1
|
Singh AK, Joshi I, Reddy NMN, Purushotham SS, Eswaramoorthy M, Vasudevan M, Banerjee S, Clement JP, Kundu TK. Epigenetic modulation rescues neurodevelopmental deficits in Syngap1 +/- mice. Aging Cell 2025; 24:e14408. [PMID: 39878322 PMCID: PMC11896221 DOI: 10.1111/acel.14408] [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: 06/12/2024] [Revised: 09/05/2024] [Accepted: 10/22/2024] [Indexed: 01/31/2025] Open
Abstract
SYNGAP1 is a Ras GTPase-activating protein that plays a crucial role during brain development and in synaptic plasticity. Sporadic heterozygous mutations in SYNGAP1 affect social and emotional behaviour observed in intellectual disability (ID) and autism spectrum disorder (ASD). Although neurophysiological deficits have been extensively studied, the epigenetic landscape of SYNGAP1 mutation-mediated intellectual disability is unexplored. Here, we have found that the p300/CBP specific acetylation marks of histones are significantly repressed in the hippocampus of adolescent Syngap1+/- mice. Additionally, we observed decreased dendritic branching of newly born DCX+ neurons in these mice, suggesting altered adult hippocampal neurogenesis. To establish the causal relationship of Syngap1+/- phenotype and the altered histone acetylation signature we have treated 2-4 months old Syngap1+/- mice with glucose-derived carbon nanosphere (CSP) conjugated potent small molecule activator (TTK21) of p300/CBP lysine acetyltransferase (CSP-TTK21). The enhancement of the p300/CBP specific acetylation marks of histones by CSP-TTK21 restored synaptic functions, increased dendritic branching of DCX+ neurons, enables the capability to reorganise cortical circuits in response to change in the sensory stimuli, and improves behavioural measures in Syngap1+/- mice that are very closely comparable to wild type littermates. Further, hippocampal RNA-Seq analysis of these mice revealed that the expression of many critical genes such as Adcy1, Ntrk3, Egr1, and Foxj1 which are key regulators of synaptic plasticity and neurogenesis and are well associated with ID/ASD reversed upon CSP-TTK21 treatment. This study could be the first demonstration of the reversal of autistic behaviour and neural wiring upon the modulation of altered epigenetic modification(s).
Collapse
Affiliation(s)
- Akash Kumar Singh
- Molecular Biology and Genetics Unit, Transcription and Disease LaboratoryJawaharlal Nehru Centre for Advanced Scientific ResearchBengaluruIndia
- Neuroscience UnitJawaharlal Nehru Centre for Advanced Scientific ResearchBangaloreIndia
| | - Ila Joshi
- Molecular Biology and Genetics Unit, Transcription and Disease LaboratoryJawaharlal Nehru Centre for Advanced Scientific ResearchBengaluruIndia
| | - Neeharika M. N. Reddy
- Neuroscience UnitJawaharlal Nehru Centre for Advanced Scientific ResearchBangaloreIndia
| | | | - M. Eswaramoorthy
- Chemistry and Physics of Materials UnitJawaharlal Nehru Centre for Advanced Scientific ResearchBengaluruIndia
| | | | | | - James P. Clement
- Neuroscience UnitJawaharlal Nehru Centre for Advanced Scientific ResearchBangaloreIndia
- Present address:
University of ExeterExeterUK
| | - Tapas K. Kundu
- Molecular Biology and Genetics Unit, Transcription and Disease LaboratoryJawaharlal Nehru Centre for Advanced Scientific ResearchBengaluruIndia
- Neuroscience UnitJawaharlal Nehru Centre for Advanced Scientific ResearchBangaloreIndia
| |
Collapse
|
2
|
Wei X, Browning JL, Olsen ML. Neuron and astrocyte specific 5mC and 5hmC signatures of BDNF's receptor, TrkB. Front Mol Neurosci 2024; 17:1463437. [PMID: 39268252 PMCID: PMC11390696 DOI: 10.3389/fnmol.2024.1463437] [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: 07/11/2024] [Accepted: 08/16/2024] [Indexed: 09/15/2024] Open
Abstract
Brain derived neurotrophic factor (BDNF) is the most studied trophic factor in the central nervous system (CNS), and its role in the maturation of neurons, including synapse development and maintenance has been investigated intensely for over three decades. The primary receptor for BDNF is the tropomyosin receptor kinase B (TrkB), which is broadly expressed as two primary isoforms in the brain; the full length TrkB (TrkB.FL) receptor, expressed mainly in neurons and the truncated TrkB (TrkB.T1) receptor. We recently demonstrated that TrkB.T1 is predominately expressed in astrocytes, and appears critical for astrocyte morphological maturation. Given the critical role of BDNF/TrkB pathway in healthy brain development and mature CNS function, we aimed to identify molecular underpinnings of cell-type specific expression of each TrkB isoform. Using Nanopore sequencing which enables direct, long read sequencing of native DNA, we profiled DNA methylation patterns of the entire TrkB gene, Ntrk2, in both neurons and astrocytes. Here, we identified robust differences in cell-type specific isoform expression associated with significantly different methylation patterns of the Ntrk2 gene in each cell type. Notably, astrocytes demonstrated lower 5mC methylation, and higher 5hmC across the entire gene when compared to neurons, including differentially methylated sites (DMSs) found in regions flanking the unique TrkB.T1 protein coding sequence (CDS). These data suggest DNA methylation patterns may provide instruction for isoform specific TrkB expression across unique CNS cell types.
Collapse
Affiliation(s)
- Xiaoran Wei
- Biomedical and Veterinary Sciences Graduate Program, Virginia Tech, Blacksburg, VA, United States
- School of Neuroscience, Virginia Tech, Blacksburg, VA, United States
| | - Jack L. Browning
- School of Neuroscience, Virginia Tech, Blacksburg, VA, United States
- Genetics, Bioinformatics and Computational Biology Graduate Program, Virginia Tech, Blacksburg, VA, United States
| | - Michelle L. Olsen
- School of Neuroscience, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
3
|
Talevi A, Bellera C. An update on the novel methods for the discovery of antiseizure and antiepileptogenic medications: where are we in 2024? Expert Opin Drug Discov 2024; 19:975-990. [PMID: 38963148 DOI: 10.1080/17460441.2024.2373165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 06/23/2024] [Indexed: 07/05/2024]
Abstract
INTRODUCTION Despite the availability of around 30 antiseizure medications, 1/3 of patients with epilepsy fail to become seizure-free upon pharmacological treatment. Available medications provide adequate symptomatic control in two-thirds of patients, but disease-modifying drugs are still scarce. Recently, though, new paradigms have been explored. AREAS COVERED Three areas are reviewed in which a high degree of innovation in the search for novel antiseizure and antiepileptogenic medications has been implemented: development of novel screening approaches, search for novel therapeutic targets, and adoption of new drug discovery paradigms aligned with a systems pharmacology perspective. EXPERT OPINION In the past, worldwide leaders in epilepsy have reiteratively stated that the lack of progress in the field may be explained by the recurrent use of the same molecular targets and screening procedures to identify novel medications. This landscape has changed recently, as reflected by the new Epilepsy Therapy Screening Program and the introduction of many in vitro and in vivo models that could possibly improve our chances of identifying first-in-class medications that may control drug-resistant epilepsy or modify the course of disease. Other milestones include the study of new molecular targets for disease-modifying drugs and exploration of a systems pharmacology perspective to design new drugs.
Collapse
Affiliation(s)
- Alan Talevi
- Laboratory of Bioactive Compound Research and Development (LIDeB), Faculty of Exact Sciences, University of La Plata (UNLP), La Plata, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), CCT La Plata, La Plata, Argentina
| | - Carolina Bellera
- Laboratory of Bioactive Compound Research and Development (LIDeB), Faculty of Exact Sciences, University of La Plata (UNLP), La Plata, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), CCT La Plata, La Plata, Argentina
| |
Collapse
|
4
|
Merighi A. Brain-Derived Neurotrophic Factor, Nociception, and Pain. Biomolecules 2024; 14:539. [PMID: 38785946 PMCID: PMC11118093 DOI: 10.3390/biom14050539] [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: 02/08/2024] [Revised: 04/26/2024] [Accepted: 04/26/2024] [Indexed: 05/25/2024] Open
Abstract
This article examines the involvement of the brain-derived neurotrophic factor (BDNF) in the control of nociception and pain. BDNF, a neurotrophin known for its essential role in neuronal survival and plasticity, has garnered significant attention for its potential implications as a modulator of synaptic transmission. This comprehensive review aims to provide insights into the multifaceted interactions between BDNF and pain pathways, encompassing both physiological and pathological pain conditions. I delve into the molecular mechanisms underlying BDNF's involvement in pain processing and discuss potential therapeutic applications of BDNF and its mimetics in managing pain. Furthermore, I highlight recent advancements and challenges in translating BDNF-related research into clinical practice.
Collapse
Affiliation(s)
- Adalberto Merighi
- Department of Veterinary Sciences, University of Turin, 10095 Turin, Italy
| |
Collapse
|
5
|
Xiong HY, Hendrix J, Schabrun S, Wyns A, Campenhout JV, Nijs J, Polli A. The Role of the Brain-Derived Neurotrophic Factor in Chronic Pain: Links to Central Sensitization and Neuroinflammation. Biomolecules 2024; 14:71. [PMID: 38254671 PMCID: PMC10813479 DOI: 10.3390/biom14010071] [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: 11/30/2023] [Revised: 01/02/2024] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
Chronic pain is sustained, in part, through the intricate process of central sensitization (CS), marked by maladaptive neuroplasticity and neuronal hyperexcitability within central pain pathways. Accumulating evidence suggests that CS is also driven by neuroinflammation in the peripheral and central nervous system. In any chronic disease, the search for perpetuating factors is crucial in identifying therapeutic targets and developing primary preventive strategies. The brain-derived neurotrophic factor (BDNF) emerges as a critical regulator of synaptic plasticity, serving as both a neurotransmitter and neuromodulator. Mounting evidence supports BDNF's pro-nociceptive role, spanning from its pain-sensitizing capacity across multiple levels of nociceptive pathways to its intricate involvement in CS and neuroinflammation. Moreover, consistently elevated BDNF levels are observed in various chronic pain disorders. To comprehensively understand the profound impact of BDNF in chronic pain, we delve into its key characteristics, focusing on its role in underlying molecular mechanisms contributing to chronic pain. Additionally, we also explore the potential utility of BDNF as an objective biomarker for chronic pain. This discussion encompasses emerging therapeutic approaches aimed at modulating BDNF expression, offering insights into addressing the intricate complexities of chronic pain.
Collapse
Affiliation(s)
- Huan-Yu Xiong
- Pain in Motion Research Group (PAIN), Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education & Physiotherapy, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (H.-Y.X.); (J.H.); (A.W.); (J.V.C.); (A.P.)
| | - Jolien Hendrix
- Pain in Motion Research Group (PAIN), Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education & Physiotherapy, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (H.-Y.X.); (J.H.); (A.W.); (J.V.C.); (A.P.)
- Department of Public Health and Primary Care, Centre for Environment & Health, KU Leuven, 3000 Leuven, Belgium
- Research Foundation—Flanders (FWO), 1000 Brussels, Belgium
| | - Siobhan Schabrun
- The School of Physical Therapy, University of Western Ontario, London, ON N6A 3K7, Canada;
- The Gray Centre for Mobility and Activity, Parkwood Institute, London, ON N6A 4V2, Canada
| | - Arne Wyns
- Pain in Motion Research Group (PAIN), Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education & Physiotherapy, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (H.-Y.X.); (J.H.); (A.W.); (J.V.C.); (A.P.)
| | - Jente Van Campenhout
- Pain in Motion Research Group (PAIN), Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education & Physiotherapy, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (H.-Y.X.); (J.H.); (A.W.); (J.V.C.); (A.P.)
| | - Jo Nijs
- Pain in Motion Research Group (PAIN), Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education & Physiotherapy, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (H.-Y.X.); (J.H.); (A.W.); (J.V.C.); (A.P.)
- Chronic Pain Rehabilitation, Department of Physical Medicine and Physiotherapy, University Hospital Brussels, 1090 Brussels, Belgium
- Department of Health and Rehabilitation, Unit of Physiotherapy, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 41390 Göterbog, Sweden
| | - Andrea Polli
- Pain in Motion Research Group (PAIN), Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education & Physiotherapy, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (H.-Y.X.); (J.H.); (A.W.); (J.V.C.); (A.P.)
- Department of Public Health and Primary Care, Centre for Environment & Health, KU Leuven, 3000 Leuven, Belgium
- Research Foundation—Flanders (FWO), 1000 Brussels, Belgium
| |
Collapse
|
6
|
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
|
7
|
Hu Y, Jia K, Zhou Y, Chen L, Wang F, Yi X, Huang Y, Ge Y, Chen X, Liao D, Peng Y, Meng Y, Liu Y, Luo Q, Cheng B, Zhao Y, Lu H, Yuan W. Rutin hydrate relieves neuroinflammation in zebrafish models: Involvement of NF-κB pathway as a central network. FISH & SHELLFISH IMMUNOLOGY 2023; 141:109062. [PMID: 37678480 DOI: 10.1016/j.fsi.2023.109062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/26/2023] [Accepted: 09/04/2023] [Indexed: 09/09/2023]
Abstract
Neuroinflammation is prevalent in multiple brain diseases and may also lead to dementia, cognitive impairment, and impaired spatial memory function associated with neurodegenerative diseases. A neuroprotective and antioxidant flavonoid, rutin hydrate (RH), was evaluated for the anti-neuroinflammatory activity mediated by copper sulfate (CuSO4) solution and lipopolysaccharide (LPS) in zebrafish. The results showed that 100 mg/L RH significantly reduced the ratio of neutrophil mobility in caudal hematopoietic tissue (CHT) region caused by CuSO4 and the number of neutrophils co-localized with facial peripheral nerves. In the LPS model, RH co-injection significantly diminished neutrophil and macrophage migration. Therefore, RH exhibited a significant rescue effect on both models. In addition, RH treatment remarkably reduced the effects of neuroinflammation on the locomotor ability, expression levels of genes associated with behavioral disorders, and acetylcholinesterase (AChE) activity. Furthermore, network pharmacology techniques were employed to investigate the potential mechanisms, and the associated genes and enzyme activities were validated in order to elucidate the underlying mechanisms. Network pharmacological analysis and zebrafish model indicated that RH regulated the expressions of NF-κB pathway-related targets (Toll-like receptor 9 (tlr9), nuclear factor kappa B subunit 1 (nfkb1), RELA proto-oncogene (RelA), nitric oxide synthase 2a, inducible (nos2a), tumour necrosis factor alpha-like (tnfα), interleukin 6 (il6), interleukin 1β (il1β), chemokine 8 (cxcl8), and macrophage migration inhibitory factor (mif)) as well as six key factors (arachidonic acid 4 alpha-lipoxygenase (alox4a), arachidonate 5-lipoxygenase a (alox5), prion protein a (prnpa), integrin, beta 2 (itgb2), catalase (CAT), and alkaline phosphatase (ALP) enzymes). Through this study, a thorough understanding of the mechanism underlying the therapeutic effects of RH in neuroinflammation has been achieved, thereby establishing a solid foundation for further research on the potential therapeutic applications of RH in neuroinflammatory disorders.
Collapse
Affiliation(s)
- Ying Hu
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Kun Jia
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Yatong Zhou
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Lixin Chen
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Fei Wang
- The First Clinical College of Gannan Medical Uinversity, Ganzhou, 341000, Jiangxi, China
| | - Xiaokun Yi
- The First Clinical College of Gannan Medical Uinversity, Ganzhou, 341000, Jiangxi, China
| | - Yong Huang
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Yurui Ge
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Xiaomei Chen
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Dalong Liao
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Yuyang Peng
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Yunlong Meng
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Ye Liu
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Qiang Luo
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Bo Cheng
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Yan Zhao
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Huiqiang Lu
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China; Affiliated Hospital of Jinggangshan University, Jinggangshan University, Ji'an, 343009, Jiangxi, China.
| | - Wei Yuan
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China.
| |
Collapse
|
8
|
Pennacchietti V, Pagano L, Malagrinò F, Diop A, Di Felice M, Di Matteo S, Marcocci L, Pietrangeli P, Toto A, Gianni S. Characterization of the folding and binding properties of the PTB domain of FRS2 with phosphorylated and unphosphorylated ligands. Arch Biochem Biophys 2023; 745:109703. [PMID: 37543351 DOI: 10.1016/j.abb.2023.109703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/07/2023]
Abstract
PTB (PhosphoTyrosine Binding) domains are protein domains that exert their function by binding phosphotyrosine residues on other proteins. They are commonly found in a variety of signaling proteins and are important for mediating protein-protein interactions in numerous cellular processes. PTB domains can also exhibit binding to unphosphorylated ligands, suggesting that they have additional binding specificities beyond phosphotyrosine recognition. Structural studies have reported that the PTB domain from FRS2 possesses this peculiar feature, allowing it to interact with both phosphorylated and unphosphorylated ligands, such as TrkB and FGFR1, through different topologies and orientations. In an effort to elucidate the dynamic and functional properties of these protein-protein interactions, we provide a complete characterization of the folding mechanism of the PTB domain of FRS2 and the binding process to peptides mimicking specific regions of TrkB and FGFR1. By analyzing the equilibrium and kinetics of PTB folding, we propose a mechanism implying the presence of an intermediate along the folding pathway. Kinetic binding experiments performed at different ionic strengths highlighted the electrostatic nature of the interaction with both peptides. The specific role of single amino acids in early and late events of binding was pinpointed by site-directed mutagenesis. These results are discussed in light of previous experimental works on these protein systems.
Collapse
Affiliation(s)
- Valeria Pennacchietti
- Dipartimento di Scienze Biochimiche "A. Rossi Fanelli", Sapienza Università di Roma, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Livia Pagano
- Dipartimento di Scienze Biochimiche "A. Rossi Fanelli", Sapienza Università di Roma, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Francesca Malagrinò
- Dipartimento di Scienze Biochimiche "A. Rossi Fanelli", Sapienza Università di Roma, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Awa Diop
- Dipartimento di Scienze Biochimiche "A. Rossi Fanelli", Sapienza Università di Roma, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Mariana Di Felice
- Dipartimento di Scienze Biochimiche "A. Rossi Fanelli", Sapienza Università di Roma, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Sara Di Matteo
- Dipartimento di Scienze Biochimiche "A. Rossi Fanelli", Sapienza Università di Roma, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Lucia Marcocci
- Dipartimento di Scienze Biochimiche "A. Rossi Fanelli", Sapienza Università di Roma, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Paola Pietrangeli
- Dipartimento di Scienze Biochimiche "A. Rossi Fanelli", Sapienza Università di Roma, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Angelo Toto
- Dipartimento di Scienze Biochimiche "A. Rossi Fanelli", Sapienza Università di Roma, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, P.le Aldo Moro 5, 00185, Rome, Italy.
| | - Stefano Gianni
- Dipartimento di Scienze Biochimiche "A. Rossi Fanelli", Sapienza Università di Roma, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, P.le Aldo Moro 5, 00185, Rome, Italy.
| |
Collapse
|
9
|
Tuwar MN, Chen WH, Chiwaya AM, Yeh HL, Nguyen MH, Bai CH. Brain-Derived Neurotrophic Factor (BDNF) and Translocator Protein (TSPO) as Diagnostic Biomarkers for Acute Ischemic Stroke. Diagnostics (Basel) 2023; 13:2298. [PMID: 37443691 DOI: 10.3390/diagnostics13132298] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/11/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) interacts with tropomyosin-related kinase B (TrkB) to promote neuronal growth, survival, differentiation, neurotransmitter release, and synaptic plasticity. The translocator protein (TSPO) is known to be found in arterial plaques, which are a symptom of atherosclerosis and a contributory cause of ischemic stroke. This study aims to determine the diagnostic accuracy of plasma BDNF and TSPO levels in discriminating new-onset acute ischemic stroke (AIS) patients from individuals without acute ischemic stroke. A total of 90 AIS patients (61% male, with a mean age of 67.7 ± 12.88) were recruited consecutively in a stroke unit, and each patient was paired with two age- and gender-matched controls. The sensitivity, specificity, and area of the curve between high plasma BDNF and TSPO and having AIS was determined using receiver operating characteristic curves. Furthermore, compared to the controls, AIS patients exhibited significantly higher levels of BDNF and TSPO, blood pressure, HbA1c, and white blood cells, as well as higher creatinine levels. The plasma levels of BDNF and TSPO can significantly discriminate AIS patients from healthy individuals (AUC 0.76 and 0.89, respectively). However, combining the two biomarkers provided little improvement in AUC (0.90). It may be possible to use elevated levels of TSPO as a diagnostic biomarker in patients with acute ischemic stroke upon admission.
Collapse
Affiliation(s)
- Mayuri N Tuwar
- School of Public Health, College of Public Health, Taipei Medical University, Taipei 106236, Taiwan
| | - Wei-Hung Chen
- Department of Neurology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei 111045, Taiwan
| | - Arthur M Chiwaya
- CLIME Group, Department of Biomedical Sciences, Division of Molecular Biology and Human Genetics, FMHS, Stellenbosch University, Francie Van Zijl Drive, Tygerberg, Cape Town 7505, South Africa
| | - Hsu-Ling Yeh
- Department of Neurology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei 111045, Taiwan
| | - Minh H Nguyen
- School of Preventive Medicine and Public Health, Hanoi Medical University, Hanoi 100000, Vietnam
| | - Chyi-Huey Bai
- School of Public Health, College of Public Health, Taipei Medical University, Taipei 106236, Taiwan
- School of Medicine, College of Medicine, Taipei Medical University, Taipei 106236, Taiwan
| |
Collapse
|
10
|
Kang JH, Guo XD, Wang YD, Kang XW. Neuroprotective Effects of N-acetylserotonin and Its Derivative. Neuroscience 2023; 517:18-25. [PMID: 36893983 DOI: 10.1016/j.neuroscience.2023.02.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 02/12/2023] [Accepted: 02/20/2023] [Indexed: 03/09/2023]
Abstract
N-acetylserotonin (NAS) is a chemical intermediate in melatonin biosynthesis. NAS and its derivative N-(2-(5-hydroxy-1H-indol-3-yl) ethyl)-2-oxopiperidine-3-carboxamide (HIOC) are potential therapeutic agents for traumatic brain injury, autoimmune encephalomyelitis, hypoxic-ischemic encephalopathy, and other diseases. Evidence shows that NAS and its derivative HIOC have neuroprotective properties, and can exert neuroprotective effects by inhibiting oxidative stress, anti-apoptosis, regulating autophagy dysfunction, and anti-inflammatory. In this review, we discussed the neuroprotective effects and related mechanisms of NAS and its derivative HIOC to provide a reference for follow-up research and applications.
Collapse
Affiliation(s)
- Ji-He Kang
- Orthopedics Department, the Second Hospital of Lanzhou University, Lanzhou, China; Orthopedics Department, the Second Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Xu-Dong Guo
- Orthopedics Department, the Second Hospital of Lanzhou University, Lanzhou, China
| | - Yi-Dian Wang
- Orthopedics Department, School of Medicine, Honghui-hospital, Xi'an Jiaotong University, Xi'an, China
| | - Xue-Wen Kang
- Orthopedics Department, the Second Hospital of Lanzhou University, Lanzhou, China.
| |
Collapse
|
11
|
Shahar O, Pereman I, Khamisie H, Ezov N, Danay O, Khattib A, Schweitzer R, Khatib S, Mahajna J. Compounds originating from the edible mushroom Auricularia auricula-judae inhibit tropomyosin receptor kinase B activity. Heliyon 2023; 9:e13756. [PMID: 36895384 PMCID: PMC9988514 DOI: 10.1016/j.heliyon.2023.e13756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 02/05/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023] Open
Abstract
Tropomyosin receptor kinase B (TrkB) serves as a pivotal factor in various cancers. To identify novel natural compounds with TrkB-inhibiting properties, a screening approach was applied using extracts from a collection of wild and cultivated mushroom fruiting bodies, and Ba/F3 cells that ectopically express TrkB (TPR-TrkB). We selected mushroom extracts that selectively inhibited proliferation of the TPR-TrkB cells. We then evaluated the ability of exogenous interleukin 3 to rescue growth inhibition by the selected TrkB-positive extracts. An ethyl acetate extract of Auricularia auricula-judae actively inhibited auto-phosphorylation of TrkB. LC-MS/MS analysis of this extract revealed substances that might be responsible for the observed activity. This screening approach demonstrates, for the first time, that extracts originating from the mushroom A. auricula-judae exhibit TrkB-inhibition properties that might hold therapeutic potential for TrkB-positive cancers.
Collapse
Affiliation(s)
- Orr Shahar
- Department of Nutrition and Natural Products, Migal - Galilee Research Institute, Kiryat Shmona, Israel.,Department of Biotechnology, Tel Hai College, Kiryat Shmona, Israel
| | - Idan Pereman
- Department of Nutrition and Natural Products, Migal - Galilee Research Institute, Kiryat Shmona, Israel.,Department of Biotechnology, Tel Hai College, Kiryat Shmona, Israel
| | - Hazem Khamisie
- Department of Nutrition and Natural Products, Migal - Galilee Research Institute, Kiryat Shmona, Israel
| | - Nirit Ezov
- Department of Nutrition and Natural Products, Migal - Galilee Research Institute, Kiryat Shmona, Israel
| | - Ofer Danay
- Department of Nutrition and Natural Products, Migal - Galilee Research Institute, Kiryat Shmona, Israel.,Department of Biotechnology, Tel Hai College, Kiryat Shmona, Israel
| | - Ali Khattib
- Department of Nutrition and Natural Products, Migal - Galilee Research Institute, Kiryat Shmona, Israel
| | - Ron Schweitzer
- Department of Nutrition and Natural Products, Migal - Galilee Research Institute, Kiryat Shmona, Israel.,Analytical Laboratory, Tel Hai College, Kiryat Shmona, Israel
| | - Soliman Khatib
- Department of Nutrition and Natural Products, Migal - Galilee Research Institute, Kiryat Shmona, Israel.,Analytical Laboratory, Tel Hai College, Kiryat Shmona, Israel
| | - Jamal Mahajna
- Department of Nutrition and Natural Products, Migal - Galilee Research Institute, Kiryat Shmona, Israel.,Department of Biotechnology, Tel Hai College, Kiryat Shmona, Israel
| |
Collapse
|
12
|
Ballesio A, Zagaria A, Curti DG, Moran R, Goadsby PJ, Rosenzweig I, Lombardo C. Peripheral brain-derived neurotrophic factor (BDNF) in insomnia: A systematic review and meta-analysis. Sleep Med Rev 2023; 67:101738. [PMID: 36577338 DOI: 10.1016/j.smrv.2022.101738] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
The brain-derived neurotrophic factor (BDNF) is associated with emotional and cognitive functioning, and it is considered a transdiagnostic biomarker for mental disorders. Literature on insomnia related BDNF changes yielded contrasting results and it has never been synthetized using meta-analysis. To fill this gap, we conducted a systematic review and meta-analysis of case-control studies examining the levels of peripheric BDNF in individuals with insomnia and healthy controls using the PRISMA guidelines. PubMed, Scopus, Medline, PsycINFO and CINAHL were searched up to Nov 2022. Nine studies met the inclusion criteria and were assessed using the Newcastle-Ottawa Scale. Eight studies reported sufficient data for meta-analysis. Random-effects models showed lower BDNF in subjects with insomnia (n = 446) than in controls (n = 706) (Hedge's g = -0.86, 95% CI: -1.39 to -0.32, p = .002). Leave-one-out sensitivity analysis confirmed that the pooled effect size was robust and not driven by any single study. However, given the small sample size, the cross-sectional nature of the measurement, and the high heterogeneity of included data, the results should be cautiously interpreted. Progress in the study of BDNF in insomnia is clinically relevant to better understand the mechanisms that may explain the relationship between disturbed sleep and mental disorders.
Collapse
Affiliation(s)
- Andrea Ballesio
- Department of Psychology, Sapienza University of Rome, Italy.
| | - Andrea Zagaria
- Department of Psychology, Sapienza University of Rome, Italy
| | | | - Rosalyn Moran
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, UK
| | - Peter J Goadsby
- NIHR-Wellcome Trust King's Clinical Research Facility, King's College London UK
| | - Ivana Rosenzweig
- Sleep and Brain Plasticity Centre, Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, UK; Sleep Disorders Centre, Guy's and St Thomas' National Health Service Foundation Trust, London, UK
| | | |
Collapse
|
13
|
Recent and Future Strategies to Overcome Resistance to Targeted Therapies and Immunotherapies in Metastatic Colorectal Cancer. J Clin Med 2022; 11:jcm11247523. [PMID: 36556139 PMCID: PMC9783354 DOI: 10.3390/jcm11247523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/06/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Colorectal cancer (CRC) is the third most common cause of cancer-related deaths worldwide, and 20% of patients with CRC present at diagnosis with metastases. The treatment of metastatic CRC is based on a fluoropyrimidine-based chemotherapy plus additional agents such as oxaliplatin and irinotecan. To date, on the basis of the molecular background, targeted therapies (e.g., monoclonal antibodies against epidermal growth factor receptor or inhibiting angiogenesis) are administered to improve the treatment of metastatic CRC. In addition, more recently, immunological agents emerged as effective in patients with a defective mismatch repair system. The administration of targeted therapies and immunotherapy lead to a significant increase in the survival of patients; however these drugs do not always prove effective. In most cases the lack of effectiveness is due to the development of primary resistance, either a resistance-inducing factor is already present before treatment or resistance is acquired when it occurs after treatment initiation. In this review we describe the most relevant targeted therapies and immunotherapies and expand on the reasons for resistance to the different approved or under development targeted drugs. Then we showed the possible mechanisms and drugs that may lead to overcoming the primary or acquired resistance in metastatic CRC.
Collapse
|
14
|
Li Y, Wei C, Wang W, Li Q, Wang Z. Tropomyosin receptor kinase B (TrkB) signalling: targeted therapy in neurogenic tumours. J Pathol Clin Res 2022; 9:89-99. [PMID: 36533776 PMCID: PMC9896160 DOI: 10.1002/cjp2.307] [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: 08/25/2022] [Revised: 11/29/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022]
Abstract
Tropomyosin receptor kinase B (TrkB), a transmembrane receptor protein, has been found to play a pivotal role in neural development. This protein is encoded by the neurotrophic receptor tyrosine kinase 2 (NTRK2) gene, and its abnormal activation caused by NTRK2 overexpression or fusion can contribute to tumour initiation, progression, and resistance to therapeutics in multiple types of neurogenic tumours. Targeted therapies for this mechanism have been designed and developed in preclinical and clinical studies, including selective TrkB inhibitors and pan-TRK inhibitors. This review describes the gene structure, biological function, abnormal TrkB activation mechanism, and current-related targeted therapies in neurogenic tumours.
Collapse
Affiliation(s)
- Yuehua Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPR China
| | - Chengjiang Wei
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPR China
| | - Wei Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPR China
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPR China
| | - Zhi‐Chao Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPR China
| |
Collapse
|
15
|
NSCLC as the Paradigm of Precision Medicine at Its Finest: The Rise of New Druggable Molecular Targets for Advanced Disease. Int J Mol Sci 2022; 23:ijms23126748. [PMID: 35743191 PMCID: PMC9223783 DOI: 10.3390/ijms23126748] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/09/2022] [Accepted: 06/14/2022] [Indexed: 02/04/2023] Open
Abstract
Standard treatment for advanced non-small cell lung cancer (NSCLC) historically consisted of systemic cytotoxic chemotherapy until the early 2000s, when precision medicine led to a revolutionary change in the therapeutic scenario. The identification of oncogenic driver mutations in EGFR, ALK and ROS1 rearrangements identified a subset of patients who largely benefit from targeted agents. However, since the proportion of patients with druggable alterations represents a minority, the discovery of new potential driver mutations is still an urgent clinical need. We provide a comprehensive review of the emerging molecular targets in NSCLC and their applications in the advanced setting.
Collapse
|
16
|
Li T, Li X, Huang X, Yu H, Li S, Zhang Z, Xie Y, Song X, Liu J, Yang X, Liu G. Mitochondriomics reveals the underlying neuroprotective mechanism of TrkB receptor agonist R13 in the 5×FAD mice. Neuropharmacology 2022; 204:108899. [PMID: 34838815 DOI: 10.1016/j.neuropharm.2021.108899] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/20/2021] [Accepted: 11/23/2021] [Indexed: 02/07/2023]
Abstract
Decreased energy metabolism and mitochondrial biogenesis defects are implicated in the pathogenesis of Alzheimer's disease (AD). In present study, mitochondriomics analysis revealed significant effects of R13, a prodrug of 7,8-dihydroxyflavone, on mitochondrial protein expression profile, including the proteins related to the biological processes: fatty acid beta-oxidation, fatty acid metabolic process, mitochondrial electron transport, and mitochondrial respiratory chain. Cluster analysis demonstrated that R13 promoted mitochondrial oxidative phosphorylation (OXPHOS). The functional analysis showed that R13 increased ATP levels and enhanced OXPHOS including complex Ⅰ, Ⅱ, Ⅲ and Ⅳ. R13 treatment increased mitochondrial biogenesis by regulating the levels of p-AMPKα, p-CREB, PGC-1α, NRF1 and TFAM as a consequence of activation of TrkB receptor in the 5 × FAD mice. Finally, R13 significantly reduced the levels of tau phosphorylation and Aβ plaque. Our data suggest that R13 may be used for treating AD via enhancing mitochondrial biogenesis and metabolism.
Collapse
Affiliation(s)
- Ting Li
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao Li
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China; Department of Pathology, Wuhan No. 1 Hospital, Wuhan, 430022, China
| | - Xi Huang
- Department of Neurology,The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, 518020, China; The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Hao Yu
- Shenzhen Polytechnic, Shenzhen, 518055, China
| | - Shupeng Li
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Zaijun Zhang
- Institute of New Drug Research and Guangzhou, Key Laboratory of Innovative Chemical Drug Research in Cardio-Cerebrovascular Diseases, Jinan University College of Pharmacy, Guangzhou, 510632, China
| | - Yongmei Xie
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Xiangrong Song
- Department of Critical Care Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jianjun Liu
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China
| | - Xifei Yang
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China.
| | - Gongping Liu
- Department of Pathology, Wuhan No. 1 Hospital, Wuhan, 430022, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, JS, China.
| |
Collapse
|
17
|
Recine F, De Vita A, Fausti V, Pieri F, Bongiovanni A, Franchini E, Casadei R, Falasconi MC, Oboldi D, Matteucci F, Pallotti MC, Mercatali L, Riva N, Gurrieri L, Vanni S, Liverani C, Miserocchi G, Spadazzi C, Cocchi C, Ibrahim T. Case Report: Adult NTRK-Rearranged Spindle Cell Neoplasm: Early Tumor Shrinkage in a Case With Bone and Visceral Metastases Treated With Targeted Therapy. Front Oncol 2022; 11:740676. [PMID: 35070960 PMCID: PMC8776642 DOI: 10.3389/fonc.2021.740676] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 12/10/2021] [Indexed: 12/12/2022] Open
Abstract
Background NTRK (neurotrophic tyrosine receptor kinase)-rearranged spindle cell neoplasms are a new group of tumors included in the new 5th edition of the World Health Organization (WHO) classification of soft Tissue and Bone Sarcomas. These tumors are characterized by NTRK gene fusions and show a wide spectrum of histologies and clinical behavior. Several targeted therapies have recently been approved for tumors harboring NTRK fusions, including STS. Case Presentation A 26-year-old male with advanced, pretreated NTRK rearranged spindle cell neoplasm and liver, lung and bone metastases was treated with larotrectinib on a continuous 28-day schedule, at a dose of 100 mg twice daily. An 18FDG-PET/CT scan performed after 7 days of treatment showed tumor shrinkage in both visceral and bone lesions. There was no drug-related toxicity. Subsequent evaluations confirmed continued tumor regression in disease sites. The patient is well and continues treatment. Conclusion The clinical and radiological response of our patient with an uncommon TPM4 (exon 7)-NTRK1 (exon 12) gene fusion tumor treated with a first-generation TRK inhibitor could contribute to a better understanding of the biology of this new STS entity and help to improve patient management.
Collapse
Affiliation(s)
- Federica Recine
- Osteoncology and Rare Tumors Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Alessandro De Vita
- Osteoncology and Rare Tumors Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Valentina Fausti
- Osteoncology and Rare Tumors Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Federica Pieri
- Pathology Unit, Morgagni-Pierantoni Hospital, Forlì, Italy
| | - Alberto Bongiovanni
- Osteoncology and Rare Tumors Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Eugenia Franchini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | | | | | - Devil Oboldi
- Radiology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Federica Matteucci
- Nuclear Medicine Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Maria Caterina Pallotti
- Palliative Care Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Laura Mercatali
- Osteoncology and Rare Tumors Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Nada Riva
- Osteoncology and Rare Tumors Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Lorena Gurrieri
- Osteoncology and Rare Tumors Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Silvia Vanni
- Osteoncology and Rare Tumors Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Chiara Liverani
- Osteoncology and Rare Tumors Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Giacomo Miserocchi
- Osteoncology and Rare Tumors Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Chiara Spadazzi
- Osteoncology and Rare Tumors Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Claudia Cocchi
- Osteoncology and Rare Tumors Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Toni Ibrahim
- Osteoncology and Rare Tumors Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| |
Collapse
|
18
|
Cristina Mendonça Nogueira T, Vinicius Nora de Souza M. New FDA oncology small molecule drugs approvals in 2020: Mechanism of action and clinical applications. Bioorg Med Chem 2021; 46:116340. [PMID: 34416511 DOI: 10.1016/j.bmc.2021.116340] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/21/2021] [Accepted: 07/26/2021] [Indexed: 12/30/2022]
Abstract
In 2020, fifty-three new drugs, including forty small-molecules (thirty-six new chemical entities and four new diagnostic agents) and thirteen biologic drugs were approved by the U.S. Food and Drug Administration (FDA). This year, small-molecules continue to play a role in innovative treatments representing around 75% of all drugs accepted by FDA. The dominant therapeutic area was oncology, accounting for twenty-three new approvals, including thirteen new chemical entities, four new diagnostic agents, and thirteen biologic drugs. Recognizing the importance of small-molecules on cancer treatment, this review aims to provide an overview regarding the clinical applications and mechanism of action of the thirteen new small-molecules (excluding new diagnostic agents) approved by FDA in 2020.
Collapse
Affiliation(s)
- Thais Cristina Mendonça Nogueira
- Instituto de Tecnologia em Fármacos-Far Manguinhos, Fundação Oswaldo Cruz, Rua Sizenando Nabuco 100, Manguinhos, Rio de Janeiro, RJ 21041- 250 Brazil
| | - Marcus Vinicius Nora de Souza
- Instituto de Tecnologia em Fármacos-Far Manguinhos, Fundação Oswaldo Cruz, Rua Sizenando Nabuco 100, Manguinhos, Rio de Janeiro, RJ 21041- 250 Brazil.
| |
Collapse
|
19
|
Ratti M, Grizzi G, Passalacqua R, Lampis A, Cereatti F, Grassia R, Hahne JC. NTRK fusions in colorectal cancer: clinical meaning and future perspective. Expert Opin Ther Targets 2021; 25:677-683. [PMID: 34488530 DOI: 10.1080/14728222.2021.1978070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
INTRODUCTION Despite the efforts of the scientific community, the prognosis of metastatic colorectal cancer (mCRC) remains poor. Actionable gene fusions such as Neurotrophic Tropomyosin Receptor Kinases (NTRK) rearrangements are rare but might represent a new target to improve outcomes in this setting. The first-generation TRK inhibitors, larotrectinib and entrectinib, have demonstrated efficacy and safety in mCRC cancer patients exhibiting NTRK pathogenic fusions. Moreover, second-generation molecules are emerging, able to overcome the acquired resistance to NTRK blocking. AREAS COVERED This review aims to report the current knowledge and the available evidence on NTRK fusion in mCRC, with a focus on molecular bases, clinical characteristics, prognostic meaning, and new therapeutic approaches, from the perspective of the clinical oncologist. EXPERT OPINION Considering the limited options associated with the treatment of mCRC patients, the possibility of identifying new molecular biomarkers is an urgent clinical need. The availability of new molecular targets and the combinations of different agents might represent the true breakthrough point, allowing for change in the clinical course of colorectal cancer patients.
Collapse
Affiliation(s)
- Margherita Ratti
- Department of Medical Oncology, Azienda Socio Sanitaria Territoriale of Cremona, Cremona, Italy
| | - Giulia Grizzi
- Department of Medical Oncology, Azienda Socio Sanitaria Territoriale of Cremona, Cremona, Italy
| | - Rodolfo Passalacqua
- Department of Medical Oncology, Azienda Socio Sanitaria Territoriale of Cremona, Cremona, Italy
| | - Andrea Lampis
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Fabrizio Cereatti
- Department of Medical Oncology, Azienda Socio Sanitaria Territoriale of Cremona, Cremona, Italy
| | - Roberto Grassia
- Department of Medical Oncology, Azienda Socio Sanitaria Territoriale of Cremona, Cremona, Italy
| | - Jens Claus Hahne
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| |
Collapse
|
20
|
Miyanishi H, Muramatsu SI, Nitta A. Striatal Shati/Nat8l-BDNF pathways determine the sensitivity to social defeat stress in mice through epigenetic regulation. Neuropsychopharmacology 2021; 46:1594-1605. [PMID: 34099867 PMCID: PMC8280178 DOI: 10.1038/s41386-021-01033-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/02/2021] [Accepted: 05/04/2021] [Indexed: 12/12/2022]
Abstract
The global number of patients with depression increases in correlation to exposure to social stress. Chronic stress does not trigger depression in all individuals, as some remain resilient. The underlying molecular mechanisms that contribute to stress sensitivity have been poorly understood, although revealing the regulation of stress sensitivity could help develop treatments for depression. We previously found that striatal Shati/Nat8l, an N-acetyltransferase, was increased in a depression mouse model. We investigated the roles of Shati/Nat8l in stress sensitivity in mice and found that Shati/Nat8l and brain-derived neurotrophic factor (BDNF) levels in the dorsal striatum were increased in stress-susceptible mice but not in resilient mice exposed to repeated social defeat stress (RSDS). Knockdown of Shati/Nat8l in the dorsal striatum induced resilience to RSDS. In addition, blockade of BDNF signaling in the dorsal striatum by ANA-12, a BDNF-specific receptor tropomyosin-receptor-kinase B (TrkB) inhibitor, also induced resilience to stress. Shati/Nat8l is correlated with BDNF expression after RSDS, and BDNF is downstream of Shati/Nat8l pathways in the dorsal striatum; Shati/Nat8l is epigenetically regulated by BDNF via histone acetylation. Our results demonstrate that striatal Shati/Nat8l-BDNF pathways determine stress sensitivity through epigenetic regulation. The striatal Shati/Nat8l-BDNF pathway could be a novel target for treatments of depression and could establish a novel therapeutic strategy for depression patients.
Collapse
Affiliation(s)
- Hajime Miyanishi
- grid.267346.20000 0001 2171 836XDepartment of Pharmaceutical Therapy and Neuropharmacology, Faculty of Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Shin-ichi Muramatsu
- grid.410804.90000000123090000Division of Neurological Gene Therapy, Open Innovation Center, Jichi Medical University, Shimotsuke, Japan ,grid.26999.3d0000 0001 2151 536XCenter for Gene and Cell Therapy, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Atsumi Nitta
- Department of Pharmaceutical Therapy and Neuropharmacology, Faculty of Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, University of Toyama, Toyama, Japan.
| |
Collapse
|
21
|
Alfei S, Schito AM, Zuccari G. Nanotechnological Manipulation of Nutraceuticals and Phytochemicals for Healthy Purposes: Established Advantages vs. Still Undefined Risks. Polymers (Basel) 2021; 13:2262. [PMID: 34301020 PMCID: PMC8309409 DOI: 10.3390/polym13142262] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 12/12/2022] Open
Abstract
Numerous foods, plants, and their bioactive constituents (BACs), named nutraceuticals and phytochemicals by experts, have shown many beneficial effects including antifungal, antiviral, anti-inflammatory, antibacterial, antiulcer, anti-cholesterol, hypoglycemic, immunomodulatory, and antioxidant activities. Producers, consumers, and the market of food- and plant-related compounds are increasingly attracted by health-promoting foods and plants, thus requiring a wider and more fruitful exploitation of the healthy properties of their BACs. The demand for new BACs and for the development of novel functional foods and BACs-based food additives is pressing from various sectors. Unfortunately, low stability, poor water solubility, opsonization, and fast metabolism in vivo hinder the effective exploitation of the potential of BACs. To overcome these issues, researchers have engineered nanomaterials, obtaining food-grade delivery systems, and edible food- and plant-related nanoparticles (NPs) acting as color, flavor, and preservative additives and natural therapeutics. Here, we have reviewed the nanotechnological transformations of several BACs implemented to increase their bioavailability, to mask any unpleasant taste and flavors, to be included as active ingredients in food or food packaging, to improve food appearance, quality, and resistance to deterioration due to storage. The pending issue regarding the possible toxic effect of NPs, whose knowledge is still limited, has also been discussed.
Collapse
Affiliation(s)
- Silvana Alfei
- Department of Pharmacy, University of Genoa, Viale Cembrano, 16148 Genoa, Italy;
| | - Anna Maria Schito
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Viale Benedetto XV 6, I-16132 Genoa, Italy;
| | - Guendalina Zuccari
- Department of Pharmacy, University of Genoa, Viale Cembrano, 16148 Genoa, Italy;
| |
Collapse
|
22
|
Li Z, Wang H, Xiao G, Du H, He S, Feng Y, Zhang B, Zhu Y. Recovery of post-stroke cognitive and motor deficiencies by Shuxuening injection via regulating hippocampal BDNF-mediated Neurotrophin/Trk Signaling. Biomed Pharmacother 2021; 141:111828. [PMID: 34146848 DOI: 10.1016/j.biopha.2021.111828] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 06/07/2021] [Accepted: 06/11/2021] [Indexed: 12/16/2022] Open
Abstract
A mild ischemic stroke may cause both debilitating locomotor and cognitive decline, for which the mechanism is not fully understood, and no therapies are currently available. In this study, a nonfatal stroke model was constructed in mice by a modified middle cerebral artery occlusion (MCAO) procedure, allowing an extended recovery period up to 28 days. The extended MCAO model successfully mimicked phenotypes of a recovery phase post-stroke, including locomotor motor and cognitive deficiencies, which were effectively improved after Shuxuening injection (SXNI) treatment. Tissue slices staining showed that SXNI repaired brain injury and reduced neuronal apoptosis, especially in the hippocampus CA3 region. Transcriptomics sequencing study revealed 565 differentially expressed genes (DEGs) in the ischemic brain after SXNI treatment. Integrated network pharmacological analysis identified Neurotrophin/Trk Signaling was the most relevant pathway, which involves 15 key genes. Related DEGs were further validated by RT-PCR. Western-blot analysis showed that SXNI reversed the abnormal expression of BDNF, TrkB, Mek3 and Jnk1after stroke. ELISA found that SXNI increased brain level of p-Erk and Creb. At sub-brain level, the expression of BDNF and TrkB was decreased and GFAP was increased on the hippocampal CA3 region in the post-stroke recovery phase and this abnormality was improved by SXNI. In vitro experiments also found that oxygen glucose deprivation reduced the expression of BDNF and TrkB, which was reversed by SXNI. In summary, we conclude that SXNI facilitates the recovery of cognitive and locomotor dysfunction by modulating Neurotrophin/Trk Signaling in a mouse model for the recovery phase of post-ischemic stroke.
Collapse
Affiliation(s)
- Zhixiong Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin 300457, China
| | - Huanyi Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin 300457, China
| | - Guangxu Xiao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin 300457, China
| | - Hongxia Du
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin 300457, China
| | - Shuang He
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin 300457, China
| | - Yuxin Feng
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin 300457, China
| | - Boli Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yan Zhu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin 300457, China.
| |
Collapse
|
23
|
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
|
24
|
Forschner A, Forchhammer S, Bonzheim I. NTRK‐
Genfusionen beim Melanom: Diagnostik, Prävalenz und mögliche Therapierelevanz. J Dtsch Dermatol Ges 2020; 18:1387-1393. [PMID: 33373127 DOI: 10.1111/ddg.14160_g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/03/2020] [Accepted: 03/09/2020] [Indexed: 11/29/2022]
Abstract
Fusionen der neurotrophen Tyrosin-Rezeptor-Kinase (NTRK) sind bekannte Treiber der Onkogenese und treten, wenn auch sehr selten, ebenfalls beim Melanom auf. Eine besonders hohe Inzidenz von NTRK-Genfusionen wird beim infantilen Fibrosarkom (> 90 %) oder der sekretorischen Form des Mammakarzinoms (> 90 %) berichtet. Erst kürzlich wurde Larotrectinib, ein Tropomyosin-Rezeptor-Kinase (TRK)-Inhibitor, zugelassen, und wir fragten uns, ob TRK-Inhibitoren auch für Melanompatienten relevant sein könnten. Aus diesem Grund haben wir die Literatur gesichtet und sind zu relevanten Ergebnissen gekommen. Beim spitzoiden Melanom sind NTRK-Fusionen mit einer Prävalenz von 21-29 % relativ häufig, verglichen mit < 1 % beim kutanen oder mukosalen und 2,5 % beim akralen Melanom. Es scheint so zu sein, dass sich Fusionsproteine und andere onkogene Treiber wie BRAF oder NRAS gegenseitig ausschließen. Ein weiterer Anhaltspunkt für eine erhöhte Wahrscheinlichkeit, NTRK-positive Tumoren zu detektieren, könnte eine geringe Tumormutationslast sein. Da für Patienten mit NTRK-Fusionen bereits TRK-Inhibitoren zur Verfügung stehen, wird die Herausforderung darin bestehen, das Screening auf NTRK-Genfusionen in die klinische Praxis umzusetzen. Ein möglicher Ansatz könnte darin bestehen, BRAF-, NRAS- und KIT-Wildtyp-Melanom-Patienten mittels Next-Generation Sequencing zu screenen, sobald sie eine systemische Therapie benötigen oder aber spätestens dann, wenn sie kein Therapieansprechen auf Checkpoint-Inhibitoren zeigen.
Collapse
Affiliation(s)
| | - Stephan Forchhammer
- Mikroskopische und molekulare Dermatologie, Universitäts-Hautklinik Tübingen
| | - Irina Bonzheim
- Institut für Pathologie und Neuropathologie, Universitätsklinikum Tübingen
| |
Collapse
|
25
|
Bartlett MJ, Flores AJ, Ye T, Smidt SI, Dollish HK, Stancati JA, Farrell DC, Parent KL, Doyle KP, Besselsen DG, Heien ML, Cowen SL, Steece-Collier K, Sherman SJ, Falk T. Preclinical evidence in support of repurposing sub-anesthetic ketamine as a treatment for L-DOPA-induced dyskinesia. Exp Neurol 2020; 333:113413. [PMID: 32717354 PMCID: PMC7518549 DOI: 10.1016/j.expneurol.2020.113413] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 02/08/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease. Pharmacotherapy with L-DOPA remains the gold-standard therapy for PD, but is often limited by the development of the common side effect of L-DOPA-induced dyskinesia (LID), which can become debilitating. The only effective treatment for disabling dyskinesia is surgical therapy (neuromodulation or lesioning), therefore effective pharmacological treatment of LID is a critical unmet need. Here, we show that sub-anesthetic doses of ketamine attenuate the development of LID in a rodent model, while also having acute anti-parkinsonian activity. The long-term anti-dyskinetic effect is mediated by brain-derived neurotrophic factor-release in the striatum, followed by activation of ERK1/2 and mTOR pathway signaling. This ultimately leads to morphological changes in dendritic spines on striatal medium spiny neurons that correlate with the behavioral effects, specifically a reduction in the density of mushroom spines, a dendritic spine phenotype that shows a high correlation with LID. These molecular and cellular changes match those occurring in hippocampus and cortex after effective sub-anesthetic ketamine treatment in preclinical models of depression, and point to common mechanisms underlying the therapeutic efficacy of ketamine for these two disorders. These preclinical mechanistic studies complement current ongoing clinical testing of sub-anesthetic ketamine for the treatment of LID by our group, and provide further evidence in support of repurposing ketamine to treat individuals with PD. Given its clinically proven therapeutic benefit for both treatment-resistant depression and several pain states, very common co-morbidities in PD, sub-anesthetic ketamine could provide multiple therapeutic benefits for PD in the future.
Collapse
Affiliation(s)
- Mitchell J Bartlett
- Department of Neurology, The University of Arizona, Tucson, AZ 85724, USA; Department of Pharmacology, The University of Arizona, Tucson, AZ 85724, USA
| | - Andrew J Flores
- Department of Neurology, The University of Arizona, Tucson, AZ 85724, USA; Graduate Interdisciplinary Program in Physiological Sciences, The University of Arizona, Tucson, AZ 85724, USA
| | - Tony Ye
- Department of Neurology, The University of Arizona, Tucson, AZ 85724, USA
| | - Saskia I Smidt
- Department of Neurology, The University of Arizona, Tucson, AZ 85724, USA
| | - Hannah K Dollish
- Graduate Interdisciplinary Program in Neuroscience, The University of Arizona, Tucson, AZ 85724, USA
| | - Jennifer A Stancati
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI 49503, USA
| | - Drew C Farrell
- Department of Chemistry & Biochemistry, The University of Arizona, Tucson, AZ 85721, USA
| | - Kate L Parent
- Department of Chemistry & Biochemistry, The University of Arizona, Tucson, AZ 85721, USA
| | - Kristian P Doyle
- Department of Neurology, The University of Arizona, Tucson, AZ 85724, USA; Department of Immunobiology, The University of Arizona, Tucson, AZ 85724, USA
| | - David G Besselsen
- University Animal Care, The University of Arizona, Tucson, AZ 85724, USA
| | - Michael L Heien
- Department of Chemistry & Biochemistry, The University of Arizona, Tucson, AZ 85721, USA
| | - Stephen L Cowen
- Department of Psychology, The University of Arizona, Tucson, AZ 85724, USA
| | - Kathy Steece-Collier
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI 49503, USA
| | - Scott J Sherman
- Department of Neurology, The University of Arizona, Tucson, AZ 85724, USA
| | - Torsten Falk
- Department of Neurology, The University of Arizona, Tucson, AZ 85724, USA; Department of Pharmacology, The University of Arizona, Tucson, AZ 85724, USA; Graduate Interdisciplinary Program in Physiological Sciences, The University of Arizona, Tucson, AZ 85724, USA; Graduate Interdisciplinary Program in Neuroscience, The University of Arizona, Tucson, AZ 85724, USA.
| |
Collapse
|
26
|
Emili M, Guidi S, Uguagliati B, Giacomini A, Bartesaghi R, Stagni F. Treatment with the flavonoid 7,8-Dihydroxyflavone: a promising strategy for a constellation of body and brain disorders. Crit Rev Food Sci Nutr 2020; 62:13-50. [DOI: 10.1080/10408398.2020.1810625] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Marco Emili
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Sandra Guidi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Beatrice Uguagliati
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Andrea Giacomini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Renata Bartesaghi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Fiorenza Stagni
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| |
Collapse
|
27
|
Lee HW, Ahmad M, Wang HW, Leenen FHH. Effects of exercise on BDNF-TrkB signaling in the paraventricular nucleus and rostral ventrolateral medulla in rats post myocardial infarction. Neuropeptides 2020; 82:102058. [PMID: 32507324 DOI: 10.1016/j.npep.2020.102058] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 05/19/2020] [Accepted: 05/20/2020] [Indexed: 02/07/2023]
Abstract
Brain-derived neurotrophic factor (BDNF)-tropomyosin-related kinase B (TrkB) signaling in the paraventricular nucleus (PVN) and rostral ventrolateral medulla (RVLM) is associated with cardiovascular regulation. Exercise increases plasma BDNF and attenuates activation of central pathways in the PVN and RVLM post myocardial infarction (MI). The present study assessed whether MI alters BDNF-TrkB signaling and intracellular factors Ca2+/calmodulin-dependent protein kinase II (CaMKII) and Akt in the PVN and RVLM of male Wistar rats with or without exercise or treatment with the TrkB blocker ANA-12. A 4-week period of treadmill exercise training was performed in MI rats. A separate experiment was conducted with 2.5 mg/kg ANA-12 in sedentary MI rats. At 5 weeks post MI, in both the PVN and RVLM, the ratio of full-length TrkB (TrkB.FL) and truncated TrkB (TrkB.T1) was decreased. 0.5 mg/kg ANA-12 did not affect BDNF-TrkB signaling and cardiac function post MI, but 2.5 mg/kg ANA-12 further decreased ejection fraction (EF). Exercise increased mature BDNF (mBDNF) and decreased Akt activity in the PVN, whereas in the RVLM, exercise did not affect mBDNF but lowered p-CaMKIIβ. ANA-12 prevented the exercise-induced increase in mBDNF in the PVN and decrease in p-CaMKIIβ in the RVLM. In conclusion, exercise decreases Akt activity in the PVN and decreases p-CaMKIIβ in the RVLM post MI. BDNF-TrkB signaling only mediates the decrease in p-CaMKIIβ in the RVLM. The exercise-induced decreases in Akt activity in the PVN and p-CaMKIIβ in the RVLM may contribute to the attenuation of the decrease in EF and sympathetic hyperactivity post MI.
Collapse
Affiliation(s)
- Heow Won Lee
- Brain and Heart Research Group, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Monir Ahmad
- Brain and Heart Research Group, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Hong-Wei Wang
- Brain and Heart Research Group, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Frans H H Leenen
- Brain and Heart Research Group, University of Ottawa Heart Institute, Ottawa, Ontario, Canada.
| |
Collapse
|
28
|
Forschner A, Forchhammer S, Bonzheim I. NTRK gene fusions in melanoma: detection, prevalence and potential therapeutic implications. J Dtsch Dermatol Ges 2020; 18:1387-1392. [PMID: 32656925 DOI: 10.1111/ddg.14160] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 03/09/2020] [Indexed: 02/06/2023]
Abstract
Fusions involving neurotrophic tyrosine receptor kinase (NTRK) are known drivers of oncogenesis and also occur in melanoma, although very rarely. A particularly high incidence of NTRK gene fusions is reported in infantile fibrosarcoma (> 90 %) or the secretory type of breast cancer (> 90 %). Recently, larotrectinib (a tropomyosin receptor kinase [TRK] inhibitor) was approved, and we wondered whether TRK inhibitors might also be helpful for melanoma patients. We therefore screened the literature and obtained relevant results. NTRK fusions are relatively common in spitzoid melanoma, with a prevalence of 21-29 % compared to < 1 % in cutaneous or mucosal melanoma and 2.5 % in acral melanoma. It appears that fusion proteins are mutually exclusive for most common oncogenic drivers such as BRAF or NRAS. A further indicator of an increased probability of detecting NTRK-positive tumors could be a low mutation load. Since TRK inhibitors are already available for patients with NTRK fusions, the challenge will be to implement screening for NTRK gene fusions in clinical practice. A possible approach could be to screen BRAF, NRAS and KIT wild-type melanoma patients with next-generation sequencing as soon as they need systemic treatment or at the latest when they have no tumor control on checkpoint inhibitors.
Collapse
Affiliation(s)
- Andrea Forschner
- Department of Dermatology, Center for Dermatooncology, University Hospital Tübingen, Tübingen, Germany
| | - Stephan Forchhammer
- Department of Dermatology, Histopathology, University Hospital Tübingen, Tübingen, Germany
| | - Irina Bonzheim
- Institute of Pathology and neuropathology, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
29
|
Glucocorticoid and brain-derived neurotrophic factor relationship: a brief investigation into the model of depression by chronic administration of corticosterone. Behav Pharmacol 2020; 31:407-412. [PMID: 32040016 DOI: 10.1097/fbp.0000000000000547] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Depression is considered a common mental disorder that affects more than 300 million people worldwide. Despite this high incidence, its etiology is not completely elucidated instigating further studies. For this purpose, different animal models are used to study routes and molecular changes involved in depression, among them the chronic administration of corticosterone. However, the knowledge about neurochemical changes after this protocol is still controversial. In this work, we evaluated serum corticosterone levels, adrenal/body weight ratio, as well as glucocorticoid receptor and brain-derived neurotrophic factor protein expression and its receptor, tropomyosin-receptor kinase B. These analyzes were performed on prefrontal cortex, hippocampus, and striatum samples taken of mice after 21 days of administration of corticosterone. Exposure to corticosterone reduced the serum corticosterone levels and the adrenal/body weight ratio. Moreover, the glucocorticoid receptor and tyrosine-receptor kinase B expression were increased in the hippocampus while the brain-derived neurotrophic factor expression was reduced in the prefrontal cortex. We also found a positive correlation between the expression of glucocorticoid receptor and tyrosine-receptor kinase B and our results suggest a possible relationship between the glucocorticoid/glucocorticoid receptor and brain-derived neurotrophic factor/tropomyosin-receptor kinase B routes after chronic corticosterone administration. To our knowledge, this is the first study that evaluate these parameters concomitantly in important mood-related structures. In addition, these results may be useful to other research groups seeking to explore new pathways and substances with therapeutic potential to treat this silent epidemic.
Collapse
|
30
|
Gambella A, Senetta R, Collemi G, Vallero SG, Monticelli M, Cofano F, Zeppa P, Garbossa D, Pellerino A, Rudà R, Soffietti R, Fagioli F, Papotti M, Cassoni P, Bertero L. NTRK Fusions in Central Nervous System Tumors: A Rare, but Worthy Target. Int J Mol Sci 2020; 21:ijms21030753. [PMID: 31979374 PMCID: PMC7037946 DOI: 10.3390/ijms21030753] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 12/11/2022] Open
Abstract
The neurotrophic tropomyosin receptor kinase (NTRK) genes (NTRK1, NTRK2, and NTRK3) code for three transmembrane high-affinity tyrosine-kinase receptors for nerve growth factors (TRK-A, TRK-B, and TRK-C) which are mainly involved in nervous system development. Loss of function alterations in these genes can lead to nervous system development problems; conversely, activating alterations harbor oncogenic potential, promoting cell proliferation/survival and tumorigenesis. Chromosomal rearrangements are the most clinically relevant alterations of pathological NTRK activation, leading to constitutionally active chimeric receptors. NTRK fusions have been detected with extremely variable frequencies in many pediatric and adult cancer types, including central nervous system (CNS) tumors. These alterations can be detected by different laboratory assays (e.g., immunohistochemistry, FISH, sequencing), but each of these approaches has specific advantages and limitations which must be taken into account for an appropriate use in diagnostics or research. Moreover, therapeutic targeting of this molecular marker recently showed extreme efficacy. Considering the overall lack of effective treatments for brain neoplasms, it is expected that detection of NTRK fusions will soon become a mainstay in the diagnostic assessment of CNS tumors, and thus in-depth knowledge regarding this topic is warranted.
Collapse
Affiliation(s)
- Alessandro Gambella
- Pathology Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (A.G.); (G.C.)
| | - Rebecca Senetta
- Pathology Unit, Department of Oncology, University of Turin, 10126 Turin, Italy; (R.S.); (M.P.)
| | - Giammarco Collemi
- Pathology Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (A.G.); (G.C.)
| | - Stefano Gabriele Vallero
- Pediatric Onco-Hematology Unit, Department of Pediatric and Public Health Sciences, University of Turin, 10126 Turin, Italy; (S.G.V.); (F.F.)
| | - Matteo Monticelli
- Neurosurgery Unit, Department of Neurosciences, University of Turin, 10126 Turin, Italy; (M.M.); (F.C.); (P.Z.); (D.G.)
| | - Fabio Cofano
- Neurosurgery Unit, Department of Neurosciences, University of Turin, 10126 Turin, Italy; (M.M.); (F.C.); (P.Z.); (D.G.)
| | - Pietro Zeppa
- Neurosurgery Unit, Department of Neurosciences, University of Turin, 10126 Turin, Italy; (M.M.); (F.C.); (P.Z.); (D.G.)
| | - Diego Garbossa
- Neurosurgery Unit, Department of Neurosciences, University of Turin, 10126 Turin, Italy; (M.M.); (F.C.); (P.Z.); (D.G.)
| | - Alessia Pellerino
- Department of Neuro-Oncology, University and City of Health and Science Hospital, 10126 Turin, Italy; (A.P.); (R.R.); (R.S.)
| | - Roberta Rudà
- Department of Neuro-Oncology, University and City of Health and Science Hospital, 10126 Turin, Italy; (A.P.); (R.R.); (R.S.)
| | - Riccardo Soffietti
- Department of Neuro-Oncology, University and City of Health and Science Hospital, 10126 Turin, Italy; (A.P.); (R.R.); (R.S.)
| | - Franca Fagioli
- Pediatric Onco-Hematology Unit, Department of Pediatric and Public Health Sciences, University of Turin, 10126 Turin, Italy; (S.G.V.); (F.F.)
| | - Mauro Papotti
- Pathology Unit, Department of Oncology, University of Turin, 10126 Turin, Italy; (R.S.); (M.P.)
| | - Paola Cassoni
- Pathology Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (A.G.); (G.C.)
| | - Luca Bertero
- Pathology Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (A.G.); (G.C.)
- Correspondence: ; Tel.: +39-011-633-5466
| |
Collapse
|
31
|
Wang H, Wei Y, Pu Y, Jiang D, Jiang X, Zhang Y, Tao J. Brain-derived neurotrophic factor stimulation of T-type Ca2+ channels in sensory neurons contributes to increased peripheral pain sensitivity. Sci Signal 2019; 12:12/600/eaaw2300. [DOI: 10.1126/scisignal.aaw2300] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Although brain-derived neurotrophic factor (BDNF) is implicated in the nociceptive signaling of peripheral sensory neurons, the underlying mechanisms remain largely unknown. Here, we elucidated the effects of BDNF on the neuronal excitability of trigeminal ganglion (TG) neurons and the pain sensitivity of rats mediated by T-type Ca2+ channels. BDNF reversibly and dose-dependently enhanced T-type channel currents through the activation of tropomyosin receptor kinase B (TrkB). Antagonism of phosphatidylinositol 3-kinase (PI3K) but not of its downstream target, the kinase AKT, abolished the BDNF-induced T-type channel response. BDNF application activated p38 mitogen-activated protein kinase (MAPK), and this effect was prevented by inhibition of PI3K but not of protein kinase A (PKA). Antagonism of either PI3K or p38 MAPK prevented the BDNF-induced stimulation of PKA activity, whereas PKA inhibition blocked the BDNF-mediated increase in T-type currents. BDNF increased the rate of action potential firing in TG neurons and enhanced the pain sensitivity of rats to mechanical stimuli. Moreover, inhibition of TrkB signaling abolished the increased mechanical sensitivity in a rat model of chronic inflammatory pain, and this effect was attenuated by either T-type channel blockade or knockdown of the channel Cav3.2. Together, our findings indicate that BDNF enhances T-type currents through the stimulation of TrkB coupled to PI3K-p38-PKA signaling, thereby inducing neuronal hyperexcitability of TG neurons and pain hypersensitivity in rats.
Collapse
|
32
|
Dedoni S, Marras L, Olianas MC, Ingianni A, Onali P. Downregulation of TrkB Expression and Signaling by Valproic Acid and Other Histone Deacetylase Inhibitors. J Pharmacol Exp Ther 2019; 370:490-503. [PMID: 31308194 DOI: 10.1124/jpet.119.258129] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 06/14/2019] [Indexed: 01/27/2023] Open
Abstract
Valproic acid (VPA) has been shown to regulate the levels of brain-derived neurotrophic factor (BDNF), but it is not known whether this drug can affect the neuronal responses to BDNF. In the present study, we show that in retinoic acid-differentiated SH-SY5Y human neuroblastoma cells, prolonged exposure to VPA reduces the expression of the BDNF receptor TrkB at the protein and mRNA levels and inhibits the intracellular signaling, neurotrophic activity, and prosurvival function of BDNF. VPA downregulates TrkB and curtails BDNF-induced signaling also in differentiated Kelly and LAN-1 neuroblastoma cells and primary mouse cortical neurons. The VPA effect is mimicked by several histone deacetylase (HDAC) inhibitors, including the class I HDAC inhibitors entinostat and romidepsin. Conversely, the class II HDAC inhibitor MC1568, the HDAC6 inhibitor tubacin, the HDAC8 inhibitor PCI-34051, and the VPA derivative valpromide have no effect. In neuroblastoma cells and primary neurons both VPA and entinostat increase the cellular levels of the transcription factor RUNX3, which negatively regulates TrkB gene expression. Treatment with RUNX3 siRNA attenuates VPA-induced RUNX3 elevation and TrkB downregulation. VPA, entinostat, HDAC1 depletion by siRNA, and 3-deazaneplanocin A (DZNep), an inhibitor of the polycomb repressor complex 2 (PRC2), decrease the PRC2 core component EZH2, a RUNX3 suppressor. Like VPA, HDAC1 depletion and DZNep increase RUNX3 and decrease TrkB expression. These results indicate that VPA downregulates TrkB through epigenetic mechanisms involving the EZH2/RUNX3 axis and provide evidence that this effect implicates relevant consequences with regard to BDNF efficacy in stimulating intracellular signaling and functional responses. SIGNIFICANCE STATEMENT: The tropomyosin-related kinase receptor B (TrkB) mediates the stimulatory effects of brain-derived neurotrophic factor (BDNF) on neuronal growth, differentiation, and survival and is highly expressed in aggressive neuroblastoma and other tumors. Here we show that exposure to valproic acid (VPA) downregulates TrkB expression and functional activity in retinoic acid-differentiated human neuroblastoma cell lines and primary mouse cortical neurons. The effects of VPA are mimicked by other histone deacetylase (HDAC) inhibitors and HDAC1 knockdown and appear to be mediated by an epigenetic mechanism involving the upregulation of RUNX3, a suppressor of TrkB gene expression. TrkB downregulation may have relevance for the use of VPA as a potential therapeutic agent in neuroblastoma and other pathologies characterized by an excessive BDNF/TrkB signaling.
Collapse
Affiliation(s)
- Simona Dedoni
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences, Department of Biomedical Sciences (S.D., M.C.O., P.O.) and Section of Microbiology, Department of Biomedical Sciences (L.M., A.I.), University of Cagliari, Cagliari, Italy
| | - Luisa Marras
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences, Department of Biomedical Sciences (S.D., M.C.O., P.O.) and Section of Microbiology, Department of Biomedical Sciences (L.M., A.I.), University of Cagliari, Cagliari, Italy
| | - Maria C Olianas
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences, Department of Biomedical Sciences (S.D., M.C.O., P.O.) and Section of Microbiology, Department of Biomedical Sciences (L.M., A.I.), University of Cagliari, Cagliari, Italy
| | - Angela Ingianni
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences, Department of Biomedical Sciences (S.D., M.C.O., P.O.) and Section of Microbiology, Department of Biomedical Sciences (L.M., A.I.), University of Cagliari, Cagliari, Italy
| | - Pierluigi Onali
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences, Department of Biomedical Sciences (S.D., M.C.O., P.O.) and Section of Microbiology, Department of Biomedical Sciences (L.M., A.I.), University of Cagliari, Cagliari, Italy
| |
Collapse
|
33
|
Almoguera B, McGinnis E, Abrams D, Vazquez L, Cederquist A, Sleiman PM, Dlugos D, Hakonarson H, Cagan A, Connolly J, Gainer VS, Garifallou J, Kaminski C, Lee YC, Mafra F, Mentch F, Pellegrino R, Qiu H, Snyder J, Tian L, Wang F, Manolio TA, Manzi S, Holm IA, Karlson EW. Drug-resistant epilepsy classified by a phenotyping algorithm associates with NTRK2. Acta Neurol Scand 2019; 140:169-176. [PMID: 31070779 DOI: 10.1111/ane.13115] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 04/16/2019] [Accepted: 05/03/2019] [Indexed: 01/31/2023]
Abstract
OBJECTIVE Up to 40% of patients with epilepsy become drug resistant (DRE). Genetic factors are likely to play a role. While efforts have focused on the transporter and target hypotheses, neither of them fully explains the pan-pharmacoresistance seen in DRE. MATERIALS AND METHODS In this study, we developed and used a phenotyping algorithm for the identification of DRE, responders, and epilepsy-free controls that were sequenced using a gene panel developed by the Pharmacogenomics Research Network (PGRN), which includes 82 genes involved in drug response. We tested the transporter hypothesis of DRE, the association between drug resistance and variants in the ATP-binding cassette family of genes previously associated with DRE, and also investigated potential new genetic factors. RESULTS In the analysis of DRE vs controls, NTRK2 was significantly associated with DRE (rs76950094; P = 1.19 × 10-7 and gene-based P-value = 1.67 × 10-4 ). NTRK2 encodes TrkB, which is involved in the development and maturation of the central nervous system, and increased activation of TrkB signaling is suggested to promote epilepsy. CONCLUSION Although the role of NTRK2 in DRE needs to be elucidated, these results support alternative mechanisms underlying DRE, complementary to the existing hypotheses, that should be evaluated.
Collapse
Affiliation(s)
- Berta Almoguera
- Center for Applied Genomics Children's Hospital of Philadelphia Philadelphia Pennsylvania
| | - Emily McGinnis
- Department of Neurology Children's Hospital of Philadelphia Philadelphia Pennsylvania
| | - Debra Abrams
- Center for Applied Genomics Children's Hospital of Philadelphia Philadelphia Pennsylvania
| | - Lyam Vazquez
- Center for Applied Genomics Children's Hospital of Philadelphia Philadelphia Pennsylvania
| | - Anna Cederquist
- Center for Applied Genomics Children's Hospital of Philadelphia Philadelphia Pennsylvania
| | - Patrick M. Sleiman
- Center for Applied Genomics Children's Hospital of Philadelphia Philadelphia Pennsylvania
- Department of Pediatrics, The Perelman School of Medicine University of Pennsylvania Philadelphia Pennsylvania
| | - Dennis Dlugos
- Department of Neurology Children's Hospital of Philadelphia Philadelphia Pennsylvania
| | - Hakon Hakonarson
- Center for Applied Genomics Children's Hospital of Philadelphia Philadelphia Pennsylvania
- Department of Pediatrics, The Perelman School of Medicine University of Pennsylvania Philadelphia Pennsylvania
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Mamdani H, Jalal SI. Breakthroughs and challenges in the management of tropomyosin receptor kinase fusion-positive tumors. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:S155. [PMID: 31576362 DOI: 10.21037/atm.2019.06.41] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Hirva Mamdani
- Karmanos Cancer Institute/Wayne State University, Detroit, MI, USA
| | - Shadia I Jalal
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN, USA
| |
Collapse
|
35
|
Qi X, Liu J, Wu J, Bi Y, Han C, Zhang G, Lou M, Lu J, Tang J. Initiating TrkB/Akt Signaling Cascade Preserves Blood-Brain Barrier after Subarachnoid Hemorrhage in Rats. Cell Transplant 2019; 28:1002-1008. [PMID: 31208230 PMCID: PMC6728706 DOI: 10.1177/0963689719857649] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The integrity of the blood–brain barrier (BBB) plays a vital role in affecting the
prognosis of subarachnoid hemorrhage (SAH). This study aimed to investigate activation of
the Tropomyosin-related kinase receptor B (TrkB) and its downstream signaling pathway on
preserving BBB breakdown after experimental SAH. An endovascular perforation SAH model was
applied. N-[2-(5-hydroxy-1H-indol-3-yl) ethyl]-2- oxopiperidine-3-carboxamide (HIOC), the
derivative of N-acetyl serotonin (NAS), was intracerebroventricularly administered 3 h
after SAH induction. The neurologic scores and brain water content were evaluated in an
outcome study. Western blot and immunofluorescence staining were used to investigate the
mechanism. The results indicated that HIOC activated the TrkB/Akt pathway, increased the
tight junction expression, improved neurologic deficits, and ameliorated brain edema after
SAH. Thus, we conclude that initiating the TrkB/Akt signaling cascade preserves BBB
breakdown after experimental SAH in rats.
Collapse
Affiliation(s)
- Xiangqian Qi
- 1 Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Both authors are the co-authors of this article
| | - Juan Liu
- 1 Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Both authors are the co-authors of this article
| | - Jiejin Wu
- 1 Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunke Bi
- 1 Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cong Han
- 1 Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guiyun Zhang
- 1 Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meiqing Lou
- 1 Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianfei Lu
- 2 Discipline of Neuroscience, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junjia Tang
- 1 Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
36
|
Abstract
Selective serotonin reuptake inhibitors are among the most prescribed antidepressants. Fluoxetine is the lead molecule which exerts its therapeutic effects, at least in part, by promoting neuroplasticity through increased brain-derived neurotrophic factor (BDNF)/tropomyosin-related receptor kinase B (TrkB) signalling. It is unclear however, to which extent the neuroplastic effects of fluoxetine are solely mediated by the inhibition of the serotonin transporter (5-HTT). To answer this question, the effects of fluoxetine on neuroplasticity were analysed in both wild type (WT) and 5-Htt knock-out (KO) mice. Using Western blotting and RT-qPCR approaches, we showed that fluoxetine 10 µM activated BDNF/TrkB signalling pathways in both CD1 and C57BL/6J mouse primary cortical neurons. Interestingly, effects on BDNF signalling were observed in primary cortical neurons from both 5-Htt WT and KO mice. In addition, a 3-week in vivo fluoxetine treatment (15 mg/kg/d; i.p.) increased the expression of plasticity genes in brains of both 5-Htt WT and KO mice, and tended to equally enhance hippocampal cell proliferation in both genotypes, without reaching significance. Our results further suggest that fluoxetine-induced neuroplasticity does not solely depend on 5-HTT blockade, but might rely, at least in part, on 5-HTT-independent direct activation of TrkB.
Collapse
|
37
|
Aydin-Abidin S, Abidin İ. 7,8-Dihydroxyflavone potentiates ongoing epileptiform activity in mice brain slices. Neurosci Lett 2019; 703:25-31. [PMID: 30880161 DOI: 10.1016/j.neulet.2019.03.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 02/22/2019] [Accepted: 03/09/2019] [Indexed: 12/14/2022]
Abstract
In the central nervous system, Tropomyosin-receptor-kinase B (TrkB) signaling is involved in neuronal survival, differentiation as well as in regulation of synaptic transmission and excitability. As its powerful potential to modulate neuronal functions, TrkB pathway is an attractive target for novel drugs and treatment of common neurological disorders. 7,8-Dihydroxyflavone (DHF), a TrkB receptor agonist, has similar properties with neurotrophin Brain Derived Neurotropic Factor (BDNF). DHF is reported to have a number of beneficial effects in neuroprotection, against depression and improving learning and memory. However, the outcome of acute application of DHF on the excitability of neuronal circuits is not clear. Especially the effects of DHF on synchronized epileptiform activity are not known. In this study, we investigated whether DHF induces epileptiform activity in brain slices and DHF has any effect on already initiated epileptiform discharges. We used acute horizontal hippocampal-entorhinal cortex slices obtained from 30 to 35 days of mice. Extracellular field potential recordings were obtained from entorhinal cortex (EC) and hippocampus CA1 region. DHF did not initiate any epileptiform activity or abnormal discharges. However, DHF increased the frequency of 4 aminopyridine (4AP) induced ictal and interictal events in both EC and CA1. The duration of induced ictal charges were also prolonged upon DHF application. In a number of slices, both EC and CA1, DHF led to ictogenesis. These results suggest that the acute activation of TrkB by DHF has a powerful potential on synchronized neuronal discharges which should be considered in future therapeutical approaches.
Collapse
Affiliation(s)
- Selcen Aydin-Abidin
- Department of Biophysics, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey.
| | - İsmail Abidin
- Department of Biophysics, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| |
Collapse
|
38
|
Levy MJF, Boulle F, Steinbusch HW, van den Hove DLA, Kenis G, Lanfumey L. Neurotrophic factors and neuroplasticity pathways in the pathophysiology and treatment of depression. Psychopharmacology (Berl) 2018; 235:2195-2220. [PMID: 29961124 PMCID: PMC6061771 DOI: 10.1007/s00213-018-4950-4] [Citation(s) in RCA: 184] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 06/18/2018] [Indexed: 02/06/2023]
Abstract
Depression is a major health problem with a high prevalence and a heavy socioeconomic burden in western societies. It is associated with atrophy and impaired functioning of cortico-limbic regions involved in mood and emotion regulation. It has been suggested that alterations in neurotrophins underlie impaired neuroplasticity, which may be causally related to the development and course of depression. Accordingly, mounting evidence suggests that antidepressant treatment may exert its beneficial effects by enhancing trophic signaling on neuronal and synaptic plasticity. However, current antidepressants still show a delayed onset of action, as well as lack of efficacy. Hence, a deeper understanding of the molecular and cellular mechanisms involved in the pathophysiology of depression, as well as in the action of antidepressants, might provide further insight to drive the development of novel fast-acting and more effective therapies. Here, we summarize the current literature on the involvement of neurotrophic factors in the pathophysiology and treatment of depression. Further, we advocate that future development of antidepressants should be based on the neurotrophin theory.
Collapse
Affiliation(s)
- Marion J F Levy
- Centre de Psychiatrie et Neurosciences (Inserm U894), Université Paris Descartes, 102-108 rue de la santé, 75014, Paris, France
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands
- EURON-European Graduate School of Neuroscience, Maastricht, The Netherlands
| | - Fabien Boulle
- Centre de Psychiatrie et Neurosciences (Inserm U894), Université Paris Descartes, 102-108 rue de la santé, 75014, Paris, France
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands
- EURON-European Graduate School of Neuroscience, Maastricht, The Netherlands
| | - Harry W Steinbusch
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands
- EURON-European Graduate School of Neuroscience, Maastricht, The Netherlands
| | - Daniël L A van den Hove
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands
- EURON-European Graduate School of Neuroscience, Maastricht, The Netherlands
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Wuerzburg, Wuerzburg, Germany
| | - Gunter Kenis
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands
- EURON-European Graduate School of Neuroscience, Maastricht, The Netherlands
| | - Laurence Lanfumey
- Centre de Psychiatrie et Neurosciences (Inserm U894), Université Paris Descartes, 102-108 rue de la santé, 75014, Paris, France.
- EURON-European Graduate School of Neuroscience, Maastricht, The Netherlands.
| |
Collapse
|
39
|
Schverer M, Lanfumey L, Baulieu EE, Froger N, Villey I. Neurosteroids: non-genomic pathways in neuroplasticity and involvement in neurological diseases. Pharmacol Ther 2018; 191:190-206. [PMID: 29953900 DOI: 10.1016/j.pharmthera.2018.06.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Neurosteroids are neuroactive brain-born steroids. They can act through non-genomic and/or through genomic pathways. Genomic pathways are largely described for steroid hormones: the binding to nuclear receptors leads to transcription regulation. Pregnenolone, Dehydroepiandrosterone, their respective sulfate esters and Allopregnanolone have no corresponding nuclear receptor identified so far whereas some of their non-genomic targets have been identified. Neuroplasticity is the capacity that neuronal networks have to change their structure and function in response to biological and/or environmental signals; it is regulated by several mechanisms, including those that involve neurosteroids. In this review, after a description of their biosynthesis, the effects of Pregnenolone, Dehydroepiandrosterone, their respective sulfate esters and Allopregnanolone on their targets will be exposed. We then shall highlight that neurosteroids, by acting on these targets, can regulate neurogenesis, structural and functional plasticity. Finally, we will discuss the therapeutic potential of neurosteroids in the pathophysiology of neurological diseases in which alterations of neuroplasticity are associated with changes in neurosteroid levels.
Collapse
Affiliation(s)
- Marina Schverer
- Inserm U894, Centre de Psychiatrie et Neurosciences, Université Paris Descartes, 75014 Paris, France
| | - Laurence Lanfumey
- Inserm U894, Centre de Psychiatrie et Neurosciences, Université Paris Descartes, 75014 Paris, France.
| | - Etienne-Emile Baulieu
- MAPREG SAS, Le Kremlin-Bicêtre, France; Inserm UMR 1195, Université Paris-Saclay, Le Kremlin Bicêtre, France
| | | | | |
Collapse
|
40
|
Martínez-Ballesta MC, Gil-Izquierdo Á, García-Viguera C, Domínguez-Perles R. Nanoparticles and Controlled Delivery for Bioactive Compounds: Outlining Challenges for New "Smart-Foods" for Health. Foods 2018; 7:E72. [PMID: 29735897 PMCID: PMC5977092 DOI: 10.3390/foods7050072] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 04/27/2018] [Accepted: 05/04/2018] [Indexed: 12/28/2022] Open
Abstract
Nanotechnology is a field of research that has been stressed as a very valuable approach for the prevention and treatment of different human health disorders. This has been stressed as a delivery system for the therapeutic fight against an array of pathophysiological situations. Actually, industry has applied this technology in the search for new oral delivery alternatives obtained upon the modification of the solubility properties of bioactive compounds. Significant works have been made in the last years for testing the input that nanomaterials and nanoparticles provide for an array of pathophysiological situations. In this frame, this review addresses general questions concerning the extent to which nanoparticles offer alternatives that improve therapeutic value, while avoid toxicity, by releasing bioactive compounds specifically to target tissues affected by specific chemical and pathophysiological settings. In this regard, to date, the contribution of nanoparticles to protect encapsulated bioactive compounds from degradation as a result of gastrointestinal digestion and cellular metabolism, to enable their release in a controlled manner, enhancing biodistribution of bioactive compounds, and to allow them to target those tissues affected by biological disturbances has been demonstrated.
Collapse
Affiliation(s)
- MCarment Martínez-Ballesta
- Department of Plant Nutrition, Centro de Edafología y Biología Aplicada del Segura-Spanish Council for Scientific Research (CEBAS-CSIC), Campus de Espinardo 25, 30100 Espinardo, Murcia, Spain.
| | - Ángel Gil-Izquierdo
- Research Group on Quality, Safety and Bioactivity of Plant Foods, Department of Food Science and Technology, Centro de Edafología y Biología Aplicada del Segura-Spanish Council for Scientific Research (CEBAS-CSIC), Campus de Espinardo 25, 30100 Espinardo, Murcia, Spain.
| | - Cristina García-Viguera
- Research Group on Quality, Safety and Bioactivity of Plant Foods, Department of Food Science and Technology, Centro de Edafología y Biología Aplicada del Segura-Spanish Council for Scientific Research (CEBAS-CSIC), Campus de Espinardo 25, 30100 Espinardo, Murcia, Spain.
| | - Raúl Domínguez-Perles
- Research Group on Quality, Safety and Bioactivity of Plant Foods, Department of Food Science and Technology, Centro de Edafología y Biología Aplicada del Segura-Spanish Council for Scientific Research (CEBAS-CSIC), Campus de Espinardo 25, 30100 Espinardo, Murcia, Spain.
| |
Collapse
|
41
|
Klein P, Dingledine R, Aronica E, Bernard C, Blümcke I, Boison D, Brodie MJ, Brooks-Kayal AR, Engel J, Forcelli PA, Hirsch LJ, Kaminski RM, Klitgaard H, Kobow K, Lowenstein DH, Pearl PL, Pitkänen A, Puhakka N, Rogawski MA, Schmidt D, Sillanpää M, Sloviter RS, Steinhäuser C, Vezzani A, Walker MC, Löscher W. Commonalities in epileptogenic processes from different acute brain insults: Do they translate? Epilepsia 2018; 59:37-66. [PMID: 29247482 PMCID: PMC5993212 DOI: 10.1111/epi.13965] [Citation(s) in RCA: 201] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2017] [Indexed: 12/12/2022]
Abstract
The most common forms of acquired epilepsies arise following acute brain insults such as traumatic brain injury, stroke, or central nervous system infections. Treatment is effective for only 60%-70% of patients and remains symptomatic despite decades of effort to develop epilepsy prevention therapies. Recent preclinical efforts are focused on likely primary drivers of epileptogenesis, namely inflammation, neuron loss, plasticity, and circuit reorganization. This review suggests a path to identify neuronal and molecular targets for clinical testing of specific hypotheses about epileptogenesis and its prevention or modification. Acquired human epilepsies with different etiologies share some features with animal models. We identify these commonalities and discuss their relevance to the development of successful epilepsy prevention or disease modification strategies. Risk factors for developing epilepsy that appear common to multiple acute injury etiologies include intracranial bleeding, disruption of the blood-brain barrier, more severe injury, and early seizures within 1 week of injury. In diverse human epilepsies and animal models, seizures appear to propagate within a limbic or thalamocortical/corticocortical network. Common histopathologic features of epilepsy of diverse and mostly focal origin are microglial activation and astrogliosis, heterotopic neurons in the white matter, loss of neurons, and the presence of inflammatory cellular infiltrates. Astrocytes exhibit smaller K+ conductances and lose gap junction coupling in many animal models as well as in sclerotic hippocampi from temporal lobe epilepsy patients. There is increasing evidence that epilepsy can be prevented or aborted in preclinical animal models of acquired epilepsy by interfering with processes that appear common to multiple acute injury etiologies, for example, in post-status epilepticus models of focal epilepsy by transient treatment with a trkB/PLCγ1 inhibitor, isoflurane, or HMGB1 antibodies and by topical administration of adenosine, in the cortical fluid percussion injury model by focal cooling, and in the albumin posttraumatic epilepsy model by losartan. Preclinical studies further highlight the roles of mTOR1 pathways, JAK-STAT3, IL-1R/TLR4 signaling, and other inflammatory pathways in the genesis or modulation of epilepsy after brain injury. The wealth of commonalities, diversity of molecular targets identified preclinically, and likely multidimensional nature of epileptogenesis argue for a combinatorial strategy in prevention therapy. Going forward, the identification of impending epilepsy biomarkers to allow better patient selection, together with better alignment with multisite preclinical trials in animal models, should guide the clinical testing of new hypotheses for epileptogenesis and its prevention.
Collapse
Affiliation(s)
- Pavel Klein
- Mid-Atlantic Epilepsy and Sleep Center, Bethesda, MD, USA
| | | | - Eleonora Aronica
- Department of (Neuro) Pathology, Academic Medical Center and Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
- Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, The Netherlands
| | - Christophe Bernard
- Aix Marseille Univ, Inserm, INS, Instit Neurosci Syst, Marseille, 13005, France
| | - Ingmar Blümcke
- Department of Neuropathology, University Hospital Erlangen, Erlangen, Germany
| | - Detlev Boison
- Robert Stone Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR, USA
| | - Martin J Brodie
- Epilepsy Unit, West Glasgow Ambulatory Care Hospital-Yorkhill, Glasgow, UK
| | - Amy R Brooks-Kayal
- Division of Neurology, Departments of Pediatrics and Neurology, University of Colorado School of Medicine, Aurora, CO, USA
- Children's Hospital Colorado, Aurora, CO, USA
- Neuroscience Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jerome Engel
- Departments of Neurology, Neurobiology, and Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, Brain Research Institute, University of California, Los Angeles, CA, USA
| | | | | | | | | | - Katja Kobow
- Department of Neuropathology, University Hospital Erlangen, Erlangen, Germany
| | | | - Phillip L Pearl
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Asla Pitkänen
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Noora Puhakka
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Michael A Rogawski
- Department of Neurology, University of California, Davis, Sacramento, CA, USA
| | | | - Matti Sillanpää
- Departments of Child Neurology and General Practice, University of Turku and Turku University Hospital, Turku, Finland
| | - Robert S Sloviter
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Christian Steinhäuser
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Annamaria Vezzani
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Institute for Pharmacological Research, Milan,, Italy
| | - Matthew C Walker
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, London, UK
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| |
Collapse
|
42
|
Boltaev U, Meyer Y, Tolibzoda F, Jacques T, Gassaway M, Xu Q, Wagner F, Zhang YL, Palmer M, Holson E, Sames D. Multiplex quantitative assays indicate a need for reevaluating reported small-molecule TrkB agonists. Sci Signal 2017; 10:10/493/eaal1670. [PMID: 28831019 DOI: 10.1126/scisignal.aal1670] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) and its receptor, tropomyosin-related kinase B (TrkB), have emerged as key regulators of brain plasticity and represent disease-modifying targets for several brain disorders, including Alzheimer's disease and major depressive disorder. Because of poor pharmacokinetic properties of BDNF, the interest in small-molecule TrkB agonists and modulators is high. Several compounds have been reported to act as TrkB agonists, and their increasing use in various nervous system disorder models creates the perception that these are reliable probes. To examine key pharmacological parameters of these compounds in detail, we have developed and optimized a series of complementary quantitative assays that measure TrkB receptor activation, TrkB-dependent downstream signaling, and gene expression in different cellular contexts. Although BDNF and other neurotrophic factors elicited robust and dose-dependent receptor activation and downstream signaling, we were unable to reproduce these activities using the reported small-molecule TrkB agonists. Our findings indicate that experimental results obtained with these compounds must be carefully interpreted and highlight the challenge of developing reliable pharmacological activators of this key molecular target.
Collapse
Affiliation(s)
- Umed Boltaev
- Department of Chemistry, Columbia University, New York, NY 10027, USA.,NeuroTechnology Center at Columbia University, New York, NY 10027, USA
| | - Yves Meyer
- Department of Chemistry, Columbia University, New York, NY 10027, USA
| | - Farangis Tolibzoda
- Department of Chemistry, Columbia University, New York, NY 10027, USA.,NeuroTechnology Center at Columbia University, New York, NY 10027, USA
| | - Teresa Jacques
- Department of Chemistry, Columbia University, New York, NY 10027, USA
| | - Madalee Gassaway
- Department of Chemistry, Columbia University, New York, NY 10027, USA.,NeuroTechnology Center at Columbia University, New York, NY 10027, USA
| | - Qihong Xu
- Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard, 7 Cambridge Center, Cambridge, MA 02142, USA
| | - Florence Wagner
- Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard, 7 Cambridge Center, Cambridge, MA 02142, USA
| | - Yan-Ling Zhang
- Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard, 7 Cambridge Center, Cambridge, MA 02142, USA
| | - Michelle Palmer
- Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard, 7 Cambridge Center, Cambridge, MA 02142, USA
| | - Edward Holson
- Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard, 7 Cambridge Center, Cambridge, MA 02142, USA
| | - Dalibor Sames
- Department of Chemistry, Columbia University, New York, NY 10027, USA. .,NeuroTechnology Center at Columbia University, New York, NY 10027, USA
| |
Collapse
|
43
|
Bar-Klein G, Klee R, Brandt C, Bankstahl M, Bascuñana P, Töllner K, Dalipaj H, Bankstahl JP, Friedman A, Löscher W. Isoflurane prevents acquired epilepsy in rat models of temporal lobe epilepsy. Ann Neurol 2017; 80:896-908. [PMID: 27761920 DOI: 10.1002/ana.24804] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 09/16/2016] [Accepted: 10/10/2016] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Acquired epilepsy is a devastating long-term risk of various brain insults, including trauma, stroke, infections, and status epilepticus (SE). There is no preventive treatment for patients at risk. Attributable to the complex alterations involved in epileptogenesis, it is likely that multitargeted approaches are required for epilepsy prevention. We report novel preclinical findings with isoflurane, which exerts various nonanesthetic effects that may be relevant for antiepileptogenesis. METHODS The effects of isoflurane were investigated in two rat models of SE-induced epilepsy: intrahippocampal kainate and systemic administration of paraoxon. Isoflurane was either administered during (kainate) or after (paraoxon) induction of SE. Magnetic resonance imaging was used to assess blood-brain barrier (BBB) dysfunction. Positron emission tomography was used to visualize neuroinflammation. Long-term electrocorticographic recordings were used to monitor spontaneous recurrent seizures. Neuronal damage was assessed histologically. RESULTS In the absence of isoflurane, spontaneous recurrent seizures were common in the majority of rats in both models. When isoflurane was administered during kainate injection, duration and severity of SE were not affected, but only few rats developed spontaneous recurrent seizures. A similar antiepileptogenic effect was found when paraoxon-treated rats were exposed to isoflurane after SE. Moreover, in the latter model, isoflurane prevented BBB dysfunction and neurodegeneration, whereas isoflurane reduced neuroinflammation in the kainate model. INTERPRETATION Given that isoflurane is a widely used volatile anesthetic, and is used for inhalational long-term sedation in critically ill patients at risk to develop epilepsy, our findings hold a promising potential to be successfully translated into the clinic. Ann Neurol 2016;80:896-908.
Collapse
Affiliation(s)
- Guy Bar-Klein
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, the Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Rebecca Klee
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Claudia Brandt
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Marion Bankstahl
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Pablo Bascuñana
- Department of Nuclear Medicine, Hannover Medical School, Germany
| | - Kathrin Töllner
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Hotjensa Dalipaj
- Department of Medical Neuroscience, Dalhousie University, Halifax, Canada
| | - Jens P Bankstahl
- Department of Nuclear Medicine, Hannover Medical School, Germany
| | - Alon Friedman
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, the Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Department of Medical Neuroscience, Dalhousie University, Halifax, Canada
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| |
Collapse
|
44
|
Tammiku-Taul J, Park R, Jaanson K, Luberg K, Dobchev DA, Kananovich D, Noole A, Mandel M, Kaasik A, Lopp M, Timmusk T, Karelson M. Indole-like Trk receptor antagonists. Eur J Med Chem 2016; 121:541-552. [DOI: 10.1016/j.ejmech.2016.06.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 06/01/2016] [Accepted: 06/02/2016] [Indexed: 01/08/2023]
|
45
|
Crowe EP, Tuzer F, Gregory BD, Donahue G, Gosai SJ, Cohen J, Leung YY, Yetkin E, Nativio R, Wang LS, Sell C, Bonini NM, Berger SL, Johnson FB, Torres C. Changes in the Transcriptome of Human Astrocytes Accompanying Oxidative Stress-Induced Senescence. Front Aging Neurosci 2016; 8:208. [PMID: 27630559 PMCID: PMC5005348 DOI: 10.3389/fnagi.2016.00208] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 08/15/2016] [Indexed: 01/23/2023] Open
Abstract
Aging is a major risk factor for many neurodegenerative disorders. A key feature of aging biology that may underlie these diseases is cellular senescence. Senescent cells accumulate in tissues with age, undergo widespread changes in gene expression, and typically demonstrate altered, pro-inflammatory profiles. Astrocyte senescence has been implicated in neurodegenerative disease, and to better understand senescence-associated changes in astrocytes, we investigated changes in their transcriptome using RNA sequencing. Senescence was induced in human fetal astrocytes by transient oxidative stress. Brain-expressed genes, including those involved in neuronal development and differentiation, were downregulated in senescent astrocytes. Remarkably, several genes indicative of astrocytic responses to injury were also downregulated, including glial fibrillary acidic protein and genes involved in the processing and presentation of antigens by major histocompatibility complex class II proteins, while pro-inflammatory genes were upregulated. Overall, our findings suggest that senescence-related changes in the function of astrocytes may impact the pathogenesis of age-related brain disorders.
Collapse
Affiliation(s)
- Elizabeth P Crowe
- Department of Pathology and Laboratory Medicine, Drexel University College of Medicine, Philadelphia PA, USA
| | - Ferit Tuzer
- Department of Pathology and Laboratory Medicine, Drexel University College of Medicine, Philadelphia PA, USA
| | - Brian D Gregory
- Department of Biology, Penn Genome Frontiers Institute, University of Pennsylvania, Philadelphia PA, USA
| | - Greg Donahue
- Epigenetics Program, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia PA, USA
| | - Sager J Gosai
- Department of Biology, Penn Genome Frontiers Institute, University of Pennsylvania, Philadelphia PA, USA
| | - Justin Cohen
- Department of Pathology and Laboratory Medicine, Drexel University College of Medicine, Philadelphia PA, USA
| | - Yuk Y Leung
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia PA, USA
| | - Emre Yetkin
- Department of Pathology and Laboratory Medicine, Drexel University College of Medicine, Philadelphia PA, USA
| | - Raffaella Nativio
- Epigenetics Program, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia PA, USA
| | - Li-San Wang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia PA, USA
| | - Christian Sell
- Department of Pathology and Laboratory Medicine, Drexel University College of Medicine, Philadelphia PA, USA
| | - Nancy M Bonini
- Department of Biology, Penn Genome Frontiers Institute, University of Pennsylvania, Philadelphia PA, USA
| | - Shelley L Berger
- Epigenetics Program, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia PA, USA
| | - F Brad Johnson
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia PA, USA
| | - Claudio Torres
- Department of Pathology and Laboratory Medicine, Drexel University College of Medicine, Philadelphia PA, USA
| |
Collapse
|
46
|
Vargas KG, Milic J, Zaciragic A, Wen KX, Jaspers L, Nano J, Dhana K, Bramer WM, Kraja B, van Beeck E, Ikram MA, Muka T, Franco OH. The functions of estrogen receptor beta in the female brain: A systematic review. Maturitas 2016; 93:41-57. [PMID: 27338976 DOI: 10.1016/j.maturitas.2016.05.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Accepted: 05/31/2016] [Indexed: 01/22/2023]
Abstract
Females have unique and additional risk factors for neurological disorders. Among classical estrogen receptors, estrogen receptor beta (ERβ) has been suggested as a therapeutic target. However, little is known about the role of ERβ in the female brain. Six electronic databases were searched for articles evaluating the role of ERβ in the female brain and the influence of age and menopause on ERβ function. After screening 3186 titles and abstracts, 49 articles were included in the review, all of which were animal studies. Of these, 19 focused on cellular signaling, 7 on neuroendocrine pathways, 8 on neurological disorders, 4 on neuroprotection and 19 on psychological and psychiatric outcomes (6 studies evaluated two or more outcomes). Our findings showed that ERβ phosphorylated and activated intracellular second messenger proteins and regulated protein expression of genes involved in neurological functions. It also promoted neurogenesis, modulated the neuroendocrine regulation of stress response, conferred neuroprotection against ischemia and inflammation, and reduced anxiety- and depression-like behaviors. Targeting ERβ may constitute a novel treatment for menopausal symptoms, including anxiety, depression, and neurological diseases. However, to establish potential therapeutic and preventive strategies targeting ERβ, future studies should be conducted in humans to further our understanding of the importance of ERβ in women's mental and cognitive health.
Collapse
Affiliation(s)
- Kris G Vargas
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Jelena Milic
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Asija Zaciragic
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Ke-Xin Wen
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Loes Jaspers
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Jana Nano
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Klodian Dhana
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | | | - Bledar Kraja
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands; Department of Biomedical Sciences, Faculty of Medicine, University of Medicine, Tirana, Albania; University Clinic of Gastrohepatology, University Hospital Center Mother Teresa, Tirana, Albania
| | - Ed van Beeck
- Department of Public Health, Erasmus University Medical Center, The Netherlands
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands; Department of Neurology, Erasmus University Medical Center, The Netherlands; Department of Radiology, Erasmus University Medical Center, The Netherlands
| | - Taulant Muka
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands.
| | - Oscar H Franco
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
47
|
Amatu A, Sartore-Bianchi A, Siena S. NTRK gene fusions as novel targets of cancer therapy across multiple tumour types. ESMO Open 2016; 1:e000023. [PMID: 27843590 PMCID: PMC5070277 DOI: 10.1136/esmoopen-2015-000023] [Citation(s) in RCA: 424] [Impact Index Per Article: 47.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 02/04/2016] [Indexed: 12/24/2022] Open
Abstract
The tropomyosin receptor kinase (Trk) receptor family comprises 3 transmembrane proteins referred to as Trk A, B and C (TrkA, TrkB and TrkC) receptors that are encoded by the NTRK1, NTRK2 and NTRK3 genes, respectively. These receptor tyrosine kinases are expressed in human neuronal tissue and play an essential role in the physiology of development and function of the nervous system through activation by neurotrophins. Gene fusions involving NTRK genes lead to transcription of chimeric Trk proteins with constitutively activated or overexpressed kinase function conferring oncogenic potential. These genetic abnormalities have recently emerged as targets for cancer therapy, because novel compounds have been developed that are selective inhibitors of the constitutively active rearranged proteins. Developments in this field are being aided by next generation sequencing methods as tools for unbiased gene fusions discovery. In this article, we review the role of NTRK gene fusions across several tumour histologies, and the promises and challenges of targeting such genetic alterations for cancer therapy.
Collapse
Affiliation(s)
- Alessio Amatu
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda , Milan , Italy
| | | | - Salvatore Siena
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Dipartimento di Oncologia e Emato-Oncologia, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
48
|
A common NTRK2 variant is associated with emotional arousal and brain white-matter integrity in healthy young subjects. Transl Psychiatry 2016; 6:e758. [PMID: 26978740 PMCID: PMC4872446 DOI: 10.1038/tp.2016.20] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 01/04/2016] [Accepted: 01/19/2016] [Indexed: 02/06/2023] Open
Abstract
Dysregulation of emotional arousal is observed in many psychiatric diseases such as schizophrenia, mood and anxiety disorders. The neurotrophic tyrosine kinase receptor type 2 gene (NTRK2) has been associated with these disorders. Here we investigated the relation between genetic variability of NTRK2 and emotional arousal in healthy young subjects in two independent samples (n1=1171; n2=707). In addition, diffusion tensor imaging (DTI) data in a subgroup of 342 participants were used to identify NTRK2-related white-matter structure differences. After correction for multiple testing, we identified a NTRK2 single nucleotide polymorphism associated with emotional arousal in both samples (n1: Pnominal=0.0003, Pcorrected=0.048; n2: Pnominal=0.0141, Pcorrected=0.036). DTI revealed significant, whole-brain corrected correlations between emotional arousal and brain white-matter mean diffusivity (MD), as well as significant, whole-brain corrected NTRK2 genotype-related differences in MD (PFWE<0.05). Our study demonstrates that genetic variability of NTRK2, a susceptibility gene for psychiatric disorders, is related to emotional arousal and-independently-to brain white-matter properties in healthy individuals.
Collapse
|
49
|
Shah AP, Carreno FR, Wu H, Chung YA, Frazer A. Role of TrkB in the anxiolytic-like and antidepressant-like effects of vagal nerve stimulation: Comparison with desipramine. Neuroscience 2016; 322:273-86. [PMID: 26899129 DOI: 10.1016/j.neuroscience.2016.02.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 01/23/2016] [Accepted: 02/09/2016] [Indexed: 12/27/2022]
Abstract
A current hypothesis regarding the mechanism of antidepressant (AD) action suggests the involvement of brain-derived neurotrophic factor (BDNF). Consistent with this hypothesis, the receptor for BDNF (and neurotrophin 4/5 (NT-4/5)), Tropomyosin-related kinase B (TrkB), is activated in rodents by treatment with classical AD drugs. Vagal nerve stimulation (VNS), a therapy for treatment resistant depression (TRD), also activates TrkB in rodents. However, the role of this receptor in the therapeutic effects of VNS is unclear. In the current study, the involvement of TrkB in the effects of VNS was investigated in rats using its inhibitor, K252a. Anxiolytic-like and AD-like effects were analyzed using the novelty suppressed feeding test (NSFT) and forced swim test (FST), respectively. K252a blocked the anxiolytic-like effect of chronic VNS treatment and the AD-like effect of acute VNS treatment. By contrast, blocking TrkB did not prevent either the anxiolytic-like or AD-like effect of chronic treatment with desipramine (DMI), a selective noradrenergic reuptake inhibitor; it did, however, block the acute effect of DMI in the FST. To examine whether the activation of TrkB caused by either VNS or DMI is ligand-dependent, use was made of TrkB-Fc, a molecular scavenger for ligands of TrkB. Intraventricular administration of TrkB-Fc blocked the acute activation of TrkB induced by either treatment, indicating that treatment-induced activation of this receptor is ligand-dependent. The behavioral results highlight differences in the involvement of TrkB in the chronic effects of an AD drug and a stimulation therapy as well as its role in acute versus chronic effects of DMI.
Collapse
Affiliation(s)
- A P Shah
- Department of Pharmacology and Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, TX, USA.
| | - F R Carreno
- Department of Pharmacology and Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, TX, USA
| | - H Wu
- Department of Pharmacology and Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, TX, USA
| | - Y A Chung
- Department of Pharmacology and Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, TX, USA
| | - A Frazer
- Department of Pharmacology and Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, TX, USA; South Texas Veterans Health Care System (STVHCS), Audie L. Murphy Division, San Antonio, TX, USA
| |
Collapse
|
50
|
Thomaz A, Jaeger M, Buendia M, Bambini-Junior V, Gregianin LJ, Brunetto AL, Brunetto AT, de Farias CB, Roesler R. BDNF/TrkB Signaling as a Potential Novel Target in Pediatric Brain Tumors: Anticancer Activity of Selective TrkB Inhibition in Medulloblastoma Cells. J Mol Neurosci 2015; 59:326-33. [PMID: 26614346 DOI: 10.1007/s12031-015-0689-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 11/18/2015] [Indexed: 12/15/2022]
Abstract
Medulloblastoma (MB) is the most common malignant pediatric brain tumor. Deregulation of brain-derived neurotrophic factor (BDNF)/tropomyosin-related kinase B (TrkB) signaling has been associated with increased proliferative capabilities, invasiveness, and chemoresistance in several types of cancer. However, the relevance of this pathway in MB remains unknown. Here, we show that the selective TrkB inhibitor N-[2-[[(hexahydro-2-oxo-1H-azepin-3-yl)amino]carbonyl]phenyl]-benzo[b]thiophene-2-carboxamide (ANA-12) markedly reduced the viability and survival of human cell lines representative of different MB molecular subgroups. These findings provide the first evidence supporting further investigation of TrkB inhibition as a potential novel strategy for MB treatment.
Collapse
Affiliation(s)
- Amanda Thomaz
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, 90035-003, Porto Alegre, RS, Brazil.,Department of Pharmacology, Institute for Basic Health Sciences, Federal University of Rio Grande do Sul, Rua Sarmento Leite, 500 (ICBS, Campus Centro/UFRGS), 90050-170, Porto Alegre, RS, Brazil
| | - Mariane Jaeger
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, 90035-003, Porto Alegre, RS, Brazil.,Department of Pharmacology, Institute for Basic Health Sciences, Federal University of Rio Grande do Sul, Rua Sarmento Leite, 500 (ICBS, Campus Centro/UFRGS), 90050-170, Porto Alegre, RS, Brazil
| | - Marienela Buendia
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, 90035-003, Porto Alegre, RS, Brazil.,Department of Pharmacology, Institute for Basic Health Sciences, Federal University of Rio Grande do Sul, Rua Sarmento Leite, 500 (ICBS, Campus Centro/UFRGS), 90050-170, Porto Alegre, RS, Brazil
| | - Victorio Bambini-Junior
- Laboratory of Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, 21040-360, Rio de Janeiro, RJ, Brazil
| | - Lauro José Gregianin
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, 90035-003, Porto Alegre, RS, Brazil.,Department of Pediatrics, Faculty of Medicine, Federal University of Rio Grande do Sul, 90035-003, Porto Alegre, RS, Brazil
| | - Algemir Lunardi Brunetto
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, 90035-003, Porto Alegre, RS, Brazil.,Children's Cancer Institute (ICI), 90420-140, Porto Alegre, RS, Brazil
| | - André T Brunetto
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, 90035-003, Porto Alegre, RS, Brazil.,Children's Cancer Institute (ICI), 90420-140, Porto Alegre, RS, Brazil
| | - Caroline Brunetto de Farias
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, 90035-003, Porto Alegre, RS, Brazil.,Children's Cancer Institute (ICI), 90420-140, Porto Alegre, RS, Brazil
| | - Rafael Roesler
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, 90035-003, Porto Alegre, RS, Brazil. .,Department of Pharmacology, Institute for Basic Health Sciences, Federal University of Rio Grande do Sul, Rua Sarmento Leite, 500 (ICBS, Campus Centro/UFRGS), 90050-170, Porto Alegre, RS, Brazil. .,Children's Cancer Institute (ICI), 90420-140, Porto Alegre, RS, Brazil.
| |
Collapse
|