1
|
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] [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
|
2
|
Dal-Pizzol F, Coelho A, Simon CS, Michels M, Corneo E, Jeremias A, Damásio D, Ritter C. Prophylactic Minocycline for Delirium in Critically Ill Patients: A Randomized Controlled Trial. Chest 2024; 165:1129-1138. [PMID: 38043911 DOI: 10.1016/j.chest.2023.11.041] [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: 07/13/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 12/05/2023] Open
Abstract
BACKGROUND Delirium is a potentially severe form of acute encephalopathy. Minocycline has neuroprotective effects in animal models of neurologic diseases; however, data from human studies remain scarce. RESEARCH QUESTION Does the neuroprotective effect of minocycline prevent delirium occurrence in critically ill patients? STUDY DESIGN AND METHODS This study was a randomized, placebo-controlled, double-anonymized trial conducted in four ICUs. Patients aged 18 years or older were eligible and randomized to receive minocycline (100 mg, twice daily) or placebo. The primary outcome was delirium incidence within 28 days or before ICU discharge. Secondary outcomes included days in delirium during ICU stay, delirium/coma-free days, length of mechanical ventilation, ICU length of stay, ICU mortality, and hospital mortality. The kinetics of various inflammatory (IL-1β, IL-6, IL-10, and C-reactive protein) and brain-related biomarkers (brain-derived neurotrophic factor and S100B) were used as exploratory outcomes. RESULTS A total of 160 patients were randomized, but one patient in the placebo group died before treatment; thus the data from 159 patients were analyzed (minocycline, n = 84; placebo, n = 75). After the COVID-19 pandemic it was decided to stop patient inclusion early. There was a small but significant decrease in delirium incidence: 17 patients (20%) in the minocycline arm compared with 26 patients (35%) in the placebo arm (P = .043). No other delirium-related outcomes were modified by minocycline treatment. Unexpectedly, there was a significant decrease in hospital mortality (39% vs. 23%; P = .029). Among all analyzed biomarkers, only plasma levels of C-reactive protein decreased significantly after minocycline treatment (F = 0.75, P = .78, within time; F = 4.09, P = .045, group × time). INTERPRETATION Our findings in this rather small study signal a possible positive effect of minocycline on delirium incidence. Further studies are needed to confirm the benefits of this drug as a preventive measure in critically ill patients. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov; No.: NCT04219735; URL: www. CLINICALTRIALS gov.
Collapse
Affiliation(s)
- Felipe Dal-Pizzol
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Universidade do Extremo Sul Catarinense, Criciúma, Brazil; Intensive Care Unit, São José Hospital, Criciúma, Brazil; São José Hospital Research Center, Criciúma, Brazil.
| | - André Coelho
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Universidade do Extremo Sul Catarinense, Criciúma, Brazil; Intensive Care Unit, São José Hospital, Criciúma, Brazil
| | - Carla S Simon
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Universidade do Extremo Sul Catarinense, Criciúma, Brazil
| | - Monique Michels
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Universidade do Extremo Sul Catarinense, Criciúma, Brazil
| | - Emily Corneo
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Universidade do Extremo Sul Catarinense, Criciúma, Brazil
| | | | | | - Cristiane Ritter
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Universidade do Extremo Sul Catarinense, Criciúma, Brazil; Intensive Care Unit, São José Hospital, Criciúma, Brazil
| |
Collapse
|
3
|
Michaelis L, Berg L, Maier L. Confounder or Confederate? The Interactions Between Drugs and the Gut Microbiome in Psychiatric and Neurological Diseases. Biol Psychiatry 2024; 95:361-369. [PMID: 37331548 DOI: 10.1016/j.biopsych.2023.06.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/06/2023] [Accepted: 06/08/2023] [Indexed: 06/20/2023]
Abstract
The gut microbiome is emerging as an important factor in signaling along the gut-brain axis. The intimate physiological connection between the gut and the brain allows perturbations in the microbiome to be directly transmitted to the central nervous system and thereby contribute to psychiatric and neurological diseases. Common microbiome perturbations result from the ingestion of xenobiotic compounds including pharmaceuticals such as psychotropic drugs. In recent years, a variety of interactions between these drug classes and the gut microbiome have been reported, ranging from direct inhibitory effects on gut bacteria to microbiome-mediated drug degradation or sequestration. Consequently, the microbiome may play a critical role in influencing the intensity, duration, and onset of therapeutic effects, as well as in influencing the side effects that patients may experience. Furthermore, because the composition of the microbiome varies from person to person, the microbiome may contribute to the frequently observed interpersonal differences in the response to these drugs. In this review, we first summarize the known interactions between xenobiotics and the gut microbiome. Then, for psychopharmaceuticals, we address the question of whether these interactions with gut bacteria are irrelevant for the host (i.e., merely confounding factors in metagenomic analyses) or whether they may even have therapeutic or adverse effects.
Collapse
Affiliation(s)
- Lena Michaelis
- Interfaculty Institute for Microbiology and Infection Medicine Tübingen, University of Tübingen, Tübingen, Germany; and the Cluster of Excellence EXC 2124 (Controlling Microbes to Fight Infections), University of Tübingen, Tübingen, Germany
| | - Lara Berg
- Interfaculty Institute for Microbiology and Infection Medicine Tübingen, University of Tübingen, Tübingen, Germany; and the Cluster of Excellence EXC 2124 (Controlling Microbes to Fight Infections), University of Tübingen, Tübingen, Germany
| | - Lisa Maier
- Interfaculty Institute for Microbiology and Infection Medicine Tübingen, University of Tübingen, Tübingen, Germany; and the Cluster of Excellence EXC 2124 (Controlling Microbes to Fight Infections), University of Tübingen, Tübingen, Germany.
| |
Collapse
|
4
|
Fond G, Mallet J, Urbach M, Benros ME, Berk M, Billeci M, Boyer L, Correll CU, Fornaro M, Kulkarni J, Leboyer M, Llorca PM, Misdrahi D, Rey R, Schürhoff F, Solmi M, Sommer IEC, Stahl SM, Pignon B, Berna F. Adjunctive agents to antipsychotics in schizophrenia: a systematic umbrella review and recommendations for amino acids, hormonal therapies and anti-inflammatory drugs. BMJ MENTAL HEALTH 2023; 26:e300771. [PMID: 37852631 PMCID: PMC10583081 DOI: 10.1136/bmjment-2023-300771] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 09/02/2023] [Indexed: 10/20/2023]
Abstract
QUESTION This umbrella review and guidelines aimed to provide evidence to support the rational choice of selected adjunctive therapies for schizophrenia. STUDY SELECTION AND ANALYSIS Following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) and World Federation of Societies of Biological Psychiatry (WFSBP)-grading recommendations, 63 randomised control trials (RCTs) (of which 4219 unique participants have completed the RCTs) and 29 meta-analyses were analysed. FINDINGS Provisional recommendations (WFSBP-grade 1) could be made for two molecules in augmentation to antipsychotics: (1) N-acetyl-cysteine (NAC, 1200-3600 mg/day, for >12 consecutive weeks) in improving negative symptoms, general psychopathology (positive and negative syndrome scale for schizophrenia (PANSS) general psychopathology factor (G)-G subscale), with the RCTs with the longer duration showing the most robust findings; (2) polyunsaturated fatty acids (3000 mg/day of eicosapentaenoic acid, for >12 weeks) in improving general psychopathology. Weaker recommendations (ie, WFSBP-grade 2) could be drawn for sarcosine (2 g/day) and minocycline (200-300 mg/day) for improving negative symptoms in chronic schizophrenia (not early schizophrenia), and NAC for improving positive symptoms and cognition. Weak recommendations are not ready for clinical practice. There is provisional evidence that oestrogens and raloxifene are effective in some patients, but further research is needed to determine their benefit/risk ratio. CONCLUSIONS The results of this umbrella review should be interpreted with caution as the number of RCTs included in the meta-analyses was generally small and the effect sizes were weak or medium. For NAC, two RCTs with low risk of bias have provided conflicting results and the WFSBP-grade recommendation included also the results of meta-analyses. These drugs could be provisionally prescribed for patients for whom no other treatments have been effective, but they should be discontinued if they prove ineffective.
Collapse
Affiliation(s)
- Guillaume Fond
- Department of psychiatry, Assistance Publique des Hôpitaux de Marseille, Marseille, France
- Fondation FondaMental, Creteil, France
- CEReSS-Health Service Research and Quality of Life Center, AMU, Marseille, France
| | - Jasmina Mallet
- Fondation FondaMental, Creteil, France
- Department of Psychiatry, Louis Mourier Hospital, Colombes, France
| | - Mathieu Urbach
- Fondation FondaMental, Creteil, France
- Department of Adult Psychiatry and Addictology, Centre Hospitalier de Versailles, Le Chesnay, France
| | - Michael Eriksen Benros
- Copenhagen Research Center for Biological and Precision Psychiatry, Mental Health Centre Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael Berk
- Deakin University, School of Medicine, and Barwon Health; IMPACT, the Institute for Mental and Physical Health and Clinical Translation; Orygen The National Centre of Excellence in Youth Mental Health, The Florey Institute of Neuroscience and Mental Health and the Department of Psychiatry, University of Melbourne and the Department of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Martina Billeci
- Department of Neuroscience, Reproductive Sciences, and Dentistry, Section of Psychiatry, Federico II University of Naples, Naples, Italy
| | - Laurent Boyer
- CEReSS-Health Service Research and Quality of Life Center, AMU, Marseille, France
- Département d'information médicale, Assistance Publique des Hôpitaux de Marseille, Marseille, France
| | - Christoph U Correll
- Department of Psychiatry, Zucker Hillside Hospital, Glen Oaks, New York, USA
- Department of Psychiatry and Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
- Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin Berlin, Berlin, Germany
- German Center for Mental Health (DZPG), partner site Berlin, Berlin, Germany
| | - Michele Fornaro
- Department of Neuroscience, Reproductive Sciences, and Dentistry, Section of Psychiatry, Federico II University of Naples, Naples, Italy
| | - Jayashri Kulkarni
- Department of Psychiatry, Monash Alfred Psychiatry Research Centre, Alfred Hospital and Monash University Central Clinical School, Monash University,607StKildaRd, Level4, Melbourne, Victoria, Australia 3004, Melbourne, Victoria, Australia
| | - Marion Leboyer
- Fondation FondaMental, Creteil, France
- Department of psychiatry, Univ Paris-Est-Créteil (UPEC), AP-HP, Hôpitaux Universitaires « H. Mondor », DMU IMPACT, INSERM U955, IMRB, translational Neuropsychiatry, F-94010 Creteil, France, Créteil, France
| | - Pierre-Michel Llorca
- Fondation FondaMental, Creteil, France
- Département de psychiatrie, Université Clermont Auvergne, CMP-B CHU, CNRS,Clermont Auvergne INP, Institut Pascal, Clermont-Ferrand, France
| | - David Misdrahi
- Fondation FondaMental, Creteil, France
- Departement de Psychiatrie Générale et Universitaire, Centre Hospitalier Charles Perrens; Univ. Bordeaux, CNRS, UMR 5287, F-33000, INCIA, Bordeaux, France
| | - Romain Rey
- Fondation FondaMental, Creteil, France
- Schizophrenia Expert Centre, Le Vinatier Hospital; INSERM, U1028; CNRS, UMR5292; University Lyon 1; Lyon Neuroscience Research Center, PSYR2 Team, Lyon, France
| | - Franck Schürhoff
- Fondation FondaMental, Creteil, France
- Department of psychiatry, Univ Paris-Est-Créteil (UPEC), AP-HP, Hôpitaux Universitaires « H. Mondor », DMU IMPACT, INSERM U955, IMRB, translational Neuropsychiatry, F-94010 Creteil, France, Créteil, France
| | - Marco Solmi
- Department of Psychiatry, University of Ottawa, Ottawa, Ontario, Canada
- Department of Mental Health, The Champlain First Episode Psychosis Program, The Ottawa Hospital, Ottawa, Ontario, Canada
- Ottawa Hospital Research Institute (OHRI) Clinical Epidemiology Program University of Ottawa, Ottawa Hospital Research Institute (OHRI), Ottawa, Ontario, Canada
- Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany
| | - Iris E C Sommer
- Department of Psychiatry, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - Stephen M Stahl
- Department of Psychiatry, University of California, San Diego, California, USA
| | - Baptiste Pignon
- Fondation FondaMental, Creteil, France
- Department of psychiatry, Univ Paris-Est-Créteil (UPEC), AP-HP, Hôpitaux Universitaires « H. Mondor », DMU IMPACT, INSERM U955, IMRB, translational Neuropsychiatry, F-94010 Creteil, France, Créteil, France
| | - Fabrice Berna
- Fondation FondaMental, Creteil, France
- Psychiatry, Strasbourg University Hospital, University of Strasbourg, Strasbourg, France
| |
Collapse
|
5
|
Ortega MA, Álvarez-Mon MA, García-Montero C, Fraile-Martínez Ó, Monserrat J, Martinez-Rozas L, Rodríguez-Jiménez R, Álvarez-Mon M, Lahera G. Microbiota-gut-brain axis mechanisms in the complex network of bipolar disorders: potential clinical implications and translational opportunities. Mol Psychiatry 2023; 28:2645-2673. [PMID: 36707651 PMCID: PMC10615769 DOI: 10.1038/s41380-023-01964-w] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 01/02/2023] [Accepted: 01/13/2023] [Indexed: 01/28/2023]
Abstract
Bipolar disorders (BD) represent a severe leading disabling mental condition worldwide characterized by episodic and often progressive mood fluctuations with manic and depressive stages. The biological mechanisms underlying the pathophysiology of BD remain incompletely understood, but it seems that there is a complex picture of genetic and environmental factors implicated. Nowadays, gut microbiota is in the spotlight of new research related to this kind of psychiatric disorder, as it can be consistently related to several pathophysiological events observed in BD. In the context of the so-called microbiota-gut-brain (MGB) axis, it is shown to have a strong influence on host neuromodulation and endocrine functions (i.e., controlling the synthesis of neurotransmitters like serotonin or mediating the activation of the hypothalamic-pituitary-adrenal axis), as well as in modulation of host immune responses, critically regulating intestinal, systemic and brain inflammation (neuroinflammation). The present review aims to elucidate pathophysiological mechanisms derived from the MGB axis disruption and possible therapeutic approaches mainly focusing on gut microbiota in the complex network of BD. Understanding the mechanisms of gut microbiota and its bidirectional communication with the immune and other systems can shed light on the discovery of new therapies for improving the clinical management of these patients. Besides, the effect of psychiatric drugs on gut microbiota currently used in BD patients, together with new therapeutical approaches targeting this ecosystem (dietary patterns, probiotics, prebiotics, and other novelties) will also be contemplated.
Collapse
Affiliation(s)
- Miguel A Ortega
- Department of Medicine and Medical Specialities, University of Alcala, Alcalá de Henares, Spain.
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain.
| | - Miguel Angel Álvarez-Mon
- Department of Medicine and Medical Specialities, University of Alcala, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Department of Psychiatry and Mental Health, Hospital Universitario Infanta Leonor, Madrid, Spain
| | - Cielo García-Montero
- Department of Medicine and Medical Specialities, University of Alcala, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Óscar Fraile-Martínez
- Department of Medicine and Medical Specialities, University of Alcala, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Jorge Monserrat
- Department of Medicine and Medical Specialities, University of Alcala, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Lucia Martinez-Rozas
- Department of Medicine and Medical Specialities, University of Alcala, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Roberto Rodríguez-Jiménez
- Department of Legal Medicine and Psychiatry, Complutense University, Madrid, Spain
- Institute for Health Research 12 de Octubre Hospital, (Imas 12)/CIBERSAM (Biomedical Research Networking Centre in Mental Health), Madrid, Spain
| | - Melchor Álvarez-Mon
- Department of Medicine and Medical Specialities, University of Alcala, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Immune System Diseases-Rheumatology, Oncology Service an Internal Medicine, University Hospital Príncipe de Asturias (CIBEREHD), Alcalá de Henares, Spain
- Psychiatry Service, Center for Biomedical Research in the Mental Health Network, University Hospital Príncipe de Asturias, Alcalá de Henares, Spain
| | - Guillermo Lahera
- Department of Medicine and Medical Specialities, University of Alcala, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Psychiatry Service, Center for Biomedical Research in the Mental Health Network, University Hospital Príncipe de Asturias, Alcalá de Henares, Spain
| |
Collapse
|
6
|
Messina A, Concerto C, Rodolico A, Petralia A, Caraci F, Signorelli MS. Is It Time for a Paradigm Shift in the Treatment of Schizophrenia? The Use of Inflammation-Reducing and Neuroprotective Drugs-A Review. Brain Sci 2023; 13:957. [PMID: 37371435 DOI: 10.3390/brainsci13060957] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/03/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Comprehending the pathogenesis of schizophrenia represents a challenge for global mental health. To date, although it is evident that alterations in dopaminergic, serotonergic, and glutamatergic neurotransmission underlie the clinical expressiveness of the disease, neuronal disconnections represent only an epiphenomenon. In recent years, several clinical studies have converged on the hypothesis of microglia hyperactivation and a consequent neuroinflammatory state as a pathogenic substrate of schizophrenia. Prenatal, perinatal, and postnatal factors can cause microglia to switch from M2 anti-inflammatory to M1 pro-inflammatory states. A continuous mild neuroinflammatory state progressively leads to neuronal loss, a reduction in dendritic spines, and myelin degeneration. The augmentation of drugs that reduce neuroinflammation to antipsychotics could be an effective therapeutic modality in managing schizophrenia. This review will consider studies in which drugs with anti-inflammatory and neuroprotective properties have been used in addition to antipsychotic treatment in patients with schizophrenia.
Collapse
Affiliation(s)
- Antonino Messina
- Department of Clinical and Experimental Medicine, Institute of Psychiatry, University of Catania, 95123 Catania, Italy
| | - Carmen Concerto
- Department of Clinical and Experimental Medicine, Institute of Psychiatry, University of Catania, 95123 Catania, Italy
| | - Alessandro Rodolico
- Department of Clinical and Experimental Medicine, Institute of Psychiatry, University of Catania, 95123 Catania, Italy
| | - Antonino Petralia
- Department of Clinical and Experimental Medicine, Institute of Psychiatry, University of Catania, 95123 Catania, Italy
| | - Filippo Caraci
- Department of Drug and Health Sciences, University of Catania, 95123 Catania, Italy
- Unit of Translational Neuropharmacology and Translational Neurosciences, Oasi Research Institute-IRCCS, 94018 Troina, Italy
| | - Maria Salvina Signorelli
- Department of Clinical and Experimental Medicine, Institute of Psychiatry, University of Catania, 95123 Catania, Italy
| |
Collapse
|
7
|
Panizzutti B, Skvarc D, Lin S, Croce S, Meehan A, Bortolasci CC, Marx W, Walker AJ, Hasebe K, Kavanagh BE, Morris MJ, Mohebbi M, Turner A, Gray L, Berk L, Walder K, Berk M, Dean OM. Minocycline as Treatment for Psychiatric and Neurological Conditions: A Systematic Review and Meta-Analysis. Int J Mol Sci 2023; 24:ijms24065250. [PMID: 36982324 PMCID: PMC10049047 DOI: 10.3390/ijms24065250] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Minocycline has anti-inflammatory, antioxidant, and anti-apoptotic properties that explain the renewed interest in its use as an adjunctive treatment for psychiatric and neurological conditions. Following the completion of several new clinical trials using minocycline, we proposed an up-to-date systematic review and meta-analysis of the data available. The PICO (patient/population, intervention, comparison and outcomes) framework was used to search 5 databases aiming to identify randomized controlled trials that used minocycline as an adjunctive treatment for psychiatric and neurological conditions. Search results, data extraction, and risk of bias were performed by two independent authors for each publication. Quantitative meta-analysis was performed using RevMan software. Literature search and review resulted in 32 studies being included in this review: 10 in schizophrenia, 3 studies in depression, and 7 in stroke, with the benefit of minocycline being used in some of the core symptoms evaluated; 2 in bipolar disorder and 2 in substance use, without demonstrating a benefit for using minocycline; 1 in obsessive-compulsive disorder, 2 in brain and spinal injuries, 2 in amyotrophic lateral sclerosis, 1 in Alzheimer’s disease, 1 in multiple systems atrophy, and 1 in pain, with mixes results. For most of the conditions included in this review the data is still limited and difficult to interpret, warranting more well-designed and powered studies. On the other hand, the studies available for schizophrenia seem to suggest an overall benefit favoring the use of minocycline as an adjunctive treatment.
Collapse
Affiliation(s)
- Bruna Panizzutti
- IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Barwon Health, School of Medicine, Deakin University, Geelong, VIC 3220, Australia
| | - David Skvarc
- School of Psychology, Faculty of Health, Deakin University, Geelong, VIC 3220, Australia
| | - Sylvia Lin
- IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Barwon Health, School of Medicine, Deakin University, Geelong, VIC 3220, Australia
- Melbourne Neuropsychiatry Centre, University of Melbourne, Parkville, VIC 3053, Australia
| | - Sarah Croce
- IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Barwon Health, School of Medicine, Deakin University, Geelong, VIC 3220, Australia
| | - Alcy Meehan
- IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Barwon Health, School of Medicine, Deakin University, Geelong, VIC 3220, Australia
| | - Chiara Cristina Bortolasci
- IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Barwon Health, School of Medicine, Deakin University, Geelong, VIC 3220, Australia
| | - Wolfgang Marx
- IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Barwon Health, School of Medicine, Deakin University, Geelong, VIC 3220, Australia
| | - Adam J. Walker
- IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Barwon Health, School of Medicine, Deakin University, Geelong, VIC 3220, Australia
| | - Kyoko Hasebe
- School of Biomedical Sciences, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Bianca E. Kavanagh
- IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Barwon Health, School of Medicine, Deakin University, Geelong, VIC 3220, Australia
| | - Margaret J. Morris
- School of Biomedical Sciences, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Mohammadreza Mohebbi
- IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Barwon Health, School of Medicine, Deakin University, Geelong, VIC 3220, Australia
- Biostatistics Unit, Faculty of Health, Deakin University, Burwood, VIC 3125, Australia
| | - Alyna Turner
- IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Barwon Health, School of Medicine, Deakin University, Geelong, VIC 3220, Australia
| | - Laura Gray
- IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Barwon Health, School of Medicine, Deakin University, Geelong, VIC 3220, Australia
| | - Lesley Berk
- IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Barwon Health, School of Medicine, Deakin University, Geelong, VIC 3220, Australia
| | - Ken Walder
- IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Barwon Health, School of Medicine, Deakin University, Geelong, VIC 3220, Australia
| | - Michael Berk
- IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Barwon Health, School of Medicine, Deakin University, Geelong, VIC 3220, Australia
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3052, Australia
- Orygen, The National Centre of Excellence in Youth Mental Health, Parkville, VIC 3052, Australia
| | - Olivia M. Dean
- IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Barwon Health, School of Medicine, Deakin University, Geelong, VIC 3220, Australia
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3052, Australia
- Correspondence: ; Tel.: +61-03-4215-3300
| |
Collapse
|
8
|
Simon MS, Arteaga-Henríquez G, Fouad Algendy A, Siepmann T, Illigens BMW. Anti-Inflammatory Treatment Efficacy in Major Depressive Disorder: A Systematic Review of Meta-Analyses. Neuropsychiatr Dis Treat 2023; 19:1-25. [PMID: 36636142 PMCID: PMC9830720 DOI: 10.2147/ndt.s385117] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 12/13/2022] [Indexed: 01/06/2023] Open
Abstract
Purpose Immune imbalances in major depressive disorder (MDD) have been targeted by anti-inflammatory treatment approaches in clinical trials to increase responsiveness to therapy. However, even after several meta-analyses, no translation of evidence into clinical practice has taken place. We performed a systematic review to evaluate meta-analytic evidence of randomized controlled trials on the use of anti-inflammatory agents for MDD to summarize efficacy estimates and elucidate shortcomings. Methods Pooled effect estimates and heterogeneity indices were primary outcomes. Characteristics of the included meta-analyses were extracted. Scientific quality of meta-analyses was assessed using the Revised Assessment of Multiple Systematic Reviews (R-AMSTAR). Results N=20 meta-analyses met the eligibility criteria. Study characteristics like outcome scales, composition of patient populations, and add-on or monotherapy regimen varied very little for celecoxib studies, varied little for minocycline studies, and were rather variable for omega 3 fatty acids studies. R-AMSTAR scores ranged from 26 to 39 out of 44 points indicating variable quality, where a comprehensive literature search was the strongest and the consideration of scientific quality in the conclusions was the weakest domain across all meta-analyses. For minocycline and celecoxib, superiority was demonstrated with medium to large effect size with substantial heterogeneity and with large to very large effect size with negligible heterogeneity, respectively. For omega 3 fatty acids, superiority was also demonstrated with mainly small and medium effect sizes with substantial heterogeneity. However, for minocycline and omega 3 fatty acids, non-significant meta-analyses were found also. Conclusion Even in our synthesized approach, no clear recommendations could be derived on the use of anti-inflammatory treatment for MDD due to several critical aspects like heterogeneity, diversity of patient populations, treatment regimen, and outcomes, and limited scientific quality. However, we observed clear inter-substance differences with meta-analytic evidence being strongest for celecoxib and weakest for omega 3 fatty acids.
Collapse
Affiliation(s)
- Maria S Simon
- Division of Health Care Sciences, Dresden International University, Dresden, Saxony, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilians-University, Munich, Bavaria, Germany
| | - Gara Arteaga-Henríquez
- Department of Psychiatry, Hospital Universitari Vall d’Hebron, Vall d’Hebron Research Institute (VHIR), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain
| | - Ahmed Fouad Algendy
- Division of Health Care Sciences, Dresden International University, Dresden, Saxony, Germany
- Clinical Pharmacy, Geriatric Department, Rumailah Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Timo Siepmann
- Division of Health Care Sciences, Dresden International University, Dresden, Saxony, Germany
- Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Saxony, Germany
| | - Ben M W Illigens
- Division of Health Care Sciences, Dresden International University, Dresden, Saxony, Germany
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
9
|
Chaves Filho AJM, Mottin M, Lós DB, Andrade CH, Macedo DS. The tetrapartite synapse in neuropsychiatric disorders: Matrix metalloproteinases (MMPs) as promising targets for treatment and rational drug design. Biochimie 2022; 201:79-99. [PMID: 35931337 DOI: 10.1016/j.biochi.2022.07.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 06/26/2022] [Accepted: 07/26/2022] [Indexed: 02/06/2023]
Abstract
Inflammation and an exacerbated immune response are widely accepted contributing mechanisms to the genesis and progression of major neuropsychiatric disorders. However, despite the impressive advances in understanding the neurobiology of these disorders, there is still no approved drug directly linked to the regulation of inflammation or brain immune responses. Importantly, matrix metalloproteinases (MMPs) comprise a group of structurally related endopeptidases primarily involved in remodeling extracellular matrix (ECM). In the central nervous system (CNS), these proteases control synaptic plasticity and strength, patency of the blood-brain barrier, and glia-neuron interactions through cleaved and non-cleaved mediators. Several pieces of evidence have pointed to a complex scenario of MMPs dysregulation triggered by neuroinflammation. Furthermore, major psychiatric disorders' affective symptoms and neurocognitive abnormalities are related to MMPs-mediated ECM changes and neuroglia activation. In the past decade, research efforts have been directed to broad-spectrum MMPs inhibitors with frustrating clinical results. However, in the light of recent advances in combinatorial chemistry and drug design technologies, specific and CNS-oriented MMPs modulators have been proposed as a new frontier of therapy for regulating ECM properties in the CNS. Therefore, here we aim to discuss the state of the art of MMPs and ECM abnormalities in major neuropsychiatric disorders, namely depression, bipolar disorder, and schizophrenia, the possible neuro-immune interactions involved in this complex scenario of MMPs dysregulation and propose these endopeptidases as promising targets for rational drug design.
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, Brazil; Laboratory for Molecular Modeling and Drug Design - LabMol, Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia, GO, Brazil.
| | - Melina Mottin
- Laboratory for Molecular Modeling and Drug Design - LabMol, Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Deniele Bezerra Lós
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Carolina Horta Andrade
- Laboratory for Molecular Modeling and Drug Design - LabMol, Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Danielle S Macedo
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| |
Collapse
|
10
|
Structural and Functional Deviations of the Hippocampus in Schizophrenia and Schizophrenia Animal Models. Int J Mol Sci 2022; 23:ijms23105482. [PMID: 35628292 PMCID: PMC9143100 DOI: 10.3390/ijms23105482] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/09/2022] [Accepted: 05/11/2022] [Indexed: 01/04/2023] Open
Abstract
Schizophrenia is a grave neuropsychiatric disease which frequently onsets between the end of adolescence and the beginning of adulthood. It is characterized by a variety of neuropsychiatric abnormalities which are categorized into positive, negative and cognitive symptoms. Most therapeutical strategies address the positive symptoms by antagonizing D2-dopamine-receptors (DR). However, negative and cognitive symptoms persist and highly impair the life quality of patients due to their disabling effects. Interestingly, hippocampal deviations are a hallmark of schizophrenia and can be observed in early as well as advanced phases of the disease progression. These alterations are commonly accompanied by a rise in neuronal activity. Therefore, hippocampal formation plays an important role in the manifestation of schizophrenia. Furthermore, studies with animal models revealed a link between environmental risk factors and morphological as well as electrophysiological abnormalities in the hippocampus. Here, we review recent findings on structural and functional hippocampal abnormalities in schizophrenic patients and in schizophrenia animal models, and we give an overview on current experimental approaches that especially target the hippocampus. A better understanding of hippocampal aberrations in schizophrenia might clarify their impact on the manifestation and on the outcome of this severe disease.
Collapse
|
11
|
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
|
12
|
Romero-Miguel D, Casquero-Veiga M, MacDowell KS, Torres-Sanchez S, Garcia-Partida JA, Lamanna-Rama N, Romero-Miranda A, Berrocoso E, Leza JC, Desco M, Soto-Montenegro ML. A Characterization of the Effects of Minocycline Treatment During Adolescence on Structural, Metabolic, and Oxidative Stress Parameters in a Maternal Immune Stimulation Model of Neurodevelopmental Brain Disorders. Int J Neuropsychopharmacol 2021; 24:734-748. [PMID: 34165516 PMCID: PMC8453277 DOI: 10.1093/ijnp/pyab036] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 06/01/2021] [Accepted: 06/18/2021] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Minocycline (MIN) is a tetracycline with antioxidant, anti-inflammatory, and neuroprotective properties. Given the likely involvement of inflammation and oxidative stress (IOS) in schizophrenia, MIN has been proposed as a potential adjuvant treatment in this pathology. We tested an early therapeutic window, during adolescence, as prevention of the schizophrenia-related deficits in the maternal immune stimulation (MIS) animal model. METHODS On gestational day 15, Poly I:C or vehicle was injected in pregnant Wistar rats. A total 93 male offspring received MIN (30 mg/kg) or saline from postnatal day (PND) 35-49. At PND70, rats were submitted to the prepulse inhibition test. FDG-PET and T2-weighted MRI brain studies were performed at adulthood. IOS markers were evaluated in frozen brain tissue. RESULTS MIN treatment did not prevent prepulse inhibition test behavioral deficits in MIS offspring. However, MIN prevented morphometric abnormalities in the third ventricle but not in the hippocampus. Additionally, MIN reduced brain metabolism in cerebellum and increased it in nucleus accumbens. Finally, MIN reduced the expression of iNOS (prefrontal cortex, caudate-putamen) and increased the levels of KEAP1 (prefrontal cortex), HO1 and NQO1 (amygdala, hippocampus), and HO1 (caudate-putamen). CONCLUSIONS MIN treatment during adolescence partially counteracts volumetric abnormalities and IOS deficits in the MIS model, likely via iNOS and Nrf2-ARE pathways, also increasing the expression of cytoprotective enzymes. However, MIN treatment during this peripubertal stage does not prevent sensorimotor gating deficits. Therefore, even though it does not prevent all the MIS-derived abnormalities evaluated, our results suggest the potential utility of early treatment with MIN in other schizophrenia domains.
Collapse
Affiliation(s)
| | - Marta Casquero-Veiga
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain,CIBER de Salud Mental (CIBERSAM), Madrid, Spain
| | - Karina S MacDowell
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain,Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense (UCM), IIS Imas12, IUIN, Madrid, Spain
| | - Sonia Torres-Sanchez
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain,Neuropsychopharmacology and Psychobiology Research Group, Psychobiology Area, Department of Psychology, Universidad de Cádiz, Puerto Real (Cádiz), Spain,Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Cádiz, Spain
| | - José Antonio Garcia-Partida
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain,Neuropsychopharmacology and Psychobiology Research Group, Psychobiology Area, Department of Psychology, Universidad de Cádiz, Puerto Real (Cádiz), Spain,Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Cádiz, Spain
| | | | | | - Esther Berrocoso
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain,Neuropsychopharmacology and Psychobiology Research Group, Psychobiology Area, Department of Psychology, Universidad de Cádiz, Puerto Real (Cádiz), Spain,Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Cádiz, Spain
| | - Juan C Leza
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain,Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense (UCM), IIS Imas12, IUIN, Madrid, Spain
| | - Manuel Desco
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain,CIBER de Salud Mental (CIBERSAM), Madrid, Spain,Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, Leganés, Spain,Centro Nacional de Investigaciones Cardiovasculares, CNIC, Madrid, Spain,Correspondence: Manuel Desco, PhD, Laboratorio de Imagen Médica, Unidad de Medicina y Cirugía Experimental, Hospital General Universitario Gregorio Marañón, Dr. Esquerdo, 46. E-28007 Madrid, Spain ()
| | - María Luisa Soto-Montenegro
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain,CIBER de Salud Mental (CIBERSAM), Madrid, Spain,High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut), University Rey Juan Carlos (URJC), Alcorcón, Spain
| |
Collapse
|
13
|
Germann M, Brederoo SG, Sommer IEC. Abnormal synaptic pruning during adolescence underlying the development of psychotic disorders. Curr Opin Psychiatry 2021; 34:222-227. [PMID: 33560023 PMCID: PMC8048735 DOI: 10.1097/yco.0000000000000696] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Excessive synaptic pruning has first been suggested by Irwin Feinberg (1982) as an important pillar in the pathophysiology in schizophrenia (SCZ). This article reviews recent developments highlighting factors implicated in aberrant synaptic pruning and its contribution to disease onset and emergence of cognitive symptoms in SCZ. Unraveling these factors provides new insights for potential prevention and treatment strategies for psychotic disorders. RECENT FINDINGS Increased pruning in SCZ was recently confirmed by a positron emission tomography-study employing the novel tracer [11C]UCB-J, demonstrating the consequential loss of synaptic density. Recent evidence supports the contributing role of astrocytes and increased complement-mediated microglial pruning in disease onset and cognitive symptoms in SCZ. Increased microglial pruning is mediated specifically by C4. Furthermore, environmental factors (e.g., infections and stress) can lead to dysbiosis which was recently linked to microglial activation and pruning in SCZ. SUMMARY Recent findings render the pruning machinery a potential target for early treatment and prevention in individuals at high risk for SCZ. Minocycline can improve cognition in SCZ, probably by reducing excessive pruning. Probiotics might also have beneficial effects on cognition, although recent findings are not encouraging. N-acetyl-cysteine recovers functional connectivity in SCZ both in vitro and in vivo, making it an interesting candidate.
Collapse
Affiliation(s)
- Monique Germann
- University of Groningen, Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, Groningen, The Netherlands
| | | | | |
Collapse
|
14
|
Wang B, Lin W, Zhu H. Minocycline improves the recovery of nerve function and alleviates blood-brain barrier damage by inhibiting endoplasmic reticulum in traumatic brain injury mice model. EUR J INFLAMM 2021. [DOI: 10.1177/20587392211010898] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) is a clinical emergency with a very high incidence, disability, and fatality rate. Minocycline, a widely used semisynthetic second-generation tetracycline antibiotic, has anti-inflammatory and bactericidal effects. However, minocycline has not been explored as a therapeutic drug in TBI and if effective, the related molecular mechanism is also unclear. In this study, we examined the neuroprotective effect and possible mechanism of minocycline, in mice TBI model by studying the trauma-related functional and morphological changes. Also, in vitro cell studies were carried out to verify the animal model data. We found that minocycline significantly improved the neurobehavioral score, inhibited apoptosis, repaired the blood-brain barrier, and reduced the levels of inflammatory factors Interleukin-6 and tumor necrosis factor-α in TBI mice. In vitro, upon oxygen and glucose deprivation, minocycline reduced the levels of cellular inflammatory factors and increased the levels of tight junction and adherens junction proteins, thereby significantly improving the cell viability. Moreover, Mino treatment prevented the loss of tight junction and adherens junction proteins which were markedly reversed by an ER stress activator (tunicamycin) both in vivo and in vitro. Our findings set an effective basis for the clinical use of Mino to treat Traumatic brain injury-induced neurological deficits.
Collapse
Affiliation(s)
- Bingbin Wang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou
| | - Wendong Lin
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou
| | - Haiping Zhu
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou
| |
Collapse
|
15
|
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
|
16
|
Sun XN, Zhou JB, Li N. Poor Oral Health in Patients with Schizophrenia: a Meta-Analysis of Case-Control Studies. Psychiatr Q 2021; 92:135-145. [PMID: 32483766 DOI: 10.1007/s11126-020-09752-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Patients with schizophrenia have high rates of comorbid physical illness, but there has been less attention to dental diseases in these patients. This meta-analysis of case-control studies systematically examined the oral health in patients with schizophrenia. Case-control studies comparing the oral health in patients with schizophrenia and healthy controls were screened and identified. Standardized mean difference (SMD) with its 95% confidence interval (CI) was calculated using RevMan version 5.3. Three case-control studies comprising 306 patients with schizophrenia and 306 healthy controls were included in this meta-analysis. All studies were rated as "high quality". Patients with schizophrenia had significantly higher scores of decayed, missing and filled teeth (SMD = 0.83, 95%CI: 0.57, 1.09, p < 0.001; I2 = 51%), missing teeth (SMD = 0.79, 95%CI: 0.59, 0.98, p < 0.001; I2 = 19%), and decayed teeth (SMD = 0.89, 95%CI: 0.24, 1.54, p = 0.008; I2 = 92%) when compared to healthy controls. Similarly, patients with schizophrenia had significantly lower filled teeth scores (SMD = -0.76, 95%CI: -1.44, -0.09, p = 0.03; I2 = 93%) when compared to healthy controls. This meta-analysis found that patients with schizophrenia were likely to have worse oral health when compared to healthy controls.
Collapse
Affiliation(s)
- Xiao-Ni Sun
- Dental Department, Ningbo Yinzhou People's Hospital Community, Zhejiang, 315040, China.
| | - Jian-Bo Zhou
- Dental Department, Ningbo Yinzhou People's Hospital Community, Zhejiang, 315040, China
| | - Na Li
- Dental Department, Ningbo Yinzhou People's Hospital Community, Zhejiang, 315040, China
| |
Collapse
|
17
|
Koola MM. Alpha7 nicotinic-N-methyl-D-aspartate hypothesis in the treatment of schizophrenia and beyond. Hum Psychopharmacol 2021; 36:1-16. [PMID: 32965756 DOI: 10.1002/hup.2758] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 09/08/2020] [Accepted: 09/09/2020] [Indexed: 12/12/2022]
Abstract
Development of novel treatments for positive, cognitive, and negative symptoms continue to be a high-priority area of schizophrenia research and a major unmet clinical need. Given that all randomized controlled trials (RCTs) conducted to date failed with one add-on medication/mechanism of action, future RCTs with the same approach are not warranted. Even if the field develops a medication for cognition, others are still needed to treat negative and positive symptoms. Therefore, fixing one domain does not completely solve the problem. Also, targeting the cholinergic system, glutamatergic system, and cholinergic plus alpha7 nicotinic and N-methyl-D-aspartate (NMDA) receptors failed independently. Hence, targeting other less important pathophysiological mechanisms/targets is unlikely to be successful. Meta-analyses of RCTs targeting major pathophysiological mechanisms have found some efficacy signal in schizophrenia; thus, combination treatments with different mechanisms of action may enhance the efficacy signal. The objective of this article is to highlight the importance of conducting RCTs with novel combination treatments in schizophrenia to develop antischizophrenia treatments. Positive RCTs with novel combination treatments that target the alpha7 nicotinic and NMDA receptors simultaneously may lead to a disease-modifying therapeutic armamentarium in schizophrenia. Novel combination treatments that concurrently improve the three domains of psychopathology and several prognostic and theranostic biomarkers may facilitate therapeutic discovery in schizophrenia.
Collapse
Affiliation(s)
- Maju Mathew Koola
- Department of Psychiatry and Behavioral Health, Stony Brook University Renaissance School of Medicine, Stony Brook, New York, USA
| |
Collapse
|
18
|
Abstract
Vagus nerve stimulation (VNS) has been increasingly studied in treating treatment-resistant depression (TRD), but the findings have been mixed. This updated meta-analysis was conducted to examine the efficacy and safety of adjunctive VNS for TRD. Controlled studies reporting on the efficacy and safety of adjunctive VNS for TRD were screened, identified and analyzed. Standardized mean difference (SMD), risk ratio (RR) and their 95% confidence intervals (CIs) were analyzed using RevMan version 5.3. Three controlled studies with a total of 1048 patients with TRD compared VNS (n = 622) with control (n = 426) groups. Only one study was rated as 'high quality' using the Jadad scale. Adjunctive VNS was significantly superior to the control group regarding study-defined response [SMD:1.96 (95%CI:1.60, 2.40), P < 0.00001, I2 = 0%]. Patient-reported voice alteration occurred more frequently with adjunctive VNS for patients with TRD. No significant group differences were found regarding discontinuation due to any reason [RR:0.50 (95%CI:0.12, 2.09), P = 0.34, I2 = 85%]. Adjunctive VNS appeared to be effective and relatively safe treatment for TRD. Further randomized controlled trials are needed to confirm the efficacy and safety of VNS for TRD.
Collapse
|
19
|
Li XM, Shi ZM, Wang PJ, Hu H. Effects of ketamine in electroconvulsive therapy for major depressive disorder: meta-analysis of randomised controlled trials. Gen Psychiatr 2020; 33:e100117. [PMID: 32596639 PMCID: PMC7299006 DOI: 10.1136/gpsych-2019-100117] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 03/30/2020] [Accepted: 04/16/2020] [Indexed: 01/20/2023] Open
Abstract
Background The use of ketamine in electroconvulsive therapy (ECT) has been examined in the treatment of major depressive disorder (MDD); however, there has been no systematic review and meta-analysis of related randomised controlled trials (RCTs). Aim To examine the efficacy and safety of ketamine augmentation of ECT in MDD treatment. Methods Two reviewers searched Chinese (China National Knowledge Infrastructure and Wanfang) and English (PubMed, PsycINFO, Embase and Cochrane Library) databases from their inception to 23 July 2019. The included studies' bias risk was evaluated using the Cochrane risk of bias assessment tool. The primary outcome of this meta-analysis was improved depressive symptoms at day 1 after a single ECT treatment session. Data were pooled to calculate the standardised mean difference and risk ratio with their 95% CIs using RevMan V.5.3. We used the Grading of Recommendations, Assessment, Development and Evaluation (GRADE) approach to assess the whole quality of evidence. Results Four RCTs (n = 239) compared ketamine alone or ketamine plus propofol (n = 149) versus propofol alone (n = 90) in patients with MDD who underwent a single ECT session. Three RCTs were considered as unclear risk with respect to random sequence generation using the Cochrane risk of bias. Compared with propofol alone, ketamine alone and the combination of ketamine and propofol had greater efficacy in the treatment of depressive symptoms at days 1, 3 and 7 after a single ECT session. Moreover, compared with propofol alone, ketamine alone and the combination of ketamine and propofol were significantly associated with increased seizure duration and seizure energy index. Compared with propofol, ketamine alone was significantly associated with increased opening-eye time. Based on the GRADE approach, the evidence level of primary and secondary outcomes ranged from very low (26.7%, 4/15) to ‘low’ (73.3%, 11/15). Conclusion Compared with propofol, there were very low or low evidence levels showing that ketamine alone and the combination of ketamine and propofol appeared to rapidly improve depressive symptoms of patients with MDD undergoing a single ECT session. There is a need for high-quality RCTs.
Collapse
Affiliation(s)
- Xiao-Mei Li
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhan-Ming Shi
- Department of Psychiatry, Chongqing Jiangbei Mental Health Center, Chongqing, China
| | - Pei-Jia Wang
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hua Hu
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
20
|
Bortolasci CC, Marx W, Walker AJ, Hasebe K, Kavanagh BE, Morris MJ, Mohebbi M, Turner A, Gray L, Berk L, Walder K, Berk M, Dean OM. Minocycline for the treatment of mental health and neurological conditions: study protocol of a systematic review and meta-analysis. BMJ Open 2020; 10:e035080. [PMID: 32198305 PMCID: PMC7103827 DOI: 10.1136/bmjopen-2019-035080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
INTRODUCTION Due to the anti-inflammatory, antioxidant and anti-apoptotic properties of minocycline, clinical trials have evaluated the potential of this drug to treat several psychiatric and neurological disorders, including major depressive disorder, schizophrenia, bipolar disorder, stroke and amyotrophic lateral sclerosis. This protocol proposes a systematic review (and potential meta-analysis) that aims to identify and critically evaluate randomised controlled trials of minocycline for treating psychiatric and neurological disorders. METHODS AND ANALYSIS PubMed, Embase, Cochrane Central Register of Controlled Clinical Trials, PsycINFO and Cumulative Index to Nursing and Allied Health Literature (CINAHL) will be used to identify randomised controlled trials that used minocycline to treat psychiatric and neurological disorders. Double-blind, randomised, controlled, clinical trials of participants aged 18 years or older and written in English will be included in the review. Data will be extracted by two independent reviewers. Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines will be followed and the Cochrane Collaboration's 'Risk of Bias' tool will be used to assess the risk of bias in all studies included in the systematic review. The Grading of Recommendations, Assessment, Development and Evaluation system will be used to access the overall quality of the level of evidence of the studies. If sufficient evidence is identified, a meta-analysis will be conducted using the standardised mean difference approach and reported with 95% CIs. Heterogeneity of evidence will be evaluated using the I2 model. ETHICS AND DISSEMINATION This systematic review will evaluate only published data; therefore, ethical approval is not required. The systematic review will be published in a peer-reviewed journal and presented at relevant research conferences. TRIAL REGISTRATION NUMBER CRD42020153292.
Collapse
Affiliation(s)
- Chiara C Bortolasci
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Barwon Health, Geelong, Victoria, Australia
| | - Wolfgang Marx
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Barwon Health, Geelong, Victoria, Australia
| | - Adam J Walker
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Barwon Health, Geelong, Victoria, Australia
| | - Kyoko Hasebe
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Barwon Health, Geelong, Victoria, Australia
- School of Medical Sciences, UNSW Sydney, Sydney, New South Wales, Australia
| | - Bianca E Kavanagh
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Barwon Health, Geelong, Victoria, Australia
| | - Margaret J Morris
- School of Medical Sciences, UNSW Sydney, Sydney, New South Wales, Australia
| | - Mohammadreza Mohebbi
- Biostatistics Unit, Faculty of Health, Deakin University, Melbourne, Victoria, Australia
| | - Alyna Turner
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Barwon Health, Geelong, Victoria, Australia
- Department of Psychiatry, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia
- School of Medicine and Public Health, Faculty of Health and Medicine, University of Newcastle & Hunter Medical Research Institute, Callaghan, New South Wales, Australia
| | - Laura Gray
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Barwon Health, Geelong, Victoria, Australia
| | - Lesley Berk
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Barwon Health, Geelong, Victoria, Australia
- Melbourne School of Population and Global Health, University of Melbourne, Carlton, Victoria, Australia
| | - Ken Walder
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Barwon Health, Geelong, Victoria, Australia
| | - Michael Berk
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Barwon Health, Geelong, Victoria, Australia
| | - Olivia M Dean
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Barwon Health, Geelong, Victoria, Australia
| |
Collapse
|
21
|
Neff AS. Technical and Theoretic Limitations of the Experimental Evidence Supporting a Gut Bacterial Etiology in Mental Illness. Clin Ther 2020; 42:e74-e81. [PMID: 32115244 DOI: 10.1016/j.clinthera.2020.02.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 02/03/2020] [Accepted: 02/04/2020] [Indexed: 02/06/2023]
Abstract
The impact of gut bacteria on the brain and behavior has become the subject of intense research. The brain is sensitive to biochemical and physiologic changes in the body, for example, changes in blood oxygenation or nutritional status. The collection of microorganisms residing within the digestive tract (the gut microbiome) is increasingly considered a major contributor to human physiology. These 2 considerations have led to the hypothesis that human psychology, including complex constructs like emotion and mental illness, could be influenced by the composition or function of gut bacteria. Five lines of evidence have been used to support the concept, including human correlational research, probiotic supplementation, antibiotic use, germ-free animal research, and fecal transplantation. Results from these experiments do not provide substantial support for the theory that complex human psychology is under the influence of gut bacteria. Placebo-controlled interventional research in humans, in particular fecal microbiota transplantation, will be required before a stronger conclusion can be reached.
Collapse
Affiliation(s)
- Andrew Steven Neff
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA; Department of Psychology and Behavioral Sciences, Rochester University, Rochester Hills, MI, USA.
| |
Collapse
|
22
|
Barnes TR, Drake R, Paton C, Cooper SJ, Deakin B, Ferrier IN, Gregory CJ, Haddad PM, Howes OD, Jones I, Joyce EM, Lewis S, Lingford-Hughes A, MacCabe JH, Owens DC, Patel MX, Sinclair JM, Stone JM, Talbot PS, Upthegrove R, Wieck A, Yung AR. Evidence-based guidelines for the pharmacological treatment of schizophrenia: Updated recommendations from the British Association for Psychopharmacology. J Psychopharmacol 2020; 34:3-78. [PMID: 31829775 DOI: 10.1177/0269881119889296] [Citation(s) in RCA: 158] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
These updated guidelines from the British Association for Psychopharmacology replace the original version published in 2011. They address the scope and targets of pharmacological treatment for schizophrenia. A consensus meeting was held in 2017, involving experts in schizophrenia and its treatment. They were asked to review key areas and consider the strength of the evidence on the risk-benefit balance of pharmacological interventions and the clinical implications, with an emphasis on meta-analyses, systematic reviews and randomised controlled trials where available, plus updates on current clinical practice. The guidelines cover the pharmacological management and treatment of schizophrenia across the various stages of the illness, including first-episode, relapse prevention, and illness that has proved refractory to standard treatment. It is hoped that the practice recommendations presented will support clinical decision making for practitioners, serve as a source of information for patients and carers, and inform quality improvement.
Collapse
Affiliation(s)
- Thomas Re Barnes
- Emeritus Professor of Clinical Psychiatry, Division of Psychiatry, Imperial College London, and Joint-head of the Prescribing Observatory for Mental Health, Centre for Quality Improvement, Royal College of Psychiatrists, London, UK
| | - Richard Drake
- Clinical Lead for Mental Health in Working Age Adults, Health Innovation Manchester, University of Manchester and Greater Manchester Mental Health NHS Foundation Trust, Manchester, UK
| | - Carol Paton
- Joint-head of the Prescribing Observatory for Mental Health, Centre for Quality Improvement, Royal College of Psychiatrists, London, UK
| | - Stephen J Cooper
- Emeritus Professor of Psychiatry, School of Medicine, Queen's University Belfast, Belfast, UK
| | - Bill Deakin
- Professor of Psychiatry, Neuroscience & Psychiatry Unit, University of Manchester and Greater Manchester Mental Health NHS Foundation Trust, Manchester, UK
| | - I Nicol Ferrier
- Emeritus Professor of Psychiatry, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - Catherine J Gregory
- Honorary Clinical Research Fellow, University of Manchester and Higher Trainee in Child and Adolescent Psychiatry, Manchester University NHS Foundation Trust, Manchester, UK
| | - Peter M Haddad
- Honorary Professor of Psychiatry, Division of Psychology and Mental Health, University of Manchester, UK and Senior Consultant Psychiatrist, Department of Psychiatry, Hamad Medical Corporation, Doha, Qatar
| | - Oliver D Howes
- Professor of Molecular Psychiatry, Imperial College London and Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Ian Jones
- Professor of Psychiatry and Director, National Centre of Mental Health, Cardiff University, Cardiff, UK
| | - Eileen M Joyce
- Professor of Neuropsychiatry, UCL Queen Square Institute of Neurology, London, UK
| | - Shôn Lewis
- Professor of Adult Psychiatry, Faculty of Biology, Medicine and Health, The University of Manchester, UK, and Mental Health Academic Lead, Health Innovation Manchester, Manchester, UK
| | - Anne Lingford-Hughes
- Professor of Addiction Biology and Honorary Consultant Psychiatrist, Imperial College London and Central North West London NHS Foundation Trust, London, UK
| | - James H MacCabe
- Professor of Epidemiology and Therapeutics, Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, and Honorary Consultant Psychiatrist, National Psychosis Service, South London and Maudsley NHS Foundation Trust, Beckenham, UK
| | - David Cunningham Owens
- Professor of Clinical Psychiatry, University of Edinburgh. Honorary Consultant Psychiatrist, Royal Edinburgh Hospital, Edinburgh, UK
| | - Maxine X Patel
- Honorary Clinical Senior Lecturer, King's College London, Institute of Psychiatry, Psychology and Neuroscience and Consultant Psychiatrist, Oxleas NHS Foundation Trust, London, UK
| | - Julia Ma Sinclair
- Professor of Addiction Psychiatry, Faculty of Medicine, University of Southampton, Southampton, UK
| | - James M Stone
- Clinical Senior Lecturer and Honorary Consultant Psychiatrist, King's College London, Institute of Psychiatry, Psychology and Neuroscience and South London and Maudsley NHS Trust, London, UK
| | - Peter S Talbot
- Senior Lecturer and Honorary Consultant Psychiatrist, University of Manchester and Greater Manchester Mental Health NHS Foundation Trust, Manchester, UK
| | - Rachel Upthegrove
- Professor of Psychiatry and Youth Mental Health, University of Birmingham and Consultant Psychiatrist, Birmingham Early Intervention Service, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - Angelika Wieck
- Honorary Consultant in Perinatal Psychiatry, Greater Manchester Mental Health NHS Foundation Trust, Manchester, UK
| | - Alison R Yung
- Professor of Psychiatry, University of Manchester, School of Health Sciences, Manchester, UK and Centre for Youth Mental Health, University of Melbourne, Australia, and Honorary Consultant Psychiatrist, Greater Manchester Mental Health NHS Foundation Trust, Manchester, UK
| |
Collapse
|
23
|
Limandri BJ. Inflammatory Response and Treatment-Resistant Mental Disorders: Should Immunotherapy Be Added to Pharmacotherapy? J Psychosoc Nurs Ment Health Serv 2020; 58:11-16. [PMID: 31895965 DOI: 10.3928/02793695-20191218-03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Treatment resistance continues to challenge and frustrate mental health clinicians and provoke psychiatric researchers to seek additional explanatory theories for psychopathology. Because the inflammatory process activates symptoms of depression, anxiety, and psychosis, it is a reasonable route to follow for primary and/or indirect contribution to mental disorders. The current article reviews the research literature regarding the role the inflammatory process and immune system play in mental disorders as well as novel treatments under investigation for resistant depression, anxiety, substance use, and psychotic disorders. [Journal of Psychosocial Nursing and Mental Health Services, 58(1), 11-16.].
Collapse
|
24
|
Abstract
People with bipolar disorder (BD) all too often have suboptimal long-term outcomes with existing treatment options. They experience relapsing episodes of depression and mania and also have interepisodic mood and anxiety symptoms. We need to have a better understanding of the pathophysiology of BD if we are to make progress in improving these outcomes. This chapter will focus on the critical role of mitochondria in human functioning, oxidative stress, and the biological mechanisms of mitochondria in BD. Additionally, this chapter will present the evidence that, at least for some people, BD is a product of mitochondrial dysregulation. We review the modulators of mitochondria, the connection between current BD medication treatments and mitochondria, and additional medications that have theoretical potential to treat BD.
Collapse
|