1
|
Ravichandran N, Iyer M, Uvarajan D, Kirola L, Kumra SM, Babu HWS, HariKrishnaReddy D, Vellingiri B, Narayanasamy A. New insights on the regulators and inhibitors of RhoA-ROCK signalling in Parkinson's disease. Metab Brain Dis 2025; 40:90. [PMID: 39775342 DOI: 10.1007/s11011-024-01500-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025]
Abstract
A multifaceted and widely prevalent neurodegenerative disease, Parkinson's disease (PD) is typified by the loss of dopaminergic neurons in the midbrain. The discovery of novel treatment(s) that can reverse or halt the course of the disease progression along with identifying the most reliable biomarker(s) in PD remains the crucial concern. RhoA in its active state has been demonstrated to interact with three distinct domains located in the central coiled-coil region of ROCK. RhoA appears to activate effectors most frequently by breaking the intramolecular autoinhibitory connections, which releases functional domains from the effector protein. Additionally, RhoA is highly expressed in the nervous system and it acts as a central molecule for its several downstream effector proteins in multiple signalling pathways both in neurons and glial cells. Mitochondrial dysfunction, vesicle transport malfunction and aggregation of α-Synuclein, a presynaptic neuronal protein genetically and neuropathologically associated with PD. While the RhoA-ROCK signalling pathway appears to have a significant role in PD symptoms, suggesting it could be a promising target for therapeutic interventions. Thus, this review article addresses the potential involvement of the RhoA-ROCK signalling system in the pathophysiology of neurodegenerative illnesses, with an emphasis on its biology and function. We also provide an overview of the state of research on RhoA regulation and its downstream biological activities, focusing on the role of RhoA signalling in neurodegenerative illnesses and the potential benefits of RhoA inhibition as a treatment for neurodegeneration.
Collapse
Affiliation(s)
- Nandita Ravichandran
- Disease Proteomics Laboratory, Department of Zoology, Bharathiar University, Coimbatore, 641046, Tamil Nadu, India
| | - Mahalaxmi Iyer
- Department of Microbiology, Central University of Punjab, Bathinda, 151401, Punjab, India
| | - Deenathayalan Uvarajan
- Department of Biochemistry, PSG College of Arts & Science, Coimbatore, Tamil Nadu, India
| | - Laxmi Kirola
- Department of Biotechnology, School of Health Sciences & Technology (SoHST), UPES Dehradun, Dehradun, India
| | - Sindduja Muthu Kumra
- Human Cytogenetics and Stem Cell Laboratory, Department of Zoology, Central University of Punjab, Bathinda, 151401, Punjab, India
| | - Harysh Winster Suresh Babu
- Human Cytogenetics and Stem Cell Laboratory, Department of Zoology, Central University of Punjab, Bathinda, 151401, Punjab, India
| | - Dibbanti HariKrishnaReddy
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, 151401, Punjab, India
| | - Balachandar Vellingiri
- Human Cytogenetics and Stem Cell Laboratory, Department of Zoology, Central University of Punjab, Bathinda, 151401, Punjab, India.
| | - Arul Narayanasamy
- Disease Proteomics Laboratory, Department of Zoology, Bharathiar University, Coimbatore, 641046, Tamil Nadu, India.
| |
Collapse
|
2
|
Zhou Y, Wang G, Liang X, Xu Z. Hindbrain networks: Exploring the hidden anxiety circuits in rodents. Behav Brain Res 2025; 476:115281. [PMID: 39374875 DOI: 10.1016/j.bbr.2024.115281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 09/23/2024] [Accepted: 10/01/2024] [Indexed: 10/09/2024]
Abstract
Anxiety disorders are multifaceted conditions that engage numerous brain regions and circuits. While the hindbrain is pivotal in fundamental biological functions, its role in modulating emotions has been underappreciated. This review will uncover critical targets and circuits within the hindbrain that are essential for both anxiety and anxiolytic effects, expanding on research obtained through behavioral tests. The bidirectional neural pathways between the hindbrain and other brain regions, with a spotlight on vagal afferent signaling, provide a crucial framework for unraveling the neural mechanisms underlying anxiety. Exploring neural circuits within the hindbrain can help to unravel the neurobiological mechanisms of anxiety and elucidate differences in the expression of these circuits between genders, thereby providing valuable insights for the development of future anxiolytic drugs.
Collapse
Affiliation(s)
- Yifu Zhou
- Department of Neurosurgery, Affiliated Hospital of Shaoxing University, Shaoxing, China
| | - Gang Wang
- Department of Neurosurgery, Affiliated Hospital of Shaoxing University, Shaoxing, China
| | - Xiaosong Liang
- Department of Neurosurgery, Affiliated Hospital of Shaoxing University, Shaoxing, China
| | - Zhidi Xu
- Department of Anesthesia and Surgery, Affiliated Hospital of Shaoxing University, Shaoxing, China.
| |
Collapse
|
3
|
Asbury S, Lai JKY, Rilett KC, Haqqee Z, Darwin BC, Ellegood J, Lerch JP, Foster JA. Host genetics maps to behaviour and brain structure in developmental mice. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2025; 21:1. [PMID: 39748372 PMCID: PMC11697848 DOI: 10.1186/s12993-024-00261-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 12/04/2024] [Indexed: 01/04/2025]
Abstract
Gene-environment interactions in the postnatal period have a long-term impact on neurodevelopment. To effectively assess neurodevelopment in the mouse, we developed a behavioural pipeline that incorporates several validated behavioural tests to measure translationally relevant milestones of behaviour in mice. The behavioral phenotype of 1060 wild type and genetically-modified mice was examined followed by structural brain imaging at 4 weeks of age. The influence of genetics, sex, and early life stress on behaviour and neuroanatomy was determined using traditional statistical and machine learning methods. Analytical results demonstrated that neuroanatomical diversity was primarily associated with genotype whereas behavioural phenotypic diversity was observed to be more susceptible to gene-environment variation. We describe a standardized mouse phenotyping pipeline, termed the Developmental Behavioural Milestones (DBM) Pipeline released alongside the 1000 Mouse Developmental Behavioural Milestones (1000 Mouse DBM) database to institute a novel framework for reproducible interventional neuroscience research.
Collapse
Affiliation(s)
- Sarah Asbury
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Jonathan K Y Lai
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Kelly C Rilett
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Zeeshan Haqqee
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Benjamin C Darwin
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, ON, Canada
| | - Jacob Ellegood
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, ON, Canada
| | - Jason P Lerch
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, ON, Canada
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Jane A Foster
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada.
- Research Institute at St. Joe's Hamilton, Hamilton, ON, Canada.
- Center for Depression Research and Clinical Care, Department of Psychiatry, O'Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
4
|
Hung H, Lin J, Teng Y, Kao C, Wang P, Soong B, Tsai T. A dominant negative Kcnd3 F227del mutation in mice causes spinocerebellar ataxia type 22 (SCA22) by impairing ER and Golgi functioning. J Pathol 2025; 265:57-68. [PMID: 39562497 PMCID: PMC11638663 DOI: 10.1002/path.6368] [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: 05/27/2024] [Revised: 09/13/2024] [Accepted: 10/13/2024] [Indexed: 11/21/2024]
Abstract
Spinocerebellar ataxia type 22 (SCA22) caused by KCND3 mutations is an autosomal dominant disorder. We established a mouse model carrying the Kcnd3 F227del mutation to study the molecular pathogenesis. Four findings were pinpointed. First, the heterozygous mice exhibited an early onset of defects in motor coordination and balance which mirror those of SCA22 patients. The degeneration and a minor loss of Purkinje cells, together with the concurrent presence of neuroinflammation, as well as the previous finding on electrophysiological changes, may all contribute to the development of the SCA22 ataxia phenotype in mice carrying the Kcnd3 F227del mutant protein. Second, the mutant protein is retained by the endoplasmic reticulum and Golgi, leading to activation of the unfolded protein response and a severe trafficking defect that affects its membrane destination. Intriguingly, profound damage of the Golgi is the earliest manifestation. Third, analysis of the transcriptome revealed that the Kcnd3 F227del mutation down-regulates a panel of genes involved in the functioning of synapses and neurogenesis which are tightly linked to the functioning of Purkinje cells. Finally, no ataxia phenotypes were detectable in knockout mice carrying a loss-of-function Kcnd3 mutation. Thus, Kcnd3 F227del is a dominant-negative mutation. This mouse model may serve as a preclinical model for exploring therapeutic strategies to treat patients. © 2024 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Hao‐Chih Hung
- Department of Life Sciences and Institute of Genome SciencesNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Jia‐Han Lin
- Department of Life Sciences and Institute of Genome SciencesNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Yuan‐Chi Teng
- Department of Life Sciences and Institute of Genome SciencesNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
- Department of Medical ResearchTaipei Veterans General HospitalTaipeiTaiwan
| | - Cheng‐Heng Kao
- Center of General EducationChang Gung UniversityTaoyuanTaiwan
| | - Pei‐Yu Wang
- Graduate Institute of Brain and Mind Sciences, College of MedicineNational Taiwan UniversityTaipeiTaiwan
| | - Bing‐Wen Soong
- Department of NeurologyTaipei Veterans General HospitalTaipeiTaiwan
- Brain Research CenterNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Ting‐Fen Tsai
- Department of Life Sciences and Institute of Genome SciencesNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
- Center for Healthy Longevity and Aging SciencesNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
- National Health Research InstitutesInstitute of Molecular and Genomic MedicineMiaoliTaiwan
| |
Collapse
|
5
|
Kundu S, Ghaemmaghami J, Sanidas G, Wolff N, Vij A, Byrd C, Simonti G, Triantafyllou M, Jablonska B, Dean T, Koutroulis I, Gallo V, Kratimenos P. Cerebellar Purkinje Cell Activity Regulates White Matter Response and Locomotor Function after Neonatal Hypoxia. J Neurosci 2025; 45:e0899242024. [PMID: 39472064 DOI: 10.1523/jneurosci.0899-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 10/14/2024] [Accepted: 10/15/2024] [Indexed: 01/03/2025] Open
Abstract
Neonatal hypoxia (Hx) causes white matter (WM) injury, particularly in the cerebellum. We previously demonstrated that Hx-induced reduction of cerebellar Purkinje cell (PC) activity results in locomotor deficits. Yet, the mechanism of Hx-induced cerebellar WM injury and associated locomotor abnormalities remains undetermined. Here, we show that the cerebellar WM injury and linked locomotor deficits are driven by PC activity and are reversed when PC activity is restored. Using optogenetics and multielectrode array recordings, we manipulated PC activity and captured the resulting cellular responses in WM oligodendrocyte precursor cells and GABAergic interneurons. To emulate the effects of Hx, we used light-activated halorhodopsin targeted specifically to the PC layer of normal mice. Suppression of PC firing activity at P13 and P21 phenocopied the locomotor deficits observed in Hx. Moreover, histopathologic analysis of the developing cerebellar WM following PC inhibition (P21) revealed a corresponding reduction in oligodendrocyte maturation and myelination, akin to our findings in Hx mice. Conversely, PC stimulation restored PC activity, promoted oligodendrocyte maturation, and enhanced myelination, resulting in reversed Hx-induced locomotor deficits. Our findings highlight the crucial role of PC activity in cerebellar WM development and locomotor performance following neonatal injury.
Collapse
Affiliation(s)
- Srikanya Kundu
- National Institutes of Health, National Center for Advancing Translational Sciences (NCATS), Bethesda, Maryland 20850
- Children's National Research Institute, Washington, DC 20012
| | - Javid Ghaemmaghami
- Children's National Research Institute, Washington, DC 20012
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109
| | | | - Nora Wolff
- Children's National Research Institute, Washington, DC 20012
| | - Abhya Vij
- Boston Children's Hospital, Boston, Massachusetts 02115
- Harvard Medical School, Boston, Massachusetts 02115
| | - Chad Byrd
- Children's National Research Institute, Washington, DC 20012
| | | | | | - Beata Jablonska
- Children's National Research Institute, Washington, DC 20012
- The George Washington University School of Medicine and Health Sciences, Washington, DC 20052
| | - Terry Dean
- Children's National Research Institute, Washington, DC 20012
- The George Washington University School of Medicine and Health Sciences, Washington, DC 20052
- Children's National Hospital, Washington, DC 20010
| | - Ioannis Koutroulis
- Children's National Research Institute, Washington, DC 20012
- The George Washington University School of Medicine and Health Sciences, Washington, DC 20052
- Children's National Hospital, Washington, DC 20010
| | - Vittorio Gallo
- Children's National Research Institute, Washington, DC 20012
- The George Washington University School of Medicine and Health Sciences, Washington, DC 20052
- Children's National Hospital, Washington, DC 20010
- Seattle Children's Research Institute, Seattle, Washington 98105
| | - Panagiotis Kratimenos
- Children's National Research Institute, Washington, DC 20012
- The George Washington University School of Medicine and Health Sciences, Washington, DC 20052
- Children's National Hospital, Washington, DC 20010
| |
Collapse
|
6
|
Wu GY, Li RX, Liu J, Sun L, Yi YL, Yao J, Tang BQ, Wen HZ, Chen PH, Lou YX, Li HL, Sui JF. An excitatory neural circuit for descending inhibition of itch processing. Cell Rep 2024; 43:115062. [PMID: 39666458 DOI: 10.1016/j.celrep.2024.115062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/07/2024] [Accepted: 11/21/2024] [Indexed: 12/14/2024] Open
Abstract
Itch serves as a self-protection mechanism against harmful external agents, whereas uncontrolled and persistent itch severely influences the quality of life of patients and aggravates their diseases. Unfortunately, the existing treatments are largely ineffective. The current difficulty in treatment may be closely related to the fact that the central neural mechanisms underlying itch processing, especially descending inhibition of itch, are poorly understood. Here, we demonstrate that an excitatory descending neural circuit from rostral anterior cingulate cortex pyramidal (rACCPy) neurons to periaqueductal gray GABAergic (PAGGABA) neurons plays a key role in the inhibition of itch. The activity of itch-tagged rACCPy neurons decreases during the itch-evoked scratching period. Artificial activation or inhibition of the neural circuits significantly impairs or enhances itch processing, respectively. Thus, an excitatory neural circuit is identified as playing a crucial inhibitory role in descending regulation of itch, suggesting that it could be a potential target for treating itch.
Collapse
Affiliation(s)
- Guang-Yan Wu
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China.
| | - Ruo-Xuan Li
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Ju Liu
- Department of Foreign Languages, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Lin Sun
- Southwest Hospital, Army Medical University, Chongqing 400038, China
| | - Yi-Lun Yi
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Juan Yao
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Bo-Qin Tang
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Hui-Zhong Wen
- Department of Neurobiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Peng-Hui Chen
- Department of Neurobiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Yun-Xiao Lou
- Department of Neurobiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Hong-Li Li
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China.
| | - Jian-Feng Sui
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China.
| |
Collapse
|
7
|
He XB, Guo F, Zhang W, Fan J, Le W, Chen Q, Ma Y, Zheng Y, Lee SH, Wang HJ, Wu Y, Zhou Q, Yang R. JMJD3 deficiency disturbs dopamine biosynthesis in midbrain and aggravates chronic inflammatory pain. Acta Neuropathol Commun 2024; 12:201. [PMID: 39716224 DOI: 10.1186/s40478-024-01912-x] [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: 09/25/2024] [Accepted: 12/09/2024] [Indexed: 12/25/2024] Open
Abstract
Midbrain dopamine (mDA) neurons participate in a wide range of brain functions through an intricate regulation of DA biosynthesis. The epigenetic factors and mechanisms in this process are not well understood. Here we report that histone demethylase JMJD3 is a critical regulator for DA biosynthesis in adult mouse mDA neurons. Mice carrying Jmjd3 conditional knockout or undergoing pharmaceutical inhibition of JMJD3 showed consistent reduction of DA content in midbrain and striatum. Histological examination of both mice confirmed that TH and NURR1, two key molecules in DA biosynthesis pathway, were decreased in mDA neurons. Mechanistic experiments in vivo and in vitro further demonstrated that the transcriptions of Th and Nurr1 in mDA neurons were suppressed by JMJD3 deficiency, because of increased repressive H3K27me3 and attenuated bindings of JMJD3 and NURR1 on the promoters of both genes. On behavioral level, a significant prolonged inflammation-induced mechanical hyperalgesia was found in conditional knockout mice regardless of sex and age, whereas motor function appeared to be intact. Our findings establish a novel link between DA level in mDA neurons with intrinsic JMJD3 activity, and suggest prolonged chronic inflammatory pain as a major loss-of-function consequence.
Collapse
Affiliation(s)
- Xi-Biao He
- Laboratory of Stem Cell Biology and Epigenetics, School of Basic Medical Sciences, Shanghai University of Medicine & Health Sciences, 279 Zhouzhu Highway, Pudong New Area, Shanghai, 201318, China.
| | - Fang Guo
- Laboratory of Stem Cell Biology and Epigenetics, School of Basic Medical Sciences, Shanghai University of Medicine & Health Sciences, 279 Zhouzhu Highway, Pudong New Area, Shanghai, 201318, China
| | - Wei Zhang
- State Key Laboratory of Microbial Metabolism, School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jiacheng Fan
- Laboratory of Stem Cell Biology and Epigenetics, School of Basic Medical Sciences, Shanghai University of Medicine & Health Sciences, 279 Zhouzhu Highway, Pudong New Area, Shanghai, 201318, China
| | - Weidong Le
- Center for Translational Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, 201318, China
| | - Qi Chen
- Laboratory of Stem Cell Biology and Epigenetics, School of Basic Medical Sciences, Shanghai University of Medicine & Health Sciences, 279 Zhouzhu Highway, Pudong New Area, Shanghai, 201318, China
| | - Yongjun Ma
- Laboratory of Stem Cell Biology and Epigenetics, School of Basic Medical Sciences, Shanghai University of Medicine & Health Sciences, 279 Zhouzhu Highway, Pudong New Area, Shanghai, 201318, China
- The Interdisciplinary Research Center of Biology and Chemistry, Chinese Academy of sciences, Shanghai, 200120, China
| | - Yong Zheng
- Laboratory of Stem Cell Biology and Epigenetics, School of Basic Medical Sciences, Shanghai University of Medicine & Health Sciences, 279 Zhouzhu Highway, Pudong New Area, Shanghai, 201318, China
| | - Sang-Hun Lee
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Hui-Jing Wang
- Laboratory of Neuropsychopharmacology, School of Basic Medical Sciences, Shanghai University of Medicine & Health Sciences, Shanghai, 201318, China
| | - Yi Wu
- Laboratory of Stem Cell Biology and Epigenetics, School of Basic Medical Sciences, Shanghai University of Medicine & Health Sciences, 279 Zhouzhu Highway, Pudong New Area, Shanghai, 201318, China
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Qinming Zhou
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Rui Yang
- Laboratory of Stem Cell Biology and Epigenetics, School of Basic Medical Sciences, Shanghai University of Medicine & Health Sciences, 279 Zhouzhu Highway, Pudong New Area, Shanghai, 201318, China
| |
Collapse
|
8
|
Ahmed MR, Inayathullah M, Morton M, Pothineni VR, Kim K, Ahmed MS, Babar MM, Rajadas J. Intranasal delivery of liposome encapsulated flavonoids ameliorates l-DOPA induced dyskinesia in hemiparkinsonian mice. Biomaterials 2024; 311:122680. [PMID: 38959534 DOI: 10.1016/j.biomaterials.2024.122680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 04/25/2024] [Accepted: 06/23/2024] [Indexed: 07/05/2024]
Abstract
In the present study, we explored the development of a novel noninvasive liposomal drug delivery material for use in intranasal drug delivery applications in human diseases. We used drug entrapment into liposomal nanoparticle assembly to efficiently deliver the drugs to the nasal mucosa to be delivered to the brain. The naturally occurring flavonoid 7,8-dihydroxyflavone (7,8-DHF) has previously been shown to have beneficial effects in ameliorating Parkinson's disease (PD). We used both naturally occurring 7,8-DHF and the chemically modified form of DHF, the DHF-ME, to be used as a drug candidate for the treatment of PD and l-DOPA induced dyskinesia (LID), which is the debilitating side effect of l-DOPA therapy in PD. The ligand-protein interaction behavior for 7,8-DHF and 6,7-DHF-ME was found to be more effective with molecular docking and molecular stimulation studies of flavonoid compounds with TrkB receptor. Our study showed that 7,8-DHF delivered via intranasal route using a liposomal formulation ameliorated LID in hemiparkinsonian mice model when these mice were chronically administered with l-DOPA, which is the only current medication for relieving the clinical symptoms of PD. The present study also demonstrated that apart from reducing the LID, 7,8-DHF delivery directly to the brain via the intranasal route also corrected some long-term signaling adaptations involving ΔFosB and α Synuclein in the brain of dopamine (DA) depleted animals.
Collapse
Affiliation(s)
- Mohamed Rafiuddin Ahmed
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory of Cardiovascular Institute/ Pulmonary and Critical Care, Department of Medicine, Stanford University, 1050 Arastradero Road, Palo Alto, CA, 94304, USA
| | - Mohammed Inayathullah
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory of Cardiovascular Institute/ Pulmonary and Critical Care, Department of Medicine, Stanford University, 1050 Arastradero Road, Palo Alto, CA, 94304, USA
| | - Mithya Morton
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory of Cardiovascular Institute/ Pulmonary and Critical Care, Department of Medicine, Stanford University, 1050 Arastradero Road, Palo Alto, CA, 94304, USA; Children's Hospital of Orange County - UC Irvine School of Medicine, Department of Pediatrics, 505 S. Main St., Suite #525, Orange, CA, 92868, USA
| | - Venkata Raveendra Pothineni
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory of Cardiovascular Institute/ Pulmonary and Critical Care, Department of Medicine, Stanford University, 1050 Arastradero Road, Palo Alto, CA, 94304, USA
| | - Kwangmin Kim
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory of Cardiovascular Institute/ Pulmonary and Critical Care, Department of Medicine, Stanford University, 1050 Arastradero Road, Palo Alto, CA, 94304, USA; Department of Physiology, Gachon University College of Medicine, Incheon, 21999, South Korea
| | - Mohamed Sohail Ahmed
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory of Cardiovascular Institute/ Pulmonary and Critical Care, Department of Medicine, Stanford University, 1050 Arastradero Road, Palo Alto, CA, 94304, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, MCN 1161 21st Ave S. Nashville TN 37232, USA
| | - Mustafeez Mujtaba Babar
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory of Cardiovascular Institute/ Pulmonary and Critical Care, Department of Medicine, Stanford University, 1050 Arastradero Road, Palo Alto, CA, 94304, USA
| | - Jayakumar Rajadas
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory of Cardiovascular Institute/ Pulmonary and Critical Care, Department of Medicine, Stanford University, 1050 Arastradero Road, Palo Alto, CA, 94304, USA.
| |
Collapse
|
9
|
Ziroldo JC, Torres LMB, Gamberini MT. Sugarcane (Saccharum officinarum L.) induces psychostimulant, anxiolytic-like effects and improvement of motor performance in rats. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118476. [PMID: 38908491 DOI: 10.1016/j.jep.2024.118476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/03/2024] [Accepted: 06/19/2024] [Indexed: 06/24/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sugarcane (Saccharum officinarum L.) is reported by traditional medicine as tonic, stimulating and beneficial in increasing resistance to fatigue. Previous preclinical studies in rats using aqueous extract of sugarcane leaves (AE) revealed pharmacological effects on the central nervous and cardiovascular systems involving the participation of dopaminergic pathways. This neurotransmission system is also related to motor, emotional and cognitive activities, which could, in part, justify the ethnopharmacological information. AIM OF STUDY The present study aimed to investigate the motor, emotional and cognitive activities of rats submitted to AE treatment using behavioral tests in order to correlate the pharmacological effects with the therapeutic benefits postulated by traditional medicine. Additionally, the chemical profile of AE was evaluated by HPLC-UV/Vis, and the presence of shikimic acid, vitexin, and ferulic acid, as possible chemical markers, was investigated through comparisons of chemical parameters with the authentic patterns, and a UV-Vis scan of known spectra. MATERIAL AND METHODS Rats received water (1.5 mL/kg, p.o.) and AE (0.5, 10 and 500 mg/kg, p.o.) in the absence and presence of haloperidol (0.5 mg/kg, i.p.), 90 min before open field; rotarod; elevated plus maze and inhibitory avoidance tests for investigation of motor; emotional and cognitive responses. As a positive control was used apomorphine (0.25 mg/kg, s.c.). The chemical profile of AE was evaluated by HPLC-UV/Vis and the presence of shikimic acid, vitexin and ferulic acid, as possible chemical markers, was investigated through comparisons with the retention times, an increase of the integral of the peak area determined by co-injection of AE with the authentic patterns, and a UV-Vis scan of known spectra. RESULTS In open field, it revealed that AE increased locomotion; reduced rearing but did not change freezing and grooming. Besides, AE increased motor performance in rotarod and reduced anxiety in elevated plus maze. A relation dose-response was observed in these tests where the lowest dose of AE was more effective in developing pharmacological responses. Previous administration of haloperidol inhibited the responses of AE. Inhibitory avoidance test revealed that AE did not modify fast-learning and associative memory. CONCLUSIONS Sugarcane induced psychostimulant, anxiolytic-like effects, and improvement of motor performance in rats, with the involvement of dopaminergic pathways. The present study points to AE as a potential adaptogen but, in addition to behavioral assessments, metabolic and molecular aspects, that involve the participation of a variety of regulatory systems, will be investigated in futures studies. Phytochemical analyses showed that AE is a complex matrix and revealed shikimic acid, vitexin, and ferulic acid as potential chemical markers.
Collapse
Affiliation(s)
- Juliana Corsini Ziroldo
- Department of Physiological Sciences, Santa Casa de Sao Paulo School of Medical Sciences - School of Medicine, São Paulo, SP, Brazil.
| | | | - Maria Thereza Gamberini
- Department of Physiological Sciences, Santa Casa de Sao Paulo School of Medical Sciences - School of Medicine, São Paulo, SP, Brazil.
| |
Collapse
|
10
|
Ubaldo-Reyes LM, Espitia-Bautista E, Barajas-Martínez A, Martínez-Tapia R, Rodríguez-Mata V, Noriega-Navarro R, Escalona R, Castillo-Hernández J, Pérez-Torres A, Navarro L. High-Fat Diet-Induced Blood-Brain Barrier Dysfunction: Impact on Allodynia and Motor Coordination in Rats. Int J Mol Sci 2024; 25:11218. [PMID: 39457000 PMCID: PMC11508281 DOI: 10.3390/ijms252011218] [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: 09/23/2024] [Revised: 10/10/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
The associations among increased pain sensitivity, obesity, and systemic inflammation have not been described as related to BBB dysfunctions. To analyze the metabolic, behavioral, and inflammatory effects of a high-fat diet (HFD) and ultrastructural modifications in brain regions, we used an in vivo experimental model. Adult male Wistar rats were randomly assigned to one of two conditions, an ad libitum control group or an HFD (60%)-fed group, for eight weeks. At the end of the protocol, glucose and insulin tolerance tests were performed. Additionally, we analyzed the response to a normally innocuous mechanical stimulus and changes in motor coordination. At the end of the protocol, HFD-fed rats presented increased HOMA-IR and metabolic syndrome (MetS) prevalence. HFD-fed rats also developed an increased nociceptive response to mechanical stimuli and neurological injury, resulting in impaired motor function. Hypothalamus and cerebellum neurons from HFD-fed rats presented with nuclear swelling, an absence of nucleoli, and karyolysis. These results reveal that HFD consumption affects vital brain structures such as the cerebellum, hippocampus, and hypothalamus. This, in turn, could be producing neuronal damage, impairing cellular communication, and consequently altering motricity and pain sensitivity. Although direct evidence of a causal link between BBB dysfunction and sensory-motor changes was not observed, understanding the association uncovered in this study could lead to targeted therapeutic strategies.
Collapse
Affiliation(s)
- Laura M. Ubaldo-Reyes
- Department of Anatomy, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Estefania Espitia-Bautista
- Laboratory of Molecular Neurophysiology, National Institute of Psychiatry Ramón de la Fuente, Mexico City 14370, Mexico;
| | - Antonio Barajas-Martínez
- Center for Complexity Science, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | - Ricardo Martínez-Tapia
- Department of Physiology, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (R.M.-T.); (R.N.-N.); (L.N.)
| | - Verónica Rodríguez-Mata
- Department of Histology, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (V.R.-M.); (A.P.-T.)
| | - Roxana Noriega-Navarro
- Department of Physiology, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (R.M.-T.); (R.N.-N.); (L.N.)
| | - Rene Escalona
- Laboratory of Embryology and Genetics, Departamento de Embriología y Genética, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | - Jesús Castillo-Hernández
- Multidisciplinary Academic Unit Middle Zone, Autonomous University of San Luis Potosí, San Luis Potosí 79615, Mexico;
| | - Armando Pérez-Torres
- Department of Histology, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (V.R.-M.); (A.P.-T.)
| | - Luz Navarro
- Department of Physiology, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (R.M.-T.); (R.N.-N.); (L.N.)
| |
Collapse
|
11
|
Zhang X, Wang Y, Xue S, Gong L, Yan J, Zheng Y, Yang X, Fan Y, Han K, Chen Y, Yao L. Chronic stress in adulthood results in microvascular dysfunction and subsequent depressive-like behavior. Sci Rep 2024; 14:24022. [PMID: 39402273 PMCID: PMC11473760 DOI: 10.1038/s41598-024-74902-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024] Open
Abstract
Depression is a prevalent mental disorder characterized by unknown pathogenesis and challenging treatment. Recent meta-analyses reveal an association between cardiovascular risk factors and an elevated risk of depression. Despite this, the precise role of vascular injury in depression development remains unclear. In this investigation, we assess vascular system function in three established animal models of depression- chronic unpredictable mild stress (CUMS), chronic social defeat stress (CSDS) and maternal separation (MS)-utilizing ultrasonography and laser Doppler measurement. All three model animals exhibit anhedonia and despair-like behavior. However, significant microvascular dysfunction (not macrovascular) is observed in animals subjected to CUMS and CSDS models, while such dysfunction is absent in the MS model. Statistical analysis further indicates that microcirculation dysfunction is not only associated with depression-like behavior but is also intricately involved in the development of depression in the CUMS and CSDS models. Furthermore, our study has proved for the first time that endothelial nitric oxide synthase-deficient (eNOS+/-) mice, which is a classic model of vascular endothelial injury, showed depression-like behavior which occurred two months later than microvascular dysfunction. Notably, the mitigation of microvascular dysfunction successfully reverses depression-like behavior in eNOS+/- mice by enhancing nitric oxide production. In conclusion, this study unveils the pivotal role of microvascular dysfunction in the onset of depression induced by chronic stress in adulthood and proposes that modulating microvascular function may serve as a potential intervention in the treatment of depression.
Collapse
Affiliation(s)
- Xiaochen Zhang
- Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
| | - Yaru Wang
- Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
| | - Song Xue
- Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
| | - Li Gong
- Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
| | - Jinglan Yan
- Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
| | - Yuanjia Zheng
- Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
| | - Xiaoyun Yang
- Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
| | - Yujing Fan
- Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
| | - Kuizhang Han
- Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
| | - Yongjun Chen
- Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China.
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China.
| | - Lin Yao
- Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China.
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China.
| |
Collapse
|
12
|
Abdelalim LR, Elnaggar YSR, Abdallah OY. Lactoferrin, chitosan double-coated oleosomes loaded with clobetasol propionate for remyelination in multiple sclerosis: Physicochemical characterization and in-vivo assessment in a cuprizone-induced demyelination model. Int J Biol Macromol 2024; 277:134144. [PMID: 39053824 DOI: 10.1016/j.ijbiomac.2024.134144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 07/10/2024] [Accepted: 07/23/2024] [Indexed: 07/27/2024]
Abstract
Multiple sclerosis is a chronic inflammatory demyelinating disorder of the CNS characterized by continuous myelin damage accompanied by deterioration in functions. Clobetasol propionate (CP) is the most potent topical corticosteroid with serious side effects related to systemic absorption. Previous studies introduced CP for remyelination without considering systemic toxicity. This work aimed at fabrication and optimization of double coated nano-oleosomes loaded with CP to achieve brain targeting through intranasal administration. The optimized formulation was coated with lactoferrin and chitosan for the first time. The obtained double-coated oleosomes had particle size (220.07 ± 0.77 nm), zeta potential (+30.23 ± 0.41 mV) along with antioxidant capacity 9.8 μM ascorbic acid equivalents. Double coating was well visualized by TEM and significantly decreased drug release. Three different doses of CP were assessed in-vivo using cuprizone-induced demyelination in C57Bl/6 mice. Neurobehavioral tests revealed improvement in motor and cognitive functions of mice in a dose-dependent manner. Histopathological examination of the brain showed about 2.3 folds increase in corpus callosum thickness in 0.3 mg/kg CP dose. Moreover, the measured biomarkers highlighted the significant antioxidant and anti-inflammatory capacity of the formulation. In conclusion, the elaborated biopolymer-integrating nanocarrier succeeded in remyelination with 6.6 folds reduction in CP dose compared to previous studies.
Collapse
Affiliation(s)
- Lamiaa R Abdelalim
- Department of Pharmaceutics and pharmaceutical technology, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| | - Yosra S R Elnaggar
- Department of Pharmaceutics and pharmaceutical technology, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt; Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt.
| | - Ossama Y Abdallah
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
13
|
Giammello F, Biella C, Priori EC, Filippo MADS, Leone R, D'Ambrosio F, Paterno' M, Cassioli G, Minetti A, Macchi F, Spalletti C, Morella I, Ruberti C, Tremonti B, Barbieri F, Lombardi G, Brambilla R, Florio T, Galli R, Rossi P, Brandalise F. Modulating voltage-gated sodium channels to enhance differentiation and sensitize glioblastoma cells to chemotherapy. Cell Commun Signal 2024; 22:434. [PMID: 39251990 PMCID: PMC11382371 DOI: 10.1186/s12964-024-01819-z] [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: 04/05/2024] [Accepted: 09/04/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM) stands as the most prevalent and aggressive form of adult gliomas. Despite the implementation of intensive therapeutic approaches involving surgery, radiation, and chemotherapy, Glioblastoma Stem Cells contribute to tumor recurrence and poor prognosis. The induction of Glioblastoma Stem Cells differentiation by manipulating the transcriptional machinery has emerged as a promising strategy for GBM treatment. Here, we explored an innovative approach by investigating the role of the depolarized resting membrane potential (RMP) observed in patient-derived GBM sphereforming cell (GSCs), which allows them to maintain a stemness profile when they reside in the G0 phase of the cell cycle. METHODS We conducted molecular biology and electrophysiological experiments, both in vitro and in vivo, to examine the functional expression of the voltage-gated sodium channel (Nav) in GSCs, particularly focusing on its cell cycle-dependent functional expression. Nav activity was pharmacologically manipulated, and its effects on GSCs behavior were assessed by live imaging cell cycle analysis, self-renewal assays, and chemosensitivity assays. Mechanistic insights into the role of Nav in regulating GBM stemness were investigated through pathway analysis in vitro and through tumor proliferation assay in vivo. RESULTS We demonstrated that Nav is functionally expressed by GSCs mainly during the G0 phase of the cell cycle, suggesting its pivotal role in modulating the RMP. The pharmacological blockade of Nav made GBM cells more susceptible to temozolomide (TMZ), a standard drug for this type of tumor, by inducing cell cycle re-entry from G0 phase to G1/S transition. Additionally, inhibition of Nav substantially influenced the self-renewal and multipotency features of GSCs, concomitantly enhancing their degree of differentiation. Finally, our data suggested that Nav positively regulates GBM stemness by depolarizing the RMP and suppressing the ERK signaling pathway. Of note, in vivo proliferation assessment confirmed the increased susceptibility to TMZ following pharmacological blockade of Nav. CONCLUSIONS This insight positions Nav as a promising prognostic biomarker and therapeutic target for GBM patients, particularly in conjunction with temozolomide treatment.
Collapse
Affiliation(s)
- Francesca Giammello
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, 27100, Italy
- PhD Program in Genetics, Molecular and Cellular Biology, University of Pavia, Pavia, Italy
| | - Chiara Biella
- IRCCS San Raffaele Hospital, Via Olgettina 58, Milan, 20132, Italy
| | - Erica Cecilia Priori
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, 27100, Italy
| | | | - Roberta Leone
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, 27100, Italy
| | | | - Martina Paterno'
- Department of Biosciences, University of Milan, Milan, 20133, Italy
| | - Giulia Cassioli
- Department of Biosciences, University of Milan, Milan, 20133, Italy
| | - Antea Minetti
- CNR Neuroscience Institute of Pisa, Via Giuseppe Moruzzi, 1, Pisa (PI), 56124, Italy
| | - Francesca Macchi
- CNR Neuroscience Institute of Pisa, Via Giuseppe Moruzzi, 1, Pisa (PI), 56124, Italy
| | - Cristina Spalletti
- CNR Neuroscience Institute of Pisa, Via Giuseppe Moruzzi, 1, Pisa (PI), 56124, Italy
| | - Ilaria Morella
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, 27100, Italy
| | - Cristina Ruberti
- Advanced Technology Platform, Department of Biosciences, University of Milan, Milan, 20133, Italy
| | - Beatrice Tremonti
- Pharmacology Unit, Department of Internal Medicine, University of Genova, Genova, 16132, Italy
| | - Federica Barbieri
- Pharmacology Unit, Department of Internal Medicine, University of Genova, Genova, 16132, Italy
| | - Giuseppe Lombardi
- Department of Oncology 1, Oncology, Veneto Institute of Oncology IOV-IRCCS, via Gattamelata 64, Padua, 35128, Italy
| | - Riccardo Brambilla
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, 27100, Italy
| | - Tullio Florio
- Pharmacology Unit, Department of Internal Medicine, University of Genova, Genova, 16132, Italy
- IRCCS Ospedale Policlinico San Martino, Genova, 16132, Italy
| | - Rossella Galli
- IRCCS San Raffaele Hospital, Via Olgettina 58, Milan, 20132, Italy
| | - Paola Rossi
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, 27100, Italy
| | | |
Collapse
|
14
|
Cavestro C, D'Amato M, Colombo MN, Cascone F, Moro AS, Levi S, Tiranti V, Di Meo I. CoA synthase plays a critical role in neurodevelopment and neurodegeneration. Front Cell Neurosci 2024; 18:1458475. [PMID: 39301217 PMCID: PMC11410578 DOI: 10.3389/fncel.2024.1458475] [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: 07/02/2024] [Accepted: 08/27/2024] [Indexed: 09/22/2024] Open
Abstract
Coenzyme A (CoA), which is widely distributed and vital for cellular metabolism, is a critical molecule essential in both synthesizing and breaking down key energy sources in the body. Inborn errors of metabolism in the cellular de novo biosynthetic pathway of CoA have been linked to human genetic disorders, emphasizing the importance of this pathway. The COASY gene encodes the bifunctional enzyme CoA synthase, which catalyzes the last two reactions of the CoA biosynthetic pathway and serves as one of the rate-limiting components of the pathway. Recessive variants of this gene cause an exceptionally rare and devastating disease called COASY protein-associated neurodegeneration (CoPAN) while complete loss-of-function variants in COASY have been identified in fetuses/neonates with Pontocerebellar Hypoplasia type 12 (PCH 12). Understanding why the different symptoms emerge in these disorders and what determines the development of one syndrome over the other is still not achieved. To shed light on the pathogenesis, we generated a new conditional animal model in which Coasy was deleted under the control of the human GFAP promoter. We used this mouse model to investigate how defects in the CoA biosynthetic pathway affect brain development. This model showed a broad spectrum of severity of the in vivo phenotype, ranging from very short survival (less than 2 weeks) to normal life expectancy in some animals. Surviving mice displayed a behavioral phenotype with sensorimotor defects. Ex vivo histological analysis revealed variable but consistent cerebral and cerebellar cortical hypoplasia, in parallel with a broad astrocytic hyper-proliferation in the cerebral cortex. In addition, primary astrocytes derived from this model exhibited lipid peroxidation, iron dyshomeostasis, and impaired mitochondrial respiration. Notably, Coasy ablation in radial glia and astrocytic lineage triggers abnormal neuronal development and chronic neuroinflammation, offering new insights into disease mechanisms.
Collapse
Affiliation(s)
- Chiara Cavestro
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Marco D'Amato
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Maria Nicol Colombo
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Floriana Cascone
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | | | - Sonia Levi
- Vita-Salute San Raffaele University, Milan, Italy
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Valeria Tiranti
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Ivano Di Meo
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|
15
|
Djabirska I, Delaval L, Tromme A, Blomet J, Desmecht D, Van Laere AS. Longitudinal quantitative assessment of TMEV-IDD-induced MS phenotypes in two inbred mouse strains using automated video tracking technology. Exp Neurol 2024; 379:114851. [PMID: 38876197 DOI: 10.1016/j.expneurol.2024.114851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/29/2024] [Accepted: 06/06/2024] [Indexed: 06/16/2024]
Abstract
Multiple sclerosis (MS) is a chronic disabling disease of the central nervous system affecting over 2.5 million people worldwide. Theiler's murine encephalomyelitis virus-induced demyelinating disease (TMEV-IDD) is a murine model that reproduces the progressive form of MS and serves as a reference model for studying virus-induced demyelination. Certain mouse strains such as SJL are highly susceptible to this virus and serve as a prototype strain for studying TMEV infection. Other strains such as SWR are also susceptible, but their disease course following TMEV infection differs from SJL's. The quantification of motor and behavioral deficits following the induction of TMEV-IDD could help identify the differences between the two strains. Motor deficits have commonly been measured with the rotarod apparatus, but a multicomponent assessment tool has so far been lacking. For that purpose, we present a novel way of quantifying locomotor deficits, gait alterations and behavioral changes in this well-established mouse model of multiple sclerosis by employing automated video analysis technology (The PhenoTyper, Noldus Information Technology). We followed 12 SJL and 12 SWR female mice and their mock-infected counterparts over a period of 9 months following TMEV-IDD induction. We demonstrated that SJL and SWR mice both suffer significant gait alterations and reduced exploration following TMEV infection. However, SJL mice also display an earlier and more severe decline in spontaneous locomotion, especially in velocity, as well as in overall activity. Maintenance behaviors such as eating and grooming are not affected in either of the two strains. The system also showed differences in mock-infected mice from both strains, highlighting an age-related decline in spontaneous locomotion in the SJL strain, as opposed to hyperactivity in the SWR strain. Our study confirms that this automated video tracking system can reliably track the progression of TMEV-IDD for 9 months. We have also shown how this system can be utilized for longitudinal phenotyping in mice by describing useful parameters that quantify locomotion, gait and behavior.
Collapse
Affiliation(s)
- Iskra Djabirska
- Department of Pathology, Fundamental and Applied Research for Animals & Health (FARAH), Faculty of Veterinary Medicine, University of Liege, Liège 4000, Belgium; Prevor Research Laboratories, Valmondois 95760, France
| | - Laetitia Delaval
- Department of Pathology, Fundamental and Applied Research for Animals & Health (FARAH), Faculty of Veterinary Medicine, University of Liege, Liège 4000, Belgium; Prevor Research Laboratories, Valmondois 95760, France
| | - Audrey Tromme
- Department of Pathology, Fundamental and Applied Research for Animals & Health (FARAH), Faculty of Veterinary Medicine, University of Liege, Liège 4000, Belgium; Prevor Research Laboratories, Valmondois 95760, France
| | - Joël Blomet
- Prevor Research Laboratories, Valmondois 95760, France
| | - Daniel Desmecht
- Department of Pathology, Fundamental and Applied Research for Animals & Health (FARAH), Faculty of Veterinary Medicine, University of Liege, Liège 4000, Belgium
| | - Anne-Sophie Van Laere
- Department of Pathology, Fundamental and Applied Research for Animals & Health (FARAH), Faculty of Veterinary Medicine, University of Liege, Liège 4000, Belgium; Prevor Research Laboratories, Valmondois 95760, France.
| |
Collapse
|
16
|
Techaniyom P, Korsirikoon C, Rungruang T, Pakaprot N, Prombutara P, Mukda S, Kettawan AK, Kettawan A. Cold-pressed perilla seed oil: Investigating its protective influence on the gut-brain axis in mice with rotenone-induced Parkinson's disease. Food Sci Nutr 2024; 12:6259-6283. [PMID: 39554352 PMCID: PMC11561828 DOI: 10.1002/fsn3.4265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/24/2024] [Accepted: 05/28/2024] [Indexed: 11/19/2024] Open
Abstract
Perilla seed oil, derived from a regional plant native to northern Thailand, undergoes cold-pressing to analyze its bioactive components, notably alpha-linolenic acid (ALA). ALA, constituting approximately 61% of the oil, serves as a precursor for therapeutic omega-3 fatty acids, EPA and DHA, with neurodegenerative disease benefits and anti-inflammatory responses. This study administered different concentrations of perilla seed oil to male C57BL/6 mice, categorized as low dose (LP 5% w/w), middle dose (MP 10% w/w), and high dose (HP 20% w/w), along with a fish oil (FP 10% w/w) diet. An experimental group received soybean oil (5% w/w). Over 42 days, these diets were administered while inducing Parkinson's disease (PD) with rotenone injections. Mice on a high perilla seed oil dose exhibited decreased Cox-2 expression in the colon, suppressed Iba-1 microglia activation, reduced alpha-synuclein accumulation in the colon and hippocampus, prevented dopaminergic cell death in the substantia nigra, and improved motor and non-motor symptoms. Mice on a middle dose showed maintenance of diverse gut microbiota, with an increased abundance of short-chain fatty acid (SCFA)-producing bacteria (Bifidobacteria, Lactobacillus, and Faecalibacteria). A reduction in bacteria correlated with PD (Turicibacter, Ruminococcus, and Akkermansia) was observed. Results suggest the potential therapeutic efficacy of high perilla seed oil doses in mitigating both intestinal and neurological aspects linked to the gut-brain axis in PD.
Collapse
Affiliation(s)
- Peerapa Techaniyom
- Doctor of Philosophy Program in Nutrition, Faculty of Medicine Ramathibodi Hospital and Institute of NutritionMahidol UniversityBangkokThailand
| | - Chawin Korsirikoon
- Doctor of Philosophy Program in Nutrition, Faculty of Medicine Ramathibodi Hospital and Institute of NutritionMahidol UniversityBangkokThailand
| | - Thanaporn Rungruang
- Department of Anatomy, Faculty of Medicine Siriraj HospitalMahidol UniversityBangkokThailand
| | - Narawut Pakaprot
- Department of Physiology, Faculty of Medicine Siriraj HospitalMahidol UniversityBangkokThailand
| | - Pinidphon Prombutara
- OMICS Sciences and Bioinformatics Center, Faculty of ScienceChulalongkorn UniversityBangkokThailand
- Mod Gut Co., Ltd.BangkokThailand
| | - Sujira Mukda
- Research Center for NeuroscienceInstitute of Molecular Biosciences, Mahidol UniversityNakhon PathomThailand
| | | | | |
Collapse
|
17
|
Altés G, Olomí A, Perramon-Güell A, Hernández S, Casanovas A, Pérez A, Díaz-Tocados JM, Valdivielso JM, Megino C, Navaridas R, Matias-Guiu X, Klein OD, Egea J, Dolcet X, Yeramian A, Encinas M. Multiple endocrine defects in adult-onset Sprouty1/2/4 triple knockout mice. Sci Rep 2024; 14:19479. [PMID: 39174793 PMCID: PMC11341818 DOI: 10.1038/s41598-024-70529-w] [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: 05/29/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024] Open
Abstract
Genes of the Sprouty family (Spry1-4) are feedback inhibitors of receptor tyrosine kinases, especially of Ret and the FGF receptors. As such, they play distinct and overlapping roles in embryo morphogenesis and are considered to be tumor suppressors in adult life. Genetic experiments in mice have defined in great detail the role of these genes during embryonic development, however their function in adult mice is less clearly established. Here we generate adult-onset, whole body Spry1/2/4 triple knockout mice. Tumor incidence in triple mutant mice is comparable to that of wild type littermates of up to one year of age, indicating that Sprouty loss per se is not sufficient to initiate tumorigenesis. On the other hand, triple knockout mice do not gain weight as they age, show less visceral fat, and have lower plasma glucose levels than wild type littermates, despite showing similar food intake and slightly reduced motor function. They also show alopecia, eyelid inflammation, and mild hyperthyroidism. Finally, triple knockout mice present phosphaturia and hypophosphatemia, suggesting exacerbated signaling downstream of FGF23. In conclusion, triple knockout mice develop a series of endocrine abnormalities but do not show increased tumor incidence.
Collapse
Affiliation(s)
- Gisela Altés
- Developmental and Oncogenic Signaling Group, Edifici Biomedicina I, Lab 2.8, Department of Experimental Medicine, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Rovira Roure, 80, 25198, Lleida, Spain
| | - Anna Olomí
- Developmental and Oncogenic Signaling Group, Edifici Biomedicina I, Lab 2.8, Department of Experimental Medicine, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Rovira Roure, 80, 25198, Lleida, Spain
| | - Aida Perramon-Güell
- Developmental and Oncogenic Signaling Group, Edifici Biomedicina I, Lab 2.8, Department of Experimental Medicine, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Rovira Roure, 80, 25198, Lleida, Spain
| | - Sara Hernández
- Experimental Neuromuscular Pathology Group, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Lleida, Spain
| | - Anna Casanovas
- Experimental Neuromuscular Pathology Group, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Lleida, Spain
| | - Aurora Pérez
- Vascular and Renal Translational Research Group, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Lleida, Spain
| | - Juan Miguel Díaz-Tocados
- Vascular and Renal Translational Research Group, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Lleida, Spain
| | - José Manuel Valdivielso
- Vascular and Renal Translational Research Group, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Lleida, Spain
| | - Cristina Megino
- Oncologic Pathology Group, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Lleida, Spain
| | - Raúl Navaridas
- Oncologic Pathology Group, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Lleida, Spain
| | - Xavier Matias-Guiu
- Oncologic Pathology Group, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Lleida, Spain
- Department of Pathology, Hospital Universitari de Bellvitge, Barcelona, Spain
| | - Ophir D Klein
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Pediatrics, Cedars-Sinai Guerin Children's, Los Angeles, CA, USA
| | - Joaquim Egea
- Developmental and Oncogenic Signaling Group, Edifici Biomedicina I, Lab 2.8, Department of Experimental Medicine, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Rovira Roure, 80, 25198, Lleida, Spain
| | - Xavi Dolcet
- Developmental and Oncogenic Signaling Group, Edifici Biomedicina I, Lab 2.8, Department of Experimental Medicine, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Rovira Roure, 80, 25198, Lleida, Spain
| | - Andrée Yeramian
- Developmental and Oncogenic Signaling Group, Edifici Biomedicina I, Lab 2.8, Department of Experimental Medicine, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Rovira Roure, 80, 25198, Lleida, Spain
| | - Mario Encinas
- Developmental and Oncogenic Signaling Group, Edifici Biomedicina I, Lab 2.8, Department of Experimental Medicine, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Rovira Roure, 80, 25198, Lleida, Spain.
| |
Collapse
|
18
|
Zhu J, Qiu W, Wei F, Zhang J, Yuan Y, Liu L, Cheng M, Xiong H, Xu R. Toll-like receptor 4 deficiency in Purkinje neurons drives cerebellar ataxia by impairing the BK channel-mediated after-hyperpolarization and cytosolic calcium homeostasis. Cell Death Dis 2024; 15:594. [PMID: 39147737 PMCID: PMC11327311 DOI: 10.1038/s41419-024-06988-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 08/17/2024]
Abstract
Toll-like receptor (TLR) 4 contributes to be the induction of neuroinflammation by recognizing pathology-associated ligands and activating microglia. In addition, numerous physiological signaling factors act as agonists or antagonists of TLR4 expressed by non-immune cells. Recently, TLR4 was found to be highly expressed in cerebellar Purkinje neurons (PNs) and involved in the maintenance of motor coordination through non-immune pathways, but the precise mechanisms remain unclear. Here we report that mice with PN specific TLR4 deletion (TLR4PKO mice) exhibited motor impairments consistent with cerebellar ataxia, reduced PN dendritic arborization and spine density, fewer parallel fiber (PF) - PN and climbing fiber (CF) - PN synapses, reduced BK channel expression, and impaired BK-mediated after-hyperpolarization, collectively leading to abnormal PN firing. Moreover, the impaired PN firing in TLR4PKO mice could be rescued with BK channel opener. The PNs of TLR4PKO mice also exhibited abnormal mitochondrial structure, disrupted mitochondrial endoplasmic reticulum tethering, and reduced cytosolic calcium, changes that may underly abnormal PN firing and ultimately drive ataxia. These results identify a previously unknown role for TLR4 in regulating PN firing and maintaining cerebellar function.
Collapse
Affiliation(s)
- Jianwei Zhu
- Department of Neurosurgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Wenqiao Qiu
- Department of Neurosurgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Fan Wei
- Department of Neurosurgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
- Department of Critical Care Medicine, Mianyang Orthopaedic Hospital, Mianyang, Sichuan Province, 621000, China
| | - Jin Zhang
- Department of Neurosurgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Ying Yuan
- Department of Neurosurgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Ling Liu
- Department of Neurosurgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Meixiong Cheng
- Department of Neurosurgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Huan Xiong
- Department of Neurosurgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Ruxiang Xu
- Department of Neurosurgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
19
|
Shah S, Pushpa Tryphena K, Singh G, Kulkarni A, Pinjala P, Kumar Khatri D. Neuroprotective role of Carvacrol via Nrf2/HO-1/NLRP3 axis in Rotenone-induced PD mice model. Brain Res 2024; 1836:148954. [PMID: 38649135 DOI: 10.1016/j.brainres.2024.148954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 04/14/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024]
Abstract
Parkinson's disease (PD) is a multifactorial neurodegenerative disorder whose cause is unclear. Neuroinflammation is recognized as one of the major pathogenic mechanisms involved in the development and progression of PD. NLRP3 inflammasome is the most widely studied inflammatory mediator in various diseases including PD. Several phytoconstituents have shown neuroprotective role in PD. Carvacrol is a phenolic monoterpene commonly found in the essential oils derived from plants belonging to Lamiaceae family. It is well known for its anti-inflammatory and antioxidant properties and has been widely explored in several diseases. In this study, we explored the role of Carvacrol in suppressing neuroinflammation by regulating NLRP3 inflammasome through Nrf2/HO-1 axis and subsequently, inflammatory cytokines like IL-1β, IL-18 in Rotenone induced PD mice model. Three doses (25 mg/kg, 50 mg/kg, 100 mg/kg p.o.) of Carvacrol were administered to, respectively, three groups (LD, MD, HD), one hour after administration of Rotenone (1.5 mg/kg, i.p.), every day, for 21 days. Treatment with Carvacrol ameliorated the motor impairment caused by Rotenone. It alleviated neurotoxicity and reduced inflammatory cytokines. Further, Carvacrol also alleviated oxidative stress and increased antioxidant enzymes. From these results, we show that Carvacrol exerts neuroprotective effects in PD via anti-inflammatory and antioxidant mechanisms and could be a potential therapeutic option in PD.
Collapse
Affiliation(s)
- Shruti Shah
- Molecular & Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Kamatham Pushpa Tryphena
- Molecular & Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Gurpreet Singh
- Molecular & Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Amrita Kulkarni
- Molecular & Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Poojitha Pinjala
- Molecular & Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Dharmendra Kumar Khatri
- Molecular & Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India.
| |
Collapse
|
20
|
Ren SY, Xia Y, Yu B, Lei QJ, Hou PF, Guo S, Wu SL, Liu W, Yang SF, Jiang YB, Chen JF, Shen KF, Zhang CQ, Wang F, Yan M, Ren H, Yang N, Zhang J, Zhang K, Lin S, Li T, Yang QW, Xiao L, Hu ZX, Mei F. Growth hormone promotes myelin repair after chronic hypoxia via triggering pericyte-dependent angiogenesis. Neuron 2024; 112:2177-2196.e6. [PMID: 38653248 DOI: 10.1016/j.neuron.2024.03.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 01/26/2024] [Accepted: 03/27/2024] [Indexed: 04/25/2024]
Abstract
White matter injury (WMI) causes oligodendrocyte precursor cell (OPC) differentiation arrest and functional deficits, with no effective therapies to date. Here, we report increased expression of growth hormone (GH) in the hypoxic neonatal mouse brain, a model of WMI. GH treatment during or post hypoxic exposure rescues hypoxia-induced hypomyelination and promotes functional recovery in adolescent mice. Single-cell sequencing reveals that Ghr mRNA expression is highly enriched in vascular cells. Cell-lineage labeling and tracing identify the GHR-expressing vascular cells as a subpopulation of pericytes. These cells display tip-cell-like morphology with kinetic polarized filopodia revealed by two-photon live imaging and seemingly direct blood vessel branching and bridging. Gain-of-function and loss-of-function experiments indicate that GHR signaling in pericytes is sufficient to modulate angiogenesis in neonatal brains, which enhances OPC differentiation and myelination indirectly. These findings demonstrate that targeting GHR and/or downstream effectors may represent a promising therapeutic strategy for WMI.
Collapse
Affiliation(s)
- Shu-Yu Ren
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yu Xia
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Bin Yu
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China; Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Qi-Jing Lei
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Peng-Fei Hou
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Sheng Guo
- Department of Immunology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Shuang-Ling Wu
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Wei Liu
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Shao-Fan Yang
- Brain Research Center, State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yi-Bin Jiang
- Department of Neurobiology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jing-Fei Chen
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Kai-Feng Shen
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Chun-Qing Zhang
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Fei Wang
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Mi Yan
- Department of Pediatrics, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing 400000, China
| | - Hong Ren
- Department of Emergence, 5(th) People's Hospital of Chongqing, Chongqing 400062, China
| | - Nian Yang
- Department of Physiology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jun Zhang
- Department of Neurobiology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Kuan Zhang
- Brain Research Center, State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Sen Lin
- Department of Neurology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Tao Li
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Qing-Wu Yang
- Department of Neurology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Lan Xiao
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Zhang-Xue Hu
- Department of Pediatrics, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing 400000, China.
| | - Feng Mei
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| |
Collapse
|
21
|
Linen SR, Chang NH, Hess EJ, Stanley GB, Waiblinger C. Sensory-Behavioral Deficits in Parkinson's Disease: Insights from a 6-OHDA Mouse Model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.05.597339. [PMID: 38895263 PMCID: PMC11185599 DOI: 10.1101/2024.06.05.597339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Parkinson's disease (PD) is characterized by the degeneration of dopaminergic neurons in the striatum, predominantly associated with motor symptoms. However, non-motor deficits, particularly sensory symptoms, often precede motor manifestations, offering a potential early diagnostic window. The impact of non-motor deficits on sensation behavior and the underlying mechanisms remains poorly understood. In this study, we examined changes in tactile sensation within a Parkinsonian state by employing a mouse model of PD induced by 6-hydroxydopamine (6-OHDA) to deplete striatal dopamine (DA). Leveraging the conserved mouse whisker system as a model for tactile-sensory stimulation, we conducted psychophysical experiments to assess sensory-driven behavioral performance during a tactile detection task in both the healthy and Parkinson-like states. Our findings reveal that DA depletion induces pronounced alterations in tactile sensation behavior, extending beyond expected motor impairments. We observed diverse behavioral deficits, spanning detection performance, task engagement, and reward accumulation, among lesioned individuals. While subjects with extreme DA depletion consistently showed severe sensory behavioral deficits, others with substantial DA depletion displayed minimal changes in sensory behavior performance. Moreover, some exhibited moderate degradation of behavioral performance, likely stemming from sensory signaling loss rather than motor impairment. The implementation of a sensory detection task is a promising approach to quantify the extent of impairments associated with DA depletion in the animal model. This facilitates the exploration of early non-motor deficits in PD, emphasizing the importance of incorporating sensory assessments in understanding the diverse spectrum of PD symptoms.
Collapse
Affiliation(s)
- Savannah R. Linen
- Program in Bioinformatics, Georgia Institute of Technology, Atlanta, GA, USA
| | - Nelson H. Chang
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Ellen J. Hess
- Departments of Pharmacology and Chemical Biology and Neurology, Emory University, Atlanta, GA USA
| | - Garrett B. Stanley
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Christian Waiblinger
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| |
Collapse
|
22
|
Droppelmann CA, Campos-Melo D, Noches V, McLellan C, Szabla R, Lyons TA, Amzil H, Withers B, Kaplanis B, Sonkar KS, Simon A, Buratti E, Junop M, Kramer JM, Strong MJ. Mitigation of TDP-43 toxic phenotype by an RGNEF fragment in amyotrophic lateral sclerosis models. Brain 2024; 147:2053-2068. [PMID: 38739752 PMCID: PMC11146434 DOI: 10.1093/brain/awae078] [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: 08/30/2023] [Revised: 01/24/2024] [Accepted: 02/08/2024] [Indexed: 05/16/2024] Open
Abstract
Aggregation of the RNA-binding protein TAR DNA binding protein (TDP-43) is a hallmark of TDP-proteinopathies including amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). As TDP-43 aggregation and dysregulation are causative of neuronal death, there is a special interest in targeting this protein as a therapeutic approach. Previously, we found that TDP-43 extensively co-aggregated with the dual function protein GEF (guanine exchange factor) and RNA-binding protein rho guanine nucleotide exchange factor (RGNEF) in ALS patients. Here, we show that an N-terminal fragment of RGNEF (NF242) interacts directly with the RNA recognition motifs of TDP-43 competing with RNA and that the IPT/TIG domain of NF242 is essential for this interaction. Genetic expression of NF242 in a fruit fly ALS model overexpressing TDP-43 suppressed the neuropathological phenotype increasing lifespan, abolishing motor defects and preventing neurodegeneration. Intracerebroventricular injections of AAV9/NF242 in a severe TDP-43 murine model (rNLS8) improved lifespan and motor phenotype, and decreased neuroinflammation markers. Our results demonstrate an innovative way to target TDP-43 proteinopathies using a protein fragment with a strong affinity for TDP-43 aggregates and a mechanism that includes competition with RNA sequestration, suggesting a promising therapeutic strategy for TDP-43 proteinopathies such as ALS and FTD.
Collapse
Affiliation(s)
- Cristian A Droppelmann
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A 5C1, Canada
| | - Danae Campos-Melo
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A 5C1, Canada
| | - Veronica Noches
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A 5C1, Canada
| | - Crystal McLellan
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A 5C1, Canada
| | - Robert Szabla
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A 5C1, Canada
| | - Taylor A Lyons
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A 5C1, Canada
| | - Hind Amzil
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A 5C1, Canada
| | - Benjamin Withers
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A 5C1, Canada
| | - Brianna Kaplanis
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A 5C1, Canada
| | - Kirti S Sonkar
- International Centre for Genetic Engineering and Biotechnology (ICGEB), AREA Science Park, 34149 Trieste, Italy
| | - Anne Simon
- Department of Biology, Faculty of Science, Western University, London, Ontario N6A 5B7, Canada
| | - Emanuele Buratti
- International Centre for Genetic Engineering and Biotechnology (ICGEB), AREA Science Park, 34149 Trieste, Italy
| | - Murray Junop
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A 5C1, Canada
| | - Jamie M Kramer
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Michael J Strong
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A 5C1, Canada
- Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A 5C1, Canada
| |
Collapse
|
23
|
O'Leary TP, Brown RE. Age-related changes in species-typical behaviours in the 5xFAD mouse model of Alzheimer's disease from 4 to 16 months of age. Behav Brain Res 2024; 465:114970. [PMID: 38531510 DOI: 10.1016/j.bbr.2024.114970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/12/2024] [Accepted: 03/23/2024] [Indexed: 03/28/2024]
Abstract
Alzheimer's disease (AD) patients show age-related decreases in the ability to perform activities of daily living and the decline in these activities is related to the severity of neurobiological deterioration underlying the disease. The 5xFAD mouse model of AD shows age-related impairments in sensory- motor and cognitive function, but little is known about changes in species-typical behaviours that may model activities of daily living in AD patients. Therefore, we examined species-typical behaviours used as indices of exploration (rearing) and compulsivity (grooming) across six tests of anxiety-like behaviour or motor function in female 5xFAD mice from 3 to 16 months of age. Robust decreases in rearing were found in 5xFAD mice across all tests after 9 months of age, although few differences were observed in grooming. A fine-scale analysis of grooming, however, revealed a previously unresolved and spatially restricted pattern of grooming in 5xFAD mice at 13-16 months of age. We then examined changes in species-typical behaviours in the home-cage, and show impaired nest building in 5xFAD mice at all ages tested. Lastly, we examined the relationship between reduced species typical behaviours in 5xFAD mice and the presentation of freezing behaviour, a commonly used measure of memory for conditioned fear. These results showed that along with cognitive and sensory-motor behaviour, 5xFAD mice have robust age-related impairments in species-typical behaviours. Therefore, species typical behaviours in 5xFAD mice may help to model the decline in activities of daily living observed in AD patients, and may provide useful behavioural phenotypes for evaluating the pre-clinical efficacy of novel therapeutics for AD.
Collapse
Affiliation(s)
- Timothy P O'Leary
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Richard E Brown
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS B3H 4R2, Canada.
| |
Collapse
|
24
|
Buvoli M, Wilson GC, Buvoli A, Gugel JF, Hau A, Bönnemann CG, Paradas C, Ryba DM, Woulfe KC, Walker LA, Buvoli T, Ochala J, Leinwand LA. A Laing distal myopathy-associated proline substitution in the β-myosin rod perturbs myosin cross-bridging activity. J Clin Invest 2024; 134:e172599. [PMID: 38690726 PMCID: PMC11060730 DOI: 10.1172/jci172599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 03/11/2024] [Indexed: 05/03/2024] Open
Abstract
Proline substitutions within the coiled-coil rod region of the β-myosin gene (MYH7) are the predominant mutations causing Laing distal myopathy (MPD1), an autosomal dominant disorder characterized by progressive weakness of distal/proximal muscles. We report that the MDP1 mutation R1500P, studied in what we believe to be the first mouse model for the disease, adversely affected myosin motor activity despite being in the structural rod domain that directs thick filament assembly. Contractility experiments carried out on isolated mutant muscles, myofibrils, and myofibers identified muscle fatigue and weakness phenotypes, an increased rate of actin-myosin detachment, and a conformational shift of the myosin heads toward the more reactive disordered relaxed (DRX) state, causing hypercontractility and greater ATP consumption. Similarly, molecular analysis of muscle biopsies from patients with MPD1 revealed a significant increase in sarcomeric DRX content, as observed in a subset of myosin motor domain mutations causing hypertrophic cardiomyopathy. Finally, oral administration of MYK-581, a small molecule that decreases the population of heads in the DRX configuration, significantly improved the limited running capacity of the R1500P-transgenic mice and corrected the increased DRX state of the myofibrils from patients. These studies provide evidence of the molecular pathogenesis of proline rod mutations and lay the groundwork for the therapeutic advancement of myosin modulators.
Collapse
Affiliation(s)
- Massimo Buvoli
- Department of Molecular, Cellular and Developmental Biology, and
- BioFrontiers Institute, Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, USA
| | - Genevieve C.K. Wilson
- Department of Molecular, Cellular and Developmental Biology, and
- BioFrontiers Institute, Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, USA
| | - Ada Buvoli
- Department of Molecular, Cellular and Developmental Biology, and
- BioFrontiers Institute, Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, USA
| | - Jack F. Gugel
- Department of Molecular, Cellular and Developmental Biology, and
- BioFrontiers Institute, Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, USA
| | - Abbi Hau
- Centre of Human and Applied Physiological Sciences, School of Basic and Medical Biosciences, Faculty of Life Sciences and Medicine, and
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, Faculty of Life Sciences and Medicine, Guy’s Campus, King’s College London, London, United Kingdom
| | - Carsten G. Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke (NINDS), NIH, Bethesda, Maryland, USA
| | - Carmen Paradas
- Neuromuscular Unit, Department of Neurology, Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | | | - Kathleen C. Woulfe
- Division of Cardiology, Department of Medicine, University of Colorado, Denver, Colorado, USA
| | - Lori A. Walker
- Division of Cardiology, Department of Medicine, University of Colorado, Denver, Colorado, USA
| | - Tommaso Buvoli
- Department of Mathematics, Tulane University, New Orleans, Louisiana, USA
| | - Julien Ochala
- Centre of Human and Applied Physiological Sciences, School of Basic and Medical Biosciences, Faculty of Life Sciences and Medicine, and
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, Faculty of Life Sciences and Medicine, Guy’s Campus, King’s College London, London, United Kingdom
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Leslie A. Leinwand
- Department of Molecular, Cellular and Developmental Biology, and
- BioFrontiers Institute, Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, USA
| |
Collapse
|
25
|
Cheng YJ, Wang F, Feng J, Yu B, Wang B, Gao Q, Wang TY, Hu B, Gao X, Chen JF, Chen YJ, Lv SQ, Feng H, Xiao L, Mei F. Prolonged myelin deficits contribute to neuron loss and functional impairments after ischaemic stroke. Brain 2024; 147:1294-1311. [PMID: 38289861 DOI: 10.1093/brain/awae029] [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: 08/06/2023] [Revised: 12/29/2023] [Accepted: 01/13/2024] [Indexed: 02/01/2024] Open
Abstract
Ischaemic stroke causes neuron loss and long-term functional deficits. Unfortunately, effective approaches to preserving neurons and promoting functional recovery remain unavailable. Oligodendrocytes, the myelinating cells in the CNS, are susceptible to oxygen and nutrition deprivation and undergo degeneration after ischaemic stroke. Technically, new oligodendrocytes and myelin can be generated by the differentiation of oligodendrocyte precursor cells (OPCs). However, myelin dynamics and their functional significance after ischaemic stroke remain poorly understood. Here, we report numerous denuded axons accompanied by decreased neuron density in sections from ischaemic stroke lesions in human brain, suggesting that neuron loss correlates with myelin deficits in these lesions. To investigate the longitudinal changes in myelin dynamics after stroke, we labelled and traced pre-existing and newly-formed myelin, respectively, using cell-specific genetic approaches. Our results indicated massive oligodendrocyte death and myelin loss 2 weeks after stroke in the transient middle cerebral artery occlusion (tMCAO) mouse model. In contrast, myelin regeneration remained insufficient 4 and 8 weeks post-stroke. Notably, neuronal loss and functional impairments worsened in aged brains, and new myelin generation was diminished. To analyse the causal relationship between remyelination and neuron survival, we manipulated myelinogenesis by conditional deletion of Olig2 (a positive regulator) or muscarinic receptor 1 (M1R, a negative regulator) in OPCs. Deleting Olig2 inhibited remyelination, reducing neuron survival and functional recovery after tMCAO. Conversely, enhancing remyelination by M1R conditional knockout or treatment with the pro-myelination drug clemastine after tMCAO preserved white matter integrity and neuronal survival, accelerating functional recovery. Together, our findings demonstrate that enhancing myelinogenesis is a promising strategy to preserve neurons and promote functional recovery after ischaemic stroke.
Collapse
Affiliation(s)
- Yong-Jie Cheng
- Department of Neurosurgery and Key Laboratory of Neurotrauma, 1st affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Fei Wang
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jie Feng
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Bin Yu
- Department of Neurosurgery, 2nd affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Bin Wang
- Department of Physiology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China
| | - Qing Gao
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, School of Mathematical Sciences, University of Electronic Science and Technology of China, Chengdu, 611731, PR China
| | - Teng-Yue Wang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, School of Mathematical Sciences, University of Electronic Science and Technology of China, Chengdu, 611731, PR China
| | - Bo Hu
- Department of Physiology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China
| | - Xing Gao
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jing-Fei Chen
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yu-Jie Chen
- Department of Neurosurgery and Key Laboratory of Neurotrauma, 1st affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Sheng-Qing Lv
- Department of Neurosurgery, 2nd affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Hua Feng
- Department of Neurosurgery and Key Laboratory of Neurotrauma, 1st affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Lan Xiao
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Department of Neurosurgery, 2nd affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Feng Mei
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| |
Collapse
|
26
|
Del Grosso A, Carpi S, De Sarlo M, Scaccini L, Colagiorgio L, Alabed HBR, Angella L, Pellegrino RM, Tonazzini I, Emiliani C, Cecchini M. Chronic Rapamycin administration via drinking water mitigates the pathological phenotype in a Krabbe disease mouse model through autophagy activation. Biomed Pharmacother 2024; 173:116351. [PMID: 38422660 DOI: 10.1016/j.biopha.2024.116351] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 02/17/2024] [Accepted: 02/23/2024] [Indexed: 03/02/2024] Open
Abstract
Krabbe disease (KD) is a rare disorder arising from the deficiency of the lysosomal enzyme galactosylceramidase (GALC), leading to the accumulation of the cytotoxic metabolite psychosine (PSY) in the nervous system. This accumulation triggers demyelination and neurodegeneration, and despite ongoing research, the underlying pathogenic mechanisms remain incompletely understood, with no cure currently available. Previous studies from our lab revealed the involvement of autophagy dysfunctions in KD pathogenesis, showcasing p62-tagged protein aggregates in the brains of KD mice and heightened p62 levels in the KD sciatic nerve. We also demonstrated that the autophagy inducer Rapamycin (RAPA) can partially reinstate the wild type (WT) phenotype in KD primary cells by decreasing the number of p62 aggregates. In this study, we tested RAPA in the Twitcher (TWI) mouse, a spontaneous KD mouse model. We administered the drug ad libitum via drinking water (15 mg/L) starting from post-natal day (PND) 21-23. We longitudinally monitored the mouse motor performance through grip strength and rotarod tests, and a set of biochemical parameters related to the KD pathogenesis (i.e. autophagy markers expression, PSY accumulation, astrogliosis and myelination). Our findings demonstrate that RAPA significantly enhances motor functions at specific treatment time points and reduces astrogliosis in TWI brain, spinal cord, and sciatic nerves. Utilizing western blot and immunohistochemistry, we observed a decrease in p62 aggregates in TWI nervous tissues, corroborating our earlier in-vitro results. Moreover, RAPA treatment partially removes PSY in the spinal cord. In conclusion, our results advocate for considering RAPA as a supportive therapy for KD. Notably, as RAPA is already available in pharmaceutical formulations for clinical use, its potential for KD treatment can be rapidly evaluated in clinical trials.
Collapse
Affiliation(s)
- Ambra Del Grosso
- Istituto Nanoscienze - CNR, Pisa, Piazza San Silvestro 12, Pisa 56127, Italy; Laboratorio NEST, Scuola Normale Superiore, Piazza S. Silvestro 12, 56127, Pisa, Italy.
| | - Sara Carpi
- Istituto Nanoscienze - CNR, Pisa, Piazza San Silvestro 12, Pisa 56127, Italy
| | - Miriam De Sarlo
- Istituto Nanoscienze - CNR, Pisa, Piazza San Silvestro 12, Pisa 56127, Italy
| | - Luca Scaccini
- Laboratorio NEST, Scuola Normale Superiore, Piazza S. Silvestro 12, 56127, Pisa, Italy
| | - Laura Colagiorgio
- Istituto Nanoscienze - CNR, Pisa, Piazza San Silvestro 12, Pisa 56127, Italy
| | - Husam B R Alabed
- Department of Chemistry, Biology, and Biotechnologies, University of Perugia, Perugia, Italy
| | - Lucia Angella
- Istituto Nanoscienze - CNR, Pisa, Piazza San Silvestro 12, Pisa 56127, Italy
| | | | - Ilaria Tonazzini
- Istituto Nanoscienze - CNR, Pisa, Piazza San Silvestro 12, Pisa 56127, Italy
| | - Carla Emiliani
- Department of Chemistry, Biology, and Biotechnologies, University of Perugia, Perugia, Italy
| | - Marco Cecchini
- Istituto Nanoscienze - CNR, Pisa, Piazza San Silvestro 12, Pisa 56127, Italy.
| |
Collapse
|
27
|
Chen S, Liu S, Huang Y, Huang S, Zhang W, Xie H, Lu L. 5Z-7-Oxozaenol attenuates cuprizone-induced demyelination in mice through microglia polarization regulation. Brain Behav 2024; 14:e3487. [PMID: 38648385 PMCID: PMC11034864 DOI: 10.1002/brb3.3487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 12/06/2023] [Accepted: 02/21/2024] [Indexed: 04/25/2024] Open
Abstract
INTRODUCTION Demyelination is a key factor in axonal degeneration and neural loss, leading to disability in multiple sclerosis (MS) patients. Transforming growth factor beta activated kinase 1 (TAK1) is a critical molecule involved in immune and inflammatory signaling pathways. Knockout of microglia TAK1 can inhibit autoimmune inflammation of the brain and spinal cord and improve the outcome of MS. However, it is unclear whether inhibiting TAK1 can alleviate demyelination. METHODS Eight-week-old male c57bl/6j mice were randomly divided into five groups: (a) the control group, (b) the group treated with cuprizone (CPZ) only, (c) the group treated with 5Z-7-Oxozaenol (OZ) only, and (d) the group treated with both cuprizone and 15 μg/30 μg OZ. Demyelination in the mice of this study was induced by administration of CPZ (ig) at a daily dose of 400 mg/kg for consecutive 5 weeks. OZ was intraperitoneally administered at mentioned doses twice a week, starting from week 3 after beginning cuprizone treatment. Histology, rotarod test, grasping test, pole test, Western blot, RT-PCR, and ELISA were used to evaluate corpus callosum demyelination, behavioral impairment, oligodendrocyte differentiation, TAK1 signaling pathway expression, microglia, and related cytokines. RESULTS Our results demonstrated that OZ protected against myelin loss and behavior impairment caused by CPZ. Additionally, OZ rescued the loss of oligodendrocytes in CPZ-induced mice. OZ inhibited the activation of JNK, p65, and p38 pathways, transformed M1 polarized microglia into M2 phenotype, and increased brain-derived neurotrophic factor (BDNF) expression to attenuate demyelination in CPZ-treated mice. Furthermore, OZ reduced the expression of proinflammatory cytokines and increases anti-inflammatory cytokines in CPZ-treated mice. CONCLUSION These findings suggest that inhibiting TAK1 may be an effective approach for treating demyelinating diseases.
Collapse
Affiliation(s)
- Shiyu Chen
- Department of NeurologyZhujiang Hospital, Southern Medical UniversityGuangzhouChina
- Department of General PracticeZhujiang Hospital, Southern Medical UniversityGuangzhouChina
| | - Siyao Liu
- Department of General PracticeZhujiang Hospital, Southern Medical UniversityGuangzhouChina
| | - Yalun Huang
- Department of NeurologyZhujiang Hospital, Southern Medical UniversityGuangzhouChina
| | - Shiwen Huang
- Department of NeurologyZhujiang Hospital, Southern Medical UniversityGuangzhouChina
| | - Wanzhou Zhang
- Department of NeurologyZhujiang Hospital, Southern Medical UniversityGuangzhouChina
| | - Huifang Xie
- Department of NeurologyZhujiang Hospital, Southern Medical UniversityGuangzhouChina
| | - Lingli Lu
- Department of General PracticeZhujiang Hospital, Southern Medical UniversityGuangzhouChina
| |
Collapse
|
28
|
Hull VL, Wang Y, McDonough J, Zhu M, Burns T, Al Ramel N, Dehghani A, Guo F, Pleasure D. Astroglial conditional Slc13a3 knockout is therapeutic in murine Canavan leukodystrophy. Ann Clin Transl Neurol 2024; 11:1059-1062. [PMID: 38282243 PMCID: PMC11021635 DOI: 10.1002/acn3.52010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/22/2023] [Accepted: 01/19/2024] [Indexed: 01/30/2024] Open
Abstract
Canavan disease is a leukodystrophy caused by ASPA mutations that diminish oligodendroglial aspartoacylase activity, and is characterized by markedly elevated brain concentrations of the aspartoacylase substrate N-acetyl-l-aspartate (NAA) and by astroglial and intramyelinic vacuolation. Astroglia express NaDC3 (encoded by SLC13A3), a sodium-coupled transporter for NAA and other dicarboxylates. Astroglial conditional Slc13a3 deletion in aspartoacylase-deficient Canavan disease model mice ("CD mice") reversed brain NAA elevation and improved motor function. These results demonstrate that astroglial NaDC3 contributes to brain NAA elevation in CD mice, and suggest that suppressing astroglial NaDC3 activity would ameliorate human Canavan disease.
Collapse
Affiliation(s)
- Vanessa L. Hull
- Department of NeurologyUC Davis School of MedicineSacramentoCaliforniaUSA
- Shriners Hospital for ChildrenSacramentoCaliforniaUSA
- Department of PhysiologyDavid Geffen School of Medicine, UCLALos AngelesCaliforniaUSA
| | - Yan Wang
- Department of NeurologyUC Davis School of MedicineSacramentoCaliforniaUSA
- Shriners Hospital for ChildrenSacramentoCaliforniaUSA
| | | | - Meina Zhu
- Department of NeurologyUC Davis School of MedicineSacramentoCaliforniaUSA
- Shriners Hospital for ChildrenSacramentoCaliforniaUSA
| | - Travis Burns
- Department of NeurologyUC Davis School of MedicineSacramentoCaliforniaUSA
- Shriners Hospital for ChildrenSacramentoCaliforniaUSA
| | - Najmah Al Ramel
- Department of Biological SciencesKent State UniversityKentOhioUSA
| | - Ali Dehghani
- Department of NeurologyUC Davis School of MedicineSacramentoCaliforniaUSA
- Shriners Hospital for ChildrenSacramentoCaliforniaUSA
| | - Fuzheng Guo
- Department of NeurologyUC Davis School of MedicineSacramentoCaliforniaUSA
- Shriners Hospital for ChildrenSacramentoCaliforniaUSA
| | - David Pleasure
- Department of NeurologyUC Davis School of MedicineSacramentoCaliforniaUSA
- Shriners Hospital for ChildrenSacramentoCaliforniaUSA
| |
Collapse
|
29
|
Zhong J, Wang C, Zhang D, Yao X, Zhao Q, Huang X, Lin F, Xue C, Wang Y, He R, Li XY, Li Q, Wang M, Zhao S, Afridi SK, Zhou W, Wang Z, Xu Y, Xu Z. PCDHA9 as a candidate gene for amyotrophic lateral sclerosis. Nat Commun 2024; 15:2189. [PMID: 38467605 PMCID: PMC10928119 DOI: 10.1038/s41467-024-46333-5] [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: 05/18/2023] [Accepted: 02/23/2024] [Indexed: 03/13/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease. To identify additional genetic factors, we analyzed exome sequences in a large cohort of Chinese ALS patients and found a homozygous variant (p.L700P) in PCDHA9 in three unrelated patients. We generated Pcdhα9 mutant mice harboring either orthologous point mutation or deletion mutation. These mice develop progressive spinal motor loss, muscle atrophy, and structural/functional abnormalities of the neuromuscular junction, leading to paralysis and early lethality. TDP-43 pathology is detected in the spinal motor neurons of aged mutant mice. Mechanistically, we demonstrate that Pcdha9 mutation causes aberrant activation of FAK and PYK2 in aging spinal cord, and dramatically reduced NKA-α1 expression in motor neurons. Our single nucleus multi-omics analysis reveals disturbed signaling involved in cell adhesion, ion transport, synapse organization, and neuronal survival in aged mutant mice. Together, our results present PCDHA9 as a potential ALS gene and provide insights into its pathogenesis.
Collapse
Affiliation(s)
- Jie Zhong
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Chaodong Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Disease, Beijing, 100053, China.
| | - Dan Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Xiaoli Yao
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Quanzhen Zhao
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xusheng Huang
- Department of Neurology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Feng Lin
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Chun Xue
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Yaqing Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Ruojie He
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xu-Ying Li
- Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Disease, Beijing, 100053, China
| | - Qibin Li
- Shenzhen Clabee Biotechnology Incorporation, Shenzhen, 518057, China
| | - Mingbang Wang
- Shanghai Key Laboratory of Birth Defects, Division of Neonatology, Children's Hospital of Fudan University, National Center for Children's Health, Shanghai, 201102, China
| | - Shaoli Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Shabbir Khan Afridi
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Wenhao Zhou
- Shanghai Key Laboratory of Birth Defects, Division of Neonatology, Children's Hospital of Fudan University, National Center for Children's Health, Shanghai, 201102, China
| | - Zhanjun Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Disease, Beijing, 100053, China
| | - Yanming Xu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Zhiheng Xu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
30
|
Bahat A, Itzhaki E, Weiss B, Tolmasov M, Tsoory M, Kuperman Y, Brandis A, Shurrush KA, Dikstein R. Lowering mutant huntingtin by small molecules relieves Huntington's disease symptoms and progression. EMBO Mol Med 2024; 16:523-546. [PMID: 38374466 PMCID: PMC10940305 DOI: 10.1038/s44321-023-00020-y] [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: 05/11/2023] [Revised: 11/21/2023] [Accepted: 12/07/2023] [Indexed: 02/21/2024] Open
Abstract
Huntington's disease (HD) is an incurable inherited disorder caused by a repeated expansion of glutamines in the huntingtin gene (Htt). The mutant protein causes neuronal degeneration leading to severe motor and psychological symptoms. Selective downregulation of the mutant Htt gene expression is considered the most promising therapeutic approach for HD. We report the identification of small molecule inhibitors of Spt5-Pol II, SPI-24 and SPI-77, which selectively lower mutant Htt mRNA and protein levels in HD cells. In the BACHD mouse model, their direct delivery to the striatum diminished mutant Htt levels, ameliorated mitochondrial dysfunction, restored BDNF expression, and improved motor and anxiety-like phenotypes. Pharmacokinetic studies revealed that these SPIs pass the blood-brain-barrier. Prolonged subcutaneous injection or oral administration to early-stage mice significantly delayed disease deterioration. SPI-24 long-term treatment had no side effects or global changes in gene expression. Thus, lowering mutant Htt levels by small molecules can be an effective therapeutic strategy for HD.
Collapse
Affiliation(s)
- Anat Bahat
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, 76100, Israel.
| | - Elad Itzhaki
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Benjamin Weiss
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Michael Tolmasov
- The Mina & Everard Goodman Faculty of Life-Sciences and The Leslie & Susan Gonda Multidisciplinary Brain Research Center Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Michael Tsoory
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Yael Kuperman
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Alexander Brandis
- Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Khriesto A Shurrush
- The Nancy and Stephen Grand Israel National Center for Personalized Medicine, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Rivka Dikstein
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, 76100, Israel.
| |
Collapse
|
31
|
Cardoso VSL, Valente-Amaral A, Monteiro RFM, Meira CLS, de Meira NS, da Silva MN, Pinheiro JDJV, Bastos GDNT, Felício JS, Yamada ES. Aqueous extract of Swietenia macrophylla leaf exerts an anti-inflammatory effect in a murine model of Parkinson's disease induced by 6-OHDA. Front Neurosci 2024; 18:1351718. [PMID: 38449740 PMCID: PMC10914943 DOI: 10.3389/fnins.2024.1351718] [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: 12/06/2023] [Accepted: 02/05/2024] [Indexed: 03/08/2024] Open
Abstract
Introduction Parkinson's disease affects 2% of the population aged over 65 years and is the second most common neurodegenerative disorder in the general population. The appearance of motor symptoms is associated with the degeneration of dopaminergic neurons in the nigrostriatal pathway. Clinically significant nonmotor symptoms are also important for severe disability with disease progression. Pharmacological treatment with levodopa, which involves dopamine restitution, results in a temporary improvement in motor symptoms. Among the mechanisms underlying the pathogenesis of the disease are exacerbated oxidative stress, mitochondrial dysfunction, and neuroinflammation. A phytochemical prospecting study showed that the aqueous extract of the leaves from Swietenia macrophylla (Melineaceae), known as mahogany, has polyphenols with antioxidant and anti-inflammatory capacity in a significantly higher percentage than leaf extracts from other Amazonian plants. Furthermore, the antioxidant and anti-inflammatory capacity of aqueous extract of mahogany leaf has already been demonstrated in an in vitro model. In this study, we hypothesized that the aqueous extract of mahogany leaf (AEML) has a neuroprotective effect in a murine model of Parkinson's disease induced by 6-hydroxidopamine (6-OHDA), due to antioxidant and anti-inflammatory properties of its phenolic compounds. Methods Mice were treated daily with the mahogany extract at a dose of 50 mg/kg, starting 7 days before 6-OHDA infusion until post-surgery day 7. Results and discussion The animals from the 6-OHDA/mahogany group, which corresponds to animals injected with the toxin and treated with aqueous extract of the mahogany leaf, presented distinct behavioral phenotypes after apomorphine challenge and were therefore subdivided into 2 groups, 6-OHDA/mahogany F1 and 6-OHDA/mahogany F2. The F1 group showed a significant increase in contralateral rotations, whereas the F2 group did not show rotations after the apomorphine stimulus. In the F1 group, there was an increase, although not significant, in motor performance in the open field and elevated plus maze tests, whereas in the F2 group, there was significant improvement, which may be related to the lesser degree of injury to the nigrostriatal dopaminergic pathway. The TH+ histopathological analysis, a dopaminergic neuron marker, confirmed that the lesion to the nigrostriatal dopaminergic pathway was more pronounced in 6-OHDA/mahogany F1 than in 6-OHDA/mahogany F2. Our main result consisted of signs of improvement in the inflammatory profile in both the F1 and F2 6-OHDA/mahogany groups, such as a lower number of IBA-1+ microglial cells in the ventral striatum and substantia nigra pars compacta and a reduction in GFAP+ expression, an astrocyte marker, in the dorsal striatum. In this study, several bioactive compounds in the aqueous extract of mahogany leaf may have contributed to the observed beneficial effects. Further studies are necessary to better characterize their applicability for treating chronic degenerative diseases with inflammatory and oxidative bases, such as Parkinson's disease.
Collapse
Affiliation(s)
- Váldina Solimar Lopes Cardoso
- Experimental Neuropathology Laboratory, João de Barros Barreto University Hospital, Federal University of Pará, Belém, Brazil
- Oncology Research Center and Graduate Program in Oncology and Medical Sciences, João de Barros Barreto University Hospital, Federal University of Pará, Belém, Brazil
| | - Anderson Valente-Amaral
- Experimental Neuropathology Laboratory, João de Barros Barreto University Hospital, Federal University of Pará, Belém, Brazil
- Graduate Program in Neuroscience and Cellular Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Rayan Fidel Martins Monteiro
- Graduate Program in Neuroscience and Cellular Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
- Neuroinflammation Laboratory, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Clarina Loius Silva Meira
- Experimental Neuropathology Laboratory, João de Barros Barreto University Hospital, Federal University of Pará, Belém, Brazil
| | - Natália Silva de Meira
- Experimental Neuropathology Laboratory, João de Barros Barreto University Hospital, Federal University of Pará, Belém, Brazil
| | - Milton Nascimento da Silva
- Liquid Chromatography Laboratory, Institute of Exact and Natural Science, Federal University of Pará, Belém, Brazil
| | - João de Jesus Viana Pinheiro
- Oncology Research Center and Graduate Program in Oncology and Medical Sciences, João de Barros Barreto University Hospital, Federal University of Pará, Belém, Brazil
- Laboratory of Pathological Anatomy and Immunohistochemistry, School of Dentistry, Federal University of Pará, Belém, Brazil
| | - Gilmara de Nazareth Tavares Bastos
- Graduate Program in Neuroscience and Cellular Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
- Neuroinflammation Laboratory, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - João Soares Felício
- Oncology Research Center and Graduate Program in Oncology and Medical Sciences, João de Barros Barreto University Hospital, Federal University of Pará, Belém, Brazil
- Endocrinology Division, University Hospital João de Barros Barreto, Federal University of Pará, Belém, Brazil
| | - Elizabeth Sumi Yamada
- Experimental Neuropathology Laboratory, João de Barros Barreto University Hospital, Federal University of Pará, Belém, Brazil
- Oncology Research Center and Graduate Program in Oncology and Medical Sciences, João de Barros Barreto University Hospital, Federal University of Pará, Belém, Brazil
- Graduate Program in Neuroscience and Cellular Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| |
Collapse
|
32
|
Wu Y, Zhu X, Jiang W, Li J, Li H, Zhang K, Yang Y, Qu S, Guan X, Bai Y, Guo H, Dai L. LMNA-related muscular dystrophy involving myoblast proliferation and apoptosis through the FOXO1/GADD45A pathway. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166943. [PMID: 37951507 DOI: 10.1016/j.bbadis.2023.166943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/07/2023] [Accepted: 10/30/2023] [Indexed: 11/14/2023]
Abstract
LMNA-related muscular dystrophy is a major disease phenotype causing mortality and morbidity in laminopathies, but its pathogenesis is still unclear. To explore the molecular pathogenesis, a knock-in mouse harbouring the Lmna-W520R mutation was modelled. Morphological and motor functional analyses showed that homozygous mutant mice revealed severe muscular atrophy, profound motor dysfunction, and shortened lifespan, while heterozygotes showed a variant arrangement of muscle bundles and mildly reduced motor capacity. Mechanistically, the FOXO1/GADD45A pathway involving muscle atrophy processes was found to be altered in vitro and in vivo assays. The expression levels of FOXO1 and its downstream regulatory molecule GADD45A significantly increased in atrophic muscle tissue. The elevated expression of FOXO1 was associated with decreased H3K27me3 in its gene promotor region. Overexpression of GADD45A induced apoptosis and cell cycle arrest of myoblasts in vitro, and it could be partially restored by the FOXO1 inhibitor AS1842856, which also slowed the muscle atrophy process with improved motor function and prolonged survival time of homozygous mutant mice in vivo. Notably, the inhibitor also partly rescued the apoptosis and cell cycle arrest of hiPSC-derived myoblasts harbouring the LMNA-W520R mutation. Together, these data suggest that the activation of the FOXO1/GADD45A pathway contributes to the pathogenesis of LMNA-related muscle atrophy, and it might serve as a potential therapeutic target for laminopathies.
Collapse
Affiliation(s)
- Yue Wu
- Department of Medical Genetics, College of Basic Medical Science, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Xintong Zhu
- Department of Medical Genetics, College of Basic Medical Science, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Wen Jiang
- Department of Medical Genetics, College of Basic Medical Science, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Jia Li
- Department of Medical Genetics, College of Basic Medical Science, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Hongyan Li
- Department of Medical Genetics, College of Basic Medical Science, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Kun Zhang
- Department of Pathogenic Biology, College of Basic Medical Science, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yixuan Yang
- Department of Medical Genetics, College of Basic Medical Science, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Song Qu
- Department of Medical Genetics, College of Basic Medical Science, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Xingying Guan
- Department of Medical Genetics, College of Basic Medical Science, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yun Bai
- Department of Medical Genetics, College of Basic Medical Science, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Hong Guo
- Department of Medical Genetics, College of Basic Medical Science, Army Medical University (Third Military Medical University), Chongqing 400038, China.
| | - Limeng Dai
- Department of Medical Genetics, College of Basic Medical Science, Army Medical University (Third Military Medical University), Chongqing 400038, China; Department of Gynecology and Obstetrics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China.
| |
Collapse
|
33
|
Fonseca A, Szysz M, Ly HT, Cordeiro C, Sepúlveda N. IgG Antibody Responses to Epstein-Barr Virus in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: Their Effective Potential for Disease Diagnosis and Pathological Antigenic Mimicry. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:161. [PMID: 38256421 PMCID: PMC10820613 DOI: 10.3390/medicina60010161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/02/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024]
Abstract
Background and Objectives: The diagnosis and pathology of myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) remain under debate. However, there is a growing body of evidence for an autoimmune component in ME/CFS caused by the Epstein-Barr virus (EBV) and other viral infections. Materials and Methods: In this work, we analyzed a large public dataset on the IgG antibodies to 3054 EBV peptides to understand whether these immune responses could help diagnose patients and trigger pathological autoimmunity; we used healthy controls (HCs) as a comparator cohort. Subsequently, we aimed at predicting the disease status of the study participants using a super learner algorithm targeting an accuracy of 85% when splitting data into train and test datasets. Results: When we compared the data of all ME/CFS patients or the data of a subgroup of those patients with non-infectious or unknown disease triggers to the data of the HC, we could not find an antibody-based classifier that would meet the desired accuracy in the test dataset. However, we could identify a 26-antibody classifier that could distinguish ME/CFS patients with an infectious disease trigger from the HCs with 100% and 90% accuracies in the train and test sets, respectively. We finally performed a bioinformatic analysis of the EBV peptides associated with these 26 antibodies. We found no correlation between the importance metric of the selected antibodies in the classifier and the maximal sequence homology between human proteins and each EBV peptide recognized by these antibodies. Conclusions: In conclusion, these 26 antibodies against EBV have an effective potential for disease diagnosis in a subset of patients. However, the peptides associated with these antibodies are less likely to induce autoimmune B-cell responses that could explain the pathogenesis of ME/CFS.
Collapse
Affiliation(s)
- André Fonseca
- Faculty of Sciences and Technology, University of Algarve, 8005-139 Faro, Portugal; (A.F.); (C.C.)
- CEAUL—Centre of Statistics and its Applications, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal
| | - Mateusz Szysz
- Faculty of Mathematics & Information Science, Warsaw University of Technology, 00-662 Warsaw, Poland; (M.S.); (H.T.L.)
| | - Hoang Thien Ly
- Faculty of Mathematics & Information Science, Warsaw University of Technology, 00-662 Warsaw, Poland; (M.S.); (H.T.L.)
| | - Clara Cordeiro
- Faculty of Sciences and Technology, University of Algarve, 8005-139 Faro, Portugal; (A.F.); (C.C.)
- CEAUL—Centre of Statistics and its Applications, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal
| | - Nuno Sepúlveda
- CEAUL—Centre of Statistics and its Applications, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal
- Faculty of Mathematics & Information Science, Warsaw University of Technology, 00-662 Warsaw, Poland; (M.S.); (H.T.L.)
| |
Collapse
|
34
|
Bidgood R, Zubelzu M, Ruiz-Ortega JA, Morera-Herreras T. Automated procedure to detect subtle motor alterations in the balance beam test in a mouse model of early Parkinson's disease. Sci Rep 2024; 14:862. [PMID: 38195974 PMCID: PMC10776624 DOI: 10.1038/s41598-024-51225-1] [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: 08/01/2023] [Accepted: 01/02/2024] [Indexed: 01/11/2024] Open
Abstract
Parkinson's disease (PD) is the most common motor neurodegenerative disorder, characterised by aggregated α-synuclein (α-syn) constituting Lewy bodies. We aimed to investigate temporal changes in motor impairments in a PD mouse model induced by overexpression of α-syn with the conventional manual analysis of the balance beam test and a novel approach using machine learning algorithms to automate behavioural analysis. We combined automated animal tracking using markerless pose estimation in DeepLabCut, with automated behavioural classification in Simple Behavior Analysis. Our automated procedure was able to detect subtle motor deficits in mouse performances in the balance beam test that the manual analysis approach could not assess. The automated model revealed time-course significant differences for the "walking" behaviour in the mean interval between each behavioural bout, the median event bout duration and the classifier probability of occurrence in male PD mice, even though no statistically significant loss of tyrosine hydroxylase in the nigrostriatal system was found in either sex. These findings are valuable for early detection of motor impairment in early PD animal models. We provide a user-friendly, step-by-step guide for automated assessment of mouse performances in the balance beam test, which aims to be replicable without any significant computational and programming knowledge.
Collapse
Affiliation(s)
- Raphaëlle Bidgood
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Barrio Sarriena S/N, 48940, Leioa, Biscay, Spain
| | - Maider Zubelzu
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Barrio Sarriena S/N, 48940, Leioa, Biscay, Spain
- Autonomic and Movement Disorders Unit, Neurodegenerative Diseases, Biobizkaia, Barakaldo, Biscay, Spain
| | - Jose Angel Ruiz-Ortega
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Barrio Sarriena S/N, 48940, Leioa, Biscay, Spain
- Autonomic and Movement Disorders Unit, Neurodegenerative Diseases, Biobizkaia, Barakaldo, Biscay, Spain
| | - Teresa Morera-Herreras
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Barrio Sarriena S/N, 48940, Leioa, Biscay, Spain.
- Autonomic and Movement Disorders Unit, Neurodegenerative Diseases, Biobizkaia, Barakaldo, Biscay, Spain.
| |
Collapse
|
35
|
Bornstein B, Watkins B, Passini FS, Blecher R, Assaraf E, Sui XM, Brumfeld V, Tsoory M, Kröger S, Zelzer E. The mechanosensitive ion channel ASIC2 mediates both proprioceptive sensing and spinal alignment. Exp Physiol 2024; 109:135-147. [PMID: 36951012 PMCID: PMC10988735 DOI: 10.1113/ep090776] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 02/20/2023] [Indexed: 03/24/2023]
Abstract
By translating mechanical forces into molecular signals, proprioceptive neurons provide the CNS with information on muscle length and tension, which is necessary to control posture and movement. However, the identities of the molecular players that mediate proprioceptive sensing are largely unknown. Here, we confirm the expression of the mechanosensitive ion channel ASIC2 in proprioceptive sensory neurons. By combining in vivo proprioception-related functional tests with ex vivo electrophysiological analyses of muscle spindles, we showed that mice lacking Asic2 display impairments in muscle spindle responses to stretch and motor coordination tasks. Finally, analysis of skeletons of Asic2 loss-of-function mice revealed a specific effect on spinal alignment. Overall, we identify ASIC2 as a key component in proprioceptive sensing and a regulator of spine alignment.
Collapse
Affiliation(s)
- Bavat Bornstein
- Department of Molecular GeneticsWeizmann Institute of ScienceRehovotIsrael
| | - Bridgette Watkins
- Department of Physiological Genomics, Biomedical CenterLudwig‐Maximilians‐UniversityPlanegg‐MartinsriedGermany
| | - Fabian S. Passini
- Department of Molecular GeneticsWeizmann Institute of ScienceRehovotIsrael
| | - Ronen Blecher
- Orthopedic DepartmentAssuta Ashdod University Hospital, Ashdod, Israel, affiliated to Ben Gurion University of the NegevBeer ShebaIsrael
| | - Eran Assaraf
- Department of Orthopedic SurgeryShamir Medical Center, Assaf HaRofeh Campus, Zeffifin, Israel, affiliated to Sackler Faculty of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Xiao Meng Sui
- Department of Chemical Research SupportWeizmann Institute of ScienceRehovotIsrael
| | - Vlad Brumfeld
- Department of Chemical Research SupportWeizmann Institute of ScienceRehovotIsrael
| | - Michael Tsoory
- Department of Veterinary ResourcesWeizmann Institute of ScienceRehovotIsrael
| | - Stephan Kröger
- Department of Physiological Genomics, Biomedical CenterLudwig‐Maximilians‐UniversityPlanegg‐MartinsriedGermany
| | - Elazar Zelzer
- Department of Molecular GeneticsWeizmann Institute of ScienceRehovotIsrael
| |
Collapse
|
36
|
Wilk CM, Cathomas F, Török O, Le Berichel J, Park MD, Bigenwald C, Heaton GR, Hamon P, Troncoso L, Scull BP, Dangoor D, Silvin A, Fleischmann R, Belabed M, Lin H, Merad Taouli E, Boettcher S, Li L, Aubry A, Manz MG, Kofler JK, Yue Z, Lira SA, Ginhoux F, Crary JF, McClain KL, Picarsic JL, Russo SJ, Allen CE, Merad M. Circulating senescent myeloid cells infiltrate the brain and cause neurodegeneration in histiocytic disorders. Immunity 2023; 56:2790-2802.e6. [PMID: 38091952 PMCID: PMC11587932 DOI: 10.1016/j.immuni.2023.11.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 09/05/2023] [Accepted: 11/14/2023] [Indexed: 12/18/2023]
Abstract
Neurodegenerative diseases (ND) are characterized by progressive loss of neuronal function. Mechanisms of ND pathogenesis are incompletely understood, hampering the development of effective therapies. Langerhans cell histiocytosis (LCH) is an inflammatory neoplastic disorder caused by hematopoietic progenitors expressing mitogen-activated protein kinase (MAPK)-activating mutations that differentiate into senescent myeloid cells that drive lesion formation. Some individuals with LCH subsequently develop progressive and incurable neurodegeneration (LCH-ND). Here, we showed that LCH-ND was caused by myeloid cells that were clonal with peripheral LCH cells. Circulating BRAFV600E+ myeloid cells caused the breakdown of the blood-brain barrier (BBB), enhancing migration into the brain parenchyma where they differentiated into senescent, inflammatory CD11a+ macrophages that accumulated in the brainstem and cerebellum. Blocking MAPK activity and senescence programs reduced peripheral inflammation, brain parenchymal infiltration, neuroinflammation, neuronal damage and improved neurological outcome in preclinical LCH-ND. MAPK activation and senescence programs in circulating myeloid cells represent targetable mechanisms of LCH-ND.
Collapse
Affiliation(s)
- C Matthias Wilk
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncology Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Flurin Cathomas
- Nash Family Department of Neuroscience, Brain & Body Research Center, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Orsolya Török
- Department of Neurology, University of Pécs, Medical School, Pécs, Hungary
| | - Jessica Le Berichel
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncology Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew D Park
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncology Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Camille Bigenwald
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncology Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Gustave Roussy Cancer Campus, Villejuif, France; Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - George R Heaton
- Department of Neurology and Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Pauline Hamon
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncology Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Leanna Troncoso
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncology Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brooks P Scull
- Texas Children's Cancer Center, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Diana Dangoor
- Department of Neurology and Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Artificial Intelligence, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Neuropathology Brain Bank and Research CoRE, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aymeric Silvin
- Gustave Roussy Cancer Campus, Villejuif, France; Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - Ryan Fleischmann
- Texas Children's Cancer Center, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Meriem Belabed
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncology Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Howard Lin
- Texas Children's Cancer Center, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Elias Merad Taouli
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncology Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Steffen Boettcher
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Long Li
- Nash Family Department of Neuroscience, Brain & Body Research Center, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Antonio Aubry
- Nash Family Department of Neuroscience, Brain & Body Research Center, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Markus G Manz
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Julia K Kofler
- Division of Neuropathology, Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zhenyu Yue
- Department of Neurology and Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sergio A Lira
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncology Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Florent Ginhoux
- Gustave Roussy Cancer Campus, Villejuif, France; Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - John F Crary
- Department of Neurology and Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Artificial Intelligence, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Neuropathology Brain Bank and Research CoRE, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kenneth L McClain
- Texas Children's Cancer Center, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Jennifer L Picarsic
- Division of Pathology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pathology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Scott J Russo
- Nash Family Department of Neuroscience, Brain & Body Research Center, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carl E Allen
- Texas Children's Cancer Center, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA.
| | - Miriam Merad
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncology Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
37
|
Khidr HY, Hassan NF, Abdelrahman SS, El-Ansary MR, El-Yamany MF, Rabie MA. Formoterol attenuated mitochondrial dysfunction in rotenone-induced Parkinson's disease in a rat model: Role of PINK-1/PARKIN and PI3K/Akt/CREB/BDNF/TrKB axis. Int Immunopharmacol 2023; 125:111207. [PMID: 37956489 DOI: 10.1016/j.intimp.2023.111207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/06/2023] [Accepted: 11/08/2023] [Indexed: 11/15/2023]
Abstract
β2-adrenoreceptors (β2AR have been identified recently as regulators of the α-synuclein gene (SNCA), one of the key milieus endorsed in injury of dopamine neurons in Parkinson's disease (PD). Accumulation of α-synuclein leads to mitochondrial dysfunction via downregulation of mitophagy proteins (PINK-1 and PARKIN) and inhibition of mitochondria biogenesis (PGC-1α) along with an increase in the master inflammatory regulator NF-κB p65 production that provokes neurodegeneration and diminishes neuroprotective signaling pathway (PI3k/Akt/CREB/BDNF). Recently, formoterol exhibited a promising neuroprotective effect against neurodegenerative conditions associated with brain inflammation. Therefore, the present investigation aims to unveil the possible neuroprotective activity of formoterol, β2AR agonist, against rotenone-induced PD in rats. Rats received rotenone (1.5 mg/kg; s.c.) every other day for 3 weeks and cured with formoterol (25 μg/kg/day; i.p.) 1 hr. after rotenone administration, starting from day 11. Formoterol treatment succeeded in upregulating β2-adrenoreceptor expression in PD rats and preserving the function and integrity of dopaminergic neurons as witnessed by enhancement of muscular performance in tests, open field, grip strength-meter, and Rotarod, besides the increment in substantia nigra and striatal tyrosine hydroxylase immunoexpression. In parallel, formoterol boosted mitophagy by activation of PINK1 and PARKIN and preserved mitochondrial membrane potential. Additionally, formoterol stimulated the neuro-survival signaling axis via stimulation of PI3k/pS473-Akt/pS133-CREB/BDNF cascade to attenuate neuronal loss. Noteworthy formoterol reduces neuro-inflammatory status by decreasing NFκBp65 immunoexpression and TNF-α content. Finally, formoterol's potential as a stimulant therapy of mitophagy via the PINK1/PARKIN axis and regulation of mitochondrial biogenesis by increasing PGC-1α to maintain mitochondrial homeostasis along with stimulation of PI3k/Akt/CREB/BDNF axis.
Collapse
Affiliation(s)
- Haneen Y Khidr
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt
| | - Noha F Hassan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt
| | - S S Abdelrahman
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Egypt
| | - Mona R El-Ansary
- Department of Biochemistry, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt
| | - Mohammed F El-Yamany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562 Cairo, Egypt
| | - Mostafa A Rabie
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562 Cairo, Egypt.
| |
Collapse
|
38
|
Chen Z, Li Y, Rasheed M, Wang H, Lei R, Zhao T, Deng Y, Ma H. Altered expression of inflammation-associated molecules in striatum: an implication for sensitivity to heavy ion radiations. Front Cell Neurosci 2023; 17:1252958. [PMID: 38107411 PMCID: PMC10725200 DOI: 10.3389/fncel.2023.1252958] [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: 07/04/2023] [Accepted: 11/06/2023] [Indexed: 12/19/2023] Open
Abstract
Background and objective Heavy ion radiation is one of the major hazards astronauts face during space expeditions, adversely affecting the central nervous system. Radiation causes severe damage to sensitive brain regions, especially the striatum, resulting in cognitive impairment and other physiological issues in astronauts. However, the intensity of brain damage and associated underlying molecular pathological mechanisms mediated by heavy ion radiation are still unknown. The present study is aimed to identify the damaging effect of heavy ion radiation on the striatum and associated underlying pathological mechanisms. Materials and methods Two parallel cohorts of rats were exposed to radiation in multiple doses and times. Cohort I was exposed to 15 Gy of 12C6+ ions radiation, whereas cohort II was exposed to 3.4 Gy and 8 Gy with 56Fe26+ ions irradiation. Physiological and behavioural tests were performed, followed by 18F-FDG-PET scans, transcriptomics analysis of the striatum, and in-vitro studies to verify the interconnection between immune cells and neurons. Results Both cohorts revealed more persistent striatum dysfunction than other brain regions under heavy ion radiation at multiple doses and time, exposed by physiological, behavioural, and 18F-FDG-PET scans. Transcriptomic analysis revealed that striatum dysfunction is linked with an abnormal immune system. In vitro studies demonstrated that radiation mediated diversified effects on different immune cells and sustained monocyte viability but inhibited its differentiation and migration, leading to chronic neuroinflammation in the striatum and might affect other associated brain regions. Conclusion Our findings suggest that striatum dysfunction under heavy ion radiation activates abnormal immune systems, leading to chronic neuroinflammation and neuronal injury.
Collapse
Affiliation(s)
- Zixuan Chen
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing, China
| | - Yumeng Li
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing, China
| | - Madiha Rasheed
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing, China
| | - Hao Wang
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing, China
| | - Runhong Lei
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing, China
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, China
| | - Tuo Zhao
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing, China
| | - Yulin Deng
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing, China
| | - Hong Ma
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
39
|
Bérard M, Martínez-Drudis L, Sheta R, El-Agnaf OMA, Oueslati A. Non-invasive systemic viral delivery of human alpha-synuclein mimics selective and progressive neuropathology of Parkinson's disease in rodent brains. Mol Neurodegener 2023; 18:91. [PMID: 38012703 PMCID: PMC10683293 DOI: 10.1186/s13024-023-00683-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 11/17/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND Alpha-synuclein (α-syn) aggregation into proteinaceous intraneuronal inclusions, called Lewy bodies (LBs), is the neuropathological hallmark of Parkinson's disease (PD) and related synucleinopathies. However, the exact role of α-syn inclusions in PD pathogenesis remains elusive. This lack of knowledge is mainly due to the absence of optimal α-syn-based animal models that recapitulate the different stages of neurodegeneration. METHODS Here we describe a novel approach for a systemic delivery of viral particles carrying human α-syn allowing for a large-scale overexpression of this protein in the mouse brain. This approach is based on the use of a new generation of adeno-associated virus (AAV), AAV-PHP.eB, with an increased capacity to cross the blood-brain barrier, thus offering a viable tool for a non-invasive and large-scale gene delivery in the central nervous system. RESULTS Using this model, we report that widespread overexpression of human α-syn induced selective degeneration of dopaminergic (DA) neurons, an exacerbated neuroinflammatory response in the substantia nigra and a progressive manifestation of PD-like motor impairments. Interestingly, biochemical analysis revealed the presence of insoluble α-syn oligomers in the midbrain. Together, our data demonstrate that a single non-invasive systemic delivery of viral particles overexpressing α-syn prompted selective and progressive neuropathology resembling the early stages of PD. CONCLUSIONS Our new in vivo model represents a valuable tool to study the role of α-syn in PD pathogenesis and in the selective vulnerability of nigral DA neurons; and offers the opportunity to test new strategies targeting α-syn toxicity for the development of disease-modifying therapies for PD and related disorders.
Collapse
Affiliation(s)
- Morgan Bérard
- CHU de Québec Research Center, Axe Neurosciences, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Laura Martínez-Drudis
- CHU de Québec Research Center, Axe Neurosciences, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Razan Sheta
- CHU de Québec Research Center, Axe Neurosciences, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Omar M A El-Agnaf
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, 34110, Qatar
| | - Abid Oueslati
- CHU de Québec Research Center, Axe Neurosciences, Quebec City, Canada.
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada.
| |
Collapse
|
40
|
Drake DM, Zhen D, Kerrebijn I, Or B, Gao S, Afsharian K, Tran J, Bhatia S, Cheng A, Wells PG. Breast cancer 1 (BRCA1) protection in altered gene expression and neurodevelopmental disorders due to physiological and ethanol-enhanced reactive oxygen species formation. Free Radic Biol Med 2023; 208:272-284. [PMID: 37541454 DOI: 10.1016/j.freeradbiomed.2023.08.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 06/08/2023] [Accepted: 08/02/2023] [Indexed: 08/06/2023]
Abstract
The breast cancer 1 (Brca1) susceptibility gene regulates the repair of reactive oxygen species (ROS)-mediated DNA damage, which is implicated in neurodevelopmental disorders. Alcohol (ethanol, EtOH) exposure during pregnancy causes fetal alcohol spectrum disorders (FASD), including abnormal brain function, associated with enhanced ROS-initiated DNA damage. Herein, oxidative DNA damage in fetal brains and neurodevelopmental disorders were enhanced in saline-exposed +/- vs. +/+ Brca1 littermates. A single EtOH exposure during gestation further enhanced oxidative DNA damage, altered the expression of developmental/DNA damage response genes in fetal brains, and resulted in neurodevelopmental disorders, all of which were BRCA1-dependent. Pretreatment with the ROS inhibitor phenylbutylnitrone (PBN) blocked DNA damage and some neurodevelopmental disorders in both saline- and EtOH-exposed progeny, corroborating a ROS-dependent mechanism. Fetal BRCA1 protects against altered gene expression and neurodevelopmental disorders caused by both physiological and EtOH-enhanced levels of ROS formation. BRCA1 deficiencies may enhance the risk for FASD.
Collapse
Affiliation(s)
- Danielle M Drake
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada; Centre for Pharmaceutical Oncology, Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Danlin Zhen
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Isabel Kerrebijn
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Benjamin Or
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Sophie Gao
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Kian Afsharian
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada; Centre for Pharmaceutical Oncology, Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Jason Tran
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Shama Bhatia
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada; Centre for Pharmaceutical Oncology, Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Ashley Cheng
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada; Centre for Pharmaceutical Oncology, Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Peter G Wells
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada; Centre for Pharmaceutical Oncology, Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
41
|
Liu X, Rao S, Chen W, Felix K, Ni J, Sahasrabudhe A, Lin S, Wang Q, Liu Y, He Z, Xu J, Huang S, Hong E, Yau T, Anikeeva P, Zhao X. Fatigue-resistant hydrogel optical fibers enable peripheral nerve optogenetics during locomotion. Nat Methods 2023; 20:1802-1809. [PMID: 37857906 PMCID: PMC11009937 DOI: 10.1038/s41592-023-02020-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 08/21/2023] [Indexed: 10/21/2023]
Abstract
We develop soft and stretchable fatigue-resistant hydrogel optical fibers that enable optogenetic modulation of peripheral nerves in naturally behaving animals during persistent locomotion. The formation of polymeric nanocrystalline domains within the hydrogels yields fibers with low optical losses of 1.07 dB cm-1, Young's modulus of 1.6 MPa, stretchability of 200% and fatigue strength of 1.4 MPa against 30,000 stretch cycles. The hydrogel fibers permitted light delivery to the sciatic nerve, optogenetically activating hindlimb muscles in Thy1::ChR2 mice during 6-week voluntary wheel running assays while experiencing repeated deformation. The fibers additionally enabled optical inhibition of pain hypersensitivity in an inflammatory model in TRPV1::NpHR mice over an 8-week period. Our hydrogel fibers offer a motion-adaptable and robust solution to peripheral nerve optogenetics, facilitating the investigation of somatosensation.
Collapse
Affiliation(s)
- Xinyue Liu
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI, USA.
| | - Siyuan Rao
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, USA.
- Department of Biomedical Engineering, Binghamton University, State University of New York, Binghamton, NY, USA.
| | - Weixuan Chen
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, USA
| | - Kayla Felix
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, USA
| | - Jiahua Ni
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Atharva Sahasrabudhe
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Shaoting Lin
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI, USA
| | - Qianbin Wang
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, USA
- Department of Biomedical Engineering, Binghamton University, State University of New York, Binghamton, NY, USA
| | - Yuanyuan Liu
- Somatosensation and Pain Unit, National Institute of Dental and Craniofacial Research, National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, USA
| | - Zhigang He
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Jingyi Xu
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, USA
| | - Sizhe Huang
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, USA
- Department of Biomedical Engineering, Binghamton University, State University of New York, Binghamton, NY, USA
| | - Eunji Hong
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, USA
- Department of Biomedical Engineering, Binghamton University, State University of New York, Binghamton, NY, USA
| | - Todd Yau
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, Amherst, MA, USA
| | - Polina Anikeeva
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA.
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
- K. Lisa Yang Brain-Body Center, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Xuanhe Zhao
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
42
|
Anshu K, Nair AK, Srinath S, Laxmi TR. Altered Developmental Trajectory in Male and Female Rats in a Prenatal Valproic Acid Exposure Model of Autism Spectrum Disorder. J Autism Dev Disord 2023; 53:4390-4411. [PMID: 35976506 DOI: 10.1007/s10803-022-05684-y] [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] [Accepted: 07/13/2022] [Indexed: 10/15/2022]
Abstract
Early motor and sensory developmental delays precede Autism Spectrum Disorder (ASD) diagnosis and may serve as early indicators of ASD. The literature on sensorimotor development in animal models is sparse, male centered, and has mixed findings. We characterized early development in a prenatal valproic acid (VPA) model of ASD and found sex-specific developmental delays in VPA rats. We created a developmental composite score combining 15 test readouts, yielding a reliable gestalt measure spanning physical, sensory, and motor development, that effectively discriminated between VPA and control groups. Considering the heterogeneity in ASD phenotype, the developmental composite offers a robust metric that can enable comparison across different animal models of ASD and can serve as an outcome measure for early intervention studies.
Collapse
Affiliation(s)
- Kumari Anshu
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Main Road, Bengaluru, Karnataka, 560029, India
- Waisman Center, University of Wisconsin-Madison, Madison, 53705, WI, USA
| | - Ajay Kumar Nair
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Main Road, Bengaluru, Karnataka, 560029, India
- Center for Healthy Minds, University of Wisconsin-Madison, Madison, 53703, WI, USA
| | - Shoba Srinath
- Department of Child and Adolescent Psychiatry, National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Main Road, Bengaluru, Karnataka, 560029, India
| | - T Rao Laxmi
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Main Road, Bengaluru, Karnataka, 560029, India.
| |
Collapse
|
43
|
Stölting G, Dinh HA, Volkert M, Hellmig N, Schewe J, Hennicke L, Seidel E, Oberacher H, Zhang J, Lifton RP, Urban I, Long M, Rivalan M, Nottoli T, Scholl UI. Isradipine therapy in Cacna1dIle772Met/+ mice ameliorates primary aldosteronism and neurologic abnormalities. JCI Insight 2023; 8:e162468. [PMID: 37698934 PMCID: PMC10619505 DOI: 10.1172/jci.insight.162468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 09/06/2023] [Indexed: 09/14/2023] Open
Abstract
Somatic gain-of-function mutations in the L-type calcium channel CaV1.3 (CACNA1D gene) cause adrenal aldosterone-producing adenomas and micronodules. De novo germline mutations are found in a syndrome of primary aldosteronism, seizures, and neurologic abnormalities (PASNA) as well as in autism spectrum disorder. Using CRISPR/Cas9, we here generated mice with a Cacna1d gain-of-function mutation found in both adenomas and PASNA syndrome (Cacna1dIle772Met/+). These mice show reduced body weight and increased mortality from weaning to approximately 100 days of age. Male mice do not breed, likely due to neuromotor impairment, and the offspring of female mice die perinatally, likely due to lack of maternal care. Mice generated by in vitro fertilization showed elevated intracellular calcium in the aldosterone-producing zona glomerulosa, an elevated aldosterone/renin ratio, and persistently elevated serum aldosterone on a high-salt diet as signs of primary aldosteronism. Anesthesia with ketamine and xylazine induced tonic-clonic seizures. Neurologic abnormalities included hyperlocomotion, impaired performance in the rotarod test, impaired nest building, and slight changes in social behavior. Intracellular calcium in the zona glomerulosa, aldosterone levels, and rotarod performance responded to treatment with the calcium channel blocker isradipine, with implications for the therapy of patients with aldosterone-producing lesions and with PASNA syndrome.
Collapse
Affiliation(s)
- Gabriel Stölting
- Center of Functional Genomics, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Hoang An Dinh
- Center of Functional Genomics, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Marina Volkert
- Center of Functional Genomics, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Nicole Hellmig
- Center of Functional Genomics, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Julia Schewe
- Center of Functional Genomics, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Luise Hennicke
- Center of Functional Genomics, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Eric Seidel
- Center of Functional Genomics, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Herbert Oberacher
- Institute of Legal Medicine and Core Facility Metabolomics, Medical University of Innsbruck, Innsbruck, Austria
| | - Junhui Zhang
- Department of Genetics and Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Richard P. Lifton
- Department of Genetics and Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, New York, USA
| | | | - Melissa Long
- Animal Behavior Phenotyping Facility (ABPF), Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Marion Rivalan
- Animal Behavior Phenotyping Facility (ABPF), Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Timothy Nottoli
- Section of Comparative Medicine, Yale Genome Editing Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Ute I. Scholl
- Center of Functional Genomics, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
44
|
Van Dam D, Valkenburg F, Van Kolen K, Pintelon I, Timmermans JP, De Deyn PP. Behavioral and Neuropathological Phenotyping of the Tau58/2 and Tau58/4 Transgenic Mouse Models for FTDP-17. Life (Basel) 2023; 13:2088. [PMID: 37895469 PMCID: PMC10608666 DOI: 10.3390/life13102088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/10/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND The Tau58/2 and Tau58/4 mouse lines expressing 0N4R tau with a P301S mutation mimic aspects of frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17). In a side-by-side comparison, we report the age-dependent development of cognitive, motor, and behavioral deficits in comparison with the spatial-temporal evolution of cellular tau pathology in both models. METHODS We applied the SHIRPA primary screen and specific neuromotor, behavioral, and cognitive paradigms. The spatiotemporal development of tau pathology was investigated immunohistochemically. Levels of sarkosyl-insoluble paired helical filaments were determined via a MesoScale Discovery biomarker assay. RESULTS Neuromotor impairments developed from age 3 months in both models. On electron microscopy, spinal cord neurofibrillary pathology was visible in mice aged 3 months; however, AT8 immunoreactivity was not yet observed in Tau58/4 mice. Behavioral abnormalities and memory deficits occurred at a later stage (>9 months) when tau pathology was fully disseminated throughout the brain. Spatiotemporally, tau pathology spread from the spinal cord via the midbrain to the frontal cortex, while the hippocampus was relatively spared, thus explaining the late onset of cognitive deficits. CONCLUSIONS Our findings indicate the face and construct validity of both Tau58 models, which may provide new, valuable insights into the pathologic effects of tau species in vivo and may consequently facilitate the development of new therapeutic targets to delay or halt neurodegenerative processes occurring in tauopathies.
Collapse
Affiliation(s)
- Debby Van Dam
- Laboratory of Neurochemistry and Behavior, Experimental Neurobiology Unit, University of Antwerp, Wilrijk, 2610 Antwerp, Belgium;
- Department of Neurology and Alzheimer Center Groningen, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Femke Valkenburg
- Laboratory of Neurochemistry and Behavior, Experimental Neurobiology Unit, University of Antwerp, Wilrijk, 2610 Antwerp, Belgium;
| | - Kristof Van Kolen
- Neuroscience Department, Janssen Research and Development, 2340 Beerse, Belgium;
| | - Isabel Pintelon
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, 2610 Antwerp, Belgium; (I.P.); (J.-P.T.)
| | - Jean-Pierre Timmermans
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, 2610 Antwerp, Belgium; (I.P.); (J.-P.T.)
| | - Peter Paul De Deyn
- Laboratory of Neurochemistry and Behavior, Experimental Neurobiology Unit, University of Antwerp, Wilrijk, 2610 Antwerp, Belgium;
- Department of Neurology and Alzheimer Center Groningen, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
45
|
Cobelo-Gómez S, Sánchez-Iglesias S, Rábano A, Senra A, Aguiar P, Gómez-Lado N, García-Varela L, Burgueño-García I, Lampón-Fernández L, Fernández-Pombo A, Díaz-López EJ, Prado-Moraña T, San Millán B, Araújo-Vilar D. A murine model of BSCL2-associated Celia's encephalopathy. Neurobiol Dis 2023; 187:106300. [PMID: 37717662 DOI: 10.1016/j.nbd.2023.106300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 09/06/2023] [Accepted: 09/14/2023] [Indexed: 09/19/2023] Open
Abstract
Celia's encephalopathy or progressive encephalopathy with/without lipodystrophy is a neurodegenerative disease with a fatal prognosis in childhood. It is generally caused by the c.985C > T variant in the BSCL2 gene, leading to the skipping of exon 7 and resulting in an aberrant seipin protein (Celia-seipin). To precisely define the temporal evolution and the mechanisms involved in neurodegeneration, lipodystrophy and fatty liver in Celia's encephalopathy, our group has generated the first global knock-in murine model for the aberrant human transcript of BSCL2 (Bscl2Celia/Celia) using a strategy based on the Cre/loxP recombination system. In order to carry out a characterization at the neurological, adipose tissue and hepatic level, behavioral studies, brain PET, metabolic, histological and molecular studies were performed. Around 12% of homozygous and 5.4% of heterozygous knock-in mice showed severe neurological symptoms early in life, and their life expectancy was dramatically reduced. Severe generalized lipodystrophy and mild hepatic steatosis were present in these affected animals, while serum triglycerides and glucose metabolism were normal, with no insulin resistance. Furthermore, the study revealed a reduction in brain glucose uptake, along with patchy loss of Purkinje cells and the presence of intranuclear inclusions in cerebellar cortex cells. Homozygous, non-severely-affected knock-in mice showed a decrease in locomotor activity and greater anxiety compared with their wild type littermates. Bscl2Celia/Celia is the first murine model of Celia's encephalopathy which partially recapitulates the phenotype and severe neurodegenerative picture suffered by these patients. This model will provide a helpful tool to investigate both the progressive encephalopathy with/without lipodystrophy and congenital generalized lipodystrophy.
Collapse
Affiliation(s)
- Silvia Cobelo-Gómez
- UETeM-Molecular Pathology Group. Department of Psychiatry, Radiology, Public Health, Nursing and Medicine, IDIS-CIMUS, University of Santiago de Compostela, Spain
| | - Sofía Sánchez-Iglesias
- UETeM-Molecular Pathology Group. Department of Psychiatry, Radiology, Public Health, Nursing and Medicine, IDIS-CIMUS, University of Santiago de Compostela, Spain
| | - Alberto Rábano
- Alzheimer's Disease Research Unit, CIEN Foundation, Queen Sofia Foundation Alzheimer Center, Madrid, Spain
| | - Ana Senra
- Department of Physiology, CIMUS, University of Santiago de Compostela, Spain
| | - Pablo Aguiar
- Molecular Imaging and Medical Physics, University of Santiago de Compostela-IDIS, Spain; Nuclear Medicine and Molecular Imaging Group, IDIS, University Clinical Hospital of Santiago de Compostela, Spain
| | - Noemí Gómez-Lado
- Molecular Imaging and Medical Physics, University of Santiago de Compostela-IDIS, Spain; Nuclear Medicine and Molecular Imaging Group, IDIS, University Clinical Hospital of Santiago de Compostela, Spain
| | - Lara García-Varela
- Molecular Imaging and Medical Physics, University of Santiago de Compostela-IDIS, Spain; Nuclear Medicine and Molecular Imaging Group, IDIS, University Clinical Hospital of Santiago de Compostela, Spain
| | - Iván Burgueño-García
- Alzheimer's Disease Research Unit, CIEN Foundation, Queen Sofia Foundation Alzheimer Center, Madrid, Spain
| | - Laura Lampón-Fernández
- UETeM-Molecular Pathology Group. Department of Psychiatry, Radiology, Public Health, Nursing and Medicine, IDIS-CIMUS, University of Santiago de Compostela, Spain
| | - Antía Fernández-Pombo
- UETeM-Molecular Pathology Group. Department of Psychiatry, Radiology, Public Health, Nursing and Medicine, IDIS-CIMUS, University of Santiago de Compostela, Spain; Division of Endocrinology and Nutrition, University Clinical Hospital of Santiago de Compostela, Spain
| | - Everardo Josué Díaz-López
- UETeM-Molecular Pathology Group. Department of Psychiatry, Radiology, Public Health, Nursing and Medicine, IDIS-CIMUS, University of Santiago de Compostela, Spain; Division of Endocrinology and Nutrition, University Clinical Hospital of Santiago de Compostela, Spain
| | - Teresa Prado-Moraña
- UETeM-Molecular Pathology Group. Department of Psychiatry, Radiology, Public Health, Nursing and Medicine, IDIS-CIMUS, University of Santiago de Compostela, Spain; Division of Endocrinology and Nutrition, University Clinical Hospital of Santiago de Compostela, Spain
| | - Beatriz San Millán
- Grupo de Enfermedades Raras y Medicina Pediátrica, Instituto de Investigación Sanitaria Galicia Sur (IISGS), Vigo, Spain; Pathology Department, Alvaro Cunqueiro Hospital, Vigo, Spain
| | - David Araújo-Vilar
- UETeM-Molecular Pathology Group. Department of Psychiatry, Radiology, Public Health, Nursing and Medicine, IDIS-CIMUS, University of Santiago de Compostela, Spain; Division of Endocrinology and Nutrition, University Clinical Hospital of Santiago de Compostela, Spain.
| |
Collapse
|
46
|
Wilk CM, Cathomas F, Török O, Le Berichel J, Park MD, Heaton GR, Hamon P, Troncoso L, Scull BP, Dangoor D, Silvin A, Fleischmann R, Belabed M, Lin H, Taouli EM, Boettcher S, Manz MG, Kofler JK, Yue Z, Lira SA, Ginhoux F, Crary JF, McClain KL, Picarsic JL, Russo SJ, Allen CE, Merad M. Circulating senescent myeloid cells drive blood brain barrier breakdown and neurodegeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.10.561744. [PMID: 37873371 PMCID: PMC10592746 DOI: 10.1101/2023.10.10.561744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Neurodegenerative diseases (ND) are characterized by progressive loss of neuronal function. Mechanisms of ND pathogenesis are incompletely understood, hampering the development of effective therapies. Langerhans cell histiocytosis (LCH) is an inflammatory neoplastic disorder caused by hematopoietic progenitors expressing MAPK activating mutations that differentiate into senescent myeloid cells that drive lesion formation. Some patients with LCH subsequently develop progressive and incurable neurodegeneration (LCH-ND). Here, we show that LCH-ND is caused by myeloid cells that are clonal with peripheral LCH cells. We discovered that circulating BRAF V600E + myeloid cells cause the breakdown of the blood-brain barrier (BBB), enhancing migration into the brain parenchyma where they differentiate into senescent, inflammatory CD11a + macrophages that accumulate in the brainstem and cerebellum. Blocking MAPK activity and senescence programs reduced parenchymal infiltration, neuroinflammation, neuronal damage and improved neurological outcome in preclinical LCH-ND. MAPK activation and senescence programs in circulating myeloid cells represent novel and targetable mechanisms of ND.
Collapse
|
47
|
Hossain SR, Karem H, Jafari Z, Kolb BE, Mohajerani MH. Early tactile stimulation influences the development of Alzheimer's disease in gestationally stressed APP NL-G-F adult offspring NL-G-F/NL-G-F mice. Exp Neurol 2023; 368:114498. [PMID: 37536439 DOI: 10.1016/j.expneurol.2023.114498] [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: 05/08/2023] [Revised: 07/20/2023] [Accepted: 07/25/2023] [Indexed: 08/05/2023]
Abstract
Alzheimer's disease (AD) is associated with cerebral plaques and tangles, reduced synapse number, and shrinkage in several brain areas and these morphological effects are associated with the onset of compromised cognitive, motor, and anxiety-like behaviours. The appearance of both anatomical and behavioural symptoms is worsened by stress. The focus of this study was to examine the effect of neonatal tactile stimulation on AD-like behavioural and neurological symptoms on APP NL-G-F/NL-G-F mice, a mouse model of AD, who have been gestationally stressed. Our findings indicate that neonatal tactile stimulation improves cognition, motor skills, and anxiety-like symptoms in both gestationally stressed and non-stressed adult APP mice and that these alterations are associated with reduced Aβ plaque formation. Thus, tactile stimulation appears to be a promising non-invasive preventative strategy for slowing the onset of dementia in aging animals.
Collapse
Affiliation(s)
- Shakhawat R Hossain
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge T1K 3M4, AB, Canada
| | - Hadil Karem
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge T1K 3M4, AB, Canada
| | - Zahra Jafari
- School of Communication Sciences and Disorders, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Bryan E Kolb
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge T1K 3M4, AB, Canada.
| | - Majid H Mohajerani
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge T1K 3M4, AB, Canada.
| |
Collapse
|
48
|
Martinez PA, Martinez VE, Rani S, Murrell M, Javors M, Gelfond J, Doorn JA, Fernandez E, Strong R. Impaired aldehyde detoxification exacerbates motor deficits in an alpha-synuclein mouse model of Parkinson's disease. Brain Behav 2023; 13:e3150. [PMID: 37452461 PMCID: PMC10498093 DOI: 10.1002/brb3.3150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 06/07/2023] [Indexed: 07/18/2023] Open
Abstract
INTRODUCTION The discovery of biogenic aldehydes in the postmortem parkinsonian brain and the ability of these aldehydes to modify and cross-link proteins has called attention to their possible role in Parkinson's disease. For example, many in vitro studies have found that the aldehyde metabolite of dopamine, 3,4-dihydroxyphenylacetaldehyde (DOPAL), induces the formation of stable, neurotoxic alpha-synuclein oligomers. METHODS To study this in vivo, mice deficient in the two aldehyde dehydrogenase enzymes (Aldh1a1 and Aldh2, DKO) primarily responsible for detoxification of DOPAL in the nigrostriatal pathway were crossed with mice that overexpress human wild-type alpha-synuclein. DKO overexpressing human wild-type alpha-synuclein (DKO/ASO) offspring were evaluated for impairment on motor tasks associated with Parkinsonism. RESULTS DKO/ASO mice developed severe motor deficits greater than that of mice overexpressing human wild-type alpha-synuclein alone. CONCLUSION These results provide evidence to support the idea that biogenic aldehydes such as DOPAL interact with human wild-type alpha-synuclein, directly or indirectly, in vivo to exacerbate locomotor deficits in Parkinson's disease.
Collapse
Affiliation(s)
- Paul Anthony Martinez
- Department of PharmacologyUniversity of Texas Health Science CenterSan AntonioTexasUSA
- Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health Science CenterSan AntonioTexasUSA
- Center for Biomedical NeuroscienceUniversity of Texas Health Science CenterSan AntonioTexasUSA
| | - Vanessa Elia Martinez
- Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health Science CenterSan AntonioTexasUSA
| | - Sheela Rani
- Department of PharmacologyUniversity of Texas Health Science CenterSan AntonioTexasUSA
- Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health Science CenterSan AntonioTexasUSA
| | - Meredith Murrell
- Department of PsychiatryUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| | - Martin Javors
- Department of PharmacologyUniversity of Texas Health Science CenterSan AntonioTexasUSA
- Department of PsychiatryUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| | - Jonathan Gelfond
- Department of MedicineUniversity of Texas Health Science CenterSan AntonioTexasUSA
- Department of Epidemiology & BiostatisticsUniversity of Texas Health Science CenterSan AntonioTexasUSA
| | - Jonathan Alan Doorn
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of PharmacyThe University of IowaIowa CityIowaUSA
| | - Elizabeth Fernandez
- Department of PharmacologyUniversity of Texas Health Science CenterSan AntonioTexasUSA
- Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health Science CenterSan AntonioTexasUSA
- Geriatric Research, Education, and Clinical CenterSouth Texas Veterans Health. Care NetworkSan AntonioTexasUSA
| | - Randy Strong
- Department of PharmacologyUniversity of Texas Health Science CenterSan AntonioTexasUSA
- Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health Science CenterSan AntonioTexasUSA
- Center for Biomedical NeuroscienceUniversity of Texas Health Science CenterSan AntonioTexasUSA
- Geriatric Research, Education, and Clinical CenterSouth Texas Veterans Health. Care NetworkSan AntonioTexasUSA
| |
Collapse
|
49
|
Yasuda T, Uchiyama T, Watanabe N, Ito N, Nakabayashi K, Mochizuki H, Onodera M. Peripheral immune system modulates Purkinje cell degeneration in Niemann-Pick disease type C1. Life Sci Alliance 2023; 6:e202201881. [PMID: 37369603 PMCID: PMC10300197 DOI: 10.26508/lsa.202201881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Niemann-Pick disease type C1 (NPC1) is a fatal lysosomal storage disorder characterized by progressive neuronal degeneration. Its key pathogenic events remain largely unknown. We have, herein, found that neonatal BM-derived cell transplantation can ameliorate Purkinje cell degeneration in NPC1 mice. We subsequently addressed the impact of the peripheral immune system on the neuropathogenesis observed in NPC1 mice. The depletion of mature lymphocytes promoted NPC1 phenotypes, thereby suggesting a neuroprotective effect of lymphocytes. Moreover, the peripheral infusion of CD4-positive cells (specifically, of regulatory T cells) from normal healthy donor ameliorated the cerebellar ataxic phenotype and enhanced the survival of Purkinje cells. Conversely, the depletion of regulatory T cells enhanced the onset of the neurological phenotype. On the other hand, circulating inflammatory monocytes were found to be involved in the progression of Purkinje cell degeneration, whereas the depletion of resident microglia had little effect. Our findings reveal a novel role of the adaptive and the innate immune systems in NPC1 neuropathology.
Collapse
Affiliation(s)
- Toru Yasuda
- Department of Human Genetics, National Center for Child Health and Development, Tokyo, Japan
| | - Toru Uchiyama
- Department of Human Genetics, National Center for Child Health and Development, Tokyo, Japan
| | - Nobuyuki Watanabe
- Department of Human Genetics, National Center for Child Health and Development, Tokyo, Japan
| | - Noriko Ito
- Department of Maternal-Fetal Biology, National Center for Child Health and Development, Tokyo, Japan
| | - Kazuhiko Nakabayashi
- Department of Maternal-Fetal Biology, National Center for Child Health and Development, Tokyo, Japan
| | - Hideki Mochizuki
- Department of Neurology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Masafumi Onodera
- Department of Human Genetics, National Center for Child Health and Development, Tokyo, Japan
| |
Collapse
|
50
|
Silva-Cardoso GK, Lazarini-Lopes W, Primini EO, Hallak JE, Crippa JA, Zuardi AW, Garcia-Cairasco N, Leite-Panissi CRA. Cannabidiol modulates chronic neuropathic pain aversion behavior by attenuation of neuroinflammation markers and neuronal activity in the corticolimbic circuit in male Wistar rats. Behav Brain Res 2023; 452:114588. [PMID: 37474023 DOI: 10.1016/j.bbr.2023.114588] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/26/2023] [Accepted: 07/16/2023] [Indexed: 07/22/2023]
Abstract
Chronic neuropathic pain (CNP) is a vast world health problem often associated with the somatosensory domain. This conceptualization is problematic because, unlike most other sensations that are usually affectively neutral and may present emotional, affective, and cognitive impairments. Neuronal circuits that modulate pain can increase or decrease painful sensitivity based on several factors, including context and expectation. The objective of this study was to evaluate whether subchronic treatment with Cannabidiol (CBD; 0.3, 3, and 10 mg/kg intraperitoneal route - i.p., once a day for 3 days) could promote pain-conditioned reversal, in the conditioned place preference (CPP) test, in male Wistar rats submitted to chronic constriction injury (CCI) of the sciatic nerve. Then, we evaluated the expression of astrocytes and microglia in animals treated with CBD through the immunofluorescence technique. Our results demonstrated that CBD promoted the reversal of CPP at 3 and 10 mg/kg. In CCI animals, CBD was able to attenuate the increase in neuronal hyperactivity, measured by FosB protein expression, in the regions of the corticolimbic circuit: anterior cingulate cortex (ACC), complex basolateral amygdala (BLA), granular layer of the dentate gyrus (GrDG), and dorsal hippocampus (DH) - adjacent to subiculum (CA1). CBD also prevented the increased expression of GFAP and IBA-1 in CCI animals. We concluded that CBD effects on CNP are linked to the modulation of the aversive component of pain. These effects decrease chronic neuronal activation and inflammatory markers in regions of the corticolimbic circuit.
Collapse
Affiliation(s)
- Gleice K Silva-Cardoso
- Department of Psychology, School of Philosophy, Science and Letters of Ribeirão Preto, University of São Paulo, Brazil; National Institute for Translational Medicine (INCT-TM; CNPq), São Paulo, Brazil
| | - Willian Lazarini-Lopes
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto School of Medicine, University of São Paulo, Brazil; Department of Pharmacology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil
| | - Eduardo Octaviano Primini
- Department of Psychology, School of Philosophy, Science and Letters of Ribeirão Preto, University of São Paulo, Brazil
| | - Jaime E Hallak
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto School of Medicine, University of São Paulo, Brazil; National Institute for Translational Medicine (INCT-TM; CNPq), São Paulo, Brazil
| | - José A Crippa
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto School of Medicine, University of São Paulo, Brazil; National Institute for Translational Medicine (INCT-TM; CNPq), São Paulo, Brazil
| | - Antônio W Zuardi
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto School of Medicine, University of São Paulo, Brazil; National Institute for Translational Medicine (INCT-TM; CNPq), São Paulo, Brazil
| | - Norberto Garcia-Cairasco
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto School of Medicine, University of São Paulo, Brazil; Department of Physiology, Ribeirão Preto School of Medicine, University of São Paulo, Brazil
| | - Christie R A Leite-Panissi
- Department of Psychology, School of Philosophy, Science and Letters of Ribeirão Preto, University of São Paulo, Brazil; National Institute for Translational Medicine (INCT-TM; CNPq), São Paulo, Brazil.
| |
Collapse
|