day 20 of gestation, fetal rats were harvested by cesarean section. The prevalence of anorectal malformations (ARMs) in the fetal rats was observed. GDNF expression in the rectal end of fetal rats was detected by immunohistochemical and Western blotting analyses. RESULTS The prevalence of ARMs in the ETU group and ETU + GDNF group was 51.4 and 52.5 %, respectively, but the difference between the two groups was not significant (P > 0.05). In the rectal end of fetal rats with an anus, GDNF expressions in the three groups were not significantly different (P > 0.05). In the rectal end of fetal rats without an anus: GDNF expression in the ETU + GDNF group was significantly higher than that in the ETU group (P = 0.036); GDNF expression in the rectal end of fetal rats without an anus from the ETU group and ETU + GDNF group was significantly lower than that of fetal rats with an anus (ETU group P = 0.001; ETU + GDNF group P = 0.028). There was a significant difference in the gray level ratio of GDNF and actin between the ETU group and ETU + GDNF group (P < 0.0001), and the expression in the ETU + GDNF group was significantly up-regulated. CONCLUSION GDNF could not totally prevent the occurrence of ETU-induced ARMs, but it up-regulated expression of the GDNF gene in the wall of the rectal end, thereby promoting the growth of a hypogenetic ENS.
Collapse
Affiliation(s)
- Yanlei Huang
- Department of Pediatric Surgery, Children's Hospital of Fudan University, 399 Wan Yuan Road, Shanghai, People's Republic of China
| | | |
Collapse
|
35
|
Boscia F, Esposito CL, Casamassa A, de Franciscis V, Annunziato L, Cerchia L. The isolectin IB4 binds RET receptor tyrosine kinase in microglia. J Neurochem 2013; 126:428-36. [PMID: 23413818 DOI: 10.1111/jnc.12209] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 02/07/2013] [Accepted: 02/08/2013] [Indexed: 02/01/2023]
Abstract
Ret receptor tyrosine kinase is the signaling component of the receptor complex for the family ligands of the glial cell line-derived neurotrophic factor (GDNF). Ret is involved in the development of enteric nervous system, of sympathetic, parasympathetic, motor and sensory neurons, and it is necessary for the post-natal maintenance of dopaminergic neurons. Ret expression has been as well demonstrated on microglia and several evidence indicate that GDNF regulates not only neuronal survival and maturation but also certain functions of microglia in the brain. Here, we demonstrated that the plant lectin Griffonia (Bandeiraea) simplicifolia lectin I, isolectin B4 (IB4), commonly used as a microglial marker in the brain, binds to the glycosylated extracellular domain of Ret on the surface of living NIH3T3 fibroblasts cells stably transfected with Ret as well as in adult rat brain as revealed by immunoblotting. Furthermore, confocal immunofluorescence analysis demonstrated a clear overlap in staining between pRet and IB4 in primary microglia cultures as well as in adult rat sections obtained from control or post-ischemic brain after permanent middle artery occlusion (pMCAO). Interestingly, IB4 staining identified activated or ameboid Ret-expressing microglia under ischemic conditions. Collectively, our data indicate Ret receptor as one of the IB4-reactive glycoconjugate accounting for the IB4 stain in microglia under physiological and ischemic conditions.
Collapse
Affiliation(s)
- Francesca Boscia
- Dipartimento di Neuroscienze, Sezione di Farmacologia, Facolta' di Medicina e Chirurgia, Universita' degli Studi di Napoli Federico II, Naples, Italy
| | | | | | | | | | | |
Collapse
|
36
|
Heermann S, Mätlik K, Hinz U, Fey J, Arumae U, Krieglstein K. Glia cell line-derived neurotrophic factor mediates survival of murine sympathetic precursors. J Neurosci Res 2013; 91:780-5. [PMID: 23426908 DOI: 10.1002/jnr.23188] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Revised: 11/12/2012] [Accepted: 11/13/2012] [Indexed: 11/11/2022]
Abstract
During embryonic development, neurons are first produced in excess, and final numbers are adjusted by apoptosis at later stages. Crucial to this end is the amount of target-derived growth factor available for the neurons. By this means, the target size correctly matches the innervating neuron number. This target-derived survival has been well studied for sympathetic neurons, and nerve growth factor (NGF) was identified to be the crucial factor for maintaining sympathetic neurons at late embryonic and early postnatal stages, with a virtual complete loss of sympathetic neurons in NGF knockout (KO) mice. This indicates that all sympathetic neurons are dependent on NGF. However, also different glia cell line-derived neurotrophic factor (GDNF) KO mice consistently presented a loss of sympathetic neurons. This was the rationale for investigating the role of GDNF for sympathetic precursor/neuron survival. Here we show that GDNF is capable of promoting survival of 30% sympathetic precursors dissociated at E13. This is in line with data from GDNF KOs in which a comparable sympathetic neuron loss was observed at late embryonic stages, although the onset of the phenotype was unclear. We further present data showing that GDNF ligand and canonical receptors are expressed in sympathetic neurons especially at embryonic stages, raising the possibility of an autocrine/paracrine GDNF action. Finally, we show that GDNF also maintained neonatal sympathetic neurons (40%) cultured for 2 days. However, the GDNF responsiveness was lost at 5 days in vitro.
Collapse
Affiliation(s)
- Stephan Heermann
- Department of Molecular Embryology Institute of Anatomy and Cell Biology, University of Freiburg, Freiburg, Germany.
| | | | | | | | | | | |
Collapse
|
37
|
Awad BI, Carmody MA, Steinmetz MP. Potential role of growth factors in the management of spinal cord injury. World Neurosurg 2013; 83:120-31. [PMID: 23334003 DOI: 10.1016/j.wneu.2013.01.042] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Revised: 01/06/2013] [Accepted: 01/11/2013] [Indexed: 12/31/2022]
Abstract
OBJECTIVE To review central nervous system growth factors and their therapeutic potential and clinical translation into spinal cord injury (SCI), as well as the challenges that have been encountered during clinical development. METHODS A systemic review of the available current and historical literature regarding central nervous system growth factors and clinical trials regarding their use in spinal cord injury was conducted. RESULTS The effectiveness of administering growth factors as a potential therapeutic strategy for SCI has been tested with the use of brain-derived neurotrophic factor, glial cell-derived neurotrophic factor, neurotrophin 3, and neurotrophin-4/5. Delivery of growth factors to injured SC has been tested by numerous methods. Unfortunately, most of clinical trials at this time are uncontrolled and have questionable results because of lack of efficacy and/or unacceptable side effects. CONCLUSIONS There is promise in the use of specific growth factors therapeutically for SCI. However, more studies involving neuronal regeneration and functional recovery are needed, as well the development of delivery methods that allow sufficient quantity of growth factors while restricting their distribution to target sites.
Collapse
Affiliation(s)
- Basem I Awad
- Department of Neurosurgery, Mansoura University School of Medicine, Mansoura, Egypt; Department of Neurosciences, MetroHealth Medical Center, Cleveland, Ohio, USA
| | - Margaret A Carmody
- Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Michael P Steinmetz
- Department of Neurosciences, MetroHealth Medical Center, Cleveland, Ohio, USA.
| |
Collapse
|
38
|
Simanainen U, Gao YRE, Desai R, Jimenez M, Spaliviero J, Keast JR, Handelsman DJ. Evidence for increased tissue androgen sensitivity in neurturin knockout mice. J Endocrinol 2013; 218:151-63. [PMID: 23678134 DOI: 10.1530/joe-13-0056] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Neurturin (NTN) is a member of the glial cell line-derived neurotrophic factor (GDNF) family and signals through GDNF family receptor alpha 2 (GFRα2). We hypothesised that epithelial atrophy reported in the reproductive organs of Ntn (Nrtn)- and Gfrα2 (Gfra2)-deficient mice could be due to NTN affecting the hormonal environment. To investigate this, we compared the reproductive organs of Ntn- and Gfrα2-deficient male mice in parallel with an analysis of their circulating reproductive hormone levels. There were no significant structural changes within the organs of the knockout mice; however, serum and intratesticular testosterone and serum LH levels were very low. To reconcile these observations, we tested androgen sensitivity by creating a dihydrotestosterone (DHT) clamp (castration plus DHT implant) to create fixed circulating levels of androgens, allowing the evaluation of androgen-sensitive endpoints. At the same serum DHT levels, serum LH levels were lower and prostate and seminal vesicle weights were higher in the Ntn knockout (NTNKO) mice than in the wild-type mice, suggesting an increased response to androgens in the accessory glands and hypothalamus and pituitary of the NTNKO mice. Testicular and pituitary responsiveness was unaffected in the NTNKO males, as determined by the response to the human chorionic gonadotrophin or GNRH analogue, leuprolide, respectively. In conclusion, our results suggest that NTN inactivation enhances androgen sensitivity in reproductive and neuroendocrine tissues, revealing a novel mechanism to influence reproductive function and the activity of other androgen-dependent tissues.
Collapse
Affiliation(s)
- Ulla Simanainen
- ANZAC Research Institute, University of Sydney, Sydney, New South Wales 2139, Australia
| | | | | | | | | | | | | |
Collapse
|
39
|
Simpson J, Keefe J, Nishi R. Differential effects of RET and TRKB on axonal branching and survival of parasympathetic neurons. Dev Neurobiol 2012; 73:45-59. [PMID: 22648743 DOI: 10.1002/dneu.22036] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2011] [Revised: 04/11/2012] [Accepted: 05/25/2012] [Indexed: 12/25/2022]
Abstract
Interactions between neurons and their targets of innervation influence many aspects of neural development. To examine how synaptic activity interacts with neurotrophic signaling, we determined the effects of blocking neuromuscular transmission on survival and axonal outgrowth of ciliary neurons from the embryonic chicken ciliary ganglion. Ciliary neurons undergo a period of cell loss due to programmed cell death between embryonic Days (E) 8 and 14 and they innervate the striated muscle of the iris. The nicotinic antagonist d-tubocurarine (dTC) induces an increase in branching measured by counting neurofilament-positive voxels (NF-VU) in the iris between E14-17 while reducing ciliary neuron survival. Blocking ganglionic transmission with dihyro-β-erythroidin and α-methyllycacontine does not mimic dTC. At E8, many trophic factors stimulate neurite outgrowth and branching of neurons placed in cell culture; however, at E13, only GDNF stimulates branching selectively in cultured ciliary neurons. The GDNF-induced branching at E13 could be inhibited by BDNF. Blocking ret signaling in vivo with a dominant negative (dn)ret decreases survival of ciliary and choroid neurons at E14 and prevents dTC induced increases in NF-VU in the iris at E17. Blocking TRKB signaling with dn TRKB increases NF-VU in the iris at E17 and decreases neuronal survival at E17, but not at E14. Thus, RET promotes survival during programmed cell death in the ciliary ganglion and contributes to promoting branching when synaptic transmission is blocked while TRKB inhibits branching and promotes maintenance of neuronal survival. These studies highlight the multifunctional nature of trophic molecule function during neuronal development.
Collapse
Affiliation(s)
- Julie Simpson
- Department of Anatomy and Neurobiology, University of Vermont College of Medicine, Burlington, Vermont 05405, USA
| | | | | |
Collapse
|
40
|
Spofford CM, Brennan TJ. Gene expression in skin, muscle, and dorsal root ganglion after plantar incision in the rat. Anesthesiology 2012; 117:161-72. [PMID: 22617252 PMCID: PMC3389501 DOI: 10.1097/aln.0b013e31825a2a2b] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
BACKGROUND Treating postoperative pain remains a significant challenge for perioperative medicine. Recent studies have shown that nerve growth factor is up-regulated and contributes to incisional pain. To date, few studies have examined expression of other neurotrophin-related mediators that may contribute to the development and/or maintenance of incisional pain. METHODS Male Sprague-Dawley rats underwent a plantar incision, and pain behaviors were examined (n = 6). In a separate group of rats, expression of neurotrophic factors were studied. At various times after incision (n = 4) or sham surgery (n = 4), the skin, muscle, and dorsal root ganglia were harvested and total RNA isolated. Real-time reverse transcription polymerase chain reaction was performed and the fold change in gene expression was analyzed using significance analysis of microarrays. RESULTS Several genes were changed (P < 0.05) as early as 1 h after incision. Expression of artemin and nerve growth factor were increased in both incised skin and muscle. Brain-derived neurotrophic factor, neurotrophin-3, and neurotrophin-5 were all down-regulated in the skin but up-regulated in the muscle 48 h after incision. Few genes changed in the dorsal root ganglion. Most changes in expression occurred in the first 48 h after incision, a timeframe when pain behavior was the greatest. CONCLUSION Surgical incision is associated with pain-related gene expression changes in skin, muscle, and, to a lesser extent, dorsal root ganglion. The gene expression profile provides clues as to mediators that are involved in peripheral sensitization and pain transmission after surgical incision and also suggest mechanisms for resolution of postoperative pain when more persistent pain syndromes like neuropathic pain continue.
Collapse
|
41
|
Specificity of peripheral nerve regeneration: interactions at the axon level. Prog Neurobiol 2012; 98:16-37. [PMID: 22609046 DOI: 10.1016/j.pneurobio.2012.05.005] [Citation(s) in RCA: 289] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 04/12/2012] [Accepted: 05/08/2012] [Indexed: 12/13/2022]
Abstract
Peripheral nerves injuries result in paralysis, anesthesia and lack of autonomic control of the affected body areas. After injury, axons distal to the lesion are disconnected from the neuronal body and degenerate, leading to denervation of the peripheral organs. Wallerian degeneration creates a microenvironment distal to the injury site that supports axonal regrowth, while the neuron body changes in phenotype to promote axonal regeneration. The significance of axonal regeneration is to replace the degenerated distal nerve segment, and achieve reinnervation of target organs and restitution of their functions. However, axonal regeneration does not always allows for adequate functional recovery, so that after a peripheral nerve injury, patients do not recover normal motor control and fine sensibility. The lack of specificity of nerve regeneration, in terms of motor and sensory axons regrowth, pathfinding and target reinnervation, is one the main shortcomings for recovery. Key factors for successful axonal regeneration include the intrinsic changes that neurons suffer to switch their transmitter state to a pro-regenerative state and the environment that the axons find distal to the lesion site. The molecular mechanisms implicated in axonal regeneration and pathfinding after injury are complex, and take into account the cross-talk between axons and glial cells, neurotrophic factors, extracellular matrix molecules and their receptors. The aim of this review is to look at those interactions, trying to understand if some of these molecular factors are specific for motor and sensory neuron growth, and provide the basic knowledge for potential strategies to enhance and guide axonal regeneration and reinnervation of adequate target organs.
Collapse
|
42
|
Takasu K, Sakai A, Hanawa H, Shimada T, Suzuki H. Overexpression of GDNF in the uninjured DRG exerts analgesic effects on neuropathic pain following segmental spinal nerve ligation in mice. THE JOURNAL OF PAIN 2012; 12:1130-9. [PMID: 21684216 DOI: 10.1016/j.jpain.2011.04.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Revised: 02/28/2011] [Accepted: 04/04/2011] [Indexed: 02/05/2023]
Abstract
UNLABELLED Glial cell line-derived neurotrophic factor (GDNF), a survival-promoting factor for a subset of nociceptive small-diameter neurons, has been shown to exert analgesic effects on neuropathic pain. However, its detailed mechanisms of action are still unknown. In the present study, we investigated the site-specific analgesic effects of GDNF in the neuropathic pain state using lentiviral vector-mediated GDNF overexpression in mice with left fifth lumbar (L5) spinal nerve ligation (SNL) as a neuropathic pain model. A lentiviral vector expressing both GDNF and enhanced green fluorescent protein (EGFP) was constructed and injected into the left dorsal spinal cord, uninjured fourth lumbar (L4) dorsal root ganglion (DRG), injured L5 DRG, or plantar skin of mice. In SNL mice, injection of the GDNF-EGFP-expressing lentivirus into the dorsal spinal cord or uninjured L4 DRG partially but significantly reduced the mechanical allodynia in association with an increase in GDNF protein expression in each virus injection site, whereas injection into the injured L5 DRG or plantar skin had no effects. These results suggest that GDNF exerts its analgesic effects in the neuropathic pain state by acting on the central terminals of uninjured DRG neurons and/or on the spinal cells targeted by the uninjured DRG neurons. PERSPECTIVE This article shows that GDNF exerts its analgesic effects on neuropathic pain by acting on the central terminals of uninjured DRG neurons and/or on the spinal cells targeted by these neurons. Therefore, research focusing on these GDNF-dependent neurons in the uninjured DRG would provide a new strategy for treating neuropathic pain.
Collapse
Affiliation(s)
- Kumiko Takasu
- Department of Pharmacology, Nippon Medical School, Tokyo, Japan
| | | | | | | | | |
Collapse
|
43
|
Hancock ML, Nowakowski DW, Role LW, Talmage DA, Flanagan JG. Type III neuregulin 1 regulates pathfinding of sensory axons in the developing spinal cord and periphery. Development 2011; 138:4887-98. [PMID: 22028026 DOI: 10.1242/dev.072306] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Sensory axons must develop appropriate connections with both central and peripheral targets. Whereas the peripheral cues have provided a classic model for neuron survival and guidance, less is known about the central cues or the coordination of central and peripheral connectivity. Here we find that type III Nrg1, in addition to its known effect on neuron survival, regulates axon pathfinding. In type III Nrg1(-/-) mice, death of TrkA(+) nociceptive/thermoreceptive neurons was increased, and could be rescued by Bax elimination. In the Bax and type III Nrg1 double mutants, axon pathfinding abnormalities were seen for TrkA(+) neurons both in cutaneous peripheral targets and in spinal cord central targets. Axon guidance phenotypes in the spinal cord included penetration of axons into ventral regions from which they would normally be repelled by Sema3A. Accordingly, sensory neurons from type III Nrg1(-/-) mice were unresponsive to the repellent effects of Sema3A in vitro, which might account, at least in part, for the central projection phenotype, and demonstrates an effect of type III Nrg1 on guidance cue responsiveness in neurons. Moreover, stimulation of type III Nrg1 back-signaling in cultured sensory neurons was found to regulate axonal levels of the Sema3A receptor neuropilin 1. These results reveal a molecular mechanism whereby type III Nrg1 signaling can regulate the responsiveness of neurons to a guidance cue, and show that type III Nrg1 is required for normal sensory neuron survival and axon pathfinding in both central and peripheral targets.
Collapse
Affiliation(s)
- Melissa L Hancock
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
44
|
Agmatine-promoted angiogenesis, neurogenesis, and inhibition of gliosis-reduced traumatic brain injury in rats. ACTA ACUST UNITED AC 2011; 71:E87-93. [PMID: 21427621 DOI: 10.1097/ta.0b013e31820932e2] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND The mechanisms of agmatine-induced neuroprotective effects in traumatic brain injury (TBI) remain unclear. This study was to test whether inhibition of gliosis, angiogenesis, and neurogenesis attenuating TBI could be agmatine stimulated. METHODS Anesthetized rats were randomly assigned to sham-operated group, TBI rats treated with saline (1 mL/kg, intraperitoneally), or TBI rats treated with agmatine (50 mg/kg, intraperitoneally). Saline or agmatine was injected 5 minutes after TBI and again once daily for the next 3 postoperative days. RESULTS Agmatine therapy in rats significantly attenuated TBI-induced motor function deficits (62° vs. 52° maximal angle) and cerebral infarction (88 mm vs. 216 mm), significantly reduced TBI-induced neuronal (9 NeuN-TUNEL double positive cells vs. 60 NeuN-TUNEL double positive cells) and glial (2 GFAP-TUNEL double positive cells vs. 20 GFAP-TUNEL double positive cells) apoptosis (increased TUNEL-positive and caspase-3-positive cells), neuronal loss (82 NeuN-positive cells vs. 60 NeuN-positive cells), gliosis (35 GFAP-positive cells vs. 72 GFAP-positive cells; 60 Iba1-positive cells vs. 90 Iba1-positive cells), and neurotoxicity (30 n-NOS-positive cells vs. 90 n-NOS-positive cells; 35 3-NT-positive cells vs. 90 3-NT-positive cells), and significantly promoted angiogenesis (3 BrdU/endothelial cells vs. 0.5 BrdU/endothelial cells; 50 vascular endothelial growth factor positive cells vs. 20 vascular endothelial growth factor-positive cells) and neurogenesis (27 BrdU/NeuN positive cells vs. 15 BrdU/NeuN positive cells). CONCLUSIONS Resultantly, agmatine therapy may attenuate TBI in rats via promoting angiogenesis, neurogenesis, and inhibition of gliosis.
Collapse
|
45
|
In vitro comparison of motor and sensory neuron outgrowth in a 3D collagen matrix. J Neurosci Methods 2011; 198:53-61. [DOI: 10.1016/j.jneumeth.2011.03.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Revised: 02/07/2011] [Accepted: 03/08/2011] [Indexed: 02/01/2023]
|
46
|
Deng LX, Hu J, Liu N, Wang X, Smith GM, Wen X, Xu XM. GDNF modifies reactive astrogliosis allowing robust axonal regeneration through Schwann cell-seeded guidance channels after spinal cord injury. Exp Neurol 2011; 229:238-50. [PMID: 21316362 DOI: 10.1016/j.expneurol.2011.02.001] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Revised: 01/28/2011] [Accepted: 02/03/2011] [Indexed: 01/19/2023]
Abstract
Reactive astrogliosis impedes axonal regeneration after injuries to the mammalian central nervous system (CNS). Here we report that glial cell line-derived neurotrophic factor (GDNF), combined with transplanted Schwann cells (SCs), effectively reversed the inhibitory properties of astrocytes at graft-host interfaces allowing robust axonal regeneration, concomitant with vigorous migration of host astrocytes into SC-seeded semi-permeable guidance channels implanted into a right-sided spinal cord hemisection at the 10th thoracic (T10) level. Within the graft, migrated host astrocytes were in close association with regenerated axons. Astrocyte processes extended parallel to the axons, implying that the migrated astrocytes were not inhibitory and might have promoted directional growth of regenerated axons. In vitro, GDNF induced migration of SCs and astrocytes toward each other in an astrocyte-SC confrontation assay. GDNF also enhanced migration of astrocytes on a SC monolayer in an inverted coverslip migration assay, suggesting that this effect is mediated by direct cell-cell contact between the two cell types. Morphologically, GDNF administration reduced astrocyte hypertrophy and induced elongated process extension of these cells, similar to what was observed in vivo. Notably, GDNF treatment significantly reduced production of glial fibrillary acidic protein (GFAP) and chondroitin sulfate proteoglycans (CSPGs), two hallmarks of astrogliosis, in both the in vivo and in vitro models. Thus, our study demonstrates a novel role of GDNF in modifying spinal cord injury (SCI)-induced astrogliosis resulting in robust axonal regeneration in adult rats.
Collapse
Affiliation(s)
- Ling-Xiao Deng
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Glial cell line-derived neurotrophic factor defines the path of developing and regenerating axons in the lateral line system of zebrafish. Proc Natl Acad Sci U S A 2010; 107:19531-6. [PMID: 20974953 DOI: 10.1073/pnas.1002171107] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
How the peripheral axons of sensory neurons are guided to distant target organs is not well understood. Here we examine this question in the case of the posterior lateral line (PLL) system of zebrafish, where sensory organs are deposited by a migrating primordium. Sensory neurites accompany this primordium during its migration and are thereby guided to their prospective target organs. We show that the inactivation of glial cell line-derived neurotrophic factor (GDNF) signaling leads to defects of innervation and that these defects are due to the inability of sensory axons to track the migrating primordium. GDNF signaling is also used as a guidance cue during axonal regeneration following nerve cut. We conclude that GDNF is a major determinant of directed neuritic growth and of target finding in this system, and we propose that GDNF acts by promoting local neurite outgrowth.
Collapse
|
48
|
Young A, Assey KS, Sturkie CD, West FD, Machacek DW, Stice SL. Glial cell line-derived neurotrophic factor enhances in vitro differentiation of mid-/hindbrain neural progenitor cells to dopaminergic-like neurons. J Neurosci Res 2010; 88:3222-32. [DOI: 10.1002/jnr.22499] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
49
|
Henriques A, Pitzer C, Schneider A. Neurotrophic growth factors for the treatment of amyotrophic lateral sclerosis: where do we stand? Front Neurosci 2010; 4:32. [PMID: 20592948 PMCID: PMC2902233 DOI: 10.3389/fnins.2010.00032] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2010] [Accepted: 05/07/2010] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease that results in progressive loss of motoneurons, motor weakness and death within 3–5 years after disease onset. Therapeutic options remain limited despite substantial number of approaches that have been tested clinically. Many neurotrophic growth factors are known to promote the survival of neurons and foster regeneration in the central nervous system. Various neurotrophic factors have been investigated pre-clinically and clinically for the treatment of ALS. Although pre-clinical data appeared promising, no neurotrophic factors succeeded yet in a clinical phase III trial. In this review we discuss the rationale behind those factors, possible reasons for clinical failures, and argue for a renewal of hope in this powerful class of drugs for the treatment of ALS.
Collapse
|
50
|
The differential axonal degradation of Ret accounts for cell-type-specific function of glial cell line-derived neurotrophic factor as a retrograde survival factor. J Neurosci 2010; 30:5149-58. [PMID: 20392937 DOI: 10.1523/jneurosci.5246-09.2010] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) is a neuronal growth factor critical for the development and maintenance of central and peripheral neurons. GDNF is expressed in targets of innervation and provides support to several populations of large, projection neurons. To determine whether GDNF promotes retrograde survival over long axonal distances to cell bodies, we used a compartmentalized culture system. GDNF supported only modest and transient survival of postnatal sympathetic neurons when applied to their distal axons, in contrast to dorsal root ganglion (DRG) sensory neurons in which GDNF promoted survival equally well from either distal axons or cell bodies. Ret, the receptor tyrosine kinase for GDNF, underwent rapid proteasomal degradation in the axons of sympathetic neurons. Interestingly, the level of activated Ret in DRG neurons was sustained in the axons and also appeared in the cell bodies, suggesting that Ret was not degraded in sensory axons and was retrogradely transported. Pharmacologic inhibition of proteasomes only in the distal axons of sympathetic neurons caused an accumulation of activated Ret in both the axons and cell bodies during GDNF stimulation. Furthermore, exposure of the distal axons of sympathetic neurons to both GDNF and proteasome inhibitors, but neither one alone, promoted robust survival, identical to GDNF applied directly to the cell bodies. This differential responsiveness of sympathetic and sensory neurons to target-derived GDNF was attributable to the differential expression and degradation of the Ret9 and Ret51 isoforms. Therefore, the local degradation of Ret in axons dictates whether GDNF family ligands act as retrograde survival factors.
Collapse
|