1
|
Lucaj C, Pitha C, Davis J, Yano H. Assessment of high-efficacy agonism in synthetic cannabinoid receptor agonists containing l- tert-leucinate. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.11.617959. [PMID: 39464043 PMCID: PMC11507767 DOI: 10.1101/2024.10.11.617959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Synthetic cannabinoid receptor agonists (SCRAs) represent a class of new psychoactive substances that pose great health risks attributed to their wide-ranging and severe adverse effects. Recent evidence has shown that SCRAs with key moieties can confer superagonism, yet this phenomenon is still not well understood. In this study, we developed a structure-activity relationship (SAR) for SCRA superagonism by comparing eight compounds differing by their head moiety (l-valinate vs. l-tert-leucinate), core moiety (indole vs. indazole), and tail moiety (5-fluoropentyl vs. 4-fluorobenzyl) through different modes of bioluminescence resonance energy transfer (BRET). We found that l-tert-leucinate head moiety and indazole core moiety conferred superagonism across multiple Gαi/o proteins and β-arrestin 2. Finally, after generating CB1R mutant constructs, we found that transmembrane 2 (TM2) interactions to the head moiety of tested SCRAs at F170, F177, and H178 are key to eliciting activity.
Collapse
Affiliation(s)
- Christopher Lucaj
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Center for Drug Discovery, Northeastern University, Boston, Massachusetts 02115, United States
| | - Charlotte Pitha
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Center for Drug Discovery, Northeastern University, Boston, Massachusetts 02115, United States
| | - Jordan Davis
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Center for Drug Discovery, Northeastern University, Boston, Massachusetts 02115, United States
| | - Hideaki Yano
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Center for Drug Discovery, Northeastern University, Boston, Massachusetts 02115, United States
| |
Collapse
|
2
|
Nebuloni F, Do QB, Cook PR, Walsh EJ, Wade-Martins R. A fluid-walled microfluidic platform for human neuron microcircuits and directed axotomy. LAB ON A CHIP 2024; 24:3252-3264. [PMID: 38841815 PMCID: PMC11198392 DOI: 10.1039/d4lc00107a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 05/27/2024] [Indexed: 06/07/2024]
Abstract
In our brains, different neurons make appropriate connections; however, there remain few in vitro models of such circuits. We use an open microfluidic approach to build and study neuronal circuits in vitro in ways that fit easily into existing bio-medical workflows. Dumbbell-shaped circuits are built in minutes in standard Petri dishes; the aqueous phase is confined by fluid walls - interfaces between cell-growth medium and an immiscible fluorocarbon, FC40. Conditions are established that ensure post-mitotic neurons derived from human induced pluripotent stem cells (iPSCs) plated in one chamber of a dumbbell remain where deposited. After seeding cortical neurons on one side, axons grow through the connecting conduit to ramify amongst striatal neurons on the other - an arrangement mimicking unidirectional cortico-striatal connectivity. We also develop a moderate-throughput non-contact axotomy assay. Cortical axons in conduits are severed by a media jet; then, brain-derived neurotrophic factor and striatal neurons in distal chambers promote axon regeneration. As additional conduits and chambers are easily added, this opens up the possibility of mimicking complex neuronal networks, and screening drugs for their effects on connectivity.
Collapse
Affiliation(s)
- Federico Nebuloni
- Osney Thermofluids Institute, Department of Engineering Science, University of Oxford, Osney Mead, Oxford OX2 0ES, UK.
- The Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Quyen B Do
- Oxford Parkinson's Disease Centre and Department of Physiology, Anatomy and Genetics, University of Oxford, South Park Road, Oxford OX1 3QU, UK.
- Kavli Institute for Neuroscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Park Road, Oxford OX1 3QU, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Peter R Cook
- The Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Edmond J Walsh
- Osney Thermofluids Institute, Department of Engineering Science, University of Oxford, Osney Mead, Oxford OX2 0ES, UK.
| | - Richard Wade-Martins
- Oxford Parkinson's Disease Centre and Department of Physiology, Anatomy and Genetics, University of Oxford, South Park Road, Oxford OX1 3QU, UK.
- Kavli Institute for Neuroscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Park Road, Oxford OX1 3QU, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| |
Collapse
|
3
|
Dell’Oste V, Palego L, Betti L, Fantasia S, Gravina D, Bordacchini A, Pedrinelli V, Giannaccini G, Carmassi C. Plasma and Platelet Brain-Derived Neurotrophic Factor (BDNF) Levels in Bipolar Disorder Patients with Post-Traumatic Stress Disorder (PTSD) or in a Major Depressive Episode Compared to Healthy Controls. Int J Mol Sci 2024; 25:3529. [PMID: 38542503 PMCID: PMC10970837 DOI: 10.3390/ijms25063529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 04/04/2024] Open
Abstract
Post-traumatic stress disorder (PTSD) is a highly disabling mental disorder arising after traumatism exposure, often revealing critical and complex courses when comorbidity with bipolar disorder (BD) occurs. To search for PTSD or depression biomarkers that would help clinicians define BD presentations, this study aimed at preliminarily evaluating circulating brain-derived-neurotrophic factor (BDNF) levels in BD subjects with PTSD or experiencing a major depressive episode versus controls. Two bloodstream BDNF components were specifically investigated, the storage (intraplatelet) and the released (plasma) ones, both as adaptogenic/repair signals during neuroendocrine stress response dynamics. Bipolar patients with PTSD (n = 20) or in a major depressive episode (n = 20) were rigorously recruited together with unrelated healthy controls (n = 24) and subsequently examined by psychiatric questionnaires and blood samplings. Platelet-poor plasma (PPP) and intraplatelet (PLT) BDNF were measured by ELISA assays. The results showed markedly higher intraplatelet vs. plasma BDNF, confirming platelets' role in neurotrophin transport/storage. No between-group PPP-BDNF difference was reported, whereas PLT-BDNF was significantly reduced in depressed BD patients. PLT-BDNF negatively correlated with mood scores but not with PTSD items like PPP-BDNF, which instead displayed opposite correlation trends with depression and manic severity. Present findings highlight PLT-BDNF as more reliable at detecting depression than PTSD in BD, encouraging further study into BDNF variability contextually with immune-inflammatory parameters in wider cohorts of differentially symptomatic bipolar patients.
Collapse
Affiliation(s)
- Valerio Dell’Oste
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (L.P.); (S.F.); (D.G.); (A.B.); (V.P.); (C.C.)
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
- UFCSMA Zona Valdinievole, Azienda USL Toscana Centro, 51016 Montecatini Terme, Italy
| | - Lionella Palego
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (L.P.); (S.F.); (D.G.); (A.B.); (V.P.); (C.C.)
- Department of Pharmacy, Section of Biochemistry, University of Pisa, 56126 Pisa, Italy; (L.B.); (G.G.)
| | - Laura Betti
- Department of Pharmacy, Section of Biochemistry, University of Pisa, 56126 Pisa, Italy; (L.B.); (G.G.)
| | - Sara Fantasia
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (L.P.); (S.F.); (D.G.); (A.B.); (V.P.); (C.C.)
| | - Davide Gravina
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (L.P.); (S.F.); (D.G.); (A.B.); (V.P.); (C.C.)
| | - Andrea Bordacchini
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (L.P.); (S.F.); (D.G.); (A.B.); (V.P.); (C.C.)
| | - Virginia Pedrinelli
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (L.P.); (S.F.); (D.G.); (A.B.); (V.P.); (C.C.)
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
- UFSMA Zona Apuana, Azienda USL Toscana Nord Ovest, 54100 Massa, Italy
| | - Gino Giannaccini
- Department of Pharmacy, Section of Biochemistry, University of Pisa, 56126 Pisa, Italy; (L.B.); (G.G.)
| | - Claudia Carmassi
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (L.P.); (S.F.); (D.G.); (A.B.); (V.P.); (C.C.)
| |
Collapse
|
4
|
Xiao J. Thirty years of BDNF study in central myelination: From biology to therapy. J Neurochem 2023; 167:321-336. [PMID: 37747083 DOI: 10.1111/jnc.15968] [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: 07/29/2023] [Revised: 09/05/2023] [Accepted: 09/07/2023] [Indexed: 09/26/2023]
Abstract
Being the highest expressed neurotrophin in the mammalian brain, the brain-derived neurotrophic factor (BDNF) is essential to neural development and plasticity in both health and diseases. Following the discovery of BDNF by Yves-Alain Barde in 1982, the main feature of BDNF's activity in myelination was first described by Cellerino et al. in 1997. Since then, genetic manipulation of the BDNF-encoding gene and its receptors in murine models has revealed the contribution of BDNF to the myelinating process in the central nervous system (CNS). The series of BDNF or receptor mouse mutants as well as the BDNF polymorphism in humans have provided new insights into the roles that BDNF signaling plays in myelination in a complex manner. 2024 marks the 30th year of BDNF's research in myelination. Here, we share our perspective on the 30-year history of BDNF in the field of CNS myelination from phenotyping to therapeutic development, focusing on genetic evidence regarding the mechanism by which BDNF regulates myelin formation and repair in the CNS. This review also discusses the current hypotheses of BDNF's action on CNS myelination: axonal- and oligodendroglial-driven mechanisms, which may be ultimately activity-dependent. Last, this review raises the challenges and opportunities of developing BDNF-based therapies for neurodegenerative diseases, opening unanswered questions for future investigation.
Collapse
Affiliation(s)
- Junhua Xiao
- School of Health Sciences, Swinburne University of Technology, Hawthorn, Victoria, Australia
- School of Allied Health, La Trobe University, Bundoora, Victoria, Australia
| |
Collapse
|
5
|
Vints WAJ, Gökçe E, Langeard A, Pavlova I, Çevik ÖS, Ziaaldini MM, Todri J, Lena O, Sakkas GK, Jak S, Zorba (Zormpa) I, Karatzaferi C, Levin O, Masiulis N, Netz Y. Myokines as mediators of exercise-induced cognitive changes in older adults: protocol for a comprehensive living systematic review and meta-analysis. Front Aging Neurosci 2023; 15:1213057. [PMID: 37520128 PMCID: PMC10374322 DOI: 10.3389/fnagi.2023.1213057] [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: 04/27/2023] [Accepted: 06/20/2023] [Indexed: 08/01/2023] Open
Abstract
Background The world's population is aging, but life expectancy has risen more than healthy life expectancy (HALE). With respect to brain and cognition, the prevalence of neurodegenerative disorders increases with age, affecting health and quality of life, and imposing significant healthcare costs. Although the effects of physical exercise on cognition in advanced age have been widely explored, in-depth fundamental knowledge of the underlying mechanisms of the exercise-induced cognitive improvements is lacking. Recent research suggests that myokines, factors released into the blood circulation by contracting skeletal muscle, may play a role in mediating the beneficial effect of exercise on cognition. Our goal in this ongoing (living) review is to continuously map the rapidly accumulating knowledge on pathways between acute or chronic exercise-induced myokines and cognitive domains enhanced by exercise. Method Randomized controlled studies will be systematically collected at baseline and every 6 months for at least 5 years. Literature search will be performed online in PubMed, EMBASE, PsycINFO, Web of Science, SportDiscus, LILACS, IBECS, CINAHL, SCOPUS, ICTRP, and ClinicalTrials.gov. Risk of bias will be assessed using the Revised Cochrane Risk of Bias tool (ROB 2). A random effects meta-analysis with mediation analysis using meta-analytic structural equation modeling (MASEM) will be performed. The primary research question is to what extent exercise-induced myokines serve as mediators of cognitive function. Secondarily, the pooled effect size of specific exercise characteristics (e.g., mode of exercise) or specific older adults' populations (e.g., cognitively impaired) on the relationship between exercise, myokines, and cognition will be assessed. The review protocol was registered in PROSPERO (CRD42023416996). Discussion Understanding the triad relationship between exercise, myokines and cognition will expand the knowledge on multiple integrated network systems communicating between skeletal muscles and other organs such as the brain, thus mediating the beneficial effects of exercise on health and performance. It may also have practical implications, e.g., if a certain myokine is found to be a mediator between exercise and cognition, the optimal exercise characteristics for inducing this myokine can be prescribed. The living review is expected to improve our state of knowledge and refine exercise regimes for enhancing cognitive functioning in diverse older adults' populations. Registration Systematic review and meta-analysis protocol was registered with the International Prospective Register of Systematic Reviews (PROSPERO) on the 24th of April 2023 (registration number CRD42023416996).
Collapse
Affiliation(s)
- Wouter A. J. Vints
- Department of Health Promotion and Rehabilitation, Lithuanian Sports University, Kaunas, Lithuania
- Department of Rehabilitation Medicine, Research School Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, Netherlands
- Adelante Zorggroep Centre of Expertise in Rehabilitation and Audiology, Hoensbroek, Netherlands
| | - Evrim Gökçe
- Sports Rehabilitation Laboratory, Ankara City Hospital, Ankara, Türkiye
| | | | - Iuliia Pavlova
- Department of Theory and Methods of Physical Culture, Lviv State University of Physical Culture, Lviv, Ukraine
| | | | | | - Jasemin Todri
- Department of Physiotherapy, Universidad Catolica San Antonio (UCAM), Murcia, Spain
| | - Orges Lena
- Department of Physiotherapy, Universidad Catolica San Antonio (UCAM), Murcia, Spain
| | - Giorgos K. Sakkas
- Lifestyle Medicine and Experimental Physiology and Myology Lab, Department of Physical Education and Sports Science, The Center of Research and Evaluation of Human Performance (CREHP), University of Thessaly, National and Kapodistrian University of Athens (TEFAA) Campus, Karyes, Greece
| | - Suzanne Jak
- Research Institute of Child Development and Education, University of Amsterdam, Amsterdam, Netherlands
| | | | - Christina Karatzaferi
- Lifestyle Medicine and Experimental Physiology and Myology Lab, Department of Physical Education and Sports Science, The Center of Research and Evaluation of Human Performance (CREHP), University of Thessaly, National and Kapodistrian University of Athens (TEFAA) Campus, Karyes, Greece
| | - Oron Levin
- Department of Health Promotion and Rehabilitation, Lithuanian Sports University, Kaunas, Lithuania
- Movement Control and Neuroplasticity Research Group, Group Biomedical Sciences, Catholic University of Leuven, Heverlee, Belgium
| | - Nerijus Masiulis
- Department of Health Promotion and Rehabilitation, Lithuanian Sports University, Kaunas, Lithuania
| | - Yael Netz
- Department of Health Promotion and Rehabilitation, Lithuanian Sports University, Kaunas, Lithuania
- The Levinsky-Wingate Academic Center, Wingate Campus, Netanya, Israel
| |
Collapse
|
6
|
Shi ZL, Fan ZY, Zhang H, Li ST, Yuan H, Tong JH. Localized delivery of brain-derived neurotrophic factor from PLGA microspheres promotes peripheral nerve regeneration in rats. J Orthop Surg Res 2022; 17:172. [PMID: 35303915 PMCID: PMC8931983 DOI: 10.1186/s13018-022-02985-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 02/03/2022] [Indexed: 01/29/2023] Open
Abstract
Background Repair of peripheral nerve defect presents a considerable challenge for reconstructive surgeons. The aim of this study is to develop a brain-derived neurotrophic factor (BDNF) from poly(D,L-lactide-co-glycolide) (PLGA) microspheres for the treatment of the peripheral nerve defect. Method BDNF microspheres were prepared by using an oil-in-water emulsification-solvent evaporation method. The morphology, particle size, encapsulation efficiency, drug loading and sustained release performance of microspheres was observed and calculated. Adipose mesenchymal stem cells (ADSCs) were isolated and expanded. ADSCs were divided into four groups: control, BDNF, blank microsphere and BDNF microsphere groups. Cell count kit-8 (CCK-8) assays were used to assess cell proliferation. Cell migration was determined by Transwell assays. Twenty-eight male Sprague–Dawley rats underwent transection damage model on the right sciatic nerve. The wet weight ratio of the gastrocnemius muscle was calculated by comparing the weight of the gastrocnemius muscle from the operated side to that of the normal side. Neuroelectrophysiological testing was performed to assess nerve function recovery. Nerve regeneration was evaluated by histological analysis and immunohistochemical staining. Results The microspheres were spherical and had uniform size (46.38 ± 1.00 μm), high encapsulation efficiency and high loading capacity. In vitro release studies showed that BDNF-loaded microspheres had good sustained release characteristics. The duration of BDNF release was extended to more than 50 days. BDNF or BDNF microsphere promote the proliferation and migration of ADSCs than control group (P < 0.05). Compared with control group, BDNF significantly decreased the nerve conduction velocity (NCV) and compound amplitude (AMP) (P < 0.05). The nerve fibers in the BDNF microsphere group were closely arranged and uniformly distributed than control group. Conclusion BDNF/PLGA sustained-release microsphere could promote the migration of ADSCs and promoted neural differentiation of ADSCs. Moreover, BDNF/PLGA sustained-release microsphere ameliorated nerve conduction velocity and prevented neuralgic amyotrophy.
Collapse
Affiliation(s)
- Zheng-Liang Shi
- Department of Orthopedics, The Second Hospital of Hebei Medical University, No. 215, Hepingxi Road, Shijiazhuang, 050000, Hebei Province, China
| | - Zhi-Yong Fan
- Department of Orthopedics, The Second Hospital of Hebei Medical University, No. 215, Hepingxi Road, Shijiazhuang, 050000, Hebei Province, China.
| | - Hua Zhang
- Department of Orthopedics, The Second Hospital of Hebei Medical University, No. 215, Hepingxi Road, Shijiazhuang, 050000, Hebei Province, China
| | - Shen-Tai Li
- Department of Orthopedics, The Second Hospital of Hebei Medical University, No. 215, Hepingxi Road, Shijiazhuang, 050000, Hebei Province, China
| | - He Yuan
- Department of Orthopedics, The Second Hospital of Hebei Medical University, No. 215, Hepingxi Road, Shijiazhuang, 050000, Hebei Province, China
| | - Jiu-Hui Tong
- Department of Orthopedics, The Second Hospital of Hebei Medical University, No. 215, Hepingxi Road, Shijiazhuang, 050000, Hebei Province, China
| |
Collapse
|
7
|
Servetti M, Pisciotta L, Tassano E, Cerminara M, Nobili L, Boeri S, Rosti G, Lerone M, Divizia MT, Ronchetto P, Puliti A. Neurodevelopmental Disorders in Patients With Complex Phenotypes and Potential Complex Genetic Basis Involving Non-Coding Genes, and Double CNVs. Front Genet 2021; 12:732002. [PMID: 34621295 PMCID: PMC8490884 DOI: 10.3389/fgene.2021.732002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/03/2021] [Indexed: 12/15/2022] Open
Abstract
Neurodevelopmental disorders (NDDs) are a heterogeneous class of brain diseases, with a complex genetic basis estimated to account for up to 50% of cases. Nevertheless, genetic diagnostic yield is about 20%. Array-comparative genomic hybridization (array-CGH) is an established first-level diagnostic test able to detect pathogenic copy number variants (CNVs), however, most identified variants remain of uncertain significance (VUS). Failure of interpretation of VUSs may depend on various factors, including complexity of clinical phenotypes and inconsistency of genotype-phenotype correlations. Indeed, although most NDD-associated CNVs are de novo, transmission from unaffected parents to affected children of CNVs with high risk for NDDs has been observed. Moreover, variability of genetic components overlapped by CNVs, such as long non-coding genes, genomic regions with long-range effects, and additive effects of multiple CNVs can make CNV interpretation challenging. We report on 12 patients with complex phenotypes possibly explained by complex genetic mechanisms, including involvement of antisense genes and boundaries of topologically associating domains. Eight among the 12 patients carried two CNVs, either de novo or inherited, respectively, by each of their healthy parents, that could additively contribute to the patients’ phenotype. CNVs overlapped either known NDD-associated or novel candidate genes (PTPRD, BUD13, GLRA3, MIR4465, ABHD4, and WSCD2). Bioinformatic enrichment analyses showed that genes overlapped by the co-occurring CNVs have synergistic roles in biological processes fundamental in neurodevelopment. Double CNVs could concur in producing deleterious effects, according to a two-hit model, thus explaining the patients’ phenotypes and the incomplete penetrance, and variable expressivity, associated with the single variants. Overall, our findings could contribute to the knowledge on clinical and genetic diagnosis of complex forms of NDD.
Collapse
Affiliation(s)
- Martina Servetti
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), University of Genoa, Genoa, Italy.,Medical Genetics Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Livia Pisciotta
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), University of Genoa, Genoa, Italy.,Child Neuropsychiatry Unit, ASST Fatebenefratelli Sacco, Milano, Italy
| | - Elisa Tassano
- Human Genetics Laboratory, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Maria Cerminara
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), University of Genoa, Genoa, Italy
| | - Lino Nobili
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), University of Genoa, Genoa, Italy.,Child Neuropsychiatry Unit, Istituto Giannina Gaslini, Genoa, Italy
| | - Silvia Boeri
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), University of Genoa, Genoa, Italy.,Child Neuropsychiatry Unit, Istituto Giannina Gaslini, Genoa, Italy
| | - Giulia Rosti
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), University of Genoa, Genoa, Italy
| | - Margherita Lerone
- Medical Genetics Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | | | - Patrizia Ronchetto
- Human Genetics Laboratory, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Aldamaria Puliti
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), University of Genoa, Genoa, Italy.,Medical Genetics Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| |
Collapse
|
8
|
Liu C, Xu Y, Yang H, Zhang J. Establishment of axon regeneration regulatory network and the role of low intensity pulsed ultrasound in the network. Saudi J Biol Sci 2020; 26:1922-1926. [PMID: 31889775 PMCID: PMC6923491 DOI: 10.1016/j.sjbs.2019.07.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 07/11/2019] [Accepted: 07/13/2019] [Indexed: 11/28/2022] Open
Abstract
Objective To establish an axon regeneration regulatory network for optimal selection, and explore the role of low intensity pulsed ultrasound in the network. Methods The axon regeneration regulatory network involving axon regeneration-related proteins NGF, BDNF and PirB was constructed by using GO and KEGG. The maximum possible pathway acting on axon regeneration was screened by Bayesian network theory. The node of low - intensity pulsed ultrasound in NGF - involved axon regeneration network was complemented by combining literature methods. Results The NGF, BDNF and PirB-involved axonal regeneration regulatory pathway was successfully constructed. The low intensity pulsed ultrasound played a role in axon regeneration by acting on ERK1/2-CREB pathway and GSK-3β. NGF-TrKA-Rap1-ERK1/2-CREB-Bcl-2 was optimized as optimal pathway by Bayesian theory. Conclusion The regulatory pathway of axon regeneration involving nerve growth related factors and low intensity pulsed ultrasound was initially established, which provided a theoretical basis for further study of axon regeneration, and also new ideas for action of low intensity pulsed ultrasound on axon regeneration regulatory pathway.
Collapse
Affiliation(s)
- Chunyang Liu
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yanhua Xu
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Hong Yang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Jianhua Zhang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| |
Collapse
|
9
|
Chen HI, Jgamadze D, Lim J, Mensah-Brown K, Wolf JA, Mills JA, Smith DH. Functional Cortical Axon Tracts Generated from Human Stem Cell-Derived Neurons. Tissue Eng Part A 2019; 25:736-745. [PMID: 30648482 DOI: 10.1089/ten.tea.2018.0270] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
IMPACT STATEMENT Axon regeneration is negligible in the adult mammalian brain, and thus, white matter damage often leads to permanent neurological deficits. A novel approach for axon repair is the generation of axon tracts in the laboratory setting followed by transplantation of these constructs. This article details a human substrate for this repair strategy. Using the technique of axon stretch growth, functional cortical axon tracts are generated from human pluripotent stem cells at rates of up to 1 mm/day. These results form the basis of a potential patient-specific protocol for cerebral axon transplantation after injury.
Collapse
Affiliation(s)
- H Isaac Chen
- 1 Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,2 Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania
| | - Dennis Jgamadze
- 1 Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - James Lim
- 1 Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kobina Mensah-Brown
- 1 Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - John A Wolf
- 1 Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,2 Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania
| | - Jason A Mills
- 3 Center for Advanced Retinal and Ocular Therapeutics, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Douglas H Smith
- 1 Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
10
|
Beker MC, Caglayan B, Yalcin E, Caglayan AB, Turkseven S, Gurel B, Kelestemur T, Sertel E, Sahin Z, Kutlu S, Kilic U, Baykal AT, Kilic E. Time-of-Day Dependent Neuronal Injury After Ischemic Stroke: Implication of Circadian Clock Transcriptional Factor Bmal1 and Survival Kinase AKT. Mol Neurobiol 2018; 55:2565-2576. [PMID: 28421530 DOI: 10.1007/s12035-017-0524-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 04/06/2017] [Indexed: 12/14/2022]
Abstract
Occurrence of stroke cases displays a time-of-day variation in human. However, the mechanism linking circadian rhythm to the internal response mechanisms against pathophysiological events after ischemic stroke remained largely unknown. To this end, temporal changes in the susceptibility to ischemia/reperfusion (I/R) injury were investigated in mice in which the ischemic stroke induced at four different Zeitgeber time points with 6-h intervals (ZT0, ZT6, ZT12, and ZT18). Besides infarct volume and brain swelling, neuronal survival, apoptosis, ischemia, and circadian rhythm related proteins were examined using immunohistochemistry, Western blot, planar surface immune assay, and liquid chromatography-mass spectrometry tools. Here, we present evidence that midnight (ZT18; 24:00) I/R injury in mice resulted in significantly improved infarct volume, brain swelling, neurological deficit score, neuronal survival, and decreased apoptotic cell death compared with ischemia induced at other time points, which were associated with increased expressions of circadian proteins Bmal1, PerI, and Clock proteins and survival kinases AKT and Erk-1/2. Moreover, ribosomal protein S6, mTOR, and Bad were also significantly increased, while the levels of PRAS40, negative regulator of AKT and mTOR, and phosphorylated p53 were decreased at this time point compared to ZT0 (06:00). Furthermore, detailed proteomic analysis revealed significantly decreased CSKP, HBB-1/2, and HBA levels, while increased GNAZ, NEGR1, IMPCT, and PDE1B at midnight as compared with early morning. Our results indicate that nighttime I/R injury results in less severe neuronal damage, with increased neuronal survival, increased levels of survival kinases and circadian clock proteins, and also alters the circadian-related proteins.
Collapse
Affiliation(s)
- Mustafa Caglar Beker
- Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Ekinciler Cad. 19, TR-34810, Istanbul, Turkey
- Department of Physiology, Istanbul Medipol University, 34810, Istanbul, Turkey
- Department of Physiology, Necmettin Erbakan University, 42060, Konya, Turkey
| | - Berrak Caglayan
- Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Ekinciler Cad. 19, TR-34810, Istanbul, Turkey
- Department of Physiology, Istanbul Medipol University, 34810, Istanbul, Turkey
| | - Esra Yalcin
- Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Ekinciler Cad. 19, TR-34810, Istanbul, Turkey
- Department of Physiology, Istanbul Medipol University, 34810, Istanbul, Turkey
| | - Ahmet Burak Caglayan
- Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Ekinciler Cad. 19, TR-34810, Istanbul, Turkey
- Department of Physiology, Istanbul Medipol University, 34810, Istanbul, Turkey
- Department of Physiology, Necmettin Erbakan University, 42060, Konya, Turkey
| | - Seyma Turkseven
- Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Ekinciler Cad. 19, TR-34810, Istanbul, Turkey
| | - Busra Gurel
- Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Ekinciler Cad. 19, TR-34810, Istanbul, Turkey
- School of Medicine, Department of Medical Biochemistry, Acibadem University, 34752, Istanbul, Turkey
| | - Taha Kelestemur
- Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Ekinciler Cad. 19, TR-34810, Istanbul, Turkey
- Department of Physiology, Istanbul Medipol University, 34810, Istanbul, Turkey
- Department of Physiology, Necmettin Erbakan University, 42060, Konya, Turkey
| | - Elif Sertel
- Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Ekinciler Cad. 19, TR-34810, Istanbul, Turkey
- Department of Physiology, Istanbul Medipol University, 34810, Istanbul, Turkey
| | - Zafer Sahin
- Department of Physiotherapy and Rehabilitation, Necmettin Erbakan University, 42060, Konya, Turkey
| | - Selim Kutlu
- Department of Physiology, Necmettin Erbakan University, 42060, Konya, Turkey
| | - Ulkan Kilic
- Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Ekinciler Cad. 19, TR-34810, Istanbul, Turkey
| | - Ahmet Tarik Baykal
- School of Medicine, Department of Medical Biochemistry, Acibadem University, 34752, Istanbul, Turkey
| | - Ertugrul Kilic
- Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Ekinciler Cad. 19, TR-34810, Istanbul, Turkey.
- Department of Physiology, Istanbul Medipol University, 34810, Istanbul, Turkey.
| |
Collapse
|
11
|
Gnaz couples the circadian and dopaminergic system to G protein-mediated signaling in mouse photoreceptors. PLoS One 2017; 12:e0187411. [PMID: 29088301 PMCID: PMC5663513 DOI: 10.1371/journal.pone.0187411] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 10/19/2017] [Indexed: 01/08/2023] Open
Abstract
The mammalian retina harbors a circadian clockwork that regulates vision and promotes healthiness of retinal neurons, mainly through directing the rhythmic release of the neurohormones dopamine—acting on dopamine D4 receptors—and melatonin—acting on MT1 and MT2 receptors. The gene Gnaz—a unique Gi/o subfamily member—was seen in the present study to be expressed in photoreceptors where its protein product Gαz shows a daily rhythm in its subcellular localization. Apart from subcellular localization, Gnaz displays a daily rhythm in expression—with peak values at night—in preparations of the whole retina, microdissected photoreceptors and photoreceptor-related pinealocytes. In retina, Gnaz rhythmicity was observed to persist under constant darkness and to be abolished in retina deficient for Clock or dopamine D4 receptors. Furthermore, circadian regulation of Gnaz was disturbed in the db/db mouse, a model of diabetic retinopathy. The data of the present study suggest that Gnaz links the circadian clockwork—via dopamine acting on D4 receptors—to G protein-mediated signaling in intact but not diabetic retina.
Collapse
|
12
|
Fenske RJ, Cadena MT, Harenda QE, Wienkes HN, Carbajal K, Schaid MD, Laundre E, Brill AL, Truchan NA, Brar H, Wisinski J, Cai J, Graham TE, Engin F, Kimple ME. The Inhibitory G Protein α-Subunit, Gαz, Promotes Type 1 Diabetes-Like Pathophysiology in NOD Mice. Endocrinology 2017; 158:1645-1658. [PMID: 28419211 PMCID: PMC5460933 DOI: 10.1210/en.2016-1700] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 04/11/2017] [Indexed: 01/23/2023]
Abstract
The α-subunit of the heterotrimeric Gz protein, Gαz, promotes β-cell death and inhibits β-cell replication when pancreatic islets are challenged by stressors. Thus, we hypothesized that loss of Gαz protein would preserve functional β-cell mass in the nonobese diabetic (NOD) model, protecting from overt diabetes. We saw that protection from diabetes was robust and durable up to 35 weeks of age in Gαz knockout mice. By 17 weeks of age, Gαz-null NOD mice had significantly higher diabetes-free survival than wild-type littermates. Islets from these mice had reduced markers of proinflammatory immune cell infiltration on both the histological and transcript levels and secreted more insulin in response to glucose. Further analyses of pancreas sections revealed significantly fewer terminal deoxynucleotidyltransferase-mediated dUTP nick end labeling (TUNEL)-positive β-cells in Gαz-null islets despite similar immune infiltration in control mice. Islets from Gαz-null mice also exhibited a higher percentage of Ki-67-positive β-cells, a measure of proliferation, even in the presence of immune infiltration. Finally, β-cell-specific Gαz-null mice phenocopy whole-body Gαz-null mice in their protection from developing hyperglycemia after streptozotocin administration, supporting a β-cell-centric role for Gαz in diabetes pathophysiology. We propose that Gαz plays a key role in β-cell signaling that becomes dysfunctional in the type 1 diabetes setting, accelerating the death of β-cells, which promotes further accumulation of immune cells in the pancreatic islets, and inhibiting a restorative proliferative response.
Collapse
MESH Headings
- Animals
- Apoptosis/genetics
- Blood Glucose/metabolism
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/pathology
- Female
- GTP-Binding Protein alpha Subunits/genetics
- Insulin-Secreting Cells/metabolism
- Insulin-Secreting Cells/physiology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, Knockout
- Mice, Transgenic
- Streptozocin
Collapse
Affiliation(s)
- Rachel J. Fenske
- Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
| | - Mark T. Cadena
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - Quincy E. Harenda
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - Haley N. Wienkes
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - Kathryn Carbajal
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - Michael D. Schaid
- Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
| | - Erin Laundre
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - Allison L. Brill
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - Nathan A. Truchan
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - Harpreet Brar
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - Jaclyn Wisinski
- Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - Jinjin Cai
- Molecular Medicine Program, Department of Medicine, Division of Endocrinology, Metabolism, and Diabetes, Department of Nutrition, and Department of Biological Chemistry, University of Utah School of Medicine, Salt Lake City, Utah 84112
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah 84112
| | - Timothy E. Graham
- Molecular Medicine Program, Department of Medicine, Division of Endocrinology, Metabolism, and Diabetes, Department of Nutrition, and Department of Biological Chemistry, University of Utah School of Medicine, Salt Lake City, Utah 84112
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah 84112
| | - Feyza Engin
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - Michelle E. Kimple
- Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin 53705
| |
Collapse
|