1
|
Biancotti JC, Sescleifer AM, Sferra SR, Penikis AB, Halbert-Elliott KM, Bubb CR, Kunisaki SM. Maternal Minocycline as Fetal Therapy in a Rat Model of Myelomeningocele. J Surg Res 2024; 301:696-703. [PMID: 39168042 DOI: 10.1016/j.jss.2024.07.088] [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: 02/27/2024] [Revised: 07/11/2024] [Accepted: 07/19/2024] [Indexed: 08/23/2024]
Abstract
INTRODUCTION This study aimed to investigate whether the maternal administration of minocycline, a tetracycline antibiotic known to have anti-inflammatory and neuroprotective properties in models of neural injury, reduces inflammation and neural cell death in a fetal rat model of myelomeningocele (MMC). METHODS E10 pregnant rats were gavaged with olive oil or olive oil + retinoic acid to induce fetal MMC. At E12, the dams were exposed to regular drinking water or water containing minocycline (range, 40-140 mg/kg/day). At E21, fetal lumbosacral spinal cords were isolated for immunohistochemistry and quantitative gene expression studies focused on microglia activity, inflammation, and apoptosis (P < 0.05). RESULTS There was a trend toward decreased activated Iba1+ microglial cells within the dorsal spinal cord of MMC pups following minocycline exposure when compared to water (H2O) alone (P = 0.052). Prenatal minocycline exposure was correlated with significantly reduced expression of the proinflammatory cytokine, IL-6 (minocycline: 1.75 versus H2O: 3.52, P = 0.04) and apoptosis gene, Bax (minocycline: 0.71 versus H2O: 1.04, P < 0.001) among MMC pups. CONCLUSIONS This study found evidence that the maternal administration of minocycline reduces selected markers of inflammation and apoptosis within the exposed dorsal spinal cords of fetal MMC rats. Further study of minocycline as a novel prenatal treatment strategy to mitigate spinal cord damage in MMC is warranted.
Collapse
Affiliation(s)
- Juan C Biancotti
- Division of General Pediatric Surgery, Department of Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Anne M Sescleifer
- Division of General Pediatric Surgery, Department of Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Shelby R Sferra
- Division of General Pediatric Surgery, Department of Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Annalise B Penikis
- Division of General Pediatric Surgery, Department of Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Kyra M Halbert-Elliott
- Division of General Pediatric Surgery, Department of Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Ciaran R Bubb
- Division of General Pediatric Surgery, Department of Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Shaun M Kunisaki
- Division of General Pediatric Surgery, Department of Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland.
| |
Collapse
|
2
|
Olivares-Costa M, Fabio MC, De la Fuente-Ortega E, Haeger PA, Pautassi R. New therapeutics for the prevention or amelioration of fetal alcohol spectrum disorders: a narrative review of the preclinical literature. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2024:1-22. [PMID: 39023419 DOI: 10.1080/00952990.2024.2361442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 05/26/2024] [Indexed: 07/20/2024]
Abstract
Background: Ethanol consumption during pregnancy induces enduring detrimental effects in the offspring, manifesting as a spectrum of symptoms collectively termed as Fetal Alcohol Spectrum Disorders (FASD). Presently, there is a scarcity of treatments for FASD.Objectives: To analyze current literature, emphasizing evidence derived from preclinical models, that could potentially inform therapeutic interventions for FASD.Methods: A narrative review was conducted focusing on four prospective treatments: nutritional supplements, antioxidants, anti-inflammatory compounds and environmental enrichment. The review also highlights innovative therapeutic strategies applied during early (e.g. folate administration, postnatal days 4-9) or late (e.g. NOX2 inhibitors given after weaning) postnatal stages that resulted in significant improvements in behavioral responses during adolescence (a critical period marked by the emergence of mental health issues in humans).Results: Our findings underscore the value of treatments centered around nutritional supplementation or environmental enrichment, aimed at mitigating oxidative stress and inflammation, implying shared mechanisms in FASD pathogenesis. Moreover, the review spotlights emerging evidence pertaining to the involvement of novel molecular components with potential pharmacological targets (such as NOX2, MCP1/CCR2, PPARJ, and PDE1).Conclusions: Preclinical studies have identified oxidative imbalance and neuroinflammation as relevant pathological mechanisms induced by prenatal ethanol exposure. The relevance of these mechanisms, which exhibit positive feedback loop mechanisms, appear to peak during early development and decreases in adulthood. These findings provide a framework for the future development of therapeutic avenues in the development of specific clinical treatments for FASD.
Collapse
Affiliation(s)
- Montserrat Olivares-Costa
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
| | - María Carolina Fabio
- Instituto de Investigación Médica M. y M. Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
- Facultad de Psicología, Universidad Nacional de Córdoba, Coquimbo, Chile
| | - Erwin De la Fuente-Ortega
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
| | - Paola A Haeger
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
- Millennium Nucleus of Neuroepigenetics and Plasticity (EpiNeuro), Santiago, Chile
| | - Ricardo Pautassi
- Instituto de Investigación Médica M. y M. Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
- Facultad de Psicología, Universidad Nacional de Córdoba, Coquimbo, Chile
| |
Collapse
|
3
|
Padilla-Valdez MM, Díaz-Iñiguez MI, Ortuño-Sahagún D, Rojas-Mayorquín AE. Neuroinflammation in fetal alcohol spectrum disorders and related novel therapeutic approaches. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166854. [PMID: 37611676 DOI: 10.1016/j.bbadis.2023.166854] [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: 04/03/2023] [Revised: 07/21/2023] [Accepted: 08/18/2023] [Indexed: 08/25/2023]
Abstract
Fetal alcohol spectrum disorders (FASD) is an umbrella term to describe the neurological effects of prenatal alcohol exposure (PAE). It has been extensively characterized that PAE causes cell proliferation disruption, heterotopias, and malformations in various brain regions and there is increasing evidence that neuroinflammation is responsible for some of these neurotoxic effects. Despite evidence of its importance, neuroinflammation is not usually considered at diagnosis or treatment for FASD. Here, we discuss the literature regarding anti- inflammatory drugs and nutraceuticals, which hold promise for future therapeutical interventions in these disorders.
Collapse
Affiliation(s)
- Mayra Madeleine Padilla-Valdez
- Departamento de Ciencias Ambientales, Universidad de Guadalajara, Centro Universitario de Ciencias Biológicas y Agropecuarias, Guadalajara 45200, Mexico; Laboratorio de Neuroinmunobiología Molecular, Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, C.P 44340 Guadalajara, JAL, Mexico
| | - María Isabel Díaz-Iñiguez
- Departamento de Ciencias Ambientales, Universidad de Guadalajara, Centro Universitario de Ciencias Biológicas y Agropecuarias, Guadalajara 45200, Mexico; Laboratorio de Neuroinmunobiología Molecular, Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, C.P 44340 Guadalajara, JAL, Mexico
| | - Daniel Ortuño-Sahagún
- Laboratorio de Neuroinmunobiología Molecular, Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, C.P 44340 Guadalajara, JAL, Mexico.
| | - Argelia Esperanza Rojas-Mayorquín
- Departamento de Ciencias Ambientales, Universidad de Guadalajara, Centro Universitario de Ciencias Biológicas y Agropecuarias, Guadalajara 45200, Mexico.
| |
Collapse
|
4
|
Mukherjee S, Tarale P, Sarkar DK. Neuroimmune Interactions in Fetal Alcohol Spectrum Disorders: Potential Therapeutic Targets and Intervention Strategies. Cells 2023; 12:2323. [PMID: 37759545 PMCID: PMC10528917 DOI: 10.3390/cells12182323] [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: 07/05/2023] [Revised: 09/13/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Fetal alcohol spectrum disorders (FASD) are a set of abnormalities caused by prenatal exposure to ethanol and are characterized by developmental defects in the brain that lead to various overt and non-overt physiological abnormalities. Growing evidence suggests that in utero alcohol exposure induces functional and structural abnormalities in gliogenesis and neuron-glia interactions, suggesting a possible role of glial cell pathologies in the development of FASD. However, the molecular mechanisms of neuron-glia interactions that lead to the development of FASD are not clearly understood. In this review, we discuss glial cell pathologies with a particular emphasis on microglia, primary resident immune cells in the brain. Additionally, we examine the involvement of several neuroimmune molecules released by glial cells, their signaling pathways, and epigenetic mechanisms responsible for FASD-related alteration in brain functions. Growing evidence suggests that extracellular vesicles (EVs) play a crucial role in the communication between cells via transporting bioactive cargo from one cell to the other. This review emphasizes the role of EVs in the context of neuron-glia interactions during prenatal alcohol exposure. Finally, some potential applications involving nutritional, pharmacological, cell-based, and exosome-based therapies in the treatment of FASD are discussed.
Collapse
Affiliation(s)
- Sayani Mukherjee
- The Endocrine Program, Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901-1573, USA; (S.M.); (P.T.)
- Hormone Laboratory Research Group, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Jonas Lies vei 91B, 5021 Bergen, Norway
| | - Prashant Tarale
- The Endocrine Program, Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901-1573, USA; (S.M.); (P.T.)
| | - Dipak K. Sarkar
- The Endocrine Program, Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901-1573, USA; (S.M.); (P.T.)
| |
Collapse
|
5
|
Dose-related shifts in proteome and function of extracellular vesicles secreted by fetal neural stem cells following chronic alcohol exposure. Heliyon 2022; 8:e11348. [PMID: 36387439 PMCID: PMC9649983 DOI: 10.1016/j.heliyon.2022.e11348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/07/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022] Open
Abstract
Accumulating evidence indicates that extracellular vesicles (EVs) mediate endocrine functions and also pathogenic effects of neurodevelopmental perturbagens like ethanol. We performed mass-spectrometry on EVs secreted by fetal murine cerebral cortical neural stem cells (NSCs), cultured ex-vivo as sex-specific neurosphere cultures, to identify overrepresented proteins and signaling pathways in EVs relative to parental NSCs in controls, and following exposure of parental NSCs to a dose range of ethanol. EV proteomes differ substantially from parental NSCs, and though EVs sequester proteins across sub-cellular compartments, they are enriched for distinct morphogenetic signals including the planar cell polarity pathway. Ethanol exposure favored selective protein sequestration in EVs and depletion in parental NSCs, and also resulted in dose-independent overrepresentation of cell-cycle and DNA replication pathways in EVs as well as dose-dependent overrepresentation of rRNA processing and mTor stress pathways. Transfer of untreated EVs to naïve cells resulted in decreased oxidative metabolism and S-phase, while EVs derived from ethanol-treated NSCs exhibited diminished effect. Collectively, these data show that NSCs secrete EVs with a distinct proteome that may have a general growth-inhibitory effect on recipient cells. Moreover, while ethanol results in selective transfer of proteins from NSCs to EVs, the efficacy of these exposure-derived EVs is diminished.
Collapse
|
6
|
Dong Y, Jiang X, Chen F, Wang D, Zhang Z. Inhibiting the aberrant PACT-p53 axis activation ameliorates spinal cord ischaemia-reperfusion injury in rats. Int Immunopharmacol 2022; 108:108745. [PMID: 35421805 DOI: 10.1016/j.intimp.2022.108745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 03/21/2022] [Accepted: 03/30/2022] [Indexed: 11/16/2022]
Abstract
Spinal cord ischaemia-reperfusion injury (SCII) induces multiple molecular and cellular changes, resulting in dyskinesia. Recently, it is reported that the p53 network plays a vital role in SCII. However, the roles of the PACT/PRKRA (interferon-inducible double-stranded RNA-dependent protein kinase activator A)-p53 axis in SCII are still unclear. The aim of this study was to elucidate the roles of the PACT-p53 axis in SCII. A Sprague-Dawley rat model of SCII was established by subjecting rats to a 14-min occlusion of the aortic arch. The Tarlov criteria, Western blotting, double immunofluorescence staining, haematoxylin and eosin (HE) staining, and transferase dUTP nick end labelling (TUNEL) assay were performed after SCII. Here, spinal cord ischaemia-reperfusion (SCI) caused hindlimb motor functional deficits as assessed by the Tarlov criteria. The protein expression of PACT was substantially upregulated at 48 h after SCII. Increased PACT fluorescence was mainly localized to neurons. Si-PACT pretreatment improved hindlimb motor function, ameliorated histological changes, and attenuated cell apoptosis after SCII. Si-PACT pretreatment reduced the protein expression of PACT, p53, Caspase-8 and IL-1β and the number of double-labelled PACT and p53. Taken together, inhibiting the aberrant PACT-p53 axis activation by si-PACT pretreatment ameliorates SCI-induced neuroapoptosis and neuroinflammation in rats. Silencing PACT expression is promising new therapeutic strategy for SCII.
Collapse
Affiliation(s)
- Yan Dong
- Department of Anesthesiology, The First Affiliated Hospital of China Medical University, Nanjingbei Street 155#, Shenyang 110001, Liaoning Province, China
| | - Xuan Jiang
- Department of Anesthesiology, The First Affiliated Hospital of China Medical University, Nanjingbei Street 155#, Shenyang 110001, Liaoning Province, China
| | - Fengshou Chen
- Department of Anesthesiology, The First Affiliated Hospital of China Medical University, Nanjingbei Street 155#, Shenyang 110001, Liaoning Province, China
| | - Dan Wang
- Department of Anesthesiology, The First Affiliated Hospital of China Medical University, Nanjingbei Street 155#, Shenyang 110001, Liaoning Province, China
| | - Zaili Zhang
- Department of Anesthesiology, The First Affiliated Hospital of China Medical University, Nanjingbei Street 155#, Shenyang 110001, Liaoning Province, China.
| |
Collapse
|
7
|
Useinovic N, Maksimovic S, Near M, Quillinan N, Jevtovic-Todorovic V. Do We Have Viable Protective Strategies against Anesthesia-Induced Developmental Neurotoxicity? Int J Mol Sci 2022; 23:ijms23031128. [PMID: 35163060 PMCID: PMC8834847 DOI: 10.3390/ijms23031128] [Citation(s) in RCA: 3] [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: 12/31/2021] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 02/07/2023] Open
Abstract
Since its invention, general anesthesia has been an indispensable component of modern surgery. While traditionally considered safe and beneficial in many pathological settings, hundreds of preclinical studies in various animal species have raised concerns about the detrimental and long-lasting consequences that general anesthetics may cause to the developing brain. Clinical evidence of anesthetic neurotoxicity in humans continues to mount as we continue to contemplate how to move forward. Notwithstanding the alarming evidence, millions of children are being anesthetized each year, setting the stage for substantial healthcare burdens in the future. Hence, furthering our knowledge of the molecular underpinnings of anesthesia-induced developmental neurotoxicity is crucially important and should enable us to develop protective strategies so that currently available general anesthetics could be safely used during critical stages of brain development. In this mini-review, we provide a summary of select strategies with primary focus on the mechanisms of neuroprotection and potential for clinical applicability. First, we summarize a diverse group of chemicals with the emphasis on intracellular targets and signal-transduction pathways. We then discuss epigenetic and transgenerational effects of general anesthetics and potential remedies, and also anesthesia-sparing or anesthesia-delaying approaches. Finally, we present evidence of a novel class of anesthetics with a distinct mechanism of action and a promising safety profile.
Collapse
Affiliation(s)
- Nemanja Useinovic
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (S.M.); (M.N.); (N.Q.); (V.J.-T.)
- Correspondence:
| | - Stefan Maksimovic
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (S.M.); (M.N.); (N.Q.); (V.J.-T.)
| | - Michelle Near
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (S.M.); (M.N.); (N.Q.); (V.J.-T.)
| | - Nidia Quillinan
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (S.M.); (M.N.); (N.Q.); (V.J.-T.)
- Neuronal Injury and Plasticity Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Vesna Jevtovic-Todorovic
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (S.M.); (M.N.); (N.Q.); (V.J.-T.)
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
8
|
Carloni E, Ramos A, Hayes LN. Developmental Stressors Induce Innate Immune Memory in Microglia and Contribute to Disease Risk. Int J Mol Sci 2021; 22:13035. [PMID: 34884841 PMCID: PMC8657756 DOI: 10.3390/ijms222313035] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 12/26/2022] Open
Abstract
Many types of stressors have an impact on brain development, function, and disease susceptibility including immune stressors, psychosocial stressors, and exposure to drugs of abuse. We propose that these diverse developmental stressors may utilize a common mechanism that underlies impaired cognitive function and neurodevelopmental disorders such as schizophrenia, autism, and mood disorders that can develop in later life as a result of developmental stressors. While these stressors are directed at critical developmental windows, their impacts are long-lasting. Immune activation is a shared pathophysiology across several different developmental stressors and may thus be a targetable treatment to mitigate the later behavioral deficits. In this review, we explore different types of prenatal and perinatal stressors and their contribution to disease risk and underlying molecular mechanisms. We highlight the impact of developmental stressors on microglia biology because of their early infiltration into the brain, their critical role in brain development and function, and their long-lived status in the brain throughout life. Furthermore, we introduce innate immune memory as a potential underlying mechanism for developmental stressors' impact on disease. Finally, we highlight the molecular and epigenetic reprogramming that is known to underlie innate immune memory and explain how similar molecular mechanisms may be at work for cells to retain a long-term perturbation after exposure to developmental stressors.
Collapse
Affiliation(s)
- Elisa Carloni
- Department of Molecular and Cellular Biology, Dartmouth College, Hanover, NH 03755, USA;
| | - Adriana Ramos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA;
| | - Lindsay N. Hayes
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
9
|
Niedzwiedz-Massey VM, Douglas JC, Rafferty T, Wight PA, Kane CJM, Drew PD. Ethanol modulation of hippocampal neuroinflammation, myelination, and neurodevelopment in a postnatal mouse model of fetal alcohol spectrum disorders. Neurotoxicol Teratol 2021; 87:107015. [PMID: 34256161 PMCID: PMC8440486 DOI: 10.1016/j.ntt.2021.107015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 06/24/2021] [Accepted: 07/08/2021] [Indexed: 01/15/2023]
Abstract
Fetal alcohol spectrum disorders (FASD) are alarmingly common and result in significant personal and societal loss. Neuropathology of the hippocampus is common in FASD leading to aberrant cognitive function. In the current study, we evaluated the effects of ethanol on the expression of a targeted set of molecules involved in neuroinflammation, myelination, neurotransmission, and neuron function in the developing hippocampus in a postnatal model of FASD. Mice were treated with ethanol from P4-P9, hippocampi were isolated 24 h after the final treatment at P10, and mRNA levels were quantitated by qRT-PCR. We evaluated the effects of ethanol on both pro-inflammatory and anti-inflammatory molecules in the hippocampus and identified novel mechanisms by which ethanol induces neuroinflammation. We further demonstrated that ethanol decreased expression of molecules associated with mature oligodendrocytes and greatly diminished expression of a lacZ reporter driven by the first half of the myelin proteolipid protein (PLP) gene (PLP1). In addition, ethanol caused a decrease in genes expressed in oligodendrocyte progenitor cells (OPCs). Together, these studies suggest ethanol may modulate pathogenesis in the developing hippocampus through effects on cells of the oligodendrocyte lineage, resulting in altered oligodendrogenesis and myelination. We also observed differential expression of molecules important in synaptic plasticity, neurogenesis, and neurotransmission. Collectively, the molecules evaluated in these studies may play a role in ethanol-induced pathology in the developing hippocampus and contribute to cognitive impairment associated with FASD. A better understanding of these molecules and their effects on the developing hippocampus may lead to novel treatment strategies for FASD.
Collapse
Affiliation(s)
- Victoria M Niedzwiedz-Massey
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - James C Douglas
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Tonya Rafferty
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Patricia A Wight
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Cynthia J M Kane
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Paul D Drew
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
10
|
Tournier N, Pottier G, Caillé F, Coulon C, Goislard M, Jégo B, Negroni J, Leroy C, Saba W. Nalmefene alleviates the neuroimmune response to repeated binge-like ethanol exposure: A TSPO PET imaging study in adolescent rats. Addict Biol 2021; 26:e12962. [PMID: 32896074 DOI: 10.1111/adb.12962] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/16/2020] [Accepted: 08/10/2020] [Indexed: 12/31/2022]
Abstract
A large body of preclinical research has shown that neuroimmunity plays a key role in the deleterious effects of alcohol (ethanol) to the brain. Translational imaging techniques are needed to monitor the efficacy of strategies to prevent or mitigate neuroinflammation and alleviate ethanol-induced neurotoxicity. Opioid receptor antagonists such as nalmefene are antagonists of the toll-like receptor 4, which may block the proinflammatory signaling cascade induced by ethanol at this specific target. Male adolescent rats received a validated protocol of ethanol injection (i.p, 3 g/kg daily for two consecutive days followed by two resting days) during 14 days. Positron emission tomography (PET) imaging with the translocator protein 18 kDa (TSPO) radioligand [18 F]DPA-714 was performed at day-15. Toxicity induced by repeated binge-like ethanol exposure (71% mortality) was drastically reduced by nalmefene pretreatment (0.4 mg/kg, 14% mortality). No mortality was observed in animals that received vehicle (control) or nalmefene alone. Compared with control animals (n = 10), a significant 2.8-fold to 4.6-fold increase in the volume of distribution (VT ) of [18 F]DPA-714 was observed among brain regions in animals exposed to ethanol only (n = 9). Pretreatment with nalmefene significantly alleviated the neuroimmune response to ethanol exposure in all brain regions (1.2-fold to 2.5-fold increase in VT ; n = 5). Nalmefene alone (n = 6) did not impact [18 F]DPA-714 VT compared with the control group. Nalmefene may protect against the neuroinflammatory response and overall toxicity associated with binge drinking. [18 F]DPA-714 PET imaging can be used to noninvasively address the neuroimmune impact of ethanol exposure and its modulation by pharmacological strategies in vivo, with translational perspectives.
Collapse
Affiliation(s)
- Nicolas Tournier
- BioMaps Université Paris‐Saclay, CEA, CNRS, Inserm Orsay France
- Institut des sciences du vivant Frédéric Joliot, CEA Service Hospitalier Frédéric Joliot Orsay France
| | - Géraldine Pottier
- Institut des sciences du vivant Frédéric Joliot, CEA Service Hospitalier Frédéric Joliot Orsay France
| | - Fabien Caillé
- BioMaps Université Paris‐Saclay, CEA, CNRS, Inserm Orsay France
- Institut des sciences du vivant Frédéric Joliot, CEA Service Hospitalier Frédéric Joliot Orsay France
| | - Christine Coulon
- BioMaps Université Paris‐Saclay, CEA, CNRS, Inserm Orsay France
- Institut des sciences du vivant Frédéric Joliot, CEA Service Hospitalier Frédéric Joliot Orsay France
| | - Maud Goislard
- BioMaps Université Paris‐Saclay, CEA, CNRS, Inserm Orsay France
- Institut des sciences du vivant Frédéric Joliot, CEA Service Hospitalier Frédéric Joliot Orsay France
| | - Benoit Jégo
- BioMaps Université Paris‐Saclay, CEA, CNRS, Inserm Orsay France
- Institut des sciences du vivant Frédéric Joliot, CEA Service Hospitalier Frédéric Joliot Orsay France
| | - Julia Negroni
- Institut des sciences du vivant Frédéric Joliot, CEA Service Hospitalier Frédéric Joliot Orsay France
| | - Claire Leroy
- BioMaps Université Paris‐Saclay, CEA, CNRS, Inserm Orsay France
- Institut des sciences du vivant Frédéric Joliot, CEA Service Hospitalier Frédéric Joliot Orsay France
| | - Wadad Saba
- BioMaps Université Paris‐Saclay, CEA, CNRS, Inserm Orsay France
- Institut des sciences du vivant Frédéric Joliot, CEA Service Hospitalier Frédéric Joliot Orsay France
| |
Collapse
|
11
|
Fujii C, Zorumski CF, Izumi Y. Ethanol, neurosteroids and cellular stress responses: Impact on central nervous system toxicity, inflammation and autophagy. Neurosci Biobehav Rev 2021; 124:168-178. [PMID: 33561510 DOI: 10.1016/j.neubiorev.2021.01.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 01/19/2021] [Indexed: 01/21/2023]
Abstract
Alcohol intake can impair brain function, in addition to other organs such as the liver and kidney. In the brain ethanol can be detrimental to memory formation, through inducing the integrated stress response/endoplasmic reticulum stress/unfolded protein response and the molecular mechanisms linking stress to other events such as NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammation and autophagy. This literature review aims to provide an overview of our current understanding of the molecular mechanisms involved in ethanol-induced damage with endoplasmic reticulum stress, integrated stress response, NLRP3 inflammation and autophagy, while discussing the impact of neurosteroids and oxysterols, including allopregnanolone, 25-hydroxycholesterol and 24S-hydroxycholesterol, on the central nervous system.
Collapse
Affiliation(s)
- Chika Fujii
- Department of Psychiatry and Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, United States
| | - Charles F Zorumski
- Department of Psychiatry and Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, United States
| | - Yukitoshi Izumi
- Department of Psychiatry and Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, United States.
| |
Collapse
|
12
|
Kane CJM, Douglas JC, Rafferty T, Johnson JW, Niedzwiedz-Massey VM, Phelan KD, Majewska AK, Drew PD. Ethanol modulation of cerebellar neuroinflammation in a postnatal mouse model of fetal alcohol spectrum disorders. J Neurosci Res 2021; 99:1986-2007. [PMID: 33533128 DOI: 10.1002/jnr.24797] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 12/28/2020] [Indexed: 01/02/2023]
Abstract
Fetal alcohol spectrum disorders (FASD) are alarmingly common, result in significant personal and societal loss, and there is no effective treatment for these disorders. Cerebellar neuropathology is common in FASD and causes aberrant cognitive and motor function. Ethanol-induced neuroinflammation is believed to contribute to neuropathological sequelae of FASD, and was previously demonstrated in the cerebellum in animal models of FASD. We now demonstrate neuroinflammation persists in the cerebellum several days following cessation of ethanol treatment in an early postnatal mouse model, with meaningful implications for timing of therapeutic intervention in FASD. We also demonstrate by Sholl analysis that ethanol decreases ramification of microglia cell processes in cells located near the Purkinje cell layer but not those near the external granule cell layer. Ethanol did not alter the expression of anti-inflammatory molecules or molecules that constitute NLRP1 and NLRP3 inflammasomes. Interestingly, ethanol decreased the expression of IL-23a (P19) and IL-12Rβ1 suggesting that ethanol may suppress IL-12 and IL-23 signaling. Fractalkine-fractalkine receptor (CX3CL1-CX3CR1) signaling is believed to suppress microglial activation and our demonstration that ethanol decreases CX3CL1 expression suggests that ethanol modulation of CX3CL1-CX3CR1 signaling may contribute to cerebellar neuroinflammation and neuropathology. We demonstrate ethanol alters the expression of specific molecules in the cerebellum understudied in FASD, but crucial for immune responses. Ethanol increases the expression of NOX-2 and NGP and decreases the expression of RAG1, NOS1, CD59a, S1PR5, PTPN22, GPR37, and Serpinb1b. These molecules represent a new horizon as potential targets for development of FASD therapy.
Collapse
Affiliation(s)
- Cynthia J M Kane
- Department of Neurobiology and Developmental Sciences, Biomedical Research Center II, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - James C Douglas
- Department of Neurobiology and Developmental Sciences, Biomedical Research Center II, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Tonya Rafferty
- Department of Neurobiology and Developmental Sciences, Biomedical Research Center II, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Jennifer W Johnson
- Department of Neurobiology and Developmental Sciences, Biomedical Research Center II, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Victoria M Niedzwiedz-Massey
- Department of Neurobiology and Developmental Sciences, Biomedical Research Center II, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Kevin D Phelan
- Department of Neurobiology and Developmental Sciences, Biomedical Research Center II, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Ania Katarzyna Majewska
- Department of Neuroscience, Center for Visual Science, University of Rochester, Rochester, NY, USA
| | - Paul D Drew
- Department of Neurobiology and Developmental Sciences, Biomedical Research Center II, University of Arkansas for Medical Sciences, Little Rock, AR, USA.,Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
13
|
Kane CJM, Drew PD. Neuroinflammatory contribution of microglia and astrocytes in fetal alcohol spectrum disorders. J Neurosci Res 2020; 99:1973-1985. [PMID: 32959429 DOI: 10.1002/jnr.24735] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 09/03/2020] [Accepted: 09/05/2020] [Indexed: 12/12/2022]
Abstract
Ethanol exposure to the fetus during pregnancy can result in fetal alcohol spectrum disorders (FASD). These disorders vary in severity, can affect multiple organ systems, and can lead to lifelong disabilities. Damage to the central nervous system (CNS) is common in FASD, and can result in altered behavior and cognition. The incidence of FASD is alarmingly high, resulting in significant personal and societal costs. There are no cures for FASD. Alcohol can directly alter the function of neurons in the developing CNS. In addition, ethanol can alter the function of CNS glial cells including microglia and astrocytes which normally maintain homeostasis in the CNS. These glial cells can function as resident immune cells in the CNS to protect against pathogens and other insults. However, activation of glia can also damage CNS cells and lead to aberrant CNS function. Ethanol exposure to the developing brain can result in the activation of glia and neuroinflammation, which may contribute to the pathology associated with FASD. This suggests that anti-inflammatory agents may be effective in the treatment of FASD.
Collapse
Affiliation(s)
- Cynthia J M Kane
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Paul D Drew
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA.,Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
14
|
Wang S, Wang C, Wang L, Cai Z. Minocycline Inhibits mTOR Signaling Activation and Alleviates Behavioral Deficits in the Wistar Rats with Acute Ischemia stroke. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2020; 19:791-799. [PMID: 32867663 DOI: 10.2174/1871527319999200831153748] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 04/06/2020] [Accepted: 07/13/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Mammalian target of rapamycin (mTOR) has been evidenced as a multimodal therapy in the pathophysiological process of Acute Ischemic Stroke (AIS). However, the pathway that minocycline targets mTOR signaling is not fully defined in the AIS pathogenesis. This study aims at the roles of minocycline on the mTOR signaling in the AIS process and further discovers the underlying mechanisms of minocycline involved in the following change of mTOR signaling-autophagy. METHODS Cerebral ischemia/reperfusion (CIR) rat animal models were established with the transient suture occlusion into the middle cerebral artery. Minocycline (50mg/kg) was given by intragastric administration. The Morris water maze was used to test the cognitive function of animals. Immunohistochemistry and immunofluorescence were introduced for testing the levels of synaptophysin and PSD-95. Western blot was conducted for investigating the levels of mTOR, p-mTOR (Ser2448), p70S6, p-p70S6 (Thr389), eEF2k, p-eEF2k (Ser366), p-eIF4B (Ser406), LC3, p62, synaptophysin and PSD-95. RESULTS Minocycline prevents the cognitive decline of the MCAO stroke rats. Minocycline limits the expression of p-mTOR (Ser2448) and the downstream targets of mTOR [p70S6, p-p70S6 (Thr389), eEF2k, p-eEF2k (Ser366) and p-eIF4B (Ser406)] (P<0.01), while minocycline has no influence on mTOR. LC3-II abundance and the LC3-II/I ratio were upregulated in the hippocampus of the MCAO stroke rats by the minocycline therapy (P<0.01). p62 was downregulated in the hippocampus from the MCAO stroke rats administrated with minocycline therapy(P<0.01). The levels of SYP and PSD-95 were upregulated in the brain of the MCAO stroke rats administrated with minocycline therapy. CONCLUSION Minocycline prevents cognitive deficits via inhibiting mTOR signaling and enhancing the autophagy process, and promoting the expression of pre- and postsynaptic proteins (synaptophysin and PSD-95) in the brain of the MCAO stroke rats. The potential neuroprotective role of minocycline in the process of cerebral ischemia may be related to mitigating ischemia-induced synapse injury via inhibiting the activation of mTOR signaling.
Collapse
Affiliation(s)
- Shengyuan Wang
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China
| | - Chuanling Wang
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China
| | - Lihua Wang
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Zhiyou Cai
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China
| |
Collapse
|
15
|
Knutson AO, Watters JJ. All roads lead to inflammation: Is maternal immune activation a common culprit behind environmental factors impacting offspring neural control of breathing? Respir Physiol Neurobiol 2019; 274:103361. [PMID: 31874263 DOI: 10.1016/j.resp.2019.103361] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 12/14/2019] [Accepted: 12/19/2019] [Indexed: 12/12/2022]
Abstract
Despite numerous studies investigating how prenatal exposures impact the developing brain, there remains very little known about how these in utero exposures impact the life-sustaining function of breathing. While some exposures such as alcohol and drugs of abuse are well-known to alter respiratory function, few studies have evaluated other common maternal environmental stimuli, such as maternal infection, inhalation of diesel exhaust particles prevalent in urban areas, or obstructive sleep apnea during pregnancy, just to name a few. The goals of this review article are thus to: 1) highlight data on gestational exposures that impair respiratory function, 2) discuss what is known about the potential role of inflammation in the effects of these maternal exposures, and 3) identify less studied but potential in utero exposures that could negatively impact CNS regions important in respiratory motor control, perhaps by impacting maternal or fetal inflammation. We highlight gaps in knowledge, summarize evidence related to the possible contributions of inflammation, and discuss the need for further studies of life-long offspring respiratory function both at baseline and after respiratory challenge.
Collapse
Affiliation(s)
- Andrew O Knutson
- Molecular and Environmental Toxicology Training Program and Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, United States
| | - Jyoti J Watters
- Molecular and Environmental Toxicology Training Program and Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, United States.
| |
Collapse
|