1
|
Wang D, Wang J, Yan D, Wang M, Yang L, Demin KA, de Abreu MS, Kalueff AV. Minocycline reduces neurobehavioral deficits evoked by chronic unpredictable stress in adult zebrafish. Brain Res 2024; 1845:149209. [PMID: 39233136 DOI: 10.1016/j.brainres.2024.149209] [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: 06/18/2024] [Revised: 08/11/2024] [Accepted: 08/28/2024] [Indexed: 09/06/2024]
Abstract
Chronic stress-related brain disorders are widespread and debilitating, and often cause lasting neurobehavioral deficits. Minocycline, a common antibiotic and an established inhibitor of microglia, emerges as potential treatment of these disorders. The zebrafish (Danio rerio) is an important emerging model organism in translational neuroscience and stress research. Here, we evaluated the potential of minocycline to correct microglia-mediated behavioral, genomic and neuroimmune responses induced by chronic unpredictable stress (CUS) in adult zebrafish. We demonstrated that CUS evoked overt behavioral deficits in the novel tank, light-dark box and shoaling tests, paralleled by elevated stress hormones (CRH, ACTH and cortisol), and upregulated brain expression of the 'neurotoxic M1' microglia-specific biomarker gene (MHC-2) and pro-inflammatory cytokine genes (IL-1β, IL-6 and IFN-γ). CUS also elevated peripheral (whole-body) pro-inflammatory (IL-1β, IFN-γ) and lowered anti-inflammatory cytokines (IL-4 and IL-10), as well as reduced whole-brain serotonin, dopamine and norepinephrine levels, and increased brain dopamine and serotonin turnover. In contrast, minocycline attenuated most of these effects, also reducing CUS-elevated peripheral levels of IL-6 and IFN-γ. Collectively, this implicates microglia in zebrafish responses to chronic stress, and suggests glial pathways as potential evolutionarily conserved drug targets for treating stress-evoked neuropathogenesis. Our findings also support the growing translational value of zebrafish models for understanding complex molecular mechanisms of brain pathogenesis and its therapy.
Collapse
Affiliation(s)
- Dongmei Wang
- School of Pharmacy, Southwest University, Chongqing, China
| | - Jingtao Wang
- School of Pharmacy, Southwest University, Chongqing, China
| | - Dongni Yan
- School of Pharmacy, Southwest University, Chongqing, China
| | - Mengyao Wang
- School of Pharmacy, Southwest University, Chongqing, China
| | - Longen Yang
- School of Pharmacy, Southwest University, Chongqing, China; Suzhou Municipal Key Laboratory of Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University (XJTLU), Suzhou, China; Department of Biological Sciences, School of Science, Xi'an Jiaotong-Liverpool University (XJTLU), Suzhou, China
| | - Konstantin A Demin
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Murilo S de Abreu
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil; Western Caspian University, Baku, Azerbaijan; Moscow Institute of Physics and Technology, Dolgoprudny, Russia.
| | - Allan V Kalueff
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Institute of Experimental Medicine, Almazov National Medical Research Center, St. Petersburg, Russia; Suzhou Municipal Key Laboratory of Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University (XJTLU), Suzhou, China; Department of Biological Sciences, School of Science, Xi'an Jiaotong-Liverpool University (XJTLU), Suzhou, China; Moscow Institute of Physics and Technology, Dolgoprudny, Russia.
| |
Collapse
|
2
|
Galstyan DS, Lebedev AS, Ilyin NP, Papulova MS, Golushko NI, Tishkina VV, Saklakova DK, Martynov D, Kolesnikova TO, Rosemberg DB, De Abreu MS, Demin KA, Kalueff AV. Acute Behavioral and Neurochemical Effects of Sulpiride in Adult Zebrafish. Neurochem Res 2024; 50:11. [PMID: 39549192 DOI: 10.1007/s11064-024-04268-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 11/18/2024]
Abstract
Affective and psychotic disorders are highly prevalent and severely debilitating mental illnesses that often remain untreated or treatment-resistant. Sulpiride is a common antipsychotic (neuroleptic) drug whose well-established additional (e.g., antidepressant) therapeutic effects call for further studies of a wider spectrum of its CNS effects. Here, we examined effects of acute 20-min exposure to sulpiride (50-200 mg/L) on anxiety- and depression-like behaviors, as well as on brain monoamines, in adult zebrafish (Danio rerio). Overall, sulpiride exerted overt anxiolytic-like effects in the novel tank test and showed tranquilizing-like effects in the zebrafish tail immobilization test, accompanied by lowered whole-brain dopamine and its elevated turnover, without affecting serotonin or norepinephrine levels and their turnover. Taken together, these findings support complex behavioral pharmacology of sulpiride in vivo and reconfirm high sensitivity of zebrafish-based screens to this and, likely, other related clinically active neuroleptics.
Collapse
Affiliation(s)
- David S Galstyan
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
- Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Andrey S Lebedev
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
- Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Nikita P Ilyin
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
- Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Maria S Papulova
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Nikita I Golushko
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Valeria V Tishkina
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Daryna K Saklakova
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Daniil Martynov
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
- Almazov National Medical Research Centre, St. Petersburg, Russia
| | | | - Dennis B Rosemberg
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, Santa Maria, Brazil
| | - Murilo S De Abreu
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil
- Western Caspian University, Baku, Azerbaijan
| | - Konstantin A Demin
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia.
- Almazov National Medical Research Centre, St. Petersburg, Russia.
| | - Allan V Kalueff
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia.
- Department of Biolosciences and Bioinformatics, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China.
- Suzhou Municipal Key Laboratory of Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China.
| |
Collapse
|
3
|
de Sousa EB, Heymbeeck JAA, Feitosa LM, Xavier AGO, Dos Santos Campos K, do Socorro Dos Santos Rodrigues L, de Freitas LM, do Carmo Silva RX, Ikeda SR, de Nazaré Dos Santos Silva S, Rocha SP, do Nascimento WL, da Silva Moraes ER, Herculano AM, Maximino C, Pereira A, Lima-Maximino M. Activation of NOS-cGMP pathways promotes stress-induced sensitization of behavioral responses in zebrafish. Pharmacol Biochem Behav 2024; 243:173816. [PMID: 38971472 DOI: 10.1016/j.pbb.2024.173816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/20/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
Nitric oxide (NO) is a molecule involved in plasticity across levels and systems. The role of NOergic pathways in stress-induced sensitization (SIS) of behavioral responses, in which a particular stressor triggers a state of hyper-responsiveness to other stressors after an incubation period, was assessed in adult zebrafish. In this model, adult zebrafish acutely exposed to a fear-inducing conspecific alarm substance (CAS) and left undisturbed for an incubation period show increased anxiety-like behavior 24 h after exposure. CAS increased forebrain glutamate immediately after stress and 30 min after stress, an effect that was accompanied by increased nitrite levels immediately after stress, 30 min after stress, 90 min after stress, and 24 h after stress. CAS also increased nitrite levels in the head kidney, where cortisol is produced in zebrafish. CAS-elicited nitrite responses in the forebrain 90 min (but not 30 min) after stress were prevented by a NOS-2 blocker. Blocking NOS-1 30 min after stress prevents SIS; blocking NOS-2 90 min after stress also prevents stress-induced sensitization, as does blocking calcium-activated potassium channels in this latter time window. Stress-induced sensitization is also prevented by blocking guanylate cyclase activation in both time windows, and cGMP-dependent channel activation in the second time window. These results suggest that different NO-related pathways converge at different time windows of the incubation period to induce stress-induced sensitization.
Collapse
Affiliation(s)
- Eveline Bezerra de Sousa
- Laboratório de Bacteriologia e Neuropatologia, Universidade do Estado do Pará, Campus VIII, Marabá, PA, Brazil; Programa de Pós-Graduação em Neurociências e Biologia Celular, Instituto de Estudos em Saúde e Biológicas, Universidade Federal do Pará (UFPA), Belém, PA, Brazil
| | - João Alphonse Apóstolo Heymbeeck
- Laboratório de Neurofarmacologia e Biofísica, Universidade do Estado do Pará, Campus VIII, Marabá, PA, Brazil; Programa de Pós-Graduação em Neurociências e Comportamento, Núcleo de Teoria e Pesquisa do Comportamento, Universidade Federal do Pará (UFPA), Belém, PA, Brazil
| | - Leonardo Miranda Feitosa
- Laboratório de Neurofarmacologia e Biofísica, Universidade do Estado do Pará, Campus VIII, Marabá, PA, Brazil; Programa de Pós-Graduação em Neurociências e Comportamento, Núcleo de Teoria e Pesquisa do Comportamento, Universidade Federal do Pará (UFPA), Belém, PA, Brazil
| | | | - Kimberly Dos Santos Campos
- Departamento de Morfologia e Ciências Fisiológicas, Universidade do Estado do Pará, Campus VIII, Marabá, PA, Brazil
| | | | - Larissa Mota de Freitas
- Departamento de Morfologia e Ciências Fisiológicas, Universidade do Estado do Pará, Campus VIII, Marabá, PA, Brazil
| | - Rhayra Xavier do Carmo Silva
- Departamento de Morfologia e Ciências Fisiológicas, Universidade do Estado do Pará, Campus VIII, Marabá, PA, Brazil
| | - Saulo Rivera Ikeda
- Departamento de Morfologia e Ciências Fisiológicas, Universidade do Estado do Pará, Campus VIII, Marabá, PA, Brazil
| | | | - Sueslene Prado Rocha
- Departamento de Morfologia e Ciências Fisiológicas, Universidade do Estado do Pará, Campus VIII, Marabá, PA, Brazil
| | - Wilker Leite do Nascimento
- Departamento de Morfologia e Ciências Fisiológicas, Universidade do Estado do Pará, Campus VIII, Marabá, PA, Brazil
| | | | - Anderson Manoel Herculano
- Laboratório de Neurofarmacologia Experimental, Instituto de Ciências Biológicas, Universidade Federal do Pará (UFPA), Belém, PA, Brazil
| | - Caio Maximino
- Laboratório de Neurociências e Comportamento "Frederico Guilherme Graeff", Instituto de Estudos em Saúde e Biológicas, Universidade Federal do Sul e Sudeste do Pará (Unifesspa), Marabá, PA, Brazil.
| | - Antonio Pereira
- Laboratório de Processamento de Sinais, Instituto de Tecnologia, Universidade Federal do Pará (UFPA), Belém, PA, Brazil
| | - Monica Lima-Maximino
- Laboratório de Neurofarmacologia e Biofísica, Universidade do Estado do Pará, Campus VIII, Marabá, PA, Brazil; Departamento de Morfologia e Ciências Fisiológicas, Universidade do Estado do Pará, Campus VIII, Marabá, PA, Brazil
| |
Collapse
|
4
|
Wang YT, Wang XL, Lei L, Zhang Y. Efficacy of ginsenoside Rg1 on rodent models of depression: A systematic review and meta-analysis. Psychopharmacology (Berl) 2024:10.1007/s00213-024-06649-y. [PMID: 39039242 DOI: 10.1007/s00213-024-06649-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 07/07/2024] [Indexed: 07/24/2024]
Abstract
RATIONALE Depression is a prevalent psychiatric disease, and ginsenoside Rg1 is a bioactive compound extracted from the root of Panax ginseng C.A.Mey. To systematically investigate the effectiveness of Rg1 in rodent models of depression and provide evidence-based references for treating depression. METHODS Electronic searches for rodent studies were performed from inception to October 2022, e.g., PUBMED and EMBASE. Data extraction and quality evaluation were performed for the references, and meta-analysis was performed on the selected data using Review Manager 5.3.5. The outcomes were analyzed via a random-effect model and presented as mean difference (MD) with 95% confidence intervals (CIs). RESULTS A total of 24 studies and 678 animals were included in this meta-analysis. Rg1 remarkably improved depressive-like symptoms of depressed rodents, including the sucrose preference test (25.08, 95% CI: 20.17-30.00, Z = 10.01, P < 0.00001), forced swimming test (MD = -37.69, 95% CI: (-45.18, -30.2); Z = 9.86, P < 0.00001), and the tail suspension test (MD = -22.93, seconds, 95% CI: (-38.49, -7.37); Z = 2.89, P = 0.004). CONCLUSIONS The main antidepressant mechanism of Rg1 was concluded to be the neurotransmitter system, oxidant stress system, and inflammation. Conclusively, this study indicated the possible protective and therapeutic effects of Rg1 for treating depression via multiple mechanisms.
Collapse
Affiliation(s)
- Ya-Ting Wang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Sunshine Southern Avenue, Fang-Shan District, Beijing, 102488, China
| | - Xiao-Le Wang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Sunshine Southern Avenue, Fang-Shan District, Beijing, 102488, China
| | - Lan Lei
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Sunshine Southern Avenue, Fang-Shan District, Beijing, 102488, China
| | - Yi Zhang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Sunshine Southern Avenue, Fang-Shan District, Beijing, 102488, China.
| |
Collapse
|
5
|
Al-Zoubi RM, Abu-Hijleh H, Zarour A, Zakaria ZZ, Yassin A, Al-Ansari AA, Al-Asmakh M, Bawadi H. Zebrafish Model in Illuminating the Complexities of Post-Traumatic Stress Disorders: A Unique Research Tool. Int J Mol Sci 2024; 25:4895. [PMID: 38732113 PMCID: PMC11084870 DOI: 10.3390/ijms25094895] [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: 01/02/2024] [Revised: 02/13/2024] [Accepted: 02/16/2024] [Indexed: 05/13/2024] Open
Abstract
Post-traumatic stress disorder (PTSD) is a debilitating psychological condition that may develop in certain individuals following exposure to life-threatening or traumatic events. Distressing symptoms, including flashbacks, are characterized by disrupted stress responses, fear, anxiety, avoidance tendencies, and disturbances in sleep patterns. The enduring effects of PTSD can profoundly impact personal and familial relationships, as well as social, medical, and financial stability. The prevalence of PTSD varies among different populations and is influenced by the nature of the traumatic event. Recently, zebrafish have emerged as a valuable model organism in studying various conditions and disorders. Zebrafish display robust behavioral patterns that can be effectively quantified using advanced video-tracking tools. Due to their relatively simple nervous system compared to humans, zebrafish are particularly well suited for behavioral investigations. These unique characteristics make zebrafish an appealing model for exploring the underlying molecular and genetic mechanisms that govern behavior, thus offering a powerful comparative platform for gaining deeper insights into PTSD. This review article aims to provide updates on the pathophysiology of PTSD and the genetic responses associated with psychological stress. Additionally, it highlights the significance of zebrafish behavior as a valuable tool for comprehending PTSD better. By leveraging zebrafish as a model organism, researchers can potentially uncover novel therapeutic interventions for the treatment of PTSD and contribute to a more comprehensive understanding of this complex condition.
Collapse
Affiliation(s)
- Raed M. Al-Zoubi
- Department of Chemistry, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan;
- Surgical Research Section, Department of Surgery, Hamad Medical Corporation, Doha P.O. Box 3050, Qatar; (A.Y.); (A.A.A.-A.)
- Department of Biomedical Sciences, QU-Health, College of Health Sciences, Qatar University, Doha 2713, Qatar
| | - Haya Abu-Hijleh
- Department of Human Nutrition, QU-Health, College of Health Sciences, Qatar University, Doha 2713, Qatar; (H.A.-H.); (M.A.-A.)
| | - Ahmad Zarour
- Department of Surgery, Acute Care Surgery, Hamad Medical Corporation, Doha P.O. Box 3050, Qatar;
| | - Zain Z. Zakaria
- Vice President for Medical and Health Sciences Office, QU-Health, Qatar University, Doha 2713, Qatar;
| | - Aksam Yassin
- Surgical Research Section, Department of Surgery, Hamad Medical Corporation, Doha P.O. Box 3050, Qatar; (A.Y.); (A.A.A.-A.)
- Center of Medicine and Health Sciences, Dresden International University, 01069 Dresden, Germany
| | - Abdulla A. Al-Ansari
- Surgical Research Section, Department of Surgery, Hamad Medical Corporation, Doha P.O. Box 3050, Qatar; (A.Y.); (A.A.A.-A.)
| | - Maha Al-Asmakh
- Department of Human Nutrition, QU-Health, College of Health Sciences, Qatar University, Doha 2713, Qatar; (H.A.-H.); (M.A.-A.)
- Biomedical Research Center, Qatar University, Doha P.O. Box 2713, Qatar
| | - Hiba Bawadi
- Department of Human Nutrition, QU-Health, College of Health Sciences, Qatar University, Doha 2713, Qatar; (H.A.-H.); (M.A.-A.)
| |
Collapse
|
6
|
Mastrogiovanni M, Martínez-Navarro FJ, Bowman TV, Cayuela ML. Inflammation in Development and Aging: Insights from the Zebrafish Model. Int J Mol Sci 2024; 25:2145. [PMID: 38396822 PMCID: PMC10889087 DOI: 10.3390/ijms25042145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 01/30/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
Zebrafish are an emergent animal model to study human diseases due to their significant genetic similarity to humans, swift development, and genetic manipulability. Their utility extends to the exploration of the involvement of inflammation in host defense, immune responses, and tissue regeneration. Additionally, the zebrafish model system facilitates prompt screening of chemical compounds that affect inflammation. This study explored the diverse roles of inflammatory pathways in zebrafish development and aging. Serving as a crucial model, zebrafish provides insights into the intricate interplay of inflammation in both developmental and aging contexts. The evidence presented suggests that the same inflammatory signaling pathways often play instructive or beneficial roles during embryogenesis and are associated with malignancies in adults.
Collapse
Affiliation(s)
- Marta Mastrogiovanni
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Francisco Juan Martínez-Navarro
- Grupo de Telomerasa, Cáncer y Envejecimiento, Hospital Clínico Universitario Virgen de la Arrixaca, 30120 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria-Arrixaca, 30120 Murcia, Spain
| | - Teresa V. Bowman
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - María L. Cayuela
- Grupo de Telomerasa, Cáncer y Envejecimiento, Hospital Clínico Universitario Virgen de la Arrixaca, 30120 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria-Arrixaca, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 30100 Murcia, Spain
| |
Collapse
|
7
|
Natsaridis E, Perdikaris P, Fokos S, Dermon CR. Neuronal and Astroglial Localization of Glucocorticoid Receptor GRα in Adult Zebrafish Brain ( Danio rerio). Brain Sci 2023; 13:861. [PMID: 37371341 DOI: 10.3390/brainsci13060861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Glucocorticoid receptor α (GRα), a ligand-regulated transcription factor, mainly activated by cortisol in humans and fish, mediates neural allostatic and homeostatic functions induced by different types of acute and chronic stress, and systemic inflammation. Zebrafish GRα is suggested to have multiple transcriptional effects essential for normal development and survival, similarly to mammals. While sequence alignments of human, monkey, rat, and mouse GRs have shown many GRα isoforms, we questioned the protein expression profile of GRα in the adult zebrafish (Danio rerio) brain using an alternative model for stress-related neuropsychiatric research, by means of Western blot, immunohistochemistry and double immunofluorescence. Our results identified four main GRα-like immunoreactive bands (95 kDa, 60 kDa, 45 kDa and 35 kDa), with the 95 kDa protein showing highest expression in forebrain compared to midbrain and hindbrain. GRα showed a wide distribution throughout the antero-posterior zebrafish brain axis, with the most prominent labeling within the telencephalon, preoptic, hypothalamus, midbrain, brain stem, central grey, locus coeruleus and cerebellum. Double immunofluorescence revealed that GRα is coexpressed in TH+, β2-AR+ and vGLUT+ neurons, suggesting the potential of GRα influences on adrenergic and glutamatergic transmission. Moreover, GRα was co-localized in midline astroglial cells (GFAP+) within the telencephalon, hypothalamus and hindbrain. Interestingly, GRα expression was evident in the brain regions involved in adaptive stress responses, social behavior, and sensory and motor integration, supporting the evolutionarily conserved features of glucocorticoid receptors in the zebrafish brain.
Collapse
Affiliation(s)
- Evangelos Natsaridis
- Laboratory of Human and Animal Physiology, Department of Biology, University of Patras, Rion, 26504 Patras, Greece
| | - Panagiotis Perdikaris
- Laboratory of Human and Animal Physiology, Department of Biology, University of Patras, Rion, 26504 Patras, Greece
| | - Stefanos Fokos
- Laboratory of Human and Animal Physiology, Department of Biology, University of Patras, Rion, 26504 Patras, Greece
| | - Catherine R Dermon
- Laboratory of Human and Animal Physiology, Department of Biology, University of Patras, Rion, 26504 Patras, Greece
| |
Collapse
|
8
|
Lawrence JM, Schardien K, Wigdahl B, Nonnemacher MR. Roles of neuropathology-associated reactive astrocytes: a systematic review. Acta Neuropathol Commun 2023; 11:42. [PMID: 36915214 PMCID: PMC10009953 DOI: 10.1186/s40478-023-01526-9] [Citation(s) in RCA: 99] [Impact Index Per Article: 99.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 02/08/2023] [Indexed: 03/16/2023] Open
Abstract
In the contexts of aging, injury, or neuroinflammation, activated microglia signaling with TNF-α, IL-1α, and C1q induces a neurotoxic astrocytic phenotype, classified as A1, A1-like, or neuroinflammatory reactive astrocytes. In contrast to typical astrocytes, which promote neuronal survival, support synapses, and maintain blood-brain barrier integrity, these reactive astrocytes downregulate supportive functions and begin to secrete neurotoxic factors, complement components like C3, and chemokines like CXCL10, which may facilitate recruitment of immune cells across the BBB into the CNS. The proportion of pro-inflammatory reactive astrocytes increases with age through associated microglia activation, and these pro-inflammatory reactive astrocytes are particularly abundant in neurodegenerative disorders. As the identification of astrocyte phenotypes progress, their molecular and cellular effects are characterized in a growing array of neuropathologies.
Collapse
Affiliation(s)
- Jill M Lawrence
- Molecular and Cell Biology and Genetics Graduate Program, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
- Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Kayla Schardien
- Molecular and Cell Biology and Genetics Graduate Program, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Brian Wigdahl
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
- Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Michael R Nonnemacher
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA.
- Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA.
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
9
|
Lipov E, Sethi Z, Nandra G, Frueh C. Efficacy of combined subanesthetic ketamine infusion and cervical sympathetic blockade as a symptomatic treatment of PTSD/TBI in a special forces patient with a 1-year follow-up: A case report. Heliyon 2023; 9:e14891. [PMID: 37089345 PMCID: PMC10119561 DOI: 10.1016/j.heliyon.2023.e14891] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 03/13/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
Co-occurrence of posttraumatic stress disorder (PTSD) and traumatic brain injury (TBI) symptoms are particularly prevalent in the special operations forces' community, along with other related conditions (e.g., endocrine dysregulation, sleep disorders, chronic pain). Ketamine infusion (KI) has been shown to increase neuroplasticity as well as memory improvement and cervical sympathetic block (CSB) has been shown to improve cognitive function, reduce sympathetic overactivity, and improve other symptoms of PTSD. We want to report the efficacious use of a single intervention consisting of bilateral CSB technique with subanesthetic KI X5 in a Special Operations Forces patient, diagnosed with PTSD with comorbid TBI, evaluated during treatment and at 1-year follow-up. We postulated KI and CSB would have a synergistic effect. Our patient received KI starting at 0.5 mg/kg, which was escalated daily. KI was combined with right-sided ultrasound-guided CSB (C6 and C4 levels). This was followed the next day by left-sided CSB and KI. Patient's PTSD symptoms were evaluated using the Posttraumatic Stress Disorder Checklist (PCL-5), Beck Depression Inventory (BDI), Beck Anxiety Inventory (BAI), suicidal ideation and other related factors by Concise Health Risk Tracking Self Report (CHRTSR). All measures were assessed prior to treatment, during treatment, and 394 days after. KI combined with CSB showed immediate and prolonged benefits 394 days later regarding the symptoms of PTSD, anxiety, depression, suicidal ideation, and cognitive deterioration (patient report). KI combined with CSB can markedly reduce symptoms of PTSD, psychiatric comorbidities, and cognitive dysfunction.
Collapse
Affiliation(s)
- Eugene Lipov
- Stella Center, Chicago, United States
- Corresponding author.
| | - Zubin Sethi
- Midwestern University Chicago College of Osteopathic Medicine, United States
| | | | - Christopher Frueh
- Department of Psychology, University of Hawaii, Hilo, HI, United States
| |
Collapse
|
10
|
Demin KA, Zabegalov KA, Kolesnikova TO, Galstyan DS, Kositsyn YMHB, Costa FV, de Abreu MS, Kalueff AV. Animal Inflammation-Based Models of Neuropsychiatric Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1411:91-104. [PMID: 36949307 DOI: 10.1007/978-981-19-7376-5_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
Mounting evidence links psychiatric disorders to central and systemic inflammation. Experimental (animal) models of psychiatric disorders are important tools for translational biopsychiatry research and CNS drug discovery. Current experimental models, most typically involving rodents, continue to reveal shared fundamental pathological pathways and biomarkers underlying the pathogenetic link between brain illnesses and neuroinflammation. Recent data also show that various proinflammatory factors can alter brain neurochemistry, modulating the levels of neurohormones and neurotrophins in neurons and microglia. The role of "active" glia in releasing a wide range of proinflammatory cytokines also implicates glial cells in various psychiatric disorders. Here, we discuss recent animal inflammation-related models of psychiatric disorders, focusing on their translational perspectives and the use of some novel promising model organisms (zebrafish), to better understand the evolutionally conservative role of inflammation in neuropsychiatric conditions.
Collapse
Affiliation(s)
- Konstantin A Demin
- Neurobiology Program, Sirius University of Science and Technology, Sochi, Russia
- Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | | | | | - David S Galstyan
- Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
| | - Yuriy M H B Kositsyn
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Fabiano V Costa
- Neurobiology Program, Sirius University of Science and Technology, Sochi, Russia
| | - Murilo S de Abreu
- Laboratory of Cell and Molecular Biology and Neurobiology, Moscow Institute of Physics and Technology, Moscow, Russia
| | - Allan V Kalueff
- Neurobiology Program, Sirius University of Science and Technology, Sochi, Russia
- Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
- Laboratory of Cell and Molecular Biology and Neurobiology, Moscow Institute of Physics and Technology, Moscow, Russia
- Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny, Russia
- Laboratory of Translational Biopsychiatry, Scientific Research Institute of Neuroscience and Medicine, Novosibirsk, Russia
- Ural Federal University, Ekaterinburg, Russia
- Novosibirsk State University, Novosibirsk, Russia
| |
Collapse
|
11
|
Kumar V, Singh C, Singh A. Neuroprotective Potential of Hydroalcoholic Extract of Centella asiatica Against 3-Nitropropionic Acid-Induced Huntington's Like Symptoms in Adult Zebrafish. Rejuvenation Res 2022; 25:260-274. [PMID: 36150031 DOI: 10.1089/rej.2022.0036] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Huntington's disease (HD) is an inherited neurodegenerative disease. 3-Nitropropionic acid (3-NP) causes increased reactive oxygen species production and neuroinflammation. Centella asiatica (CA) is a strong antioxidant. The aim of this study is to investigate the effect of hydroalcoholic extract of C. asiatica (HA-CA) on 3-NP-induced HD in adult zebrafish. Adult zebrafish (∼5-6 months old) weighing 470 to 530 mg was used and treated with 3-NP (5 mg/kg intraperitoneal [i.p.]). The animals received HA-CA (80 and 100 mg/L) daily for up to 28 days in water. Tetrabenazine (3 mg/kg i.p.) was used as a standard drug. We have done an open field test (for locomotor activity), a novel tank diving test (for anxiety), and a light and dark tank test (for memory), followed by biochemical analysis (acetyl-cholinesterase [AchEs], nitrite, lipid peroxidation [LPO], and glutathione [GSH]) and histopathology to further confirm memory dysfunctions. 3-NP-treated zebrafish exhibit reductions in body weight, progressive neuronal damage, cognition, and locomotor activity. The HA-CA group significantly reduced the 3-NP-induced increase in LPO, AchEs, and nitrite levels while decreasing GSH levels. Oral administration of HA-CA (80 or 100 mg/L) significantly reduces 3-NP-induced changes in body weight and behaviors, in addition to neuroinflammation in the brain by lowering tumor necrosis factor-α and interleukin-1β levels. Moreover, HA-CA significantly decreases the 3-NP-induced neuronal damage in the brain. HA-CA ameliorates neurotoxicity and neurobehavioral deficits in 3-NP-induced HD-like symptoms in adult zebrafish.
Collapse
Affiliation(s)
- Vishal Kumar
- Department of Pharmacology, ISF College of Pharmacy, Moga, Affiliated to IK Gujral Punjab Technical University, Jalandhar, India
| | - Charan Singh
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Affiliated to IK Gujral Punjab Technical University, Jalandhar, India
| | - Arti Singh
- Department of Pharmacology, ISF College of Pharmacy, Moga, Affiliated to IK Gujral Punjab Technical University, Jalandhar, India
| |
Collapse
|
12
|
Su Y, Lian J, Chen S, Zhang W, Deng C. Epigenetic histone acetylation modulating prenatal Poly I:C induced neuroinflammation in the prefrontal cortex of rats: a study in a maternal immune activation model. Front Cell Neurosci 2022; 16:1037105. [DOI: 10.3389/fncel.2022.1037105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 11/08/2022] [Indexed: 11/29/2022] Open
Abstract
Introduction: Neuroinflammation in the central nervous system, particularly the prefrontal cortex (PFC), plays a role in the pathogenesis of schizophrenia, which has been found to be associated with maternal immune activation (MIA). Recent evidence suggests that epigenetic regulation involves in the MIA-induced neurodevelopmental disturbance. However, it is not well-understood how epigenetic modulation is involved in the neuroinflammation and pathogenesis of schizophrenia.Methods: This study explored the modulation of histone acetylation in both neuroinflammation and neurotransmission using an MIA rat model induced by prenatal polyriboinosinic-polyribocytidylic acid (Poly I:C) exposure, specifically examining those genes that were previously observed to be impacted by the exposure, including a subunit of nuclear factor kappa-B (Rela), Nod-Like-Receptor family Pyrin domain containing 3 (Nlrp3), NMDA receptor subunit 2A (Grin2a), 5-HT2A (Htr2a), and GABAA subunit β3 (Gabrb3).Results: Our results revealed global changes of histone acetylation on H3 (H3ace) and H4 (H4ace) in the PFC of offspring rats with prenatal Poly I:C exposure. In addition, it revealed enhancement of both H3ace and H4ace binding on the promoter region of Rela, as well as positive correlations between Rela and genes encoding histone acetyltransferases (HATs) including CREB-binding protein (CBP) and E1A-associated protein p300 (EP300). Although there was no change in H3ace or H4ace enrichment on the promoter region of Nlrp3, a significant enhancement of histone deacetylase 6 (HDAC6) binding on the promoter region of Nlrp3 and a positive correlation between Nlrp3 and Hdac6 were also observed. However, prenatal Poly I:C treatment did not lead to any specific changes of H3ace and H4ace on the promoter region of the target genes encoding neurotransmitter receptors in this study.Discussion: These findings demonstrated that epigenetic modulation contributes to NF-κB/NLRP3 mediated neuroinflammation induced by prenatal Poly I:C exposure via enhancement of histone acetylation of H3ace and H4ace on Rela and HDAC6-mediated NLRP3 transcriptional activation. This may further lead to deficits in neurotransmissions and schizophrenia-like behaviors observed in offspring.
Collapse
|
13
|
Im J, Eom HJ, Choi J. Effect of Early-Life Exposure of Polystyrene Microplastics on Behavior and DNA Methylation in Later Life Stage of Zebrafish. ARCHIVES OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2022; 82:558-568. [PMID: 35469368 DOI: 10.1007/s00244-022-00924-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 03/09/2022] [Indexed: 06/14/2023]
Abstract
Microplastic contamination has received increasing attention in recent years, and concern regarding the toxicity of microplastics to the environment and humans has increased. In this study, we investigated the neurodevelopmental toxicity of polystyrene microplastics (PSMPs) in the zebrafish Danio rerio under different exposure scenarios. Zebrafish were exposed to PSMPs during embryonic stage and then allowed the fish to recover. The neurodevelopmental toxic responses were investigated using fish behavior and behavior-related gene expression. Early-life exposure to PSMPs did not alter fish behavior at the early stage; however, it led to hyperactivity later life stage. Generally, oxidative stress (i.e., sod2 and nrf2a)- and nervous system (i.e., slc6a4b, npy, and nrbf2)-related gene expression increased in all PSMPs-exposed fish. DNA hypomethylation was observed in fish challenged for a second time using the same PSMPs. Taken together, the current results imply that PSMPs have neurodevelopmental toxic potential when introduced early in life.
Collapse
Affiliation(s)
- Jeongeun Im
- School of Environmental Engineering, University of Seoul, 163 Siripdaero, Dongdaemun-gu, Seoul, 02504, Republic of Korea
| | - Hyun-Jeong Eom
- School of Environmental Engineering, University of Seoul, 163 Siripdaero, Dongdaemun-gu, Seoul, 02504, Republic of Korea
| | - Jinhee Choi
- School of Environmental Engineering, University of Seoul, 163 Siripdaero, Dongdaemun-gu, Seoul, 02504, Republic of Korea.
| |
Collapse
|
14
|
He Y, Han Y, Liao X, Zou M, Wang Y. Biology of cyclooxygenase-2: An application in depression therapeutics. Front Psychiatry 2022; 13:1037588. [PMID: 36440427 PMCID: PMC9684729 DOI: 10.3389/fpsyt.2022.1037588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/25/2022] [Indexed: 11/11/2022] Open
Abstract
Depressive Disorder is a common mood disorder or affective disorder that is dominated by depressed mood. It is characterized by a high incidence and recurrence. The onset of depression is related to genetic, biological and psychosocial factors. However, the pathogenesis is still unclear. In recent years, there has been an increasing amount of research on the inflammatory hypothesis of depression, in which cyclo-oxygen-ase 2 (COX-2), a pro-inflammatory cytokine, is closely associated with depression. A variety of chemical drugs and natural products have been found to exert therapeutic effects by modulating COX-2 levels. This paper summarizes the relationship between COX-2 and depression in terms of neuroinflammation, intestinal flora, neurotransmitters, HPA axis, mitochondrial dysfunction and hippocampal neuronal damage, which can provide a reference for further preventive control, clinical treatment and scientific research on depression.
Collapse
Affiliation(s)
- Ying He
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Yuanshan Han
- Department of Scientific Research, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Xiaolin Liao
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Manshu Zou
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Yuhong Wang
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China.,Hunan Provincial Key Laboratory for the Prevention and Treatment of Depressive Diseases with Traditional Chinese Medicine, Changsha, China.,Hunan Key Laboratory of Power and Innovative Drugs State Key Laboratory of Ministry Training Bases, Changsha, China
| |
Collapse
|
15
|
de Abreu MS, Demin KA, Giacomini ACVV, Amstislavskaya TG, Strekalova T, Maslov GO, Kositsin Y, Petersen EV, Kalueff AV. Understanding how stress responses and stress-related behaviors have evolved in zebrafish and mammals. Neurobiol Stress 2021; 15:100405. [PMID: 34722834 PMCID: PMC8536782 DOI: 10.1016/j.ynstr.2021.100405] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 09/12/2021] [Accepted: 09/27/2021] [Indexed: 12/27/2022] Open
Abstract
Stress response is essential for the organism to quickly restore physiological homeostasis disturbed by various environmental insults. In addition to well-established physiological cascades, stress also evokes various brain and behavioral responses. Aquatic animal models, including the zebrafish (Danio rerio), have been extensively used to probe pathobiological mechanisms of stress and stress-related brain disorders. Here, we critically discuss the use of zebrafish models for studying mechanisms of stress and modeling its disorders experimentally, with a particular cross-taxon focus on the potential evolution of stress responses from zebrafish to rodents and humans, as well as its translational implications.
Collapse
Affiliation(s)
- Murilo S de Abreu
- Bioscience Institute, University of Passo Fundo (UPF), Passo Fundo, RS, Brazil
- Laboratory of Cell and Molecular Biology and Neurobiology, Moscow Institute of Physics and Technology, Moscow, Russia
| | - Konstantin A Demin
- Institute of Experimental Medicine, Almazov National Medcial Research Center, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
- Granov Russian Scientific Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
| | - Ana C V V Giacomini
- Bioscience Institute, University of Passo Fundo (UPF), Passo Fundo, RS, Brazil
- Postgraduate Program in Environmental Sciences, University of Passo Fundo (UPF), Passo Fundo, RS, Brazil
| | - Tamara G Amstislavskaya
- Scientific Research Institute of Physiology and Basic Medcicine, Novosibirsk, Russia
- Novosibirsk State University, Novosibirsk, Russia
| | | | - Gleb O Maslov
- Neuroscience Program, Sirius University, Sochi, Russia
| | - Yury Kositsin
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
- Neuroscience Program, Sirius University, Sochi, Russia
| | - Elena V Petersen
- Laboratory of Cell and Molecular Biology and Neurobiology, Moscow Institute of Physics and Technology, Moscow, Russia
| | - Allan V Kalueff
- School of Pharmacy, Southwest University, Chongqing, China
- Ural Federal University, Ekaterinburg, Russia
| |
Collapse
|
16
|
Dai Y, Wei T, Shen Z, Bei Y, Lin H, Dai H. Classical HDACs in the regulation of neuroinflammation. Neurochem Int 2021; 150:105182. [PMID: 34509559 DOI: 10.1016/j.neuint.2021.105182] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 08/22/2021] [Accepted: 09/07/2021] [Indexed: 12/15/2022]
Abstract
Neuroinflammation is a key factor of the pathology of various neurological diseases (brain injury, depression, neurodegenerative diseases). It is a complex and orderly process that relies on various types of glial cells and peripheral immune cells. Inhibition of neuroinflammation can reduce the severity of neurological diseases. The initiation, progression, and termination of inflammation require gene activation, epigenetic modification, transcriptional translation, and post-translational regulation, all of which are tightly regulated by different enzymes. Epigenetics refers to the regulation of epigenetic gene expression by epigenetic changes (DNA methylation, histone modification, and non-coding RNAs such as miRNA) that are not dependent on changes in gene sequence and are heritable. Histone deacetylases (HDACs) are a group of important enzymes that regulate epigenetics. They can remove the acetyl group on the lysine ϵ-amino group of the target protein, thereby affecting gene transcription or altering protein activity. HDACs are involved in the regulation of immunity and inflammation. HDAC inhibitor (HDACi) has also become a new hotspot in the research of anti-inflammatory drugs. Therefore, the aim of the current review is to discuss and summarize the role and mechanism of different HDACs in neuroinflammation and the corresponding role of HDACi in neurological diseases, and to providing new ideas for future research on neuroinflammation-related diseases and drug development.
Collapse
Affiliation(s)
- Yunjian Dai
- Department of Pharmacy, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang Province, China
| | - Taofeng Wei
- Department of Pharmacy, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang Province, China
| | - Zexu Shen
- Department of Pharmacy, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang Province, China
| | - Yun Bei
- Department of Pharmacy, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang Province, China
| | - Haoran Lin
- Department of Pharmacy, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang Province, China
| | - Haibin Dai
- Department of Pharmacy, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang Province, China.
| |
Collapse
|
17
|
Lachowicz J, Niedziałek K, Rostkowska E, Szopa A, Świąder K, Szponar J, Serefko A. Zebrafish as an Animal Model for Testing Agents with Antidepressant Potential. Life (Basel) 2021; 11:life11080792. [PMID: 34440536 PMCID: PMC8401799 DOI: 10.3390/life11080792] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 08/01/2021] [Accepted: 08/03/2021] [Indexed: 12/28/2022] Open
Abstract
Depression is a serious mental disease that, according to statistics, affects 320 million people worldwide. Additionally, a current situation related to the COVID-19 pandemic has led to a significant deterioration of mental health in people around the world. So far, rodents have been treated as basic animal models used in studies on this disease, but in recent years, Danio rerio has emerged as a new organism that might serve well in preclinical experiments. Zebrafish have a lot of advantages, such as a quick reproductive cycle, transparent body during the early developmental stages, high genetic and physiological homology to humans, and low costs of maintenance. Here, we discuss the potential of the zebrafish model to be used in behavioral studies focused on testing agents with antidepressant potential.
Collapse
Affiliation(s)
- Joanna Lachowicz
- Student’s Scientific Circle at Laboratory of Preclinical Testing, Medical University of Lublin, Chodźki 1, 20-093 Lublin, Poland; (J.L.); (K.N.)
| | - Karolina Niedziałek
- Student’s Scientific Circle at Laboratory of Preclinical Testing, Medical University of Lublin, Chodźki 1, 20-093 Lublin, Poland; (J.L.); (K.N.)
| | | | - Aleksandra Szopa
- Laboratory of Preclinical Testing, Chair and Department of Applied and Social Pharmacy, Medical University of Lublin, Chodźki 1, 20-093 Lublin, Poland
- Correspondence: (A.S.); (A.S.)
| | - Katarzyna Świąder
- Chair and Department of Applied and Social Pharmacy, Medical University of Lublin, Chodźki 1, 20-093 Lublin, Poland;
| | - Jarosław Szponar
- Clinical Department of Toxicology and Cardiology, Medical University of Lublin, Chodźki 1, 20-093 Lublin, Poland;
- Toxicology Clinic, Stefan Wyszyński Regional Specialist Hospital in Lublin, Al. Kraśnicka 100, 20-718 Lublin, Poland
| | - Anna Serefko
- Laboratory of Preclinical Testing, Chair and Department of Applied and Social Pharmacy, Medical University of Lublin, Chodźki 1, 20-093 Lublin, Poland
- Correspondence: (A.S.); (A.S.)
| |
Collapse
|
18
|
Fontana BD, Müller TE, Cleal M, de Abreu MS, Norton WHJ, Demin KA, Amstislavskaya TG, Petersen EV, Kalueff AV, Parker MO, Rosemberg DB. Using zebrafish (Danio rerio) models to understand the critical role of social interactions in mental health and wellbeing. Prog Neurobiol 2021; 208:101993. [PMID: 33440208 DOI: 10.1016/j.pneurobio.2021.101993] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 11/24/2020] [Accepted: 01/05/2021] [Indexed: 02/07/2023]
Abstract
Social behavior represents a beneficial interaction between conspecifics that is critical for maintaining health and wellbeing. Dysfunctional or poor social interaction are associated with increased risk of physical (e.g., vascular) and psychiatric disorders (e.g., anxiety, depression, and substance abuse). Although the impact of negative and positive social interactions is well-studied, their underlying mechanisms remain poorly understood. Zebrafish have well-characterized social behavior phenotypes, high genetic homology with humans, relative experimental simplicity and the potential for high-throughput screens. Here, we discuss the use of zebrafish as a candidate model organism for studying the fundamental mechanisms underlying social interactions, as well as potential impacts of social isolation on human health and wellbeing. Overall, the growing utility of zebrafish models may improve our understanding of how the presence and absence of social interactions can differentially modulate various molecular and physiological biomarkers, as well as a wide range of other behaviors.
Collapse
Affiliation(s)
- Barbara D Fontana
- Brain and Behaviour Laboratory, School of Pharmacy and Biomedical Sciences, University of Portsmouth, UK.
| | - Talise E Müller
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Natural and Exact Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil; Laboratory of Experimental Neuropscychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Madeleine Cleal
- Brain and Behaviour Laboratory, School of Pharmacy and Biomedical Sciences, University of Portsmouth, UK
| | - Murilo S de Abreu
- Bioscience Institute, University of Passo Fundo, Passo Fundo, Brazil
| | - William H J Norton
- Department of Neuroscience, Psychology and Behaviour, College of Medicine, Biological Sciences and Psychology, University of Leicester, Leicester, UK; The International Zebrafish Neuroscience Research Consortium (ZNRC), Slidell, LA, USA
| | - Konstantin A Demin
- Institute of Experimental Medicine, Almazov National Medical Research Center, St. Petersburg, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Scientific Research Center of Radiology and Surgical Technologies, St. Petersburg, Russia
| | | | - Elena V Petersen
- Laboratory of Molecular Biology, Neuroscience and Bioscreening, Moscow Institute of Physics and Technology, Moscow, Russia
| | - Allan V Kalueff
- School of Pharmacy, Southwest University, Beibei, Chongqing, China; Ural Federal University, Ekaterinburg, Russia
| | - Matthew O Parker
- Brain and Behaviour Laboratory, School of Pharmacy and Biomedical Sciences, University of Portsmouth, UK; The International Zebrafish Neuroscience Research Consortium (ZNRC), Slidell, LA, USA
| | - Denis B Rosemberg
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Natural and Exact Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil; Laboratory of Experimental Neuropscychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil; The International Zebrafish Neuroscience Research Consortium (ZNRC), Slidell, LA, USA.
| |
Collapse
|
19
|
Zabegalov KN, Wang D, Yang L, Wang J, Hu G, Serikuly N, Alpyshov ET, Khatsko SL, Zhdanov A, Demin KA, Galstyan DS, Volgin AD, de Abreu MS, Strekalova T, Song C, Amstislavskaya TG, Sysoev Y, Musienko PE, Kalueff AV. Decoding the role of zebrafish neuroglia in CNS disease modeling. Brain Res Bull 2020; 166:44-53. [PMID: 33027679 DOI: 10.1016/j.brainresbull.2020.09.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 09/14/2020] [Accepted: 09/25/2020] [Indexed: 12/19/2022]
Abstract
Neuroglia, including microglia and astrocytes, is a critical component of the central nervous system (CNS) that interacts with neurons to modulate brain activity, development, metabolism and signaling pathways. Thus, a better understanding of the role of neuroglia in the brain is critical. Complementing clinical and rodent data, the zebrafish (Danio rerio) is rapidly becoming an important model organism to probe the role of neuroglia in brain disorders. With high genetic and physiological similarity to humans and rodents, zebrafish possess some common (shared), as well as some specific molecular biomarkers and features of neuroglia development and functioning. Studying these common and zebrafish-specific aspects of neuroglia may generate important insights into key brain mechanisms, including neurodevelopmental, neurodegenerative, neuroregenerative and neurological processes. Here, we discuss the biology of neuroglia in humans, rodents and fish, its role in various CNS functions, and further directions of translational research into the role of neuroglia in CNS disorders using zebrafish models.
Collapse
Affiliation(s)
- Konstantin N Zabegalov
- School of Pharmacy, Southwest University, Chongqing, China; Ural Federal University, Ekaterinburg, Russia
| | - Dongmei Wang
- School of Pharmacy, Southwest University, Chongqing, China
| | - LongEn Yang
- School of Pharmacy, Southwest University, Chongqing, China
| | - Jingtao Wang
- School of Pharmacy, Southwest University, Chongqing, China
| | - Guojun Hu
- School of Pharmacy, Southwest University, Chongqing, China
| | - Nazar Serikuly
- School of Pharmacy, Southwest University, Chongqing, China
| | | | | | | | - Konstantin A Demin
- Institute of Experimental Medicine, Almazov National Medical Research Centre, St. Petersburg, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - David S Galstyan
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Laboratory of Cell and Molecular Biology and Neurobiology, Moscow Institute of Physics and Technology, Moscow, Russia
| | - Andrey D Volgin
- Scientific Research Institute of Neurosciences and Medicine, Novosibirsk, Russia; Laboratory of Cell and Molecular Biology and Neurobiology, Moscow Institute of Physics and Technology, Moscow, Russia
| | - Murilo S de Abreu
- Bioscience Institute, University of Passo Fundo, Passo Fundo, Brazil; Laboratory of Cell and Molecular Biology and Neurobiology, Moscow Institute of Physics and Technology, Moscow, Russia.
| | - Tatyana Strekalova
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands; Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia; Division of Molecular Psychiatry, Centre of Mental Health, University of Würzburg, Würzburg, Germany
| | - Cai Song
- Institute for Marine Drugs and Nutrition, Guangdong Ocean University, Zhanjiang, China; Marine Medicine Development Center, Shenzhen Institute, Guangdong Ocean University, Shenzhen, China
| | - Tamara G Amstislavskaya
- Scientific Research Institute of Neurosciences and Medicine, Novosibirsk, Russia; Zelman Institute of Medicine and Psychology, Novosibirsk State University, Novosibirsk, Russia
| | - Yury Sysoev
- Laboratory of Neuroprosthetics, Institute of Translational Biomedicine, Petersburg State University, St. Petersburg, Russia; Department of Pharmacology and Clinical Pharmacology, St. Petersburg State Chemical Pharmaceutical University, St. Petersburg, Russia
| | - Pavel E Musienko
- Laboratory of Neuroprosthetics, Institute of Translational Biomedicine, Petersburg State University, St. Petersburg, Russia; Institute of Phthisiopulmonology, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
| | - Allan V Kalueff
- School of Pharmacy, Southwest University, Chongqing, China; Ural Federal University, Ekaterinburg, Russia.
| |
Collapse
|