Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
Add more filters

Publication year range
1.
Transl Psychiatry ; 14(1): 14, 2024 Jan 08.
Article in English | MEDLINE | ID: mdl-38191622

ABSTRACT

Schizophrenia is a chronic neurodevelopmental disorder with an inflammatory/prooxidant component. N-acetylcysteine (NAC) has been evaluated in schizophrenia as an adjuvant to antipsychotics, but its role as a preventive strategy has not been sufficiently explored. We aimed to evaluate the potential of NAC administration in two-time windows before the onset of symptoms in a schizophrenia-like maternal immune stimulation (MIS) rat model. Pregnant Wistar rats were injected with Poly I:C or Saline on gestational day (GD) 15. Three different preventive approaches were evaluated: 1) NAC treatment during periadolescence in the offspring (from postnatal day [PND] 35 to 49); 2) NAC treatment during pregnancy after MIS challenge until delivery (GD15-21); and 3) NAC treatment throughout all pregnancy (GD1-21). At postnatal day (PND) 70, prepulse inhibition (PPI) and anxiety levels were evaluated. In vivo magnetic resonance (MR) imaging was acquired on PND100 to assess structural changes in gray and white matter, and brain metabolite concentrations. Additionally, inflammation and oxidative stress (IOS) markers were measured ex vivo in selected brain regions. MIS offspring showed behavioral, neuroanatomical, and biochemical alterations. Interestingly, NAC treatment during periadolescence prevented PPI deficits and partially counteracted some biochemical imbalances. Moreover, NAC treatments during pregnancy not only replicated the beneficial outcomes reported by the treatment in periadolescence, but also prevented some neuroanatomical deficits, including reductions in hippocampal and corpus callosum volumes. This study suggests that early reduction of inflammation and prooxidation could help prevent the onset of schizophrenia-like symptoms, supporting the importance of anti-IOS compounds in ameliorating this disorder.


Subject(s)
Acetylcysteine , Schizophrenia , Female , Pregnancy , Rats , Animals , Rats, Wistar , Acetylcysteine/pharmacology , Schizophrenia/drug therapy , Schizophrenia/prevention & control , Poly I-C , Inflammation
2.
Int J Mol Sci ; 23(16)2022 Aug 20.
Article in English | MEDLINE | ID: mdl-36012679

ABSTRACT

Symptoms of schizophrenia (SZ) typically emerge during adolescence to young adulthood, which gives a window before full-blown psychosis for early intervention. Strategies for preventing the conversion from the prodromal phase to the psychotic phase are warranted. Heterozygous (Het) Disc1 mutant mice are considered a prodromal model of SZ, suitable for studying psychotic conversion. We evaluated the preventive effect of chronic N-acetylcysteine (NAC) administration, covering the prenatal era to adulthood, on the reaction following the Amph challenge, which mimics the outbreak or conversion of psychosis, in adult Het Disc1 mice. Biochemical and morphological features were examined in the striatum of NAC-treated mice. Chronic NAC treatment normalized the Amph-induced activity in the Het Disc1 mice. Furthermore, the striatal phenotypes of Het Disc1 mice were rescued by NAC including dopamine receptors, the expression of GSK3s, MSN dendritic impairments, and striatal PV density. The current study demonstrated a potent preventive effect of chronic NAC treatment in Disc1 Het mice on the acute Amph test, which mimics the outbreak of psychosis. Our findings not only support the benefit of NAC as a dietary supplement for SZ prodromes, but also advance our knowledge of striatal dopamine receptors, PV neurons, and GSK3 signaling pathways as therapeutic targets for treating or preventing the pathogenesis of mental disorders.


Subject(s)
Amphetamine , Schizophrenia , Acetylcysteine/pharmacology , Amphetamine/pharmacology , Animals , Disease Models, Animal , Dopamine/metabolism , Female , Glycogen Synthase Kinase 3 , Humans , Mice , Nerve Tissue Proteins , Pregnancy , Receptors, Dopamine , Schizophrenia/drug therapy , Schizophrenia/genetics , Schizophrenia/prevention & control
3.
J Ethnopharmacol ; 295: 115432, 2022 Sep 15.
Article in English | MEDLINE | ID: mdl-35659625

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Carpolobia lutea decoction is widely used as a phytotherapeutic against central nervous system-related disorders including insomnia, migraine headache, and mental illness in West and Central Tropical Africa. AIM: This study was designed to investigate the antipsychotic activity of Carpolobia lutea (EECL) in mice models of psychosis. METHODS: Male Swiss mice (n = 5/group) were given EECL (100, 200, 400, and 800 mg/kg), haloperidol (1 mg/kg), clozapine (5 mg/kg) and vehicle (10 mL/kg) orally before amphetamine (5 mg/kg)-induced hyperlocomotion and stereotypy, apomorphine (2 mg/kg)-induced stereotypy, or ketamine (10, 30, and 100 mg/kg)-induced hyperlocomotion, enhancement of immobility and cognitive impairment. RESULTS: EECL (200, 400, and 800 mg/kg) prevented amphetamine- and apomorphine-induced stereotypies, as well as reduced hyperlocomotion induced by amphetamine and ketamine, all of which are predictors of positive symptoms. Regardless of the dose administered, EECL prevented the index of negative symptoms induced by ketamine. Furthermore, higher doses of EECL (400 and 800 mg/kg) also prevented ketamine-induced cognitive impairment, a behavioral phenotype of cognitive symptoms. CONCLUSION: Pretreatment with EECL demonstrated antipsychotic activity in mice, preventing amphetamine-, apomorphine-, and ketamine-induced schizophrenia-like symptoms, with 800 mg/kg being the most effective dose.


Subject(s)
Antipsychotic Agents , Ketamine , Psychotic Disorders , Schizophrenia , Amphetamine , Animals , Antipsychotic Agents/pharmacology , Apomorphine/pharmacology , Ethanol/therapeutic use , Ketamine/pharmacology , Male , Mice , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Psychotic Disorders/drug therapy , Psychotic Disorders/prevention & control , Schizophrenia/chemically induced , Schizophrenia/drug therapy , Schizophrenia/prevention & control
4.
Psychopharmacology (Berl) ; 239(7): 2263-2275, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35294602

ABSTRACT

RATIONALE: At present, the research on the prevention of schizophrenia is still in its infancy. Pyrroloquinoline quinone (PQQ) has potential to treat psychological and neurological diseases including schizophrenia. However, the preventive effect of PQQ on schizophrenia remains unclear. OBJECTIVES: In this study, we aimed to examine the preventive effect of supplementation of dietary PQQ from pregnancy or after birth on dizocilpine (MK-801)-induced schizophrenia-like behaviors in mice. RESULTS: Supplementation of dietary PQQ from pregnancy could effectively prevent MK-801-induced weight gain decrease, hyperlocomotion, stereotypical behavior, ataxia, exploratory activity decrease, social interaction disorder, memory deficit, and depression in mice. Supplementation of dietary PQQ after birth could effectively prevent MK-801-induced weight gain decrease, stereotypical behavior, ataxia, and memory deficit in mice. Female mice responded to a greater degree than males in preventing MK-801-induced weight gain decrease in both forms of PQQ supplementation. For mice that began PQQ supplementation after birth, females performed better than males in preventing MK-801-induced ataxia, memory deficit, and depression. For mice that began PQQ supplementation from pregnancy, males performed better than females in preventing MK-801-induced memory deficit. In vitro experiments indicated that PQQ supplementation in the earlier stage of life contributed to the growth of neurons and the development of neurites. CONCLUSIONS: Our current study suggested that PQQ supplementation from pregnancy or postpartum could prevent some schizophrenia-like behaviors induced by MK-801 in mice. Our work supported the potential usage of dietary supplement of PQQ in preventing or alleviating symptoms associated with schizophrenia.


Subject(s)
Dizocilpine Maleate , Schizophrenia , Animals , Ataxia , Dietary Supplements , Dizocilpine Maleate/pharmacology , Female , Male , Memory Disorders , Mice , PQQ Cofactor/pharmacology , Pregnancy , Schizophrenia/chemically induced , Schizophrenia/prevention & control , Weight Gain
5.
Pharmacol Res ; 174: 105938, 2021 12.
Article in English | MEDLINE | ID: mdl-34655773

ABSTRACT

The recent shift in socio-political debates and growing liberalization of Cannabis use across the globe has raised concern regarding its impact on vulnerable populations such as adolescents. Concurrent with declining perception of Cannabis harms, more adolescents are using it daily in several countries and consuming marijuana strains with high content of psychotropic delta (9)-tetrahydrocannabinol (THC). These dual, related trends seem to facilitate the development of compromised social and cognitive performance at adulthood, which are described in preclinical and human studies. Cannabis exerts its effects via altering signalling within the endocannabinoid system (ECS), which modulates the stress circuitry during the neurodevelopment. In this context early interventions appear to circumvent the emergence of adult neurodevelopmental deficits. Accordingly, Cannabis sativa second-most abundant compound, cannabidiol (CBD), emerges as a potential therapeutic agent to treat neuropsychiatric disorders. We first focus on human and preclinical studies on the long-term effects induced by adolescent THC exposure as a "critical window" of enhanced neurophysiological vulnerability, which could be involved in the pathophysiology of schizophrenia and related primary psychotic disorders. Then, we focus on adolescence as a "window of opportunity" for early pharmacological treatment, as novel risk reduction strategy for neurodevelopmental disorders. Thus, we review current preclinical and clinical evidence regarding the efficacy of CBD in terms of positive, negative and cognitive symptoms treatment, safety profile, and molecular targets.


Subject(s)
Cannabinoids , Phytochemicals , Psychoses, Substance-Induced , Schizophrenia , Adolescent , Animals , Cannabinoids/adverse effects , Cannabinoids/pharmacology , Cannabinoids/therapeutic use , Humans , Phytochemicals/adverse effects , Phytochemicals/pharmacology , Phytochemicals/therapeutic use , Psychoses, Substance-Induced/drug therapy , Psychoses, Substance-Induced/metabolism , Schizophrenia/chemically induced , Schizophrenia/drug therapy , Schizophrenia/metabolism , Schizophrenia/prevention & control
6.
Int J Mol Sci ; 22(13)2021 Jun 26.
Article in English | MEDLINE | ID: mdl-34206945

ABSTRACT

Schizophrenia typically emerges during adolescence, with progression from an ultra-high risk state (UHR) to the first episode of psychosis (FEP) followed by a chronic phase. The detailed pathophysiology of schizophrenia and the factors leading to progression across these stages remain relatively unknown. The current treatment relies on antipsychotics, which are effective for FEP and chronic schizophrenia but ineffective for UHR patients. Antipsychotics modulate dopaminergic and glutamatergic neurotransmission, inflammation, oxidative stress, and membrane lipids pathways. Many of these biological pathways intercommunicate and play a role in schizophrenia pathophysiology. In this context, research of preventive treatment in early stages has explored the antipsychotic effects of omega-3 supplementation in UHR and FEP patients. This review summarizes the action of omega-3 in various biological systems involved in schizophrenia. Similar to antipsychotics, omega-3 supplementation reduces inflammation and oxidative stress, improves myelination, modifies the properties of cell membranes, and influences dopamine and glutamate pathways. Omega-3 supplementation also modulates one-carbon metabolism, the endocannabinoid system, and appears to present neuroprotective properties. Omega-3 has little side effects compared to antipsychotics and may be safely prescribed for UHR patients and as an add-on for FEP patients. This could to lead to more efficacious individualised treatments, thus contributing to precision medicine in psychiatry.


Subject(s)
Antipsychotic Agents/pharmacokinetics , Fatty Acids, Omega-3/metabolism , Schizophrenia/metabolism , Animals , Antipsychotic Agents/therapeutic use , Folic Acid/metabolism , Humans , Methionine/metabolism , Myelin Sheath/metabolism , Schizophrenia/drug therapy , Schizophrenia/prevention & control , Synaptic Transmission
7.
Life Sci ; 277: 119417, 2021 Jul 15.
Article in English | MEDLINE | ID: mdl-33794248

ABSTRACT

AIMS: Schizophrenia (SZ) is recognized as a neuropsychiatric disorder in humans with accelerated mortality and profound morbidity followed with impairments in social as well as vocational functioning. Though various antipsychotics are being considered as approved treatment therapy for the psychotic symptoms of SZ but they also exert adverse effects and also lack efficacy in treating full spectrum of the disorder. Spirulina platensis (blue-green algae), a nutritional supplement, constitutes a variety of multi-nutrients and possesses a large number of neuroprotective activities. Therefore, present experimental work was designed to evaluate the neuroprotective effects of spirulina in ameliorating the psychosis-like symptoms in dizocilpine-induced rat model of SZ. MATERIALS AND METHODS: The spirulina was tested as preventive and therapeutic regimen at the dose of 180 mg/kg. After pre- and post-treatment with spirulina, rats were subjected to behavioral assessments followed by biochemical and neurochemical estimations. Biomarkers including APO-E, RTN-4, TNF-α, and IL-6 were also estimated using ELISA. KEY FINDINGS: Present results showed that administration of spirulina not only improved behavioral deficits induced by dizocilpine but it also regulates neurotransmission, oligodendrocyte dysfunction and APO-E over expression. Moreover, it also restores the immune response dysfunction by reducing inflammatory cytokines. SIGNIFICANCE: Thus, from present findings it may be suggested that spirulina aids in ameliorating the psychosis-like symptoms induced by dizocilpine in animal model possibly via regulation of neurotransmission and other biomarkers that are extensively used to uncover the etiopathology of SZ. Hence, blue-green algae can be used as an effective therapy for preventive or therapeutic measures in SZ.


Subject(s)
Apolipoproteins E/metabolism , Gene Expression Regulation/drug effects , Neuroprotective Agents/pharmacology , Nogo Proteins/metabolism , Prefrontal Cortex/drug effects , Schizophrenia/prevention & control , Spirulina/physiology , Animals , Apolipoproteins E/genetics , Behavior, Animal/drug effects , Dietary Supplements , Disease Models, Animal , Male , Nogo Proteins/genetics , Oxidative Stress , Prefrontal Cortex/metabolism , Rats , Rats, Wistar , Schizophrenia/genetics , Schizophrenia/metabolism , Schizophrenia/pathology
8.
Int J Dev Neurosci ; 81(5): 461-467, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33786893

ABSTRACT

INTRODUCTION: Schizophrenia is considered one of the most disabling and severe human diseases worldwide. The etiology of schizophrenia is thought to be multifactorial and evidence suggests that DNA methylation can play an important role in underlying pivotal neurobiological alterations of this disorder. Some studies have demonstrated the effects of dietary supplementation as an alternative approach to the prevention of schizophrenia, including folic acid. However, no study has ever investigated the role of such supplementation in altering the DNA methylation system in the context of schizophrenia. OBJECTIVES: The present study aims to investigate the effects of maternal folic acid supplementation at different doses on nuclear methyltransferase activity of adult rat offspring subjected to an animal model schizophrenia induced by ketamine. METHODS: Adult female Wistar rats, (60 days old) received folic acid-deficient diet, control diet, or control diet plus folic acid supplementation (at 5, 10, or 50 mg/kg) during pregnancy and lactation. After reaching adulthood (60 days), the male offspring of these dams were subjected to the animal model of schizophrenia induced by 7 days of ketamine intraperitoneal injection (25 mg/kg). After the 7-day protocol, the activity of nuclear methyltransferase was evaluated in the brains of the offspring. RESULTS: Maternal folic acid supplementation at 50 mg/kg increased methyltransferase activity in the frontal cortex, while 10 mg/kg increased methyltransferase activity in the hippocampus. In the striatum of offspring treated with ketamine, maternal deficient diet, control diet, and folic acid supplementation at 5 mg/kg decreased methyltransferase activity compared to the control group. The folic acid supplementation at 10 and 50 mg/kg reversed this ketamine effect. CONCLUSIONS: Maternal FA deficiency could be related to schizophrenia pathophysiology, while FA supplementation could present a protective effect since it demonstrated persistent effects in epigenetic parameters in adult offspring.


Subject(s)
Cell Nucleus/enzymology , Folic Acid/therapeutic use , Methyltransferases/metabolism , Schizophrenia/prevention & control , Animals , Cell Nucleus/drug effects , DNA Methylation/drug effects , Diet , Dietary Supplements , Female , Folic Acid Deficiency/complications , Ketamine , Male , Pregnancy , Rats , Rats, Wistar , Schizophrenia/chemically induced , Schizophrenia/enzymology , Schizophrenic Psychology
9.
Eur Neuropsychopharmacol ; 46: 14-27, 2021 05.
Article in English | MEDLINE | ID: mdl-33735708

ABSTRACT

The likely involvement of inflammation and oxidative stress (IOS) in mental disease has led to advocate anti-oxidant and anti-inflammatory drugs as therapeutic strategies in the treatment of schizophrenia. Since omega-3 fatty acids (ω-3) show anti-inflammatory/neuroprotective properties, we aim to evaluate whether ω-3 treatment during adolescence in the maternal immune stimulation (MIS) animal model of schizophrenia could prevent the brain and behavioural deficits described in adulthood. At gestational day 15, PolyI:C (4 mg/kg) or saline (VH) were injected to pregnant Wistar rats. Male offspring received ω-3 (800 mg/kg) or saline (Sal) daily from postnatal day (PND) 35-49, defining 4 groups: MIS-ω-3; MIS-Sal; VH-ω-3 and VH-Sal. At PND70, rats were submitted to prepulse inhibition test (PPI). FDG-PET and T2-weighted MRI brain studies were performed in adulthood and analyzed by means of SPM12. IOS markers were measured in selected brain areas. MIS-offspring showed a PPI deficit compared with VH-offspring and ω-3 treatment prevented this deficit. Also, ω-3 reduced the brain metabolism in the deep mesencephalic area and prevented the volumetric abnormalities in the hippocampus but not in the ventricles in MIS-offspring. Besides, ω-3 reduced the expression of iNOS and Keap1 and increased the activity/concentration of HO1, NQO1 and GPX. Our study demonstrates that administration of ω-3 during adolescence prevents PPI behavioural deficits and hippocampal volumetric abnormalities, and partially counteracts IOS deficits via iNOS and Nrf2-ARE pathways in the MIS model. This study highlights the need for novel strategies based on anti-inflammatory/anti-oxidant compounds to alter the disease course in high-risk populations at early stages.


Subject(s)
Fatty Acids, Omega-3 , Prenatal Exposure Delayed Effects , Schizophrenia , Virus Diseases , Animals , Anti-Inflammatory Agents/therapeutic use , Antioxidants , Disease Models, Animal , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/therapeutic use , Female , Kelch-Like ECH-Associated Protein 1 , Male , NF-E2-Related Factor 2/therapeutic use , Poly I-C , Pregnancy , Prenatal Exposure Delayed Effects/prevention & control , Rats , Rats, Wistar , Schizophrenia/diagnostic imaging , Schizophrenia/drug therapy , Schizophrenia/prevention & control , Virus Diseases/drug therapy
10.
Eur J Pharmacol ; 897: 173949, 2021 Apr 15.
Article in English | MEDLINE | ID: mdl-33607108

ABSTRACT

Schizophrenia is a devastating neurodevelopmental disorder. The animal model based on perinatal immune activation, as first-hit, combined with peripubertal stress, as a second hit, has gained evidence in recent years. Omega-3 polyunsaturated fatty acids (n3-PUFAs) is being a promise for schizophrenia prevention. Nevertheless, the influence of sex in schizophrenia neurobiology and prevention has been neglected. Thus, the present study evaluates the preventive effects of n3-PUFAs in both sexes' mice submitted to the two-hit model and the participation of oxidative changes in this mechanism. The two-hit consisted of polyI:C administration from postnatal days (PNs) 5-7, and unpredictable stress from PNs35-43. n3-PUFAs were administered from PNs30-60. Prepulse inhibition of the startle reflex (PPI), social interaction, and Y-maze tests were conducted between PNs70-72 to evaluate positive-, negative-, and cognitive-like schizophrenia symptoms. We assessed brain oxidative changes in brain areas and plasma. Both sexes' two-hit mice presented deficits in PPI, social interaction, and working memory that were prevented by n3-PUFAs. In two-hit females, n3-PUFAs prevented increments in nitrite levels in the prefrontal cortex (PFC), hippocampus, striatum, and plasma TBARS levels. In two-hit males, n3-PUFAs prevented the increase in TBARS in the PFC, hippocampus, and striatum. Notably, male mice that received only n3-PUFAs without hit exposure presented impairments in working memory and social interaction. These results add further preclinical evidence for n3-PUFAs as an accessible and effective alternative in preventing behavioral and oxidative changes related to schizophrenia but call attention to the need for precaution in this indication due to hit- and sex-sensitive issues.


Subject(s)
Antioxidants/pharmacology , Behavior, Animal/drug effects , Brain/drug effects , Fatty Acids, Omega-3/pharmacology , Oxidative Stress/drug effects , Schizophrenia/prevention & control , Schizophrenic Psychology , Age Factors , Animals , Brain/metabolism , Brain/physiopathology , Dietary Supplements , Disease Models, Animal , Female , Male , Maze Learning/drug effects , Mice , Poly I-C , Prepulse Inhibition/drug effects , Reflex, Startle/drug effects , Schizophrenia/etiology , Schizophrenia/metabolism , Schizophrenia/physiopathology , Sex Factors , Sexual Development , Social Behavior , Stress, Psychological/complications
11.
Int J Dev Neurosci ; 81(1): 26-36, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32780510

ABSTRACT

Schizophrenia is a chronic neuropsychiatric disorder with a poorly understood pathophysiology. The theories about the disorder are mainly about dysregulation in one or more systems of neurotransmitters, and the progression triggers the presence of inflammatory markers indicates the possibility that the disorder is initially an inflammatory disease. The objective was to evaluate the ascorbic acid supplementation in an animal model of schizophrenia, on behavioral parameters, and cytokines involved in inflammation IL-1ß, IL-10. Wistar rats with 60 days of age were used which were supplemented with ascorbic acid at 0.1, 1, and 10 mg/kg or saline for 14 days via orogastric gavage. Subsequently, four groups were given ketamine (25 mg/kg) and four groups received intraperitoneal saline from the 9th-15th day of the experiment. After 30 min of the last administration of ketamine/saline, and behavioral test, rats were killed by guillotine decapitation and the brain structures were carefully dissected for biochemical analysis. Results showed that ascorbic acid supplementation prevented motor sensory loss but nor alter other parameters evaluated. We concluded that ascorbic acid may be used as a therapeutic adjuvant in schizophrenia and may help to improve the schizophrenic patient's life quality.


Subject(s)
Anesthetics, Dissociative , Ascorbic Acid/therapeutic use , Dietary Supplements , Ketamine , Schizophrenia/chemically induced , Schizophrenia/prevention & control , Vitamins/therapeutic use , Animals , Behavior, Animal/drug effects , Brain/pathology , Cytokines , Dose-Response Relationship, Drug , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Male , Rats , Rats, Wistar , Schizophrenia/pathology , Schizophrenic Psychology
13.
BMJ Open ; 10(4): e033711, 2020 04 29.
Article in English | MEDLINE | ID: mdl-32354777

ABSTRACT

OBJECTIVES: To evaluate whether a newly developed care pathway, Treatment and Recovery In PsycHosis (TRIumPH), is feasible, acceptable and effective in meeting National Institute of Health and Care Excellence (NICE) quality standards in a timely manner. METHODS: This is a pragmatic, non-randomised, prospective, mixed methods study comparing an implementation (TRIumPH) and comparator site (not implementing TRIumPH) across three cohorts to assess feasibility, acceptability and effectiveness of the integrated pathway. SETTING: Early intervention in psychosis (EIP) services at two National Health Service organisations in South of England. PARTICIPANTS: All patients accepted into EIP services between 1 June 2014 and 31 May 2017 were each followed up for 1 year within their respective cohorts. METHODOLOGY: Quantitative data consisted of routinely collected clinical data retrieved from patient records to assess whether the implementation of TRIumPH achieved better concordance to NICE standards. These included time to access services, physical health assessments, clinical outcomes based timeliness of delivery and acute data. The controlled trial has evaluated the effect of TRIumPH (Intervention) with Care As Usual (Comparator). Qualitative measures consisted of questionnaires, interviews and focus groups to assess acceptability and satisfaction. Outcome measures were compared within the baseline, year 1 and year 2 cohorts and between the two sites. Quantitative data were statistically analysed by comparing means and proportions. RESULTS: Time to assessment improved in the implementation site and remained within the target in comparator site. Meeting of quality standards increased substantially in the implementation site but was more variable and reached lower levels in the comparator site especially for physical health standards. Cognitive therapy for psychosis, family intervention and carer and employment support were all offered to a greater extent in the implementation site and uptake increased over the period. CONCLUSIONS: Pathway implementation generally led to greater improvements in achievement of access and quality standards compared with comparator site. TRIAL REGISTRATION NUMBER: UK Clinical Research Network Portfolio (19187).


Subject(s)
Early Medical Intervention , Psychotic Disorders/therapy , Schizophrenia/therapy , Time-to-Treatment , Adult , Delivery of Health Care, Integrated , England , Feasibility Studies , Female , Focus Groups , Humans , Length of Stay , Male , Outcome Assessment, Health Care , Patient Discharge , Prospective Studies , Psychotic Disorders/prevention & control , Psychotic Disorders/rehabilitation , Recovery of Function , Schizophrenia/prevention & control , Schizophrenia/rehabilitation , Secondary Prevention/methods , Treatment Outcome , Young Adult
14.
Neuropsychobiology ; 79(1): 20-42, 2020.
Article in English | MEDLINE | ID: mdl-30359969

ABSTRACT

INTRODUCTION: Schizophrenia spectrum disorders (SSD) represent a cluster of severe mental illnesses. Diet has been identified as a modifiable risk factor and opportunity for intervention in many physical illnesses and more recently in mental illnesses such as unipolar depression; however, no dietary guidelines exist for patients with SSD. OBJECTIVE: This review sought to systematically scope the existing literature in order to identify nutritional interventions for the prevention or treatment of mental health symptoms in SSD as well as gaps and opportunities for further research. METHODS: This review followed established methodological approaches for scoping reviews including an extensive a priori search strategy and duplicate screening. Because of the large volume of results, an online program (Abstrackr) was used for screening and tagging. Data were extracted based on the dietary constituents and analyzed. RESULTS: Of 55,330 results identified by the search, 822 studies met the criteria for inclusion. Observational evidence shows a connection between the presence of psychotic disorders and poorer quality dietary patterns, higher intake of refined carbohydrates and total fat, and lower intake or levels of fibre, ω-3 and ω-6 fatty acids, vegetables, fruit, and certain vitamins and minerals (vitamin B12 and B6, folate, vitamin C, zinc, and selenium). Evidence illustrates a role of food allergy and sensitivity as well as microbiome composition and specific phytonutrients (such as L-theanine, sulforaphane, and resveratrol). Experimental studies have demonstrated benefit using healthy diet patterns and specific vitamins and minerals (vitamin B12 and B6, folate, and zinc) and amino acids (serine, lysine, glycine, and tryptophan). DISCUSSION: Overall, these findings were consistent with many other bodies of knowledge about healthy dietary patterns. Many limitations exist related to the design of the individual studies and the ability to extrapolate the results of studies using dietary supplements to dietary interventions (food). Dietary recommendations are presented as well as recommendations for further research including more prospective observational studies and intervention studies that modify diet constituents or entire dietary patterns with statistical power to detect mental health outcomes.


Subject(s)
Diet , Nutritional Physiological Phenomena , Psychotic Disorders , Schizophrenia , Humans , Psychotic Disorders/diet therapy , Psychotic Disorders/etiology , Psychotic Disorders/prevention & control , Schizophrenia/diet therapy , Schizophrenia/etiology , Schizophrenia/prevention & control
15.
J Pharm Pharmacol ; 72(1): 149-160, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31713882

ABSTRACT

OBJECTIVES: A botanical drug derived from the ethanolic extract composed of Clematis chinensis Osbeck (Ranunculaceae), Trichosanthes kirilowii Maximowicz (Cucurbitaceae) and Prunella vulgaris Linné (Lamiaceae) has been used to ameliorate rheumatoid arthritis as an ethical drug in Korea. In our study, we investigated the effect of this herbal complex extract (HCE) on schizophrenia-like behaviours induced by MK-801. METHODS: HCE (30, 100 or 300 mg/kg, p.o) was orally administered to male ICR mice to a schizophrenia-like animal model induced by MK-801. We conducted an acoustic startle response task, an open-field task, a novel object recognition task and a social novelty preference task. KEY FINDINGS: We found that a single administration of HCE (100 or 300 mg/kg) ameliorated MK-801-induced abnormal behaviours including sensorimotor gating deficits and social or object recognition memory deficits. In addition, MK-801-induced increases in phosphorylated Akt and GSK-3ß expression levels in the prefrontal cortex were reversed by HCE (30, 100 or 300 mg/kg). CONCLUSIONS: These results imply that HCE ameliorates MK-801-induced dysfunctions in prepulse inhibition, social interactions and cognitive function, partly by regulating the Akt and GSK-3ß signalling pathways.


Subject(s)
Antipsychotic Agents/pharmacology , Behavior, Animal/drug effects , Cognition/drug effects , Cognitive Dysfunction/prevention & control , Gait Disorders, Neurologic/prevention & control , Plant Extracts/pharmacology , Prefrontal Cortex/drug effects , Schizophrenia/prevention & control , Sensory Gating/drug effects , Animals , Clematis , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/physiopathology , Cognitive Dysfunction/psychology , Disease Models, Animal , Dizocilpine Maleate , Gait Disorders, Neurologic/chemically induced , Gait Disorders, Neurologic/physiopathology , Gait Disorders, Neurologic/psychology , Glycogen Synthase Kinase 3 beta/metabolism , Locomotion/drug effects , Male , Mice, Inbred ICR , Phosphorylation , Prefrontal Cortex/metabolism , Prefrontal Cortex/physiopathology , Proto-Oncogene Proteins c-akt/metabolism , Prunella , Recognition, Psychology/drug effects , Reflex, Startle/drug effects , Schizophrenia/chemically induced , Schizophrenic Psychology , Social Behavior , Trichosanthes
16.
J Abnorm Psychol ; 128(8): 840-854, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31343181

ABSTRACT

The stress-vulnerability-protective factors model is often used to explain the etiology and known risk and protective factors of initial psychotic symptoms and symptomatic relapses. Over the past 40 years since its initial conception, the model has evolved and gathered a plethora of evidence of varying quality for its different components. The objective of this metareview is to analyze the quality of the evidence and the effect sizes for each component of the model not previously reviewed. Recent meta-analyses covering each component of the model in relation to the onset of psychotic symptoms or symptomatic relapse in schizophrenia were reviewed with the grading of recommendations, assessment, development, and evaluation system. Thirty-one meta-analyses were kept, from 3,044 papers reviewed. We did not add to previous metareviews in terms of obstetric/prenatal or genetic vulnerabilities. For stressors, moderate to strong research evidence was found for childhood adversity, cannabis, methamphetamine abuse, and expressed emotions as triggers of psychotic relapse or as linked to the onset of psychotic symptoms. For protective factors, moderate to strong evidence was found for antipsychotic medication in adults, family interventions, social skills training, as well as interventions focusing on recovery management skills. Poor evidence or no evidence (i.e., absence of meta-analyses) were found for the other components of the model. More rigorous studies and systematic reviews are needed in order to validate the various components of the model in regard to symptom onset and relapse. (PsycINFO Database Record (c) 2019 APA, all rights reserved).


Subject(s)
Antipsychotic Agents/therapeutic use , Psychotherapy/methods , Schizophrenia/therapy , Schizophrenic Psychology , Secondary Prevention/methods , Adult , Child , Female , Humans , Recurrence , Schizophrenia/drug therapy , Schizophrenia/prevention & control
17.
Mol Med Rep ; 18(4): 3834-3842, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30132543

ABSTRACT

To understand maternal immune activation (MIA) during prenatal development, the synthetic double­stranded RNA polyriboinosinic­polyribocytidylic acid [poly(I:C)] has been widely used in animal models to induce behavioral deficits similar to those in schizophrenia and other psychotic disorders. Panax ginseng C.A. Meyer (PG) extract is widely used to treat various kinds of nervous system disorders in Asia particularly China and Korea. The present study aimed to examine the effects of PG extract on MIA offspring using behavioral activity tests and protein expression analyses. Pregnant mice were exposed to poly(I:C) (5 mg/kg) or vehicle treatment on gestation day 9, and the resulting MIA offspring were subjected to vehicle or PG (300 mg/kg) treatment. In the acoustic startle response test, MIA­induced sensorimotor gating deficit was ameliorated by PG. The majority of behavioral parameters measured in the social interaction (non­aggressive or/and aggressive pattern), open field (number/duration of behavior) and forced swimming test (immobility behavior) were significantly altered in the MIA offspring. Western blot and immunohistochemical analyses of the medial prefrontal cortex indicated that the expression levels of certain neurodevelopmental proteins, including dihydropyrimidinase­related 2, LIM and SH3 domain 1, neurofilament medium, and discs large homolog 4, were decreased in the untreated MIA offspring, whereas PG treatment improved behavioral impairments and increased neurodevelopmental protein expression in MIA offspring. These results suggested that PG may be useful in neurodevelopmental disorder therapy, including psychiatric disorders such as schizophrenia, owing to its antipsychotic effects.


Subject(s)
Antipsychotic Agents/therapeutic use , Panax , Plant Extracts/therapeutic use , Prenatal Exposure Delayed Effects/etiology , Prenatal Exposure Delayed Effects/prevention & control , Schizophrenia/etiology , Schizophrenia/prevention & control , Animals , Antipsychotic Agents/chemistry , Behavior, Animal/drug effects , Disease Models, Animal , Female , Mice , Mice, Inbred C57BL , Panax/chemistry , Plant Extracts/chemistry , Poly I-C , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/immunology , Schizophrenia/chemically induced , Schizophrenia/immunology
18.
Schizophr Res ; 202: 297-302, 2018 12.
Article in English | MEDLINE | ID: mdl-29925476

ABSTRACT

OBJECTIVES: Studies have suggested a possible autoimmune contribution in a subset of patients with schizophrenia. The purpose of this study was to determine if a history of autoimmune diseases (AD) is associated with an increased risk of later onset of schizophrenia. METHODS: Taiwan's National Health Insurance Research Database was used to identify a total of 64,817 AD patients and an equal number of age-matched control patients. The incidence rates of schizophrenia with a maximum follow-up period of 10 years between patients with and without AD were compared using a Cox proportional hazard model to estimate the hazard ratio (HR) and 95% confidence interval (95% CI). RESULTS: The main finding was the discovery of a higher incidence of subsequent schizophrenia in patients with AD (HR: 1.72, 95% CI: 1.23-2.4) after adjustment for other demographic characteristics. Specifically, the risk of schizophrenia was observed to be a significant increase in systemic lupus erythematosus (3.73, 2.07-6.72), rheumatoid arthritis (2.89, 1.97-4.23), dermatomyositis (5.85, 1.32-25.94) and autoimmune vasculitis (2.44, 1.17-5.06). Also, this study revealed some potential risk factors for developing schizophrenia, including younger age (less than or equal to 50 years) and some comorbidities (hypertension, chronic obstructive pulmonary disease, and alcohol use disorder). Conversely, this study found that steroid use was a potential protective factor for the development of schizophrenia. CONCLUSIONS: This study found that AD were associated with an increased risk of developing schizophrenia, suggesting that the abnormal autoimmune process was associated with an increase in the expression of psychiatric disturbances.


Subject(s)
Autoimmune Diseases/epidemiology , Registries/statistics & numerical data , Schizophrenia/epidemiology , Adult , Age Factors , Autoimmune Diseases/drug therapy , Comorbidity , Dermatomyositis/epidemiology , Female , Follow-Up Studies , Humans , Incidence , Lupus Erythematosus, Systemic/epidemiology , Male , Middle Aged , National Health Programs , Proportional Hazards Models , Rheumatic Fever/epidemiology , Risk , Schizophrenia/prevention & control , Steroids/therapeutic use , Taiwan/epidemiology , Vasculitis/epidemiology , Young Adult
19.
Psychiatriki ; 29(1): 64-73, 2018.
Article in English | MEDLINE | ID: mdl-29754122

ABSTRACT

Schizophrenia is associated with increased risk for type 2 diabetes mellitus, resulting in elevated cardiovascular risk and limited life expectancy, translated into a weighted average of 14.5 years of potential life lost and an overall weighted average life expectancy of 64.7 years. The exact prevalence of type 2 diabetes among people with schizophrenia varies across studies and ranges 2-5fold higher than in the general population, whereas the aetiology is complex and multifactorial. Besides common diabetogenic factors, applied similarly in the general population, such as obesity, hyperlipidemia, smoking, hypertension, poor diet and limited physical activity, the co-occurrence of schizophrenia and diabetes is also attributed to unique conditions. Specifically, excessive sedentary lifestyle, social determinants, adverse effects of antipsychotic drugs and limited access to medical care are considered aggravating factors for diabetes onset and low quality of diabetes management. Schizophrenia itself is further proposed as causal factor for diabetes, given the observed higher prevalence of diabetes in young patients, newly diagnosed with schizophrenia and unexposed to antipsychotics. Furthermore, studies support genetic predisposition to diabetes among people with schizophrenia, suggesting shared genetic risk and disclosing a number of overlapped risk loci. Therefore, special attention should be paid in preventing diabetes in people with schizophrenia, through intervention in all possible modifiable risk factors. Implementation of careful antipsychotic prescription, provision of adequate motivation for balanced diet and physical activity and facilitating access to primary health care, could serve in reducing diabetes prevalence. On the other hand, increasing calls are made for early diagnosis of diabetes, application of the appropriate anti-diabetic therapy and strict inspection of therapy adherence, to limit the excess mortality due to cardiovascular events in people with schizophrenia. Moreover, population health programs could help counseling and preventing diabetes risk, additionally to early screening and diagnosis set, aiming to reduce disparities in populations. Finally, mental health-care providers might greatly promote offered health services to patients with schizophrenia, through a holistic individualized approach, considering additionally the physical health of the patients and working closely, preventively and therapeutically, in collaboration with the physicians and diabetologists.


Subject(s)
Diabetes Mellitus, Type 2/epidemiology , Schizophrenia/epidemiology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/prevention & control , Diabetes Mellitus, Type 2/therapy , Humans , Obesity , Prevalence , Risk , Risk Factors , Schizophrenia/complications , Schizophrenia/prevention & control , Schizophrenia/therapy
20.
Trials ; 18(1): 524, 2017 Nov 09.
Article in English | MEDLINE | ID: mdl-29121974

ABSTRACT

BACKGROUND: The at-risk mental state (ARMS) describes individuals at high risk of developing schizophrenia or psychosis. The use of antipsychotics in this population is not supported, because most individuals with ARMS are unlikely to develop psychosis. Anti-inflammatory treatments and polyunsaturated fatty acids (PUFAs) may have some beneficial effects in the treatment of ARMS. There have been no controlled clinical trials in which researchers have investigated the use of minocycline for ARMS and no trials involving PUFAs in combination with other proposed treatments. There is a need to find effective, tolerable and inexpensive interventions for individuals with ARMS that are available in high-, low- and middle-income countries. METHODS/DESIGN: A 6-month intervention study of minocycline and/or omega-3 fatty acids added to treatment as usual (TAU) in patients with ARMS will be conducted in Pakistan using a randomised, placebo-controlled, double-blind factorial design. A total of 320 consenting patients with capacity will be recruited from the community, general practitioner clinics and psychiatric units. Allowing for a 25% dropout rate, we will recruit 59 completing participants into each study arm, and in total 236 will complete the study. We will determine whether the addition of minocycline and/or omega-3 fatty acids to TAU attenuates the rate of transition from ARMS to first-episode psychosis and improves symptoms and/or level of functioning in ARMS. We will also investigate whether any candidate risk factors, such as negative symptoms, influence treatment response in the ARMS group. The primary efficacy endpoint is conversion to psychotic disorder at 12 months after study entry. Analysis will be done according to the intention to treat principle using analysis of variance, chi-square tests and adjusted ORs to assess between-group differences. Cox regression analysis will be used to evaluate potential between-group differences in time to onset of psychosis. DISCUSSION: The outcomes of this trial will provide evidence of the potential benefits of minocycline and PUFAs in the treatment of ARMS. Both minocycline and PUFAs are inexpensive, are readily available in low-/middle-income countries such as Pakistan, and if proven, may be safe and effective for treating individuals with ARMS. TRIAL REGISTRATION: ClinicalTrials.gov, NCT02569307 . Registered on 3 October 2015.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Dietary Supplements , Fatty Acids, Omega-3/therapeutic use , Mental Health , Minocycline/therapeutic use , Psychotic Disorders/prevention & control , Schizophrenia/prevention & control , Schizophrenic Psychology , Adolescent , Adult , Anti-Inflammatory Agents/adverse effects , Chi-Square Distribution , Clinical Protocols , Dietary Supplements/adverse effects , Double-Blind Method , Fatty Acids, Omega-3/adverse effects , Female , Humans , Intention to Treat Analysis , Male , Minocycline/adverse effects , Pakistan , Proportional Hazards Models , Psychotic Disorders/diagnosis , Psychotic Disorders/psychology , Research Design , Risk Assessment , Risk Factors , Schizophrenia/diagnosis , Time Factors , Treatment Outcome , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL