1
|
Yu Q, Li Y, Yan W, Han W, Liu Q, Zhang J, Li X, Shi Y, Wang Y, Yin J, Yan S. Left superior cervical ganglia lymph node mimicry and its role in rat ventricular arrhythmias following myocardial infarction. Acta Physiol (Oxf) 2025; 241:e14279. [PMID: 39803764 DOI: 10.1111/apha.14279] [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: 04/10/2024] [Revised: 11/25/2024] [Accepted: 01/01/2025] [Indexed: 05/02/2025]
Abstract
AIM Sympathetic overactivation may lead to severe ventricular arrhythmias (VAs) post-myocardial infarction (MI). The superior cervical ganglion (SCG) is an extracardiac sympathetic ganglion which regulates cardiac autonomic tone. We aimed to investigate the characteristics and functional significance of SCG on neuro-cardiac communication post-MI. METHODS Constructed MI rat model by left anterior descending coronary artery ligation, and electrophysiological, SCG sympathetic nerve activity testing, echocardiography and histology study were performed. The proteins and gene expression were detected using RNA-seq, spatial transcriptomics, quantitative PCR, and western blotting. RESULTS The SCG neuronal remodeling was recognized by significant increase in adrenergic tyrosine hydroxylase (TH) (+) neurons and decrease in neuronal size. Top differentially expressed genes enriched in pro-inflammatory profile and nerve regulatory factor in left SCG (LSCG) post-MI. Interleukin (IL)-1β and IL-6 increased significantly at Day 3, ahead of nerve growth factor (NGF) which peaked at Day 7 post-MI. Spatial transcriptomics further identified the relativity of TH enrichment with macrophages and cytokines. Therapeutic LSCG-ectomy successfully triggered cardiac denervation and improved VA vulnerability. Eventually, cardiac denervation attenuated macrophage/mast cell infiltration at para-infarct regions, thus improved cardiac dysfunction. Mechanism study revealed that genetic knockdown of NGF receptor trkA in LSCG reversed sympathetic remodeling and cardiac inflammation, which may be partially mediated by substance P and calcitonin gene-related peptide (CGRP). CONCLUSION Extracardiac sympathetic LSCG remodeling participated in arrhythmogenesis and cardiac inflammation/function post-MI. NGF bridged neuro-immune crosstalk between pro-inflammatory shifting and sympathetic overdrive. Targeting LSCG modification facilitated cardiac protection and prevented VAs post-MI.
Collapse
Affiliation(s)
- Qingxia Yu
- Department of Cardiology, Cheeloo College of Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
- Department of Critical Care Medicine, Yantai Yuhuangding Hospital Affiliated with Medical Colledge of Qingdao University, Yantai, China
| | - Yan Li
- Shandong Province University Clinical Immunology Translational Medicine Laboratory, Medical Research Center, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated with Shandong First Medical University, Jinan, China
| | - Wenju Yan
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated with Shandong First Medical University, Jinan, China
| | - Weizhong Han
- Department of Cardiology, Shandong Provincial Hospital Affiliated with Shandong First Medical University, Jinan, China
| | - Qian Liu
- Department of Cardiology, Cheeloo College of Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Junyi Zhang
- Department of Cardiology, Cheeloo College of Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Xiaolu Li
- Department of Emergency Medicine, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated with Shandong First Medical University, Jinan, China
- Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Emergency Medicine, Jinan, China
| | - Yugen Shi
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated with Shandong First Medical University, Jinan, China
| | - Yu Wang
- Department of Emergency Medicine, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated with Shandong First Medical University, Jinan, China
| | - Jie Yin
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated with Shandong First Medical University, Jinan, China
- Department of Cardiology, Shandong Provincial Hospital Affiliated with Shandong First Medical University, Jinan, China
| | - Suhua Yan
- Department of Cardiology, Cheeloo College of Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated with Shandong First Medical University, Jinan, China
| |
Collapse
|
2
|
Testa L, Dotta S, Vercelli A, Marvaldi L. Communicating pain: emerging axonal signaling in peripheral neuropathic pain. Front Neuroanat 2024; 18:1398400. [PMID: 39045347 PMCID: PMC11265228 DOI: 10.3389/fnana.2024.1398400] [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: 03/09/2024] [Accepted: 05/21/2024] [Indexed: 07/25/2024] Open
Abstract
Peripheral nerve damage often leads to the onset of neuropathic pain (NeuP). This condition afflicts millions of people, significantly burdening healthcare systems and putting strain on families' financial well-being. Here, we will focus on the role of peripheral sensory neurons, specifically the Dorsal Root Ganglia neurons (DRG neurons) in the development of NeuP. After axotomy, DRG neurons activate regenerative signals of axons-soma communication to promote a gene program that activates an axonal branching and elongation processes. The results of a neuronal morphological cytoskeleton change are not always associated with functional recovery. Moreover, any axonal miss-targeting may contribute to NeuP development. In this review, we will explore the epidemiology of NeuP and its molecular causes at the level of the peripheral nervous system and the target organs, with major focus on the neuronal cross-talk between intrinsic and extrinsic factors. Specifically, we will describe how failures in the neuronal regenerative program can exacerbate NeuP.
Collapse
Affiliation(s)
- Livia Testa
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano (Torino), Torino, Italy
- Department of Neuroscience “Rita Levi-Montalcini”, Torino, Italy
| | - Sofia Dotta
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano (Torino), Torino, Italy
- Department of Neuroscience “Rita Levi-Montalcini”, Torino, Italy
| | - Alessandro Vercelli
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano (Torino), Torino, Italy
- Department of Neuroscience “Rita Levi-Montalcini”, Torino, Italy
| | - Letizia Marvaldi
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano (Torino), Torino, Italy
- Department of Neuroscience “Rita Levi-Montalcini”, Torino, Italy
| |
Collapse
|
3
|
Iorga RE, Moraru AD, Costin D, Munteanu-Dănulescu RS, Brănișteanu DC. Current trends in targeting the oxidative stress in glaucoma (Review). Eur J Ophthalmol 2024; 34:328-337. [PMID: 37974458 DOI: 10.1177/11206721231214297] [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] [Indexed: 11/19/2023]
Abstract
Glaucoma is a progressive optic neuropathy characterised by retinal ganglion cell degeneration and visual field loss. Glaucoma is considered to be the leading cause of blindness in the industrialised countries. Oxidative damage is an important pathogenic factor in glaucoma, which triggers trabecular meshwork (TM) degeneration, which then leads to intraocular hypertension. Neurodegenerative insults during glaucomatous neurodegeneration initiate an immune response to restore tissue homeostasis. However, the oxidative stress (OS) that develops during the pathogenic processes of glaucoma, along with the agerelated OS, plays a critical role in shifting the physiological equilibrium. In the TM from glaucoma donors, proinflammatory markers were found, which were induced by the activation of a stress response. Chronic changes in the composition of antioxidants found in aqueous humour may induce alterations in TM as well as in the optic nerve head cells. Highlighting the pathogenic role of reactive oxygen species (ROS) in glaucoma has implications in preventing this disease. Various clinical trials are available to test the efficacy of antioxidant drugs in glaucoma management. In this review, we discuss the OS as a therapeutic target, suggesting that the modulation of a pro-oxidant/antioxidant status might be a relevant target for glaucoma prevention and therapy.
Collapse
Affiliation(s)
- Raluca Eugenia Iorga
- Department of Ophthalmology, "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania
| | - Andreea Dana Moraru
- Department of Ophthalmology, "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania
| | - Dănuț Costin
- Department of Ophthalmology, "N. Oblu" Clinical Hospital, Iasi, Romania
| | | | | |
Collapse
|
4
|
Shalaby WS, Ahmed OM, Waisbourd M, Katz LJ. A Review of Potential Novel Glaucoma Therapeutic Options Independent of Intraocular Pressure. Surv Ophthalmol 2021; 67:1062-1080. [PMID: 34890600 DOI: 10.1016/j.survophthal.2021.12.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 12/01/2021] [Accepted: 12/06/2021] [Indexed: 02/06/2023]
Abstract
Glaucoma, a progressive optic neuropathy characterized by retinal ganglion cell degeneration and visual field loss, is the leading cause of irreversible blindness worldwide. Intraocular pressure (IOP) is presently the only modifiable risk factor demonstrated to slow or halt disease progression; however, glaucomatous damage persists in almost 50% of patients despite significant IOP reduction. Many studies have investigated the non-IOP-related risk factors that contribute to glaucoma progression as well as interventions that can prevent or delay glaucomatous neurodegeneration and preserve vision throughout life, independently of IOP. A vast number of experimental studies have reported effective neuroprotection in glaucoma, and clinical studies are ongoing attempting to provide strong evidence of effectiveness of these interventions. In this review, we look into the current understanding of the pathophysiology of glaucoma and explore the recent advances in non-IOP related strategies for neuroprotection and neuroregeneration in glaucoma.
Collapse
Key Words
- AMD, Age-related macular degeneration
- BDNF, Brain derived neurotrophic factor
- CNTF, Ciliary neurotrophic factor
- GDNF, Glial‐derived neurotrophic factor
- Glaucoma
- IOP, Intraocular pressure
- LoGTS, Low-Pressure Glaucoma Treatment Study
- MRI, Magnetic resonance imaging
- MSCs, Mesenchymal stem cells
- NGF, Nerve growth factor
- NTG, Normal tension glaucoma
- OCTA, Optical coherence tomography angiography
- PBM, hotobiomodulation
- PDGF, Platelet derived growth factor
- POAG, Primary open angle glaucoma
- RGCs, Retinal ganglion cells
- TNF-α, Tumor necrosis factor- α
- bFGF, Basic fibroblast growth factor
- gene therapy
- intracranial pressure
- intraocular pressure
- neuroprotection
- ocular blood flow
- oxidative stress
- retinal ganglion cells
- stem cell therapy
Collapse
Affiliation(s)
- Wesam Shamseldin Shalaby
- Glaucoma Research Center, Wills Eye Hospital, Philadelphia, PA, USA; Department of Ophthalmology, Tanta Medical School, Tanta University, Tanta, Gharbia, Egypt
| | - Osama M Ahmed
- Glaucoma Research Center, Wills Eye Hospital, Philadelphia, PA, USA; Yale University School of Medicine, New Haven, CT, USA
| | - Michael Waisbourd
- Glaucoma Research Center, Wills Eye Hospital, Philadelphia, PA, USA; Department of Ophthalmology, Tel Aviv Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Israel
| | - L Jay Katz
- Glaucoma Research Center, Wills Eye Hospital, Philadelphia, PA, USA.
| |
Collapse
|
5
|
Kaur S, Verma H, Dhiman M, Tell G, Gigli GL, Janes F, Mantha AK. Brain Exosomes: Friend or Foe in Alzheimer's Disease? Mol Neurobiol 2021; 58:6610-6624. [PMID: 34595669 DOI: 10.1007/s12035-021-02547-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 08/23/2021] [Indexed: 01/18/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease. It is known to be a multifactorial disease and several causes are associated with its occurrence as well as progression. However, the accumulation of amyloid beta (Aβ) is widely considered its major pathogenic hallmark. Additionally, neurofibrillary tangles (NFT), mitochondrial dysfunction, oxidative stress, and aging (cellular senescence) are considered as additional hits affecting the disease pathology. Several studies are now suggesting important role of inflammation in AD, which shifts our thought towards the brain's resident immune cells, microglia, and astrocytes; how they interact with neurons; and how these interactions are affected by intra and extracellular stressful factors. These interactions can be modulated by different mechanisms and pathways, in which exosomes could play an important role. Exosomes are multivesicular bodies secreted by nearly all types of cells. The exosomes secreted by glial cells or neurons affect the interactions and thus the physiology of these cells by transmitting miRNAs, proteins, and lipids. Exosomes can serve as a friend or foe to the neuron function, depending upon the carried signals. Exosomes, from the healthy microenvironment, may assist neuron function and health, whereas, from the stressed microenvironment, they carry oxidative and inflammatory signals to the neurons and thus prove detrimental to the neuronal function. Furthermore, exosomes can cross the blood-brain barrier (BBB), and from the blood plasma they can enter the brain cells and activate microglia and astrocytes. Exosomes can transport Aβ or Tau, cytokines, miRNAs between the cells, and alter the physiology of recipient cells. They can also assist in Aβ clearance and regulation of synaptic activity. The exosomes derived from different cells play different roles, and this field is still in its infancy stage. This review advocates exosomes' role as a friend or foe in neurodegenerative diseases, especially in the case of Alzheimer's disease.
Collapse
Affiliation(s)
- Sharanjot Kaur
- Department of Microbiology, School of Biological Sciences , Central University of Punjab, Bathinda, Punjab, India
| | - Harkomal Verma
- Department of Zoology, School of Biological Sciences, Central University of Punjab, Village Ghudda151 401, Punjab, Bathinda, India
| | - Monisha Dhiman
- Department of Microbiology, School of Biological Sciences , Central University of Punjab, Bathinda, Punjab, India
| | - Gianluca Tell
- Department of Medicine, University of Udine, Udine, Italy
| | - Gian Luigi Gigli
- Department of Medicine, University of Udine, Udine, Italy
- Clinical Neurology, Udine University Hospital, Udine, Italy
| | | | - Anil K Mantha
- Department of Zoology, School of Biological Sciences, Central University of Punjab, Village Ghudda151 401, Punjab, Bathinda, India.
| |
Collapse
|
6
|
Markworth R, Dambeck V, Steinbeck LM, Koufali A, Bues B, Dankovich TM, Wichmann C, Burk K. Tubular microdomains of Rab7-positive endosomes retrieve TrkA, a mechanism disrupted in Charcot-Marie-Tooth disease 2B. J Cell Sci 2021; 134:272650. [PMID: 34486665 DOI: 10.1242/jcs.258559] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 08/23/2021] [Indexed: 01/04/2023] Open
Abstract
Axonal survival and growth requires signalling from tropomyosin receptor kinases (Trks). To transmit their signals, receptor-ligand complexes are endocytosed and undergo retrograde trafficking to the soma, where downstream signalling occurs. Vesicles transporting neurotrophic receptors to the soma are reported to be Rab7-positive late endosomes and/or multivesicular bodies (MVBs), where receptors localize within so-called intraluminal vesicles (herein Rab7 corresponds to Rab7A unless specified otherwise). Therefore, one challenging question is how downstream signalling is possible given the insulating properties of intraluminal vesicles. In this study, we report that Rab7-positive endosomes and MVBs retrieve TrkA (also known as NTRK1) through tubular microdomains. Interestingly, this phenotype is absent for the EGF receptor. Furthermore, we found that endophilinA1, endophilinA2 and endophilinA3, together with WASH1 (also known as WASHC1), are involved in the tubulation process. In Charcot-Marie-Tooth disease 2B (CMT2B), a neuropathy of the peripheral nervous system, this tubulating mechanism is disrupted. In addition, the ability to tubulate correlates with the phosphorylation levels of TrkA as well as with neurite length in neuronal cultures from dorsal root ganglia. In all, we report a new retrieval mechanism of late Rab7-positive endosomes, which enables TrkA signalling and sheds new light onto how neurotrophic signalling is disrupted in CMT2B. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Ronja Markworth
- Department of Neurology, University Medical Center Göttingen, Robert Koch Straße 40, 37075 Göttingen, Germany.,European Neuroscience Institute, Grisebachstraße 5, 37077 Göttingen, Germany.,Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Von-Siebold Straße 3A, 37075 Göttingen, Germany
| | - Vivian Dambeck
- Department of Neurology, University Medical Center Göttingen, Robert Koch Straße 40, 37075 Göttingen, Germany.,Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Von-Siebold Straße 3A, 37075 Göttingen, Germany
| | - Lars Malte Steinbeck
- Department of Neurology, University Medical Center Göttingen, Robert Koch Straße 40, 37075 Göttingen, Germany.,Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Von-Siebold Straße 3A, 37075 Göttingen, Germany
| | - Angeliki Koufali
- Department of Neurology, University Medical Center Göttingen, Robert Koch Straße 40, 37075 Göttingen, Germany.,Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Von-Siebold Straße 3A, 37075 Göttingen, Germany
| | - Bastian Bues
- Department of Neurology, University Medical Center Göttingen, Robert Koch Straße 40, 37075 Göttingen, Germany.,Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Von-Siebold Straße 3A, 37075 Göttingen, Germany
| | - Tal M Dankovich
- Institute for Neuro- and Sensory Physiology, Humboldtallee 23, 37073 Göttingen, Germany
| | - Carolin Wichmann
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Von-Siebold Straße 3A, 37075 Göttingen, Germany.,Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,Collaborative Research Centers 889 'Cellular Mechanisms of Sensory Processing' and 1286 'Quantitative Synaptology', 37099 Göttingen, Germany
| | - Katja Burk
- Department of Neurology, University Medical Center Göttingen, Robert Koch Straße 40, 37075 Göttingen, Germany.,European Neuroscience Institute, Grisebachstraße 5, 37077 Göttingen, Germany.,Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Von-Siebold Straße 3A, 37075 Göttingen, Germany
| |
Collapse
|
7
|
The cellular and molecular basis of somatosensory neuron development. Neuron 2021; 109:3736-3757. [PMID: 34592169 DOI: 10.1016/j.neuron.2021.09.004] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/23/2021] [Accepted: 09/01/2021] [Indexed: 11/23/2022]
Abstract
Primary somatosensory neurons convey salient information about our external environment and internal state to the CNS, allowing us to detect, perceive, and react to a wide range of innocuous and noxious stimuli. Pseudo-unipolar in shape, and among the largest (longest) cells of most mammals, dorsal root ganglia (DRG) somatosensory neurons have peripheral axons that extend into skin, muscle, viscera, or bone and central axons that innervate the spinal cord and brainstem, where they synaptically engage the central somatosensory circuitry. Here, we review the diversity of mammalian DRG neuron subtypes and the intrinsic and extrinsic mechanisms that control their development. We describe classical and contemporary advances that frame our understanding of DRG neurogenesis, transcriptional specification of DRG neurons, and the establishment of morphological, physiological, and synaptic diversification across somatosensory neuron subtypes.
Collapse
|
8
|
Kaplan DR, Mobley WC. (H)Elping nerve growth factor: Elp1 inhibits TrkA's phosphatase to maintain retrograde signaling. J Clin Invest 2021; 130:2195-2198. [PMID: 32281945 DOI: 10.1172/jci136162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Nerve growth factor (NGF) regulates many aspects of neuronal biology by retrogradely propagating signals along axons to the targets of those axons. How this occurs when axons contain a plethora of proteins that can silence those signals has long perplexed the neurotrophin field. In this issue of the JCI, Li et al. suggest an answer to this vexing problem, while exploring why the Elp1 gene that is mutated in familial dysautonomia (FD) causes peripheral neuropathy. They describe a distinctive function of Elp1 as a protein that is required to sustain NGF signaling by blocking the activity of its phosphatase that shuts off those signals. This finding helps explain the innervation deficits prominent in FD and reveals a unique role for Elp1 in the regulation of NGF-dependent TrkA activity.
Collapse
Affiliation(s)
- David R Kaplan
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
9
|
Neurotrophic Factors in Glaucoma and Innovative Delivery Systems. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10249015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Glaucoma is a neurodegenerative disease and a worldwide leading cause of irreversible vision loss. In the last decades, high efforts have been made to develop novel treatments effective in inducing protection and/or recovery of neural function in glaucoma, including neurotrophic factors (NTFs). These approaches have shown encouraging data in preclinical setting; however, the challenge of sustained, targeted delivery to the retina and optic nerve still prevents the clinical translation. In this paper, the authors review and discuss the most recent advances for the use of NTFs treatment in glaucoma, including intraocular delivery. Novel strategies in drug and gene delivery technology for NTFs are proving effective in promoting long-term retinal ganglion cells (RGCs) survival and related functional improvements. Results of experimental and clinical studies evaluating the efficacy and safety of biodegradable slow-release NTF-loaded microparticle devices, encapsulated NTF-secreting cells implants, mimetic ligands for NTF receptors, and viral and non-viral NTF gene vehicles are discussed. NTFs are able to prevent and even reverse apoptotic ganglion cell death. Nevertheless, neuroprotection in glaucoma remains an open issue due to the unmet need of sustained delivery to the posterior segment of the eye. The recent advances in intraocular delivery systems pave the way for possible future use of NTFs in clinical practice for the treatment of glaucoma.
Collapse
|
10
|
Pijuan J, Rodríguez-Sanz M, Natera-de Benito D, Ortez C, Altimir A, Osuna-López M, Roura M, Ugalde M, Van de Vondel L, Reina-Castillón J, Fons C, Benítez R, Nascimento A, Hoenicka J, Palau F. Translational Diagnostics: An In-House Pipeline to Validate Genetic Variants in Children with Undiagnosed and Rare Diseases. J Mol Diagn 2020; 23:71-90. [PMID: 33223419 DOI: 10.1016/j.jmoldx.2020.10.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 09/10/2020] [Accepted: 10/13/2020] [Indexed: 12/26/2022] Open
Abstract
Diagnosis is essential for the management and treatment of patients with rare diseases. In a group of patients, the genetic study identifies variants of uncertain significance or inconsistent with the phenotype; therefore, it is urgent to develop novel strategies to reach the definitive diagnosis. Herein, we develop the in-house Translational Diagnostics Program (TDP) to validate genetic variants as part of the diagnostic process with the close collaboration of physicians, clinical scientists, and research scientists. The first 7 of 33 consecutive patients for whom exome-based tests were not diagnostic were investigated. The TDP pipeline includes four steps: (i) phenotype assessment, (ii) literature review and prediction of in silico pathogenicity, (iii) experimental functional studies, and (iv) diagnostic decision-making. Re-evaluation of the phenotype and re-analysis of the exome allowed the diagnosis in one patient. In the remaining patients, the studies included either cDNA cloning or PCR-amplified genomic DNA, or the use of patients' fibroblasts. A comparative computational analysis of confocal microscopy images and studies related to the protein function was performed. In five of these six patients, evidence of pathogenicity of the genetic variant was found, which was validated by physicians. The current research demonstrates the feasibility of the TDP to support and resolve intramural medical problems when the clinical significance of the patient variant is unknown or inconsistent with the phenotype.
Collapse
Affiliation(s)
- Jordi Pijuan
- Laboratory of Neurogenetics and Molecular Medicine-Pediatric Institute of Rare Diseases, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - María Rodríguez-Sanz
- Laboratory of Neurogenetics and Molecular Medicine-Pediatric Institute of Rare Diseases, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Daniel Natera-de Benito
- Neuromuscular Unit, Department of Pediatric Neurology, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Carlos Ortez
- Neuromuscular Unit, Department of Pediatric Neurology, Hospital Sant Joan de Déu, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras, Barcelona, Spain
| | - Arola Altimir
- Laboratory of Neurogenetics and Molecular Medicine-Pediatric Institute of Rare Diseases, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Mireia Osuna-López
- Laboratory of Neurogenetics and Molecular Medicine-Pediatric Institute of Rare Diseases, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Montserrat Roura
- Laboratory of Neurogenetics and Molecular Medicine-Pediatric Institute of Rare Diseases, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Maddi Ugalde
- Laboratory of Neurogenetics and Molecular Medicine-Pediatric Institute of Rare Diseases, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Liedewei Van de Vondel
- Laboratory of Neurogenetics and Molecular Medicine-Pediatric Institute of Rare Diseases, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Judith Reina-Castillón
- Laboratory of Neurogenetics and Molecular Medicine-Pediatric Institute of Rare Diseases, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Carme Fons
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Barcelona, Spain; Department of Pediatric Neurology, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Raúl Benítez
- Automatic Control Department and Biomedical Engineering Research Center, Universitat Politècnica de Catalunya, Barcelona, Spain
| | - Andrés Nascimento
- Neuromuscular Unit, Department of Pediatric Neurology, Hospital Sant Joan de Déu, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras, Barcelona, Spain
| | - Janet Hoenicka
- Laboratory of Neurogenetics and Molecular Medicine-Pediatric Institute of Rare Diseases, Institut de Recerca Sant Joan de Déu, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras, Barcelona, Spain.
| | - Francesc Palau
- Laboratory of Neurogenetics and Molecular Medicine-Pediatric Institute of Rare Diseases, Institut de Recerca Sant Joan de Déu, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras, Barcelona, Spain; Department of Genetic Medicine-IPER, Hospital Sant Joan de Déu, Barcelona, Spain; Clinic Institute of Medicine and Dermatology, Hospital Clínic, Barcelona, Spain; Division of Pediatrics, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
11
|
Chi H, Sun L, Shiu RH, Han R, Hsieh CP, Wei TM, Lo CC, Chang HY, Sang TK. Cleavage of human tau at Asp421 inhibits hyperphosphorylated tau induced pathology in a Drosophila model. Sci Rep 2020; 10:13482. [PMID: 32778728 PMCID: PMC7417559 DOI: 10.1038/s41598-020-70423-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 07/29/2020] [Indexed: 11/09/2022] Open
Abstract
Hyperphosphorylated and truncated tau variants are enriched in neuropathological aggregates in diseases known as tauopathies. However, whether the interaction of these posttranslational modifications affects tau toxicity as a whole remains unresolved. By expressing human tau with disease-related Ser/Thr residues to simulate hyperphosphorylation, we show that despite severe neurodegeneration in full-length tau, with the truncation at Asp421, the toxicity is ameliorated. Cytological and biochemical analyses reveal that hyperphosphorylated full-length tau distributes in the soma, the axon, and the axonal terminal without evident distinction, whereas the Asp421-truncated version is mostly restricted from the axonal terminal. This discrepancy is correlated with the fact that fly expressing hyperphosphorylated full-length tau, but not Asp421-cleaved one, develops axonopathy lesions, including axonal spheroids and aberrant actin accumulations. The reduced presence of hyperphosphorylated tau in the axonal terminal is corroborated with the observation that flies expressing Asp421-truncated variants showed less motor deficit, suggesting synaptic function is preserved. The Asp421 cleavage of tau is a proteolytic product commonly found in the neurofibrillary tangles. Our finding suggests the coordination of different posttranslational modifications on tau may have an unexpected impact on the protein subcellular localization and cytotoxicity, which may be valuable when considering tau for therapeutic purposes.
Collapse
Affiliation(s)
- Hao Chi
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Lee Sun
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Ren-Huei Shiu
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Rui Han
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Chien-Ping Hsieh
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Tzu-Min Wei
- Institute of Systems Neuroscience, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Chung-Chuan Lo
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 30013, Taiwan.,Institute of Systems Neuroscience, National Tsing Hua University, Hsinchu, 30013, Taiwan.,Brain Research Center, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Hui-Yun Chang
- Institute of Systems Neuroscience, National Tsing Hua University, Hsinchu, 30013, Taiwan.,Department of Medical Science, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Tzu-Kang Sang
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, 30013, Taiwan. .,Brain Research Center, National Tsing Hua University, Hsinchu, 30013, Taiwan. .,Department of Life Science, National Tsing Hua University, Hsinchu, 30013, Taiwan.
| |
Collapse
|
12
|
|
13
|
Neurotrophic effects of G M1 ganglioside, NGF, and FGF2 on canine dorsal root ganglia neurons in vitro. Sci Rep 2020; 10:5380. [PMID: 32214122 PMCID: PMC7096396 DOI: 10.1038/s41598-020-61852-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 03/04/2020] [Indexed: 01/26/2023] Open
Abstract
Dogs share many chronic morbidities with humans and thus represent a powerful model for translational research. In comparison to rodents, the canine ganglioside metabolism more closely resembles the human one. Gangliosides are components of the cell plasma membrane playing a role in neuronal development, intercellular communication and cellular differentiation. The present in vitro study aimed to characterize structural and functional changes induced by GM1 ganglioside (GM1) in canine dorsal root ganglia (DRG) neurons and interactions of GM1 with nerve growth factor (NGF) and fibroblast growth factor (FGF2) using immunofluorescence for several cellular proteins including neurofilaments, synaptophysin, and cleaved caspase 3, transmission electron microscopy, and electrophysiology. GM1 supplementation resulted in increased neurite outgrowth and neuronal survival. This was also observed in DRG neurons challenged with hypoxia mimicking neurodegenerative conditions due to disruptions of energy homeostasis. Immunofluorescence indicated an impact of GM1 on neurofilament phosphorylation, axonal transport, and synaptogenesis. An increased number of multivesicular bodies in GM1 treated neurons suggested metabolic changes. Electrophysiological changes induced by GM1 indicated an increased neuronal excitability. Summarized, GM1 has neurotrophic and neuroprotective effects on canine DRG neurons and induces functional changes. However, further studies are needed to clarify the therapeutic value of gangliosides in neurodegenerative diseases.
Collapse
|
14
|
Dutta R, Sarkar SR. Role of Dynein and Dynactin (DCTN-1) in Neurodegenerative Diseases. ACTA ACUST UNITED AC 2019. [DOI: 10.33805/2641-8991.123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The pathophysiology and concept of degeneration in central nervous system is very complex and overwhelming at times. There is a complex mechanism which exists among different molecules in the cytoplasm of cell bodies of neurons, antegrade and retrograde axonal transport of cargoes and accumulation of certain substances and proteins which can influence the excitatory neurotransmitter like glutamate initiating the process of neurodegeneration. Neurons have extensive processes and communication between those processes and the cell body is crucial to neuronal function, viability and survival over time with progression of age. Researchers believe neurons are uniquely dependent on microtubule-based cargo transport. There is enough evidence to support that deficits in retrograde axonal transport contribute to pathogenesis in multiple neurodegenerative diseases. Cytoplasmic dynein and its regulation by Dynactin (DCTN1) is the major molecular motor cargo involved in autophagy, mitosis and neuronal cell survival. Mutation in dynactin gene located in 2p13.1,is indeed studied very extensively and is considered to be involved directly or indirectly to various conditions like Perry syndrome, familial and sporadic Amyotrophic lateral sclerosis, Hereditary spastic paraplegia, Spinocerebellar Ataxia (SCA-5), Huntingtons disease, Alzheimers disease, Charcot marie tooth disease, Hereditary motor neuropathy 7B, prion disease, parkinsons disease, malformation of cortical development, polymicrogyria to name a few with exception of Multiple Sclerosis (MS).
Collapse
|
15
|
Claes M, De Groef L, Moons L. Target-Derived Neurotrophic Factor Deprivation Puts Retinal Ganglion Cells on Death Row: Cold Hard Evidence and Caveats. Int J Mol Sci 2019; 20:E4314. [PMID: 31484425 PMCID: PMC6747494 DOI: 10.3390/ijms20174314] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 08/28/2019] [Accepted: 08/30/2019] [Indexed: 12/14/2022] Open
Abstract
Glaucoma and other optic neuropathies are characterized by axonal transport deficits. Axonal cargo travels back and forth between the soma and the axon terminus, a mechanism ensuring homeostasis and the viability of a neuron. An example of vital molecules in the axonal cargo are neurotrophic factors (NTFs). Hindered retrograde transport can cause a scarcity of those factors in the retina, which in turn can tilt the fate of retinal ganglion cells (RGCs) towards apoptosis. This postulation is one of the most widely recognized theories to explain RGC death in the disease progression of glaucoma and is known as the NTF deprivation theory. For several decades, research has been focused on the use of NTFs as a novel neuroprotective glaucoma treatment. Until now, results in animal models have been promising, but translation to the clinic has been highly disappointing. Are we lacking important knowledge to lever NTF therapies towards the therapeutic armamentarium? Or did we get the wrong end of the stick regarding the NTF deprivation theory? In this review, we will tackle the existing evidence and caveats advocating for and against the target-derived NTF deprivation theory in glaucoma, whilst digging into associated therapy efforts.
Collapse
Affiliation(s)
- Marie Claes
- Laboratory of Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, 3000 Leuven, Belgium
| | - Lies De Groef
- Laboratory of Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, 3000 Leuven, Belgium
| | - Lieve Moons
- Laboratory of Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
16
|
Tellone E, Galtieri A, Ficarra S. Reviewing Biochemical Implications of Normal and Mutated Huntingtin in Huntington's Disease. Curr Med Chem 2019; 27:5137-5158. [PMID: 31223078 DOI: 10.2174/0929867326666190621101909] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/08/2019] [Accepted: 05/22/2019] [Indexed: 12/17/2022]
Abstract
Huntingtin (Htt) is a multi-function protein of the brain. Normal Htt shows a common alpha-helical structure but conformational changes in the form with beta strands are the principal cause of Huntington's disease. Huntington's disease is a genetic neurological disorder caused by a repeated expansion of the CAG trinucleotide, causing instability in the N-terminal of the gene coding for the Huntingtin protein. The mutation leads to the abnormal expansion of the production of the polyglutamine tract (polyQ) resulting in the form of an unstable Huntingtin protein commonly referred to as mutant Huntingtin. Mutant Huntingtin is the cause of the complex neurological metabolic alteration of Huntington's disease, resulting in both the loss of all the functions of normal Huntingtin and the genesis of abnormal interactions due to the presence of this mutation. One of the problems arising from the misfolded Huntingtin is the increase in oxidative stress, which is common in many neurological diseases such as Alzheimer's, Parkinson's, Amyotrophic Lateral Sclerosis and Creutzfeldt-Jakob disease. In the last few years, the use of antioxidants had a strong incentive to find valid therapies for defence against neurodegenerations. Although further studies are needed, the use of antioxidant mixtures to counteract neuronal damages seems promising.
Collapse
Affiliation(s)
- Ester Tellone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, V.le Ferdinando Stagno d'Alcontres 31, 98166 Messina, Italy
| | - Antonio Galtieri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, V.le Ferdinando Stagno d'Alcontres 31, 98166 Messina, Italy
| | - Silvana Ficarra
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, V.le Ferdinando Stagno d'Alcontres 31, 98166 Messina, Italy
| |
Collapse
|
17
|
Abstract
PURPOSE OF REVIEW The current review analyzes recent findings that suggest that axon degeneration is a druggable process in the treatment of neurodegenerative disorders and a subset of traumas. RECENT FINDINGS Emerging evidence reveals that axon degeneration is an active and regulated process in the early progression of some neurodegenerative diseases and acute traumas, which is orchestrated through a combination of axon-intrinsic and somatically derived signaling events. The identification of these pathways has presented appealing drug targets whose specificity for the nervous system and phenotypes in mouse models offers significant clinical opportunity. SUMMARY As the biology of axon degeneration becomes clear, so too has the realization that the pathways driving axon degeneration overlap in part with those that drive neuronal apoptosis and, importantly, axon regeneration. Axon-specific disorders like those seen in CIPN, where injury signaling to the nucleus is not a prominent feature, have been shown to benefit from disruption of Sarm1. In injury and disease contexts, where involvement of somatic events is prominent, inhibition of the MAP Kinase DLK exhibits promise for neuroprotection. Here, however, interfering with somatic signaling may preclude the ability of an axon or a circuit to regenerate or functionally adapt following acute injuries.
Collapse
|
18
|
Pietak A, Bischof J, LaPalme J, Morokuma J, Levin M. Neural control of body-plan axis in regenerating planaria. PLoS Comput Biol 2019; 15:e1006904. [PMID: 30990801 PMCID: PMC6485777 DOI: 10.1371/journal.pcbi.1006904] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 04/26/2019] [Accepted: 02/26/2019] [Indexed: 01/01/2023] Open
Abstract
Control of axial polarity during regeneration is a crucial open question. We developed a quantitative model of regenerating planaria, which elucidates self-assembly mechanisms of morphogen gradients required for robust body-plan control. The computational model has been developed to predict the fraction of heteromorphoses expected in a population of regenerating planaria fragments subjected to different treatments, and for fragments originating from different regions along the anterior-posterior and medio-lateral axis. This allows for a direct comparison between computational and experimental regeneration outcomes. Vector transport of morphogens was identified as a fundamental requirement to account for virtually scale-free self-assembly of the morphogen gradients observed in planarian homeostasis and regeneration. The model correctly describes altered body-plans following many known experimental manipulations, and accurately predicts outcomes of novel cutting scenarios, which we tested. We show that the vector transport field coincides with the alignment of nerve axons distributed throughout the planarian tissue, and demonstrate that the head-tail axis is controlled by the net polarity of neurons in a regenerating fragment. This model provides a comprehensive framework for mechanistically understanding fundamental aspects of body-plan regulation, and sheds new light on the role of the nervous system in directing growth and form.
Collapse
Affiliation(s)
- Alexis Pietak
- Allen Discovery Center, Tufts University, Medford, Massachusetts, United States of America
| | - Johanna Bischof
- Allen Discovery Center, Tufts University, Medford, Massachusetts, United States of America
- Department of Biology, Tufts University, Medford, Massachusetts, United States of America
| | - Joshua LaPalme
- Allen Discovery Center, Tufts University, Medford, Massachusetts, United States of America
- Department of Biology, Tufts University, Medford, Massachusetts, United States of America
| | - Junji Morokuma
- Allen Discovery Center, Tufts University, Medford, Massachusetts, United States of America
- Department of Biology, Tufts University, Medford, Massachusetts, United States of America
| | - Michael Levin
- Allen Discovery Center, Tufts University, Medford, Massachusetts, United States of America
- Department of Biology, Tufts University, Medford, Massachusetts, United States of America
| |
Collapse
|
19
|
Sampayo RG, Toscani AM, Rubashkin MG, Thi K, Masullo LA, Violi IL, Lakins JN, Cáceres A, Hines WC, Coluccio Leskow F, Stefani FD, Chialvo DR, Bissell MJ, Weaver VM, Simian M. Fibronectin rescues estrogen receptor α from lysosomal degradation in breast cancer cells. J Cell Biol 2018; 217:2777-2798. [PMID: 29980625 PMCID: PMC6080927 DOI: 10.1083/jcb.201703037] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 12/20/2017] [Accepted: 05/03/2018] [Indexed: 02/07/2023] Open
Abstract
Estrogen receptor α (ERα) is expressed in tissues as diverse as brains and mammary glands. In breast cancer, ERα is a key regulator of tumor progression. Therefore, understanding what activates ERα is critical for cancer treatment in particular and cell biology in general. Using biochemical approaches and superresolution microscopy, we show that estrogen drives membrane ERα into endosomes in breast cancer cells and that its fate is determined by the presence of fibronectin (FN) in the extracellular matrix; it is trafficked to lysosomes in the absence of FN and avoids the lysosomal compartment in its presence. In this context, FN prolongs ERα half-life and strengthens its transcriptional activity. We show that ERα is associated with β1-integrin at the membrane, and this integrin follows the same endocytosis and subcellular trafficking pathway triggered by estrogen. Moreover, ERα+ vesicles are present within human breast tissues, and colocalization with β1-integrin is detected primarily in tumors. Our work unravels a key, clinically relevant mechanism of microenvironmental regulation of ERα signaling.
Collapse
Affiliation(s)
- Rocío G Sampayo
- Universidad de Buenos Aires, Instituto de Oncología "Ángel H. Roffo", Área Investigación, Buenos Aires, Argentina .,Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Fisiología y Biología Molecular y Celular, Ciudad Universitaria, Buenos Aires, Argentina.,Universidad Nacional de San Martín, Instituto de Nanosistemas, Campus Miguelete, San Martín, Argentina
| | - Andrés M Toscani
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, IQUIBICEN UBA-CONICET y Universidad Nacional de Luján, Departamento de Ciencias Básicas, Buenos Aires, Argentina
| | - Matthew G Rubashkin
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA
| | - Kate Thi
- Division of Biological Systems and Engineering, Lawrence Berkeley National Laboratory, Berkeley, CA
| | - Luciano A Masullo
- Centro de Investigaciones en Bionanociencias, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina.,Departamento de Física, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ianina L Violi
- Centro de Investigaciones en Bionanociencias, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Jonathon N Lakins
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA
| | - Alfredo Cáceres
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, Córdoba, Argentina
| | - William C Hines
- Division of Biological Systems and Engineering, Lawrence Berkeley National Laboratory, Berkeley, CA
| | - Federico Coluccio Leskow
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, IQUIBICEN UBA-CONICET y Universidad Nacional de Luján, Departamento de Ciencias Básicas, Buenos Aires, Argentina
| | - Fernando D Stefani
- Centro de Investigaciones en Bionanociencias, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina.,Departamento de Física, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Dante R Chialvo
- Center for Complex Systems and Brain Sciences, Escuela de Ciencia y Tecnología, Universidad Nacional de San Martín and Consejo Nacional de Investigaciones Científicas y Tecnológicas, San Martín, Argentina
| | - Mina J Bissell
- Division of Biological Systems and Engineering, Lawrence Berkeley National Laboratory, Berkeley, CA
| | - Valerie M Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA
| | - Marina Simian
- Universidad de Buenos Aires, Instituto de Oncología "Ángel H. Roffo", Área Investigación, Buenos Aires, Argentina .,Universidad Nacional de San Martín, Instituto de Nanosistemas, Campus Miguelete, San Martín, Argentina
| |
Collapse
|
20
|
Abstract
Exosomes are secreted extracellular vesicles (EVs) that carry micro RNAs and other factors to reprogram cancer cells and tissues affected by cancer. Exosomes are exchanged between cancer cells and other tissues, often to prepare a premetastatic niche, escape immune surveillance, or spread multidrug resistance. Only a few studies investigated the function of lipids in exosomes although their lipid composition is different from that of the secreting cells. Ceramide is one of the lipids critical for exosome formation, and it is also enriched in these EVs. New research suggests that lipids in the exosomal membrane may organize and transmit "mobile rafts" that turn exosomes into extracellular signalosomes spreading activation of cell signaling pathways in oncogenesis and metastasis. Ceramide may modulate the function of mobile rafts and their effect on these cell signaling pathways. The critical role of lipids and, in particular, ceramide for formation, secretion, and function of exosomes may lead to a radically new understanding of cancer biology and therapy.
Collapse
Affiliation(s)
- Ahmed Elsherbini
- Department of Physiology, University of Kentucky, Lexington, KY, United States
| | - Erhard Bieberich
- Department of Physiology, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
21
|
Bakhtiarzadeh F, Nahavandi A, Goudarzi M, Shirvalilou S, Rakhshan K, Niknazar S. Axonal transport proteins and depressive like behavior, following Chronic Unpredictable Mild Stress in male rat. Physiol Behav 2018; 194:9-14. [PMID: 29698729 DOI: 10.1016/j.physbeh.2018.04.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 04/20/2018] [Accepted: 04/20/2018] [Indexed: 11/27/2022]
Abstract
BACKGROUND A common mood disorder, depression has long been considered a leading cause of disability worldwide. Chronic stress is involved in the development of various psychiatric diseases including major depressive disorder. Stress can induce depressive-like symptoms and initiate neurodegenerative processes in the brain. The neurodegenerative theory of depression holds impaired axonal transport as a negative factor in neural survival. Axonal transport is a critical mechanism for normal neuronal function, playing crucial roles in axon growth, neurotransmitter secretion, normal mitochondrial function and neural survival. METHODS AND MATERIALS To investigate the effects of stress-induced depression, in the present study, we evaluated behavior by forced swimming test (FST), corticosterone plasma level by ELISA assay, hippocampal mRNA expression of three genes (NGF, kinesin and dynein) via real-time PCR and hippocamp count by Nissl staining in male Wistar rats. RESULTS Our data demonstrated a significant decrease in the expression of NGF, kinesin and dynein genes in CUMS groups compared to the control group (non-stressed) (p < 0.05). CUMS also caused an elevation in immobility time and corticosterone plasma level in the stressed group compared to the controls (p < 0.01 and p < 0.05, respectively). CONCLUSION The results suggested that the possibility of stress-induced depressive behavior associated with hippocampal neurodegeneration process is correlated with a low expression of kinesin and dynein, the two most important proteins in axonal transport.
Collapse
Affiliation(s)
| | - Arezo Nahavandi
- Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran; Neuroscience Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Mina Goudarzi
- Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran; Neuroscience Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Sakine Shirvalilou
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Kamran Rakhshan
- Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Somayeh Niknazar
- ENT Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
22
|
Gannon SM, Hawk K, Walsh BF, Coulibaly A, Isaacson LG. Retrograde influences of SCG axotomy on uninjured preganglionic neurons. Brain Res 2018; 1691:44-54. [PMID: 29679543 DOI: 10.1016/j.brainres.2018.04.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 04/06/2018] [Accepted: 04/13/2018] [Indexed: 10/17/2022]
Abstract
There is evidence that neuronal injury can affect uninjured neurons in the same neural circuit. The overall goal of this study was to understand the effects of peripheral nerve injury on uninjured neurons located in the central nervous system (CNS). As a model, we examined whether axotomy (transection of postganglionic axons) of the superior cervical ganglion (SCG) affected the uninjured, preganglionic neurons that innervate the SCG. At 7 days post-injury a reduction in choline acetyltransferase (ChAT) and synaptophysin immunoreactivity in the SCG, both markers for preganglionic axons, was observed, and this reduction persisted at 8 and 12 weeks post-injury. No changes were observed in the number or size of the parent cell bodies in the intermediolateral cell column (IML) of the spinal cord, yet synaptic input to the IML neurons was decreased at both 8 and 12 weeks post-injury. In order to understand the mechanisms underlying these changes, protein levels of brain-derived neurotrophic factor (BDNF) and tyrosine receptor kinase B (TrkB) were examined and reductions were observed at 7 days post-injury in both the SCG and spinal cord. Taken together these results suggest that axotomy of the SCG led to reduced BDNF in the SCG and spinal cord, which in turn influenced ChAT and synaptophysin expression in the SCG and also contributed to the altered synaptic input to the IML neurons. More generally these findings provide evidence that the effects of peripheral injury can cascade into the CNS and affect uninjured neurons.
Collapse
Affiliation(s)
- Sean M Gannon
- Center for Neuroscience and Behavior, Miami University, Oxford, OH 45056, United States; Department of Biology, Miami University, Oxford, OH 45056, United States
| | - Kiel Hawk
- Center for Neuroscience and Behavior, Miami University, Oxford, OH 45056, United States; Graduate Program in Cell, Molecular, and Structural Biology, Miami University, Oxford, OH 45056, United States
| | - Brian F Walsh
- Department of Biology, Miami University, Oxford, OH 45056, United States
| | - Aminata Coulibaly
- Center for Neuroscience and Behavior, Miami University, Oxford, OH 45056, United States; Graduate Program in Cell, Molecular, and Structural Biology, Miami University, Oxford, OH 45056, United States
| | - Lori G Isaacson
- Center for Neuroscience and Behavior, Miami University, Oxford, OH 45056, United States; Graduate Program in Cell, Molecular, and Structural Biology, Miami University, Oxford, OH 45056, United States; Department of Biology, Miami University, Oxford, OH 45056, United States.
| |
Collapse
|
23
|
Liang Y, Sigrist S. Autophagy and proteostasis in the control of synapse aging and disease. Curr Opin Neurobiol 2018; 48:113-121. [DOI: 10.1016/j.conb.2017.12.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 10/25/2017] [Accepted: 12/10/2017] [Indexed: 11/30/2022]
|
24
|
Ye M, Lehigh KM, Ginty DD. Multivesicular bodies mediate long-range retrograde NGF-TrkA signaling. eLife 2018; 7:33012. [PMID: 29381137 PMCID: PMC5811214 DOI: 10.7554/elife.33012] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Accepted: 01/30/2018] [Indexed: 12/25/2022] Open
Abstract
The development of neurons in the peripheral nervous system is dependent on target-derived, long-range retrograde neurotrophic factor signals. The prevailing view is that target-derived nerve growth factor (NGF), the prototypical neurotrophin, and its receptor TrkA are carried retrogradely by early endosomes, which serve as TrkA signaling platforms in cell bodies. Here, we report that the majority of retrograde TrkA signaling endosomes in mouse sympathetic neurons are ultrastructurally and molecularly defined multivesicular bodies (MVBs). In contrast to MVBs that carry non-TrkA cargoes from distal axons to cell bodies, retrogradely transported TrkA+ MVBs that arrive in cell bodies evade lysosomal fusion and instead evolve into TrkA+ single-membrane vesicles that are signaling competent. Moreover, TrkA kinase activity associated with retrogradely transported TrkA+ MVBs determines TrkA+ endosome evolution and fate. Thus, MVBs deliver long-range retrograde NGF signals and serve as signaling and sorting platforms in the cell soma, and MVB cargoes dictate their vesicular fate.
Collapse
Affiliation(s)
- Mengchen Ye
- Human Genetics Training Program, The Johns Hopkins University, School of Medicine, Baltimore, United States.,Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, United States
| | - Kathryn M Lehigh
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, United States.,Department of Neuroscience, The Johns Hopkins University, School of Medicine, Baltimore, United States
| | - David D Ginty
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, United States
| |
Collapse
|
25
|
Caldieri G, Malabarba MG, Di Fiore PP, Sigismund S. EGFR Trafficking in Physiology and Cancer. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2018; 57:235-272. [PMID: 30097778 DOI: 10.1007/978-3-319-96704-2_9] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Signaling from the epidermal growth factor receptor (EGFR) elicits multiple biological responses, including cell proliferation, migration, and survival. Receptor endocytosis and trafficking are critical physiological processes that control the strength, duration, diversification, and spatial restriction of EGFR signaling through multiple mechanisms, which we review in this chapter. These mechanisms include: (i) regulation of receptor density and activation at the cell surface; (ii) concentration of receptors into distinct nascent endocytic structures; (iii) commitment of the receptor to different endocytic routes; (iv) endosomal sorting and postendocytic trafficking of the receptor through distinct pathways, and (v) recycling to restricted regions of the cell surface. We also highlight how communication between organelles controls EGFR activity along the endocytic route. Finally, we illustrate how abnormal trafficking of EGFR oncogenic mutants, as well as alterations of the endocytic machinery, contributes to aberrant EGFR signaling in cancer.
Collapse
Affiliation(s)
- Giusi Caldieri
- Dipartimento di Oncologia ed Emato-oncologia, Università degli Studi di Milano, Via Santa Sofia 9/1, 20122, Milan, Italy
- Istituto Europeo di Oncologia, Via Ripamonti 435, 20141, Milan, Italy
| | - Maria Grazia Malabarba
- Dipartimento di Oncologia ed Emato-oncologia, Università degli Studi di Milano, Via Santa Sofia 9/1, 20122, Milan, Italy
- Istituto Europeo di Oncologia, Via Ripamonti 435, 20141, Milan, Italy
| | - Pier Paolo Di Fiore
- Dipartimento di Oncologia ed Emato-oncologia, Università degli Studi di Milano, Via Santa Sofia 9/1, 20122, Milan, Italy
- Istituto Europeo di Oncologia, Via Ripamonti 435, 20141, Milan, Italy
| | - Sara Sigismund
- Dipartimento di Oncologia ed Emato-oncologia, Università degli Studi di Milano, Via Santa Sofia 9/1, 20122, Milan, Italy.
- Istituto Europeo di Oncologia, Via Ripamonti 435, 20141, Milan, Italy.
| |
Collapse
|
26
|
Chitranshi N, Dheer Y, Abbasi M, You Y, Graham SL, Gupta V. Glaucoma Pathogenesis and Neurotrophins: Focus on the Molecular and Genetic Basis for Therapeutic Prospects. Curr Neuropharmacol 2018; 16:1018-1035. [PMID: 29676228 PMCID: PMC6120108 DOI: 10.2174/1570159x16666180419121247] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 04/10/2018] [Accepted: 04/18/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Retinal ganglion cell (RGC) degeneration is a major feature of glaucoma pathology. Neuroprotective approaches that delay or halt the progression of RGC loss are needed to prevent vision loss which can occur even after conventional medical or surgical treatments to lower intraocular pressure. OBJECTIVE The aim of this review was to examine the progress in genetics and cellular mechanisms associated with endoplasmic reticulum (ER) stress, RGC dysfunction and cell death pathways in glaucoma. MATERIALS AND METHODS Here, we review the involvement of neurotrophins like brain derived neurotrophic factor (BDNF) and its high affinity receptor tropomyosin receptor kinase (TrkB) in glaucoma. The role of ER stress markers in human and animal retinas in health and disease conditions is also discussed. Further, we analysed the literature highlighting genetic linkage in the context of primary open angle glaucoma and suggested mechanistic insights into potential therapeutic options relevant to glaucoma management. RESULTS The literature review of the neurobiology underlying neurotrophin pathways, ER stress and gene associations provide critical insights into association of RGCs death in glaucoma. Alteration in signalling pathway is associated with increased risk of misfolded protein aggregation in ER promoting RGC apoptosis. Several genes that are linked with neurotrophin signalling pathways have been reported to be associated with glaucoma pathology. CONCLUSION Understanding genetic heterogeneity and involvement of neurotrophin biology in glaucoma could help to understand the complex pathophysiology of glaucoma. Identification of novel molecular targets will be critical for drug development and provide neuroprotection to the RGCs and optic nerve.
Collapse
Affiliation(s)
- Nitin Chitranshi
- Address correspondence to this author at the Faculty of Medicine and Health Sciences, 75, Talavera Road, Macquarie University, Sydney, NSW 2109, Australia; Tel: +61-298502760; E-mail:
| | | | | | | | | | | |
Collapse
|
27
|
Wehner AB, Abdesselem H, Dickendesher TL, Imai F, Yoshida Y, Giger RJ, Pierchala BA. Semaphorin 3A is a retrograde cell death signal in developing sympathetic neurons. Development 2017; 143:1560-70. [PMID: 27143756 DOI: 10.1242/dev.134627] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 02/29/2016] [Indexed: 12/30/2022]
Abstract
During development of the peripheral nervous system, excess neurons are generated, most of which will be lost by programmed cell death due to a limited supply of neurotrophic factors from their targets. Other environmental factors, such as 'competition factors' produced by neurons themselves, and axon guidance molecules have also been implicated in developmental cell death. Semaphorin 3A (Sema3A), in addition to its function as a chemorepulsive guidance cue, can also induce death of sensory neurons in vitro The extent to which Sema3A regulates developmental cell death in vivo, however, is debated. We show that in compartmentalized cultures of rat sympathetic neurons, a Sema3A-initiated apoptosis signal is retrogradely transported from axon terminals to cell bodies to induce cell death. Sema3A-mediated apoptosis utilizes the extrinsic pathway and requires both neuropilin 1 and plexin A3. Sema3A is not retrogradely transported in older, survival factor-independent sympathetic neurons, and is much less effective at inducing apoptosis in these neurons. Importantly, deletion of either neuropilin 1 or plexin A3 significantly reduces developmental cell death in the superior cervical ganglia. Taken together, a Sema3A-initiated apoptotic signaling complex regulates the apoptosis of sympathetic neurons during the period of naturally occurring cell death.
Collapse
Affiliation(s)
- Amanda B Wehner
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA Neuroscience Program, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Houari Abdesselem
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| | - Travis L Dickendesher
- Neuroscience Program, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA Department of Cell and Developmental Biology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Fumiyasu Imai
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45299, USA
| | - Yutaka Yoshida
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45299, USA
| | - Roman J Giger
- Neuroscience Program, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA Department of Cell and Developmental Biology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA Department of Neurology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Brian A Pierchala
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA Neuroscience Program, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| |
Collapse
|
28
|
Retrograde apoptotic signaling by the p75 neurotrophin receptor. Neuronal Signal 2017; 1:NS20160007. [PMID: 32714573 PMCID: PMC7373242 DOI: 10.1042/ns20160007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 01/23/2017] [Accepted: 01/25/2017] [Indexed: 02/06/2023] Open
Abstract
Neurotrophins are target-derived factors necessary for mammalian nervous system development and maintenance. They are typically produced by neuronal target tissues and interact with their receptors at axonal endings. Therefore, locally generated neurotrophin signals must be conveyed from the axon back to the cell soma. Retrograde survival signaling by neurotrophin binding to Trk receptors has been extensively studied. However, neurotrophins also bind to the p75 receptor, which can induce apoptosis in a variety of contexts. Selective activation of p75 at distal axon ends has been shown to generate a retrograde apoptotic signal, although the mechanisms involved are poorly understood. The present review summarizes the available evidence for retrograde proapoptotic signaling in general and the role of the p75 receptor in particular, with discussion of unanswered questions in the field. In-depth knowledge of the mechanisms of retrograde apoptotic signaling is essential for understanding the etiology of neurodegeneration in many diseases and injuries.
Collapse
|
29
|
Goshima Y, Yamashita N, Nakamura F, Sasaki Y. Regulation of dendritic development by semaphorin 3A through novel intracellular remote signaling. Cell Adh Migr 2016; 10:627-640. [PMID: 27392015 DOI: 10.1080/19336918.2016.1210758] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Numerous cell adhesion molecules, extracellular matrix proteins and axon guidance molecules participate in neuronal network formation through local effects at axo-dendritic, axo-axonic or dendro-dendritic contact sites. In contrast, neurotrophins and their receptors play crucial roles in neural wiring by sending retrograde signals to remote cell bodies. Semaphorin 3A (Sema3A), a prototype of secreted type 3 semaphorins, is implicated in axon repulsion, dendritic branching and synapse formation via binding protein neuropilin-1 (NRP1) and the signal transducing protein PlexinAs (PlexAs) complex. This review focuses on Sema3A retrograde signaling that regulates dendritic localization of AMPA-type glutamate receptor GluA2 and dendritic patterning. This signaling is elicited by activation of NRP1 in growth cones and is propagated to cell bodies by dynein-dependent retrograde axonal transport of PlexAs. It also requires interaction between PlexAs and a high-affinity receptor for nerve growth factor, toropomyosin receptor kinase A. We propose a control mechanism by which retrograde Sema3A signaling regulates the glutamate receptor localization through trafficking of cis-interacting PlexAs with GluA2 along dendrites; this remote signaling may be an alternative mechanism to local adhesive contacts for neural network formation.
Collapse
Affiliation(s)
- Yoshio Goshima
- a Department of Molecular Pharmacology and Neurobiology , Yokohama City University Graduate School of Medicine , Yokohama , Japan
| | - Naoya Yamashita
- a Department of Molecular Pharmacology and Neurobiology , Yokohama City University Graduate School of Medicine , Yokohama , Japan.,c Department of Biology , Johns Hopkins University , Baltimore , MD , USA
| | - Fumio Nakamura
- a Department of Molecular Pharmacology and Neurobiology , Yokohama City University Graduate School of Medicine , Yokohama , Japan
| | - Yukio Sasaki
- b Functional Structural, Biology Laboratory, Department of Medical Life Science , Yokohama City University Graduate School of Medical Life Science , Suehirocho, Tsurumi-ku, Yokohama , Japan
| |
Collapse
|
30
|
White JA, Banerjee R, Gunawardena S. Axonal Transport and Neurodegeneration: How Marine Drugs Can Be Used for the Development of Therapeutics. Mar Drugs 2016; 14:E102. [PMID: 27213408 PMCID: PMC4882576 DOI: 10.3390/md14050102] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 04/19/2016] [Accepted: 04/26/2016] [Indexed: 11/23/2022] Open
Abstract
Unlike virtually any other cells in the human body, neurons are tasked with the unique problem of transporting important factors from sites of synthesis at the cell bodies, across enormous distances, along narrow-caliber projections, to distally located nerve terminals in order to maintain cell viability. As a result, axonal transport is a highly regulated process whereby necessary cargoes of all types are packaged and shipped from one end of the neuron to the other. Interruptions in this finely tuned transport have been linked to many neurodegenerative disorders including Alzheimer's (AD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS) suggesting that this pathway is likely perturbed early in disease progression. Therefore, developing therapeutics targeted at modifying transport defects could potentially avert disease progression. In this review, we examine a variety of potential compounds identified from marine aquatic species that affect the axonal transport pathway. These compounds have been shown to function in microtubule (MT) assembly and maintenance, motor protein control, and in the regulation of protein degradation pathways, such as the autophagy-lysosome processes, which are defective in many degenerative diseases. Therefore, marine compounds have great potential in developing effective treatment strategies aimed at early defects which, over time, will restore transport and prevent cell death.
Collapse
Affiliation(s)
- Joseph A White
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, NY 14260, USA.
| | - Rupkatha Banerjee
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, NY 14260, USA.
| | - Shermali Gunawardena
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, NY 14260, USA.
| |
Collapse
|
31
|
Nirschl JJ, Magiera MM, Lazarus JE, Janke C, Holzbaur ELF. α-Tubulin Tyrosination and CLIP-170 Phosphorylation Regulate the Initiation of Dynein-Driven Transport in Neurons. Cell Rep 2016; 14:2637-52. [PMID: 26972003 PMCID: PMC4819336 DOI: 10.1016/j.celrep.2016.02.046] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 01/08/2016] [Accepted: 02/05/2016] [Indexed: 11/18/2022] Open
Abstract
Motor-cargo recruitment to microtubules is often the rate-limiting step of intracellular transport, and defects in this recruitment can cause neurodegenerative disease. Here, we use in vitro reconstitution assays with single-molecule resolution, live-cell transport assays in primary neurons, computational image analysis, and computer simulations to investigate the factors regulating retrograde transport initiation in the distal axon. We find that phosphorylation of the cytoskeletal-organelle linker protein CLIP-170 and post-translational modifications of the microtubule track combine to precisely control the initiation of retrograde transport. Computer simulations of organelle dynamics in the distal axon indicate that while CLIP-170 primarily regulates the time to microtubule encounter, the tyrosination state of the microtubule lattice regulates the likelihood of binding. These mechanisms interact to control transport initiation in the axon in a manner sensitive to the specialized cytoskeletal architecture of the neuron.
Collapse
Affiliation(s)
- Jeffrey J Nirschl
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Maria M Magiera
- Institut Curie, PSL Research University, CNRS UMR3348, 91405 Orsay, France; Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, 91405 Orsay, France
| | - Jacob E Lazarus
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Carsten Janke
- Institut Curie, PSL Research University, CNRS UMR3348, 91405 Orsay, France; Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, 91405 Orsay, France
| | - Erika L F Holzbaur
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
32
|
LIN RUHUI, CHEN JIXIANG, LI XIAOJIE, MAO JINGJIE, WU YUNAN, ZHUO PEIYUAN, ZHANG YINZHENG, LIU WEILIN, HUANG JIA, TAO JING, CHEN LIDIAN. Electroacupuncture at the Baihui acupoint alleviates cognitive impairment and exerts neuroprotective effects by modulating the expression and processing of brain-derived neurotrophic factor in APP/PS1 transgenic mice. Mol Med Rep 2016; 13:1611-7. [PMID: 26739187 PMCID: PMC4732857 DOI: 10.3892/mmr.2015.4751] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 12/02/2015] [Indexed: 01/01/2023] Open
Abstract
Alzheimer's disease (AD) is a common human neurodegenerative disorder characterized by progressive deterioration of cognition and memory. Acupuncture at the Baihui (DU20) acupoint has long been used in China to clinically treat cognitive impairment. However, the precise mechanism underlying its neuroprotective effects remains to be elucidated. In the present study, electroacupuncture (EA) at the Baihui (DU20) acupoint was observed to markedly ameliorate cognitive impairments, reduce the aberrant overexpression of β-amyloid(1-42), and inhibit neuronal apoptosis in APP/PS1 mice. As brain-derived neurotrophic factor (BDNF) has been implicated in the pathogenesis of AD, the expression and processing of BDNF in APP/PS1 mice was investigated. EA at the Baihui (DU20) acupoint was indicated to significantly enhance the expression levels of mature BDNF and proBDNF in APP/PS1 mice. Furthermore, an increase in the BDNF/proBDNF ratio, upregulation of the expression levels of phosphorylated tropomyosin receptor kinase B and a decrease in the expression level of p75 neurotrophin receptor were also observed in the APP/PS1 mice. The present study demonstrates the efficacy of EA at the Baihui (DU20) acupoint in the treatment of cognitive impairments in APP/PS1 transgenic mice. The present study hypothesized that modulation of BDNF expression and processing may be the underlying mechanism by which stimulation of the Baihui (DU20) acupoint exerts its neuroprotective effect.
Collapse
Affiliation(s)
- RUHUI LIN
- College of Rehabilitation Medicine, Fujian University of Traditional Medicine, Fuzhou, Fujian 350122, P.R. China
- Academy of Integrative Medicine Biomedical Research Center, Fuzhou, Fujian 350122, P.R. China
| | - JIXIANG CHEN
- College of Rehabilitation Medicine, Fujian University of Traditional Medicine, Fuzhou, Fujian 350122, P.R. China
- Department of Rehabilitation Medicine, Guangzhou First People's Hospital, Guangzhou, Guangdong 510180, P.R. China
| | - XIAOJIE LI
- College of Rehabilitation Medicine, Fujian University of Traditional Medicine, Fuzhou, Fujian 350122, P.R. China
| | - JINGJIE MAO
- Academy of Integrative Medicine Biomedical Research Center, Fuzhou, Fujian 350122, P.R. China
| | - YUNAN WU
- College of Rehabilitation Medicine, Fujian University of Traditional Medicine, Fuzhou, Fujian 350122, P.R. China
| | - PEIYUAN ZHUO
- College of Rehabilitation Medicine, Fujian University of Traditional Medicine, Fuzhou, Fujian 350122, P.R. China
| | - YINZHENG ZHANG
- College of Rehabilitation Medicine, Fujian University of Traditional Medicine, Fuzhou, Fujian 350122, P.R. China
| | - WEILIN LIU
- College of Rehabilitation Medicine, Fujian University of Traditional Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Exercise Rehabilitation, Fuzhou, Fujian 350122, P.R. China
| | - JIA HUANG
- College of Rehabilitation Medicine, Fujian University of Traditional Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Exercise Rehabilitation, Fuzhou, Fujian 350122, P.R. China
| | - JING TAO
- College of Rehabilitation Medicine, Fujian University of Traditional Medicine, Fuzhou, Fujian 350122, P.R. China
- TCM Rehabilitation Research Center of SATCM, Fuzhou, Fujian 350122, P.R. China
| | - LI-DIAN CHEN
- College of Rehabilitation Medicine, Fujian University of Traditional Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Rehabilitation Tech Co-innovation Center, Fujian University of Traditional Medicine, Fuzhou, Fujian 350122, P.R. China
| |
Collapse
|
33
|
Godo O, Gaskell K, K. Pathak G, R. Kyrtsos C, H. Ehrman S, B. Shah S. Characterization of fluorescent iron nanoparticles—candidates for multimodal tracking of neuronal transport. AIMS BIOENGINEERING 2016. [DOI: 10.3934/bioeng.2016.3.362] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
34
|
Activity-dependent BDNF release via endocytic pathways is regulated by synaptotagmin-6 and complexin. Proc Natl Acad Sci U S A 2015. [PMID: 26216953 DOI: 10.1073/pnas.1511830112] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is known to modulate synapse development and plasticity, but the source of synaptic BDNF and molecular mechanisms regulating BDNF release remain unclear. Using exogenous BDNF tagged with quantum dots (BDNF-QDs), we found that endocytosed BDNF-QDs were preferentially localized to postsynaptic sites in the dendrite of cultured hippocampal neurons. Repetitive neuronal spiking induced the release of BDNF-QDs at these sites, and this process required activation of glutamate receptors. Down-regulating complexin 1/2 (Cpx1/2) expression eliminated activity-induced BDNF-QD secretion, although the overall activity-independent secretion was elevated. Among eight synaptotagmin (Syt) isoforms examined, down-regulation of only Syt6 impaired activity-induced BDNF-QD secretion. In contrast, activity-induced release of endogenously synthesized BDNF did not depend on Syt6. Thus, neuronal activity could trigger the release of endosomal BDNF from postsynaptic dendrites in a Cpx- and Syt6-dependent manner, and endosomes containing BDNF may serve as a source of BDNF for activity-dependent synaptic modulation.
Collapse
|
35
|
Yang ML, Shin J, Kearns CA, Langworthy MM, Snell H, Walker MB, Appel B. CNS myelination requires cytoplasmic dynein function. Dev Dyn 2015; 244:134-45. [PMID: 25488883 DOI: 10.1002/dvdy.24238] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 11/26/2014] [Accepted: 12/03/2014] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Cytoplasmic dynein provides the main motor force for minus-end-directed transport of cargo on microtubules. Within the vertebrate central nervous system (CNS), proliferation, neuronal migration, and retrograde axon transport are among the cellular functions known to require dynein. Accordingly, mutations of DYNC1H1, which encodes the heavy chain subunit of cytoplasmic dynein, have been linked to developmental brain malformations and axonal pathologies. Oligodendrocytes, the myelinating glial cell type of the CNS, migrate from their origins to their target axons and subsequently extend multiple long processes that ensheath axons with specialized insulating membrane. These processes are filled with microtubules, which facilitate molecular transport of myelin components. However, whether oligodendrocytes require cytoplasmic dynein to ensheath axons with myelin is not known. RESULTS We identified a mutation of zebrafish dync1h1 in a forward genetic screen that caused a deficit of oligodendrocytes. Using in vivo imaging and gene expression analyses, we additionally found evidence that dync1h1 promotes axon ensheathment and myelin gene expression. CONCLUSIONS In addition to its well known roles in axon transport and neuronal migration, cytoplasmic dynein contributes to neural development by promoting myelination.
Collapse
|
36
|
Nuschke AC, Farrell SR, Levesque JM, Chauhan BC. Assessment of retinal ganglion cell damage in glaucomatous optic neuropathy: Axon transport, injury and soma loss. Exp Eye Res 2015; 141:111-24. [PMID: 26070986 DOI: 10.1016/j.exer.2015.06.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 06/01/2015] [Accepted: 06/06/2015] [Indexed: 02/07/2023]
Abstract
Glaucoma is a disease characterized by progressive axonal pathology and death of retinal ganglion cells (RGCs), which causes structural changes in the optic nerve head and irreversible vision loss. Several experimental models of glaucomatous optic neuropathy (GON) have been developed, primarily in non-human primates and, more recently and commonly, in rodents. These models provide important research tools to study the mechanisms underlying glaucomatous damage. Moreover, experimental GON provides the ability to quantify and monitor risk factors leading to RGC loss such as the level of intraocular pressure, axonal health and the RGC population. Using these experimental models we are able to gain a better understanding of GON, which allows for the development of potential neuroprotective strategies. Here we review the advantages and disadvantages of the relevant and most often utilized methods for evaluating axonal degeneration and RGC loss in GON. Axonal pathology in GON includes functional disruption of axonal transport (AT) and structural degeneration. Horseradish peroxidase (HRP), rhodamine-B-isothiocyanate (RITC) and cholera toxin-B (CTB) fluorescent conjugates have proven to be effective reporters of AT. Also, immunohistochemistry (IHC) for endogenous AT-associated proteins is often used as an indicator of AT function. Similarly, structural degeneration of axons in GON can be investigated via changes in the activity and expression of key axonal enzymes and structural proteins. Assessment of axonal degeneration can be measured by direct quantification of axons, qualitative grading, or a combination of both methods. RGC loss is the most frequently quantified variable in studies of experimental GON. Retrograde tracers can be used to quantify RGC populations in rodents via application to the superior colliculus (SC). In addition, in situ IHC for RGC-specific proteins is a common method of RGC quantification used in many studies. Recently, transgenic mouse models that express fluorescent proteins under the Thy-1 promoter have been examined for their potential to provide specific and selective labeling of RGCs for the study of GON. While these methods represent important advances in assessing the structural and functional integrity of RGCs, each has its advantages and disadvantages; together they provide an extensive toolbox for the study of GON.
Collapse
Affiliation(s)
- Andrea C Nuschke
- Retina and Optic Nerve Research Laboratory, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Spring R Farrell
- Retina and Optic Nerve Research Laboratory, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada; Capital District Health Authority, Halifax, Nova Scotia, Canada
| | - Julie M Levesque
- Retina and Optic Nerve Research Laboratory, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Balwantray C Chauhan
- Retina and Optic Nerve Research Laboratory, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada; Capital District Health Authority, Halifax, Nova Scotia, Canada; Department of Ophthalmology and Visual Sciences, Dalhousie University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
37
|
G-protein-coupled estrogen receptor 1 is anatomically positioned to modulate synaptic plasticity in the mouse hippocampus. J Neurosci 2015; 35:2384-97. [PMID: 25673833 DOI: 10.1523/jneurosci.1298-14.2015] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Both estrous cycle and sex affect the numbers and types of neuronal and glial profiles containing the classical estrogen receptors α and β, and synaptic levels in the rodent dorsal hippocampus. Here, we examined whether the membrane estrogen receptor, G-protein-coupled estrogen receptor 1 (GPER1), is anatomically positioned in the dorsal hippocampus of mice to regulate synaptic plasticity. By light microscopy, GPER1-immunoreactivity (IR) was most noticeable in the pyramidal cell layer and interspersed interneurons, especially those in the hilus of the dentate gyrus. Diffuse GPER1-IR was found in all lamina but was most dense in stratum lucidum of CA3. Ultrastructural analysis revealed discrete extranuclear GPER1-IR affiliated with the plasma membrane and endoplasmic reticulum of neuronal perikarya and dendritic shafts, synaptic specializations in dendritic spines, and clusters of vesicles in axon terminals. Moreover, GPER1-IR was found in unmyelinated axons and glial profiles. Overall, the types and amounts of GPER1-labeled profiles were similar between males and females; however, in females elevated estrogen levels generally increased axonal labeling. Some estradiol-induced changes observed in previous studies were replicated by the GPER agonist G1: G1 increased PSD95-IR in strata oriens, lucidum, and radiatum of CA3 in ovariectomized mice 6 h after administration. In contrast, estradiol but not G1 increased Akt phosphorylation levels. Instead, GPER1 actions in the synapse may be due to interactions with synaptic scaffolding proteins, such as SAP97. These results suggest that although estrogen's actions via GPER1 may converge on the same synaptic elements, different pathways are used to achieve these actions.
Collapse
|
38
|
Lam So RW, Wong HS, Ko KM. A Traditional Chinese Medicine Approach in Treating Depression by Promoting Liver Qi Circulation: A Western Medicine Perspective. Chin Med 2015. [DOI: 10.4236/cm.2015.64021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
39
|
NGF in Early Embryogenesis, Differentiation, and Pathology in the Nervous and Immune Systems. Curr Top Behav Neurosci 2015; 29:125-152. [PMID: 26695167 DOI: 10.1007/7854_2015_420] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The physiology of NGF is extremely complex, and although the study of this neurotrophin began more than 60 years ago, it is far from being concluded. NGF, its precursor molecule pro-NGF, and their different receptor systems (i.e., TrkA, p75NTR, and sortilin) have key roles in the development and adult physiology of both the nervous and immune systems. Although the NGF receptor system and the pathways activated are similar for all types of cells sensitive to NGF, the effects exerted during embryonic differentiation and in committed mature cells are strikingly different and sometimes opposite. Bearing in mind the pleiotropic effects of NGF, alterations in its expression and synthesis, as well as variations in the types of receptor available and in their respective levels of expression, may have profound effects and play multiple roles in the development and progression of several diseases. In recent years, the use of NGF or of inhibitors of its receptors has been prospected as a therapeutic tool in a variety of neurological diseases and injuries. In this review, we outline the different roles played by the NGF system in various moments of nervous and immune system differentiation and physiology, from embryonic development to aging. The data collected over the past decades indicate that NGF activities are highly integrated among systems and are necessary for the maintenance of homeostasis. Further, more integrated and multidisciplinary studies should take into consideration these multiple and interactive aspects of NGF physiology in order to design new therapeutic strategies based on the manipulation of NGF and its intracellular pathways.
Collapse
|
40
|
Sakuma C, Kawauchi T, Haraguchi S, Shikanai M, Yamaguchi Y, Gelfand VI, Luo L, Miura M, Chihara T. Drosophila Strip serves as a platform for early endosome organization during axon elongation. Nat Commun 2014; 5:5180. [PMID: 25312435 PMCID: PMC4197811 DOI: 10.1038/ncomms6180] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 09/05/2014] [Indexed: 12/19/2022] Open
Abstract
Early endosomes are essential for regulating cell signalling and controlling the amount of cell surface molecules during neuronal morphogenesis. Early endosomes undergo retrograde transport (clustering) before their homotypic fusion. Small GTPase Rab5 is known to promote early endosomal fusion, but the mechanism linking the transport/clustering with Rab5 activity is unclear. Here we show that Drosophila Strip is a key regulator for neuronal morphogenesis. strip knockdown disturbs the early endosome clustering and Rab5-positive early endosomes become smaller and scattered. Strip genetically and biochemically interacts with both Glued (the regulator of dynein-dependent transport) and Sprint (the guanine nucleotide exchange factor for Rab5), suggesting that Strip is a molecular linker between retrograde transport and Rab5 activation. Overexpression of an active form of Rab5 in strip mutant neurons suppresses the axon elongation defects. Thus, Strip acts as a molecular platform for the early endosome organization that plays important roles in neuronal morphogenesis.
Collapse
Affiliation(s)
- Chisako Sakuma
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Takeshi Kawauchi
- 1] Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan [2] PRESTO, Japan Science and Technology Agency (JST), 7, Gobancho, Chiyoda-ku, Tokyo 102-0076, Japan
| | - Shuka Haraguchi
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Mima Shikanai
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Yoshifumi Yamaguchi
- 1] Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan [2] PRESTO, Japan Science and Technology Agency (JST), 7, Gobancho, Chiyoda-ku, Tokyo 102-0076, Japan
| | - Vladimir I Gelfand
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - Liqun Luo
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, California 94305, USA
| | - Masayuki Miura
- 1] Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan [2] CREST, Japan Science and Technology Agency (JST), 7, Gobancho, Chiyoda-ku, Tokyo 102-0076, Japan
| | - Takahiro Chihara
- 1] Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan [2] PRESTO, Japan Science and Technology Agency (JST), 7, Gobancho, Chiyoda-ku, Tokyo 102-0076, Japan [3] CREST, Japan Science and Technology Agency (JST), 7, Gobancho, Chiyoda-ku, Tokyo 102-0076, Japan
| |
Collapse
|
41
|
Kang MJ, Hansen TJ, Mickiewicz M, Kaczynski TJ, Fye S, Gunawardena S. Disruption of axonal transport perturbs bone morphogenetic protein (BMP)--signaling and contributes to synaptic abnormalities in two neurodegenerative diseases. PLoS One 2014; 9:e104617. [PMID: 25127478 PMCID: PMC4134223 DOI: 10.1371/journal.pone.0104617] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 07/15/2014] [Indexed: 01/14/2023] Open
Abstract
Formation of new synapses or maintenance of existing synapses requires the delivery of synaptic components from the soma to the nerve termini via axonal transport. One pathway that is important in synapse formation, maintenance and function of the Drosophila neuromuscular junction (NMJ) is the bone morphogenetic protein (BMP)-signaling pathway. Here we show that perturbations in axonal transport directly disrupt BMP signaling, as measured by its downstream signal, phospho Mad (p-Mad). We found that components of the BMP pathway genetically interact with both kinesin-1 and dynein motor proteins. Thick vein (TKV) vesicle motility was also perturbed by reductions in kinesin-1 or dynein motors. Interestingly, dynein mutations severely disrupted p-Mad signaling while kinesin-1 mutants showed a mild reduction in p-Mad signal intensity. Similar to mutants in components of the BMP pathway, both kinesin-1 and dynein motor protein mutants also showed synaptic morphological defects. Strikingly TKV motility and p-Mad signaling were disrupted in larvae expressing two human disease proteins; expansions of glutamine repeats (polyQ77) and human amyloid precursor protein (APP) with a familial Alzheimer's disease (AD) mutation (APPswe). Consistent with axonal transport defects, larvae expressing these disease proteins showed accumulations of synaptic proteins along axons and synaptic abnormalities. Taken together our results suggest that similar to the NGF-TrkA signaling endosome, a BMP signaling endosome that directly interacts with molecular motors likely exist. Thus problems in axonal transport occurs early, perturbs BMP signaling, and likely contributes to the synaptic abnormalities observed in these two diseases.
Collapse
Affiliation(s)
- Min Jung Kang
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Timothy J. Hansen
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Monique Mickiewicz
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Tadeusz J. Kaczynski
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Samantha Fye
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Shermali Gunawardena
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, New York, United States of America
- * E-mail:
| |
Collapse
|
42
|
Kondapalli KC, Prasad H, Rao R. An inside job: how endosomal Na(+)/H(+) exchangers link to autism and neurological disease. Front Cell Neurosci 2014; 8:172. [PMID: 25002837 PMCID: PMC4066934 DOI: 10.3389/fncel.2014.00172] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 06/04/2014] [Indexed: 12/02/2022] Open
Abstract
Autism imposes a major impediment to childhood development and a huge emotional and financial burden on society. In recent years, there has been rapidly accumulating genetic evidence that links the eNHE, a subset of Na(+)/H(+) exchangers that localize to intracellular vesicles, to a variety of neurological conditions including autism, attention deficit hyperactivity disorder (ADHD), intellectual disability, and epilepsy. By providing a leak pathway for protons pumped by the V-ATPase, eNHE determine luminal pH and regulate cation (Na(+), K(+)) content in early and recycling endosomal compartments. Loss-of-function mutations in eNHE cause hyperacidification of endosomal lumen, as a result of imbalance in pump and leak pathways. Two isoforms, NHE6 and NHE9 are highly expressed in brain, including hippocampus and cortex. Here, we summarize evidence for the importance of luminal cation content and pH on processing, delivery and fate of cargo. Drawing upon insights from model organisms and mammalian cells we show how eNHE affect surface expression and function of membrane receptors and neurotransmitter transporters. These studies lead to cellular models of eNHE activity in pre- and post-synaptic neurons and astrocytes, where they could impact synapse development and plasticity. The study of eNHE has provided new insight on the mechanism of autism and other debilitating neurological disorders and opened up new possibilities for therapeutic intervention.
Collapse
Affiliation(s)
| | | | - Rajini Rao
- Department of Physiology, The Johns Hopkins University School of MedicineBaltimore, MD, USA
| |
Collapse
|
43
|
Vermehren-Schmaedick A, Krueger W, Jacob T, Ramunno-Johnson D, Balkowiec A, Lidke KA, Vu TQ. Heterogeneous intracellular trafficking dynamics of brain-derived neurotrophic factor complexes in the neuronal soma revealed by single quantum dot tracking. PLoS One 2014; 9:e95113. [PMID: 24732948 PMCID: PMC3986401 DOI: 10.1371/journal.pone.0095113] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 03/23/2014] [Indexed: 01/19/2023] Open
Abstract
Accumulating evidence underscores the importance of ligand-receptor dynamics in shaping cellular signaling. In the nervous system, growth factor-activated Trk receptor trafficking serves to convey biochemical signaling that underlies fundamental neural functions. Focus has been placed on axonal trafficking but little is known about growth factor-activated Trk dynamics in the neuronal soma, particularly at the molecular scale, due in large part to technical hurdles in observing individual growth factor-Trk complexes for long periods of time inside live cells. Quantum dots (QDs) are intensely fluorescent nanoparticles that have been used to study the dynamics of ligand-receptor complexes at the plasma membrane but the value of QDs for investigating ligand-receptor intracellular dynamics has not been well exploited. The current study establishes that QD conjugated brain-derived neurotrophic factor (QD-BDNF) binds to TrkB receptors with high specificity, activates TrkB downstream signaling, and allows single QD tracking capability for long recording durations deep within the soma of live neurons. QD-BDNF complexes undergo internalization, recycling, and intracellular trafficking in the neuronal soma. These trafficking events exhibit little time-synchrony and diverse heterogeneity in underlying dynamics that include phases of sustained rapid motor transport without pause as well as immobility of surprisingly long-lasting duration (several minutes). Moreover, the trajectories formed by dynamic individual BDNF complexes show no apparent end destination; BDNF complexes can be found meandering over long distances of several microns throughout the expanse of the neuronal soma in a circuitous fashion. The complex, heterogeneous nature of neuronal soma trafficking dynamics contrasts the reported linear nature of axonal transport data and calls for models that surpass our generally limited notions of nuclear-directed transport in the soma. QD-ligand probes are poised to provide understanding of how the molecular mechanisms underlying intracellular ligand-receptor trafficking shape cell signaling under conditions of both healthy and dysfunctional neurological disease models.
Collapse
Affiliation(s)
- Anke Vermehren-Schmaedick
- Department of Biomedical Engineering and Center for Spatial Systems Biomedicine, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Wesley Krueger
- Department of Physics & Astronomy, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Thomas Jacob
- Department of Biomedical Engineering and Center for Spatial Systems Biomedicine, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Damien Ramunno-Johnson
- Department of Biomedical Engineering and Center for Spatial Systems Biomedicine, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Agnieszka Balkowiec
- Department of Integrative Biosciences, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Keith A. Lidke
- Department of Physics & Astronomy, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Tania Q. Vu
- Department of Biomedical Engineering and Center for Spatial Systems Biomedicine, Oregon Health & Science University, Portland, Oregon, United States of America
- * E-mail:
| |
Collapse
|
44
|
A defect in the CLIP1 gene (CLIP-170) can cause autosomal recessive intellectual disability. Eur J Hum Genet 2014; 23:331-6. [PMID: 24569606 DOI: 10.1038/ejhg.2014.13] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 01/12/2014] [Accepted: 01/16/2014] [Indexed: 12/29/2022] Open
Abstract
In the context of a comprehensive research project, investigating novel autosomal recessive intellectual disability (ARID) genes, linkage analysis based on autozygosity mapping helped identify an intellectual disability locus on Chr.12q24, in an Iranian family (LOD score = 3.7). Next-generation sequencing (NGS) following exon enrichment in this novel interval, detected a nonsense mutation (p.Q1010*) in the CLIP1 gene. CLIP1 encodes a member of microtubule (MT) plus-end tracking proteins, which specifically associates with the ends of growing MTs. These proteins regulate MT dynamic behavior and are important for MT-mediated transport over the length of axons and dendrites. As such, CLIP1 may have a role in neuronal development. We studied lymphoblastoid and skin fibroblast cell lines established from healthy and affected patients. RT-PCR and western blot analyses showed the absence of CLIP1 transcript and protein in lymphoblastoid cells derived from affected patients. Furthermore, immunofluorescence analyses showed MT plus-end staining only in fibroblasts containing the wild-type (and not the mutant) CLIP1 protein. Collectively, our data suggest that defects in CLIP1 may lead to ARID.
Collapse
|
45
|
Abstract
The distinctive morphology of neurons, with complex dendritic arbors and extensive axons, presents spatial challenges for intracellular signal transduction. The endosomal system provides mechanisms that enable signaling molecules initiated by extracellular cues to be trafficked throughout the expanse of the neuron, allowing intracellular signals to be sustained over long distances. Therefore endosomes are critical for many aspects of neuronal signaling that regulate cell survival, axonal growth and guidance, dendritic branching, and cell migration. An intriguing characteristic of neuronal signal transduction is that endosomal trafficking enables physiological responses that vary based on the subcellular location of signal initiation. In this review, we will discuss the specialized mechanisms and the functional significance of endosomal signaling in neurons, both during normal development and in disease.
Collapse
Affiliation(s)
- Katharina E Cosker
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115
| | | |
Collapse
|
46
|
Harrison BJ, Flight RM, Gomes C, Venkat G, Ellis SR, Sankar U, Twiss JL, Rouchka EC, Petruska JC. IB4-binding sensory neurons in the adult rat express a novel 3' UTR-extended isoform of CaMK4 that is associated with its localization to axons. J Comp Neurol 2014; 522:308-36. [PMID: 23817991 PMCID: PMC3855891 DOI: 10.1002/cne.23398] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 06/13/2013] [Accepted: 06/19/2013] [Indexed: 01/22/2023]
Abstract
Calcium/calmodulin-dependent protein kinase 4 (gene and transcript: CaMK4; protein: CaMKIV) is the nuclear effector of the Ca(2+) /calmodulin kinase (CaMK) pathway where it coordinates transcriptional responses. However, CaMKIV is present in the cytoplasm and axons of subpopulations of neurons, including some sensory neurons of the dorsal root ganglia (DRG), suggesting an extranuclear role for this protein. We observed that CaMKIV was expressed strongly in the cytoplasm and axons of a subpopulation of small-diameter DRG neurons, most likely cutaneous nociceptors by virtue of their binding the isolectin IB4. In IB4+ spinal nerve axons, 20% of CaMKIV was colocalized with the endocytic marker Rab7 in axons that highly expressed CAM-kinase-kinase (CAMKK), an upstream activator of CaMKIV, suggesting a role for CaMKIV in signaling though signaling endosomes. Using fluorescent in situ hybridization (FISH) with riboprobes, we also observed that small-diameter neurons expressed high levels of a novel 3' untranslated region (UTR) variant of CaMK4 mRNA. Using rapid amplification of cDNA ends (RACE), reverse-transcription polymerase chain reaction (RT-PCR) with gene-specific primers, and cDNA sequencing analyses we determined that the novel transcript contains an additional 10 kb beyond the annotated gene terminus to a highly conserved alternate polyadenylation site. Quantitative PCR (qPCR) analyses of fluorescent-activated cell sorted (FACS) DRG neurons confirmed that this 3'-UTR-extended variant was preferentially expressed in IB4-binding neurons. Computational analyses of the 3'-UTR sequence predict that UTR-extension introduces consensus sites for RNA-binding proteins (RBPs) including the embryonic lethal abnormal vision (ELAV)/Hu family proteins. We consider the possible implications of axonal CaMKIV in the context of the unique properties of IB4-binding DRG neurons.
Collapse
Affiliation(s)
- Benjamin J. Harrison
- Anatomical Sciences and Neurobiology, University of Louisville, Louisville, Kentucky, 40202, USA
- Kentucky Spinal Cord Injury Research Center (KSCIRC), University of Louisville, Louisville, Kentucky, 40292, USA
| | - Robert M. Flight
- Anatomical Sciences and Neurobiology, University of Louisville, Louisville, Kentucky, 40202, USA
| | - Cynthia Gomes
- Department of Biochemistry and Molecular Bi ology, University of Louisville School of Medicine, Kentucky, 40202, USA
| | - Gayathri Venkat
- Anatomical Sciences and Neurobiology, University of Louisville, Louisville, Kentucky, 40202, USA
- Kentucky Spinal Cord Injury Research Center (KSCIRC), University of Louisville, Louisville, Kentucky, 40292, USA
| | - Steven R Ellis
- Department of Biochemistry and Molecular Bi ology, University of Louisville School of Medicine, Kentucky, 40202, USA
| | - Uma Sankar
- James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, 40292, USA
- Owensboro Cancer Research Program, University of Louisville, Owensboro, KY 42303, USA
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, 40292, USA
| | - Jeffery L. Twiss
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, 19104, USA
| | - Eric C. Rouchka
- Department of Computer Engineering and Computer Science, University of Louisville, Louisville, Kentucky, 40292, USA
| | - Jeffrey C. Petruska
- Anatomical Sciences and Neurobiology, University of Louisville, Louisville, Kentucky, 40202, USA
- Kentucky Spinal Cord Injury Research Center (KSCIRC), University of Louisville, Louisville, Kentucky, 40292, USA
- Department of Neurological Surgery, University of Louisville, Louisville, Kentucky, 40202, USA
| |
Collapse
|
47
|
BDNF and NT4 play interchangeable roles in gustatory development. Dev Biol 2013; 386:308-20. [PMID: 24378336 DOI: 10.1016/j.ydbio.2013.12.031] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 12/17/2013] [Accepted: 12/20/2013] [Indexed: 01/10/2023]
Abstract
A limited number of growth factors are capable of regulating numerous developmental processes, but how they accomplish this is unclear. The gustatory system is ideal for examining this issue because the neurotrophins brain-derived neurotrophic factor (BDNF) and neurotrophin-4 (NT4) have different developmental roles although both of them activate the same receptors, TrkB and p75. Here we first investigated whether the different roles of BDNF and NT4 are due to their differences in temporal and spatial expression patterns. Then, we asked whether or not these two neurotrophins exert their unique roles on the gustatory system by regulating different sets of downstream genes. By using Bdnf(Nt4/Nt4) mice, in which the coding region for BDNF is replaced with NT4, we examined whether the different functions of BDNF and NT4 are interchangeable during taste development. Our results demonstrated that NT4 could mediate most of the unique roles of BDNF during taste development. Specifically, caspase-3-mediated cell death, which was increased in the geniculate ganglion in Bdnf(-/-) mice, was rescued in Bdnf(Nt4/Nt4) mice. In BDNF knockout mice, tongue innervation was disrupted, and gustatory axons failed to reach their targets. However, disrupted innervation was rescued and target innervation is normal when NT4 replaced BDNF. Genome wide expression analyses revealed that BDNF and NT4 mutant mice exhibited different gene expression profiles in the gustatory (geniculate) ganglion. Compared to wild type, the expression of differentiation-, apoptosis- and axon guidance-related genes was changed in BDNF mutant mice, which is consistent with their different roles during taste development. However, replacement of BDNF by NT4 rescued these gene expression changes. These findings indicate that the functions of BDNF and NT4 in taste development are interchangeable. Spatial and temporal differences in BDNF and NT4 expression can regulate differential gene expression in vivo and determine their specific roles during development.
Collapse
|
48
|
Pea3 transcription factor family members Etv4 and Etv5 mediate retrograde signaling and axonal growth of DRG sensory neurons in response to NGF. J Neurosci 2013; 33:15940-51. [PMID: 24089499 DOI: 10.1523/jneurosci.0928-13.2013] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Nerve growth factor (NGF) is a target-derived neurotrophic growth factor that controls many aspects of sensory and sympathetic neuronal development. The identification of transcription factors and downstream target genes that mediate NGF-dependent neuronal differentiation and target field innervation is currently a major challenge. Here, we show that the Pea3 transcription factor family members Etv4 and Etv5 are expressed by developing TrkA-positive dorsal root ganglion (DRG) neurons during the period of target innervation. Real-time PCR assays indicated that Etv4 and Etv5 mRNAs are significantly induced by NGF in different neuronal cells, suggesting that they could be involved in the biological responses induced by this neurotrophin. Interestingly, distal axon application of NGF in compartmentalized cultures of rat DRG sensory neurons was sufficient to induce a significant increase in Etv4 and Etv5 mRNA expression. Pharmacological assays also revealed that activation of MEK/ERK (MAPK) pathway is required for Etv4 and Etv5 gene induction in response to NGF. Downregulation of Etv4 and Etv5 using small interference RNA knockdown experiments inhibited NGF-induced neurite outgrowth of rat sensory neurons, while overexpression of full-length Etv4 or Etv5 potentiated neuronal differentiation in response to this neurotrophin. Together, these data establish Etv4 and Etv5 as essential molecules of the transcriptional program linking neurotrophin signaling to sensory neuronal differentiation, and suggest that they can be involved in NGF-mediated target innervation.
Collapse
|
49
|
Miaczynska M. Effects of membrane trafficking on signaling by receptor tyrosine kinases. Cold Spring Harb Perspect Biol 2013; 5:a009035. [PMID: 24186066 DOI: 10.1101/cshperspect.a009035] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The intracellular trafficking machinery contributes to the spatial and temporal control of signaling by receptor tyrosine kinases (RTKs). The primary role in this process is played by endocytic trafficking, which regulates the localization of RTKs and their downstream effectors, as well as the duration and the extent of their activity. The key regulatory points along the endocytic pathway are internalization of RTKs from the plasma membrane, their sorting to degradation or recycling, and their residence in various endosomal compartments. Here I will review factors and mechanisms that modulate RTK signaling by (1) affecting receptor internalization, (2) regulating the balance between degradation and recycling of RTK, and (3) compartmentalization of signals in endosomes and other organelles. Cumulatively, these mechanisms illustrate a multilayered control of RTK signaling exerted by the trafficking machinery.
Collapse
Affiliation(s)
- Marta Miaczynska
- International Institute of Molecular and Cell Biology, Laboratory of Cell Biology, 02-109 Warsaw, Poland
| |
Collapse
|
50
|
Releasing the brake: restoring fast axonal transport in neurodegenerative disorders. Trends Cell Biol 2013; 23:634-43. [PMID: 24091156 DOI: 10.1016/j.tcb.2013.08.007] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Revised: 08/26/2013] [Accepted: 08/27/2013] [Indexed: 12/15/2022]
Abstract
Emerging evidence suggests that the dysregulation of fast axonal transport (FAT) plays a crucial role in several neurodegenerative disorders. Some of these diseases are caused by mutations affecting the molecular motors or adaptors that mediate FAT, and transport defects in organelles such as mitochondria and vesicles are observed in most, if not all neurodegenerative disorders. The relationship between neurodegenerative disorders and FAT is probably due to the extreme polarization of neurons, which extend long processes such as axons and dendrites. These characteristics render neurons particularly sensitive to transport alterations. Here we review the impact of such alterations on neuronal survival. We also discuss various strategies that might restore FAT, potentially slowing disease progression.
Collapse
|