51
|
Kim KT, Kwak YJ, Han SC, Hwang JH. Impairment of motor coordination and interneuron migration in perinatal exposure to glufosinate-ammonium. Sci Rep 2020; 10:20647. [PMID: 33244012 PMCID: PMC7691990 DOI: 10.1038/s41598-020-76869-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 08/03/2020] [Indexed: 11/09/2022] Open
Abstract
Glufosinate-ammonium (GLA) is a broad-spectrum herbicide for agricultural weed control and crop desiccation. Due to many GLA-resistant crops being developed to effectively control weeds and increase harvest yields, herbicide usage and the residual GLA in food has increased significantly. Though perinatal exposure by the residual GLA in food might affect brain development, the developmental neurotoxicity of GLA is still unclear. Therefore, this study aimed to investigate the effects of perinatal exposure to GLA on cortical development. The analysis revealed that perinatal GLA exposure altered behavioral changes in offspring, especially motor functional behavior. Moreover, perinatal GLA exposure affected cortical development, particularly by disrupting interneuron migration. These results provide new evidence that early life exposure to GLA alters cortical development.
Collapse
Affiliation(s)
- Kyung-Tai Kim
- Jeonbuk Branch Institute, Korea Institute of Toxicology, 30 Baekhak1-gil, Jeongeup, Jeollabuk-do, 56212, Republic of Korea
| | - Ye-Jung Kwak
- Jeonbuk Branch Institute, Korea Institute of Toxicology, 30 Baekhak1-gil, Jeongeup, Jeollabuk-do, 56212, Republic of Korea
| | - Su-Cheol Han
- Jeonbuk Branch Institute, Korea Institute of Toxicology, 30 Baekhak1-gil, Jeongeup, Jeollabuk-do, 56212, Republic of Korea.
| | - Jeong Ho Hwang
- Jeonbuk Branch Institute, Korea Institute of Toxicology, 30 Baekhak1-gil, Jeongeup, Jeollabuk-do, 56212, Republic of Korea.
| |
Collapse
|
52
|
Therapeutic candidates for the Zika virus identified by a high-throughput screen for Zika protease inhibitors. Proc Natl Acad Sci U S A 2020; 117:31365-31375. [PMID: 33229545 DOI: 10.1073/pnas.2005463117] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
When Zika virus emerged as a public health emergency there were no drugs or vaccines approved for its prevention or treatment. We used a high-throughput screen for Zika virus protease inhibitors to identify several inhibitors of Zika virus infection. We expressed the NS2B-NS3 Zika virus protease and conducted a biochemical screen for small-molecule inhibitors. A quantitative structure-activity relationship model was employed to virtually screen ∼138,000 compounds, which increased the identification of active compounds, while decreasing screening time and resources. Candidate inhibitors were validated in several viral infection assays. Small molecules with favorable clinical profiles, especially the five-lipoxygenase-activating protein inhibitor, MK-591, inhibited the Zika virus protease and infection in neural stem cells. Members of the tetracycline family of antibiotics were more potent inhibitors of Zika virus infection than the protease, suggesting they may have multiple mechanisms of action. The most potent tetracycline, methacycline, reduced the amount of Zika virus present in the brain and the severity of Zika virus-induced motor deficits in an immunocompetent mouse model. As Food and Drug Administration-approved drugs, the tetracyclines could be quickly translated to the clinic. The compounds identified through our screening paradigm have the potential to be used as prophylactics for patients traveling to endemic regions or for the treatment of the neurological complications of Zika virus infection.
Collapse
|
53
|
Childers WE, Elokely KM, Abou-Gharbia M. The Resurrection of Phenotypic Drug Discovery. ACS Med Chem Lett 2020; 11:1820-1828. [PMID: 33062159 DOI: 10.1021/acsmedchemlett.0c00006] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 03/06/2020] [Indexed: 12/12/2022] Open
Abstract
Prior to genetic mapping, the majority of drug discovery efforts involved phenotypic screening, wherein compounds were screened in either in vitro or in vivo models thought to mimic the disease state of interest. While never completely abandoning phenotypic approaches, the labor intensive nature of such tests encouraged the pharmaceutical industry to move away from them in favor of target-based drug discovery, which facilitated throughput and allowed for the efficient screening of large numbers of compounds. However, a consequence of reliance on target-based screening was an increased number of failures in clinical trials due to poor correlation between novel mechanistic targets and the actual disease state. As a result, the field has seen a recent resurrection in phenotypic drug discovery approaches. In this work, we highlight some recent phenotypic projects from our industrial past and in our current academic drug discovery environment that have provided encouraging results.
Collapse
Affiliation(s)
- Wayne E. Childers
- Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, 3307 N. Broad Street, Philadelphia, Pennsylvania 19140, United States
| | - Khaled M. Elokely
- Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, 3307 N. Broad Street, Philadelphia, Pennsylvania 19140, United States
- Department of Chemistry, College of Science and Technology, Temple University, 1925 N. 12th Street, Philadelphia, Pennsylvania 19122, United States
- Department of Pharmaceutical Chemistry, Tanta University, Tanta 31527, Egypt
| | - Magid Abou-Gharbia
- Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, 3307 N. Broad Street, Philadelphia, Pennsylvania 19140, United States
| |
Collapse
|
54
|
Koike Y, Li B, Ganji N, Zhu H, Miyake H, Chen Y, Lee C, Janssen Lok M, Zozaya C, Lau E, Lee D, Chusilp S, Zhang Z, Yamoto M, Wu RY, Inoue M, Uchida K, Kusunoki M, Delgado-Olguin P, Mertens L, Daneman A, Eaton S, Sherman PM, Pierro A. Remote ischemic conditioning counteracts the intestinal damage of necrotizing enterocolitis by improving intestinal microcirculation. Nat Commun 2020; 11:4950. [PMID: 33009377 PMCID: PMC7532542 DOI: 10.1038/s41467-020-18750-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 09/09/2020] [Indexed: 02/07/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a devastating disease of premature infants with high mortality rate, indicating the need for precision treatment. NEC is characterized by intestinal inflammation and ischemia, as well derangements in intestinal microcirculation. Remote ischemic conditioning (RIC) has emerged as a promising tool in protecting distant organs against ischemia-induced damage. However, the effectiveness of RIC against NEC is unknown. To address this gap, we aimed to determine the efficacy and mechanism of action of RIC in experimental NEC. NEC was induced in mouse pups between postnatal day (P) 5 and 9. RIC was applied through intermittent occlusion of hind limb blood flow. RIC, when administered in the early stages of disease progression, decreases intestinal injury and prolongs survival. The mechanism of action of RIC involves increasing intestinal perfusion through vasodilation mediated by nitric oxide and hydrogen sulfide. RIC is a viable and non-invasive treatment strategy for NEC. Necrotizing enterocolitis (NEC) is one of the most lethal gastrointestinal emergencies in neonates needing precision treatment. Here the authors show that remote ischemic conditioning is a non-invasive therapeutic method that enhances blood flow in the intestine, reduces damage, and improves NEC outcome.
Collapse
Affiliation(s)
- Yuhki Koike
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada.,Departments of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Bo Li
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Niloofar Ganji
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Haitao Zhu
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Hiromu Miyake
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Yong Chen
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Carol Lee
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Maarten Janssen Lok
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Carlos Zozaya
- Division of Neonatology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Ethan Lau
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Dorothy Lee
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Sinobol Chusilp
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Zhen Zhang
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Masaya Yamoto
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Richard Y Wu
- Cell Biology Program, Research Institute, Division of Gastroenterology, Hepatology and Nutrition, Hospital for Sick Children, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Mikihiro Inoue
- Departments of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Keiichi Uchida
- Departments of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Masato Kusunoki
- Departments of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Paul Delgado-Olguin
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.,Heart & Stroke Richard Lewar Centre of Excellence, Toronto, ON, Canada
| | - Luc Mertens
- The Labatt Family Heart Center, Cardiology, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Alan Daneman
- Department of Diagnostic Imaging, Division of Nuclear Medicine, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Simon Eaton
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Philip M Sherman
- Cell Biology Program, Research Institute, Division of Gastroenterology, Hepatology and Nutrition, Hospital for Sick Children, Toronto, ON, Canada.,Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Agostino Pierro
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada. .,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada. .,Department of Surgery, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
55
|
Roper RJ, Goodlett CR, Martínez de Lagrán M, Dierssen M. Behavioral Phenotyping for Down Syndrome in Mice. ACTA ACUST UNITED AC 2020; 10:e79. [PMID: 32780566 DOI: 10.1002/cpmo.79] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Down syndrome (DS) is the most frequent genetic cause of intellectual disability, characterized by alterations in different behavioral symptom domains: neurodevelopment, motor behavior, and cognition. As mouse models have the potential to generate data regarding the neurological basis for the specific behavioral profile of DS, and may indicate pharmacological treatments with the potential to affect their behavioral phenotype, it is important to be able to assess disease-relevant behavioral traits in animal models in order to provide biological plausibility to the potential findings. The field is at a juncture that requires assessments that may effectively translate the findings acquired in mouse models to humans with DS. In this article, behavioral tests are described that are relevant to the domains affected in DS. A neurodevelopmental behavioral screen, the balance beam test, and the Multivariate Concentric Square Field test to assess multiple behavioral phenotypes and locomotion are described, discussing the ways to merge these findings to more fully understand cognitive strengths and weaknesses in this population. New directions for approaches to cognitive assessment in mice and humans are discussed. © 2020 Wiley Periodicals LLC. Basic Protocol 1: Preweaning neurodevelopmental battery Basic Protocol 2: Balance beam Basic Protocol 3: Multivariate concentric square field test (MCSF).
Collapse
Affiliation(s)
| | | | - María Martínez de Lagrán
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Mara Dierssen
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain.,Biomedical Research Networking Center on Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
56
|
Demaestri C, Pan T, Critz M, Ofray D, Gallo M, Bath KG. Type of early life adversity confers differential, sex-dependent effects on early maturational milestones in mice. Horm Behav 2020; 124:104763. [PMID: 32407728 PMCID: PMC7487052 DOI: 10.1016/j.yhbeh.2020.104763] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/16/2020] [Accepted: 04/23/2020] [Indexed: 12/24/2022]
Abstract
Early life adversity (ELA) increases risk for negative health outcomes, with sex disparities in prevalence and form of ELA experienced and risk for neuropsychiatric pathology. ELA comes in many forms (e.g. parental neglect/loss, limited access to resources) but whether disparate forms of ELA have common effects on outcomes, and if males and females are equally affected, remains unknown. Epidemiological studies often fail to accurately account for differences in type, timing, and duration of adversity experienced. Rodent models allow precise control of many of these variables. However, differences in the form of ELA, species, strain, housing, and testing paradigms used may contribute to differences in outcomes leading to questions of whether differences are the result of the form of ELA or these other variables. Here, we directly compared two mouse models of ELA, maternal separation (MS) and limited bedding (LB) in males and females on development of the body, motor and visual milestones, stress physiology, and anxiety-like behavior. LB affected timing of early milestones, somatic growth, and stress physiology in both sexes, yet only females showed later anxiety-like behaviors. MS rearing affected males and females similarly in early milestone development, yet only males showed changes in stress physiology and anxiety-like outcomes. These studies provide a platform to directly compare MS and LB models within one lab. The current work advances our understanding of the unique features of ELA that shape early neurodevelopmental events and risk for later pathology, increasing the translational relevance of these ELA models.
Collapse
Affiliation(s)
- Camila Demaestri
- Department of Cognitive, Linguistic, and Psychological Sciences, 190 Thayer St., Box 1821, Brown University, Providence, RI 02912, USA
| | - Tracy Pan
- Department of Cognitive, Linguistic, and Psychological Sciences, 190 Thayer St., Box 1821, Brown University, Providence, RI 02912, USA
| | - Madalyn Critz
- Department of Cognitive, Linguistic, and Psychological Sciences, 190 Thayer St., Box 1821, Brown University, Providence, RI 02912, USA
| | - Dayshalis Ofray
- Department of Cognitive, Linguistic, and Psychological Sciences, 190 Thayer St., Box 1821, Brown University, Providence, RI 02912, USA
| | - Meghan Gallo
- Department of Cognitive, Linguistic, and Psychological Sciences, 190 Thayer St., Box 1821, Brown University, Providence, RI 02912, USA
| | - Kevin G Bath
- Department of Cognitive, Linguistic, and Psychological Sciences, 190 Thayer St., Box 1821, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
57
|
Aimiuwu OV, Fowler AM, Sah M, Teoh JJ, Kanber A, Pyne NK, Petri S, Rosenthal-Weiss C, Yang M, Harper SQ, Frankel WN. RNAi-Based Gene Therapy Rescues Developmental and Epileptic Encephalopathy in a Genetic Mouse Model. Mol Ther 2020; 28:1706-1716. [PMID: 32353324 PMCID: PMC7335739 DOI: 10.1016/j.ymthe.2020.04.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/19/2020] [Accepted: 04/08/2020] [Indexed: 01/23/2023] Open
Abstract
Developmental and epileptic encephalopathy (DEE) associated with de novo variants in the gene encoding dynamin-1 (DNM1) is a severe debilitating disease with no pharmacological remedy. Like most genetic DEEs, the majority of DNM1 patients suffer from therapy-resistant seizures and comorbidities such as intellectual disability, developmental delay, and hypotonia. We tested RNAi gene therapy in the Dnm1 fitful mouse model of DEE using a Dnm1-targeted therapeutic microRNA delivered by a self-complementary adeno-associated virus vector. Untreated or control-injected fitful mice have growth delay, severe ataxia, and lethal tonic-clonic seizures by 3 weeks of age. These major impairments are mitigated following a single treatment in newborn mice, along with key underlying cellular features including gliosis, cell death, and aberrant neuronal metabolic activity typically associated with recurrent seizures. Our results underscore the potential for RNAi gene therapy to treat DNM1 disease and other genetic DEEs where treatment would require inhibition of the pathogenic gene product.
Collapse
Affiliation(s)
- Osasumwen V Aimiuwu
- Institute for Genomic Medicine and Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Allison M Fowler
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Megha Sah
- Institute for Genomic Medicine and Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Jia Jie Teoh
- Institute for Genomic Medicine and Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Ayla Kanber
- Institute for Genomic Medicine and Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Nettie K Pyne
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Sabrina Petri
- Institute for Genomic Medicine and Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Chana Rosenthal-Weiss
- Institute for Genomic Medicine and Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Mu Yang
- Institute for Genomic Medicine and Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Scott Q Harper
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA; Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43205, USA
| | - Wayne N Frankel
- Institute for Genomic Medicine and Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
58
|
Nusinersen ameliorates motor function and prevents motoneuron Cajal body disassembly and abnormal poly(A) RNA distribution in a SMA mouse model. Sci Rep 2020; 10:10738. [PMID: 32612161 PMCID: PMC7330045 DOI: 10.1038/s41598-020-67569-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 06/08/2020] [Indexed: 11/09/2022] Open
Abstract
Spinal muscular atrophy (SMA) is a devastating autosomal recessive neuromuscular disease characterized by degeneration of spinal cord alpha motor neurons (αMNs). SMA is caused by the homozygous deletion or mutation of the survival motor neuron 1 (SMN1) gene, resulting in reduced expression of SMN protein, which leads to αMN degeneration and muscle atrophy. The majority of transcripts of a second gene (SMN2) generate an alternative spliced isoform that lacks exon 7 and produces a truncated nonfunctional form of SMN. A major function of SMN is the biogenesis of spliceosomal snRNPs, which are essential components of the pre-mRNA splicing machinery, the spliceosome. In recent years, new potential therapies have been developed to increase SMN levels, including treatment with antisense oligonucleotides (ASOs). The ASO-nusinersen (Spinraza) promotes the inclusion of exon 7 in SMN2 transcripts and notably enhances the production of full-length SMN in mouse models of SMA. In this work, we used the intracerebroventricular injection of nusinersen in the SMN∆7 mouse model of SMA to evaluate the effects of this ASO on the behavior of Cajal bodies (CBs), nuclear structures involved in spliceosomal snRNP biogenesis, and the cellular distribution of polyadenylated mRNAs in αMNs. The administration of nusinersen at postnatal day (P) 1 normalized SMN expression in the spinal cord but not in skeletal muscle, rescued the growth curve and improved motor behavior at P12 (late symptomatic stage). Importantly, this ASO recovered the number of canonical CBs in MNs, significantly reduced the abnormal accumulation of polyadenylated RNAs in nuclear granules, and normalized the expression of the pre-mRNAs encoding chondrolectin and choline acetyltransferase, two key factors for αMN homeostasis. We propose that the splicing modulatory function of nusinersen in SMA αMN is mediated by the rescue of CB biogenesis, resulting in enhanced polyadenylated pre-mRNA transcription and splicing and nuclear export of mature mRNAs for translation. Our results support that the selective restoration of SMN expression in the spinal cord has a beneficial impact not only on αMNs but also on skeletal myofibers. However, the rescue of SMN expression in muscle appears to be necessary for the complete recovery of motor function.
Collapse
|
59
|
Gamdzyk M, Doycheva DM, Araujo C, Ocak U, Luo Y, Tang J, Zhang JH. cGAS/STING Pathway Activation Contributes to Delayed Neurodegeneration in Neonatal Hypoxia-Ischemia Rat Model: Possible Involvement of LINE-1. Mol Neurobiol 2020; 57:2600-2619. [PMID: 32253733 PMCID: PMC7260114 DOI: 10.1007/s12035-020-01904-7] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 03/19/2020] [Indexed: 12/12/2022]
Abstract
cGAS is a sensor of cytosolic DNA and responds equally to exogenous and endogenous DNA. After recognition of cytosolic dsDNA or ssDNA, cGAS synthesizes the second messenger 2'3'-cGAMP, which then binds to and activates stimulator of interferon genes (STING). STING plays an essential role in responding to pathogenic DNA and self-DNA in the context of autoimmunity. In pathologic conditions, such as stroke or hypoxia-ischemia (HI), DNA can gain access into the cytoplasm of the cell and leak from the dying cells into the extracellular environment, which potentially activates cGAS/STING. Recent in vivo studies of myocardial ischemia, traumatic brain injury, and liver damage models suggest that activation of cGAS/STING is not only a side-effect of the injury, but it can also actively contribute to cell death and apoptosis. We found, for the first time, that cGAS/STING pathway becomes activated between 24 and 48 h after HI in a 10-day-old rat model. Silencing STING with siRNA resulted in decreased infarction area, reduced cortical neurodegeneration, and improved neurobehavior at 48 h, suggesting that STING can contribute to injury progression after HI. STING colocalized with lysosomal marker LAMP-1 and blocking STING reduced the expression of cathepsin B and decreased the expression of Bax and caspase 3 cleavage. We observed similar protective effects after intranasal treatment with cGAS inhibitor RU.521, which were reversed by administration of STING agonist 2'3'-cGAMP. Additionally, we showed that long interspersed element 1 (LINE-1) retrotransposon, a potential upstream activator of cGAS/STING pathway was induced at 48 h after HI, which was evidenced by increased expression of ORF1p and ORF2p proteins and increased LINE-1 DNA content in the cytosol. Blocking LINE-1 with the nucleoside analog reverse-transcriptase inhibitor (NRTI) stavudine reduced infarction area, neuronal degeneration in the cerebral cortex, and reduced the expression of Bax and cleaved caspase 3. Thus, our results identify the cGAS/STING pathway as a potential therapeutic target to inhibit delayed neuronal death after HI.
Collapse
Affiliation(s)
- Marcin Gamdzyk
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, 11041 Campus St, Risley Hall, Room 219, Loma Linda, CA, 92354, USA
| | - Desislava Met Doycheva
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, 11041 Campus St, Risley Hall, Room 219, Loma Linda, CA, 92354, USA
| | - Camila Araujo
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, 11041 Campus St, Risley Hall, Room 219, Loma Linda, CA, 92354, USA
| | - Umut Ocak
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, 11041 Campus St, Risley Hall, Room 219, Loma Linda, CA, 92354, USA
- Department of Emergency Medicine, Bursa Yuksek Ihtisas Training and Research Hospital, University of Health Sciences, 16310, Bursa, Turkey
| | - Yujie Luo
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, 11041 Campus St, Risley Hall, Room 219, Loma Linda, CA, 92354, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, 11041 Campus St, Risley Hall, Room 219, Loma Linda, CA, 92354, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, 11041 Campus St, Risley Hall, Room 219, Loma Linda, CA, 92354, USA.
- Department of Anesthesiology, Neurosurgery and Neurology, Loma Linda University School of Medicine, 11041 Campus St, Risley Hall, Room 219, Loma Linda, CA, 92354, USA.
| |
Collapse
|
60
|
Sase S, Almad AA, Boecker CA, Guedes-Dias P, Li JJ, Takanohashi A, Patel A, McCaffrey T, Patel H, Sirdeshpande D, Curiel J, Shih-Hwa Liu J, Padiath Q, Holzbaur EL, Scherer SS, Vanderver A. TUBB4A mutations result in both glial and neuronal degeneration in an H-ABC leukodystrophy mouse model. eLife 2020; 9:52986. [PMID: 32463361 PMCID: PMC7255805 DOI: 10.7554/elife.52986] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 04/13/2020] [Indexed: 12/20/2022] Open
Abstract
Mutations in TUBB4A result in a spectrum of leukodystrophy including Hypomyelination with Atrophy of Basal Ganglia and Cerebellum (H-ABC), a rare hypomyelinating leukodystrophy, often associated with a recurring variant p.Asp249Asn (D249N). We have developed a novel knock-in mouse model harboring heterozygous (Tubb4aD249N/+) and the homozygous (Tubb4aD249N/D249N) mutation that recapitulate the progressive motor dysfunction with tremor, dystonia and ataxia seen in H-ABC. Tubb4aD249N/D249N mice have myelination deficits along with dramatic decrease in mature oligodendrocytes and their progenitor cells. Additionally, a significant loss occurs in the cerebellar granular neurons and striatal neurons in Tubb4aD249N/D249N mice. In vitro studies show decreased survival and dysfunction in microtubule dynamics in neurons from Tubb4aD249N/D249N mice. Thus Tubb4aD249N/D249N mice demonstrate the complex cellular physiology of H-ABC, likely due to independent effects on oligodendrocytes, striatal neurons, and cerebellar granule cells in the context of altered microtubule dynamics, with profound neurodevelopmental deficits.
Collapse
Affiliation(s)
- Sunetra Sase
- Department of Neurology, The Children's Hospital of Philadelphia, Philadelphia, United States
| | - Akshata A Almad
- Department of Neurology, The Children's Hospital of Philadelphia, Philadelphia, United States
| | - C Alexander Boecker
- Department of Physiology, the Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Pedro Guedes-Dias
- Department of Physiology, the Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Jian J Li
- Department of Neurology, the Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Asako Takanohashi
- Department of Neurology, The Children's Hospital of Philadelphia, Philadelphia, United States
| | - Akshilkumar Patel
- Department of Neurology, The Children's Hospital of Philadelphia, Philadelphia, United States
| | - Tara McCaffrey
- Department of Neurology, The Children's Hospital of Philadelphia, Philadelphia, United States
| | - Heta Patel
- Department of Neurology, The Children's Hospital of Philadelphia, Philadelphia, United States
| | - Divya Sirdeshpande
- Department of Neurology, The Children's Hospital of Philadelphia, Philadelphia, United States
| | - Julian Curiel
- Department of Neurology, The Children's Hospital of Philadelphia, Philadelphia, United States
| | - Judy Shih-Hwa Liu
- Department of Neurology, Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, United States
| | - Quasar Padiath
- Department of Human Genetics and Neurobiology, University of Pittsburgh, Pittsburgh, United States
| | - Erika Lf Holzbaur
- Department of Physiology, the Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Steven S Scherer
- Department of Neurology, the Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Adeline Vanderver
- Department of Neurology, The Children's Hospital of Philadelphia, Philadelphia, United States.,Department of Neurology, the Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| |
Collapse
|
61
|
Obuchowicz E, Bielecka-Wajdman A, Zieliński M, Machnik G, Gołyszny M, Ludyga T. Imipramine and Venlafaxine Differentially Affect Primary Glial Cultures of Prenatally Stressed Rats. Front Pharmacol 2020; 10:1687. [PMID: 32076407 PMCID: PMC7006619 DOI: 10.3389/fphar.2019.01687] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 12/24/2019] [Indexed: 12/25/2022] Open
Abstract
Here, we examine the effects of prenatal administration of two antidepressants—imipramine (IMI) and venlafaxine (VEN)—on morphology and activity of a primary glial culture. Microglia are targeted by antidepressants used for antenatal depression and are important regulators of central nervous system development. In this study, female Wistar rats were assigned to one of four groups: a control group that received water ad libitum (1), and groups that received additionally once daily either water (2), IMI (10 mg/kg) (3), or VEN (20 mg/kg) (4) by oral gavage from gestation day 7 to 22. Oral gavage administration induced prenatal stress. Cell cultures were obtained from the brains of 1-day-old pups. Prenatal stress caused a disturbance of sensorimotor function in pups. Prenatal stress also produced alterations in the glial cultures, specifically, an increased percentage of microglia in the mixed glial cultures and an increased percentage of dead cells. Moreover, increased levels of IL1-β, TNF-α, NO, and an increased expression of CX3CR1 mRNA were found in microglia. However, the ratio of Bax/Bcl2 mRNA was reduced. Prenatal stress increased the vulnerability of microglia to lipopolysaccharide (LPS). The mixed glial culture derived from pups exposed to IMI showed greater morphological changes and the highest percentage of microglia. Microglia were characterized by the largest increase in the production of pro-inflammatory cytokines and NO, and the greatest reduction in the expression of CX3CR1 mRNA. Exposure to IMI reduced the effects of LPS on IL-1β production and Bax/Bcl2 mRNA, and exacerbated the effects of LPS on CX3CR1 mRNA expression. Prenatal administration of VEN induced protective effects on microglia, as measured by all studied parameters. Taken together, our data suggest that, by disturbing microglia function, exposure to even mild forms of chronic prenatal stress may predispose individuals to psychiatric or neurodevelopmental disorders. These data also indicate that chronic mild stress sensitizes microglia to immune challenges, which may lead to enhanced neuronal damage in the embryonic brain. The observed detrimental effects of IMI on microglial activity under conditions of prenatal stress may help to explain the teratogenic effects of IMI reported in the literature.
Collapse
Affiliation(s)
- Ewa Obuchowicz
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Anna Bielecka-Wajdman
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Michał Zieliński
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Grzegorz Machnik
- Department of Internal Medicine and Clinical Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Miłosz Gołyszny
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Tomasz Ludyga
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| |
Collapse
|
62
|
Arakawa H. Sensorimotor developmental factors influencing the performance of laboratory rodents on learning and memory. Behav Brain Res 2019; 375:112140. [PMID: 31401145 PMCID: PMC6741784 DOI: 10.1016/j.bbr.2019.112140] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 07/31/2019] [Accepted: 08/01/2019] [Indexed: 02/08/2023]
Abstract
Behavioral studies in animal models have advanced our knowledge of brain function and the neural mechanisms of human diseases. Commonly used laboratory rodents, such as mice and rats, provide a useful tool for studying the behaviors and mechanisms associated with learning and memory processes which are cooperatively regulated by multiple underlying factors, including sensory and motor performance and emotional/defense innate components. Each of these factors shows unique ontogeny and governs the sustainment of behavioral performance in learning tasks, and thus, understanding the integrative processes of behavioral development are crucial in the accurate interpretation of the functional meaning of learning and memory behaviors expressed in commonly employed behavioral test paradigms. In this review, we will summarize the major findings in the developmental processes of rodent behavior on the basis of the emergence of fundamental components for sustaining learning and memory behaviors. Briefly, most sensory modalities (except for vision) and motor abilities are functional at the juvenile stage, in which several defensive components, including active and passive defensive strategies and risk assessment behavior, emerge. Sex differences are detectable from the juvenile stage through adulthood and are considerable factors that influence behavioral tests. The test paradigms addressed in this review include associative learning (with an emphasis on fear conditioning), spatial learning, and recognition. This basic background information will aid in accurately performing behavioral studies in laboratory rodents and will therefore contribute to reducing inappropriate interpretations of behavioral data and further advance research on learning and memory in rodent models.
Collapse
Affiliation(s)
- Hiroyuki Arakawa
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn St. HSF2/S251, Baltimore, MD, 21201, USA.
| |
Collapse
|
63
|
Janakiraman U, Yu J, Moutal A, Chinnasamy D, Boinon L, Batchelor SN, Anandhan A, Khanna R, Nelson MA. TAF1-gene editing alters the morphology and function of the cerebellum and cerebral cortex. Neurobiol Dis 2019; 132:104539. [PMID: 31344492 PMCID: PMC7197880 DOI: 10.1016/j.nbd.2019.104539] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 06/20/2019] [Accepted: 07/19/2019] [Indexed: 10/26/2022] Open
Abstract
TAF1/MRSX33 intellectual disability syndrome is an X-linked disorder caused by loss-of-function mutations in the TAF1 gene. How these mutations cause dysmorphology, hypotonia, intellectual and motor defects is unknown. Mouse models which have embryonically targeted TAF1 have failed, possibly due to TAF1 being essential for viability, preferentially expressed in early brain development, and intolerant of mutation. Novel animal models are valuable tools for understanding neuronal pathology. Here, we report the development and characterization of a novel animal model for TAF1 ID syndrome in which the TAF1 gene is deleted in embryonic rats using clustered regularly interspaced short palindromic repeats (CRISPR) associated protein 9 (Cas9) technology and somatic brain transgenesis mediated by lentiviral transduction. Rat pups, post-natal day 3, were subjected to intracerebroventricular (ICV) injection of either gRNA-control or gRNA-TAF1 vectors. Rats were subjected to a battery of behavioral tests followed by histopathological analyses of brains at post-natal day 14 and day 35. TAF1-edited rats exhibited behavioral deficits at both the neonatal and juvenile stages of development. Deletion of TAF1 lead to a hypoplasia and loss of the Purkinje cells. We also observed a decreased in GFAP positive astrocytes and an increase in Iba1 positive microglia within the granular layer of the cerebellum in TAF1-edited animals. Immunostaining revealed a reduction in the expression of the CaV3.1 T-type calcium channel. Abnormal motor symptoms in TAF1-edited rats were associated with irregular cerebellar output caused by changes in the intrinsic activity of the Purkinje cells due to loss of pre-synaptic CaV3.1. This animal model provides a powerful new tool for studies of neuronal dysfunction in conditions associated with TAF1 abnormalities and should prove useful for developing therapeutic strategies to treat TAF1 ID syndrome.
Collapse
Affiliation(s)
- Udaiyappan Janakiraman
- Department of Pathology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA
| | - Jie Yu
- Department of Pharmacology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA; College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou 310058, China
| | - Aubin Moutal
- Department of Pharmacology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA
| | - Dhanalakshmi Chinnasamy
- Department of Pathology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA
| | - Lisa Boinon
- Department of Pharmacology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA
| | - Shelby N Batchelor
- Department of Pathology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA
| | - Annaduri Anandhan
- Department of Pharmacology and Toxicology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA
| | - Rajesh Khanna
- Department of Pathology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA; Department of Pharmacology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA; The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, AZ, United States of America; The BIO5 Institute, University of Arizona, United States of America
| | - Mark A Nelson
- Department of Pathology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA.
| |
Collapse
|
64
|
Kadam P, Ntemou E, Onofre J, Van Saen D, Goossens E. Does co-transplantation of mesenchymal and spermatogonial stem cells improve reproductive efficiency and safety in mice? Stem Cell Res Ther 2019; 10:310. [PMID: 31640769 PMCID: PMC6805426 DOI: 10.1186/s13287-019-1420-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 09/03/2019] [Accepted: 09/16/2019] [Indexed: 12/12/2022] Open
Abstract
Background Spermatogonial stem cell transplantation (SSCT) is a promising therapy in restoring the fertility of childhood cancer survivors. However, the low efficiency of SSCT is a significant concern. SSCT could be improved by co-transplanting transforming growth factor beta 1 (TGFβ1)-induced mesenchymal stem cells (MSCs). In this study, we investigated the reproductive efficiency and safety of co-transplanting spermatogonial stem cells (SSCs) and TGFβ1-induced MSCs. Methods A mouse model for long-term infertility was used to transplant SSCs (SSCT, n = 10) and a combination of SSCs and TGFβ1-treated MSCs (MSi-SSCT, n = 10). Both transplanted groups and a fertile control group (n = 7) were allowed to mate naturally to check the reproductive efficiency after transplantation. Furthermore, the testes from transplanted males and donor-derived male offspring were analyzed for the epigenetic markers DNA methyltransferase 3A (DNMT3A) and histone 4 lysine 5 acetylation (H4K5ac). Results The overall tubular fertility index (TFI) after SSCT (76 ± 12) was similar to that after MSi-SSCT (73 ± 14). However, the donor-derived TFI after MSi-SSCT (26 ± 14) was higher compared to the one after SSCT (9 ± 5; P = 0.002), even after injecting half of the number of SSCs in MSi-SSCT. The litter sizes after SSCT (3.7 ± 3.7) and MSi-SSCT (3.7 ± 3.6) were similar but differed significantly with the control group (7.6 ± 1.0; P < 0.001). The number of GFP+ offspring per litter obtained after SSCT (1.6 ± 0.5) and MSi-SSCT (2.0 ± 1.0) was also similar. The expression of DNMT3A and H4K5ac in germ cells of transplanted males was found to be significantly reduced compared to the control group. However, in donor-derived offspring, DNMT3A and H4K5ac followed the normal pattern. Conclusion Co-transplanting SSCs and TGFβ1-treated MSCs results in reproductive efficiency as good as SSCT, even after transplanting half the number of SSCs. Although transplanted males showed lower expression of DNMT3A and H4K5ac in donor-derived germ cells, the expression was restored to normal levels in germ cells of donor-derived offspring. This procedure could become an efficient method to restore fertility in a clinical setup, but more studies are needed to ensure safety in the long term.
Collapse
Affiliation(s)
- Prashant Kadam
- Biology of the Testis (BITE) Laboratory, Department of Reproduction, Genetics and Regenerative Medicine, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Elissavet Ntemou
- Biology of the Testis (BITE) Laboratory, Department of Reproduction, Genetics and Regenerative Medicine, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Jaime Onofre
- Biology of the Testis (BITE) Laboratory, Department of Reproduction, Genetics and Regenerative Medicine, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Dorien Van Saen
- Biology of the Testis (BITE) Laboratory, Department of Reproduction, Genetics and Regenerative Medicine, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Ellen Goossens
- Biology of the Testis (BITE) Laboratory, Department of Reproduction, Genetics and Regenerative Medicine, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090, Brussels, Belgium.
| |
Collapse
|
65
|
Early skeletal muscle pathology and disease progress in the dy 3K/dy 3K mouse model of congenital muscular dystrophy with laminin α2 chain-deficiency. Sci Rep 2019; 9:14324. [PMID: 31586140 PMCID: PMC6778073 DOI: 10.1038/s41598-019-50550-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 09/12/2019] [Indexed: 11/09/2022] Open
Abstract
Deficiency of laminin α2 chain leads to a severe form of congenital muscular dystrophy (LAMA2-CMD), and dystrophic symptoms progress rapidly in early childhood. Currently, there is no treatment for this detrimental disorder. Development of therapies is largely hindered by lack of understanding of mechanisms involved in the disease initiation and progress, both in patients but also in mouse models that are commonly used in the preclinical setup. Here, we unveil the first pathogenic events and characterise the disease development in a mouse model for LAMA2-CMD (dy3K/dy3K), by analysing muscles at perinatal, neonatal and postnatal stages. We found that apoptotic muscle fibres were present as early as postnatal day 1. Other typical dystrophic hallmarks (muscle degeneration, inflammation, and extensive production of the extracellular matrix proteins) were clearly evident already at postnatal day 4, and the highest degree of muscle deterioration was reached by day 7. Interestingly, the severe phenotype of limb muscles partially recovered on days 14 and 21, despite worsening of the general condition of the dy3K/dy3K mouse by that age. We found that masticatory muscles were severely affected in dy3K/dy3K mice and this may be an underlying cause of their malnutrition, which contributes to death around day 21. We also showed that several signalling pathways were affected already in 1-day-old dy3K/dy3K muscle. Therapeutic tests in the dy3K/dy3K mouse model should therefore be initiated shortly after birth, but should also take into account timing and correlation between regenerative and pathogenic events.
Collapse
|
66
|
Hydrogen-rich saline promotes microglia M2 polarization and complement-mediated synapse loss to restore behavioral deficits following hypoxia-ischemic in neonatal mice via AMPK activation. J Neuroinflammation 2019; 16:104. [PMID: 31103039 PMCID: PMC6525972 DOI: 10.1186/s12974-019-1488-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 04/25/2019] [Indexed: 12/23/2022] Open
Abstract
Background Hypoxia-ischemia (HI) during the perinatal period is one of the most common causes of acute mortality and chronic neurologic morbidity. Hydrogen-rich saline (HS) treatment in neonatal mice has been reported to alleviate brain injury following HI, but the mechanisms involved are not known. Methods A modified version of the Rice-Vannucci method for the induction of neonatal HI brain injury was performed on postnatal day 7 mouse pups. Animals or BV2-cells received HS and an AMPK inhibitor at indicative time post-injury. Results In the current study, we show that HS treatment attenuated the accumulation of CD11b+/CD45high cells, suppressed HI-induced neuro-inflammation, induced microglial anti-inflammatory M2 polarization, was associated with promoting AMPK activation, and inhibited nuclear factor-κB activation as demonstrated both in vivo and in vitro. In addition, HS treatment reversed HI-induced neurological disabilities, was associated with improving damaged synapses, and restored the expression levels of synaptophysin and postsynaptic density protein 95 following HI insult. Furthermore, HI insult which increased levels of complement component C1q, C3, and C3aR1 was observed. Importantly, C1q deposited in the infarct core and lesion boundary zone following HI injury, was found to co-localize within regions of synapse loss, whereas HS treatment reversed these effects of HI on synapse loss and complement component levels. Notably, the AMPK inhibitor reversed the beneficial effects of HS as described above. Conclusions These results demonstrate that HS restored behavioral deficits after HI in neonatal mice. These beneficial effects, in part, involve promoting microglia M2 polarization and complement-mediated synapse loss via AMPK activation. Electronic supplementary material The online version of this article (10.1186/s12974-019-1488-2) contains supplementary material, which is available to authorized users.
Collapse
|
67
|
Naik AK, Pandey U, Mukherjee R, Mukhopadhyay S, Chakraborty S, Ghosh A, Aich P. Lactobacillus rhamnosus GG reverses mortality of neonatal mice against Salmonella challenge. Toxicol Res (Camb) 2019; 8:361-372. [PMID: 31160970 PMCID: PMC6505381 DOI: 10.1039/c9tx00006b] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 01/30/2019] [Indexed: 12/12/2022] Open
Abstract
Pathogenic infection is one of the major causes of death in newborns. Antibiotic based therapies are still the major mode of treatment for infection. Increased usage of antibiotics leads to selective evolution of microorganisms and causes diseases in adulthood. Attempts to develop alternatives to antibiotics did not yield much success. A recent viable trend is to identify novel probiotics that could alleviate problems associated with over usage of antibiotics. We screened three different Lactobacillus species to establish their efficacy in neonates in protecting against Salmonella challenge. The methodologies employed are metagenomics, metabonomics, transcriptional profiling, molecular assays and behavioral studies. Among the three probiotics used, only Lactobacillus rhamnosus GG (LGG) treatment of the neonates resulted in rescuing of 80% of the Salmonella-infected mice. We have shown that LGG (MTCC #1408) can prevent Salmonella mediated infection in neonates. In the current report, results from histopathology, gene expression, neutrophil infiltration, metabolite and metataxonomic profiling, and protein level data suggested that LGG treatment of the neonates enhanced anti-inflammatory cytokine expression and increased the gut barrier function. The current report establishes the potential use of LGG in clinical intervention of infectious diseases.
Collapse
Affiliation(s)
- Aman Kumar Naik
- School of Biological Sciences , National Institute of Science Education and Research (NISER) , HBNI , P.O. - Bhimpur-Padanpur , Jatni - 752050 , Dist. - Khurdha , Odisha , India . ; ; Tel: +916742494133
| | - Uday Pandey
- School of Biological Sciences , National Institute of Science Education and Research (NISER) , HBNI , P.O. - Bhimpur-Padanpur , Jatni - 752050 , Dist. - Khurdha , Odisha , India . ; ; Tel: +916742494133
| | - Raktim Mukherjee
- School of Biological Sciences , National Institute of Science Education and Research (NISER) , HBNI , P.O. - Bhimpur-Padanpur , Jatni - 752050 , Dist. - Khurdha , Odisha , India . ; ; Tel: +916742494133
| | - Sohini Mukhopadhyay
- School of Biological Sciences , National Institute of Science Education and Research (NISER) , HBNI , P.O. - Bhimpur-Padanpur , Jatni - 752050 , Dist. - Khurdha , Odisha , India . ; ; Tel: +916742494133
| | - Subhayan Chakraborty
- School of Chemical Sciences , National Institute of Science Education and Research (NISER) , HBNI , P.O. - Bhimpur-Padanpur , Jatni - 752050 , Dist. - Khurdha , Odisha , India
| | - Arindam Ghosh
- School of Chemical Sciences , National Institute of Science Education and Research (NISER) , HBNI , P.O. - Bhimpur-Padanpur , Jatni - 752050 , Dist. - Khurdha , Odisha , India
| | - Palok Aich
- School of Biological Sciences , National Institute of Science Education and Research (NISER) , HBNI , P.O. - Bhimpur-Padanpur , Jatni - 752050 , Dist. - Khurdha , Odisha , India . ; ; Tel: +916742494133
| |
Collapse
|
68
|
Jiang X, Liu H, Shao Y, Peng M, Zhang W, Li D, Li X, Cai Y, Tan T, Lu X, Xu J, Su X, Lin Y, Liu Z, Huang Y, Zeng C, Tang YP, Liu L. A novel GTPCH deficiency mouse model exhibiting tetrahydrobiopterin-related metabolic disturbance and infancy-onset motor impairments. Metabolism 2019; 94:96-104. [PMID: 30742839 DOI: 10.1016/j.metabol.2019.02.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/22/2019] [Accepted: 02/05/2019] [Indexed: 11/17/2022]
Abstract
BACKGROUND GTP cyclohydrolase I (GTPCH) deficiency could impair the synthesis of tetrahydrobiopterin and causes metabolic diseases involving phenylalanine catabolism, neurotransmitter synthesis, nitric oxide production and so on. Though improvements could be achieved by tetrahydrobiopterin and neurotransmitter precursor levodopa supplementation, residual motor and mental deficits remain in some patients. An appropriate GTPCH deficiency animal model with clinical symptoms, especially the motor impairments, is still not available for mechanism and therapy studies yet. OBJECTIVES AND METHODS To investigate whether the heterozygous GTPCH missense mutation p.Leu117Arg identified from a patient with severe infancy-onset dopa-responsive motor impairments is causative and establish a clinical relevant GTPCH deficiency mouse model, we generated a mouse mutant mimicking this missense mutation using the CRISPR/Cas9 technology. Series of characterization experiments on the heterozygous and homozygous mutants were conducted. RESULTS The expressions of GTPCH were not significantly changed in the mutants, but the enzyme activities were impaired in the homozygous mutants. BH4 reduction and phenylalanine accumulation were observed both in the liver and brain of the homozygous mutants. Severer metabolic disturbance occurred in the brain than in the liver. Significant reduction of neurotransmitter dopamine, norepinephrine and serotonin was observed in the brains of homozygous mutants. Live-born homozygous mutants exhibited infancy-onset motor and vocalization deficits similar to the disease symptoms observed in the patient, while no obvious symptoms were observed in the young heterozygous mutant mice. With benserazide-levodopa treatment, survival of the homozygous mutants was improved but not completely rescued. CONCLUSIONS The GTPCH p.Leu117Arg missense mutation is deleterious and could cause tetrahydrobiopterin, phenylalanine and neurotransmitter metabolic disturbances and infancy-onset motor dysfunctions recessively. This is the first GTPCH deficiency mouse model which could be live-born and exhibits significant motor impairments. The different extents of BH4 reduction and phenylalanine accumulation observed between liver and brain in response to GTPCH deficiency gives potential new insights into the vulnerability of brain to GTPCH deficiency.
Collapse
Affiliation(s)
- Xiaoling Jiang
- Department of Genetics and Endocrine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Huazhen Liu
- Department of Genetics and Endocrine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Yongxian Shao
- Department of Genetics and Endocrine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Mingzhi Peng
- Department of Genetics and Endocrine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Wen Zhang
- Department of Genetics and Endocrine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Duan Li
- Department of Genetics and Endocrine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Xiuzhen Li
- Department of Genetics and Endocrine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Yanna Cai
- Department of Genetics and Endocrine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Ting Tan
- Lab of Neural Development and Behavior Genetics, Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Xinshuo Lu
- Department of Genetics and Endocrine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Jianan Xu
- Department of Genetics and Endocrine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Xueying Su
- Department of Genetics and Endocrine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Yunting Lin
- Department of Genetics and Endocrine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Zongcai Liu
- Department of Genetics and Endocrine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Yonglan Huang
- Department of Neonatal Screening, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Chunhua Zeng
- Department of Genetics and Endocrine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Ya-Ping Tang
- Lab of Neural Development and Behavior Genetics, Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China.
| | - Li Liu
- Department of Genetics and Endocrine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China.
| |
Collapse
|
69
|
Exploring conditions that make cortical bone geometry optimal for physiological loading. Biomech Model Mechanobiol 2019; 18:1335-1349. [PMID: 30953214 DOI: 10.1007/s10237-019-01147-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 04/02/2019] [Indexed: 10/27/2022]
Abstract
While physiological loading on lower long bones changes during bone development, the bone cross section either remains circular or slowly changes from nearly circular to other shapes such as oval and roughly triangular. Bone is said to be an optimal structure, where strength is maximized using the optimal distribution of bone mass (also called Wolff's law). One of the most appropriate mathematical validations of this law would be a structural optimization-based formulation where total strain energy is minimized against a mass and a space constraint. Assuming that the change in cross section during bone development and homeostasis after adulthood is direct result of the change in physiological loading, this work investigates what optimization problem formulation (collectively, design variables, objective function, constraints, loading conditions, etc.) results in mathematically optimal solutions that resemble bones under actual physiological loading. For this purpose, an advanced structural optimization-based computational model for cortical bone development and defect repair is presented. In the optimization problem, overall bone stiffness is maximized first against a mass constraint, and then also against a polar first moment of area constraint that simultaneously constrains both mass and space. The investigation is completed in two stages. The first stage is developmental stage when physiological loading on lower long bones (tibia) is a random combination of axial, bending and torsion. The topology optimization applied to this case with the area moment constraint results into circular and elliptical cross sections similar to that found in growing mouse or human. The second investigation stage is bone homeostasis reached in adulthood when the physiological loading has a fixed pattern. A drill hole defect is applied to the adult mouse bone, which would disrupt the homeostasis. The optimization applied after the defect interestingly brings the damaged section back to the original intact geometry. The results, however, show that cortical bone geometry is optimal for the physiological loading only when there is also a constraint on polar moment of area. Further numerical experiments show that application of torsion along with the gait-analysis-based physiological loading improves the results, which seems to indicate that the cortical bone geometry is optimal for some amount of torsion in addition to the gait-based physiological loading. This work has a potential to be extended to bone growth/development models and fracture healing models, where topology optimization and polar moment of area constraint have not been introduced earlier.
Collapse
|
70
|
Buratti P, Covatti C, Centenaro LA, Brancalhão RMC, Torrejais MM. Morphofunctional characteristics of skeletal muscle in rats with cerebral palsy. Int J Exp Pathol 2019; 100:49-59. [PMID: 30773727 DOI: 10.1111/iep.12304] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 10/24/2018] [Accepted: 12/27/2018] [Indexed: 01/10/2023] Open
Abstract
Knowledge of skeletal muscle adaptations is important to understand the functional deficits in cerebral palsy (CP). This study aimed to investigate the morphofunctional characteristics of skeletal muscle in a CP animal model. Initially, pregnant Wistar rats were injected intraperitoneally with saline or lipopolysaccharide over the last five days of pregnancy. The control group (n = 8) consisted of male pups born to females injected with saline. The CP group (n = 8) consisted of male pups born to females injected with lipopolysaccharide, which were submitted to perinatal anoxia [day of birth, postnatal day 0 (P0)] and sensorimotor restriction (P1-P30). The open-field test was undertaken on P29 and P45. On P48, the animals were weighed, and the plantaris muscle was collected and its weight and length were measured. Transverse sections were stained with haematoxylin-eosin, NADH-TR, Masson's trichrome and non-specific esterase reaction for analysis. and transmission electron microscopy was performed. In the CP group, reductions were observed in mobility time, number of crossings and rearing frequency, body weight, muscle weight and length, and nucleus-to-fibre and capillary-to-fibre ratios. There was a statistically significant increase in the percentage area of the muscle section occupied by collagen; reduction in the area and increase in the number of type I muscle fibres; increase in myofibrillar disorganization and Z-line disorganization and dissolution; and reduction in the area and largest and smallest diameters of neuromuscular junctions. Thus this animal model of CP produced morphofunctional alterations in skeletal muscle, that were associated with evidence of motor deficits as demonstrated by the open-field test.
Collapse
Affiliation(s)
- Pâmela Buratti
- Programa de Pós-Graduação em Biociências e Saúde, Universidade Estadual do Oeste do Paraná - UNIOESTE, Cascavel, Paraná, Brazil
| | - Caroline Covatti
- Programa de Pós-Graduação em Biociências e Saúde, Universidade Estadual do Oeste do Paraná - UNIOESTE, Cascavel, Paraná, Brazil
| | - Lígia Aline Centenaro
- Centro de Ciências Médicas e Farmacêuticas, Universidade Estadual do Oeste do Paraná - UNIOESTE, Cascavel, Paraná, Brazil
| | - Rose Meire Costa Brancalhão
- Programa de Pós-Graduação em Biociências e Saúde, Universidade Estadual do Oeste do Paraná - UNIOESTE, Cascavel, Paraná, Brazil
| | - Marcia Miranda Torrejais
- Programa de Pós-Graduação em Biociências e Saúde, Universidade Estadual do Oeste do Paraná - UNIOESTE, Cascavel, Paraná, Brazil
| |
Collapse
|
71
|
Glycine Protects against Hypoxic-Ischemic Brain Injury by Regulating Mitochondria-Mediated Autophagy via the AMPK Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4248529. [PMID: 30881590 PMCID: PMC6381570 DOI: 10.1155/2019/4248529] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 08/10/2018] [Accepted: 09/14/2018] [Indexed: 12/25/2022]
Abstract
Hypoxic-ischemic encephalopathy (HIE) is detrimental to newborns and is associated with high mortality and poor prognosis. Thus, the primary aim of the present study was to determine whether glycine could (1) attenuate HIE injury in rats and hypoxic stress in PC12 cells and (2) downregulate mitochondria-mediated autophagy dependent on the adenosine monophosphate- (AMP-) activated protein kinase (AMPK) pathway. Experiments conducted using an in vivo HIE animal model and in vitro hypoxic stress to PC12 cells revealed that intense autophagy associated with mitochondrial function occurred during in vivo HIE injury and in vitro hypoxic stress. However, glycine treatment effectively attenuated mitochondria-mediated autophagy. Additionally, after identifying alterations in proteins within the AMPK pathway in rats and PC12 cells following glycine treatment, cyclosporin A (CsA) and 5-aminoimidazole-4-carboxamide-1-b-4-ribofuranoside (AICAR) were administered in these models and indicated that glycine protected against HIE and CoCl2 injury by downregulating mitochondria-mediated autophagy that was dependent on the AMPK pathway. Overall, glycine attenuated hypoxic-ischemic injury in neurons via reductions in mitochondria-mediated autophagy through the AMPK pathway both in vitro and in vivo.
Collapse
|
72
|
Effect of Maternal Exposure to Zinc Oxide Nanoparticles on Reflexive Motor Behaviors in Mice Offspring. Int J Pept Res Ther 2018. [DOI: 10.1007/s10989-018-9752-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
73
|
Mulder CL, Serrano JB, Catsburg LAE, Roseboom TJ, Repping S, van Pelt AMM. A practical blueprint to systematically study life-long health consequences of novel medically assisted reproductive treatments. Hum Reprod 2018; 33:784-792. [PMID: 29635479 PMCID: PMC5925779 DOI: 10.1093/humrep/dey070] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 02/27/2018] [Indexed: 01/27/2023] Open
Abstract
In medicine, safety and efficacy are the two pillars on which the implementation of novel treatments rest. To protect the patient from unnecessary or unsafe treatments, usually, a stringent path of (pre) clinical testing is followed before a treatment is introduced into routine patient care. However, in reproductive medicine several techniques have been clinically introduced without elaborate preclinical studies. Moreover, novel reproductive techniques may harbor safety risks not only for the patients undergoing treatment, but also for the offspring conceived through these techniques. If preclinical (animal) studies were performed, efficacy and functionality the upper hand. When a new medically assisted reproduction (MAR) treatment was proven effective (i.e. if it resulted in live birth) the treatment was often rapidly implemented in the clinic. For IVF, the first study on the long-term health of IVF children was published a decade after its clinical implementation. In more recent years, prospective follow-up studies have been conducted that provided the opportunity to study the health of large groups of children derived from different reproductive techniques. Although such studies have indicated differences between children conceived through MAR and children conceived naturally, results are often difficult to interpret due to the observational nature of these studies (and the associated risk of confounding factors, e.g. subfertility of the parents), differences in definitions of clinical outcome measures, lack of uniformity in assessment protocols and heterogeneity of the underlying reasons for fertility treatment. With more novel MARs waiting at the horizon, there is a need for a framework on how to assess safety of novel reproductive techniques in a preclinical (animal) setting before they are clinically implemented. In this article, we provide a blueprint for preclinical testing of safety and health of offspring generated by novel MARs using a mouse model involving an array of tests that comprise the entire lifespan. We urge scientists to perform the proposed extensive preclinical tests for novel reproductive techniques with the goal to acquire knowledge on efficacy and the possible health effects of to-be implemented reproductive techniques to safeguard quality of novel MARs.
Collapse
Affiliation(s)
- Callista L Mulder
- Center for Reproductive Medicine, Amsterdam Research Institute Reproduction and Development, Academic Medical Centre, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Joana B Serrano
- Center for Reproductive Medicine, Amsterdam Research Institute Reproduction and Development, Academic Medical Centre, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Lisa A E Catsburg
- Center for Reproductive Medicine, Amsterdam Research Institute Reproduction and Development, Academic Medical Centre, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Tessa J Roseboom
- Department of Obstetrics and Gynaecology, Amsterdam Reproduction and Development Research Institute, Academic Medical Centre, Meibergdeef 9, 1105 AZ, Amsterdam, The Netherlands
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam Public Health Research Institute, Academic Medical Centre, Meibergdeef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Sjoerd Repping
- Center for Reproductive Medicine, Amsterdam Research Institute Reproduction and Development, Academic Medical Centre, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Ans M M van Pelt
- Center for Reproductive Medicine, Amsterdam Research Institute Reproduction and Development, Academic Medical Centre, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|