1
|
Zhao J, Zhang J, Yang C, Yin L, Hou L, Jiang L. Sodium butyrate aids brain injury repair in neonatal rats. Open Life Sci 2025; 20:20221046. [PMID: 40291783 PMCID: PMC12032984 DOI: 10.1515/biol-2022-1046] [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: 06/18/2024] [Revised: 12/10/2024] [Accepted: 12/14/2024] [Indexed: 04/30/2025] Open
Abstract
The aim of this study is to investigate the effects and mechanism of action of sodium butyrate (SB) on brain injury repair in neonatal rats. 126 neonatal SD rats were randomly allocated to 7 groups, and necrotizing enterocolitis (NEC) and hypoxic-ischemic brain injury (HIBI) rat models were established. Hematoxylin and eosin staining showed that SB intervention alleviated intestinal and brain injuries in the HIBI + SB, NEC + SB, and NEC + HIBI + SB groups. Compared to the NEC and NEC + HIBI groups, the NEC + SB and NEC + HIBI + SB groups had significantly higher interleukin (IL)-10 and lower IL-17 levels (P < 0.05). Immunohistochemistry revealed increased Bcl-2 expression and decreased Bax expression in the NEC + SB and NEC + HIBI + SB groups compared to the NEC and NEC + HIBI groups in intestinal and brain tissues (P < 0.05). Compared to the control group (CG), gut microbiota diversity decreased in the HIBI, NEC, and NEC + HIBI groups, and increased significantly in the HIBI + SB, NEC + SB, and NEC + HIBI + SB groups. SB may alleviate brain injury by modulating gut microbiota, affecting IL-10 and IL-17 levels, and regulating Bcl-2 and Bax expression in intestinal and brain tissues.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Neonatology, Affiliated Hospital of North Sichuan Medical College, No. 1, Mao Yuan South Road, Shunqing District, Nanchong, Sichuan, 637000, China
| | - Jun Zhang
- Department of Neonatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Can Yang
- Department of Neonatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Linlin Yin
- Department of Neonatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Li Hou
- Department of Neonatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Lin Jiang
- Department of Neonatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| |
Collapse
|
2
|
Zhong J, Yu X, Lin Z. Phosphodiesterase 4 inhibition as a novel treatment for stroke. PeerJ 2025; 13:e18905. [PMID: 39897494 PMCID: PMC11786714 DOI: 10.7717/peerj.18905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 01/06/2025] [Indexed: 02/04/2025] Open
Abstract
The incidence of stroke ranks third among the leading causes of mortality worldwide. It has the characteristics of high morbidity, high disability rate and high recurrence rate. The current risk associated with stroke surgery is exceedingly high. It may potentially outweigh the benefits and fail to ameliorate the cerebral tissue damage following ischemia. Therefore, pharmacological intervention assumes paramount importance. The use of thrombolytic drugs is most common in the treatment of stroke; however, its efficacy is limited due to its time-sensitive nature and propensity for increased bleeding. Over the past few years, the treatment of stroke has witnessed a surge in interest towards neuroprotective drugs that possess the potential to enhance neurological function. The PDE4D gene has been demonstrated to have a positive correlation with the risk of ischemic stroke. Additionally, the utilization of phosphodiesterase 4 inhibitors can enhance synaptic plasticity within the neural circuitry, regulate cellular metabolism, and prevent secondary brain injury caused by impaired blood flow. These mechanisms collectively facilitate the recovery of functional neurons, thereby serving as potential therapeutic interventions. Therefore, the comprehensive investigation of phosphodiesterase 4 as an innovative pharmacological target for stroke injury provides valuable insights into the development of therapeutic interventions in stroke treatment. This review is intended for, but not limited to, pharmacological researchers, drug target researchers, neurologists, neuromedical researchers, and behavioral scientists.
Collapse
Affiliation(s)
- Jiahong Zhong
- Department of Clinical Pharmacy, Meizhou People’s Hospital, Meizhou, Guangdong, China
| | - Xihui Yu
- Department of Pharmacy, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Zhuomiao Lin
- Department of Clinical Pharmacy, Meizhou People’s Hospital, Meizhou, Guangdong, China
| |
Collapse
|
3
|
Zheng Y, Huang Z, Zhao Y, Huang L, Wang J, Li H, Chen X, Wang J, Xie J. Mechanism of ameliorating cerebral ischemia/reperfusion injury by antioxidant inhibition of autophagy based on network pharmacology and experimental verification. Aging (Albany NY) 2024; 16:7474-7486. [PMID: 38669115 PMCID: PMC11087111 DOI: 10.18632/aging.205773] [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: 11/20/2023] [Accepted: 03/18/2024] [Indexed: 04/28/2024]
Abstract
Cerebral ischemia-reperfusion injury (CIRI) is one of the most difficult challenges in cerebrovascular disease research. It is primarily caused by excessive autophagy induced by oxidative stress. Previously, a novel compound X5 was found, and the excellent antioxidant activity of it was verified in this study. Moreover, network pharmacological analysis suggested that compound X5 was closely associated with autophagy and the mTOR pathway. In vitro, X5 could significantly inhibit the expression of autophagy proteins Beclin-1 and LC3-β, which are induced by H2O2, and promote the expression of SIRT1. In vivo, compound X5 significantly reduced the infarct size and improved the neurological function scores in the middle cerebral artery occlusion (MCAO) model of rats. In conclusion, ROS-induced autophagy is closely related to mTOR, SIRT1 and others, and X5 holds promise as a candidate for the treatment of CIRI.
Collapse
Affiliation(s)
- Yuantie Zheng
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of the Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhicheng Huang
- Department of Pharmacy, Ezhou Central Hospital, Ezhou, Hubei, China
| | - Yang Zhao
- Department of Pharmacy, Guangyuan Central Hospital, Guangyuan, Sichuan, China
| | - Lili Huang
- Department of Pharmacy, Lihuili Hospital Affiliated to Ningbo University, Ningbo, Zhejiang, China
| | - Jun Wang
- Department of Pharmacy, Guangyuan Central Hospital, Guangyuan, Sichuan, China
| | - Heping Li
- Department of Pharmacy, Guangyuan Central Hospital, Guangyuan, Sichuan, China
| | - Xing Chen
- Department of Pharmacy, Guangyuan Central Hospital, Guangyuan, Sichuan, China
| | - Jingsong Wang
- Department of Pharmacy, Guangyuan Central Hospital, Guangyuan, Sichuan, China
| | - Jingwen Xie
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of the Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Health, Chongqing Industry and Trade Polytechnic, Chongqing, China
| |
Collapse
|
4
|
Di Martino E, Ambikan A, Ramsköld D, Umekawa T, Giatrellis S, Vacondio D, Romero AL, Galán MG, Sandberg R, Ådén U, Lauschke VM, Neogi U, Blomgren K, Kele J. Inflammatory, metabolic, and sex-dependent gene-regulatory dynamics of microglia and macrophages in neonatal hippocampus after hypoxia-ischemia. iScience 2024; 27:109346. [PMID: 38500830 PMCID: PMC10945260 DOI: 10.1016/j.isci.2024.109346] [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: 07/10/2023] [Revised: 01/02/2024] [Accepted: 02/22/2024] [Indexed: 03/20/2024] Open
Abstract
Neonatal hypoxia-ischemia (HI) is a major cause of perinatal death and long-term disabilities worldwide. Post-ischemic neuroinflammation plays a pivotal role in HI pathophysiology. In the present study, we investigated the temporal dynamics of microglia (CX3CR1GFP/+) and infiltrating macrophages (CCR2RFP/+) in the hippocampi of mice subjected to HI at postnatal day 9. Using inflammatory pathway and transcription factor (TF) analyses, we identified a distinct post-ischemic response in CCR2RFP/+ cells characterized by differential gene expression in sensome, homeostatic, matrisome, lipid metabolic, and inflammatory molecular signatures. Three days after injury, transcriptomic signatures of CX3CR1GFP/+ and CCR2RFP/+ cells isolated from hippocampi showed a partial convergence. Interestingly, microglia-specific genes in CX3CR1GFP/+ cells showed a sexual dimorphism, where expression returned to control levels in males but not in females during the experimental time frame. These results highlight the importance of further investigations on metabolic rewiring to pave the way for future interventions in asphyxiated neonates.
Collapse
Affiliation(s)
- Elena Di Martino
- Department of Women’s and Children’s Health, Karolinska Institutet, 17177 Stockholm, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, 58183 Linköping, Sweden
| | - Anoop Ambikan
- The Systems Virology Lab, Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, 14152 Huddinge, Sweden
| | - Daniel Ramsköld
- Department of Cell and Molecular Biology, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Takashi Umekawa
- Department of Women’s and Children’s Health, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Sarantis Giatrellis
- Department of Cell and Molecular Biology, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Davide Vacondio
- Department of Women’s and Children’s Health, Karolinska Institutet, 17177 Stockholm, Sweden
| | | | - Marta Gómez Galán
- Department of Physiology and Pharmacology, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Rickard Sandberg
- Department of Cell and Molecular Biology, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Ulrika Ådén
- Department of Women’s and Children’s Health, Karolinska Institutet, 17177 Stockholm, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, 58183 Linköping, Sweden
- Neonatology, Karolinska University Hospital, Stockholm, Sweden
| | - Volker M. Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, 17165 Stockholm, Sweden
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany
- University of Tuebingen, 72074 Tuebingen, Germany
| | - Ujjwal Neogi
- The Systems Virology Lab, Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, 14152 Huddinge, Sweden
| | - Klas Blomgren
- Department of Women’s and Children’s Health, Karolinska Institutet, 17177 Stockholm, Sweden
- Pediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Julianna Kele
- Department of Physiology and Pharmacology, Karolinska Institutet, 17165 Stockholm, Sweden
- Team Neurovascular Biology and Health, Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institutet, 14152 Huddinge, Sweden
| |
Collapse
|
5
|
Shabanov P, Samorodov A, Urakova N, Fisher E, Shchemeleva A. Low Fetal Resistance to Hypoxia as a Cause of Stillbirth and Neonatal Encephalopathy. CLIN EXP OBSTET GYN 2024; 51. [DOI: 10.31083/j.ceog5102033] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
Objective: Low fetal resistance to hypoxia is a factor in stillbirth and neonatal encephalopathy. This review examines fetal movement patterns in response to hypoxia as a predictor of the likelihood of stillbirth. Monitoring the dynamics of fetal movements during maternal apnea could allow the assessment of fetal resistance to hypoxia. The goal of this study is to describe the practical application of this method by doctors and pregnant women. Mechanism: We searched relevant keywords in the international scientific literature databases Scopus and Web of Science, as well as databases for patents granted in China, India, USA, Japan, Germany, Russia and other countries. Devices, drugs and medical technologies that provide diagnosis, modeling, prevention and treatment of intrauterine fetal hypoxia, stillbirth and neonatal encephalopathy were considered. Findings in Brief: During apnea by a pregnant woman in the second half of normal pregnancy, if the maximum duration of fetal immobility exceeds 30 seconds from the onset of breath-holding, then the fetus is considered to show good resistance to hypoxia, thus preserving its health and life during vaginal delivery. On the other hand, excessive fetal movements <10 seconds after the onset of apnea in a pregnant woman indicates low fetal resistance to hypoxia. When fetal resistance to hypoxia is low, there is no alternative to immediate cesarean section for the preservation of fetal life and health. Conclusions: The monitoring of fetal movements during apnea in pregnant women allows real-time assessment of fetal resistance to intrauterine hypoxia. Obtaining timely information on fetal resistance to hypoxia is critical for determining the optimal timing and type of delivery in order to prevent encephalopathy and stillbirth.
Collapse
Affiliation(s)
- Petr Shabanov
- Department of Psychopharmacology, Institute of Experimental Medicine, 197376 Saint Petersburg, Russia
| | - Aleksandr Samorodov
- Department of Pharmacology with a Course of Сlinical Pharmacology, Bashkir State Medical University, 450008 Ufa, Bashkortostan, Russia
| | - Natalya Urakova
- Department of Obstetrics and Gynecology, Izhevsk State Medical Academy, 426034 Izhevsk, Udmurt Republic, Russia
- Department of Experimental and Clinical Research and Inventive, Institute of Thermology, 426054 Izhevsk, Udmurt Republic, Russia
| | - Evgeny Fisher
- Department of Experimental and Clinical Research and Inventive, Institute of Thermology, 426054 Izhevsk, Udmurt Republic, Russia
| | - Albina Shchemeleva
- Department of Experimental and Clinical Research and Inventive, Institute of Thermology, 426054 Izhevsk, Udmurt Republic, Russia
| |
Collapse
|
6
|
Pluta R. The Dual Role of Autophagy in Postischemic Brain Neurodegeneration of Alzheimer's Disease Proteinopathy. Int J Mol Sci 2023; 24:13793. [PMID: 37762096 PMCID: PMC10530906 DOI: 10.3390/ijms241813793] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/02/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Autophagy is a self-defense and self-degrading intracellular system involved in the recycling and elimination of the payload of cytoplasmic redundant components, aggregated or misfolded proteins and intracellular pathogens to maintain cell homeostasis and physiological function. Autophagy is activated in response to metabolic stress or starvation to maintain homeostasis in cells by updating organelles and dysfunctional proteins. In neurodegenerative diseases, such as cerebral ischemia, autophagy is disturbed, e.g., as a result of the pathological accumulation of proteins associated with Alzheimer's disease and their structural changes. Postischemic brain neurodegeneration, such as Alzheimer's disease, is characterized by the accumulation of amyloid and tau protein. After cerebral ischemia, autophagy was found to be activated in neuronal, glial and vascular cells. Some studies have shown the protective properties of autophagy in postischemic brain, while other studies have shown completely opposite properties. Thus, autophagy is now presented as a double-edged sword with possible therapeutic potential in brain ischemia. The exact role and regulatory pathways of autophagy that are involved in cerebral ischemia have not been conclusively elucidated. This review aims to provide a comprehensive look at the advances in the study of autophagy behavior in neuronal, glial and vascular cells for ischemic brain injury. In addition, the importance of autophagy in neurodegeneration after cerebral ischemia has been highlighted. The review also presents the possibility of modulating the autophagy machinery through various compounds on the development of neurodegeneration after cerebral ischemia.
Collapse
Affiliation(s)
- Ryszard Pluta
- Department of Pathophysiology, Medical University of Lublin, 20-090 Lublin, Poland
| |
Collapse
|
7
|
Kanal HD, Levison SW. Neuroprotective Effects of Delayed TGF-β1 Receptor Antagonist Administration on Perinatal Hypoxic-Ischemic Brain Injury. Dev Neurosci 2023; 46:188-200. [PMID: 37348472 DOI: 10.1159/000531650] [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: 11/10/2022] [Accepted: 06/19/2023] [Indexed: 06/24/2023] Open
Abstract
Hypoxic-ischemic (HI) brain injury in neonatal encephalopathy triggers a wave of neuroinflammatory events attributed to causing the progressive degeneration and functional deficits seen weeks after the primary damage. The cellular processes mediating this prolonged neurodegeneration in HI injury are not sufficiently understood. Consequently, current therapies are not fully protective. In a recent study, we found significant improvements in neurologic outcomes when a small molecule antagonist for activin-like kinase 5 (ALK5), a transforming growth factor beta (TGF-β) receptor was used as a therapeutic in a rat model of moderate term HI. Here, we have extended those studies to a mouse preterm pup model of HI. For these studies, postnatal day 7 CD1 mice of both sexes were exposed to 35-40 min of HI. Beginning 3 days later, SB505124, the ALK5 receptor antagonist, was administered systemically through intraperitoneal injections performed every 12 h for 5 days. When evaluated 23 days later, SB505124-treated mice had ∼2.5-fold more hippocampal area and ∼2-fold more thalamic tissue. Approximately 90% of the ipsilateral hemisphere (ILH) was preserved in the SB505124-treated HI mice compared to the vehicle-treated HI mice, where the ILH was ∼60% of its normal size. SB505124 also preserved the subcortical white matter. SB505124 treatment preserved levels of aquaporin-4 and n-cadherin, key proteins associated with blood-brain barrier function. Importantly, SB505124 administration improved sensorimotor function as assessed by a battery of behavioral tests. Altogether, these data lend additional support to the conclusion that SB505124 is a candidate neuroprotective molecule that could be an effective treatment for HI-related encephalopathy in moderately injured preterm infants.
Collapse
Affiliation(s)
- Hur Dolunay Kanal
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Steven W Levison
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| |
Collapse
|
8
|
Choi SG, Shin J, Lee KY, Park H, Kim SI, Yi YY, Kim DW, Song HJ, Shin HJ. PINK1 siRNA-loaded poly(lactic-co-glycolic acid) nanoparticles provide neuroprotection in a mouse model of photothrombosis-induced ischemic stroke. Glia 2023; 71:1294-1310. [PMID: 36655313 DOI: 10.1002/glia.24339] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 12/14/2022] [Accepted: 01/05/2023] [Indexed: 01/20/2023]
Abstract
PTEN-induced kinase 1 (PINK1) is a well-known critical marker in the pathway for mitophagy regulation as well as mitochondrial dysfunction. Evidence suggests that mitochondrial dynamics and mitophagy flux play an important role in the development of brain damage from stroke pathogenesis. In this study, we propose a treatment strategy using nanoparticles that can control PINK1. We used a murine photothrombotic ischemic stroke (PTS) model in which clogging of blood vessels is induced with Rose Bengal (RB) to cause brain damage. We targeted PINK1 with poly(lactic-co-glycolic acid) (PLGA)-based nanoparticles loaded with PINK1 siRNA (PINK1 NPs). After characterizing siRNA loading in the nanoparticles, we assessed the efficacy of PINK1 NPs in mice with PTS using immunohistochemistry, 1% 2,3,5-triphenyltetrazolium chloride staining, measurement of motor dysfunction, and Western blot. PINK1 was highly expressed in microglia 24 h after PTS induction. PINK1 siRNA treatment increased phagocytic activity, migration, and expression of an anti-inflammatory state in microglia. In addition, the PLGA nanoparticles were selectively taken up by microglia and specifically regulated PINK1 expression in those cells. Treatment with PINK1 NPs prior to stroke induction reduced expression of mitophagy-inducing factors, infarct volume, and motor dysfunction in mice with photothrombotic ischemia. Experiments with PINK1-knockout mice and microglia depletion with PLX3397 confirmed a decrease in stroke-induced infarct volume and behavioral dysfunction. Application of nanoparticles for PINK1 inhibition attenuates RB-induced photothrombotic ischemic injury by inhibiting microglia responses, suggesting that a nanomedical approach targeting the PINK1 pathway may provide a therapeutic avenue for stroke treatment.
Collapse
Affiliation(s)
- Seung Gyu Choi
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea
- Department of Anatomy and Cell Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Juhee Shin
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea
- Department of Anatomy and Cell Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Ka Young Lee
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea
- Department of Anatomy and Cell Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Hyewon Park
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea
- Department of Anatomy and Cell Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Song I Kim
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea
- Department of Anatomy and Cell Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Yoon Young Yi
- Department of Pediatrics, College of Medicine, Hallym University and Gangdong Sacred Heart Hospital, Seoul, Republic of Korea
| | - Dong Woon Kim
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea
- Department of Anatomy and Cell Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Hee-Jung Song
- Department of Neurology, Chungnam National University Sejong Hospital and College of Medicine, Republic of Korea
| | - Hyo Jung Shin
- Brain Research Institute, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
9
|
Wu Y, Sun Y, Wang X, Zhu C. The Regulated Cell Death and Potential Interventions in Preterm Infants after Intracerebral Hemorrhage. Curr Neuropharmacol 2023; 21:1488-1503. [PMID: 36397619 PMCID: PMC10472811 DOI: 10.2174/1570159x21666221117155209] [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: 04/21/2022] [Revised: 07/14/2022] [Accepted: 07/18/2022] [Indexed: 11/21/2022] Open
Abstract
Intracerebral hemorrhage (ICH) in preterm infants is one of the major co-morbidities of preterm birth and is associated with long-term neurodevelopmental deficits. There are currently no widely accepted treatments to prevent ICH or therapies for the neurological sequelae. With studies broadening the scope of cell death, the newly defined concept of regulated cell death has enriched our understanding of the underlying mechanisms of secondary brain injury after ICH and has suggested potential interventions in preterm infants. In this review, we will summarize the current evidence for regulated cell death pathways in preterm infants after ICH, including apoptosis, necroptosis, pyroptosis, ferroptosis, autophagy, and PANoptosis as well as several potential intervention strategies that may protect the immature brain from secondary injury after ICH through regulating regulated cell death.
Collapse
Affiliation(s)
- Yanan Wu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience of Zhengzhou University, Zhengzhou 450052, China
| | - Yanyan Sun
- Department of Human Anatomy, School of Basic Medical Science, Zhengzhou University, Zhengzhou, China
| | - Xiaoyang Wang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience of Zhengzhou University, Zhengzhou 450052, China
- Centre for Perinatal Medicine and Health, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience of Zhengzhou University, Zhengzhou 450052, China
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
10
|
Miller JA, Drouet DE, Yermakov LM, Elbasiouny MS, Bensabeur FZ, Bottomley M, Susuki K. Distinct Changes in Calpain and Calpastatin during PNS Myelination and Demyelination in Rodent Models. Int J Mol Sci 2022; 23:15443. [PMID: 36499770 PMCID: PMC9737575 DOI: 10.3390/ijms232315443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 11/19/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Myelin forming around axons provides electrical insulation and ensures rapid and efficient transmission of electrical impulses. Disruptions to myelinated nerves often result in nerve conduction failure along with neurological symptoms and long-term disability. In the central nervous system, calpains, a family of calcium dependent cysteine proteases, have been shown to have a role in developmental myelination and in demyelinating diseases. The roles of calpains in myelination and demyelination in the peripheral nervous system remain unclear. Here, we show a transient increase of activated CAPN1, a major calpain isoform, in postnatal rat sciatic nerves when myelin is actively formed. Expression of the endogenous calpain inhibitor, calpastatin, showed a steady decrease throughout the period of peripheral nerve development. In the sciatic nerves of Trembler-J mice characterized by dysmyelination, expression levels of CAPN1 and calpastatin and calpain activity were significantly increased. In lysolecithin-induced acute demyelination in adult rat sciatic nerves, we show an increase of CAPN1 and decrease of calpastatin expression. These changes in the calpain-calpastatin system are distinct from those during central nervous system development or in acute axonal degeneration in peripheral nerves. Our results suggest that the calpain-calpastatin system has putative roles in myelination and demyelinating diseases of peripheral nerves.
Collapse
Affiliation(s)
- John A. Miller
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA
| | - Domenica E. Drouet
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA
| | - Leonid M. Yermakov
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA
| | - Mahmoud S. Elbasiouny
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA
| | - Fatima Z. Bensabeur
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA
| | - Michael Bottomley
- Department of Mathematics and Statistics, Wright State University, Dayton, OH 45435, USA
| | - Keiichiro Susuki
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA
| |
Collapse
|
11
|
He Y, Ying J, Tang J, Zhou R, Qu H, Qu Y, Mu D. Neonatal Arterial Ischaemic Stroke: Advances in Pathologic Neural Death, Diagnosis, Treatment, and Prognosis. Curr Neuropharmacol 2022; 20:2248-2266. [PMID: 35193484 PMCID: PMC9890291 DOI: 10.2174/1570159x20666220222144744] [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: 11/29/2021] [Revised: 02/04/2022] [Accepted: 02/18/2022] [Indexed: 12/29/2022] Open
Abstract
Neonatal arterial ischaemic stroke (NAIS) is caused by focal arterial occlusion and often leads to severe neurological sequelae. Neural deaths after NAIS mainly include necrosis, apoptosis, necroptosis, autophagy, ferroptosis, and pyroptosis. These neural deaths are mainly caused by upstream stimulations, including excitotoxicity, oxidative stress, inflammation, and death receptor pathways. The current clinical approaches to managing NAIS mainly focus on supportive treatments, including seizure control and anticoagulation. In recent years, research on the pathology, early diagnosis, and potential therapeutic targets of NAIS has progressed. In this review, we summarise the latest progress of research on the pathology, diagnosis, treatment, and prognosis of NAIS and highlight newly potential diagnostic and treatment approaches.
Collapse
Affiliation(s)
- Yang He
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Junjie Ying
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Jun Tang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Ruixi Zhou
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Haibo Qu
- Department of Radiology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yi Qu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Dezhi Mu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| |
Collapse
|
12
|
Li T, Sun Y, Zhang S, Xu Y, Li K, Xie C, Wang Y, Wang Y, Cao J, Wang X, Penninger JM, Kroemer G, Blomgren K, Zhu C. AIF Overexpression Aggravates Oxidative Stress in Neonatal Male Mice After Hypoxia-Ischemia Injury. Mol Neurobiol 2022; 59:6613-6631. [PMID: 35974295 PMCID: PMC9525408 DOI: 10.1007/s12035-022-02987-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 08/01/2022] [Indexed: 11/03/2022]
Abstract
There are sex differences in the severity, mechanisms, and outcomes of neonatal hypoxia-ischemia (HI) brain injury, and apoptosis-inducing factor (AIF) may play a critical role in this discrepancy. Based on previous findings that AIF overexpression aggravates neonatal HI brain injury, we further investigated potential sex differences in the severity and molecular mechanisms underlying the injury using mice that overexpress AIF from homozygous transgenes. We found that the male sex significantly aggravated AIF-driven brain damage, as indicated by the injury volume in the gray matter (2.25 times greater in males) and by the lost volume of subcortical white matter (1.71 greater in males) after HI. As compared to females, male mice exhibited more severe brain injury, correlating with reduced antioxidant capacities, more pronounced protein carbonylation and nitration, and increased neuronal cell death. Under physiological conditions (without HI), the doublecortin-positive area in the dentate gyrus of females was 1.15 times larger than in males, indicating that AIF upregulation effectively promoted neurogenesis in females in the long term. We also found that AIF stimulated carbohydrate metabolism in young males. Altogether, these findings corroborate earlier studies and further demonstrate that AIF is involved in oxidative stress, which contributes to the sex-specific differences observed in neonatal HI brain injury.
Collapse
Affiliation(s)
- Tao Li
- Henan Children's Neurodevelopment Engineering Research Center, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China.,Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, China.,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, 40530, Gothenburg, Sweden
| | - Yanyan Sun
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, China.,Department of Human Anatomy, School of Basic Medicine and Institute of Neuroscience, Zhengzhou University, Zhengzhou, 450052, China
| | - Shan Zhang
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, China.,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, 40530, Gothenburg, Sweden
| | - Yiran Xu
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, China.,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, 40530, Gothenburg, Sweden
| | - Kenan Li
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, China.,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, 40530, Gothenburg, Sweden
| | - Cuicui Xie
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, 40530, Gothenburg, Sweden
| | - Yong Wang
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, China.,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, 40530, Gothenburg, Sweden
| | - Yafeng Wang
- Henan Children's Neurodevelopment Engineering Research Center, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China.,Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, China.,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, 40530, Gothenburg, Sweden
| | - Jing Cao
- Department of Human Anatomy, School of Basic Medicine and Institute of Neuroscience, Zhengzhou University, Zhengzhou, 450052, China
| | - Xiaoyang Wang
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, China.,Centre of Perinatal Medicine and Health, Institute of Clinical Science, University of Gothenburg, 40530, Gothenburg, Sweden
| | - Josef M Penninger
- Institute of Molecular Biotechnology, Austrian Academy of Sciences, 1030, Vienna, Austria.,Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Guido Kroemer
- Centre de Recherche Des Cordeliers, Equipe Labellisée Par La Ligue Contre Le Cancer, Inserm U1138, Université de Paris Cité, Sorbonne Université, Institut Universitaire de France, Paris, France.,Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.,Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Klas Blomgren
- Pediatric Oncology, Karolinska University Hospital, Stockholm, Sweden.,Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, China. .,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, 40530, Gothenburg, Sweden. .,Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
13
|
Ding X, Pan T, Tian Q, Huang W, Hayashi LS, Liu Q, Li F, Xu LX, Miao P, Yang X, Sun B, Feng CX, Feng X, Li M, Huang J. Profiling Temporal Changes of the Pineal Transcriptomes at Single Cell Level Upon Neonatal HIBD. Front Cell Dev Biol 2022; 10:794012. [PMID: 35350377 PMCID: PMC8958010 DOI: 10.3389/fcell.2022.794012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 01/24/2022] [Indexed: 12/21/2022] Open
Abstract
Neonatal hypoxic-ischemic brain damage (HIBD) often results in various neurological deficits. Among them, a common, yet often neglected, symptom is circadian rhythm disorders. Previous studies revealed that the occurrence of cysts in the pineal gland, an organ known to regulate circadian rhythm, is associated with circadian problems in children with HIBD. However, the underlying mechanisms of pineal dependent dysfunctions post HIBD remain largely elusive. Here, by performing 10x single cell RNA sequencing, we firstly molecularly identified distinct pineal cell types and explored their transcriptome changes at single cell level at 24 and 72 h post neonatal HIBD. Bioinformatic analysis of cell prioritization showed that both subtypes of pinealocytes, the predominant component of the pineal gland, were mostly affected. We then went further to investigate how distinct pineal cell types responded to neonatal HIBD. Within pinealocytes, we revealed a molecularly defined β to α subtype conversion induced by neonatal HIBD. Within astrocytes, we discovered that all three subtypes responded to neonatal HIBD, with differential expression of reactive astrocytes markers. Two subtypes of microglia cells were both activated by HIBD, marked by up-regulation of Ccl3. Notably, microglia cells showed substantial reduction at 72 h post HIBD. Further investigation revealed that pyroptosis preferentially occurred in pineal microglia through NLRP3-Caspase-1-GSDMD signaling pathway. Taken together, our results delineated temporal changes of molecular and cellular events occurring in the pineal gland following neonatal HIBD. By revealing pyroptosis in the pineal gland, our study also provided potential therapeutic targets for preventing extravasation of pineal pathology and thus improving circadian rhythm dysfunction in neonates with HIBD.
Collapse
Affiliation(s)
- Xin Ding
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury, Children's Hospital of Soochow University, Suzhou, China
| | - Tao Pan
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury, Children's Hospital of Soochow University, Suzhou, China
| | - Qiuyan Tian
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, China
| | - Wenxi Huang
- Undergraduate Program, University of Virginia, Charlottesville, VA, United States
| | - Lauren S Hayashi
- IRTA Fellow, National Institutes of Health, Bethesda, MD, United States
| | - Qin Liu
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, China
| | - Fuyong Li
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, China
| | - Li-Xiao Xu
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, China
| | - Po Miao
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury, Children's Hospital of Soochow University, Suzhou, China
| | - Xiaofeng Yang
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury, Children's Hospital of Soochow University, Suzhou, China
| | - Bin Sun
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury, Children's Hospital of Soochow University, Suzhou, China
| | - Chen-Xi Feng
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, China
| | - Xing Feng
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury, Children's Hospital of Soochow University, Suzhou, China.,Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, China.,Undergraduate Program, University of Virginia, Charlottesville, VA, United States.,IRTA Fellow, National Institutes of Health, Bethesda, MD, United States.,School of Basic Medicine and Biological Sciences, Medical College of Soochow University, Suzhou, China
| | - Mei Li
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, China
| | - Jian Huang
- School of Basic Medicine and Biological Sciences, Medical College of Soochow University, Suzhou, China
| |
Collapse
|
14
|
Ovcjak A, Xiao A, Kim JS, Xu B, Szeto V, Turlova E, Abussaud A, Chen NH, Miller SP, Sun HS, Feng ZP. Ryanodine receptor inhibitor dantrolene reduces hypoxic-ischemic brain injury in neonatal mice. Exp Neurol 2022; 351:113985. [DOI: 10.1016/j.expneurol.2022.113985] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 01/07/2022] [Accepted: 01/13/2022] [Indexed: 11/04/2022]
|
15
|
Gupta R, Ambasta RK, Pravir Kumar. Autophagy and apoptosis cascade: which is more prominent in neuronal death? Cell Mol Life Sci 2021; 78:8001-8047. [PMID: 34741624 PMCID: PMC11072037 DOI: 10.1007/s00018-021-04004-4] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 10/16/2021] [Accepted: 10/20/2021] [Indexed: 02/06/2023]
Abstract
Autophagy and apoptosis are two crucial self-destructive processes that maintain cellular homeostasis, which are characterized by their morphology and regulated through signal transduction mechanisms. These pathways determine the fate of cellular organelle and protein involved in human health and disease such as neurodegeneration, cancer, and cardiovascular disease. Cell death pathways share common molecular mechanisms, such as mitochondrial dysfunction, oxidative stress, calcium ion concentration, reactive oxygen species, and endoplasmic reticulum stress. Some key signaling molecules such as p53 and VEGF mediated angiogenic pathway exhibit cellular and molecular responses resulting in the triggering of apoptotic and autophagic pathways. Herein, based on previous studies, we describe the intricate relation between cell death pathways through their common genes and the role of various stress-causing agents. Further, extensive research on autophagy and apoptotic machinery excavates the implementation of selective biomarkers, for instance, mTOR, Bcl-2, BH3 family members, caspases, AMPK, PI3K/Akt/GSK3β, and p38/JNK/MAPK, in the pathogenesis and progression of neurodegenerative diseases. This molecular phenomenon will lead to the discovery of possible therapeutic biomolecules as a pharmacological intervention that are involved in the modulation of apoptosis and autophagy pathways. Moreover, we describe the potential role of micro-RNAs, long non-coding RNAs, and biomolecules as therapeutic agents that regulate cell death machinery to treat neurodegenerative diseases. Mounting evidence demonstrated that under stress conditions, such as calcium efflux, endoplasmic reticulum stress, the ubiquitin-proteasome system, and oxidative stress intermediate molecules, namely p53 and VEGF, activate and cause cell death. Further, activation of p53 and VEGF cause alteration in gene expression and dysregulated signaling pathways through the involvement of signaling molecules, namely mTOR, Bcl-2, BH3, AMPK, MAPK, JNK, and PI3K/Akt, and caspases. Alteration in gene expression and signaling cascades cause neurotoxicity and misfolded protein aggregates, which are characteristics features of neurodegenerative diseases. Excessive neurotoxicity and misfolded protein aggregates lead to neuronal cell death by activating death pathways like autophagy and apoptosis. However, autophagy has a dual role in the apoptosis pathways, i.e., activation and inhibition of the apoptosis signaling. Further, micro-RNAs and LncRNAs act as pharmacological regulators of autophagy and apoptosis cascade, whereas, natural compounds and chemical compounds act as pharmacological inhibitors that rescue neuronal cell death through inhibition of apoptosis and autophagic cell death.
Collapse
Affiliation(s)
- Rohan Gupta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Mechanical Engineering Building, Delhi Technological University (Formerly Delhi College of Engineering), Room# FW4TF3, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Mechanical Engineering Building, Delhi Technological University (Formerly Delhi College of Engineering), Room# FW4TF3, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Mechanical Engineering Building, Delhi Technological University (Formerly Delhi College of Engineering), Room# FW4TF3, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India.
- , Delhi, India.
| |
Collapse
|
16
|
Fuente-García C, Aldai N, Sentandreu E, Oliván M, Franco D, García-Torres S, R Barron LJ, Sentandreu MÁ. Caspase activity in post mortem muscle and its relation to cattle handling practices. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2021; 101:6258-6264. [PMID: 33937991 DOI: 10.1002/jsfa.11293] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 04/20/2021] [Accepted: 05/03/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Animal handling practices are one of the factors majorly affecting animal metabolism prior to slaughter. This phenomenon increases the occurrence of meat quality defects such as dark cutting-beef, causing high economical losses in the meat industry. Under this framework, the assessment of apoptosis onset in post mortem muscle was proposed as a novel approach to reveal biochemical characteristics in several Spanish bovine breeds (Asturiana de los Valles, Retinta and Rubia Gallega) managed under different production systems (intensive versus semi-extensive) and transport/lairage conditions (mixing versus not mixing with unfamiliar animals). To do so, the activities of initiator caspase 9 and executioner caspases 3/7 were determined in Longissimus thoracis et lumborum muscle at three early post mortem times (2, 8, and 24 h). RESULTS Breed effect and transport/lairage conditions were the most relevant factors that influenced both caspase activities over post mortem time, showing Rubia Gallega breed a completely different behavior compared to Asturiana de los Valles and Retinta breeds. Moreover, it is postulated that apoptosis cascade is initiated via the activation of caspase 9 under hypoxic or metabolic stress followed by the activation of executioner caspases 3/7. CONCLUSIONS Assessment of apoptosis on post mortem muscle can be a novel approach to study the influence of animal handling on muscle metabolism and post mortem cell death and its consequences on meat quality traits. © 2021 The Authors. Journal of The Science of Food and Agriculture published by John Wiley & Sons Ltd on behalf of Society of Chemical Industry.
Collapse
Affiliation(s)
- Claudia Fuente-García
- Instituto de Agroquímica y Tecnología de Alimentos (CSIC), Calle del Catedràtic Agustín Escardino Benlloch 7, Paterna, Valencia, 46980, Spain
- Lactiker Research Group, Department of Pharmacy and Food Sciences, University of the Basque Country (UPV-EHU), Paseo de la Universidad 7, Vitoria-Gasteiz, 01006, Spain
| | - Noelia Aldai
- Lactiker Research Group, Department of Pharmacy and Food Sciences, University of the Basque Country (UPV-EHU), Paseo de la Universidad 7, Vitoria-Gasteiz, 01006, Spain
| | - Enrique Sentandreu
- Instituto de Agroquímica y Tecnología de Alimentos (CSIC), Calle del Catedràtic Agustín Escardino Benlloch 7, Paterna, Valencia, 46980, Spain
| | - Mamen Oliván
- Servicio Regional de Investigación y Desarrollo Alimentario (SERIDA), Apdo 13, Villaviciosa, Asturias, 33300, Spain
| | - Daniel Franco
- Centro Tecnológico de la Carne (CETECA), Rúa Galicia 4, San Cibrao das Viñas, Ourense, 32900, Spain
| | - Susana García-Torres
- Centro de Investigaciones Científicas y Tecnológicas de Extremadura (CICYTEX), Autovía A5, P.K. 372, Guadajira, Badajoz, 06187, Spain
| | - Luis Javier R Barron
- Lactiker Research Group, Department of Pharmacy and Food Sciences, University of the Basque Country (UPV-EHU), Paseo de la Universidad 7, Vitoria-Gasteiz, 01006, Spain
| | - Miguel Ángel Sentandreu
- Instituto de Agroquímica y Tecnología de Alimentos (CSIC), Calle del Catedràtic Agustín Escardino Benlloch 7, Paterna, Valencia, 46980, Spain
| |
Collapse
|
17
|
Nie BX, Zhao G, Yuan XF, Yu LX, Zhang J, Yuan Y, Liu Y, Hu J, Song E, Zhou YC, Shu J. Inhibition of CDK1 attenuates neuronal apoptosis and autophagy and confers neuroprotection after chronic spinal cord injury in vivo. J Chem Neuroanat 2021; 119:102053. [PMID: 34839004 DOI: 10.1016/j.jchemneu.2021.102053] [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: 08/15/2021] [Revised: 11/18/2021] [Accepted: 11/21/2021] [Indexed: 10/19/2022]
Abstract
Chronic spinal cord injury (CSCI) results from progressive compression of the spinal cord over time. A variety of factors cause CSCI, and its exact pathogenesis is unknown. Cyclin-dependent kinase 1 (CDK1) is closely related to the apoptosis pathway, but no CSCI-related studies on CDK1 have been conducted. In this study, the role of CDK1 in CSCI was explored in a rat model. The CSCI model was established by screw compression using the cervical anterior approach for twelve weeks. The neurological function of the rats was evaluated using the neurological severity scores (NSS) and motor evoked potentials (MEPs). Pathological changes in spinal cord tissue were observed by hematoxylin-eosin (HE) staining, and Nissl staining was performed to assess the survival of motor neurons in the anterior horn of the spinal cord. Changes in autophagy and apoptosis in anterior horn of spinal cord tissue were detected using transmission electron microscopy and the terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay, respectively. The expression levels of glial fibrillary acidic protein (GFAP), ionized calcium-binding adaptor (IBA) and choline acetyltransferase (CHAT) in the anterior horn were determined using immunohistochemistry assays to investigate astrocytes, microglia and motor neurons, respectively, in the anterior horn. Western blot assays were used to detect the expression levels of CDK1, Bcl-2, Bax, Caspase 3, LC3 and Beclin1. Changes in the expression of CDK1, LC3 and Beclin1 were also observed using immunohistochemistry. The results indicated that CSCI resulted in neuronal injury and a decrease in the NSS. In the CSCI model group, anterior horn astrocytes and microglia were activated, and motor neurons were decreased. Neuronal apoptosis was promoted, and the number of autophagic vacuoles was elevated. Rats treated with the CDK1 shRNA lentivirus exhibited better NSS, more surviving motor neurons, and fewer apoptotic neurons than the model rats. The occurrence of autophagy and the expression of proapoptotic and autophagy-related proteins were lower in the CDK1 shRNA group than the model group. In conclusion, CDK1 downregulation suppressed the activation of anterior horn astrocytes and microglia, promoted motor neuron repair, and inhibited neurons apoptosis and autophagy to promote the recovery of motor function after spinal cord injury.
Collapse
Affiliation(s)
- Bang-Xu Nie
- Traumatology Surgery, Second Affiliated Hospital of Kunming Medical University, Kunming 650106, Yunnan, China
| | - Gang Zhao
- Traumatology Surgery, Second Affiliated Hospital of Kunming Medical University, Kunming 650106, Yunnan, China
| | - Xiao-Feng Yuan
- Department of Orthopedics, Affiliated Calmette Hospital of Kunming Medical University, Kunming 650224, Yunnan, China
| | - Lin-Xin Yu
- Department of Orthopedics, Affiliated Calmette Hospital of Kunming Medical University, Kunming 650224, Yunnan, China
| | - Jin Zhang
- Department of Orthopedics, Affiliated Calmette Hospital of Kunming Medical University, Kunming 650224, Yunnan, China
| | - Yong Yuan
- Traumatology Surgery, Second Affiliated Hospital of Kunming Medical University, Kunming 650106, Yunnan, China
| | - Yao Liu
- College of Rehabilitation, Kunming Medical University, Kunming 650504, Yunnan, China
| | - Jun Hu
- Department of Orthopedics, Affiliated Calmette Hospital of Kunming Medical University, Kunming 650224, Yunnan, China
| | - En Song
- Department of Sports Medicine, First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Yu-Cheng Zhou
- Department of Orthopedics, Yunnan Provincial Rehabilitation Center for the Disabled Persons, Kunming 650034, Yunnan, China
| | - Jun Shu
- Traumatology Surgery, Second Affiliated Hospital of Kunming Medical University, Kunming 650106, Yunnan, China.
| |
Collapse
|
18
|
Davidson JO, Gonzalez F, Gressens P, Gunn AJ. Update on mechanisms of the pathophysiology of neonatal encephalopathy. Semin Fetal Neonatal Med 2021; 26:101267. [PMID: 34274259 DOI: 10.1016/j.siny.2021.101267] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Therapeutic hypothermia is now well established to significantly improve survival without disability after neonatal encephalopathy (NE). To further improve outcomes, we need to better understand the mechanisms of brain injury. The central finding, which offers the potential for neuroprotective and neurorestorative interventions, is that brain damage after perinatal hypoxia-ischemia evolves slowly over time. Although brain cells may die during profound hypoxia-ischemia, even after surprisingly severe insults many cells show transient recovery of oxidative metabolism during a "latent" phase characterized by actively suppressed neural metabolism and activity. Critically, after moderate to severe hypoxia-ischemia, this transient recovery is followed after ~6 h by a phase of secondary deterioration, with delayed seizures, failure of mitochondrial function, cytotoxic edema, and cell death over ~72 h. This is followed by a tertiary phase of remodeling and recovery. This review discusses the mechanisms of injury that occur during the primary, latent, secondary and tertiary phases of injury and potential treatments that target one or more of these phases. By analogy with therapeutic hypothermia, treatment as early as possible in the latent phase is likely to have the greatest potential to prevent injury ("neuroprotection"). In the secondary phase of injury, anticonvulsants can attenuate seizures, but show limited neuroprotection. Encouragingly, there is now increasing preclinical evidence that late, neurorestorative interventions have potential to improve long-term outcomes.
Collapse
Affiliation(s)
- Joanne O Davidson
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand.
| | - Fernando Gonzalez
- Department of Pediatrics, University of California, San Francisco, CA, USA.
| | | | - Alistair J Gunn
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand.
| | | |
Collapse
|
19
|
Kolmogorova D, Ah-Yen EG, Taylor BC, Vaggas T, Liang J, Davis T, Ismail N. Sex-specific responses of the pubertal neuroimmune axis in CD-1 mice. Brain Behav Immun Health 2021; 13:100229. [PMID: 34589744 PMCID: PMC8474685 DOI: 10.1016/j.bbih.2021.100229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 02/18/2021] [Indexed: 12/30/2022] Open
Abstract
The mechanistic relationship between the sexually dimorphic neuroimmune system and the sex-specific outcomes of a pubertal immune challenge is unclear. Therefore, we examined sex differences in the progression of cytotoxic microglial responses and blood-brain barrier (BBB) disruption to a peripubertal lipopolysaccharide (LPS) treatment in brain regions relevant to stress responses and cognitive function. Six-week-old (i.e., stress-sensitive pubertal period) male and female CD-1 mice were treated with LPS (1.5 mg/kg body weight, ip) or 0.9% saline (LPS-matched volume, ip). Sex and treatment differences in microglial (Iba1+) and apoptotic neuronal (caspase-3+/NeuN+) and non-neuronal (caspase-3+/NeuN−) expression were examined in the hippocampus, medial prefrontal cortex (mPFC), and paraventricular nucleus 24 h (sickness), one week (symptomatic recovery) and four weeks (early adulthood) post-treatment (n = 8/group). Microglial morphology was quantified with fractal analyses. Group differences in BBB permeability to 14C-sucrose were examined 24 h (whole-brain, hippocampus, prefrontal cortex, hypothalamus, and cerebellum) and one week (whole-brain) post-treatment. The acute effects of pubertal LPS were specific to females (i.e., global BBB disruption, altered microglial expression and morphology in the mPFC and hippocampus, increased hippocampal apoptosis). The residual effects of pubertal LPS-induced sickness observed in microglia persisted into adulthood in a sex- and region-specific manner. In addition to highlighting these sex-specific responses of the pubertal neuroimmune system, we report baseline region-specific sex differences in microglia spanning puberty through adulthood. We propose that these sex differences in neuroimmune-neurovascular interactions during the stress-sensitive pubertal period create sex biases in stress-related disorders of brain and behaviour. Pubertal LPS alters baseline sex differences in microglial numbers and morphology. Pubertal CD-1 mice mount sexually dimorphic neuroimmune responses to systemic LPS. Treatment effects on microglial expression and morphology differ by sex and region. The acute LPS-induced effects were specific to females.
Collapse
Affiliation(s)
- Daria Kolmogorova
- NISE Laboratory, School of Psychology, University of Ottawa, Ottawa, Ontario, Canada
| | - Emily Grace Ah-Yen
- NISE Laboratory, School of Psychology, University of Ottawa, Ottawa, Ontario, Canada
| | | | - Tiffany Vaggas
- School of Biosciences, Cardiff University, Cardiff, Wales, United Kingdom
| | - Jacky Liang
- NISE Laboratory, School of Psychology, University of Ottawa, Ottawa, Ontario, Canada
| | - Tama Davis
- NISE Laboratory, School of Psychology, University of Ottawa, Ottawa, Ontario, Canada
| | - Nafissa Ismail
- NISE Laboratory, School of Psychology, University of Ottawa, Ottawa, Ontario, Canada.,Brain and Mind Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
20
|
Orekhova K, Mazzariol S, Sussan B, Bucci M, Bonsembiante F, Verin R, Centelleghe C. Immunohistochemical Markers of Apoptotic and Hypoxic Damage Facilitate Evidence-Based Assessment in Pups with Neurological Disorders. Vet Sci 2021; 8:203. [PMID: 34679033 PMCID: PMC8537515 DOI: 10.3390/vetsci8100203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/14/2021] [Accepted: 09/18/2021] [Indexed: 12/15/2022] Open
Abstract
Seizures in puppies often present a diagnostic challenge in terms of identifying and treating the underlying cause. Dog breeds with mutations of the MDR1-gene are known to show adverse reactions to certain drugs, yet metabolic imbalance exacerbated by physiologically immature organs and other contributing pathologies require consideration before arriving at a diagnosis. This study analysed the brains of two male, 5-week-old Australian Shepherd siblings that died after displaying severe neurological symptoms upon administration of MilproVet® to treat severe intestinal helminth infection. Despite the initial symptoms being similar, their case histories varied in terms of the symptom duration, access to supportive therapy and post-mortem interval. Histopathology and immunohistochemistry were used to obtain more information about the phase of the pathological processes in the brain, employing protein markers associated with acute hypoxic damage (β-amyloid precursor protein/APP) and apoptosis (diacylglycerolkinase-ζ/DGK-ζ, apoptotic protease activating factor 1/Apaf1, and B-cell lymphoma related protein 2/Bcl-2). The results seem to reflect the course of the animals' clinical deterioration, implicating that the hypoxic damage to the brains was incompatible with life, and suggesting the usefulness of the mentioned immunohistochemical markers in clarifying the cause of death in animals with acute neurological deficits.
Collapse
Affiliation(s)
- Ksenia Orekhova
- Department of Comparative Biomedicine and Food Science, University of Padova AGRIPOLIS, viale dell’Università 16, 35020 Legnaro, Italy; (S.M.); (F.B.); (R.V.); (C.C.)
| | - Sandro Mazzariol
- Department of Comparative Biomedicine and Food Science, University of Padova AGRIPOLIS, viale dell’Università 16, 35020 Legnaro, Italy; (S.M.); (F.B.); (R.V.); (C.C.)
| | - Beatrice Sussan
- Department of Animal Medicine, Production and Health, University of Padova AGRIPOLIS, viale dell’Università 16, 35020 Legnaro, Italy; (B.S.); (M.B.)
| | - Massimo Bucci
- Department of Animal Medicine, Production and Health, University of Padova AGRIPOLIS, viale dell’Università 16, 35020 Legnaro, Italy; (B.S.); (M.B.)
| | - Federico Bonsembiante
- Department of Comparative Biomedicine and Food Science, University of Padova AGRIPOLIS, viale dell’Università 16, 35020 Legnaro, Italy; (S.M.); (F.B.); (R.V.); (C.C.)
- Department of Animal Medicine, Production and Health, University of Padova AGRIPOLIS, viale dell’Università 16, 35020 Legnaro, Italy; (B.S.); (M.B.)
| | - Ranieri Verin
- Department of Comparative Biomedicine and Food Science, University of Padova AGRIPOLIS, viale dell’Università 16, 35020 Legnaro, Italy; (S.M.); (F.B.); (R.V.); (C.C.)
| | - Cinzia Centelleghe
- Department of Comparative Biomedicine and Food Science, University of Padova AGRIPOLIS, viale dell’Università 16, 35020 Legnaro, Italy; (S.M.); (F.B.); (R.V.); (C.C.)
| |
Collapse
|
21
|
Samaiya PK, Krishnamurthy S, Kumar A. Mitochondrial dysfunction in perinatal asphyxia: role in pathogenesis and potential therapeutic interventions. Mol Cell Biochem 2021; 476:4421-4434. [PMID: 34472002 DOI: 10.1007/s11010-021-04253-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 08/20/2021] [Indexed: 01/13/2023]
Abstract
Perinatal asphyxia (PA)-induced brain injury may present as hypoxic-ischemic encephalopathy in the neonatal period, and long-term sequelae such as spastic motor deficits, intellectual disability, seizure disorders and learning disabilities. The brain injury is secondary to both the hypoxic-ischemic event and oxygenation-reperfusion following resuscitation. Following PA, a time-dependent progression of neuronal insult takes place in terms of transition of cell death from necrosis to apoptosis. This transition is the result of time-dependent progression of pathomechanisms which involve excitotoxicity, oxidative stress, and ultimately mitochondrial dysfunction in developing brain. More precisely mitochondrial respiration is suppressed and calcium signalling is dysregulated. Consequently, Bax-dependent mitochondrial permeabilization occurs leading to release of cytochrome c and activation of caspases leading to transition of cell death in developing brain. The therapeutic window lies within this transition process. At present, therapeutic hypothermia (TH) is the only clinical treatment available for treating moderate as well as severe asphyxia in new-born as it attenuates secondary loss of high-energy phosphates (ATP) (Solevåg et al. in Free Radic Biol Med 142:113-122, 2019; Gunn et al. in Pediatr Res 81:202-209, 2017), improving both short- and long-term outcomes. Mitoprotective therapies can offer a new avenue of intervention alone or in combination with therapeutic hypothermia for babies with birth asphyxia. This review will explore these mitochondrial pathways, and finally will summarize past and current efforts in targeting these pathways after PA, as a means of identifying new avenues of therapeutic intervention.
Collapse
Affiliation(s)
- Puneet K Samaiya
- Department of Pharmacy, Shri G.S. Institute of Technology and Science, Indore, MP, 452003, India.
| | - Sairam Krishnamurthy
- Neurotherapeutics Lab, Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ashok Kumar
- Department of Pediatrics, Institute of Medical Sciences, BHU, Varanasi, UP, India
| |
Collapse
|
22
|
Ajoolabady A, Wang S, Kroemer G, Penninger JM, Uversky VN, Pratico D, Henninger N, Reiter RJ, Bruno A, Joshipura K, Aslkhodapasandhokmabad H, Klionsky DJ, Ren J. Targeting autophagy in ischemic stroke: From molecular mechanisms to clinical therapeutics. Pharmacol Ther 2021; 225:107848. [PMID: 33823204 PMCID: PMC8263472 DOI: 10.1016/j.pharmthera.2021.107848] [Citation(s) in RCA: 174] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 03/23/2021] [Accepted: 04/01/2021] [Indexed: 01/18/2023]
Abstract
Stroke constitutes the second leading cause of death and a major cause of disability worldwide. Stroke is normally classified as either ischemic or hemorrhagic stroke (HS) although 87% of cases belong to ischemic nature. Approximately 700,000 individuals suffer an ischemic stroke (IS) in the US each year. Recent evidence has denoted a rather pivotal role for defective macroautophagy/autophagy in the pathogenesis of IS. Cellular response to stroke includes autophagy as an adaptive mechanism that alleviates cellular stresses by removing long-lived or damaged organelles, protein aggregates, and surplus cellular components via the autophagosome-lysosomal degradation process. In this context, autophagy functions as an essential cellular process to maintain cellular homeostasis and organismal survival. However, unchecked or excessive induction of autophagy has been perceived to be detrimental and its contribution to neuronal cell death remains largely unknown. In this review, we will summarize the role of autophagy in IS, and discuss potential strategies, particularly, employment of natural compounds for IS treatment through manipulation of autophagy.
Collapse
Affiliation(s)
- Amir Ajoolabady
- University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | - Shuyi Wang
- University of Wyoming College of Health Sciences, Laramie, WY 82071, USA; School of Medicine Shanghai University, Shanghai 200444, China
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France; Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France; Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China; Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Vienna, Austria; Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Vladimir N Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA; Institute for Biological Instrumentation of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino, Moscow region 142290, Russia
| | - Domenico Pratico
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Nils Henninger
- Department of Neurology, University of Massachusetts, Worcester, Massachusetts, USA; Department of Psychiatry, University of Massachusetts, Worcester, Massachusetts, USA
| | - Russel J Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Askiel Bruno
- Department of Neurology, Medical College of Georgia, Augusta University, GA 30912, USA
| | - Kaumudi Joshipura
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Center for Clinical Research and Health Promotion, University of Puerto Rico Medical Sciences Campus, San Juan, PR 00936-5067, Puerto Rico
| | | | - Daniel J Klionsky
- Life Sciences Institute and Departments of Molecular, Cellular and Developmental Biology and Biological Chemistry, University of Michigan, Ann Arbor 48109, USA.
| | - Jun Ren
- Department of Laboratory Medicine and Pathology, University of Washington Seattle, Seattle, WA 98195, USA; Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
23
|
Winstanley EL, Mahoney JJ, Castillo F, Comer SD. Neurocognitive impairments and brain abnormalities resulting from opioid-related overdoses: A systematic review. Drug Alcohol Depend 2021; 226:108838. [PMID: 34271512 PMCID: PMC8889511 DOI: 10.1016/j.drugalcdep.2021.108838] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/05/2021] [Accepted: 05/07/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Non-fatal opioid-related overdoses have increased significantly over the past two decades and there have been increasing reports of brain injuries and/or neurocognitive impairments following overdose events. Limited preclinical research suggests that opioid overdoses may cause brain injury; however, little is known about such injuries in humans. The purpose this systematic review is to summarize existing studies on neurocognitive impairments and/or brain abnormalities associated with an opioid-related overdose in humans. METHODS PubMed, Web of Science, Ovid MEDLINE and PsyINFO were searched, without year restrictions, and identified 3099 articles. An additional 24 articles were identified by reviewing references. Articles were included if they were published in English, reported study findings in humans, included individuals 18 years of age or older, and reported an objective measure of neurocognitive impairments and/or brain abnormalities resulting from an opioid-related overdose. Six domains of bias (selection, performance, attrition, detection (two dimensions) and reporting were evaluated and themes were summarized. RESULTS Seventy-nine journal articles, published between 1973-2020, were included in the review. More than half of the articles were case reports (n = 44) and there were 11 cohort studies, 18 case series, and 6 case-control studies. All of the studies were categorized as at-risk of bias, few controlled for confounding factors, and methodological differences made direct comparisons difficult. Less than half of the studies reported toxicology results confirming an opioid-related overdose; 64.6 % reported brain MRI results and 27.8 % reported results of neuropsychological testing. Only two studies had within subject comparative data to document changes in the brain possibly associated with an overdose. Despite these limitations, existing publications suggest that brain injuries and neurocognitive impairments are associated with opioid overdose. Additional research is needed to establish the incidence of overdose-related brain injuries and the potential impact on functioning, as well as engagement in treatment of substance use disorders. CONCLUSIONS Respiratory depression is a defining characteristic of opioid overdose and prolonged cerebral hypoxia may cause brain injuries and/or neurocognitive impairments. The onset, characteristics, and duration of such injuries is variable and additional research is needed to understand their clinical implications.
Collapse
Affiliation(s)
- Erin L. Winstanley
- Department of Behavioral Medicine and Psychiatry, School of Medicine and Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA,Department of Neuroscience, West Virginia University, Morgantown, WV, USA,Corresponding author at: West Virginia University, School of Medicine, Rockefeller Neuroscience Institute, Department of Behavioral Medicine and Psychiatry, 930 Chestnut Ridge Road, Morgantown, WV 26505, USA. (E.L. Winstanley)
| | - James J. Mahoney
- Department of Behavioral Medicine and Psychiatry, School of Medicine and Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA,Department of Neuroscience, West Virginia University, Morgantown, WV, USA
| | - Felipe Castillo
- Columbia University, Department of Psychiatry and New York State Psychiatric Institute, New York, NY, USA
| | - Sandra D. Comer
- Columbia University, Department of Psychiatry and New York State Psychiatric Institute, New York, NY, USA
| |
Collapse
|
24
|
Umbilical cord blood therapy modulates neonatal hypoxic ischemic brain injury in both females and males. Sci Rep 2021; 11:15788. [PMID: 34349144 PMCID: PMC8338979 DOI: 10.1038/s41598-021-95035-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 07/20/2021] [Indexed: 12/12/2022] Open
Abstract
Preclinical and clinical studies have shown that sex is a significant risk factor for perinatal morbidity and mortality, with males being more susceptible to neonatal hypoxic ischemic (HI) brain injury. No study has investigated sexual dimorphism in the efficacy of umbilical cord blood (UCB) cell therapy. HI injury was induced in postnatal day 10 (PND10) rat pups using the Rice-Vannucci method of carotid artery ligation. Pups received 3 doses of UCB cells (PND11, 13, 20) and underwent behavioural testing. On PND50, brains were collected for immunohistochemical analysis. Behavioural and neuropathological outcomes were assessed for sex differences. HI brain injury resulted in a significant decrease in brain weight and increase in tissue loss in females and males. Females and males also exhibited significant cell death, region-specific neuron loss and long-term behavioural deficits. Females had significantly smaller brains overall compared to males and males had significantly reduced neuron numbers in the cortex compared to females. UCB administration improved multiple aspects of neuropathology and functional outcomes in males and females. Females and males both exhibited injury following HI. This is the first preclinical evidence that UCB is an appropriate treatment for neonatal brain injury in both female and male neonates.
Collapse
|
25
|
Gao L, Yang L, Cui H. GSK-3β inhibitor TWS119 alleviates hypoxic-ischemic brain damage via a crosstalk with Wnt and Notch signaling pathways in neonatal rats. Brain Res 2021; 1768:147588. [PMID: 34310937 DOI: 10.1016/j.brainres.2021.147588] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 07/15/2021] [Accepted: 07/19/2021] [Indexed: 02/06/2023]
Abstract
Preterm infant brain injury is a leading cause of morbidity and disability in survivors of preterm infants. Unfortunately, the effective treatment remains absent. Recent evidence suggests that GSK-3β inhibitor TWS119 has a neuroprotectiverole in adult brain injury by activation of Wnt/β-catenin signaling pathway. However, the role on neonatal brain injury is not yet explored. The study aims to evaluate the effect of TWS119 at 7 d after hypoxic-ischemic brain damage and investigate the mechanism that it regulates Wnt and Notch signaling pathways at 24 h after hypoxic-ischemic brain damage in neonatal rats. Three-day-old rats were randomly divided into 3 groups: sham group, HI group and TWS119 group. The neonatal rats were subjected to left carotid artery ligation followed by 2 h of hypoxia (8.0% O2). A single dose of TWS119 (30 mg/kg) was intraperitoneally injected 20 min prior to hypoxia-ischemia (HI). At 7 d after HI, TWS119 improved the tissue structure, reduced cell apoptosis, up-regulated bcl-2 expression, up-regulated the expression of PSD-95 and Synapsin-1. At 24 h after HI, it activated Wnt/β-catenin signaling pathway by up-regulation of β-catenin protein expression and wnt3a/wnt5a/wnt7a mRNA expression. Simultaneously, it suppressed Notch signaling pathway by down-regulation of Notch1 and HES-1 proteins expression. Our study suggested that TWS119 performed a neuroprotective function at 7 d after hypoxic-ischemic brain damage via a crosstalk with Wnt/β-catenin and Notch signaling pathways at 24 h after hypoxic-ischemic brain damage in neonatal rats.
Collapse
Affiliation(s)
- Limin Gao
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, No. 95 Yongan Road, Xicheng District, Beijing 100050, China
| | - Lijun Yang
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, No. 95 Yongan Road, Xicheng District, Beijing 100050, China.
| | - Hong Cui
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, No. 95 Yongan Road, Xicheng District, Beijing 100050, China.
| |
Collapse
|
26
|
Li K, van Delft MF, Dewson G. Too much death can kill you: inhibiting intrinsic apoptosis to treat disease. EMBO J 2021; 40:e107341. [PMID: 34037273 DOI: 10.15252/embj.2020107341] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/11/2021] [Accepted: 03/18/2021] [Indexed: 02/06/2023] Open
Abstract
Apoptotic cell death is implicated in both physiological and pathological processes. Since many types of cancerous cells intrinsically evade apoptotic elimination, induction of apoptosis has become an attractive and often necessary cancer therapeutic approach. Conversely, some cells are extremely sensitive to apoptotic stimuli leading to neurodegenerative disease and immune pathologies. However, due to several challenges, pharmacological inhibition of apoptosis is still only a recently emerging strategy to combat pathological cell loss. Here, we describe several key steps in the intrinsic (mitochondrial) apoptosis pathway that represent potential targets for inhibitors in disease contexts. We also discuss the mechanisms of action, advantages and limitations of small-molecule and peptide-based inhibitors that have been developed to date. These inhibitors serve as important research tools to dissect apoptotic signalling and may foster new treatments to reduce unwanted cell loss.
Collapse
Affiliation(s)
- Kaiming Li
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Royal Parade, Melbourne, VIC, Australia
| | - Mark F van Delft
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Royal Parade, Melbourne, VIC, Australia
| | - Grant Dewson
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Royal Parade, Melbourne, VIC, Australia
| |
Collapse
|
27
|
Different types of cell death in vascular diseases. Mol Biol Rep 2021; 48:4687-4702. [PMID: 34013393 DOI: 10.1007/s11033-021-06402-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/08/2021] [Indexed: 10/21/2022]
Abstract
In a mature organism, tissue homeostasis is regulated by cell division and cell demise as the two major physiological procedures. There is increasing evidence that deregulation of these processes is important in the pathogenicity of main diseases, including myocardial infarction, stroke, atherosclerosis, and inflammatory diseases. Therefore, there are ongoing efforts to discover modulating factors of the cell cycle and cell demise planners aiming at shaping innovative therapeutically modalities to the therapy of such diseases. Although the life of a cell is terminated by several modes of action, a few cell deaths exist-some of which resemble apoptosis and/or necrosis, and most of them are different from one another-that contribute to a wide range of functions to either support or disrupt the homoeostasis. Even in normal physiological conditions, cell life is severe within the cardiovascular system. Cells are persistently undergoing stretch, contraction, injurious metabolic byproducts, and hemodynamic forces, and a few of cells sustain decade-long lifetimes. The duration of vascular disease causes further exposure of vascular cells to a novel range of offences, most of which induce cell death. There is growing evidence on consequences of direct damage to a cell, as well as on responses of adjacent and infiltrating cells, which also have an effect on the pathology. In this study, by focusing on different pathways of cell death in different vascular diseases, an attempt is made to open a new perspective on the therapeutic goals associated with cell death in these diseases.
Collapse
|
28
|
Radiation Triggers a Dynamic Sequence of Transient Microglial Alterations in Juvenile Brain. Cell Rep 2021; 31:107699. [PMID: 32492415 DOI: 10.1016/j.celrep.2020.107699] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 10/08/2019] [Accepted: 05/06/2020] [Indexed: 11/21/2022] Open
Abstract
Cranial irradiation (IR), an effective tool to treat malignant brain tumors, triggers a chronic pro-inflammatory microglial response, at least in the adult brain. Using single-cell and bulk RNA sequencing, combined with histology, we show that the microglial response in the juvenile mouse hippocampus is rapid but returns toward normal within 1 week. The response is characterized by a series of temporally distinct homeostasis-, sensome-, and inflammation-related molecular signatures. We find that a single microglial cell simultaneously upregulates transcripts associated with pro- and anti-inflammatory microglial phenotypes. Finally, we show that juvenile and adult irradiated microglia are already transcriptionally distinct in the early phase after IR. Our results indicate that microglia are involved in the initial stages but may not be responsible for driving long-term inflammation in the juvenile brain.
Collapse
|
29
|
Valencia M, Kim SR, Jang Y, Lee SH. Neuronal Autophagy: Characteristic Features and Roles in Neuronal Pathophysiology. Biomol Ther (Seoul) 2021; 29:605-614. [PMID: 33875624 PMCID: PMC8551733 DOI: 10.4062/biomolther.2021.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/02/2021] [Accepted: 03/23/2021] [Indexed: 11/12/2022] Open
Abstract
Autophagy is an important degradative pathway that eliminates misfolded proteins and damaged organelles from cells. Autophagy is crucial for neuronal homeostasis and function. A lack of or deficiency in autophagy leads to the accumulation of protein aggregates, which are associated with several neurodegenerative diseases. Compared with non-neuronal cells, neurons exhibit rapid autophagic flux because damaged organelles or protein aggregates cannot be diluted in post-mitotic cells; because of this, these cells exhibit characteristic features of autophagy, such as compartment-specific autophagy, which depends on polarized structures and rapid autophagy flux. In addition, neurons exhibit compartment-specific autophagy, which depends on polarized structures. Neuronal autophagy may have additional physiological roles other than amino acid recycling. In this review, we focus on the characteristics and regulatory factors of neuronal autophagy. We also describe intracellular selective autophagy in neurons and its association with neurodegenerative diseases.
Collapse
Affiliation(s)
- McNeil Valencia
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Sung Rae Kim
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Yeseul Jang
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Sung Hoon Lee
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| |
Collapse
|
30
|
Lee SH, Shim KS, Kim CY, Park TK. Characterization of the role of autophagy in retinal ganglion cell survival over time using a rat model of chronic ocular hypertension. Sci Rep 2021; 11:5767. [PMID: 33707562 PMCID: PMC7952572 DOI: 10.1038/s41598-021-85181-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 02/22/2021] [Indexed: 12/14/2022] Open
Abstract
Autophagy is an essential cellular process for the degradation and recycling of cellular components, and its dysregulation has been linked to neuronal cell death and neurodegeneration. In glaucoma, the role of autophagy in retinal ganglion cell (RGC) survival remains contradictory. Moreover, the effects of autophagy modulation at different time-points on RGC survival in a glaucoma model have not been investigated. In this study, we assessed the time-dependent role of autophagy in RGC survival in a circumlimbal suture-induced ocular hypertensive (OHT) rat model. Intraocular pressure (IOP) elevation led to a gradual autophagy induction, which reached a maximum between 1 and 4 weeks after OHT induction. On the other hand, early autophagy was impaired between 1 and 3 days after circumlimbal suturing, indicated by increased p62 levels due to reduced autophagosomal turnover. The intravitreal administration of rapamycin at different time-points after the application of the circumlimbal suture indicated that autophagy induction early during OHT development had potent survival-promoting effects in RGCs. In conclusion, our findings suggest that the role of autophagy in RGCs during OHT development might differ in a time-dependent manner. Modulating autophagy at the appropriate time might serve as a potential therapeutic approach to enhance RGC survival in OHT.
Collapse
Affiliation(s)
- Si Hyung Lee
- Department of Ophthalmology, College of Medicine, Soonchunhyang University, Cheonan, Republic of Korea
- Department of Ophthalmology, Soonchunhyang University Hospital Bucheon, 170 Jomaru-ro, Wonmi-gu, Bucheon, 14584, Republic of Korea
| | - Kyung Sun Shim
- Department of Ophthalmology, College of Medicine, Soonchunhyang University, Cheonan, Republic of Korea
- Department of Ophthalmology, Soonchunhyang University Hospital Bucheon, 170 Jomaru-ro, Wonmi-gu, Bucheon, 14584, Republic of Korea
| | - Chan Yun Kim
- Institute of Vision Research, Department of Ophthalmology, College of Medicine, Severance Hospital, Yonsei University, Seoul, South Korea
| | - Tae Kwann Park
- Department of Ophthalmology, College of Medicine, Soonchunhyang University, Cheonan, Republic of Korea.
- Department of Ophthalmology, Soonchunhyang University Hospital Bucheon, 170 Jomaru-ro, Wonmi-gu, Bucheon, 14584, Republic of Korea.
| |
Collapse
|
31
|
Zhang B, Ran Y, Wu S, Zhang F, Huang H, Zhu C, Zhang S, Zhang X. Inhibition of Colony Stimulating Factor 1 Receptor Suppresses Neuroinflammation and Neonatal Hypoxic-Ischemic Brain Injury. Front Neurol 2021; 12:607370. [PMID: 33679579 PMCID: PMC7930561 DOI: 10.3389/fneur.2021.607370] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 01/28/2021] [Indexed: 12/17/2022] Open
Abstract
Hypoxic-ischemic (HI) brain injury is a major cause of neonatal death or lifetime disability without widely accepted effective pharmacological treatments. It has been shown that the survival of microglia requires colony-stimulating factor 1 receptor (CSF1R) signaling and microglia participate in neonatal HI brain injury. We therefore hypothesize that microglia depletion during a HI insult period could reduce immature brain injury. In this study, CD1 mouse pups were treated with a CSF1R inhibitor (PLX3397, 25 mg/kg/daily) or a vehicle from postnatal day 4 to day 11 (P4-11), and over 90% of total brain microglia were deleted at P9. Unilateral hemisphere HI injury was induced at P9 by permanently ligating the left common carotid arteries and exposing the pups to 10% oxygen for 30 min to produce moderate left hemisphere injury. We found that the PLX3397 treatment reduced HI brain injury by 46.4%, as evaluated by the percentage of brain infarction at 48 h after HI. Furthermore, CSF1R inhibition suppressed the infiltration of neutrophils (69.7% reduction, p = 0.038), macrophages (77.4% reduction, p = 0.009), and T cells (72.9% reduction, p = 0.008) to the brain, the production of cytokines and chemokines (such as CCL12, CCL6, CCL21, CCL22, CCL19, IL7, CD14, and WISP-1), and reduced neuronal apoptosis as indicated by active caspase-3 labeled cells at 48 h after HI (615.20 ± 156.84/mm2 vs. 1,205.00 ± 99.15/mm2, p = 0.013). Our results suggest that CSF1R inhibition suppresses neuroinflammation and neonatal brain injury after acute cerebral hypoxia-ischemia in neonatal mice.
Collapse
Affiliation(s)
- Bohao Zhang
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Center of Advanced Analysis & Gene Sequencing, Zhengzhou University, Zhengzhou, China
| | - Yunwei Ran
- Medical Research Center, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Siting Wu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Fang Zhang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Huachen Huang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Clinical Neuroscience, Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Shusheng Zhang
- Center of Advanced Analysis & Gene Sequencing, Zhengzhou University, Zhengzhou, China
| | - Xiaoan Zhang
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
32
|
Manual Kollareth DJ, Zirpoli H, Ten VS, Deckelbaum RJ. Acute Injection of Omega-3 Triglyceride Emulsion Provides Very Similar Protection as Hypothermia in a Neonatal Mouse Model of Hypoxic-Ischemic Brain Injury. Front Neurol 2021; 11:618419. [PMID: 33519700 PMCID: PMC7843448 DOI: 10.3389/fneur.2020.618419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/21/2020] [Indexed: 11/17/2022] Open
Abstract
Therapeutic hypothermia (HT) is a currently accepted treatment for neonatal asphyxia and is a promising strategy in adult stroke therapy. We previously reported that acute administration of docosahexaenoic acid (DHA) triglyceride emulsion (tri-DHA) protects against hypoxic-ischemic (HI) injury in neonatal mice. We questioned if co-treatment with HT and tri-DHA would achieve synergic effects in protecting the brain from HI injury. Neonatal mice (10-day old) subjected to HI injury were placed in temperature-controlled chambers for 4 h of either HT (rectal temperature 31–32°C) or normothermia (NT, rectal temperature 37°C). Mice were treated with tri-DHA (0.375 g tri-DHA/kg bw, two injections) before and 1 h after initiation of HT. We observed that HT, beginning immediately after HI injury, reduced brain infarct volume similarly to tri-DHA treatment (~50%). Further, HT delayed 2 h post-HI injury provided neuroprotection (% infarct volume: 31.4 ± 4.1 vs. 18.8 ± 4.6 HT), while 4 h delayed HT did not protect against HI insult (% infarct volume: 30.7 ± 5.0 vs. 31.3 ± 5.6 HT). HT plus tri-DHA combination treatment beginning at 0 or 2 h after HI injury did not further reduce infarct volumes compared to HT alone. Our results indicate that HT offers similar degrees of neuroprotection against HI injury compared to tri-DHA treatment. HT can only be provided in tertiary care centers, requires intense monitoring and can have adverse effects. In contrast, tri-DHA treatment may be advantageous in providing a feasible and effective strategy in patients after HI injury.
Collapse
Affiliation(s)
| | - Hylde Zirpoli
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY, United States
| | - Vadim S Ten
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, United States
| | - Richard J Deckelbaum
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY, United States.,Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, United States
| |
Collapse
|
33
|
Age-related injury responses of human oligodendrocytes to metabolic insults: link to BCL-2 and autophagy pathways. Commun Biol 2021; 4:20. [PMID: 33398046 PMCID: PMC7782481 DOI: 10.1038/s42003-020-01557-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 12/01/2020] [Indexed: 01/29/2023] Open
Abstract
Myelin destruction and oligodendrocyte (OL) death consequent to metabolic stress is a feature of CNS disorders across the age spectrum. Using cells derived from surgically resected tissue, we demonstrate that young (<age 5) pediatric-aged sample OLs are more resistant to in-vitro metabolic injury than fetal O4+ progenitor cells, but more susceptible to cell death and apoptosis than adult-derived OLs. Pediatric but not adult OLs show measurable levels of TUNEL+ cells, a feature of the fetal cell response. The ratio of anti- vs pro-apoptotic BCL-2 family genes are increased in adult vs pediatric (<age 5) mature OLs and in more mature OL lineage cells. Lysosomal gene expression was increased in adult and pediatric compared to fetal OL lineage cells. Cell death of OLs was increased by inhibiting pro-apoptotic BCL-2 gene and autophagy activity. These distinct age-related injury responses should be considered in designing therapies aimed at reducing myelin injury.
Collapse
|
34
|
Rodriguez J, Li T, Xu Y, Sun Y, Zhu C. Role of apoptosis-inducing factor in perinatal hypoxic-ischemic brain injury. Neural Regen Res 2021; 16:205-213. [PMID: 32859765 PMCID: PMC7896227 DOI: 10.4103/1673-5374.290875] [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] [Indexed: 11/11/2022] Open
Abstract
Perinatal complications, such as asphyxia, can cause brain injuries that are often associated with subsequent neurological deficits, such as cerebral palsy or mental retardation. The mechanisms of perinatal brain injury are not fully understood, but mitochondria play a prominent role not only due to their central function in metabolism but also because many proteins with apoptosis-related functions are located in the mitochondrion. Among these proteins, apoptosis-inducing factor has already been shown to be an important factor involved in neuronal cell death upon hypoxia-ischemia, but a better understanding of the mechanisms behind these processes is required for the development of more effective treatments during the early stages of perinatal brain injury. In this review, we focus on the molecular mechanisms of hypoxic-ischemic encephalopathy, specifically on the importance of apoptosis-inducing factor. The relevance of apoptosis-inducing factor is based not only because it participates in the caspase-independent apoptotic pathway but also because it plays a crucial role in mitochondrial energetic functionality, especially with regard to the maintenance of electron transport during oxidative phosphorylation and in oxidative stress, acting as a free radical scavenger. We also discuss all the different apoptosis-inducing factor isoforms discovered, focusing especially on apoptosis-inducing factor 2, which is only expressed in the brain and the functions of which are starting now to be clarified. Finally, we summarized the interaction of apoptosis-inducing factor with several proteins that are crucial for both apoptosis-inducing factor functions (pro-survival and pro-apoptotic) and that are highly important in order to develop promising therapeutic targets for improving outcomes after perinatal brain injury.
Collapse
Affiliation(s)
- Juan Rodriguez
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Tao Li
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yiran Xu
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yanyan Sun
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Anatomy, School of Basic Medical Science, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Changlian Zhu
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China; Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
35
|
Combined the SMAC mimetic and BCL2 inhibitor sensitizes neoadjuvant chemotherapy by targeting necrosome complexes in tyrosine aminoacyl-tRNA synthase-positive breast cancer. Breast Cancer Res 2020; 22:130. [PMID: 33239070 PMCID: PMC7687715 DOI: 10.1186/s13058-020-01367-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 11/04/2020] [Indexed: 11/10/2022] Open
Abstract
Background Chemotherapy is the standard treatment for breast cancer; however, the response to chemotherapy is disappointingly low. Here, we investigated the alternative therapeutic efficacy of novel combination treatment with necroptosis-inducing small molecules to overcome chemotherapeutic resistance in tyrosine aminoacyl-tRNA synthetase (YARS)-positive breast cancer. Methods Pre-chemotherapeutic needle biopsy of 143 invasive ductal carcinomas undergoing the same chemotherapeutic regimen was subjected to proteomic analysis. Four different machine learning algorithms were employed to determine signature protein combinations. Immunoreactive markers were selected using three common candidate proteins from the machine-learning algorithms and verified by immunohistochemistry using 123 cases of independent needle biopsy FFPE samples. The regulation of chemotherapeutic response and necroptotic cell death was assessed using lentiviral YARS overexpression and depletion 3D spheroid formation assay, viability assays, LDH release assay, flow cytometry analysis, and transmission electron microscopy. The ROS-induced metabolic dysregulation and phosphorylation of necrosome complex by YARS were assessed using oxygen consumption rate analysis, flow cytometry analysis, and 3D cell viability assay. The therapeutic roles of SMAC mimetics (LCL161) and a pan-BCL2 inhibitor (ABT-263) were determined by 3D cell viability assay and flow cytometry analysis. Additional biologic process and protein-protein interaction pathway analysis were performed using Gene Ontology annotation and Cytoscape databases. Results YARS was selected as a potential biomarker by proteomics-based machine-learning algorithms and was exclusively associated with good response to chemotherapy by subsequent immunohistochemical validation. In 3D spheroid models of breast cancer cell lines, YARS overexpression significantly improved chemotherapy response via phosphorylation of the necrosome complex. YARS-induced necroptosis sequentially mediated mitochondrial dysfunction through the overproduction of ROS in breast cancer cell lines. Combination treatment with necroptosis-inducing small molecules, including a SMAC mimetic (LCL161) and a pan-BCL2 inhibitor (ABT-263), showed therapeutic efficacy in YARS-overexpressing breast cancer cells. Conclusions Our results indicate that, before chemotherapy, an initial screening of YARS protein expression should be performed, and YARS-positive breast cancer patients might consider the combined treatment with LCL161 and ABT-263; this could be a novel stepwise clinical approach to apply new targeted therapy in breast cancer patients in the future.
Collapse
|
36
|
Ross-Munro E, Kwa F, Kreiner J, Khore M, Miller SL, Tolcos M, Fleiss B, Walker DW. Midkine: The Who, What, Where, and When of a Promising Neurotrophic Therapy for Perinatal Brain Injury. Front Neurol 2020; 11:568814. [PMID: 33193008 PMCID: PMC7642484 DOI: 10.3389/fneur.2020.568814] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 09/18/2020] [Indexed: 12/21/2022] Open
Abstract
Midkine (MK) is a small secreted heparin-binding protein highly expressed during embryonic/fetal development which, through interactions with multiple cell surface receptors promotes growth through effects on cell proliferation, migration, and differentiation. MK is upregulated in the adult central nervous system (CNS) after multiple types of experimental injury and has neuroprotective and neuroregenerative properties. The potential for MK as a therapy for developmental brain injury is largely unknown. This review discusses what is known of MK's expression and actions in the developing brain, areas for future research, and the potential for using MK as a therapeutic agent to ameliorate the effects of brain damage caused by insults such as birth-related hypoxia and inflammation.
Collapse
Affiliation(s)
- Emily Ross-Munro
- Neurodevelopment in Health and Disease Research Program, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT), Melbourne, VIC, Australia
| | - Faith Kwa
- Neurodevelopment in Health and Disease Research Program, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT), Melbourne, VIC, Australia.,School of Health Sciences, Swinburne University of Technology, Melbourne, VIC, Australia
| | - Jenny Kreiner
- Neurodevelopment in Health and Disease Research Program, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT), Melbourne, VIC, Australia
| | - Madhavi Khore
- Neurodevelopment in Health and Disease Research Program, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT), Melbourne, VIC, Australia
| | - Suzanne L Miller
- The Ritchie Centre, Hudson Institute of Medical Research, Monash University, Clayton, VIC, Australia
| | - Mary Tolcos
- Neurodevelopment in Health and Disease Research Program, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT), Melbourne, VIC, Australia
| | - Bobbi Fleiss
- Neurodevelopment in Health and Disease Research Program, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT), Melbourne, VIC, Australia.,Neurodiderot, Inserm U1141, Universita de Paris, Paris, France
| | - David W Walker
- Neurodevelopment in Health and Disease Research Program, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT), Melbourne, VIC, Australia
| |
Collapse
|
37
|
Sowmithra S, Jain NK, Datta I. Evaluating In Vitro Neonatal Hypoxic-Ischemic Injury Using Neural Progenitors Derived from Human Embryonic Stem Cells. Stem Cells Dev 2020; 29:929-951. [DOI: 10.1089/scd.2020.0018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Sowmithra Sowmithra
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Institute of National Importance, Bengaluru, India
| | - Nishtha Kusum Jain
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Institute of National Importance, Bengaluru, India
| | - Indrani Datta
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Institute of National Importance, Bengaluru, India
| |
Collapse
|
38
|
Preterm birth and sustained inflammation: consequences for the neonate. Semin Immunopathol 2020; 42:451-468. [PMID: 32661735 PMCID: PMC7508934 DOI: 10.1007/s00281-020-00803-2] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 06/24/2020] [Indexed: 12/15/2022]
Abstract
Almost half of all preterm births are caused or triggered by an inflammatory process at the feto-maternal interface resulting in preterm labor or rupture of membranes with or without chorioamnionitis (“first inflammatory hit”). Preterm babies have highly vulnerable body surfaces and immature organ systems. They are postnatally confronted with a drastically altered antigen exposure including hospital-specific microbes, artificial devices, drugs, nutritional antigens, and hypoxia or hyperoxia (“second inflammatory hit”). This is of particular importance to extremely preterm infants born before 28 weeks, as they have not experienced important “third-trimester” adaptation processes to tolerate maternal and self-antigens. Instead of a balanced adaptation to extrauterine life, the delicate co-regulation between immune defense mechanisms and immunosuppression (tolerance) to allow microbiome establishment is therefore often disturbed. Hence, preterm infants are predisposed to sepsis but also to several injurious conditions that can contribute to the onset or perpetuation of sustained inflammation (SI). This is a continuing challenge to clinicians involved in the care of preterm infants, as SI is regarded as a crucial mediator for mortality and the development of morbidities in preterm infants. This review will outline the (i) role of inflammation for short-term consequences of preterm birth and (ii) the effect of SI on organ development and long-term outcome.
Collapse
|
39
|
Kumar AJ, Martins DO, Arruda BP, Lee VY, Chacur M, Nogueira MI. Impairment of nociceptive responses after neonatal anoxia correlates with somatosensory thalamic damage: A study in rats. Behav Brain Res 2020; 390:112690. [DOI: 10.1016/j.bbr.2020.112690] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 05/01/2020] [Accepted: 05/02/2020] [Indexed: 10/24/2022]
|
40
|
Protective effects of FGF10 on neurovascular unit in a rat model of neonatal hypoxic-ischemic brain injury. Exp Neurol 2020; 332:113393. [PMID: 32610105 DOI: 10.1016/j.expneurol.2020.113393] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 06/02/2020] [Accepted: 06/25/2020] [Indexed: 11/20/2022]
Abstract
Neonatal hypoxic-ischemic (HI) brain injury remains a devastating clinical disease associated with high mortality and lifetime disability. Neonatal HI injury damages the architecture of neurovascular unit (NVU), thus, therapy targeting the NVU may provide effective neuroprotection against HI. This study was designed to investigate whether fibroblast growth factor 10 (FGF10) protected the NVU against HI and afforded observable neuroprotection in a rat model of neonatal HI brain injury. The results showed that FGF10 treatment significantly reduced brain damage post HI, characterized by reduction in brain infarct volume and tissue loss. Further interesting findings showed that FGF10 treatment exerted neuroprotective effects on HI brain injury in neonate rats through protecting the NVU against HI, evidenced by inhibition of neuronal cell apoptosis, suppression of gliosis, and amelioration of blood-brain barrier disruption. Collectively, our study indicates that FGF10 treatment exhibits great potential for protecting NVU against HI and attenuates neonatal brain injury, suggesting a potential novel therapeutic agent to this disease.
Collapse
|
41
|
Wang L, Xiong X, Zhang X, Ye Y, Jian Z, Gao W, Gu L. Sodium Tanshinone IIA Sulfonate Protects Against Cerebral Ischemia-reperfusion Injury by Inhibiting Autophagy and Inflammation. Neuroscience 2020; 441:46-57. [PMID: 32505745 DOI: 10.1016/j.neuroscience.2020.05.054] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 05/28/2020] [Accepted: 05/28/2020] [Indexed: 12/19/2022]
Abstract
Sodium tanshinone IIA sulfonate (STS) can protect against brain damage induced by stroke. However, the neural protection mechanism of STS remains unclear. We investigated whether STS performs its protective function by suppressing autophagy and inflammatory activity during brain injury. We established a transient middle cerebral artery occlusion and reperfusion (MCAO/R) model by blocking the left middle cerebral artery with a thread inserted through the internal carotid artery for 1 h, followed by reperfusion for 48 h either with or without STS and the autophagy inhibitor 3-methyladenine (3-MA). Neuroprotective effects were determined by evaluating infarction, brain edema, and neurological deficits. The numbers of microglia-derived macrophages, monocyte-derived microglia, T cells, and B cells in the brains were measured, based on the surface marker analyses of CD45, CD11b, B220, CD3, and CD4 using fluorescence-assisted cell sorting. STS (10, 20, 40 mg/kg) was able to significantly reduce infarct volumes, improve neurological deficits, and reduce brain water contents. STS treatment reduced neuroinflammation, as assessed by the infiltration of macrophages and neutrophils, corresponding with reduced numbers of macrophages, T cells, and B cells in ischemia/reperfusion (I/R) brains. In addition, STS treatment also attenuated the upregulation of autophagy associated proteins, such as LC3-II, Beclin-1 and Sirt 6, which was induced by MCAO. These results demonstrated that STS can provide remarkable protection against ischemic stroke, possibly via the inhibition of autophagy and inflammatory activity.
Collapse
Affiliation(s)
- Lei Wang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, P.O. Box 430060, No. 238 Jiefang Road, Wuhan, China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, P.O. Box 430060, No. 238 Jiefang Road, Wuhan, China
| | - Xu Zhang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, P.O. Box 430060, No. 238 Jiefang Road, Wuhan, China
| | - Yingze Ye
- Central Laboratory, Renmin Hospital of Wuhan University, P.O. Box 430060, No. 238 Jiefang Road, Wuhan, China
| | - Zhihong Jian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, P.O. Box 430060, No. 238 Jiefang Road, Wuhan, China
| | - Wenwei Gao
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, P.O. Box 430060, No. 238 Jiefang Road, Wuhan, China.
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, P.O. Box 430060, No. 238 Jiefang Road, Wuhan, China.
| |
Collapse
|
42
|
Gravina G, Svedin P, Ardalan M, Levy O, Ek CJ, Mallard C, Lai JCY. Staphylococcus epidermidis Sensitizes Perinatal Hypoxic-Ischemic Brain Injury in Male but Not Female Mice. Front Immunol 2020; 11:516. [PMID: 32373108 PMCID: PMC7186320 DOI: 10.3389/fimmu.2020.00516] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 03/06/2020] [Indexed: 12/21/2022] Open
Abstract
Background: Staphylococcus epidermidis is the most common nosocomial infection and the predominant pathogen in late-onset sepsis in preterm infants. Infection and inflammation are linked to neurological and developmental sequelae and bacterial infections increase the vulnerability of the brain to hypoxia-ischemia (HI). We thus tested the hypothesis that S. epidermidis exacerbates HI neuropathology in neonatal mice. Methods: Male and female C57Bl/6 mice were injected intraperitoneally with sterile saline or 3.5 × 107 colony-forming units of S. epidermidis on postnatal day (PND) 4 and then subjected to HI on PND5 (24 h after injection) or PND9 (5 d after injection) by left carotid artery ligation and exposure to 10% O2. White and gray matter injury was assessed on PND14-16. In an additional group of animals, the plasma, brain, and liver were collected on PND5 or PND9 after infection to evaluate cytokine and chemokine profiles, C5a levels and C5 signaling. Results: HI induced 24 h after injection of S. epidermidis resulted in greater gray and white matter injury compared to saline injected controls in males, but not in females. Specifically, males demonstrated increased gray matter injury in the cortex and striatum, and white matter loss in the subcortical region, hippocampal fimbria and striatum. In contrast, there was no potentiation of brain injury when HI occurred 5 d after infection in either sex. In the plasma, S. epidermidis-injected mice demonstrated increased levels of pro- and anti-inflammatory cytokines and chemokines and a reduction of C5a at 24 h, but not 5 d after infection. Brain CCL2 levels were increased in both sexes 24 h after infection, but increased only in males at 5 d post infection. Conclusion: Ongoing S. epidermidis infection combined with neonatal HI increases the vulnerability of the developing brain in male but not in female mice. These sex-dependent effects were to a large extent independent of expression of systemic cytokines or brain CCL2 expression. Overall, we provide new insights into how systemic S. epidermidis infection affects the developing brain and show that the time interval between infection and HI is a critical sensitizing factor in males.
Collapse
Affiliation(s)
- Giacomo Gravina
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Pernilla Svedin
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Maryam Ardalan
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ofer Levy
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States.,Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - C Joakim Ek
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Carina Mallard
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jacqueline C Y Lai
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
43
|
Corti O, Blomgren K, Poletti A, Beart PM. Autophagy in neurodegeneration: New insights underpinning therapy for neurological diseases. J Neurochem 2020; 154:354-371. [PMID: 32149395 DOI: 10.1111/jnc.15002] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/27/2020] [Accepted: 03/05/2020] [Indexed: 12/13/2022]
Abstract
In autophagy long-lived proteins, protein aggregates or damaged organelles are engulfed by vesicles called autophagosomes prior to lysosomal degradation. Autophagy dysfunction is a hallmark of several neurodegenerative diseases in which misfolded proteins or dysfunctional mitochondria accumulate. Excessive autophagy can also exacerbate brain injury under certain conditions. In this review, we provide specific examples to illustrate the critical role played by autophagy in pathological conditions affecting the brain and discuss potential therapeutic implications. We show how a singular type of autophagy-dependent cell death termed autosis has attracted attention as a promising target for improving outcomes in perinatal asphyxia and hypoxic-ischaemic injury to the immature brain. We provide evidence that autophagy inhibition may be protective against radiotherapy-induced damage to the young brain. We describe a specialized form of macroautophagy of therapeutic relevance for motoneuron and neuromuscular diseases, known as chaperone-assisted selective autophagy, in which heat shock protein B8 is used to deliver aberrant proteins to autophagosomes. We summarize studies pinpointing mitophagy mediated by the serine/threonine kinase PINK1 and the ubiquitin-protein ligase Parkin as a mechanism potentially relevant to Parkinson's disease, despite debate over the physiological conditions in which it is activated in organisms. Finally, with the example of the autophagy-inducing agent rilmenidine and its discrepant effects in cell culture and mouse models of motor neuron disorders, we illustrate the importance of considering aspects such a disease stage and aggressiveness, type of insult and load of damaged or toxic cellular components, when choosing the appropriate drug, timepoint and duration of treatment.
Collapse
Affiliation(s)
- Olga Corti
- Institut National de la Santé et de la Recherche Médicale, Paris, France.,Centre National de la Recherche Scientifique, Paris, France.,Sorbonne Universités, Paris, France.,Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| | - Klas Blomgren
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Department of Paediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Angelo Poletti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, Milan, Italy
| | - Philip M Beart
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Vic, Australia.,Department of Pharmacology, University of Melbourne, Parkville, Vic, Australia
| |
Collapse
|
44
|
Tsuji S, Di Martino E, Mukai T, Tsuji S, Murakami T, Harris RA, Blomgren K, Åden U. Aggravated brain injury after neonatal hypoxic ischemia in microglia-depleted mice. J Neuroinflammation 2020; 17:111. [PMID: 32276642 PMCID: PMC7149909 DOI: 10.1186/s12974-020-01792-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 03/27/2020] [Indexed: 01/17/2023] Open
Abstract
Background Neuroinflammation plays an important role in neonatal hypoxic-ischemic encephalopathy (HIE). Although microglia are largely responsible for injury-induced inflammatory response, they play beneficial roles in both normal and disease states. However, the effects of microglial depletion on neonatal HIE remain unclear. Methods Tamoxifen was administered to Cx3cr1CreER/+Rosa26DTA/+ (microglia-depleted model) and Cx3cr1CreER/+Rosa26DTA/− (control) mice at P8 and P9 to assess the effect of microglial depletion. The density of microglia was quantified using Iba-1 staining. Moreover, the proportion of resident microglia after the HI insult was analyzed using flow cytometric analysis. At P10, the HI insult was conducted using the Rice-Vannucci procedure at P10. The infarct size and apoptotic cells were analyzed at P13. Cytokine analyses were performed using quantitative polymerase chain reaction and enzyme-linked immunosorbent assay (ELISA) at P13. Results At P10, tamoxifen administration induced > 99% microglial depletion in DTA+ mice. Following HI insult, there was persisted microglial depletion over 97% at P13. Compared to male DTA− mice, male DTA+ mice exhibited significantly larger infarct volumes; however, there were no significant differences among females. Moreover, compared to male DTA− mice, male DTA+ mice had a significantly higher density of TUNEL+ cells in the caudoputamen, cerebral cortex, and thalamus. Moreover, compared to female DTA− mice, female DTA+ mice showed a significantly greater number of TUNEL+ cells in the hippocampus and thalamus. Compared to DTA− mice, ELISA revealed significantly lower IL-10 and TGF-β levels in both male and female DTA+ mice under both normal conditions and after HI (more pronounced). Conclusion We established a microglial depletion model that aggravated neuronal damage and apoptosis after the HI insult, which was predominantly observed in males.
Collapse
Affiliation(s)
- Shunichiro Tsuji
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Department of Obstetrics and Gynecology, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu City, Shiga, 520-2192, Japan
| | - Elena Di Martino
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Takeo Mukai
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Shoko Tsuji
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Takashi Murakami
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu City, Shiga, 520-2192, Japan
| | - Robert A Harris
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska Hospital Solna, Stockholm, Sweden
| | - Klas Blomgren
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Department of Pediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Ulrika Åden
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Department of Neonatal Medicine, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
45
|
Inhibiting the interaction between apoptosis-inducing factor and cyclophilin A prevents brain injury in neonatal mice after hypoxia-ischemia. Neuropharmacology 2020; 171:108088. [PMID: 32277944 DOI: 10.1016/j.neuropharm.2020.108088] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 03/07/2020] [Accepted: 04/02/2020] [Indexed: 01/10/2023]
Abstract
The interaction between apoptosis-inducing factor (AIF) and cyclophilin A (CypA) has been shown to contribute to caspase-independent apoptosis. Blocking the AIF/CypA interaction protects against glutamate-induced neuronal cell death in vitro, and the purpose of this study was to determine the in vivo effect of an AIF/CypA interaction blocking peptide (AIF(370-394)-TAT) on neonatal mouse brain injury after hypoxia-ischemia (HI). The pups were treated with AIF (370-394)-TAT peptide intranasally prior to HI. Brain injury was significantly reduced at 72 h after HI in the AIF(370-394)-TAT peptide treatment group compared to vehicle-only treatment for both the gray matter and the subcortical white matter, and the neuroprotection was more pronounced in males than in females. Neuronal cell death was evaluated in males at 8 h and 24 h post-HI, and it was decreased significantly in the CA1 region of the hippocampus and the nucleus habenularis region after AIF(370-394)-TAT treatment. Caspase-independent apoptosis was decreased in the cortex, striatum, and nucleus habenularis after AIF(370-394)-TAT treatment, but no significant change was found on caspase-dependent apoptosis as indicated by the number of active caspase-3-labeled cells. Further analysis showed that both AIF and CypA nuclear accumulation were decreased after treatment with the AIF(370-394)-TAT peptide. These results suggest that AIF(370-394)-TAT inhibited AIF/CypA translocation to the nucleus and reduced HI-induced caspase-independent apoptosis and brain injury in young male mice, suggesting that blocking AIF/CypA might be a potential therapeutic target for neonatal brain injury.
Collapse
|
46
|
Romeo-Guitart D, Marcos-DeJuana C, Marmolejo-Martínez-Artesero S, Navarro X, Casas C. Novel neuroprotective therapy with NeuroHeal by autophagy induction for damaged neonatal motoneurons. Theranostics 2020; 10:5154-5168. [PMID: 32308774 PMCID: PMC7163445 DOI: 10.7150/thno.43765] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 03/20/2020] [Indexed: 01/03/2023] Open
Abstract
Rationale: Protective mechanisms allow healthy neurons to cope with diverse stresses. Excessive damage as well as aging can lead to defective functioning of these mechanisms. We recently designed NeuroHeal using artificial intelligence with the goal of bolstering endogenous neuroprotective mechanisms. Understanding the key nodes involved in neuroprotection will allow us to identify even more effective strategies for treatment of neurodegenerative diseases. Methods: We used a model of peripheral nerve axotomy in rat pups, that induces retrograde apoptotic death of motoneurons. Nourishing mothers received treatment with vehicle, NeuroHeal or NeuroHeal plus nicotinamide, an inhibitor of sirtuins, and analysis of the pups were performed by immunohistochemistry, electron microscopy, and immunoblotting. In vitro, the post-translational status of proteins of interest was detailed using organotypic spinal cord cultures and genetic modifications in cell lines to unravel the neuroprotective mechanisms involved. Results: We found that the concomitant activation of the NAD+-dependent deacetylase SIRT1 and the PI3K/AKT signaling pathway converge to increase the presence of deacetylated and phosphorylated FOXO3a, a transcription factor, in the nucleus. This favors the activation of autophagy, a pro-survival process, and prevents pro-apoptotic PARP1/2 cleavage. Major conclusion: NeuroHeal is a neuroprotective agent for neonatal motoneurons that fine-tunes autophagy on by converging SIRT1/AKT/FOXO3a axis. NeuroHeal is a combo of repurposed drugs that allow its readiness for prospective pediatric use.
Collapse
|
47
|
Zeng SS, Bai JJ, Jiang H, Zhu JJ, Fu CC, He MZ, Zhu JH, Chen SQ, Li PJ, Fu XQ, Lin ZL. Treatment With Liraglutide Exerts Neuroprotection After Hypoxic-Ischemic Brain Injury in Neonatal Rats via the PI3K/AKT/GSK3β Pathway. Front Cell Neurosci 2020; 13:585. [PMID: 32082121 PMCID: PMC7003644 DOI: 10.3389/fncel.2019.00585] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 12/23/2019] [Indexed: 12/16/2022] Open
Abstract
Neonatal hypoxic–ischemic (HI) brain injury is a detrimental disease, which results in high mortality and long-term neurological deficits. Nevertheless, the treatment options for this disease are limited. Thus, the aim of the present study was to assess the role of liraglutide in neonatal HI brain injury in rats and investigate the associated mechanisms. The results showed that treatment with liraglutide significantly reduced infarct volume and ameliorated cerebral edema, decreased inflammatory response, promoted the recovery of tissue structure, and improved prognosis following HI brain injury. Moreover, treatment with liraglutide inhibited apoptosis and promoted neuronal survival both in the rat model and following oxygen-glucose deprivation (OGD) insult. LY294002, an inhibitor of phosphoinositide 3-kinase (PI3K), partially reversed these therapeutic effects, suggesting that the PI3K/protein kinase B (Akt) pathway was involved. In conclusion, our data revealed that treatment with liraglutide exerts neuroprotection after neonatal HI brain injury via the PI3K/Akt/glycogen synthase kinase-3β (GSK3β) pathway and may be a promising therapy for this disease.
Collapse
Affiliation(s)
- Shan-Shan Zeng
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jun-Jie Bai
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Huai Jiang
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jin-Jin Zhu
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Chang-Chang Fu
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Min-Zhi He
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jiang-Hu Zhu
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Shang-Qin Chen
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Pei-Jun Li
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xiao-Qin Fu
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Zhen-Lang Lin
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
48
|
Li T, Li K, Zhang S, Wang Y, Xu Y, Cronin SJF, Sun Y, Zhang Y, Xie C, Rodriguez J, Zhou K, Hagberg H, Mallard C, Wang X, Penninger JM, Kroemer G, Blomgren K, Zhu C. Overexpression of apoptosis inducing factor aggravates hypoxic-ischemic brain injury in neonatal mice. Cell Death Dis 2020; 11:77. [PMID: 32001673 PMCID: PMC6992638 DOI: 10.1038/s41419-020-2280-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 01/14/2020] [Accepted: 01/16/2020] [Indexed: 12/21/2022]
Abstract
Apoptosis inducing factor (AIF) has been shown to be a major contributor to neuron loss in the immature brain after hypoxia-ischemia (HI). Indeed, mice bearing a hypomorphic mutation causing reduced AIF expression are protected against neonatal HI. To further investigate the possible molecular mechanisms of this neuroprotection, we generated an AIF knock-in mouse by introduction of a latent transgene coding for flagged AIF protein into the Rosa26 locus, followed by its conditional activation by a ubiquitously expressed Cre recombinase. Such AIF transgenic mice overexpress the pro-apoptotic splice variant of AIF (AIF1) at both the mRNA (5.9 times higher) and protein level (2.4 times higher), but not the brain-specific AIF splice-isoform (AIF2). Excessive AIF did not have any apparent effects on the phenotype or physiological functions of the mice. However, brain injury (both gray and white matter) after neonatal HI was exacerbated in mice overexpressing AIF, coupled to enhanced translocation of mitochondrial AIF to the nucleus as well as enhanced caspase-3 activation in some brain regions, as indicated by immunohistochemistry. Altogether, these findings corroborate earlier studies demonstrating that AIF plays a causal role in neonatal HI brain injury.
Collapse
Affiliation(s)
- Tao Li
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, China.,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden.,Department of Pediatrics, Children's Hospital Affiliated of Zhengzhou University, Zhengzhou, 450018, China
| | - Kenan Li
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, China.,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden
| | - Shan Zhang
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, China.,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden
| | - Yafeng Wang
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, China.,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden.,Department of Pediatrics, Children's Hospital Affiliated of Zhengzhou University, Zhengzhou, 450018, China
| | - Yiran Xu
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, China.,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden
| | - Shane J F Cronin
- Institute of Molecular Biotechnology, Austrian Academy of Sciences, 1030, Vienna, Austria
| | - Yanyan Sun
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, China.,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden
| | - Yaodong Zhang
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden.,Department of Pediatrics, Children's Hospital Affiliated of Zhengzhou University, Zhengzhou, 450018, China
| | - Cuicui Xie
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden.,Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Juan Rodriguez
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden
| | - Kai Zhou
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden.,Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Henrik Hagberg
- Centre of Perinatal Medicine and Health, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden
| | - Carina Mallard
- Centre of Perinatal Medicine and Health, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden.,Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden
| | - Xiaoyang Wang
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, China.,Centre of Perinatal Medicine and Health, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden.,Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden
| | - Josef M Penninger
- Institute of Molecular Biotechnology, Austrian Academy of Sciences, 1030, Vienna, Austria.,Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Guido Kroemer
- Equipe labellisée par la Ligue contre le cancer, Université Paris Descartes, Université Sorbonne Paris Cité, Université Paris Diderot, Sorbonne Université, INSERM U1138, Centre de Recherche des Cordeliers, Paris, France.,Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Suzhou Institute for Systems Biology, Chinese Academy of Sciences, Suzhou, China.,Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Klas Blomgren
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Pediatric Hematology and Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, China. .,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden. .,Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden. .,Centre of Perinatal Medicine and Health, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden.
| |
Collapse
|
49
|
Autophagy as a Cellular Stress Response Mechanism in the Nervous System. J Mol Biol 2020; 432:2560-2588. [PMID: 31962122 DOI: 10.1016/j.jmb.2020.01.017] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 12/11/2019] [Accepted: 01/10/2020] [Indexed: 12/13/2022]
Abstract
Cells of an organism face with various types of insults during their lifetime. Exposure to toxins, metabolic problems, ischaemia/reperfusion, physical trauma, genetic diseases, neurodegenerative diseases are among the conditions that trigger cellular stress responses. In this context, autophagy is one of the mechanisms that supports cell survival under stressful conditions. Autophagic vesicle engulfs the cargo and transports it to lysosome for degradation and turnover. As such, autophagy eliminates abnormal proteins, clears damaged organelles, limits oxidative stress and helps to improve metabolic balance. Nervous system cells and particularly postmitotic neurons are highly sensitive to a spectrum of insults, and autophagy emerges as one of the key stress response mechanism, ensuring health and survival of these vulnerable cell types. In this review, we will overview mechanisms through which cells cope with stress, and how these stress responses regulate autophagy, with a special focus on the nervous system.
Collapse
|
50
|
Pagida MA, Konstantinidou AE, Chrysanthou-Piterou MA, Patsouris ES, Panayotacopoulou MT. Apoptotic Markers in the Midbrain of the Human Neonate After Perinatal Hypoxic/Ischemic Injury. J Neuropathol Exp Neurol 2020; 79:86-101. [PMID: 31803912 DOI: 10.1093/jnen/nlz114] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/01/2019] [Accepted: 10/23/2019] [Indexed: 11/13/2022] Open
Abstract
Our previous postmortem studies on neonates with neuropathological injury of perinatal hypoxia/ischemia (PHI) showed a dramatic reduction of tyrosine hydroxylase expression (dopamine synthesis enzyme) in substantia nigra (SN) neurons, with reduction of their cellular size. In order to investigate if the above observations represent an early stage of SN degeneration, we immunohistochemically studied the expression of cleaved caspase-3 (CCP3), apoptosis inducing factor (AIF), and DNA fragmentation by using terminal deoxynucleotidyltransferase-mediated dUTP-biotin 3'-end-labeling (TUNEL) technique in the SN of 22 autopsied neonates (corrected age ranging from 34 to 46.5 gestational weeks), in relation to the severity/duration of PHI injury, as estimated by neuropathological criteria. No CCP3-immunoreactive neurons and a limited number of apoptotic TUNEL-positive neurons with pyknotic characteristics were found in the SN. Nuclear AIF staining was revealed only in few SN neurons, indicating the presence of early signs of AIF-mediated degeneration. By contrast, motor neurons of the oculomotor nucleus showed higher cytoplasmic AIF expression and nuclear translocation, possibly attributed to the combined effect of developmental processes and increased oxidative stress induced by antemortem and postmortem factors. Our study indicates the activation of AIF, but not CCP3, in the SN and oculomotor nucleus of the human neonate in the developmentally critical perinatal period.
Collapse
Affiliation(s)
- Marianna A Pagida
- 1st Department of Psychiatry (MPag, MC-P, MPan); Laboratory of Neurobiology and Histochemistry, University Mental Health Research Institute (MPag, MC-P, MPan); and 1st Department of Pathology (AK, EP), National and Kapodistrian University of Athens, Athens, Greece
| | - Anastasia E Konstantinidou
- 1st Department of Psychiatry (MPag, MC-P, MPan); Laboratory of Neurobiology and Histochemistry, University Mental Health Research Institute (MPag, MC-P, MPan); and 1st Department of Pathology (AK, EP), National and Kapodistrian University of Athens, Athens, Greece
| | - Margarita A Chrysanthou-Piterou
- 1st Department of Psychiatry (MPag, MC-P, MPan); Laboratory of Neurobiology and Histochemistry, University Mental Health Research Institute (MPag, MC-P, MPan); and 1st Department of Pathology (AK, EP), National and Kapodistrian University of Athens, Athens, Greece
| | - Efstratios S Patsouris
- 1st Department of Psychiatry (MPag, MC-P, MPan); Laboratory of Neurobiology and Histochemistry, University Mental Health Research Institute (MPag, MC-P, MPan); and 1st Department of Pathology (AK, EP), National and Kapodistrian University of Athens, Athens, Greece
| | - Maria T Panayotacopoulou
- 1st Department of Psychiatry (MPag, MC-P, MPan); Laboratory of Neurobiology and Histochemistry, University Mental Health Research Institute (MPag, MC-P, MPan); and 1st Department of Pathology (AK, EP), National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|