1
|
Silva AO, Ribeiro JM, Soares NP, Oliveira KCM, Espuri PF, Belo TCA, Reis LFCD, Aguiar DCD, Paula FBDA, Ruginsk SG, Almeida LAD, Marques MJ, José AR, Elias LLK, Torres LHL, Cau S, Ceron CS. Minocycline treatment attenuates high-refined carbohydrate diet-induced gut bacterial dysbiosis, anxiety-like behaviour, and cardiac damage in mice. Eur J Pharmacol 2025; 996:177552. [PMID: 40154569 DOI: 10.1016/j.ejphar.2025.177552] [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: 12/10/2024] [Revised: 03/12/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
The high-refined carbohydrate diet (HC diet) is linked to anxiety development and oxidative damage to heart tissue. However, little is known about how the gut microbiota profile is modulated in this diet model. Minocycline is an antibiotic with anti-inflammatory, antioxidant, and matrix metalloproteinases (MMPs) inhibitor properties. Therefore, we evaluated the effects of minocycline treatment on HC diet-induced cardiac damage, anxiety-like behaviour, and bacterial gut dysbiosis in mice. Male BALB/C mice were divided into two groups, which received standard diet or HC diet for 12 weeks. In the 10th week, both groups were subdivided and received water or minocycline (50 mg/kg) by gavage for 15 days. The gut bacterial populations, behavioural parameters, adiposity index, biochemical profile, cardiac oxidative stress indicators, MMPs, cardiac remodelling, and contractile analyses by Langendorff-perfused hearts were analysed. The HC diet induced bacterial gut dysbiosis and anxiety-like behaviour increased the adiposity index with changes in the lipid profile and creatine kinase fraction MB (CK-MB). In the heart, the HC diet increased tissue oxidative stress, MMP-2 and MMP-9 activity, collagen deposition, and altered cardiac performance. Minocycline treatment reversed diet-induced bacterial gut dysbiosis and anxiety-like behaviour, ameliorated the biochemical profile, diminished oxidative stress, MMP activity, cardiac collagen deposition, and improved cardiac performance. These findings suggest that minocycline treatment modulated the microbiota and attenuated behavioural changes and cardiac damage caused by the HC diet, suggesting an interplay between the gut-microbiota-brain axis and cardiac damage caused by the HC diet consumption.
Collapse
Affiliation(s)
- Alessandra Oliveira Silva
- Department of Food and Medicine, Faculty of Pharmaceutical Sciences, Federal University of Alfenas, Alfenas, Brazil
| | - Jéssyca Milene Ribeiro
- Department of Food and Medicine, Faculty of Pharmaceutical Sciences, Federal University of Alfenas, Alfenas, Brazil
| | - Nícia Pedreira Soares
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Patrícia Ferreira Espuri
- Department of Pathology and Parasitology, Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, Brazil
| | - Thiago Caetano Andrade Belo
- Department of Microbiology and Immunology, Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, Brazil
| | - Luis Felipe Cunha Dos Reis
- Department of Structural Biology, Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, Brazil
| | - Daniele Cristina de Aguiar
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Fernanda Borges de Araújo Paula
- Department of Clinical and Toxicological Analysis, Faculty of Pharmaceutical Sciences, Federal University of Alfenas, Alfenas, Brazil
| | - Sílvia Graciela Ruginsk
- Department of Physiological Sciences, Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, Brazil
| | - Leonardo Augusto de Almeida
- Department of Microbiology and Immunology, Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, Brazil
| | - Marcos José Marques
- Department of Pathology and Parasitology, Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, Brazil
| | - Antunes-Rodrigues José
- Department of Physiology, Faculty of Medicine, University of São Paulo, Ribeirão Preto, SP, Brazil
| | | | - Larissa Helena Lobo Torres
- Department of Food and Medicine, Faculty of Pharmaceutical Sciences, Federal University of Alfenas, Alfenas, Brazil
| | - Stefany Cau
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Carla Speroni Ceron
- Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, MG, Brazil.
| |
Collapse
|
2
|
Hosseini M, Bardaghi Z, Askarpour H, Rajabian A, Mahmoudabady M, Shabab S, Samadi‐Noshahr Z, Salmani H. Minocycline mitigates sepsis-induced neuroinflammation and promotes recovery in male mice: Insights into neuroprotection and inflammatory modulation. Physiol Rep 2024; 12:e70032. [PMID: 39370294 PMCID: PMC11456363 DOI: 10.14814/phy2.70032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/13/2024] [Accepted: 08/26/2024] [Indexed: 10/08/2024] Open
Abstract
Sepsis is associated with brain injury and acute brain inflammation, which can potentially transition into chronic inflammation, triggering a cascade of inflammatory responses that may lead to neurological disorders. Minocycline, recognized for its potent anti-inflammatory properties, was investigated in this study for its protective effects against sepsis-induced brain injury. Adult male C57 mice pretreated with minocycline (12.5, 25, and 50 mg/kg) 3 days before sepsis induction. An intraperitoneal injection of 5 mg/kg LPS was used to induce sepsis. Spontaneous locomotor activity (SLA) and weight changes were assessed over several days post-sepsis to monitor the recovery of the mice. The expression of inflammatory mediators and oxidative stress markers was assessed 24 h post sepsis. Septic mice exhibited significant weight loss and impaired SLA. Initially, minocycline did not attenuate the severity of weight loss (1 day) or SLA (4 h post-sepsis), but it significantly accelerated the recovery of the mice in later days. Minocycline dose-dependently mitigated sepsis-induced brain inflammation and oxidative stress. Our findings demonstrate that pretreatment with minocycline has the potential to prevent brain tissue damage and accelerate recovery from sepsis in mice, suggesting that minocycline may serve as a promising therapeutic intervention to protect against sepsis-induced neurological complications.
Collapse
Affiliation(s)
- Mahmoud Hosseini
- Psychiatry and Behavioral Sciences Research CenterMashhad University of Medical SciencesMashhadIran
- Applied Biomedical Research CenterMashhad University of Medical SciencesMashhadIran
| | - Zahra Bardaghi
- Psychiatry and Behavioral Sciences Research CenterMashhad University of Medical SciencesMashhadIran
- Department of Developmental BiologyWashington University School of MedicineSt. LouisMissouriUSA
| | - Hedyeh Askarpour
- Bioenvironmental Health Hazards Research CenterJiroft University of Medical SciencesJiroftIran
| | - Arezoo Rajabian
- Neuroscience Research CenterMashhad University of Medical SciencesMashhadIran
| | - Maryam Mahmoudabady
- Applied Biomedical Research CenterMashhad University of Medical SciencesMashhadIran
| | - Sadegh Shabab
- Applied Biomedical Research CenterMashhad University of Medical SciencesMashhadIran
| | - Zahra Samadi‐Noshahr
- Chabahar Faculty of Medical Sciences, School of MedicineIranshahr University of Medical SciencesChabaharIran
| | - Hossein Salmani
- Bioenvironmental Health Hazards Research CenterJiroft University of Medical SciencesJiroftIran
- Department of Physiology and Pharmacology, Faculty of MedicineSabzevar University of Medical SciencesMashhadIran
| |
Collapse
|
3
|
Gholami M, Ghelichkhani Z, Aghakhani R, Klionsky DJ, Motaghinejad O, Motaghinejad M, Koohi MK, Hassan J. Minocycline Acts as a Neuroprotective Agent Against Tramadol-Induced Neurodegeneration: Behavioral and Molecular Evidence. Int J Prev Med 2024; 15:47. [PMID: 39539580 PMCID: PMC11559692 DOI: 10.4103/ijpvm.ijpvm_10_24] [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: 01/04/2024] [Accepted: 06/04/2024] [Indexed: 11/16/2024] Open
Abstract
Background Previous evidence indicates that tramadol (TRA) can lead to neurodegenerative events and minocycline (MIN) has neuroprotective properties. Aim of the Study The current research evaluated the neuroprotective effects of MIN for TRA-promoted neurodegeneration. Methods Sixty adult male rats were placed into the following groups: 1 (received 0.7 ml/rat of normal saline, IP), 2 (received 50 mg/kg of TRA, i.p.), 3, 4, 5 (administered TRA as 50 mg/kg simultaneously with MIN at 20, 40, and 60 mg/kg, IP, respectively), and 6 (received MIN alone as 60 mg/kg, IP). The treatment procedure was 21 days. An open field test (OFT) was used to measure motor activity and anxiety-related behavior. Furthermore, oxidative stress; hippocampal inflammation; apoptotic parameters as well as activity of mitochondrial complexes I, II, III, and IV; ATP levels; and mitochondrial membrane potential (MMP) were evaluated. In addition, histomorphological alteration was assessed in two regions of the hippocampus: Cornu Ammonis (CA1) and dentate gyrus (DG). Results MIN treatment could inhibit TRA-induced anxiety and motor activity disturbances (P < 0.05). In addition, MIN could attenuate reactive oxygen species (ROS), H2O2, oxidized glutathione (GSSG), and malondialdehyde (MDA) level (P < 0.05), while there was increased reduced glutathione (GSH), total antioxidant capacity (TAC), ATP, MMP, and BCL2 levels (P < 0.05) and also elevation of SOD, GPX, GSR (P < 0.05), and mitochondrial complexes I, II, III, and IV activity (P < 0.05) in TRA-treated rats. In consistence with these findings, MIN could reduce TNF/TNF-α, IL1B/IL1-β, BAX, and CASP3 levels (P < 0.05) in TRA-treated rats. MIN also restored the quantitative (P < 0.05) and qualitative histomorphological sequels of TRA in both CA1 and DG areas of the hippocampus. Conclusions MIN probably has repositioning capability for inhibition of TRA-induced neurodegeneration via modulation of inflammation, oxidative stress, apoptosis, and mitochondrial disorders.
Collapse
Affiliation(s)
- Mina Gholami
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Reza Aghakhani
- Faculty of Veterinary Medicine, Semnan University, Semnan, Iran
| | | | - Ozra Motaghinejad
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Majid Motaghinejad
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Kazem Koohi
- Department of Comparative Bioscience, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Jalal Hassan
- Division of Toxicology, Department of Comparative Bioscience, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| |
Collapse
|
4
|
Mori R, Abe M, Saimoto Y, Shinto S, Jodai S, Tomomatsu M, Tazoe K, Ishida M, Enoki M, Kato N, Yamashita T, Itabashi Y, Nakanishi I, Ohkubo K, Kaidzu S, Tanito M, Matsuoka Y, Morimoto K, Yamada KI. Construction of a screening system for lipid-derived radical inhibitors and validation of hit compounds to target retinal and cerebrovascular diseases. Redox Biol 2024; 73:103186. [PMID: 38744193 PMCID: PMC11109892 DOI: 10.1016/j.redox.2024.103186] [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: 04/16/2024] [Accepted: 05/07/2024] [Indexed: 05/16/2024] Open
Abstract
Recent studies have highlighted the indispensable role of oxidized lipids in inflammatory responses, cell death, and disease pathogenesis. Consequently, inhibitors targeting oxidized lipids, particularly lipid-derived radicals critical in lipid peroxidation, which are known as radical-trapping antioxidants (RTAs), have been actively pursued. We focused our investigation on nitroxide compounds that have rapid second-order reaction rate constants for reaction with lipid-derived radicals. A novel screening system was developed by employing competitive reactions between library compounds and a newly developed profluorescence nitroxide probe with lipid-derived radicals to identify RTA compounds. A PubMed search of the top hit compounds revealed their wide application as repositioned drugs. Notably, the inhibitory efficacy of methyldopa, selected from these compounds, against retinal damage and bilateral common carotid artery stenosis was confirmed in animal models. These findings underscore the efficacy of our screening system and suggest that it is an effective approach for the discovery of RTA compounds.
Collapse
Affiliation(s)
- Ryota Mori
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Masami Abe
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yuma Saimoto
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Saki Shinto
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Sara Jodai
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Manami Tomomatsu
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kaho Tazoe
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Minato Ishida
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Masataka Enoki
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Nao Kato
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Tomohiro Yamashita
- Department of Drug Discovery Structural Biology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yuki Itabashi
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Ikuo Nakanishi
- Quantum RedOx Chemistry Team, Institute for Quantum Life Science (iQLS), Quantum Life and Medical Science Directorate (QLMS), National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| | - Kei Ohkubo
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan; Quantum RedOx Chemistry Team, Institute for Quantum Life Science (iQLS), Quantum Life and Medical Science Directorate (QLMS), National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan; Institute for Advanced Co-Creation Studies, Osaka University, 1-6 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Sachiko Kaidzu
- Department of Ophthalmology, Shimane University Faculty of Medicine, 89-1 Enya Izumo, Shimane, 693-8501, Japan
| | - Masaki Tanito
- Department of Ophthalmology, Shimane University Faculty of Medicine, 89-1 Enya Izumo, Shimane, 693-8501, Japan
| | - Yuta Matsuoka
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kazushi Morimoto
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Ken-Ichi Yamada
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
5
|
Zhao K, Wang P, Tang X, Chang N, Shi H, Guo L, Wang B, Yang P, Zhu T, Zhao X. The mechanisms of minocycline in alleviating ischemic stroke damage and cerebral ischemia-reperfusion injury. Eur J Pharmacol 2023; 955:175903. [PMID: 37422120 DOI: 10.1016/j.ejphar.2023.175903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/08/2023] [Accepted: 07/05/2023] [Indexed: 07/10/2023]
Abstract
Stroke is a group of diseases resulting from cerebral vascular rupture or obstruction and subsequent brain blood circulation disorder, leading to rapid neurological deficits. Ischemic stroke accounts for the majority of all stroke cases. The current treatments for ischemic stroke mainly include t-PA thrombolytic therapy and surgical thrombectomy. However, these interventions aimed at recanalizing cerebral vessels can paradoxically lead to ischemia-reperfusion injury, which exacerbates the severity of brain damage. Minocycline, a semi-synthetic tetracycline antibiotic, has been shown to possess a wide range of neuroprotective effects independent of its antibacterial activity. Here we summarize the mechanisms underlying the protective effects of minocycline against cerebral ischemia-reperfusion injury based on the pathogenesis of cerebral ischemia-reperfusion injury, including its modulation of oxidative stress, inflammatory response, excitotoxicity, programmed cell death and blood-brain barrier injury, and also introduce the role of minocycline in alleviating stroke-related complications, in order to provide a theoretical basis for the clinical application of minocycline in cerebral ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Kemeng Zhao
- Basic Medical College, Xinxiang Medical University, Xinxiang, China; College of First Clinical, Xinxiang Medical University, Xinxiang, China
| | - Pengwei Wang
- Department of Pharmacy, The First Affiliated Hospital of Xinxiang Medical University, No. 88 Jiankang Road, Weihui, 453100, Henan, China
| | - Xiaoguang Tang
- College of Pharamacy, Xinxiang Medical University, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Na Chang
- College of Pharamacy, Xinxiang Medical University, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Haonan Shi
- Sanquan Medical College, Xinxiang Medical University, Xinxiang, China
| | - Longfei Guo
- College of Pharamacy, Xinxiang Medical University, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Bingyi Wang
- College of Pharamacy, Xinxiang Medical University, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Pengfei Yang
- College of Pharamacy, Xinxiang Medical University, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.
| | - Tiantian Zhu
- College of Pharamacy, Xinxiang Medical University, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.
| | - Xinghua Zhao
- Basic Medical College, Xinxiang Medical University, Xinxiang, China.
| |
Collapse
|
6
|
de Medeiros Borges H, Dagostin CS, Córneo E, Dondossola ER, Bernardo HT, Pickler KDP, da Costa Pereira B, de Oliveira MA, Scussel R, Michels M, Machado-de-Ávila RA, Dal-Pizzol F, Rico EP. Zebrafish as a potential model for stroke: A comparative study with standardized models. Life Sci 2022; 312:121200. [PMID: 36435227 DOI: 10.1016/j.lfs.2022.121200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/11/2022] [Accepted: 11/12/2022] [Indexed: 11/25/2022]
Abstract
Animal models of cerebral ischemia have improved our understanding of the pathophysiology and mechanisms involved in stroke, as well as the investigation of potential therapies. The potential of zebrafish to model human diseases has become increasingly evident. The availability of these models allows for an increased understanding of the role of chemical exposure in human conditions and provides essential tools for mechanistic studies of disease. To evaluate the potential neuroprotective properties of minocycline against ischemia and reperfusion injury in zebrafish and compare them with other standardized models. In vitro studies with BV-2 cells were performed, and mammalian transient middle cerebral artery occlusion (tMCAO) was used as a comparative standard with the zebrafish stroke model. Animals were subjected to ischemia and reperfusion injury protocols and treated with minocycline. Infarction size, cytokine levels, oxidative stress, glutamate toxicity, and immunofluorescence for microglial activation, and behavioral test results were determined and compared. Administration of minocycline provided significant protection in the three stroke models in different parameters analyzed. Both experimental models complement each other in their particularities. The proposal also strengthens the findings in the literature in rodent models and allows the validation of alternative models so that they can be used in further research involving diseases with ischemia and reperfusion injury.
Collapse
Affiliation(s)
- Heloisa de Medeiros Borges
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil; Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Caroline Serafim Dagostin
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Emily Córneo
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Eduardo Ronconi Dondossola
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Henrique Teza Bernardo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Karolyne De Pieri Pickler
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Bárbara da Costa Pereira
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Mariane Amanda de Oliveira
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Rahisa Scussel
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Monique Michels
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil; Gabbia Biotechnology Company, Barra Velha, Santa Catarina, Brazil
| | - Ricardo Andrez Machado-de-Ávila
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Felipe Dal-Pizzol
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Eduardo Pacheco Rico
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| |
Collapse
|
7
|
Marcatili M, Borgonovo R, Cimminiello N, Cornaggia RD, Casati G, Pellicioli C, Maggioni L, Motta F, Redaelli C, Ledda L, Pozzi FE, Krivosova M, Pagano J, Nava R, Colmegna F, Dakanalis A, Caldiroli A, Capuzzi E, Benatti B, Dell’Osso B, Bertola F, Villa N, Piperno A, Ippolito S, Appollonio I, Sala C, Conti L, Clerici M. Possible Use of Minocycline in Adjunction to Intranasal Esketamine for the Management of Difficult to Treat Depression following Extensive Pharmacogenomic Testing: Two Case Reports. J Pers Med 2022; 12:jpm12091524. [PMID: 36143309 PMCID: PMC9503866 DOI: 10.3390/jpm12091524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/11/2022] [Accepted: 09/12/2022] [Indexed: 11/17/2022] Open
Abstract
The advent of intra-nasal esketamine (ESK), one of the first so called fast-acting antidepressant, promises to revolutionize the management of treatment resistant depression (TRD). This NMDA receptor antagonist has proven to be rapidly effective in the short- and medium-term course of the illness, revealing its potential in targeting response in TRD. Although many TRD ESK responders are able to achieve remission, a considerable portion of them undergo a metamorphosis of their depression into different clinical presentations, characterized by instable responses and high recurrence rates that can be considered closer to the concept of Difficult to Treat Depression (DTD) than to TRD. The management of these DTD patients usually requires a further complex multidisciplinary approach and can benefit from the valuable contribution of new personalized medicine tools such as therapeutic drug monitoring and pharmacogenetics. Despite this, these patients usually come with long and complex previous treatments history and, often, advanced and sophisticated ongoing pharmacological schemes that can make the finding of new alternative options to face the current recurrences extremely challenging. In this paper, we describe two DTD patients—already receiving intranasal ESK but showing an instable course—who were clinically stabilized by the association with minocycline, a semisynthetic second-generation tetracycline with known and promising antidepressant properties.
Collapse
Affiliation(s)
- Matteo Marcatili
- Psychiatric Department, San Gerardo Hospital, ASST Monza, 20900 Monza, Italy
- Correspondence:
| | - Riccardo Borgonovo
- Department of Medicine and Surgery, University of Milano Bicocca, 20900 Monza, Italy
| | - Noemi Cimminiello
- Department of Medicine and Surgery, University of Milano Bicocca, 20900 Monza, Italy
| | | | - Giulia Casati
- Department of Medicine and Surgery, University of Milano Bicocca, 20900 Monza, Italy
| | - Cristian Pellicioli
- Department of Medicine and Surgery, University of Milano Bicocca, 20900 Monza, Italy
| | - Laura Maggioni
- Department of Medicine and Surgery, University of Milano Bicocca, 20900 Monza, Italy
| | - Federico Motta
- Department of Medicine and Surgery, University of Milano Bicocca, 20900 Monza, Italy
| | - Chiara Redaelli
- Department of Medicine and Surgery, University of Milano Bicocca, 20900 Monza, Italy
| | - Luisa Ledda
- Department of Medicine and Surgery, University of Milano Bicocca, 20900 Monza, Italy
| | - Federico Emanuele Pozzi
- Department of Medicine and Surgery, University of Milano Bicocca, 20900 Monza, Italy
- Department of Neurology, San Gerardo Hospital, ASST Monza, 20900 Monza, Italy
| | - Michaela Krivosova
- Department of Medicine and Surgery, University of Milano Bicocca, 20900 Monza, Italy
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Jessica Pagano
- National Research Council Neuroscience Institute, 20100 Milan, Italy
| | - Roberto Nava
- Psychiatric Department, San Gerardo Hospital, ASST Monza, 20900 Monza, Italy
| | - Fabrizia Colmegna
- Psychiatric Department, San Gerardo Hospital, ASST Monza, 20900 Monza, Italy
| | - Antonios Dakanalis
- Department of Medicine and Surgery, University of Milano Bicocca, 20900 Monza, Italy
| | - Alice Caldiroli
- Psychiatric Department, San Gerardo Hospital, ASST Monza, 20900 Monza, Italy
| | - Enrico Capuzzi
- Psychiatric Department, San Gerardo Hospital, ASST Monza, 20900 Monza, Italy
| | - Beatrice Benatti
- Psychiatry Unit, Department of Biomedical and Clinical Sciences “Luigi Sacco”, University of Milan, 20100 Milan, Italy
- CRC “Aldo Ravelli” for Neurotechnology and Experimental Brain Therapeutics, University of Milan, 20100 Milan, Italy
| | - Bernardo Dell’Osso
- Psychiatry Unit, Department of Biomedical and Clinical Sciences “Luigi Sacco”, University of Milan, 20100 Milan, Italy
- CRC “Aldo Ravelli” for Neurotechnology and Experimental Brain Therapeutics, University of Milan, 20100 Milan, Italy
| | - Francesca Bertola
- Cytogenetics and Medical Genetics Unit, Centre for Disorders of Iron Metabolism, San Gerardo Hospital, ASST Monza, 20900 Monza, Italy
| | - Nicoletta Villa
- Cytogenetics and Medical Genetics Unit, Centre for Disorders of Iron Metabolism, San Gerardo Hospital, ASST Monza, 20900 Monza, Italy
| | - Alberto Piperno
- Department of Medicine and Surgery, University of Milano Bicocca, 20900 Monza, Italy
- Cytogenetics and Medical Genetics Unit, Centre for Disorders of Iron Metabolism, San Gerardo Hospital, ASST Monza, 20900 Monza, Italy
| | - Silvia Ippolito
- Clinical Chemistry Laboratory, San Gerardo Hospital, ASST Monza, 20900 Monza, Italy
| | - Ildebrando Appollonio
- Department of Medicine and Surgery, University of Milano Bicocca, 20900 Monza, Italy
- Department of Neurology, San Gerardo Hospital, ASST Monza, 20900 Monza, Italy
| | - Carlo Sala
- National Research Council Neuroscience Institute, 20100 Milan, Italy
| | - Luciano Conti
- Laboratory of Stem Cell Biology, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38122 Trento, Italy
| | - Massimo Clerici
- Psychiatric Department, San Gerardo Hospital, ASST Monza, 20900 Monza, Italy
- Department of Medicine and Surgery, University of Milano Bicocca, 20900 Monza, Italy
| |
Collapse
|
8
|
Motaghinejad M, Motevalian M. Neuroprotective Properties of Minocycline Against Methylphenidate-Induced Neurodegeneration: Possible Role of CREB/BDNF and Akt/GSK3 Signaling Pathways in Rat Hippocampus. Neurotox Res 2022; 40:689-713. [PMID: 35446003 DOI: 10.1007/s12640-021-00454-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/17/2021] [Accepted: 11/29/2021] [Indexed: 12/19/2022]
Abstract
Neurodegeneration is a side effect of methylphenidate (MPH), and minocycline possesses neuroprotective properties. This study aimed to investigate the neuroprotective effects of minocycline against methylphenidate-induced neurodegeneration mediated by signaling pathways of CREB/BDNF and Akt/GSK3. Seven groups of seventy male rats were randomly distributed in seven groups (n = 10). Group 1 received 0.7 ml/rat of normal saline (i.p.), and group 2 was treated with MPH (10 mg/kg, i.p.). Groups 3, 4, 5, and 6 were simultaneously administered MPH (10 mg/kg) and minocycline (10, 20, 30, and 40 mg/kg, i.p.) for 21 days. Minocycline alone (40 mg/kg, i.p.) was administrated to group 7. Open field test (OFT) (on day 22), forced swim test (FST) (on day 24), and elevated plus maze (on day 26) were conducted to analyze the mood-related behaviors; hippocampal oxidative stress, inflammatory, and apoptotic parameters, as well as the levels of protein kinase B (Akt-1), glycogen synthase kinase 3 (GSK3), cAMP response element-binding protein (CREB), and brain-derived neurotrophic factor (BDNF), were also assessed. Furthermore, localization of total CREB, Akt, and GSK3 in the DG and CA1 areas of the hippocampus were measured using immunohistochemistry (IHC). Histological changes in the mentioned areas were also evaluated. Minocycline treatment inhibited MPH-induced mood disorders and decreased lipid peroxidation, oxidized form of glutathione (GSSG), interleukin 1 beta (IL-1β), alpha tumor necrosis factor (TNF-α), Bax, and GSK3 levels. In the contrary, it increased the levels of reduced form of glutathione (GSH), Bcl-2, CREB, BDNF, and Akt-1 and superoxide dismutase (SOD), glutathione peroxidase (GPx), and glutathione reductase (GR) activities in the experimental animals' hippocampus. IHC data showed that minocycline also improved the localization and expression of CREB and Akt positive cells and decreased the GSK3 positive cells in the DG and CA1 regions of the hippocampus of MPH-treated rats. Minocycline also inhibited MPH-induced changes of hippocampal cells' density and shape in both DG and CA1 areas of the hippocampus. According to obtained data, it can be concluded that minocycline probably via activation of the P-CREB/BDNF or Akt/GSK3 signaling pathway can confer its neuroprotective effects against MPH-induced neurodegeneration.
Collapse
Affiliation(s)
- Majid Motaghinejad
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Shahid Hemmat High way, Iran Univ. Med. Sci., P.O. Box 14496-14525, Tehran, Iran.
| | - Manijeh Motevalian
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Shahid Hemmat High way, Iran Univ. Med. Sci., P.O. Box 14496-14525, Tehran, Iran.
- Department of Pharmacology, School of Medicine, Tehran, Iran University of Medical Sciences, Shahid Hemmat High way, Iran Univ. Med. Sci., P.O. Box 14496-14525, Tehran, Iran.
| |
Collapse
|
9
|
Entezari Z, Jahanabadi S. Anticonvulsant Effect of Minocycline on Pentylenetetrazole-Induced Seizure in Mice: Involvement of 5-HT3 Receptor. Drug Res (Stuttg) 2022; 72:268-273. [PMID: 35426093 DOI: 10.1055/a-1783-7836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Minocycline, widely used as an antibiotic, has recently been found to have an anti-inflammatory, neuroprotective and anticonvulsant effects. This study was aimed to investigate the anticonvulsant effect of acute administration of minocycline on pentylenetetrazole (PTZ)-induced seizures considering the possible involvement of 5-HT3 receptor in this effect. For this purpose, seizures were induced by intravenous PTZ infusion. All drugs were administrated by intraperitoneal (i.p.) route before PTZ injection. Also, 1-(m-chlorophenyl)-biguanide (mCPBG, a 5-HT3 receptor agonist) and Tropisetron (a 5-HT3 receptor antagonist) were used 45 minutes before minocycline treatment. Our results demonstrate that acute minocycline treatment (80 and 120 mg/kg) increased the seizure threshold. In addition, the 5-HT3 antagonist, tropisetron, at doses that had no effect on seizure threshold, augmented the anticonvulsant effect of minocycline (40 mg/kg), while mCPBG (0.2 mg/kg) blunted the anticonvulsant effect of minocycline (80 mg/kg). In conclusion, our findings revealed that the anticonvulsant effect of minocycline is mediated, at least in part, by inhibition of 5-HT3 receptor.
Collapse
Affiliation(s)
- Zahra Entezari
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Pharmaceutical Science Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Samane Jahanabadi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Pharmaceutical Science Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
10
|
Jurcau A, Simion A. Neuroinflammation in Cerebral Ischemia and Ischemia/Reperfusion Injuries: From Pathophysiology to Therapeutic Strategies. Int J Mol Sci 2021; 23:14. [PMID: 35008440 PMCID: PMC8744548 DOI: 10.3390/ijms23010014] [Citation(s) in RCA: 242] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 12/18/2021] [Accepted: 12/18/2021] [Indexed: 02/07/2023] Open
Abstract
Its increasing incidence has led stroke to be the second leading cause of death worldwide. Despite significant advances in recanalization strategies, patients are still at risk for ischemia/reperfusion injuries in this pathophysiology, in which neuroinflammation is significantly involved. Research has shown that in the acute phase, neuroinflammatory cascades lead to apoptosis, disruption of the blood-brain barrier, cerebral edema, and hemorrhagic transformation, while in later stages, these pathways support tissue repair and functional recovery. The present review discusses the various cell types and the mechanisms through which neuroinflammation contributes to parenchymal injury and tissue repair, as well as therapeutic attempts made in vitro, in animal experiments, and in clinical trials which target neuroinflammation, highlighting future therapeutic perspectives.
Collapse
Affiliation(s)
- Anamaria Jurcau
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania;
- Neurology Ward, Clinical Municipal Hospital “dr. G. Curteanu” Oradea, 410154 Oradea, Romania
| | - Aurel Simion
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania;
- Neurorehabilitation Ward, Clinical Municipal Hospital “dr. G. Curteanu” Oradea, 410154 Oradea, Romania
| |
Collapse
|
11
|
Mosaferi B, Jand Y, Salari AA. Gut microbiota depletion from early adolescence alters anxiety and depression-related behaviours in male mice with Alzheimer-like disease. Sci Rep 2021; 11:22941. [PMID: 34824332 PMCID: PMC8617202 DOI: 10.1038/s41598-021-02231-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 11/12/2021] [Indexed: 12/13/2022] Open
Abstract
The gut-microbiota-brain axis plays an important role in stress-related disorders, and dysfunction of this complex bidirectional system is associated with Alzheimer's disease. This study aimed to assess the idea that whether gut microbiota depletion from early adolescence can alter anxiety- and depression-related behaviours in adult mice with or without Alzheimer-like disease. Male C57BL/6 mice were treated with an antibiotic cocktail from weaning to adulthood. Adult mice received an intracerebroventricular injection of amyloid-beta (Aβ)1-42, and were subjected to anxiety and depression tests. We measured, brain malondialdehyde and glutathione following anxiety tests, and assessed brain oxytocin and the hypothalamic-pituitary-adrenal (HPA) axis function by measuring adrenocorticotrophic hormone (ACTH) and corticosterone following depression tests. Healthy antibiotic-treated mice displayed significant decreases in anxiety-like behaviours, whereas they did not show any alterations in depression-like behaviours and HPA axis function. Antibiotic treatment from early adolescence prevented the development of anxiety- and depression-related behaviours, oxidative stress and HPA axis dysregulation in Alzheimer-induced mice. Antibiotic treatment increased oxytocin in the brain of healthy but not Alzheimer-induced mice. Taken together, these findings suggest that gut microbiota depletion following antibiotic treatment from early adolescence might profoundly affect anxiety- and depression-related behaviours, and HPA axis function in adult mice with Alzheimer-like disease.
Collapse
Affiliation(s)
- Belal Mosaferi
- Department of Basic Sciences, School of Nursing and Midwifery, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Yahya Jand
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali-Akbar Salari
- Salari Institute of Cognitive and Behavioral Disorders (SICBD), P.O. Box 31396-45999, Karaj, Alborz, Iran.
| |
Collapse
|
12
|
Chaves Filho AJM, Mottin M, Soares MVR, Jucá PM, Andrade CH, Macedo DS. Tetracyclines, a promise for neuropsychiatric disorders: from adjunctive therapy to the discovery of new targets for rational drug design in psychiatry. Behav Pharmacol 2021; 32:123-141. [PMID: 33595954 DOI: 10.1097/fbp.0000000000000585] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Major mental disorders, such as schizophrenia, bipolar disorder, and major depressive disorder, represent the leading cause of disability worldwide. Nevertheless, the current pharmacotherapy has several limitations, and a large portion of patients do not respond appropriately to it or remain with disabling symptoms overtime. Traditionally, pharmacological interventions for psychiatric disorders modulate dysfunctional neurotransmitter systems. In the last decades, compelling evidence has advocated for chronic inflammatory mechanisms underlying these disorders. Therefore, the repurposing of anti-inflammatory agents has emerged as an attractive therapeutic tool for mental disorders. Minocycline (MINO) and doxycycline (DOXY) are semisynthetic second-generation tetracyclines with neuroprotective and anti-inflammatory properties. More recently, the most promising results obtained in clinical trials using tetracyclines for major psychiatric disorders were for schizophrenia. In a reverse translational approach, tetracyclines inhibit microglial reactivity and toxic inflammation by mechanisms related to the inhibition of nuclear factor kappa B signaling, cyclooxygenase 2, and matrix metalloproteinases. However, the molecular mechanism underlying the effects of these tetracyclines is not fully understood. Therefore, the present review sought to summarize the latest findings of MINO and DOXY use for major psychiatric disorders and present the possible targets to their molecular and behavioral effects. In conclusion, tetracyclines hold great promise as (ready-to-use) agents for being used as adjunctive therapy for human neuropsychiatric disorders. Hence, the understanding of their molecular mechanisms may contribute to the discovery of new targets for the rational drug design of novel psychoactive agents.
Collapse
Affiliation(s)
- Adriano José Maia Chaves Filho
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE
- Laboratory for Molecular Modeling and Drug Design, LabMol, Faculdade de Farmácia, Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia, GO
| | - Melina Mottin
- Laboratory for Molecular Modeling and Drug Design, LabMol, Faculdade de Farmácia, Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia, GO
| | - Michele Verde-Ramo Soares
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE
| | - Paloma Marinho Jucá
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE
| | - Carolina Horta Andrade
- Laboratory for Molecular Modeling and Drug Design, LabMol, Faculdade de Farmácia, Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia, GO
| | - Danielle S Macedo
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE
- National Institute for Translational Medicine (INCT-TM, CNPq), Ribeirão Preto, SP, Brazil
| |
Collapse
|
13
|
Motaghinejad M, Mashayekh R, Motevalian M, Safari S. The possible role of CREB-BDNF signaling pathway in neuroprotective effects of minocycline against alcohol-induced neurodegeneration: molecular and behavioral evidences. Fundam Clin Pharmacol 2021; 35:113-130. [PMID: 32579730 DOI: 10.1111/fcp.12584] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 05/02/2020] [Accepted: 06/18/2020] [Indexed: 12/17/2022]
Abstract
Abuse of alcohol triggers neurodegeneration in human brain. Minocycline has characteristics conferring neuroprotection. Current study evaluates the role of the CREB-BDNF signaling pathway in mediating minocycline's neuroprotective effects against alcohol-induced neurodegeneration. Seventy adult male rats were randomly split into groups 1 and 2 that received saline and alcohol (2 g/kg/day by gavage, once daily), respectively, and groups 3, 4, 5, and 6 were treated simultaneously with alcohol and minocycline (10, 20, 30 and 40 mg/kg I.P, respectively) for 21 days. Group 7 received minocycline alone (40 mg/kg, i.p) for 21 days. Morris water maze (MWM) has been used to assess cognitive activity. Hippocampal neurodegenerative and histological parameters as well as cyclic AMP response element-binding protein (CREB) and brain-derived neurotrophic factor (BDNF) levels were assessed. Alcohol impaired cognition, and concurrent therapy with various minocycline doses attenuated alcohol-induced cognition disturbances. Additionally, alcohol administration boosted lipid peroxidation and levels of glutathione in oxidized form (GSSG), tumor necrosis factor alpha (TNF-α), interleukin 1 beta (IL-1β), and Bax protein, while decreased reducing type of glutathione (GSH), Bcl-2 protein, phosphorylated CREB, and BDNF levels in rat hippocampus. Alcohol also decreased the activity in the hippocampus of superoxide dismutase (SOD), glutathione peroxidase (GPx), and glutathione reductase (GR). In comparison, minocycline attenuated alcohol-induced neurodegeneration; elevating expression levels of P-CREB and BDNF and inhibited alcohol induced histopathological changes in both dentate gyrus (DG) and CA1 of hippocampus. Thus, minocycline is likely to provide neuroprotection against alcohol-induced neurodegeneration through mediation of the P-CREB/BDNF signaling pathway.
Collapse
Affiliation(s)
- Majid Motaghinejad
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Roya Mashayekh
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Chemistry, Pharmaceutical Sciences Branch, Islamic Azad University (IUAPS), Tehran, Iran
| | - Manijeh Motevalian
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sepideh Safari
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Filling the gaps on stroke research: Focus on inflammation and immunity. Brain Behav Immun 2021; 91:649-667. [PMID: 33017613 PMCID: PMC7531595 DOI: 10.1016/j.bbi.2020.09.025] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/10/2020] [Accepted: 09/23/2020] [Indexed: 02/08/2023] Open
Abstract
For the last two decades, researchers have placed hopes in a new era in which a combination of reperfusion and neuroprotection would revolutionize the treatment of stroke. Nevertheless, despite the thousands of papers available in the literature showing positive results in preclinical stroke models, randomized clinical trials have failed to show efficacy. It seems clear now that the existing data obtained in preclinical research have depicted an incomplete picture of stroke pathophysiology. In order to ameliorate bench-to-bed translation, in this review we first describe the main actors on stroke inflammatory and immune responses based on the available preclinical data, highlighting the fact that the link between leukocyte infiltration, lesion volume and neurological outcome remains unclear. We then describe what is known on neuroinflammation and immune responses in stroke patients, and summarize the results of the clinical trials on immunomodulatory drugs. In order to understand the gap between clinical trials and preclinical results on stroke, we discuss in detail the experimental results that served as the basis for the summarized clinical trials on immunomodulatory drugs, focusing on (i) experimental stroke models, (ii) the timing and selection of outcome measuring, (iii) alternative entry routes for leukocytes into the ischemic region, and (iv) factors affecting stroke outcome such as gender differences, ageing, comorbidities like hypertension and diabetes, obesity, tobacco, alcohol consumption and previous infections like Covid-19. We can do better for stroke treatment, especially when targeting inflammation following stroke. We need to re-think the design of stroke experimental setups, notably by (i) using clinically relevant models of stroke, (ii) including both radiological and neurological outcomes, (iii) performing long-term follow-up studies, (iv) conducting large-scale preclinical stroke trials, and (v) including stroke comorbidities in preclinical research.
Collapse
|
15
|
González-Lizárraga F, Ploper D, Ávila CL, Socías SB, Dos-Santos-Pereira M, Machín B, Del-Bel E, Michel PP, Pietrasanta LI, Raisman-Vozari R, Chehín R. CMT-3 targets different α-synuclein aggregates mitigating their toxic and inflammogenic effects. Sci Rep 2020; 10:20258. [PMID: 33219264 PMCID: PMC7679368 DOI: 10.1038/s41598-020-76927-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 11/03/2020] [Indexed: 12/22/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder for which only symptomatic treatments are available. Repurposing drugs that target α-synuclein aggregation, considered one of the main drivers of PD progression, could accelerate the development of disease-modifying therapies. In this work, we focused on chemically modified tetracycline 3 (CMT-3), a derivative with reduced antibiotic activity that crosses the blood–brain barrier and is pharmacologically safe. We found that CMT-3 inhibited α-synuclein amyloid aggregation and led to the formation of non-toxic molecular species, unlike minocycline. Furthermore, CMT-3 disassembled preformed α-synuclein amyloid fibrils into smaller fragments that were unable to seed in subsequent aggregation reactions. Most interestingly, disaggregated species were non-toxic and less inflammogenic on brain microglial cells. Finally, we modelled the interactions between CMT-3 and α-synuclein aggregates by molecular simulations. In this way, we propose a mechanism for fibril disassembly. Our results place CMT-3 as a potential disease modifier for PD and possibly other synucleinopathies.
Collapse
Affiliation(s)
- Florencia González-Lizárraga
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), Pasaje Dorrego 1080, 4000, San Miguel de Tucumán, Argentina
| | - Diego Ploper
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), Pasaje Dorrego 1080, 4000, San Miguel de Tucumán, Argentina
| | - César L Ávila
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), Pasaje Dorrego 1080, 4000, San Miguel de Tucumán, Argentina
| | - Sergio B Socías
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), Pasaje Dorrego 1080, 4000, San Miguel de Tucumán, Argentina
| | | | - Belén Machín
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), Pasaje Dorrego 1080, 4000, San Miguel de Tucumán, Argentina
| | - Elaine Del-Bel
- Faculdade de Odontologia de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Patrick Pierre Michel
- Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne Université UM75, Paris, France
| | - Lía I Pietrasanta
- Departamento de Física-Instituto de Física de Buenos Aires (IFIBA, UBA-CONICET) and Centro de Microscopías Avanzadas (CMA), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1428EHA, Buenos Aires, Argentina
| | - Rita Raisman-Vozari
- Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne Université UM75, Paris, France.
| | - Rosana Chehín
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), Pasaje Dorrego 1080, 4000, San Miguel de Tucumán, Argentina.
| |
Collapse
|
16
|
Motaghinejad M, Farokhi N, Motevalian M, Safari S. Molecular, histological and behavioral evidences for neuroprotective effects of minocycline against nicotine-induced neurodegeneration and cognition impairment: Possible role of CREB-BDNF signaling pathway. Behav Brain Res 2020; 386:112597. [PMID: 32198107 DOI: 10.1016/j.bbr.2020.112597] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 03/02/2020] [Indexed: 12/25/2022]
Abstract
AIM Neurodegeneration is one of the serious adverse effects of stimulant agents such as nicotine. Minocycline possess established neuroprotective properties. The role of CREB-BDNF signaling pathway in mediating the neuroprotective effects of minocycline against nicotine-induced neurodegeneration in rats was evaluated in current study. METHODS Seventy adult male rats were divided randomly into seven groups. Group 1 and 2, received 0.7 ml/rat of normal saline (i.p) and nicotine (10 mg/kg, s.c) respectively. Groups 3, 4, 5 and 6, treated concurrently with nicotine (10 mg/kg) and minocycline (10, 20, 30 and 40 mg/kg, i.p, respectively) for 21 days. Group 7 received minocycline alone (40 mg/kg, i.p) for 21 days. From 17th to 21 st days of experiment, Morris water maze (MWM) was used to evaluate learning and spatial memory in rats treated in different groups. According to the critical role of hippocampus in cognitive behavior, hippocampal neurodegenerative parameters (oxidative stress and inflammatory biomarkers) and also cyclic AMP response element binding protein (CREB) and brain-derived neurotrophic factor (BDNF) levels were evaluated in isolated hippocampus in day 22 of experiment and after drug treatment. Also hippocampal cell density and tissue changes were evaluated by hematoxylin and eosin staining. RESULT Nicotine administration impaired the learning and spatial memory in rats and simultaneous treatment with various doses of minocycline attenuated the nicotine-induced cognition disturbances. In addition, nicotine treatment increased lipid peroxidation and the levels of oxidized form of glutathione (GSSG), interleukin 1 beta (IL-1β), tumor necrosis factor alpha (TNF-α), and Bax protein, while decreasing reduced form of glutathione (GSH), Bcl-2 protein, P-CREB and BDNF levels in the hippocampus of experimental animals. Nicotine also reduced the activity of superoxide dismutase (SOD), glutathione peroxidase (GPx) and glutathione reductase (GR) in the hippocampus. Minocycline attenuated nicotine-induced neurodegeneration and elevating CREB (both forms) and BDNF levels. Also minocycline treatment alone increases the cognitive activity and increased CREB (both forms) and BDNF levels and decreased oxidative stress, inflammation and apoptotic biomarkers. Minocycline at high doses cause inhibition of nicotine induced cell density and changes in both area of dentate gyrus (DG) and CA1 in hippocampus. CONCLUSION It can be concluded that minocycline, probably through activation of P-CREB/BDNF signaling pathway, confers neuroprotection against nicotine-induced neurodegeneration in rat hippocampus.
Collapse
Affiliation(s)
- Majid Motaghinejad
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Negin Farokhi
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Chemistry, Pharmaceutical Sciences Branch, Islamic Azad University (IUAPS), Tehran, Iran
| | - Manijeh Motevalian
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sepideh Safari
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Carabelli B, Delattre AM, Waltrick APF, Araújo G, Suchecki D, Machado RB, de Souza LER, Zanata SM, Zanoveli JM, Ferraz AC. Fish-oil supplementation decreases Indoleamine-2,3-Dioxygenase expression and increases hippocampal serotonin levels in the LPS depression model. Behav Brain Res 2020; 390:112675. [PMID: 32407816 DOI: 10.1016/j.bbr.2020.112675] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 04/07/2020] [Accepted: 04/24/2020] [Indexed: 10/24/2022]
Abstract
AIM To test the hypothesis that the antidepressant-like effect of omega-3 polyunsaturated fatty acids is related to the Indoleamine-2,3-Dioxygenase (IDO) inhibition. METHODS Animals were supplemented for 50 days with 3.0 g/kg of Fish Oil (FO) or received water (Control group - C), via gavage. At the end of this period, both groups were injected with LPS 24 h before the modified forced swim test (MFST) and the open field. To assess the possible involvement of IDO in the FO effects, we performed two independent experiments, using two IDO inhibitors: the direct inhibitor 1-methyl-DL-tryptophan (1-MT) and the anti-inflammatory drug minocycline (MINO), administered 23 h, 5 h and 1 h before the tests. After the tests, the animals' hippocampi were removed for quantification of serotonin (5-HT) and its metabolite 5-hydroxyindoleacetic acid (5-HIAA) by HPLC, and for IDO expression by western blot. RESULTS LPS induced a depressive-like state in the animals, and this effect was blocked by 1-MT, MINO and FO. Regardless of IDO inhibition, FO supplemented animals displayed an antidepressant-like response by increasing swimming and decreasing immobility frequencies in the MFST when compared to the control group. The immune challenge induced an over-expression of IDO and reduced hippocampal 5-HT levels, both of which were reversed by MINO and FO. CONCLUSION FO induced a pronounced antidepressant-like effect and prevented LPS-induced depressive-like behavior, and this effect was related to decreased IDO expression and increased 5-HT levels in the hippocampus.
Collapse
Affiliation(s)
- Bruno Carabelli
- Departamento de Fisiologia, Universidade Federal do Paraná, Curitiba, PR, Brazil.
| | - Ana Márcia Delattre
- Departamento de Fisiologia, Universidade Federal do Paraná, Curitiba, PR, Brazil
| | | | - Giulia Araújo
- Departamento de Fisiologia, Universidade Federal do Paraná, Curitiba, PR, Brazil
| | - Deborah Suchecki
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | | | | | - Silvio M Zanata
- Departamento de Patologia, Universidade Federal do Paraná, Curitiba, PR, Brazil
| | | | - Anete Curte Ferraz
- Departamento de Fisiologia, Universidade Federal do Paraná, Curitiba, PR, Brazil
| |
Collapse
|
18
|
Yuan T, Manohar K, Latorre R, Orock A, Greenwood-Van Meerveld B. Inhibition of Microglial Activation in the Amygdala Reverses Stress-Induced Abdominal Pain in the Male Rat. Cell Mol Gastroenterol Hepatol 2020; 10:527-543. [PMID: 32408032 PMCID: PMC7394753 DOI: 10.1016/j.jcmgh.2020.04.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 04/29/2020] [Accepted: 04/30/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Psychological stress is a trigger for the development of irritable bowel syndrome and associated symptoms including abdominal pain. Although irritable bowel syndrome patients show increased activation in the limbic brain, including the amygdala, the underlying molecular and cellular mechanisms regulating visceral nociception in the central nervous system are incompletely understood. In a rodent model of chronic stress, we explored the role of microglia in the central nucleus of the amygdala (CeA) in controlling visceral sensitivity. Microglia are activated by environmental challenges such as stress, and are able to modify neuronal activity via synaptic remodeling and inflammatory cytokine release. Inflammatory gene expression and microglial activity are regulated negatively by nuclear glucocorticoid receptors (GR), which are suppressed by the stress-activated pain mediator p38 mitogen-activated protein kinases (MAPK). METHODS Fisher-344 male rats were exposed to water avoidance stress (WAS) for 1 hour per day for 7 days. Microglia morphology and the expression of phospho-p38 MAPK and GR were analyzed via immunofluorescence. Microglia-mediated synaptic remodeling was investigated by quantifying the number of postsynaptic density protein 95-positive puncta. Cytokine expression levels in the CeA were assessed via quantitative polymerase chain reaction and a Luminex assay (Bio-Rad, Hercules, CA). Stereotaxic infusion into the CeA of minocycline to inhibit, or fractalkine to activate, microglia was followed by colonic sensitivity measurement via a visceromotor behavioral response to isobaric graded pressures of tonic colorectal distension. RESULTS WAS induced microglial deramification in the CeA. Moreover, WAS induced a 3-fold increase in the expression of phospho-p38 and decreased the ratio of nuclear GR in the microglia. The number of microglia-engulfed postsynaptic density protein 95-positive puncta in the CeA was increased 3-fold by WAS, while cytokine levels were unchanged. WAS-induced changes in microglial morphology, microglia-mediated synaptic engulfment in the CeA, and visceral hypersensitivity were reversed by minocycline whereas in stress-naïve rats, fractalkine induced microglial deramification and visceral hypersensitivity. CONCLUSIONS Our data show that chronic stress induces visceral hypersensitivity in male rats and is associated with microglial p38 MAPK activation, GR dysfunction, and neuronal remodeling in the CeA.
Collapse
Affiliation(s)
- Tian Yuan
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Krishna Manohar
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Rocco Latorre
- Department of Basic Science and Craniofacial Biology, New York University, New York City, New York
| | - Albert Orock
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Beverley Greenwood-Van Meerveld
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma,Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma,Oklahoma City VA Health Care System, Oklahoma City, Oklahoma,Correspondence Address correspondence to: Beverley Greenwood-Van Meerveld, PhD, O’Donoghue Building, Room 332, 1122 NE 13th Street, Oklahoma City, Oklahoma 73117.
| |
Collapse
|
19
|
Naderi Y, Panahi Y, Barreto GE, Sahebkar A. Neuroprotective effects of minocycline on focal cerebral ischemia injury: a systematic review. Neural Regen Res 2020; 15:773-782. [PMID: 31719236 PMCID: PMC6990777 DOI: 10.4103/1673-5374.268898] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
To review the neuroprotective effects of minocycline in focal cerebral ischemia in animal models. By searching in the databases of PubMed, ScienceDirect, and Scopus, and considering the inclusion and exclusion criteria of the study. Studies were included if focal cerebral ischemia model was performed in mammals and including a control group that has been compared with a minocycline group. Written in languages other than English; duplicate data; in vitro studies and combination of minocycline with other neuroprotective agents were excluded. Neurological function of patients was assessed by National Institute of Health Stroke Scale, modified Rankin Scale, and modified Barthel Index. Neuroprotective effects were assessed by detecting the expression of inflammatory cytokines. We examined 35 papers concerning the protective effects of minocycline in focal cerebral ischemia in animal models and 6 clinical trials which had evaluated the neuroprotective effects of minocycline in ischemic stroke. These studies revealed that minocycline increases the viability of neurons and decreases the infarct volume following cerebral ischemia. The mechanisms that were reported in these studies included anti-inflammatory, antioxidant, as well as anti-apoptotic effects. Minocycline also increases the neuronal regeneration following cerebral ischemia. Minocycline has considerable neuroprotective effects against cerebral ischemia-induced neuronal damages. However, larger clinical trials may be required before using minocycline as a neuroprotective drug in ischemic stroke.
Collapse
Affiliation(s)
- Yazdan Naderi
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Yunes Panahi
- Pharmacotherapy Department, Faculty of Pharmacy, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
| | - Amirhosein Sahebkar
- Halal Research Center of IRI, FDA, Tehran; Biotechnology Research Center, Pharmaceutical Technology Institute; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
20
|
Janata A, Magnet IAM, Schreiber KL, Wilson CD, Stezoski JP, Janesko-Feldman K, Kochanek PM, Drabek T. Minocycline fails to improve neurologic and histologic outcome after ventricular fibrillation cardiac arrest in rats. World J Crit Care Med 2019; 8:106-119. [PMID: 31853446 PMCID: PMC6918046 DOI: 10.5492/wjccm.v8.i7.106] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 09/17/2019] [Accepted: 10/27/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Prolonged cardiac arrest (CA) produces extensive neuronal death and microglial proliferation and activation resulting in neuro-cognitive disabilities. Among other potential mechanisms, microglia have been implicated as triggers of neuronal death after hypoxic-ischemic insults. Minocycline is neuroprotective in some brain ischemia models, either by blunting the microglial response or by a direct effect on neurons. AIM To improve survival, attenuate neurologic deficits, neuroinflammation, and histological damage after ventricular fibrillation (VF) CA in rats. METHODS Adult male isoflurane-anesthetized rats were subjected to 6 min VF CA followed by 2 min resuscitation including chest compression, epinephrine, bicarbonate, and defibrillation. After return of spontaneous circulation (ROSC), rats were randomized to two groups: (1) Minocycline 90 mg/kg intraperitoneally (i.p.) at 15 min ROSC, followed by 22.5 mg/kg i.p. every 12 h for 72 h; and (2) Controls, receiving the same volume of vehicle (phosphate-buffered saline). The rats were kept normothermic during the postoperative course. Neurologic injury was assessed daily using Overall Performance Category (OPC; 1 = normal, 5 = dead) and Neurologic Deficit Score (NDS; 0% = normal, 100% = dead). Rats were sacrificed at 72 h. Neuronal degeneration (Fluoro-Jade C staining) and microglia proliferation (anti-Iba-1 staining) were quantified in four selectively vulnerable brain regions (hippocampus, striatum, cerebellum, cortex) by three independent reviewers masked to the group assignment. RESULTS In the minocycline group, 8 out of 14 rats survived to 72 h compared to 8 out of 19 rats in the control group (P = 0.46). The degree of neurologic deficit at 72 h [median, (interquartile range)] was not different between survivors in minocycline vs controls: OPC 1.5 (1-2.75) vs 2 (1.25-3), P = 0.442; NDS 12 (2-20) vs 17 (7-51), P = 0.328) or between all studied rats. The number of degenerating neurons (minocycline vs controls, mean ± SEM: Hippocampus 58 ± 8 vs 76 ± 8; striatum 121 ± 43 vs 153 ± 32; cerebellum 20 ± 7 vs 22 ± 8; cortex 0 ± 0 vs 0 ± 0) or proliferating microglia (hippocampus 157 ± 15 vs 193 cortex 0 ± 0 vs 0 ± 0; 16; striatum 150 ± 22 vs 161 ± 23; cerebellum 20 ± 7 vs 22 ± 8; cortex 26 ± 6 vs 31 ± 7) was not different between groups in any region (all P > 0.05). Numerically, there were approximately 20% less degenerating neurons and proliferating microglia in the hippocampus and striatum in the minocycline group, with a consistent pattern of histological damage across the individual regions of interest. CONCLUSION Minocycline did not improve survival and failed to confer substantial benefits on neurologic function, neuronal loss or microglial proliferation across multiple brain regions in our model of rat VF CA.
Collapse
Affiliation(s)
- Andreas Janata
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, United States
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, United States
- Emergency Department, KA Rudolfstiftung, Vienna 1030, Austria
| | - Ingrid AM Magnet
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, United States
- Department of Emergency Medicine, Vienna General Hospital, Medical University of Vienna, Vienna 1090, Austria
| | - Kristin L Schreiber
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, United States
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, United States
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Caleb D Wilson
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, United States
- Wyoming Otolaryngology, Wyoming Medical Center, Casper, WY 82604, United States
| | - Jason P Stezoski
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, United States
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, United States
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, United States
| | - Keri Janesko-Feldman
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, United States
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, United States
| | - Patrick M Kochanek
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, United States
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, United States
| | - Tomas Drabek
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, United States
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, United States
| |
Collapse
|
21
|
Drieu A, Buendia I, Levard D, Hélie P, Brodin C, Vivien D, Rubio M. Immune Responses and Anti-inflammatory Strategies in a Clinically Relevant Model of Thromboembolic Ischemic Stroke with Reperfusion. Transl Stroke Res 2019; 11:481-495. [PMID: 31522409 DOI: 10.1007/s12975-019-00733-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 08/20/2019] [Accepted: 08/23/2019] [Indexed: 01/12/2023]
Abstract
The poor clinical relevance of experimental models of stroke contributes to the translational failure between preclinical and clinical studies testing anti-inflammatory molecules for ischemic stroke. Here, we (i) describe the time course of inflammatory responses triggered by a thromboembolic model of ischemic stroke and (ii) we examine the efficacy of two clinically tested anti-inflammatory drugs: Minocycline or anti-CD49d antibodies (tested in stroke patients as Natalizumab) administered early (1 h) or late (48 h) after stroke onset. Radiological (lesion volume) and neurological (grip test) outcomes were evaluated at 24 h and 5 days after stroke. Immune cell responses peaked 48 h after stroke onset. Myeloid cells (microglia/macrophages, dendritic cells, and neutrophils) were already increased 24 h after stroke onset, peaked at 48 h, and remained increased-although to a lesser extent-5 days after stroke onset. CD8+ and CD4+ T-lymphocytes infiltrated the ipsilateral hemisphere later on (only from 48 h). These responses occurred together with a progressive blood-brain barrier leakage at the lesion site, starting 24 h after stroke onset. Lesion volume was maximal 24-48 h after stroke onset. Minocycline reduced both lesion volume and neurological deficit only when administered early after stroke onset. The blockade of leukocyte infiltration by anti-CD49d had no impact on lesion volume or long-term neurological deficit, independently of the timing of treatment. Our data are in accordance with the results of previous clinical reports on the use of Minocycline and Natalizumab on ischemic stroke. We thus propose the use of this clinically relevant model of thromboembolic stroke with recanalization for future testing of anti-inflammatory strategies for stroke.
Collapse
Affiliation(s)
- Antoine Drieu
- Normandie Univ, UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders", Institut Blood and Brain @ Caen-Normandie, Cyceron, 14000 Caen, France, Normandie Université, 14000, Caen, France
| | - Izaskun Buendia
- Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria, Madrid, Spain
| | - Damien Levard
- Normandie Univ, UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders", Institut Blood and Brain @ Caen-Normandie, Cyceron, 14000 Caen, France, Normandie Université, 14000, Caen, France
| | - Pauline Hélie
- Normandie Univ, UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders", Institut Blood and Brain @ Caen-Normandie, Cyceron, 14000 Caen, France, Normandie Université, 14000, Caen, France
| | - Camille Brodin
- Normandie Univ, UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders", Institut Blood and Brain @ Caen-Normandie, Cyceron, 14000 Caen, France, Normandie Université, 14000, Caen, France
| | - Denis Vivien
- Normandie Univ, UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders", Institut Blood and Brain @ Caen-Normandie, Cyceron, 14000 Caen, France, Normandie Université, 14000, Caen, France
- Department of Clinical Research, CHU de Caen Normandy, 14000, Caen, France
| | - Marina Rubio
- Normandie Univ, UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders", Institut Blood and Brain @ Caen-Normandie, Cyceron, 14000 Caen, France, Normandie Université, 14000, Caen, France.
| |
Collapse
|
22
|
Oh J, Lee J, Piao C, Jeong JH, Lee M. A self-assembled DNA-nanoparticle with a targeting peptide for hypoxia-inducible gene therapy of ischemic stroke. Biomater Sci 2019; 7:2174-2190. [PMID: 30900719 DOI: 10.1039/c8bm01621f] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
A self-assembled nanoparticle composed of hypoxia-specific anti-RAGE peptide (HSAP), heme oxygenase-1 plasmid (pHO1), and deoxycholate-conjugated polyethylenimine-2k (DP2k) was developed for ischemic stroke therapy. RAGE is over-expressed and induces inflammation in the ischemic brain. To inhibit RAGE-mediated signal transduction, HSAP was produced by recombinant DNA technology, based on the RAGE-binding domain of high mobility group box-1. Because of the specific binding to RAGE, the nanoparticle with HSAP (HSAP-NP) may have dual roles as a cytoprotective reagent and a specific ligand to RAGE for receptor-mediated transfection. As a cytoprotective reagent, the HSAP-NP reduced RAGE expression on the surface of the brain cells by inhibiting the positive feedback of RAGE-mediated signal transduction. As a result, inflammation, apoptosis, and reactive oxygen species were decreased in hypoxic cells. As a gene carrier, HSAP-NP showed a higher transfection efficiency than polyethylenimine-25k, DP2k, and Lipofectamine. Particularly, HSAP-NP enhanced gene delivery to hypoxic cells. In the stroke animal models, HSAP-NP reduced the levels of RAGE, inducible nitric oxide synthase, and inflammation. Additionally, HSAP-NP with pHO1 (HSAP-NP/pHO1) increased HO1 expression in the ischemic brain. Gene expression was higher in hypoxia-inducible factor-1α (HIF-1α)-positive cells than in HIF-1α-negative cells, suggesting that HSAP-NP delivered the genes to ischemic tissues more efficiently. Cell death and infarct volume in the stroke models were significantly decreased by HSAP-NP/pHO1 compared with HSAP alone or the DP2k/pHO1 complex. Therefore, HSAP-NP may be a useful gene and peptide therapy system for stroke therapy with dual functions of hypoxia-specific gene delivery and cytoprotective effects.
Collapse
Affiliation(s)
- Jungju Oh
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, Korea.
| | | | | | | | | |
Collapse
|
23
|
Sato T, Hirohara A, Izumi Y, Nakatomi T, Iwayama K, Ohtaki KI, Hayase N. Effects of Minocycline on H2O2-induced Cell Death and Interleukin-8 Production in Human Small Airway Epithelial Cells. INT J PHARMACOL 2019. [DOI: 10.3923/ijp.2019.509.514] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
24
|
Salehi P, Shahmirzadi ZY, Mirrezaei FS, Shirvani Boushehri F, Mayahi F, Songhori M, Abofazeli M, Motaghinejad M, Safari S. A hypothetic role of minocycline as a neuroprotective agent against methylphenidate-induced neuronal mitochondrial dysfunction and tau protein hyper-phosphorylation: Possible role of PI3/Akt/GSK3β signaling pathway. Med Hypotheses 2019; 128:6-10. [PMID: 31203911 DOI: 10.1016/j.mehy.2019.04.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 04/14/2019] [Accepted: 04/23/2019] [Indexed: 02/08/2023]
Abstract
The underlining mechanism in neural mitochondrial dysfunction and consequences neurotoxicity, and cognitive behavior after methylphenidate (MPH) prolonged uses is unclear and proposing of therapeutic approaches for treatment of these types of neurotoxicity is one of the main goals of scientist in this manner. MPH-induced mitochondrial dysfunction in neural cells caused induction of oxidative stress, apoptosis, inflammation and cognition impairment, which leads to neurotoxicity, was reported previously but role of key neural cells proteins and involved signaling pathway in this manner remained indeterminate. Tau protein aggregation is a biomarker for mitochondrial dysfunction, neurodegenerative event and cognition impairment. Tau aggregation occur by stimulation effects of Glycogen synthase kinase-3(GSK3β) and phosphatidylinositol 3-kinase (PI3K) which activates protein kinase B(Akt) and causes inhibition of phosphorylation(activation) of GSK3β, thus Akt activation can cause inhibition of tau aggregation (hyper-phosphorylation). Management of mentioned MPH-induced mitochondrial dysfunction and consequences of neurotoxicity, and cognitive behavior through a new generation neuroprotective combination, based on modulation of disturbed in Akt function and inhibition of GSK3β and tau hyper-phosphorylation can be a prefect therapeutic interventions. Therefore, finding, introduction and development of new neuroprotective properties and explanation of their effects with potential capacity for modulation of tau hyper-phosphorylation via PI3/Akt/GSK signaling pathway is necessitated. During recent years, using new neuroprotective compounds with therapeutic probability for treatment of psychostimulant-induced mitochondrial dysfunction, neurotoxicity and cognitive malicious effects have been amazingly increased. Many previous studies have reported the neuroprotective roles of minocycline (a broad-spectrum and long-acting antibiotic) in multiple neurodegenerative events and diseases in animal model. But the role of neuroprotective effects of this agent against MPH induced mitochondrial dysfunction, neurotoxicity and cognitive malicious and also role of tau hyper-phosphorylation by modulation of PI3/Akt/GSK signaling pathway in this manner remain unknown. Thus we suggested and theorized that by using minocycline in MPH addicted subject, it would provide neuroprotection against MPH-induced mitochondrial dysfunction, neurotoxicity and cognitive malicious. Also we hypothesized that minocycline, via modulation of PI3/Akt/GSK and inhibition of tau hyper-phosphorylation, can inhibit MPH-induced mitochondrial dysfunction, neurotoxicity and cognitive malicious. In this article, we tried to discuss our hypothesis regarding the possible role of minocycline, as a powerful neuroprotective agent, and also role of tau hyper-phosphorylation related to PI3/Akt/GSK signaling pathway in treatment of MPH-induced mitochondrial dysfunction, neurotoxicity and cognitive disturbance.
Collapse
Affiliation(s)
- Pegah Salehi
- Research Center for Addiction and Risky Behaviors (ReCARB), Iran Psychiatric Center, Iran University of Medical Sciences, Tehran, Iran
| | - Zhara Yaraei Shahmirzadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Chemistry, Pharmaceutical Sciences Branch, Islamic Azad University (IUAPS), Tehran, Iran
| | - Fatemeh Sadat Mirrezaei
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Chemistry, Pharmaceutical Sciences Branch, Islamic Azad University (IUAPS), Tehran, Iran
| | - Farima Shirvani Boushehri
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Chemistry, Pharmaceutical Sciences Branch, Islamic Azad University (IUAPS), Tehran, Iran
| | - Fatemeh Mayahi
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Chemistry, Pharmaceutical Sciences Branch, Islamic Azad University (IUAPS), Tehran, Iran
| | - Mojtaba Songhori
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Chemistry, Pharmaceutical Sciences Branch, Islamic Azad University (IUAPS), Tehran, Iran
| | - Maryam Abofazeli
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Chemistry, Pharmaceutical Sciences Branch, Islamic Azad University (IUAPS), Tehran, Iran
| | - Majid Motaghinejad
- Research Center for Addiction and Risky Behaviors (ReCARB), Iran Psychiatric Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Sepideh Safari
- Razi Drug Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
25
|
Vadokas G, Koehler S, Weiland J, Lilla N, Stetter C, Westermaier T. Early Antiinflammatory Therapy Attenuates Brain Damage After Sah in Rats. Transl Neurosci 2019; 10:104-111. [PMID: 31098320 PMCID: PMC6487785 DOI: 10.1515/tnsci-2019-0018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 03/18/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Early inflammatory processes may play an important role in the development of early brain injury (EBI) after subarachnoid hemorrhage (SAH). Experimental studies suggest that anti-inflammatory and membrane-stabilizing drugs might have beneficial effects, although the underlying mechanisms are not fully understood. The aim of this study was to investigate the effect of early treatment with methylprednisolone and minocycline on cerebral perfusion and EBI after experimental SAH. METHODS Male Sprague-Dawley rats were subjected to SAH using the endovascular filament model. 30 minutes after SAH, they were randomly assigned to receive an intravenous injection of methylprednisolone (16mg/kg body weight, n=10), minocycline (45mg/kg body weight, n=10) or saline (n=11). Mean arterial blood pressure (MABP), intracranial pressure (ICP) and local cerebral blood flow (LCBF) over both hemispheres were recorded continuously for three hours following SAH. Neurological assessment was performed after 24 hours. Hippocampal damage was analyzed by immunohistochemical staining (caspase 3). RESULTS Treatment with methylprednisolone or minocycline did not result in a significant improvement of MABP, ICP or LCBF. Animals of both treatment groups showed a non-significant trend to better neurological recovery compared to animals of the control group. Mortality was reduced and hippocampal damage significantly attenuated in both methylprednisolone and minocycline treated animals. CONCLUSION The results of this study suggest that inflammatory processes may play an important role in the pathophysiology of EBI after SAH. Early treatment with the anti-inflammatory drugs methylprednisolone or minocycline in the acute phase of SAH has the potential to reduce brain damage and exert a neuroprotective effect.
Collapse
Affiliation(s)
- Georg Vadokas
- Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider-Straße 11, 97080Würzburg, Germany
- Department of Urology, Canisius Wilhelmina Hospital Nijmegen, Weg door Jonkerbos 100, 6532 SZ Nijmegen, Netherlands
| | - Stefan Koehler
- Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider-Straße 11, 97080Würzburg, Germany
| | - Judith Weiland
- Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider-Straße 11, 97080Würzburg, Germany
| | - Nadine Lilla
- Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider-Straße 11, 97080Würzburg, Germany
| | - Christian Stetter
- Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider-Straße 11, 97080Würzburg, Germany
| | - Thomas Westermaier
- Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider-Straße 11, 97080Würzburg, Germany
| |
Collapse
|
26
|
Amini-Khoei H, Kordjazy N, Haj-Mirzaian A, Amiri S, Haj-Mirzaian A, Shirzadian A, Hasanvand A, Balali-Dehkordi S, Hassanipour M, Dehpour AR. Anticonvulsant effect of minocycline on pentylenetetrazole-induced seizure in mice: involvement of nitric oxide and N-methyl-d-aspartate receptor. Can J Physiol Pharmacol 2018; 96:742-750. [DOI: 10.1139/cjpp-2017-0673] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Anticonvulsant effects of minocycline have been explored recently. This study was designed to examine the anticonvulsant effect of acute administration of minocycline on pentylenetetrazole-induced seizures in mouse considering the possible role of the nitric oxide/N-methyl-d-aspartate (NMDA) pathway. We induced seizure using intravenous administration of pentylenetetrazole. Our results showed that acute administration of minocycline increased the seizure threshold. Furthermore, co-administration of subeffective doses of the nonselective nitric oxide synthase (NOS) inhibitor NG-l-arginine methyl ester (10 mg/kg) and the neuronal NOS inhibitor 7-nitroindazole (40 mg/kg) enhanced the anticonvulsant effect of subeffective doses of minocycline (40 mg/kg). We found that inducible NOS inhibitor aminoguanidine (100 mg/kg) had no effect on the antiseizure effect of minocycline. Moreover, l-arginine (60 mg/kg), as a NOS substrate, reduced the anticonvulsant effect of minocycline. We also demonstrated that pretreatment with the NMDA receptor antagonists ketamine (0.5 mg/kg) and MK-801 (0.05 mg/kg) increased the anticonvulsant effect of subeffective doses of minocycline. Results showed that minocycline significantly decreased the hippocampal nitrite level. Furthermore, co-administration of a neuronal NOS inhibitor like NMDA receptor antagonists augmented the effect of minocycline on the hippocampal nitrite level. In conclusion, we revealed that anticonvulsant effect of minocycline might be, at least in part, due to a decline in constitutive hippocampal nitric oxide activity as well as inhibition of NMDA receptors.
Collapse
Affiliation(s)
- Hossein Amini-Khoei
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Department of Physiology and Pharmacology, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Nastaran Kordjazy
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Arvin Haj-Mirzaian
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shayan Amiri
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Regenerative Medicine Program, Department of Biochemistry and Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Arya Haj-Mirzaian
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Armin Shirzadian
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Amin Hasanvand
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Shima Balali-Dehkordi
- Department of Basic Sciences, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Mahsa Hassanipour
- Physiology-Pharmacology Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Physiology and Pharmacology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Ahmad Reza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
27
|
Drieu A, Levard D, Vivien D, Rubio M. Anti-inflammatory treatments for stroke: from bench to bedside. Ther Adv Neurol Disord 2018; 11:1756286418789854. [PMID: 30083232 PMCID: PMC6066814 DOI: 10.1177/1756286418789854] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 06/19/2018] [Indexed: 12/11/2022] Open
Abstract
So far, intravenous tissue-type plasminogen activator (tPA) and mechanical
removal of arterial blood clot (thrombectomy) are the only available treatments
for acute ischemic stroke. However, the short therapeutic window and the lack of
specialized stroke unit care make the overall availability of both treatments
limited. Additional agents to combine with tPA administration or thrombectomy to
enhance efficacy and improve outcomes associated with stroke are needed.
Stroke-induced inflammatory processes are a response to the tissue damage due to
the absence of blood supply but have been proposed also as key contributors to
all the stages of the ischemic stroke pathophysiology. Despite promising results
in experimental studies, inflammation-modulating treatments have not yet been
translated successfully into the clinical setting. This review will (a) describe
the timing of the stroke immune pathophysiology; (b) detail the immune responses
to stroke sift-through cell type; and (c) discuss the pitfalls on the
translation from experimental studies to clinical trials testing the therapeutic
pertinence of immune modulators.
Collapse
Affiliation(s)
- Antoine Drieu
- Pathophysiology and Imaging of Neurological Disorders, Normandy University, Caen, France
| | - Damien Levard
- Pathophysiology and Imaging of Neurological Disorders, Normandy University, Caen, France
| | - Denis Vivien
- Pathophysiology and Imaging of Neurological Disorders, Normandy University, Caen, France Pathophysiology and Imaging of Neurological Disorders, Centre Hospitalier Universitaire de Caen, Caen, France
| | - Marina Rubio
- Pathophysiology and Imaging of Neurological Disorders, Normandy University, Boulevard Henri Becquerel BP 5229, Caen Cedex, 14000, France
| |
Collapse
|
28
|
|
29
|
Abstract
Accumulating research substantiates the statement that inflammation plays an important role in the development of stroke. Both proinflammatory and anti-inflammatory mediators are involved in the pathogenesis of stroke, an imbalance of which leads to inflammation. Anti-inflammation is a kind of hopeful strategy for the prevention and treatment of stroke. Substantial studies have demonstrated that minocycline, a second-generation semisynthetic antibiotic belonging to the tetracycline family, can inhibit neuroinflammation, inflammatory mediators and microglia activation, and improve neurological outcome. Experimental and clinical data have found the preclinical and clinical potential of minocycline in the treatment of stroke due to its anti-inflammation properties and anti-inflammation-induced pathogeneses, including antioxidative stress, antiapoptosis, inhibiting leukocyte migration and microglial activation, and decreasing matrix metalloproteinases activity. Hence, it suggests a great future for minocycline in the therapeutics of stroke that diminish the inflammatory progress of stroke.
Collapse
|
30
|
Thymelaea lythroides extract attenuates microglial activation and depressive-like behavior in LPS-induced inflammation in adult male rats. Biomed Pharmacother 2018; 99:655-663. [PMID: 29710462 DOI: 10.1016/j.biopha.2018.01.125] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 01/13/2018] [Accepted: 01/24/2018] [Indexed: 12/29/2022] Open
Abstract
Thymelaea lythroides extract is widely used as a traditional folk medicine in Morocco, especially for the treatment of diabetes, rheumatism and Inflammatory disease. The aim of the study is to evaluate the possible effect of methanolic extract of Thymelaea lythroides in repressing the inflammatory responses and long-lasting depression-like behavior associated with neuroinflammation in adult rats after neonatal LPS exposure. Male rat pups were treated systemically with either LPS (250??g/kg) or vehicle (phosphate buffer saline) on postnatal day 14. Six hours later, the LPS groups were assigned to intraperitoneal (ip) injection of Minocycline (50?mg/kg) or Thymelaea lythroides (200?mg/kg). Thereafter, in adulthood (postnatal days 90-97), the spontaneous locomotor activity and depression-like behavior were assessed successively in open field and forced swim tests. The levels of proinflammatory cytokines, oxidative damage, and activation of microglia were determined in the hippocampus (HP) of male rats on (PND90-97). Our results showed that open field hypoactivity and increased immobility period in LPS-induced adult rats were normalized on treatment with Thymelaea lythroides and minocycline. Both treatments attenuate the overactivated microglial cells in the CA1 and CA3 of hippocampus (HP) and significantly reduced the oxidative-nitrosative stress markers and cytokine (TNF ?) production in the HP. Thymelaea lythroides seems to have similar neuroprotective effects to Minocycline, and such protection may be due to: reduction of oxidative stress, upregulation of inflammatory mediators production, antidepressant behavior which all are associated with neuroinflammation.
Collapse
|
31
|
Neuroprotective effects of pretreatment with minocycline on memory impairment following cerebral ischemia in rats. Behav Pharmacol 2018; 28:214-222. [PMID: 28257293 DOI: 10.1097/fbp.0000000000000297] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Cerebral ischemia leads to memory impairment that is associated with loss of hippocampal CA1 pyramidal neurons. Neuroinflammation and oxidative stress may be implicated in the pathogenesis of ischemia/reperfusion damage. Minocycline has anti-inflammatory and antioxidant properties. We investigated the neuroprotective effects of minocycline in rats subjected to cerebral ischemia/reperfusion injury. Thirty male rats were divided into three groups: control, sham, and minocycline-pretreated group. Minocycline (40 mg/kg) was injected intraperitoneally immediately before surgery, and then ischemia was induced by occlusion of common carotid arteries for 20 min. Seven days after reperfusion, the Morris water-maze task was used to evaluate memory. Nissl staining was also performed to analyze pyramidal cell damage. We measured the contents of malondialdehyde and proinflammatory cytokines in the hippocampus by the thiobarbituric acid method and enzyme-linked immunosorbent assay, respectively. Microglial activation was also investigated by Iba1 immunostaining. The results showed that pretreatment with minocycline prevented memory impairment induced by cerebral ischemia/reperfusion. Minocycline pretreatment also significantly attenuated ischemia-induced pyramidal cell death and microglial activation in the CA1 region and reduced the levels of malondialdehyde and proinflammatory cytokines (interleukin-1β and tumor necrosis factor-α) in the hippocampus of ischemic rats. Minocycline showed neuroprotective effects on cerebral ischemia-induced memory deficit probably through its anti-inflammatory and antioxidant activities.
Collapse
|
32
|
de Queiroz AIG, Chaves Filho AJM, Araújo TDS, Lima CNC, Machado MDJS, Carvalho AF, Vasconcelos SMM, de Lucena DF, Quevedo J, Macedo D. Antimanic activity of minocycline in a GBR12909-induced model of mania in mice: Possible role of antioxidant and neurotrophic mechanisms. J Affect Disord 2018; 225:40-51. [PMID: 28783519 DOI: 10.1016/j.jad.2017.07.053] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 07/27/2017] [Accepted: 07/31/2017] [Indexed: 10/19/2022]
Abstract
BACKGROUND Mania/hypomania is the cardinal feature of bipolar disorder. Recently, single administration of the dopamine transporter (DAT) inhibitor, GBR12909, was related to mania-like alterations. In the present study we aimed at testing behavioral and brain oxidant/neurotrophic alterations induced by the repeated administration of GBR12909 and its prevention/reversal by the mood stabilizing drugs, lithium (Li) and valproate (VAL) as well as by the neuroprotective drug, minocycline (Mino). METHODS Adult Swiss mice were submitted to 14 days protocols namely prevention and reversal. In the reversal protocol mice were given GBR12909 or saline and between days 8 and 14 received Li, VAL, Mino (25 or 50mg/kg) or saline. In the prevention treatment, mice were pretreated with Li, VAL, Mino or saline prior to GBR12909. RESULTS GBR12909 repeated administration induced hyperlocomotion and increased risk taking behavior that were prevented and reversed by the mood stabilizers and both doses of Mino. Li, VAL or Mino were more effective in the reversal of striatal GSH alterations induced by GBR12909. Regarding lipid peroxidation Mino was more effective in the prevention and reversal of lipid peroxidation in the hippocampus whereas Li and VAL prevented this alteration in the striatum and PFC. Li, VAL and Mino25 reversed the decrease in BDNF levels induced by GBR12909. CONCLUSION GBR12909 repeated administration resembles manic phenotype. Similarly to classical mood-stabilizing agents, Mino prevented and reversed GBR12909 manic-like behavior in mice. Thus, our data provide preclinical support to the design of trials investigating Mino's possible antimanic effects.
Collapse
Affiliation(s)
- Ana Isabelle G de Queiroz
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Adriano José Maia Chaves Filho
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Tatiane da Silva Araújo
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Camila Nayane Carvalho Lima
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Michel de Jesus Souza Machado
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - André F Carvalho
- Translational Psychiatry Research Group and the Department of Clinical Medicine, Faculty of Medicine, Fortaleza, CE, Brazil
| | - Silvania Maria Mendes Vasconcelos
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - David Freitas de Lucena
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - João Quevedo
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Neuroscience Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA; Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Danielle Macedo
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil.
| |
Collapse
|
33
|
Haghi-Aminjan H, Asghari MH, Goharbari MH, Abdollahi M. A systematic review on potential mechanisms of minocycline in kidney diseases. Pharmacol Rep 2017; 69:602-609. [DOI: 10.1016/j.pharep.2017.02.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 01/06/2017] [Accepted: 02/02/2017] [Indexed: 12/16/2022]
|
34
|
Wang W, Sidoli S, Zhang W, Wang Q, Wang L, Jensen ON, Guo L, Zhao X, Zheng L. Abnormal levels of histone methylation in the retinas of diabetic rats are reversed by minocycline treatment. Sci Rep 2017; 7:45103. [PMID: 28338045 PMCID: PMC5364468 DOI: 10.1038/srep45103] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 02/17/2017] [Indexed: 02/07/2023] Open
Abstract
In this study we quantified the alterations of retinal histone post-translational modifications (PTMs) in diabetic rats using a liquid chromatography - tandem mass spectrometry (LC-MS/MS) approach. Some diabetic rats were subsequently treated with minocycline, a tetracycline antibiotic, which has been shown to inhibit the diabetes-induced chronic inflammation in the retinas of rodents. We quantified 266 differentially modified histone peptides, including 48 out of 83 methylation marks with significantly different abundancein retinas of diabetic rats as compared to non-diabetic controls. About 67% of these marks had their relative abundance restored to non-diabetic levels after minocycline treatment. Mono- and di-methylation states of histone H4 lysine 20 (H4K20me1/me2), markers related to DNA damage response, were found to be up-regulated in the retinas of diabetic rats and restored to control levels upon minocycline treatment. DNA damage response biomarkers showed the same pattern once quantified by western blotting. Collectively, this study indicates that alteration of some histone methylation levels is associated with the development of diabetic retinopathy in rodents, and the beneficial effect of minocycline on the retinas of diabetic rodents is partially through its ability to normalize the altered histone methylation levels.
Collapse
Affiliation(s)
- Wenjun Wang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, P.R. China
| | - Simone Sidoli
- Department of Biochemistry and Molecular Biology and VILLUM Center for Bioanalytical Sciences, University of Southern Denmark, DK-5230 Odense M, Denmark
| | - Wenquan Zhang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, P.R. China
| | - Qing Wang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, P.R. China
| | - Leilei Wang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, P.R. China
| | - Ole N Jensen
- Department of Biochemistry and Molecular Biology and VILLUM Center for Bioanalytical Sciences, University of Southern Denmark, DK-5230 Odense M, Denmark
| | - Lin Guo
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, P.R. China
| | - Xiaolu Zhao
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, P.R. China
| | - Ling Zheng
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, P.R. China
| |
Collapse
|
35
|
Shultz RB, Zhong Y. Minocycline targets multiple secondary injury mechanisms in traumatic spinal cord injury. Neural Regen Res 2017; 12:702-713. [PMID: 28616020 PMCID: PMC5461601 DOI: 10.4103/1673-5374.206633] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Minocycline hydrochloride (MH), a semi-synthetic tetracycline derivative, is a clinically available antibiotic and anti-inflammatory drug that also exhibits potent neuroprotective activities. It has been shown to target multiple secondary injury mechanisms in spinal cord injury, via its anti-inflammatory, anti-oxidant, and anti-apoptotic properties. The secondary injury mechanisms that MH can potentially target include inflammation, free radicals and oxidative stress, glutamate excitotoxicity, calcium influx, mitochondrial dysfunction, ischemia, hemorrhage, and edema. This review discusses the potential mechanisms of the multifaceted actions of MH. Its anti-inflammatory and neuroprotective effects are partially achieved through conserved mechanisms such as modulation of p38 mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K)/Akt signaling pathways as well as inhibition of matrix metalloproteinases (MMPs). Additionally, MH can directly inhibit calcium influx through the N-methyl-D-aspartate (NMDA) receptors, mitochondrial calcium uptake, poly(ADP-ribose) polymerase-1 (PARP-1) enzymatic activity, and iron toxicity. It can also directly scavenge free radicals. Because it can target many secondary injury mechanisms, MH treatment holds great promise for reducing tissue damage and promoting functional recovery following spinal cord injury.
Collapse
Affiliation(s)
- Robert B Shultz
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Yinghui Zhong
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA
| |
Collapse
|
36
|
Figueira I, Fernandes A, Mladenovic Djordjevic A, Lopez-Contreras A, Henriques CM, Selman C, Ferreiro E, Gonos ES, Trejo JL, Misra J, Rasmussen LJ, Xapelli S, Ellam T, Bellantuono I. Interventions for age-related diseases: Shifting the paradigm. Mech Ageing Dev 2016; 160:69-92. [DOI: 10.1016/j.mad.2016.09.009] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 09/18/2016] [Accepted: 09/28/2016] [Indexed: 12/14/2022]
|
37
|
Esalatmanesh S, Abrishami Z, Zeinoddini A, Rahiminejad F, Sadeghi M, Najarzadegan MR, Shalbafan MR, Akhondzadeh S. Minocycline combination therapy with fluvoxamine in moderate-to-severe obsessive-compulsive disorder: A placebo-controlled, double-blind, randomized trial. Psychiatry Clin Neurosci 2016; 70:517-526. [PMID: 27488081 DOI: 10.1111/pcn.12430] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 07/16/2016] [Accepted: 07/31/2016] [Indexed: 01/12/2023]
Abstract
AIM Several lines of evidence implicate glutamatergic dysfunction in the pathophysiology of obsessive-compulsive disorder (OCD), presenting this neurotransmitter as a target for the development of novel pharmacotherapy. The objective of this study was to assess the efficacy of minocycline as an augmentative agent to fluvoxamine in the treatment of patients with OCD. METHODS One hundred and two patients with the diagnosis of moderate-to-severe OCD were recruited to this study. A randomized double-blind trial was designed and patients received either L-carnosine or placebo as adjuvant to fluvoxamine for 10 weeks. The patients randomly received either minocycline 100 mg twice per day or placebo for 10 weeks. All patients received fluvoxamine (100 mg/day) for the first 4 weeks, followed by 200 mg/day for the rest of the trial, regardless of their treatment groups. Participants were evaluated using the Yale-Brown Obsessive Compulsive Scale (Y-BOCS). The main outcome measure was to assess the efficacy of minocycline in improving the OCD symptoms. RESULTS General linear model repeated measures demonstrated significant effect for time × treatment interaction on the Y-BOCS total scores, F(1.49, 137.93) = 7.1, P = 0.003, and Y-BOCS Obsession subscale score, F(1.54, 141.94) = 9.72, P = 0.001, and near significant effect for the Y-BOCS Compulsion subscale score, F(1.27, 117.47) = 2.92, P = 0.08. A significantly greater rate of partial and complete response was observed in the minocycline group (P < 0.001). The frequency of side-effects was not significantly different between the treatment arms. CONCLUSION The results of this study suggest that minocycline could be a tolerable and effective adjuvant in the management of patients with OCD.
Collapse
Affiliation(s)
- Sophia Esalatmanesh
- Psychiatric Research Center, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Zoha Abrishami
- Psychiatric Research Center, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Atefeh Zeinoddini
- Psychiatric Research Center, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Rahiminejad
- Psychiatric Research Center, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Majid Sadeghi
- Psychiatric Research Center, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mohammad-Reza Shalbafan
- Mental Health Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shahin Akhondzadeh
- Psychiatric Research Center, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
38
|
Abstract
BACKGROUND Cognitive dysfunction in major depressive disorder (MDD) encompasses several domains, including but not limited to executive function, verbal memory, and attention. Furthermore, cognitive dysfunction is a frequent residual manifestation in depression and may persist during the remitted phase. Cognitive deficits may also impede functional recovery, including workforce performance, in patients with MDD. The overarching aims of this opinion article are to critically evaluate the effects of available antidepressants as well as novel therapeutic targets on neurocognitive dysfunction in MDD. DISCUSSION Conventional antidepressant drugs mitigate cognitive dysfunction in some people with MDD. However, a significant proportion of MDD patients continue to experience significant cognitive impairment. Two multicenter randomized controlled trials (RCTs) reported that vortioxetine, a multimodal antidepressant, has significant precognitive effects in MDD unrelated to mood improvement. Lisdexamfetamine dimesylate was shown to alleviate executive dysfunction in an RCT of adults after full or partial remission of MDD. Preliminary evidence also indicates that erythropoietin may alleviate cognitive dysfunction in MDD. Several other novel agents may be repurposed as cognitive enhancers for MDD treatment, including minocycline, insulin, antidiabetic agents, angiotensin-converting enzyme inhibitors, S-adenosyl methionine, acetyl-L-carnitine, alpha lipoic acid, omega-3 fatty acids, melatonin, modafinil, galantamine, scopolamine, N-acetylcysteine, curcumin, statins, and coenzyme Q10. The management of cognitive dysfunction remains an unmet need in the treatment of MDD. However, it is hoped that the development of novel therapeutic targets will contribute to 'cognitive remission', which may aid functional recovery in MDD.
Collapse
|
39
|
da Silva Dias IC, Carabelli B, Ishii DK, de Morais H, de Carvalho MC, Rizzo de Souza LE, Zanata SM, Brandão ML, Cunha TM, Ferraz AC, Cunha JM, Zanoveli JM. Indoleamine-2,3-Dioxygenase/Kynurenine Pathway as a Potential Pharmacological Target to Treat Depression Associated with Diabetes. Mol Neurobiol 2015; 53:6997-7009. [PMID: 26671617 DOI: 10.1007/s12035-015-9617-0] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 12/07/2015] [Indexed: 12/20/2022]
Abstract
Diabetes is a chronic disease associated with depression whose pathophysiological mechanisms that associate these conditions are not fully elucidated. However, the activation of the indoleamine-2,3-dioxygenase (IDO), an enzyme that participate of the tryptophan metabolism leading to a decrease of serotonin (5-HT) levels and whose expression is associated with an immune system activation, has been proposed as a common mechanism that links depression and diabetes. To test this hypothesis, diabetic (DBT) and normoglycemic (NGL) groups had the cytokines (TNFα, IL-1β, and IL-6) and 5-HT and norepinephrine (NE) levels in the hippocampus (HIP) evaluated. Moreover, the effect of the selective serotonin reuptake inhibitor fluoxetine (FLX), IDO direct inhibitor 1-methyl-tryptophan (1-MT), anti-inflammatory and IDO indirect inhibitor minocycline (MINO), or non-selective cyclooxygenase inhibitor ibuprofen (IBU) was evaluated in DBT rats submitted to the modified forced swimming test (MFST). After the behavioral test, the HIP was obtained for IDO expression by Western blotting analysis. DBT rats exhibited a significant increase in HIP levels of TNFα, IL-1β, and IL-6 and a decrease in HIP 5-HT and NA levels. They also presented a depressive-like behavior which was reverted by all employed treatments. Interestingly, treatment with MINO, IBU, or FLX but not with 1-MT reduced the increased IDO expression in the HIP from DBT animals. Taken together, our data support our hypothesis that neuroinflammation in the HIP followed by IDO activation with a consequent decrease in the 5-HT levels can be a possible pathophysiological mechanism that links depression to diabetes.
Collapse
Affiliation(s)
- Isabella Caroline da Silva Dias
- Department of Pharmacology, Federal University of Paraná, Rua Coronel H dos Santos S/N, P.O. Box 19031, Curitiba, PR, 81540-990, Brazil
| | - Bruno Carabelli
- Department of Physiology, Federal University of Paraná, Curitiba, PR, 81540-990, Brazil
| | - Daniela Kaori Ishii
- Department of Pharmacology, Federal University of Paraná, Rua Coronel H dos Santos S/N, P.O. Box 19031, Curitiba, PR, 81540-990, Brazil
| | - Helen de Morais
- Department of Pharmacology, Federal University of Paraná, Rua Coronel H dos Santos S/N, P.O. Box 19031, Curitiba, PR, 81540-990, Brazil
| | - Milene Cristina de Carvalho
- Institute of Neurosciences and Behavior (INeC) and Laboratory of Neuropsychopharmacology of Faculty of Philosophy, Sciences and Letters of University of São Paulo, Ribeirão Preto, SP, 14040-901, Brazil
| | - Luiz E Rizzo de Souza
- Department of Basic Pathology, Laboratory of Neurobiology, Federal University of Paraná, Curitiba, PR, 81531-990, Brazil
| | - Silvio M Zanata
- Department of Basic Pathology, Laboratory of Neurobiology, Federal University of Paraná, Curitiba, PR, 81531-990, Brazil
| | - Marcus Lira Brandão
- Institute of Neurosciences and Behavior (INeC) and Laboratory of Neuropsychopharmacology of Faculty of Philosophy, Sciences and Letters of University of São Paulo, Ribeirão Preto, SP, 14040-901, Brazil
| | - Thiago Mattar Cunha
- Department of Pharmacology, Faculty of Medicine, University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Anete Curte Ferraz
- Department of Physiology, Federal University of Paraná, Curitiba, PR, 81540-990, Brazil
| | - Joice Maria Cunha
- Department of Pharmacology, Federal University of Paraná, Rua Coronel H dos Santos S/N, P.O. Box 19031, Curitiba, PR, 81540-990, Brazil
| | - Janaina Menezes Zanoveli
- Department of Pharmacology, Federal University of Paraná, Rua Coronel H dos Santos S/N, P.O. Box 19031, Curitiba, PR, 81540-990, Brazil.
| |
Collapse
|
40
|
Suppression of microglia activation after hypoxia-ischemia results in age-dependent improvements in neurologic injury. J Neuroimmunol 2015; 291:18-27. [PMID: 26857490 DOI: 10.1016/j.jneuroim.2015.12.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 12/05/2015] [Accepted: 12/09/2015] [Indexed: 12/14/2022]
Abstract
We previously found increased microglial proliferation and pro-inflammatory cytokine release in infant mice compared to juvenile mice after hypoxia-ischemia (HI). The aim of the current study was to assess for differences in the effect of microglial suppression on HI-induced brain injury in infant and juvenile mice. HI was induced in neonatal (P9) and juvenile (P30) mice and minocycline or vehicle was administered at 2h and 24h post-HI. P9 minocycline-treated mice demonstrated early but transient improvements in neurologic injury, while P30 minocycline-treated mice demonstrated sustained improvements in cerebral atrophy and Morris Water Maze performance at 60days post-HI.
Collapse
|
41
|
Flowers SA, Ellingrod VL. The Microbiome in Mental Health: Potential Contribution of Gut Microbiota in Disease and Pharmacotherapy Management. Pharmacotherapy 2015; 35:910-6. [DOI: 10.1002/phar.1640] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Stephanie A. Flowers
- College of Pharmacy, Clinical Social and Administrative Sciences; University of Michigan; Ann Arbor Michigan
| | - Vicki L. Ellingrod
- College of Pharmacy, Clinical Social and Administrative Sciences; University of Michigan; Ann Arbor Michigan
- Department of Psychiatry; School of Medicine; University of Michigan; Ann Arbor Michigan
| |
Collapse
|
42
|
Aras M, Altas M, Motor S, Dokuyucu R, Yilmaz A, Ozgiray E, Seraslan Y, Yilmaz N. Protective effects of minocycline on experimental spinal cord injury in rats. Injury 2015; 46:1471-4. [PMID: 26052053 DOI: 10.1016/j.injury.2015.05.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 04/07/2015] [Accepted: 05/01/2015] [Indexed: 02/02/2023]
Abstract
BACKGROUND The effects of minocycline on neuronal injury after spinal cord injury (SCI) are limited and controversial. Therefore we aimed to investigate the protective effects of minocycline on tissue and on serum concentrations of malondialdehyde (MDA) levels, superoxide dismutase (SOD) activity, glutathione peroxidase (GSH-Px) activity, tissue total antioxidant and oxidant status (TAS and TOS, respectively), and AST and LDH levels in rats with SCI. METHODS This study was performed on 7-8 weeks 38 male Wistar albino rats. The animals were randomly divided into five groups: group 1, Sham (n=8); group 2, SCI (spinal cord injury)/control (n=8); group 3, SCI+minocycline3 (n=7); group 4, SCI+minocycline30 (n=8) and group 5 SCI+minocycline90 (n=7). Blood and tissue samples were analysed for MDA, SOD, GSH-Px, TAS, TOS, AST and LDH levels. RESULTS The MDA levels were significantly higher in SCI group compared to sham group (p<0.001), and MDA levels were also significantly higher in SCI group compared to SCI+M3, SCI+M30, SCI+M90 (p<0.05). SOD levels were significantly higher in SCI+M30 when compared to SCI and SCI+M3 groups (p<0.05). GSH-Px levels decreased significantly in SCI and SCI+M3 groups compared to sham (p<0.05). SCI+M3 group showed significantly decreased levels of TAS and TOS compared to SCI group (p<0.05). TAS and TOS levels significantly increased in SCI+M90 group compared to SCI+M3 and SCI+M30 groups (p<0.05). CONCLUSIONS The present study demonstrates the dose-dependent antioxidant activity of minocycline against spinal cord injury in rats. Minocycline administration increased antioxidant enzyme levels and improved total antioxidant status.
Collapse
Affiliation(s)
- Mustafa Aras
- Mustafa Kemal University, Department of Neurosurgery, Hatay, Turkey.
| | - Murat Altas
- Mustafa Kemal University, Department of Neurosurgery, Hatay, Turkey
| | - Sedat Motor
- Mustafa Kemal University, Department of Biochemistry, Hatay, Turkey
| | - Recep Dokuyucu
- Mustafa Kemal University, Department of Physiology, Hatay, Turkey
| | - Atilla Yilmaz
- Mustafa Kemal University, Department of Neurosurgery, Hatay, Turkey
| | - Erkin Ozgiray
- Ege University, Department of Neurosurgery, Izmir, Turkey
| | - Yurdal Seraslan
- Mustafa Kemal University, Department of Neurosurgery, Hatay, Turkey
| | - Nebi Yilmaz
- Mustafa Kemal University, Department of Neurosurgery, Hatay, Turkey
| |
Collapse
|
43
|
Réus GZ, Abelaira HM, Maciel AL, Dos Santos MAB, Carlessi AS, Steckert AV, Ferreira GK, De Prá SD, Streck EL, Macêdo DS, Quevedo J. Minocycline protects against oxidative damage and alters energy metabolism parameters in the brain of rats subjected to chronic mild stress. Metab Brain Dis 2015; 30:545-53. [PMID: 25112549 DOI: 10.1007/s11011-014-9602-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 08/06/2014] [Indexed: 01/30/2023]
Abstract
Studies have been suggested that minocycline can be a potential new agent for the treatment of depression. In addition, both oxidative stress and energy metabolism present an important role in pathophysiology of depression. So, the present study was aimed to evaluate the effects of minocycline on stress oxidative parameters and energy metabolism in the brain of adult rats submitted to the chronic mild stress protocol (CMS). After CMS Wistar, both stressed animals as controls received twice ICV injection of minocycline (160 μg) or vehicle. The oxidative stress and energy metabolism parameters were assessed in the prefrontal cortex (PF), hippocampus (HIP), amygdala (AMY) and nucleus accumbens (Nac). Our findings showed that stress induced an increase on protein carbonyl in the PF, AMY and NAc, and mynocicline injection reversed this alteration. The TBARS was increased by stress in the PF, HIP and NAc, however, minocycline reversed the alteration in the PF and HIP. The Complex I was incrased in AMY by stress, and minocycline reversed this effect, however reduced Complex I activity in the NAc; Complex II reduced in PF and AMY by stress or minocycline; the Complex II-III increased in the HIP in stress plus minocycline treatment and in the NAc with minocycline; in the PF and HIP there were a reduced in Complex IV with stress and minocycline. The creatine kinase was reduced in AMY and NAc with stress and minocycline. In conclusion, minocycline presented neuroprotector effects by reducing oxidative damage and regulating energy metabolism in specific brain areas.
Collapse
Affiliation(s)
- Gislaine Z Réus
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Dean OM, Maes M, Ashton M, Berk L, Kanchanatawan B, Sughondhabirom A, Tangwongchai S, Ng C, Dowling N, Malhi GS, Berk MI. Protocol and rationale-the efficacy of minocycline as an adjunctive treatment for major depressive disorder: a double blind, randomised, placebo controlled trial. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2014; 12:180-8. [PMID: 25598820 PMCID: PMC4293162 DOI: 10.9758/cpn.2014.12.3.180] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 04/29/2014] [Accepted: 04/30/2014] [Indexed: 01/28/2023]
Abstract
While current pharmacotherapies are efficacious, there remain a clear shortfall between symptom remission and functional recovery. With the explosion in our understanding of the biology of these disorders, the time is ripe for the investigation of novel therapies. Recently depression is conceptualized as an immune-inflammatory and nitro-oxidative stress related disorder. Minocycline is a tetracycline antibiotic that has anti-inflammatory, pro-oxidant, glutamatergic, neurotrophic and neuroprotective properties that make it a viable target to explore as a new therapy. This double blind, randomised, placebo controlled adjunctive trial will investigate the benefits of 200 mg/day of minocycline treatment, in addition to any usual treatment, as an adjunctive treatment for moderate-severe major depressive disorder. Sixty adults are being randomised to 12 weeks of treatment (with a 4 week follow-up post-discontinuation). The primary outcome measure for the study is mean change on the Montgomery-Asberg Depression Rating Scale (MADRS), with secondary outcomes including the Social and Occupational Functioning Assessment Scale (SOFAS), Clinical Global Impressions (CGI), Hamilton Rating Scale for Anxiety (HAM-A), Patient Global Impression (PGI), Quality of Life Enjoyment and Satisfaction Questionnaire (Q-LES-Q) and Range of Impaired Functioning Tool (LIFE-RIFT). Biomarker analyses will also be conducted at baseline and week 12. The study has the potential to provide new treatment targets, both by showing efficacy with a new class of 'antidepressant' but also through the analysis of biomarkers that may further inform our understanding of the pathophysiology of unipolar depression.
Collapse
Affiliation(s)
- Olivia May Dean
- Innovations in Mental and Physical Health and Clinical Treatments Strategic Research Centre, Deakin University, School of Medicine, Geelong, Australia. ; Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, Australia. ; Department of Psychiatry, University of Melbourne, Parkville, Australia
| | - Michael Maes
- Department of Psychiatry, Chulalongkorn University, Bangkok, Thailand
| | - Melanie Ashton
- Innovations in Mental and Physical Health and Clinical Treatments Strategic Research Centre, Deakin University, School of Medicine, Geelong, Australia
| | - Lesley Berk
- Department of Psychiatry, Chulalongkorn University, Bangkok, Thailand
| | - Buranee Kanchanatawan
- Centre for Mental Health and Wellbeing Research, Deakin University, School of Psychology, Burwood, Australia
| | - Atapol Sughondhabirom
- Centre for Mental Health and Wellbeing Research, Deakin University, School of Psychology, Burwood, Australia
| | - Sookjareon Tangwongchai
- Centre for Mental Health and Wellbeing Research, Deakin University, School of Psychology, Burwood, Australia
| | - Chee Ng
- Department of Psychiatry, University of Melbourne, The Melbourne Clinic, Richmond, Australia
| | - Nathan Dowling
- Department of Psychiatry, University of Melbourne, The Melbourne Clinic, Richmond, Australia
| | - Gin S Malhi
- Discipline of Psychiatry, Sydney Medical School, University of Sydney, Sydney, Australia. ; Department of Psychiatry, CADE Clinic, St. Leonards, Australia
| | - MIchael Berk
- Innovations in Mental and Physical Health and Clinical Treatments Strategic Research Centre, Deakin University, School of Medicine, Geelong, Australia. ; Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, Australia. ; Department of Psychiatry, University of Melbourne, Parkville, Australia. ; Orygen Youth Health Research Centre, Parkville, Australia. ; Centre of Youth Mental Health, University of Melbourne, Parkville, Australia
| |
Collapse
|
45
|
Goossens J, Hachimi-Idrissi S. Combination of therapeutic hypothermia and other neuroprotective strategies after an ischemic cerebral insult. Curr Neuropharmacol 2014; 12:399-412. [PMID: 25426009 PMCID: PMC4243031 DOI: 10.2174/1570159x12666140424233036] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 03/14/2014] [Accepted: 04/22/2014] [Indexed: 12/31/2022] Open
Abstract
Abrupt deprivation of substrates to neuronal tissue triggers a number of pathological events (the “ischemic cascade”) that lead to cell death. As this is a process of delayed neuronal cell death and not an instantaneous event, several pharmacological and non-pharmacological strategies have been developed to attenuate or block this cascade. The most promising neuroprotectant so far is therapeutic hypothermia and its beneficial effects have inspired researchers to further improve its protective benefit by combining it with other neuroprotective agents. This review provides an overview of all neuroprotective strategies that have been combined with therapeutic hypothermia in rodent models of focal cerebral ischemia. A distinction is made between drugs interrupting only one event of the ischemic cascade from those mitigating different pathways and having multimodal effects. Also the combination of therapeutic hypothermia with hemicraniectomy, gene therapy and protein therapy is briefly discussed. Furthermore, those combinations that have been studied in a clinical setting are also reviewed.
Collapse
Affiliation(s)
- Joline Goossens
- Critical Care Department and Cerebral Resuscitation Research Group, Ghent University, De Pintelaan 185, 9000 Ghent, Belgium
| | - Saïd Hachimi-Idrissi
- Critical Care Department and Cerebral Resuscitation Research Group, Ghent University, De Pintelaan 185, 9000 Ghent, Belgium
| |
Collapse
|
46
|
Fond G, Hamdani N, Kapczinski F, Boukouaci W, Drancourt N, Dargel A, Oliveira J, Le Guen E, Marlinge E, Tamouza R, Leboyer M. Effectiveness and tolerance of anti-inflammatory drugs' add-on therapy in major mental disorders: a systematic qualitative review. Acta Psychiatr Scand 2014; 129:163-79. [PMID: 24215721 DOI: 10.1111/acps.12211] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/20/2013] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To provide a systematic review of the literature regarding the efficacy of anti-inflammatory drugs in three major mental disorders [major depressive disorder (MDD), schizophrenia and bipolar disorders]. METHOD Four databases were explored, without any year or language restrictions. The baseline search paradigm was limited to open-labelled clinical and randomized controlled trials (RCTs). RESULTS Four major classes of anti-inflammatory drugs were identified, namely polyunsaturated fatty acids (PUFAs), cyclooxygenase (COX) inhibitors, anti-TNFalpha and minocycline. Effectiveness and benefit/risk ratio of each class in MDD, bipolar disorders and schizophrenia was detailed when data were available. Several meta-analyses indicated effectiveness of PUFAs in MDD with a good tolerance profile. One meta-analysis indicated that COX-2 specific inhibitors showed effectiveness in schizophrenia. Anti-TNFalpha showed important effectiveness in resistant MDD with blood inflammatory abnormalities. Minocycline showed effectiveness in schizophrenia. CONCLUSION Polyunsaturated fatty acids seem to have the best benefit/risk ratio profile but proved their effectiveness only in MDD. A number of anti-inflammatory drugs are available as adjunct treatment for treatment-resistant patients with MDD, schizophrenia and bipolar disorder. If used with caution regarding their possible side-effects, they may be reasonable therapeutic alternatives for resistant symptomatology.
Collapse
Affiliation(s)
- G Fond
- Pôle de psychiatrie des hôpitaux universitaires H Mondor, University Paris Est-Créteil, INSERM U955, Eq Psychiatrie Génétique, Fondation FondaMental Fondation de coopération scientifique en santé mentale, Créteil, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
Accumulating evidence suggests that neuroinflammation affecting microglia plays an important role in the etiology of schizophrenia, and appropriate control of microglial activation may be a promising therapeutic strategy for schizophrenia. Minocycline, a second-generation tetracycline that inhibits microglial activation, has been shown to have a neuroprotective effect in various models of neurodegenerative disease, including anti-inflammatory, antioxidant, and antiapoptotic properties, and an ability to modulate glutamate-induced excitotoxicity. Given that these mechanisms overlap with neuropathologic pathways, minocycline may have a potential role in the adjuvant treatment of schizophrenia, and improve its negative symptoms. Here, we review the relevant studies of minocycline, ranging from preclinical research to human clinical trials.
Collapse
Affiliation(s)
- Lulu Zhang
- Mental Health Institute of the Second Xiangya Hospital, Key Laboratory of Psychiatry and Mental Health of Hunan Province, Central South University, Changsha, Hunan, People's Republic of China ; Department of Psychology, Guangzhou First People's Hospital, Guangzhou, Guangdong, People's Republic of China
| | - Jingping Zhao
- Mental Health Institute of the Second Xiangya Hospital, Key Laboratory of Psychiatry and Mental Health of Hunan Province, Central South University, Changsha, Hunan, People's Republic of China
| |
Collapse
|
48
|
Chen YI, Lee YJ, Wilkie DA, Lin CT. Evaluation of potential topical and systemic neuroprotective agents for ocular hypertension-induced retinal ischemia-reperfusion injury. Vet Ophthalmol 2013; 17:432-42. [PMID: 24171811 DOI: 10.1111/vop.12105] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To evaluate for drugs with superior neuroprotective efficacy and investigate their underlying mechanisms related to antioxidation. PROCEDURES Brinzolamide (1%), timolol (0.5%), minocycline (22 mg/kg), lidocaine (1.5 mg/kg), and methylprednisolone (30 mg/kg) were administered to Sprague-Dawley (SD) rats. The retina was evaluated by electroretinography and histological analysis. The antioxidative capacity of drugs was evaluated to clarify the underlying mechanism. The oxidant/antioxidant profiles of plasma, red blood cells, and retina were analyzed by lipid peroxidation (malondialdehyde) and by measuring the activities of antioxidants. Proteomic analysis was used to investigate the possible protective mechanisms of the drug against ischemia-reperfusion injury. RESULTS The results suggested that timolol, methylprednisolone, and minocycline protected retinal function. Methylprednisolone and minocycline possessed good antioxidative activity. Brinzolamide and lidocaine preserved the structural integrity of the retina, but not retinal function. CONCLUSION Methylprednisolone, minocycline, and timolol have potential acute or delayed benefit in retinal ischemia-reperfusion injury. Their neuroprotective actions depend at least partially on the ability to alleviate oxidative stress.
Collapse
Affiliation(s)
- Yi-Ing Chen
- Institute of Veterinary Clinical Sciences, School of Veterinary Medicine, National Taiwan University, No. 1, Section 4, Roosevelt Road, Taipei, 106, Taiwan
| | | | | | | |
Collapse
|
49
|
Yui J, Xie L, Fujinaga M, Yamasaki T, Hatori A, Kumata K, Nengaki N, Zhang MR. Monitoring Neuroprotective Effects Using Positron Emission Tomography With [
11
C]ITMM, a Radiotracer for Metabotropic Glutamate 1 Receptor. Stroke 2013; 44:2567-72. [DOI: 10.1161/strokeaha.113.001178] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background and Purpose—
Recent pharmacological evidence shows that antagonists for the metabotropic glutamate 1 (mGlu1) receptor exhibit neuroprotective effects in an ischemic brain. The aim of this study was to visualize the mGlu1 receptor and to monitor neuroprotective effects in a rat model of mild focal ischemia using positron emission tomography (PET) with
N
-[4-[6-(isopropylamino)pyrimidin-4-yl]-1,3-thiazol-2-yl]-4-[
11
C]methoxy-
N-
methylbenzamide ([
11
C]ITMM), a radiotracer for mGlu1.
Methods—
Rats were subjected to a 30-minute transient right middle cerebral artery occlusion. Saline or minocycline, a neuroprotective agent, was intravenously injected immediately after surgery and then daily during the subsequent 7 days. PET imaging with [
11
C]ITMM was performed on the rats on days 1 to 7 after ischemia. In vitro autoradiography and histopathologic staining were conducted to confirm the results of in vivo PET.
Results—
PET with [
11
C]ITMM demonstrated a gradual decrease of radioactivity in the ipsilateral sides of the ischemic brains. The radioactivity uptake ratio between the ipsilateral and contralateral sides also decreased with time. Minocycline treatment slowed down the decrease in the radioactivity level in the ipsilateral sides. Pretreatment with JNJ16259685, an mGlu1-selective ligand, significantly reduced brain radioactivity, confirming that the uptake of [
11
C]ITMM primarily reflects mGlu1 levels in the brain regions, including the ischemic area. In vitro autoradiography and histopathology confirmed the changes in mGlu1 levels in the brains.
Conclusions—
[
11
C]ITMM-PET may be a useful technique for characterizing the change in mGlu1 level during the occurrence and progression of neuronal damage and for evaluating the neuroprotective effects of drugs after ischemia.
Collapse
Affiliation(s)
- Joji Yui
- From the Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan (J.Y., L.X., M.F., T.Y., A.H., K.K., N.N., M.-R.Z.); and SHI Accelerator Service Co Ltd, Tokyo, Japan (N.N.)
| | - Lin Xie
- From the Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan (J.Y., L.X., M.F., T.Y., A.H., K.K., N.N., M.-R.Z.); and SHI Accelerator Service Co Ltd, Tokyo, Japan (N.N.)
| | - Masayuki Fujinaga
- From the Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan (J.Y., L.X., M.F., T.Y., A.H., K.K., N.N., M.-R.Z.); and SHI Accelerator Service Co Ltd, Tokyo, Japan (N.N.)
| | - Tomoteru Yamasaki
- From the Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan (J.Y., L.X., M.F., T.Y., A.H., K.K., N.N., M.-R.Z.); and SHI Accelerator Service Co Ltd, Tokyo, Japan (N.N.)
| | - Akiko Hatori
- From the Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan (J.Y., L.X., M.F., T.Y., A.H., K.K., N.N., M.-R.Z.); and SHI Accelerator Service Co Ltd, Tokyo, Japan (N.N.)
| | - Katsushi Kumata
- From the Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan (J.Y., L.X., M.F., T.Y., A.H., K.K., N.N., M.-R.Z.); and SHI Accelerator Service Co Ltd, Tokyo, Japan (N.N.)
| | - Nobuki Nengaki
- From the Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan (J.Y., L.X., M.F., T.Y., A.H., K.K., N.N., M.-R.Z.); and SHI Accelerator Service Co Ltd, Tokyo, Japan (N.N.)
| | - Ming-Rong Zhang
- From the Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan (J.Y., L.X., M.F., T.Y., A.H., K.K., N.N., M.-R.Z.); and SHI Accelerator Service Co Ltd, Tokyo, Japan (N.N.)
| |
Collapse
|
50
|
Kohler E, Prentice DA, Bates TR, Hankey GJ, Claxton A, van Heerden J, Blacker D. Intravenous minocycline in acute stroke: a randomized, controlled pilot study and meta-analysis. Stroke 2013; 44:2493-9. [PMID: 23868273 DOI: 10.1161/strokeaha.113.000780] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Minocycline, in animal models and 2 small randomized controlled human trials, is a promising neuroprotective agent in acute stroke. We analyzed the efficacy and safety of intravenous minocycline in acute ischemic and hemorrhagic stroke. METHODS A multicenter prospective randomized open-label blinded end point evaluation pilot study of minocycline 100 mg administered intravenously, commenced within 24 hours of onset of stroke, and continued 12 hourly for a total of 5 doses, versus no minocycline. All participants received routine stroke care. Primary end point was survival free of handicap (modified Rankin Scale, ≤2) at day 90. RESULTS Ninety-five participants were randomized; 47 to minocycline and 48 to no minocycline. In the intention-to-treat population, 29 of 47 (65.9%) allocated minocycline survived free of handicap compared with 33 of 48 (70.2%) allocated no minocycline (rate ratio, 0.94; 95% confidence interval, 0.71-1.25 and odds ratio, 0.73; 95% CI, 0.31-1.71). A meta-analysis of the 3 human trials suggests minocycline may increase the odds of handicap-free survival by 3-fold (odds ratio, 2.99; 95% CI, 1.74-5.16) but there was substantial heterogeneity among the trials. CONCLUSIONS In this pilot study of a small sample of acute stroke patients, intravenous minocycline was safe but not efficacious. The study was not powered to identify reliably or exclude a modest but clinically important treatment effect of minocycline. Larger trials would improve the precision of the estimates of any treatment effect of minocycline. CLINICAL TRIAL REGISTRATION URL http://www.anzctr.org.au. Unique identifier: ACTRN12612000237886.
Collapse
|