1
|
Li Y, Ong JWX, See YM, Yee JY, Tang C, Zheng S, Ng BT, Lee BTK, Rotzschke O, Andiappan AK, Lee J. Immunophenotyping schizophrenia subtypes stratified by antipsychotic response. Brain Behav Immun 2025; 123:656-671. [PMID: 39414177 DOI: 10.1016/j.bbi.2024.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 09/02/2024] [Accepted: 10/11/2024] [Indexed: 10/18/2024] Open
Abstract
Immune dysfunction has been proposed to play a role in the pathophysiology behind the development and persistence of psychosis. Current immunophenotyping studies are limited by small sample sizes and the number of immune markers investigated. Pharmacological subtypes in schizophrenia based on antipsychotic response have been proposed, but few studies have investigated immunophenotypes in treatment-resistant schizophrenia. In this study, we perform comprehensive immunophenotyping on 196 subjects comprising 147 schizophrenia patients stratified by antipsychotic response (49 antipsychotic-responsive, 70 clozapine-responsive, 28 clozapine-resistant) and 49 healthy controls. We aim to identify significant immune cell populations associated with schizophrenia and increasing treatment resistance, as potential modulators of underlying psychosis and/or treatment response. Patients with schizophrenia were recruited and assessed on the Clinical Global Impression - Schizophrenia (CGI-SCH). Treatment response was defined as a rating of three (mild severity) or less on the CGI-SCH positive symptom item after at least 8 weeks of adequate antipsychotic or clozapine treatment. Peripheral blood mononuclear cells were collected and flow cytometry was performed to identify 66 immune cell populations. Differences in cell population proportions were compared between schizophrenia cases and controls, and across all 4 groups, with post-hoc pairwise comparisons. Mucosal-associated invariant T (MAIT) cells (specifically CD8 + and DN double-negative subsets), total, exhausted and memory CD8 + T cells, VD1 + ϒδ T cells, plasmablasts, IgG + B cells and conventional dendritic cells 2 (cDC2) were among the top cell populations downregulated in schizophrenia. We observed increased downregulation with increasing treatment resistance. Conversely, naïve and exhausted CD4 + T cells, CD4/CD8 ratio and CCR5 + CCR2 + HLA DR + Myeloid cells were found to be upregulated in schizophrenia - we observed increased upregulation with increasing treatment resistance. We show significant immunophenotypic differences between schizophrenia cases and healthy controls, and consistent trend differences across varying degrees of antipsychotic resistance. We also examined immune cell populations not previously reported in schizophrenia. Future studies may explore immune markers identified as potential biomarkers of treatment resistance, and clarify on the relationship between immunological changes and pharmacological subtypes in schizophrenia.
Collapse
Affiliation(s)
- Yanhui Li
- Institute of Mental Health, Singapore. 10 Buangkok View, Buangkok Green Medical Park, Singapore 539747, Singapore
| | - Jocelyn Wen Xin Ong
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore. 8A Biomedical Grove Level 3 & 4. Immunos Building Singapore 138648, Singapore
| | - Yuen Mei See
- Institute of Mental Health, Singapore. 10 Buangkok View, Buangkok Green Medical Park, Singapore 539747, Singapore
| | - Jie Yin Yee
- Institute of Mental Health, Singapore. 10 Buangkok View, Buangkok Green Medical Park, Singapore 539747, Singapore
| | - Charmaine Tang
- Institute of Mental Health, Singapore. 10 Buangkok View, Buangkok Green Medical Park, Singapore 539747, Singapore
| | - Shushan Zheng
- Institute of Mental Health, Singapore. 10 Buangkok View, Buangkok Green Medical Park, Singapore 539747, Singapore
| | - Boon Tat Ng
- Institute of Mental Health, Singapore. 10 Buangkok View, Buangkok Green Medical Park, Singapore 539747, Singapore
| | - Bernett Teck Kwong Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore. 1 Mandalay Rd, Singapore 308232, Singapore
| | - Olaf Rotzschke
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore. 8A Biomedical Grove Level 3 & 4. Immunos Building Singapore 138648, Singapore
| | - Anand Kumar Andiappan
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore. 8A Biomedical Grove Level 3 & 4. Immunos Building Singapore 138648, Singapore
| | - Jimmy Lee
- Institute of Mental Health, Singapore. 10 Buangkok View, Buangkok Green Medical Park, Singapore 539747, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore. 1 Mandalay Rd, Singapore 308232, Singapore.
| |
Collapse
|
2
|
Uliana DL, Martinez A, Grace AA. THPP-1 PDE10A inhibitor reverses the cognitive deficits and hyperdopaminergic state in a neurodevelopment model of schizophrenia. Schizophr Res 2024; 274:315-326. [PMID: 39437478 DOI: 10.1016/j.schres.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 09/26/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024]
Abstract
Schizophrenia (SCZ) is a complex neuropsychiatric disorder characterized by positive, negative, and cognitive symptoms. The neurodevelopmental methylazoxy-methanol acetate (MAM) rodent model replicates key neurobiological features of SCZ which includes hyperdopaminergic states in the ventral tegmental area (VTA) and cognitive deficits. Typical and atypical antipsychotics are primarily effective in treating the positive symptoms of SCZ but often fall short of addressing cognitive deficits. A promising therapeutic approach for treating all symptoms of SCZ has emerged through the inhibition of phosphodiesterase 10 A (PDE10A). Our study aim was to investigate the impact of acute and chronic THPP-1 (PDE10A inhibitor) treatment, in MAM rats, focusing on cognitive deficits and VTA dopamine (DA) activity. Adult offspring of pregnant rats treated with Saline or MAM (20 mg/kg) on gestational day 17 were treated with THPP-1 acutely (male/female rats; 3 mg/kg) at postnatal day (PD) 70-80 or chronically (males; 3 weeks; 2-3 mg/kg) from PD 70-91 and tested in the novel object recognition test and electrophysiological recording of DA neurons in the VTA. Acute THPP-1 treatment reversed cognitive impairments and normalized the increased number of active DA neurons in the VTA of male and female MAM rats, without affecting control rats. Also, chronic THPP-1 treatment reversed cognitive deficits and normalized DA hyperactivity in the VTA of male MAM rats. The efficacy of THPP-1 in reversing MAM-induced impairments underscores its ability to target disease-specific circuitry without affecting normal regulated systems in control rats. Our findings highlight the therapeutic potential of THPP-1 for addressing cognitive deficits and DA dysregulation in SCZ.
Collapse
Affiliation(s)
- Daniela L Uliana
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Angela Martinez
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anthony A Grace
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
3
|
He Z, He Q, Tang X, Huang K, Lin Y, Xu J, Chen Q, Xu N, Yao L. Prenatal Exposure to MAM Impairs mPFC and Hippocampal Inhibitory Function in Mice during Adolescence and Adulthood. eNeuro 2024; 11:ENEURO.0362-24.2024. [PMID: 39500572 PMCID: PMC11625879 DOI: 10.1523/eneuro.0362-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/26/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Neurodevelopmental abnormalities are considered to be one of the important causes of schizophrenia. The offspring of methylazoxymethanol acetate (MAM)-exposed mice are recognized for the dysregulation of neurodevelopment and are well-characterized with schizophrenia-like phenotypes. However, the inhibition-related properties of the medial prefrontal cortex (mPFC) and hippocampus throughout adolescence and adulthood have not been systematically elucidated. In this study, both 10 and 15 mg/kg MAM-exposed mice exhibited schizophrenia-related phenotypes in both adolescence and adulthood, including spontaneous locomotion hyperactivity and deficits in prepulse inhibition. We observed that there was an obvious parvalbumin (PV) loss in the mPFC and hippocampus of MAM-exposed mice, extending from adolescence to adulthood. Moreover, the frequency of spontaneous inhibitory postsynaptic currents (sIPSCs) in pyramidal neurons at mPFC and hippocampus was significantly dampened in the 10 and 15 mg/kg MAM-exposed mice. Furthermore, the firing rate of putative pyramidal neurons in mPFC and hippocampus was increased, while that of putative inhibitory neurons was decreased during both adolescence and adulthood. In conclusion, PV loss in mPFC and hippocampus of MAM-exposed mice may contribute to the impaired inhibitory function leading to the attenuation of inhibition in the brain both in vitro and in vivo.
Collapse
Affiliation(s)
- Zhiyin He
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Qian He
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xiaorong Tang
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Keni Huang
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yiwen Lin
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Jianrui Xu
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Qiliang Chen
- School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Nenggui Xu
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Lulu Yao
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| |
Collapse
|
4
|
Khalid J, Aparasu RR. Adverse effects associated with antipsychotic use in older adults. Expert Opin Drug Saf 2024; 23:1157-1171. [PMID: 39076106 DOI: 10.1080/14740338.2024.2386377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 06/17/2024] [Accepted: 07/16/2024] [Indexed: 07/31/2024]
Abstract
INTRODUCTION Antipsychotic (AP) medications are extensively utilized for diverse psychiatric and non-psychiatric conditions, but they are associated with significant adverse effects in older adults. This expert opinion review provides an updated profile of adverse effects associated with AP medications in older adults. AREAS COVERED This review specifically examines real-world evidence for adverse events associated with AP use, including all-cause mortality, falls and fractures, diabetes, cardiovascular and cerebrovascular events, acute kidney injury, venous thromboembolism/pulmonary embolism, extrapyramidal symptoms, and infections based on real-world evidence. EXPERT OPINION This comprehensive safety review of AP revealed multiple adverse events in older adults. All-cause mortality, more associated with typical APs than atypicals, underscores a dose-dependent risk, urging cautious prescribing practices and avoidance of typical APs. Falls/fractures associated with atypical APs highlight the importance of judicious dosing and -close monitoring, especially after initiation. Cardiovascular risks, particularly stroke, with AP use emphasize the need for vigilant monitoring. Extrapyramidal symptoms, more linked to typical APs, also pose a significant risk in older adults. Pneumonia and pulmonary embolism/venous thromboembolism demand cautious consideration of APs, prompting the need for additional real-world studies to inform clinical practice. Conflicting evidence on diabetes and acute kidney injury necessitates ongoing studies and further research in older adults.
Collapse
Affiliation(s)
- Javeria Khalid
- Department of Pharmaceutical Health Outcomes and Policy, College of Pharmacy, University of Houston, Houston, Texas, USA
| | | |
Collapse
|
5
|
Bojesen KB, Rostrup E, Sigvard AK, Mikkelsen M, Edden RAE, Ebdrup BH, Glenthøj B. The Trajectory of Prefrontal GABA Levels in Initially Antipsychotic-Naïve Patients With Psychosis During 2 Years of Treatment and Associations With Striatal Cerebral Blood Flow and Outcome. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2024; 9:703-713. [PMID: 38145706 DOI: 10.1016/j.bpsc.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 11/03/2023] [Accepted: 12/15/2023] [Indexed: 12/27/2023]
Abstract
BACKGROUND GABAergic (gamma-aminobutyric acidergic) function in the prefrontal cortex seems dysfunctional in patients with first-episode psychosis, but the impact of longer-term treatment and relationship to clinical outcomes and striatal activity are unknown. METHODS A longitudinal study of 39 antipsychotic-naïve and benzodiazepine-free patients with psychosis (22.4 ± 5.4 years, 64% women) and 54 matched healthy control participants (HCs) (22.2 ± 4.3 years, 61% women) who were followed up after 6 weeks (28 patients, 51 HCs), 6 months (17 patients, 47 HCs), and 2 years (21 patients, 43 HCs) was completed. GABA levels in the dorsal anterior cingulate cortex and striatal resting cerebral blood flow were assessed on a 3T magnetic resonance scanner at all visits. RESULTS GABA levels in the dorsal anterior cingulate cortex were significantly lower in patients at baseline and after 6 weeks but not after 6 months or 2 years. Analyses of groups separately revealed decreased GABA levels after 2 years in HCs but stable levels in patients. Treatment increased striatal resting cerebral blood flow after 6 weeks and 6 months but not after 2 years. GABA levels were negatively associated with striatal resting cerebral blood flow in both groups at all visits. Last, lower baseline GABA levels in patients were related to less functional improvement after 2 years. CONCLUSIONS The findings suggest a different trajectory of GABA levels and striatal perfusion in first-episode patients over 2 years of antipsychotic treatment compared with HCs and indicate a downregulatory role of prefrontal GABAergic function on the striatum. Moreover, abnormally low prefrontal GABA level at illness onset may be a marker for a more severe prognosis.
Collapse
Affiliation(s)
- Kirsten Borup Bojesen
- Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Center Glostrup, University of Copenhagen, Glostrup, Denmark.
| | - Egill Rostrup
- Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Center Glostrup, University of Copenhagen, Glostrup, Denmark; Functional Imaging Unit, Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet Glostrup, University of Copenhagen, Copenhagen, Denmark
| | - Anne Korning Sigvard
- Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Center Glostrup, University of Copenhagen, Glostrup, Denmark
| | - Mark Mikkelsen
- Department of Radiology, Weill Cornell Medicine, New York, New York
| | - Richard A E Edden
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland; F.M. Kirby Research Center for Functional Brain Imaging, Baltimore, Maryland
| | - Bjørn Hylsebeck Ebdrup
- Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Center Glostrup, University of Copenhagen, Glostrup, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Birte Glenthøj
- Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Center Glostrup, University of Copenhagen, Glostrup, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
6
|
Tsugawa S, Honda S, Noda Y, Wannan C, Zalesky A, Tarumi R, Iwata Y, Ogyu K, Plitman E, Ueno F, Mimura M, Uchida H, Chakravarty M, Graff-Guerrero A, Nakajima S. Associations Between Structural Covariance Network and Antipsychotic Treatment Response in Schizophrenia. Schizophr Bull 2024; 50:382-392. [PMID: 37978044 PMCID: PMC10919786 DOI: 10.1093/schbul/sbad160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
BACKGROUND AND HYPOTHESIS Schizophrenia is associated with widespread cortical thinning and abnormality in the structural covariance network, which may reflect connectome alterations due to treatment effect or disease progression. Notably, patients with treatment-resistant schizophrenia (TRS) have stronger and more widespread cortical thinning, but it remains unclear whether structural covariance is associated with treatment response in schizophrenia. STUDY DESIGN We organized a multicenter magnetic resonance imaging study to assess structural covariance in a large population of TRS and non-TRS, who had been resistant and responsive to non-clozapine antipsychotics, respectively. Whole-brain structural covariance for cortical thickness was assessed in 102 patients with TRS, 77 patients with non-TRS, and 79 healthy controls (HC). Network-based statistics were used to examine the difference in structural covariance networks among the 3 groups. Moreover, the relationship between altered individual differentiated structural covariance and clinico-demographics was also explored. STUDY RESULTS Patients with non-TRS exhibited greater structural covariance compared with HC, mainly in the fronto-temporal and fronto-occipital regions, while there were no significant differences in structural covariance between TRS and non-TRS or HC. Higher individual differentiated structural covariance was associated with lower general scores of the Positive and Negative Syndrome Scale in the non-TRS group, but not in the TRS group. CONCLUSIONS These findings suggest that reconfiguration of brain networks via coordinated cortical thinning is related to treatment response in schizophrenia. Further longitudinal studies are warranted to confirm if greater structural covariance could serve as a marker for treatment response in this disease.
Collapse
Affiliation(s)
- Sakiko Tsugawa
- Department of Neuropsychiatry, Keio University, Tokyo, Japan
| | - Shiori Honda
- Department of Neuropsychiatry, Keio University, Tokyo, Japan
| | - Yoshihiro Noda
- Department of Neuropsychiatry, Keio University, Tokyo, Japan
| | - Cassandra Wannan
- Department of Psychiatry, University of Melbourne, Melbourne, Australia
| | - Andrew Zalesky
- Department of Biomedical Engineering, Melbourne School of Engineering, the University of Melbourne, Melbourne, Australia
| | - Ryosuke Tarumi
- Department of Neuropsychiatry, Keio University, Tokyo, Japan
- Department of Psychiatry, Komagino Hospital, Tokyo, Japan
| | - Yusuke Iwata
- Department of Neuropsychiatry, University of Yamanashi, Yamanashi, Japan
| | - Kamiyu Ogyu
- Department of Neuropsychiatry, Keio University, Tokyo, Japan
- Department of Psychiatry, National Hospital Organization Shimofusa Psychiatric Medical Center, Chiba, Japan
| | - Eric Plitman
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Fumihiko Ueno
- Department of Neuropsychiatry, Keio University, Tokyo, Japan
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Masaru Mimura
- Department of Neuropsychiatry, Keio University, Tokyo, Japan
| | - Hiroyuki Uchida
- Department of Neuropsychiatry, Keio University, Tokyo, Japan
| | - Mallar Chakravarty
- Department of Psychiatry, McGill University, Montreal, QC, Canada
- Computational Brain Anatomy Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, QC, Canada
| | | | | |
Collapse
|
7
|
de Almeida V, Mendes ND, Zuccoli GS, Reis-de-Oliveira G, Almeida GM, Podolsky-Gondim GG, Neder L, Martins-de-Souza D, Sebollela A. NMDA glutamate receptor antagonist MK-801 induces proteome changes in adult human brain slices which are partially counteracted by haloperidol and clozapine. J Neurochem 2024; 168:238-250. [PMID: 38332572 DOI: 10.1111/jnc.16059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 11/27/2023] [Accepted: 01/04/2024] [Indexed: 02/10/2024]
Abstract
Deciphering the molecular pathways associated with N-methyl-D-aspartate receptor (NMDAr) hypofunction and its interaction with antipsychotics is necessary to advance our understanding of the basis of schizophrenia, as well as our capacity to treat this disease. In this regard, the development of human brain-derived models that are amenable to studying the neurobiology of schizophrenia may contribute to filling the gaps left by the widely employed animal models. Here, we assessed the proteomic changes induced by the NMDA glutamate receptor antagonist MK-801 on human brain slice cultures obtained from adult donors submitted to respective neurosurgery. Initially, we demonstrated that MK-801 diminishes NMDA glutamate receptor signaling in human brain slices in culture. Next, using mass-spectrometry-based proteomics and systems biology in silico analyses, we found that MK-801 led to alterations in proteins related to several pathways previously associated with schizophrenia pathophysiology, including ephrin, opioid, melatonin, sirtuin signaling, interleukin 8, endocannabinoid, and synaptic vesicle cycle. We also evaluated the impact of both typical and atypical antipsychotics on MK-801-induced proteome changes. Interestingly, the atypical antipsychotic clozapine showed a more significant capacity to counteract the protein alterations induced by NMDAr hypofunction than haloperidol. Finally, using our dataset, we identified potential modulators of the MK-801-induced proteome changes, which may be considered promising targets to treat NMDAr hypofunction in schizophrenia. This dataset is publicly available and may be helpful in further studies aimed at evaluating the effects of MK-801 and antipsychotics in the human brain.
Collapse
Affiliation(s)
- Valéria de Almeida
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Niele Dias Mendes
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
- Department of Pathology and Forensic Medicine, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
- Division of Neurosurgery, Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Sao Paulo, Brazil
| | - Giuliana S Zuccoli
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Guilherme Reis-de-Oliveira
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Glaucia M Almeida
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - Guilherme Gozzoli Podolsky-Gondim
- Division of Neurosurgery, Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Sao Paulo, Brazil
| | - Luciano Neder
- Department of Pathology and Forensic Medicine, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
- Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBION) Conselho Nacional de Desenvolvimento Científico e Tecnológico, Sao Paulo, Brazil
- Experimental Medicine Research Cluster (EMRC), University of Campinas, Campinas, Sao Paulo, Brazil
- D'Or Institute for Research and Education (IDOR), Sao Paulo, Brazil
| | - Adriano Sebollela
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
8
|
Duncan L, Deisseroth K. Are novel treatments for brain disorders hiding in plain sight? Neuropsychopharmacology 2024; 49:276-281. [PMID: 37422511 PMCID: PMC10700299 DOI: 10.1038/s41386-023-01636-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/11/2023] [Accepted: 05/19/2023] [Indexed: 07/10/2023]
Abstract
Psychiatric diseases are strongly influenced by genetics, but genetically guided treatments have been slow to develop, and precise molecular mechanisms remain mysterious. Although individual locations in the genome tend to not contribute powerfully to psychiatric disease incidence, genome-wide association studies (GWAS) have now successfully linked hundreds of specific genetic loci to psychiatric disorders [1-3]. Here, building upon results from well-powered GWAS of four phenotypes relevant to psychiatry, we motivate an exploratory workflow leading from GWAS screening, through causal testing in animal models using methods such as optogenetics, to new therapies in human beings. We focus on schizophrenia and the dopamine D2 receptor (DRD2), hot flashes and the neurokinin B receptor (TACR3), cigarette smoking and receptors bound by nicotine (CHRNA5, CHRNA3, CHRNB4), and alcohol use and enzymes that help to break down alcohol (ADH1B, ADH1C, ADH7). A single genomic locus may not powerfully determine disease at the level of the population, but the same locus may nevertheless represent a potent treatment target suitable for population-wide therapeutic approaches.
Collapse
Affiliation(s)
- Laramie Duncan
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA.
| | - Karl Deisseroth
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA.
- Department of Bioengineering and Howard Hughes Medial Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
9
|
Bright Y, Maas DA, Verheij MM, Paladini MS, Amatdjais-Groenen HI, Molteni R, Riva MA, Martens GJ, Homberg JR. The Natural Protoalkaloid Methyl-2-Amino-3-Methoxybenzoate (MAM) Alleviates Positive as well as Cognitive Symptoms in Rat and Mouse Schizophrenia Models. Curr Neuropharmacol 2024; 22:323-338. [PMID: 37475559 PMCID: PMC10788887 DOI: 10.2174/1570159x21666230720122354] [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: 10/13/2022] [Revised: 01/19/2023] [Accepted: 01/19/2023] [Indexed: 07/22/2023] Open
Abstract
The development of new antipsychotics with pro-cognitive properties and less side effects represents a priority in schizophrenia drug research. In this study, we present for the first time a preclinical exploration of the effects of the promising natural atypical antipsychotic Methyl-2-Amino-3- Methoxybenzoate (MAM), a brain-penetrable protoalkaloid from the seed of the plant Nigella damascena. Using animal models related to hyperdopaminergic activity, namely the pharmacogenetic apomorphine (D2/D1 receptor agonist)-susceptible (APO-SUS) rat model and pharmacologically induced mouse and rat models of schizophrenia, we found that MAM reduced gnawing stereotypy and climbing behaviours induced by dopaminergic agents. This predicts antipsychotic activity. In line, MAM antagonized apomorphine-induced c-Fos and NPAS4 mRNA levels in post-mortem brain nucleus accumbens and dorsolateral striatum of APO-SUS rats. Furthermore, phencyclidine (PCP, an NMDA receptor antagonist) and 2,5-Dimethoxy-4-iodoamphetamine (DOI, a 5HT2A/2C receptor agonist) induced prepulse inhibition deficits, reflecting the positive symptoms of schizophrenia, which were rescued by treatment with MAM and atypical antipsychotics alike. Post-mortem brain immunostaining revealed that MAM blocked the strong activation of both PCP- and DOI-induced c-Fos immunoreactivity in a number of cortical areas. Finally, during a 28-day subchronic treatment regime, MAM did not induce weight gain, hyperglycemia, hyperlipidemia or hepato- and nephrotoxic effects, side effects known to be induced by atypical antipsychotics. MAM also did not show any cataleptic effects. In conclusion, its brain penetrability, the apparent absence of preclinical side effects, and its ability to antagonize positive and cognitive symptoms associated with schizophrenia make MAM an exciting new antipsychotic drug that deserves clinical testing.
Collapse
Affiliation(s)
- Yami Bright
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Dorien A. Maas
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, The Netherlands
- Department of Molecular Animal Physiology, Donders Institute for Brain, Cognition and Behaviour, Faculty of Science, Nijmegen, The Netherlands
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Michel M.M. Verheij
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Maria S. Paladini
- Department of Pharmacological and Biomolecular Sciences, Universita’ degli Studi di Milano, Milan, Italy
- Altos Labs Bay Area Institute of Science, Altos Labs, Inc., Redwood City, CA, USA
| | | | - Raffaella Molteni
- Department of Medical Biotechnology and Translational Medicine, Universita’ degli Studi di Milano, Milan, Italy
| | - Marco A. Riva
- Department of Pharmacological and Biomolecular Sciences, Universita’ degli Studi di Milano, Milan, Italy
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Gerard J.M. Martens
- Department of Molecular Animal Physiology, Donders Institute for Brain, Cognition and Behaviour, Faculty of Science, Nijmegen, The Netherlands
| | - Judith R. Homberg
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
10
|
Maćkowiak M. Psychedelics action and schizophrenia. Pharmacol Rep 2023; 75:1350-1361. [PMID: 37899392 PMCID: PMC10661800 DOI: 10.1007/s43440-023-00546-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/09/2023] [Accepted: 10/16/2023] [Indexed: 10/31/2023]
Abstract
Psychedelics are compounds acting by serotonin 5-hydroxytryptamine (5-HT)2A receptor activation and induce several behavioral responses. They are of special interest because of their positive effects on neuropsychiatric disorders (depression and posttraumatic stress disorder). However, several findings revealed that some psychedelic actions are similar to symptoms observed in schizophrenia (psychosis, sensorimotor gating impairments, attention, and working memory deficits) which might limit their clinical applications. Psychedelics activate some neurotransmitters, i.e., serotonergic, and glutamatergic, that are also impaired in schizophrenia. Therefore, the neurobiological background of psychedelics and schizophrenia is partially similar. Another important aspect to discuss is the perspective of using psychedelics in schizophrenia therapy. Postmortem studies showed a loss of synapses in schizophrenia, and the positive effects of psychedelics on neuroplasticity (synaptogenesis, neurogenesis, and neuritogenesis) might be essential in the context of schizophrenia therapy. However, because of psychedelics' psychotic action, the recommended doses of psychedelics in schizophrenia treatment are not established, and subpsychedelic dosing or microdosing are considered. Exploratory studies are needed to determine the tolerability of treatment and appropriate dosing regimen. Another therapeutic option is using non-hallucinogenic psychedelic analogs that also induce neuroplastic outcomes but do not have psychotogenic effects. Further preclinical and clinical studies are needed to recognize the potential effectiveness of 5-HT2A agonists in schizophrenia therapy.
Collapse
Affiliation(s)
- Marzena Maćkowiak
- Laboratory of Pharmacology and Brain Biostructure, Pharmacology Department, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland.
| |
Collapse
|
11
|
Taylor D, Chithiramohan R, Grewal J, Gupta A, Hansen L, Reynolds GP, Pappa S. Dopamine partial agonists: a discrete class of antipsychotics. Int J Psychiatry Clin Pract 2023; 27:272-284. [PMID: 36495086 DOI: 10.1080/13651501.2022.2151473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 11/04/2022] [Accepted: 11/08/2022] [Indexed: 12/14/2022]
Abstract
Worldwide, there are now three marketed dopamine D2 partial agonists: aripiprazole, brexpiprazole and cariprazine. These three drugs share a number of properties other than their action at D2 receptors. Pharmacologically, they are 5HT2 antagonists and D3 and 5HT1A partial agonists but with little or no alpha-adrenergic, anticholinergic or antihistaminic activity. They also share a long duration of action. Clinically, D2 partial agonists are effective antipsychotics and generally have useful antimanic and antidepressant activity. They are usually well tolerated, causing akathisia and insomnia only at the start of treatment, and are non-sedating. These drugs also share a very low risk of increased prolactin and of weight gain and accompanying metabolic effects. They may also have a relatively low risk of tardive dyskinesia. There is some evidence that they are preferred by patients to dopamine antagonists. Individual dopamineD2 partial agonists have much in common and as a group they differ importantly from dopamine D2 antagonists. Dopamine D2 partial agonists should be considered a distinct class of antipsychotics.Key pointsD2 partial agonists share many pharmacological and clinical propertiesD2 partial agonists differ in several important respects from D2 antagonistsD2 partial agonists should be considered a discrete class of antipsychotics.
Collapse
Affiliation(s)
- David Taylor
- Institute of Pharmaceutical Science, King's College London, London, UK
- Pharmacy Department, South London and Maudsley NHS Foundation Trust, London, UK
| | | | | | - Avirup Gupta
- South London and Maudsley NHS Foundation Trust, London, UK
| | - Lars Hansen
- Southampton University, Hartley Library B12, Southampton, UK
- Southern Health NHS Foundation Trust, Southampton, UK
| | - Gavin P Reynolds
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK
| | - Sofia Pappa
- Department of Brain Sciences, Imperial College London, London, UK
- West London NHS Trust, London, UK
| |
Collapse
|
12
|
Jiang S, Huang H, Zhou J, Li H, Duan M, Yao D, Luo C. Progressive trajectories of schizophrenia across symptoms, genes, and the brain. BMC Med 2023; 21:237. [PMID: 37400838 PMCID: PMC10318676 DOI: 10.1186/s12916-023-02935-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 06/12/2023] [Indexed: 07/05/2023] Open
Abstract
BACKGROUND Schizophrenia is characterized by complex psychiatric symptoms and unclear pathological mechanisms. Most previous studies have focused on the morphological changes that occur over the development of the disease; however, the corresponding functional trajectories remain unclear. In the present study, we aimed to explore the progressive trajectories of patterns of dysfunction after diagnosis. METHODS Eighty-six patients with schizophrenia and 120 healthy controls were recruited as the discovery dataset. Based on multiple functional indicators of resting-state brain functional magnetic resonance imaging, we conducted a duration-sliding dynamic analysis framework to investigate trajectories in association with disease progression. Neuroimaging findings were associated with clinical symptoms and gene expression data from the Allen Human Brain Atlas database. A replication cohort of patients with schizophrenia from the University of California, Los Angeles, was used as the replication dataset for the validation analysis. RESULTS Five stage-specific phenotypes were identified. A symptom trajectory was characterized by positive-dominated, negative ascendant, negative-dominated, positive ascendant, and negative surpassed stages. Dysfunctional trajectories from primary and subcortical regions to higher-order cortices were recognized; these are associated with abnormal external sensory gating and a disrupted internal excitation-inhibition equilibrium. From stage 1 to stage 5, the importance of neuroimaging features associated with behaviors gradually shifted from primary to higher-order cortices and subcortical regions. Genetic enrichment analysis identified that neurodevelopmental and neurodegenerative factors may be relevant as schizophrenia progresses and highlighted multiple synaptic systems. CONCLUSIONS Our convergent results indicate that progressive symptoms and functional neuroimaging phenotypes are associated with genetic factors in schizophrenia. Furthermore, the identification of functional trajectories complements previous findings of structural abnormalities and provides potential targets for drug and non-drug interventions in different stages of schizophrenia.
Collapse
Affiliation(s)
- Sisi Jiang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, People's Republic of China
- Research Unit of NeuroInformation, Chinese Academy of Medical Sciences, 2019RU035, Chengdu, People's Republic of China
| | - Huan Huang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, People's Republic of China
| | - Jingyu Zhou
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, People's Republic of China
| | - Hechun Li
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, People's Republic of China
| | - Mingjun Duan
- High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, Center for Information in Medicine, University of Electronic Science and Technology of China, No. 2006, Xiyuan Ave., West Hi-Tech Zone, 611731, Chengdu, Sichuan, People's Republic of China
| | - Dezhong Yao
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, People's Republic of China
- Research Unit of NeuroInformation, Chinese Academy of Medical Sciences, 2019RU035, Chengdu, People's Republic of China
- High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, Center for Information in Medicine, University of Electronic Science and Technology of China, No. 2006, Xiyuan Ave., West Hi-Tech Zone, 611731, Chengdu, Sichuan, People's Republic of China
| | - Cheng Luo
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, People's Republic of China.
- Research Unit of NeuroInformation, Chinese Academy of Medical Sciences, 2019RU035, Chengdu, People's Republic of China.
- High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, Center for Information in Medicine, University of Electronic Science and Technology of China, No. 2006, Xiyuan Ave., West Hi-Tech Zone, 611731, Chengdu, Sichuan, People's Republic of China.
| |
Collapse
|
13
|
Yang C, Zhang W, Liu J, Yao L, Bishop JR, Lencer R, Gong Q, Yang Z, Lui S. Disrupted subcortical functional connectome gradient in drug-naïve first-episode schizophrenia and the normalization effects after antipsychotic treatment. Neuropsychopharmacology 2023; 48:789-796. [PMID: 36496508 PMCID: PMC10066388 DOI: 10.1038/s41386-022-01512-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 11/18/2022] [Accepted: 11/23/2022] [Indexed: 12/14/2022]
Abstract
Antipsychotics are thought to improve schizophrenia symptoms through the antagonism of dopamine D2 receptors, which are abundant mainly in subcortical regions. By introducing functional gradient, a novel approach to identify hierarchy alterations by capturing the similarity of whole brain fucntional connectivity (FC) profiles between two voxels, the present study aimed to characterize how the subcortical gradient is associated with treatment effects and response in first-episode schizophrenia in vivo. Two independent samples of first-episode schizophrenia (FES) patients with matched healthy controls (HC) were obtained: the discovery dataset included 71 patients (FES0W) and 64 HC at baseline, and patients were re-scanned after either 6 weeks (FES6W, N = 33) or 12 months (FES12M, N = 57) of antipsychotic treatment, of which 19 patients finished both 6-week and 12-month evaluation. The validation dataset included 22 patients and 24 HC at baseline and patients were re-scanned after 6 weeks. Gradient metrics were calculated using BrainSpace Toolbox. Voxel-based gradient values were generated and group-averaged gradient values were further extracted across all voxels (global), three systems (thalamus, limbic and striatum) and their subcortical subfields. The comparisons were conducted separately between FES0W and HC for investigating illness effects, and between FES6W/FES12M and FES0W for treatment effects. Correlational analyses were then conducted between the longitudinal gradient alterations and the improvement of clinical ratings. Before treatment, schizophrenia patients exhibited an expanded range of global gradient scores compared to HC which indicated functional segregation within subcortical systems. The increased gradient in limbic system and decreased gradient in thalamic and striatal system contributed to the baseline abnormalities and led to the disruption of the subcortical functional integration. After treatment, these disruptions were normalized and the longitudinal changes of gradient scores in limbic system were significantly associated with symptom improvement. Similar illness and treatment effects were also observed in the validation dataset. By measuring functional hierarchy of subcortical organization, our findings provide a novel imaging marker that is sensitive to treatment effects and may make a promising indicator of treatment response in schizophrenia.
Collapse
Affiliation(s)
- Chengmin Yang
- Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, China
| | - Wenjing Zhang
- Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, China
| | - Jiajun Liu
- College of Electronic Engineering, Chengdu University of Information Technology, Chengdu, China
| | - Li Yao
- Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, China
| | - Jeffrey R Bishop
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Rebekka Lencer
- Department of Psychiatry and Psychotherapy, University of Lübeck, Lübeck, Germany
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, China
| | - Zhipeng Yang
- College of Electronic Engineering, Chengdu University of Information Technology, Chengdu, China.
| | - Su Lui
- Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, Department of Radiology, West China Hospital, Sichuan University, Chengdu, China.
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, China.
| |
Collapse
|
14
|
Free RB, Nilson AN, Boldizsar NM, Doyle TB, Rodriguiz RM, Pogorelov VM, Machino M, Lee KH, Bertz JW, Xu J, Lim HD, Dulcey AE, Mach RH, Woods JH, Lane JR, Shi L, Marugan JJ, Wetsel WC, Sibley DR. Identification and Characterization of ML321: A Novel and Highly Selective D 2 Dopamine Receptor Antagonist with Efficacy in Animal Models That Predict Atypical Antipsychotic Activity. ACS Pharmacol Transl Sci 2023; 6:151-170. [PMID: 36654757 PMCID: PMC9841785 DOI: 10.1021/acsptsci.2c00202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Indexed: 12/31/2022]
Abstract
We have developed and characterized a novel D2R antagonist with exceptional GPCR selectivity - ML321. In functional profiling screens of 168 different GPCRs, ML321 showed little activity beyond potent inhibition of the D2R and to a lesser extent the D3R, demonstrating excellent receptor selectivity. The D2R selectivity of ML321 may be related to the fact that, unlike other monoaminergic ligands, ML321 lacks a positively charged amine group and adopts a unique binding pose within the orthosteric binding site of the D2R. PET imaging studies in non-human primates demonstrated that ML321 penetrates the CNS and occupies the D2R in a dose-dependent manner. Behavioral paradigms in rats demonstrate that ML321 can selectively antagonize a D2R-mediated response (hypothermia) while not affecting a D3R-mediated response (yawning) using the same dose of drug, thus indicating exceptional in vivo selectivity. We also investigated the effects of ML321 in animal models that are predictive of antipsychotic efficacy in humans. We found that ML321 attenuates both amphetamine- and phencyclidine-induced locomotor activity and restored pre-pulse inhibition (PPI) of acoustic startle in a dose-dependent manner. Surprisingly, using doses that were maximally effective in both the locomotor and PPI studies, ML321 was relatively ineffective in promoting catalepsy. Kinetic studies revealed that ML321 exhibits slow-on and fast-off receptor binding rates, similar to those observed with atypical antipsychotics with reduced extrapyramidal side effects. Taken together, these observations suggest that ML321, or a derivative thereof, may exhibit ″atypical″ antipsychotic activity in humans with significantly fewer side effects than observed with the currently FDA-approved D2R antagonists.
Collapse
Affiliation(s)
- R. Benjamin Free
- Molecular
Neuropharmacology Section, National Institute of Neurological Disorders
and Stroke, Intramural Research Program, National Institutes of Health, 35 Convent Drive, MSC-3723, Bethesda, Maryland20892, United States
| | - Ashley N. Nilson
- Molecular
Neuropharmacology Section, National Institute of Neurological Disorders
and Stroke, Intramural Research Program, National Institutes of Health, 35 Convent Drive, MSC-3723, Bethesda, Maryland20892, United States
| | - Noelia M. Boldizsar
- Molecular
Neuropharmacology Section, National Institute of Neurological Disorders
and Stroke, Intramural Research Program, National Institutes of Health, 35 Convent Drive, MSC-3723, Bethesda, Maryland20892, United States
| | - Trevor B. Doyle
- Molecular
Neuropharmacology Section, National Institute of Neurological Disorders
and Stroke, Intramural Research Program, National Institutes of Health, 35 Convent Drive, MSC-3723, Bethesda, Maryland20892, United States
| | - Ramona M. Rodriguiz
- Department
of Psychiatry and Behavioral Sciences, Mouse Behavioral and Neuroendocrine
Analysis Core Facility, Duke University
Medical Center, 354 Sands Building, 303 Research Drive, Durham, North Carolina27710, United States
| | - Vladimir M. Pogorelov
- Department
of Psychiatry and Behavioral Sciences, Mouse Behavioral and Neuroendocrine
Analysis Core Facility, Duke University
Medical Center, 354 Sands Building, 303 Research Drive, Durham, North Carolina27710, United States
| | - Mayako Machino
- Computational
Chemistry and Molecular Biophysics Section, Molecular Targets and
Medications Discovery Branch, National Institute on Drug Abuse, Intramural
Research Program, National Institutes of
Health, 333 Cassell Drive, Baltimore, Maryland21224, United
States
| | - Kuo Hao Lee
- Computational
Chemistry and Molecular Biophysics Section, Molecular Targets and
Medications Discovery Branch, National Institute on Drug Abuse, Intramural
Research Program, National Institutes of
Health, 333 Cassell Drive, Baltimore, Maryland21224, United
States
| | - Jeremiah W. Bertz
- Department
of Pharmacology, University of Michigan
Medical School, 1150 W. Medical Center Dr., Ann Arbor, Michigan48109, United States
| | - Jinbin Xu
- Division
of Radiological Sciences, Department of Radiology, Mallinckrodt Institute
of Radiology, Washington University School
of Medicine, St. Louis, Missouri63110, United States
| | - Herman D. Lim
- Drug Discovery
Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, VIC3052, Australia
| | - Andrés E. Dulcey
- Division
of Pre-Clinical Innovation, National Center for Advancing Translational
Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, Maryland20850, United States
| | - Robert H. Mach
- Department
of Radiology, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, Pennsylvania19104, United States
| | - James H. Woods
- Department
of Pharmacology, University of Michigan
Medical School, 1150 W. Medical Center Dr., Ann Arbor, Michigan48109, United States
| | - J Robert Lane
- Centre
of Membrane Proteins and Receptors, Universities
of Birmingham and Nottingham, NottinghamNG7 2UH, United Kingdom
| | - Lei Shi
- Computational
Chemistry and Molecular Biophysics Section, Molecular Targets and
Medications Discovery Branch, National Institute on Drug Abuse, Intramural
Research Program, National Institutes of
Health, 333 Cassell Drive, Baltimore, Maryland21224, United
States
| | - Juan J. Marugan
- Division
of Pre-Clinical Innovation, National Center for Advancing Translational
Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, Maryland20850, United States
| | - William C. Wetsel
- Department
of Psychiatry and Behavioral Sciences, Mouse Behavioral and Neuroendocrine
Analysis Core Facility, Duke University
Medical Center, 354 Sands Building, 303 Research Drive, Durham, North Carolina27710, United States
- Departments
of Neurobiology and Cell Biology, Duke University
Medical Center, 354 Sands Building, 303 Research Drive, Durham, North Carolina27710, United States
| | - David R. Sibley
- Molecular
Neuropharmacology Section, National Institute of Neurological Disorders
and Stroke, Intramural Research Program, National Institutes of Health, 35 Convent Drive, MSC-3723, Bethesda, Maryland20892, United States
| |
Collapse
|
15
|
Tang Y, Wu Y, Li X, Hao Q, Deng W, Yue W, Yan H, Zhang Y, Tan L, Chen Q, Yang G, Lu T, Wang L, Yang F, Zhang F, Yang J, Li K, Lv L, Tan Q, Zhang H, Ma X, Li L, Wang C, Ma X, Zhang D, Yu H, Zhao L, Ren H, Wang Y, Zhang G, Li C, Du X, Hu X, Li T, Wang Q. Early Efficacy of Antipsychotic Medications at Week 2 Predicts Subsequent Responses at Week 6 in a Large-scale Randomized Controlled Trial. Curr Neuropharmacol 2023; 21:424-436. [PMID: 36411567 PMCID: PMC10190139 DOI: 10.2174/1570159x21666221118164612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 09/25/2022] [Accepted: 10/18/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Since the early clinical efficacy of antipsychotics has not yet been well perceived, this study sought to decide whether the efficacy of antipsychotics at week 2 can predict subsequent responses at week 6 and identify how such predictive capacities vary among different antipsychotics and psychotic symptoms. METHODS A total of 3010 patients with schizophrenia enrolled in a randomized controlled trial (RCT) and received a 6-week treatment with one antipsychotic drug randomly chosen from five atypical antipsychotics (risperidone 2-6 mg/d, olanzapine 5-20 mg/d, quetiapine 400-750 mg/d, aripiprazole 10-30 mg/d, and ziprasidone 80-160 mg/d) and two typical antipsychotics (perphenazine 20-60 mg/d and haloperidol 6-20 mg/d). Early efficacy was defined as the reduction rate using the Positive and Negative Syndrome Scale (PANSS) total score at week 2. With cut-offs at 50% reduction, logistic regression, receiver operating characteristic (ROC) and random forests were adopted. RESULTS The reduction rate of PANSS total score and improvement of psychotic symptoms at week 2 enabled subsequent responses to 7 antipsychotics to be predicted, in which improvements in delusions, lack of judgment and insight, unusual thought content, and suspiciousness/ persecution were endowed with the greatest weight. CONCLUSION It is robust enough to clinically predict treatment responses to antipsychotics at week 6 using the reduction rate of PANSS total score and symptom relief at week 2. Psychiatric clinicians had better determine whether to switch the treatment plan by the first 2 weeks.
Collapse
Affiliation(s)
- Yiguo Tang
- Mental Health Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Sichuan Clinical Medical Research Center for Mental Disorders, Chengdu, China
| | - Yulu Wu
- Mental Health Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Sichuan Clinical Medical Research Center for Mental Disorders, Chengdu, China
| | - Xiaojing Li
- Department of Neurobiology, Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - QinJian Hao
- The Center of Gerontology and Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Wei Deng
- Department of Neurobiology, Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Weihua Yue
- Peking University Sixth Hospital, Institute of Mental Health, Beijing, China
- National Clinical Research Center for Mental Disorders and Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, China
| | - Hao Yan
- Peking University Sixth Hospital, Institute of Mental Health, Beijing, China
- National Clinical Research Center for Mental Disorders and Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, China
| | - Yamin Zhang
- Department of Neurobiology, Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Liwen Tan
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qi Chen
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guigang Yang
- Beijing Anding Hospital, Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Tianlan Lu
- Peking University Sixth Hospital, Institute of Mental Health, Beijing, China
- National Clinical Research Center for Mental Disorders and Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, China
| | - Lifang Wang
- Peking University Sixth Hospital, Institute of Mental Health, Beijing, China
- National Clinical Research Center for Mental Disorders and Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, China
| | - Fude Yang
- Beijing HuiLongGuan Hospital, Beijing, China
| | - Fuquan Zhang
- Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - Jianli Yang
- Institute of Mental Health, Tianjin Anding Hospital, Tianjin, China
- Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
| | - Keqing Li
- Hebei Mental Health Center, Baoding, Hebei, China
| | - Luxian Lv
- Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Qingrong Tan
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, Shanxi, China
| | - Hongyan Zhang
- Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - Xin Ma
- Beijing Anding Hospital, Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Lingjiang Li
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chuanyue Wang
- Beijing Anding Hospital, Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Xiaohong Ma
- Mental Health Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Sichuan Clinical Medical Research Center for Mental Disorders, Chengdu, China
| | - Dai Zhang
- Peking University Sixth Hospital, Institute of Mental Health, Beijing, China
- National Clinical Research Center for Mental Disorders and Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, China
| | - Hao Yu
- Department of Psychiatry, Jining Medical University, Jining, China
| | - Liansheng Zhao
- Mental Health Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Sichuan Clinical Medical Research Center for Mental Disorders, Chengdu, China
| | - Hongyan Ren
- Mental Health Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Sichuan Clinical Medical Research Center for Mental Disorders, Chengdu, China
| | - Yingcheng Wang
- Mental Health Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Sichuan Clinical Medical Research Center for Mental Disorders, Chengdu, China
| | - Guangya Zhang
- Department of Psychiatry, Suzhou Psychiatric Hospital, Suzhou, China
- The Affiliated Guangji Hospital of Soochow University, Suzhou, China
| | - Chuanwei Li
- Department of Psychiatry, Suzhou Psychiatric Hospital, Suzhou, China
- The Affiliated Guangji Hospital of Soochow University, Suzhou, China
| | - Xiangdong Du
- Department of Psychiatry, Suzhou Psychiatric Hospital, Suzhou, China
- The Affiliated Guangji Hospital of Soochow University, Suzhou, China
| | - Xun Hu
- The Clinical Research Center and the Department of Pathology, Affiliated Second Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Tao Li
- Department of Neurobiology, Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qiang Wang
- Mental Health Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Sichuan Clinical Medical Research Center for Mental Disorders, Chengdu, China
| |
Collapse
|
16
|
Lencz T, Moyett A, Argyelan M, Barber AD, Cholewa J, Birnbaum ML, Gallego JA, John M, Szeszko PR, Robinson DG, Malhotra AK. Frontal lobe fALFF measured from resting-state fMRI as a prognostic biomarker in first-episode psychosis. Neuropsychopharmacology 2022; 47:2245-2251. [PMID: 36198875 PMCID: PMC9630308 DOI: 10.1038/s41386-022-01470-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/05/2022] [Accepted: 09/21/2022] [Indexed: 11/09/2022]
Abstract
Clinical response to antipsychotic drug treatment is highly variable, yet prognostic biomarkers are lacking. The goal of the present study was to test whether the fractional amplitude of low-frequency fluctuations (fALFF), as measured from baseline resting-state fMRI data, can serve as a potential biomarker of treatment response to antipsychotics. Patients in the first episode of psychosis (n = 126) were enrolled in two prospective studies employing second-generation antipsychotics (risperidone or aripiprazole). Patients were scanned at the initiation of treatment on a 3T MRI scanner (Study 1, GE Signa HDx, n = 74; Study 2, Siemens Prisma, n = 52). Voxelwise fALFF derived from baseline resting-state fMRI scans served as the primary measure of interest, providing a hypothesis-free (as opposed to region-of-interest) search for regions of the brain that might be predictive of response. At baseline, patients who would later meet strict criteria for clinical response (defined as two consecutive ratings of much or very much improved on the CGI, as well as a rating of ≤3 on psychosis-related items of the BPRS-A) demonstrated significantly greater baseline fALFF in bilateral orbitofrontal cortex compared to non-responders. Thus, spontaneous activity in orbitofrontal cortex may serve as a prognostic biomarker of antipsychotic treatment.
Collapse
Affiliation(s)
- Todd Lencz
- Department of Psychiatry, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, 11550, USA.
- Department of Psychiatry, Division of Research, The Zucker Hillside Hospital Division of Northwell Health, Glen Oaks, NY, 11004, USA.
- Institute for Behavioral Science, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA.
| | - Ashley Moyett
- Department of Psychiatry, Division of Research, The Zucker Hillside Hospital Division of Northwell Health, Glen Oaks, NY, 11004, USA
| | - Miklos Argyelan
- Department of Psychiatry, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, 11550, USA
- Department of Psychiatry, Division of Research, The Zucker Hillside Hospital Division of Northwell Health, Glen Oaks, NY, 11004, USA
- Institute for Behavioral Science, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
| | - Anita D Barber
- Department of Psychiatry, Division of Research, The Zucker Hillside Hospital Division of Northwell Health, Glen Oaks, NY, 11004, USA
- Institute for Behavioral Science, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
| | - John Cholewa
- Department of Psychiatry, Division of Research, The Zucker Hillside Hospital Division of Northwell Health, Glen Oaks, NY, 11004, USA
| | - Michael L Birnbaum
- Department of Psychiatry, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, 11550, USA
- Department of Psychiatry, Division of Research, The Zucker Hillside Hospital Division of Northwell Health, Glen Oaks, NY, 11004, USA
- Institute for Behavioral Science, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
| | - Juan A Gallego
- Department of Psychiatry, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, 11550, USA
- Department of Psychiatry, Division of Research, The Zucker Hillside Hospital Division of Northwell Health, Glen Oaks, NY, 11004, USA
- Institute for Behavioral Science, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
| | - Majnu John
- Department of Psychiatry, Division of Research, The Zucker Hillside Hospital Division of Northwell Health, Glen Oaks, NY, 11004, USA
- Institute for Behavioral Science, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
- Department of Mathematics, Hofstra University, Hempstead, NY, 11549, USA
| | - Philip R Szeszko
- James J. Peters VA Medical Center, Bronx, NY, 10468, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Delbert G Robinson
- Department of Psychiatry, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, 11550, USA
- Department of Psychiatry, Division of Research, The Zucker Hillside Hospital Division of Northwell Health, Glen Oaks, NY, 11004, USA
- Institute for Behavioral Science, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
| | - Anil K Malhotra
- Department of Psychiatry, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, 11550, USA
- Department of Psychiatry, Division of Research, The Zucker Hillside Hospital Division of Northwell Health, Glen Oaks, NY, 11004, USA
- Institute for Behavioral Science, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
| |
Collapse
|
17
|
Abstract
Growing evidence indicates that a suboptimal intrauterine environment confers risk for schizophrenia. The developmental model of schizophrenia posits that aberrant brain growth during early brain development and adolescence may interact to contribute to this psychiatric disease in adulthood. Although a variety of factors may perturb the environment of the developing fetus and predispose for schizophrenia later, a common mechanism has yet to be elucidated. Micronutrient deficiencies during the perinatal period are known to induce potent effects on brain development by altering neurodevelopmental processes. Iron is an important candidate nutrient to consider because of its role in energy metabolism, monoamine synthesis, synaptogenesis, myelination, and the high prevalence of iron deficiency (ID) in the mother-infant dyad. Understanding the current state of science regarding perinatal ID as an early risk factor for schizophrenia is imperative to inform empirical work investigating the etiology of schizophrenia and develop prevention and intervention programs. In this narrative review, we focus on perinatal ID as a common mechanism underlying the fetal programming of schizophrenia. First, we review the neural aberrations associated with perinatal ID that indicate risk for schizophrenia in adulthood, including disruptions in dopaminergic neurotransmission, hippocampal-dependent learning and memory, and sensorimotor gating. Second, we review the pathophysiology of perinatal ID as a function of maternal ID during pregnancy and use epidemiological and cohort studies to link perinatal ID with risk of schizophrenia. Finally, we review potential confounding phenotypes, including nonanemic causes of perinatal brain ID and future risk of schizophrenia.
Collapse
Affiliation(s)
- Andrea M. Maxwell
- Medical Scientist Training Program, University of Minnesota, Minneapolis, MN 55455 (USA)
| | - Raghavendra B. Rao
- Department of Pediatrics, Division of Neonatology, University of Minnesota Medical School, Minneapolis, MN 55455 (USA)
- Center for Neurobehavioral Development, University of Minnesota, Minneapolis, MN 55455 (USA)
| |
Collapse
|
18
|
Phosphorylation Signals Downstream of Dopamine Receptors in Emotional Behaviors: Association with Preference and Avoidance. Int J Mol Sci 2022; 23:ijms231911643. [PMID: 36232945 PMCID: PMC9570387 DOI: 10.3390/ijms231911643] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 09/26/2022] [Accepted: 09/28/2022] [Indexed: 11/09/2022] Open
Abstract
Dopamine regulates emotional behaviors, including rewarding and aversive behaviors, through the mesolimbic dopaminergic pathway, which projects dopamine neurons from the ventral tegmental area to the nucleus accumbens (NAc). Protein phosphorylation is critical for intracellular signaling pathways and physiological functions, which are regulated by neurotransmitters in the brain. Previous studies have demonstrated that dopamine stimulated the phosphorylation of intracellular substrates, such as receptors, ion channels, and transcription factors, to regulate neuronal excitability and synaptic plasticity through dopamine receptors. We also established a novel database called KANPHOS that provides information on phosphorylation signals downstream of monoamines identified by our kinase substrate screening methods, including dopamine, in addition to those reported in the literature. Recent advances in proteomics techniques have enabled us to clarify the mechanisms through which dopamine controls rewarding and aversive behaviors through signal pathways in the NAc. In this review, we discuss the intracellular phosphorylation signals regulated by dopamine in these two emotional behaviors.
Collapse
|
19
|
Al Abadey A, Connor B, Flamme ACL, Robichon K. Clozapine reduces chemokine-mediated migration of lymphocytes by targeting NF-κB and AKT phosphorylation. Cell Signal 2022; 99:110449. [PMID: 36031090 DOI: 10.1016/j.cellsig.2022.110449] [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: 06/20/2022] [Revised: 08/11/2022] [Accepted: 08/22/2022] [Indexed: 11/27/2022]
Abstract
Multiple sclerosis is a disease characterised by demyelination of axons in the central nervous system. The atypical antipsychotic drug clozapine has been shown to attenuate disease severity in experimental autoimmune encephalomyelitis (EAE), a mouse model that is useful for the study of multiple sclerosis. However, the mechanism of action by which clozapine reduces disease in EAE is poorly understood. To better understand how clozapine exerts its protective effects, we investigated the underlying signalling pathways by which clozapine may reduce immune cell migration by evaluating chemokine and dopamine receptor-associated signalling pathways. We found that clozapine inhibits migration of immune cells by reducing chemokine production in microglia cells by targeting NF-κB phosphorylation and promoting an anti-inflammatory milieu. Furthermore, clozapine directly targets immune cell migration by changing Ca2+ levels within immune cells and reduces the phosphorylation of signalling protein AKT. Linking these pathways to the antagonising effect of clozapine on dopamine and serotonin receptors, we provide insight into how clozapine alters immune cells migration by directly targeting the underlying migration-associated pathways.
Collapse
Affiliation(s)
- Afnan Al Abadey
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand; Centre for Biodiscovery Wellington, Victoria University of Wellington, Wellington, New Zealand
| | - Bronwen Connor
- Department of Pharmacology and Clinical Pharmacology, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Anne Camille La Flamme
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand; Centre for Biodiscovery Wellington, Victoria University of Wellington, Wellington, New Zealand; Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Katharina Robichon
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand; Centre for Biodiscovery Wellington, Victoria University of Wellington, Wellington, New Zealand.
| |
Collapse
|
20
|
Li Y, Zhu Z, Sun L, Xiang Z, Chang C, Fan C. Physicochemical Insights on Terahertz Wave Diminished Side Effects of Drugs from Slow Dissociation. ACS NANO 2022; 16:8419-8426. [PMID: 35438483 DOI: 10.1021/acsnano.2c02952] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Dopamine D2 receptors (D2Rs) are one of the most intensely investigated and well-established drug targets for neuropsychiatric disorders. Selective D2R antagonists have been developed as efficacious antipsychotic drugs. Nevertheless, the potent drugs with necessarily high affinity are prone to slow dissociation, which invokes a plethora of severe side effects such as extrapyramidal symptoms, substantial weight gain, associated diabetes, etc. This has become a major barrier in treating psychiatric patients. In this work, we propose a physical method, terahertz wave modulation, to promote the dissociation of high-affinity antipsychotics and thus diminish the side effects. We have proven that a 4.0 THz wave could reduce the affinity by 71% between the D2R and a risperidone ligand and meanwhile expand the exit via conformation modulation, which promises an accelerated dissociation of risperidone. In addition, it is estimated that the enhancement of the dissociation rate due to lowered binding by terahertz irritation could constitute up to 8 orders of magnitude, which is fairly impressive and resembles the enzyme's catalysis. Also, acceleration of the dissociation rate could be adjusted by the irritation strength. This work elaborates the terahertz wave-modulated noncovalent interactions critical in cell signaling pathways. Most importantly, it demonstrates the feasibility of terahertz technologies intervening in receptor-ligand complex regulated diseases such as neurodegenerative disorders, metabolic diseases, etc.
Collapse
Affiliation(s)
- Yangmei Li
- Innovation Laboratory of Terahertz Biophysics, National Innovation Institute of Defense Technology, Beijing 100071, China
| | - Zhi Zhu
- School of Optical-Electrical Computer Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Lan Sun
- Innovation Laboratory of Terahertz Biophysics, National Innovation Institute of Defense Technology, Beijing 100071, China
| | - Zuoxian Xiang
- Innovation Laboratory of Terahertz Biophysics, National Innovation Institute of Defense Technology, Beijing 100071, China
| | - Chao Chang
- Innovation Laboratory of Terahertz Biophysics, National Innovation Institute of Defense Technology, Beijing 100071, China
- School of Physics, Peking University, Beijing 100871, China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, and Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
21
|
Motamed M, Karimi H, Sanjari Moghaddam H, Taherzadeh Boroujeni S, Sanatian Z, Hasanzadeh A, Khodaei Ardakani MR, Akhondzadeh S. Risperidone combination therapy with adalimumab for treatment of chronic schizophrenia: a randomized, double-blind, placebo-controlled clinical trial. Int Clin Psychopharmacol 2022; 37:92-101. [PMID: 35258035 DOI: 10.1097/yic.0000000000000399] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
This study aimed to investigate the efficacy and safety of antitumor necrosis factor-alpha (TNF-α) therapy using adalimumab in patients with chronic schizophrenia. This is a randomized, double-blind, placebo-controlled clinical trial carried out at Roozbeh Hospital (Tehran, Iran) from June 2020 to October 2021. The patients were randomly divided into two parallel adalimumab + risperidone and placebo + risperidone groups. Participants in the intervention group received adalimumab subcutaneous injection (40 mg) by pen-injector at weeks 0 and 4. Using the Positive and Negative Symptoms Scale (PANSS), patients' positive and negative symptoms were assessed at weeks 0, 4, and 8. Forty patients (20 in each group) were included. PANSS total (t = 4.43, df = 38, P < 0.001), negative (t = 2.88, df = 38, P = 0.006), and general psychopathology (t = 4.06, df = 38, P < 0.001) scores demonstrated a significantly greater decline in adalimumab compared with the placebo group from baseline study endpoint. However, improvement of PANSS positive subscale scores showed no significant difference from the baseline study endpoint. There was no significant between-group difference regarding levels of C-reactive protein, interleukin (IL)-1β, TNF-α, IL-6, and IL-8 at baseline and also at the week 8 visit (P > 0.05 for all). The current study found adalimumab adjunctive therapy effective in treating schizophrenia, particularly its negative and general psychopathology symptoms, with no side effects.
Collapse
Affiliation(s)
- Mahsa Motamed
- Psychiatric Research Center, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences
| | - Hanieh Karimi
- Psychiatric Research Center, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences
| | | | | | - Zahra Sanatian
- Psychiatric Research Center, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences
| | - Alireza Hasanzadeh
- Psychiatric Research Center, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences
| | | | - Shahin Akhondzadeh
- Psychiatric Research Center, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences
| |
Collapse
|
22
|
Jaitpal V, Gawande S. Olanzapine-Induced Parkinsonism and Akathisia: A Case Report. Cureus 2022; 14:e21354. [PMID: 35198269 PMCID: PMC8851297 DOI: 10.7759/cureus.21354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2022] [Indexed: 11/09/2022] Open
Abstract
This article sheds light on the case of a 22-year-old patient with paranoid schizophrenia. A treatment regimen of olanzapine (an atypical antipsychotic) of 15 mg/day was initiated for the patient and it was associated with extrapyramidal symptoms (EPS) such as drug-induced parkinsonism (DIP) and akathisia. Based on the different treatment regimens available, the patient was switched to an alternate antipsychotic along with anticholinergics, antihistamines, and benzodiazepines, following which the patient recovered well and did not report any further side effects. The highlighted case stresses the need for proper monitoring of the drug dose and sequential periodic examination of the patient to nullify the risk of adverse effects and poor compliance.
Collapse
|
23
|
Maini K, Hollier JW, Gould H, Bollich V, John LaForge J, Cornett EM, Edinoff AN, Kaye AM, Kaye AD. Lumateperone tosylate, A Selective and Concurrent Modulator of Serotonin, Dopamine, and Glutamate, in the Treatment of Schizophrenia. Health Psychol Res 2021; 9:24932. [PMID: 34746489 DOI: 10.52965/001c.24932] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 06/17/2021] [Indexed: 11/06/2022] Open
Abstract
Purpose of Review This is a comprehensive review of the literature regarding the use of Lumateperone tosylate for schizophrenia. This review presents the background, evidence, and indications for the use of lumateperone tosylate in the treatment of schizophrenia. Recent Findings Schizophrenia is a chronic mental health disorder that affects approximately 3.3 million people in the United States. Its symptoms, which must be present more than six months, are comprised of disorganized behavior and speech, a diminished capacity to comprehend reality, hearing voices unheard by others, seeing things unseen by others, delusions, decreased social commitment, and decreased motivation. The majority of these symptoms can be managed with antipsychotic medication. Lumateperone is a selective and concurrent modulator of serotonin, dopamine, and glutamate, which all mediate or modulate serious mental illness. Summary Schizophrenia is a complex, severe mental illness that affects how the brain processes information. There are many medications used to treat schizophrenia. One antipsychotic agent, lumateperone tosylate, is a newer agent that the FDA recently approved. The most common adverse effects are shown to be mild such as somnolence, constipation, sedation, and fatigue, with the 42 mg recommended dose. Lumateperone tosylate is an FDA-approved drug that can be given only at the 42mg dose once daily with no titration requirements.
Collapse
Affiliation(s)
| | | | - Haley Gould
- Louisiana State University Shreveport School of Medicine, Shreveport, LA
| | - Victoria Bollich
- Louisiana State University Shreveport School of Medicine, Shreveport, LA
| | - John John LaForge
- Louisiana State University Shreveport School of Medicine, Shreveport, LA
| | | | | | - Adam M Kaye
- Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Stockton, CA
| | | |
Collapse
|
24
|
Trace Amine-Associated Receptor 1 as a Target for the Development of New Antipsychotics: Current Status of Research and Future Directions. CNS Drugs 2021; 35:1153-1161. [PMID: 34655036 DOI: 10.1007/s40263-021-00864-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/20/2021] [Indexed: 10/20/2022]
Abstract
Schizophrenia is a mental illness associated with an array of symptoms that often result in disability. The primary treatments for schizophrenia are termed antipsychotics. Although antipsychotics modulate a number of different receptor types and subtypes, all currently regulatory agency-approved antipsychotics share in common direct or functional antagonism at the dopamine type 2 receptor (D2R). The majority of people with schizophrenia do not achieve full resolution of their symptoms with antipsychotics, suggesting the need for alternative or complementary approaches. The primary focus of this review is to assess the evidence for the role of the trace amine-associated receptor 1 (TAAR-1) in schizophrenia and the role of TAAR-1 modulators as novel-mechanism antipsychotics. Topics include an overview of TAAR-1 physiology and pathophysiology in schizophrenia, interaction with other neurotransmitter systems, including the dopaminergic, glutamatergic and serotonergic system, and finally, a review of investigational TAAR-1 compounds that have reached Phase II clinical studies in schizophrenia: SEP-363856 (ulotaront) and RO6889450 (ralmitaront). Thus far, results are publicly available only for ulotaront in a relatively young (18-40 years) and acutely exacerbated cohort. These results showed positive effects for overall schizophrenia symptoms without significant tolerability concerns. An ongoing study of ralmitaront will assess specific efficacy in patients with persistent negative symptoms. If trials of TAAR-1 modulators, and other novel-mechanism targets for schizophrenia that are under active study, continue to show positive results, the definition of an antipsychotic may need to be expanded beyond the D2R target in the near future.
Collapse
|
25
|
Free RB, Cuoco CA, Xie B, Namkung Y, Prabhu VV, Willette BKA, Day MM, Sanchez-Soto M, Lane JR, Laporte SA, Shi L, Allen JE, Sibley DR. Pharmacological Characterization of the Imipridone Anticancer Drug ONC201 Reveals a Negative Allosteric Mechanism of Action at the D 2 Dopamine Receptor. Mol Pharmacol 2021; 100:372-387. [PMID: 34353882 PMCID: PMC8626643 DOI: 10.1124/molpharm.121.000336] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 07/26/2021] [Indexed: 11/22/2022] Open
Abstract
ONC201 is a first-in-class imipridone compound that is in clinical trials for the treatment of high-grade gliomas and other advanced cancers. Recent studies identified that ONC201 antagonizes D2-like dopamine receptors at therapeutically relevant concentrations. In the current study, characterization of ONC201 using radioligand binding and multiple functional assays revealed that it was a full antagonist of the D2 and D3 receptors (D2R and D3R) with low micromolar potencies, similar to its potency for antiproliferative effects. Curve-shift experiments using D2R-mediated β-arrestin recruitment and cAMP assays revealed that ONC201 exhibited a mixed form of antagonism. An operational model of allostery was used to analyze these data, which suggested that the predominant modulatory effect of ONC201 was on dopamine efficacy with little to no effect on dopamine affinity. To investigate how ONC201 binds to the D2R, we employed scanning mutagenesis coupled with a D2R-mediated calcium efflux assay. Eight residues were identified as being important for ONC201's functional antagonism of the D2R. Mutation of these residues followed by assessing ONC201 antagonism in multiple signaling assays highlighted specific residues involved in ONC201 binding. Together with computational modeling and simulation studies, our results suggest that ONC201 interacts with the D2R in a bitopic manner where the imipridone core of the molecule protrudes into the orthosteric binding site, but does not compete with dopamine, whereas a secondary phenyl ring engages an allosteric binding pocket that may be associated with negative modulation of receptor activity. SIGNIFICANCE STATEMENT: ONC201 is a novel antagonist of the D2 dopamine receptor with demonstrated efficacy in the treatment of various cancers, especially high-grade glioma. This study demonstrates that ONC201 antagonizes the D2 receptor with novel bitopic and negative allosteric mechanisms of action, which may explain its high selectivity and some of its clinical anticancer properties that are distinct from other D2 receptor antagonists widely used for the treatment of schizophrenia and other neuropsychiatric disorders.
Collapse
Affiliation(s)
- R Benjamin Free
- Molecular Neuropharmacology Section, National Institute of Neurologic Disorders and Stroke, National Institutes of Health, Bethesda, Maryland (R.B.F., C.A.C., B.K.A.W., M.M.D., M.S-S., D.R.S.); Chimerix, Inc., Durham, North Carolina (V.V.P., J.E.A.); Computational Chemistry and Molecular Biophysics Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland (B.X., L.S.); Department of Medicine, Research Institute of the McGill University Health Center (RI-MUHC), McGill University, Montréal, Canada (Y.N., S.A.P.); and Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, Nottingham, Midlands, United Kingdom (J.R.L.)
| | - Caroline A Cuoco
- Molecular Neuropharmacology Section, National Institute of Neurologic Disorders and Stroke, National Institutes of Health, Bethesda, Maryland (R.B.F., C.A.C., B.K.A.W., M.M.D., M.S-S., D.R.S.); Chimerix, Inc., Durham, North Carolina (V.V.P., J.E.A.); Computational Chemistry and Molecular Biophysics Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland (B.X., L.S.); Department of Medicine, Research Institute of the McGill University Health Center (RI-MUHC), McGill University, Montréal, Canada (Y.N., S.A.P.); and Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, Nottingham, Midlands, United Kingdom (J.R.L.)
| | - Bing Xie
- Molecular Neuropharmacology Section, National Institute of Neurologic Disorders and Stroke, National Institutes of Health, Bethesda, Maryland (R.B.F., C.A.C., B.K.A.W., M.M.D., M.S-S., D.R.S.); Chimerix, Inc., Durham, North Carolina (V.V.P., J.E.A.); Computational Chemistry and Molecular Biophysics Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland (B.X., L.S.); Department of Medicine, Research Institute of the McGill University Health Center (RI-MUHC), McGill University, Montréal, Canada (Y.N., S.A.P.); and Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, Nottingham, Midlands, United Kingdom (J.R.L.)
| | - Yoon Namkung
- Molecular Neuropharmacology Section, National Institute of Neurologic Disorders and Stroke, National Institutes of Health, Bethesda, Maryland (R.B.F., C.A.C., B.K.A.W., M.M.D., M.S-S., D.R.S.); Chimerix, Inc., Durham, North Carolina (V.V.P., J.E.A.); Computational Chemistry and Molecular Biophysics Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland (B.X., L.S.); Department of Medicine, Research Institute of the McGill University Health Center (RI-MUHC), McGill University, Montréal, Canada (Y.N., S.A.P.); and Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, Nottingham, Midlands, United Kingdom (J.R.L.)
| | - Varun V Prabhu
- Molecular Neuropharmacology Section, National Institute of Neurologic Disorders and Stroke, National Institutes of Health, Bethesda, Maryland (R.B.F., C.A.C., B.K.A.W., M.M.D., M.S-S., D.R.S.); Chimerix, Inc., Durham, North Carolina (V.V.P., J.E.A.); Computational Chemistry and Molecular Biophysics Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland (B.X., L.S.); Department of Medicine, Research Institute of the McGill University Health Center (RI-MUHC), McGill University, Montréal, Canada (Y.N., S.A.P.); and Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, Nottingham, Midlands, United Kingdom (J.R.L.)
| | - Blair K A Willette
- Molecular Neuropharmacology Section, National Institute of Neurologic Disorders and Stroke, National Institutes of Health, Bethesda, Maryland (R.B.F., C.A.C., B.K.A.W., M.M.D., M.S-S., D.R.S.); Chimerix, Inc., Durham, North Carolina (V.V.P., J.E.A.); Computational Chemistry and Molecular Biophysics Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland (B.X., L.S.); Department of Medicine, Research Institute of the McGill University Health Center (RI-MUHC), McGill University, Montréal, Canada (Y.N., S.A.P.); and Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, Nottingham, Midlands, United Kingdom (J.R.L.)
| | - Marilyn M Day
- Molecular Neuropharmacology Section, National Institute of Neurologic Disorders and Stroke, National Institutes of Health, Bethesda, Maryland (R.B.F., C.A.C., B.K.A.W., M.M.D., M.S-S., D.R.S.); Chimerix, Inc., Durham, North Carolina (V.V.P., J.E.A.); Computational Chemistry and Molecular Biophysics Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland (B.X., L.S.); Department of Medicine, Research Institute of the McGill University Health Center (RI-MUHC), McGill University, Montréal, Canada (Y.N., S.A.P.); and Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, Nottingham, Midlands, United Kingdom (J.R.L.)
| | - Marta Sanchez-Soto
- Molecular Neuropharmacology Section, National Institute of Neurologic Disorders and Stroke, National Institutes of Health, Bethesda, Maryland (R.B.F., C.A.C., B.K.A.W., M.M.D., M.S-S., D.R.S.); Chimerix, Inc., Durham, North Carolina (V.V.P., J.E.A.); Computational Chemistry and Molecular Biophysics Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland (B.X., L.S.); Department of Medicine, Research Institute of the McGill University Health Center (RI-MUHC), McGill University, Montréal, Canada (Y.N., S.A.P.); and Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, Nottingham, Midlands, United Kingdom (J.R.L.)
| | - J Robert Lane
- Molecular Neuropharmacology Section, National Institute of Neurologic Disorders and Stroke, National Institutes of Health, Bethesda, Maryland (R.B.F., C.A.C., B.K.A.W., M.M.D., M.S-S., D.R.S.); Chimerix, Inc., Durham, North Carolina (V.V.P., J.E.A.); Computational Chemistry and Molecular Biophysics Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland (B.X., L.S.); Department of Medicine, Research Institute of the McGill University Health Center (RI-MUHC), McGill University, Montréal, Canada (Y.N., S.A.P.); and Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, Nottingham, Midlands, United Kingdom (J.R.L.)
| | - Stéphane A Laporte
- Molecular Neuropharmacology Section, National Institute of Neurologic Disorders and Stroke, National Institutes of Health, Bethesda, Maryland (R.B.F., C.A.C., B.K.A.W., M.M.D., M.S-S., D.R.S.); Chimerix, Inc., Durham, North Carolina (V.V.P., J.E.A.); Computational Chemistry and Molecular Biophysics Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland (B.X., L.S.); Department of Medicine, Research Institute of the McGill University Health Center (RI-MUHC), McGill University, Montréal, Canada (Y.N., S.A.P.); and Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, Nottingham, Midlands, United Kingdom (J.R.L.)
| | - Lei Shi
- Molecular Neuropharmacology Section, National Institute of Neurologic Disorders and Stroke, National Institutes of Health, Bethesda, Maryland (R.B.F., C.A.C., B.K.A.W., M.M.D., M.S-S., D.R.S.); Chimerix, Inc., Durham, North Carolina (V.V.P., J.E.A.); Computational Chemistry and Molecular Biophysics Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland (B.X., L.S.); Department of Medicine, Research Institute of the McGill University Health Center (RI-MUHC), McGill University, Montréal, Canada (Y.N., S.A.P.); and Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, Nottingham, Midlands, United Kingdom (J.R.L.)
| | - Joshua E Allen
- Molecular Neuropharmacology Section, National Institute of Neurologic Disorders and Stroke, National Institutes of Health, Bethesda, Maryland (R.B.F., C.A.C., B.K.A.W., M.M.D., M.S-S., D.R.S.); Chimerix, Inc., Durham, North Carolina (V.V.P., J.E.A.); Computational Chemistry and Molecular Biophysics Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland (B.X., L.S.); Department of Medicine, Research Institute of the McGill University Health Center (RI-MUHC), McGill University, Montréal, Canada (Y.N., S.A.P.); and Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, Nottingham, Midlands, United Kingdom (J.R.L.)
| | - David R Sibley
- Molecular Neuropharmacology Section, National Institute of Neurologic Disorders and Stroke, National Institutes of Health, Bethesda, Maryland (R.B.F., C.A.C., B.K.A.W., M.M.D., M.S-S., D.R.S.); Chimerix, Inc., Durham, North Carolina (V.V.P., J.E.A.); Computational Chemistry and Molecular Biophysics Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland (B.X., L.S.); Department of Medicine, Research Institute of the McGill University Health Center (RI-MUHC), McGill University, Montréal, Canada (Y.N., S.A.P.); and Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, Nottingham, Midlands, United Kingdom (J.R.L.)
| |
Collapse
|
26
|
Remington G, Hahn MK, Agarwal SM, Chintoh A, Agid O. Schizophrenia: Antipsychotics and drug development. Behav Brain Res 2021; 414:113507. [PMID: 34352293 DOI: 10.1016/j.bbr.2021.113507] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 07/25/2021] [Accepted: 07/28/2021] [Indexed: 12/21/2022]
Abstract
The introduction of chlorpromazine and the work that ensued provided the foundation to reposition schizophrenia as a biological illness. The present paper follows the evolution of antipsychotics and their shift from 'typical' to 'atypical'. Atypicality is reviewed in reference to its original definition, clozapine's role, and developments that now leave the concept's utility in question. In a similar fashion, drug development is reviewed in the context of the illness' multiple symptom domains, as well as differences captured by clinical staging and phenotyping. Collectively, the evidence argues for a more nuanced approach to drug development that aligns with the illness' heterogeneity and complexity. Just as 'atypical' as a descriptor for antipsychotics may be outdated, it may be time to set aside the notion of developing drugs that treat 'schizophrenia'.
Collapse
Affiliation(s)
- Gary Remington
- University of Toronto, Department of Psychiatry, University of Toronto, Toronto, Canada; Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada.
| | - Margaret K Hahn
- University of Toronto, Department of Psychiatry, University of Toronto, Toronto, Canada; Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada
| | - Sri Mahavir Agarwal
- University of Toronto, Department of Psychiatry, University of Toronto, Toronto, Canada; Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada
| | - Araba Chintoh
- University of Toronto, Department of Psychiatry, University of Toronto, Toronto, Canada; Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada
| | - Ofer Agid
- University of Toronto, Department of Psychiatry, University of Toronto, Toronto, Canada; Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada
| |
Collapse
|
27
|
Amani P, Habibpour R, Karami L, Hofmann A. Docking Screens of Noncovalent Interaction Motifs of the Human Subtype-D2 Receptor-75 Schizophrenia Antipsychotic Complexes with Physicochemical Appraisal of Antipsychotics. ACS Chem Neurosci 2021; 12:2218-2232. [PMID: 34061513 DOI: 10.1021/acschemneuro.1c00229] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Chemoinformatics appraisal and molecular docking were employed to investigate 225 complexes of 75 schizophrenia antipsychotics with the dopamine receptor subtypes D2R, D3R, and D4R. Considering the effective noncovalent interactions in the subtype-D2 receptor selectivity of antipsychotics, this study evaluated the possible physicochemical properties of ligands underlying the design of safer and more effective antipsychotics. The pan-assay interference compounds (PAINs) include about 25% of typical antipsychotics and 5% of atypicals. Popular antipsychotics like haloperidol, clozapine, risperidone, and aripiprazole are not PAINs. They have stronger interactions with D2R and D4R, but their interactions with D3R are slightly weaker, which is similar to the behavior of dopamine. In contrast to typical antipsychotics, atypical antipsychotics exhibit more noncovalent interactions with D4R than with D2R. These results suggest that selectivity to D2R and D4R comes from the synergy between hydrophobic and hydrogen-bonding interactions through their concomitant occurrence in the form of a hydrogen-bonding site adorned with hydrophobic contacts in antipsychotic-receptor complexes. All the antipsychotics had more synergic interactions with D2R and D4R in comparison with D3R. The atypical antipsychotics made a good distinction between the subtype D2 receptors with high selectivity to D4R. Among the popular antipsychotics, haloperidol, clozapine, and risperidone have hydrophobic-hydrogen-bonding synergy with D4R, while aripiprazole profits with D2R. The most important residue participating in the synergic interactions was threonine for D2R and cysteine for D4R. This work could be useful in informing and guiding future drug discovery and development studies aimed at receptor-specific antipsychotics.
Collapse
Affiliation(s)
- Parisa Amani
- Department of Chemical Technology, Iranian Research Organization for Science and Technology, Tehran 3313193685, Iran
| | - Razieh Habibpour
- Department of Chemical Technology, Iranian Research Organization for Science and Technology, Tehran 3313193685, Iran
| | - Leila Karami
- Department of Cell and Molecular Biology, Kharazmi University, Tehran 1571914911, Iran
| | - Andreas Hofmann
- Griffith Institute for Drug Discovery, Griffith University, Nathan 4111, Australia
- Department of Veterinary Biosciences, The University of Melbourne, Parkville 3010, Australia
| |
Collapse
|
28
|
Gómez-Revuelta M, Pelayo-Terán JM, Vázquez-Bourgon J, Ortiz-García de la Foz V, Mayoral-van Son J, Ayesa-Arriola R, Crespo-Facorro B. Aripiprazole vs Risperidone for the acute-phase treatment of first-episode psychosis: A 6-week randomized, flexible-dose, open-label clinical trial. Eur Neuropsychopharmacol 2021; 47:74-85. [PMID: 33678469 DOI: 10.1016/j.euroneuro.2021.02.009] [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] [Received: 10/07/2020] [Revised: 02/14/2021] [Accepted: 02/17/2021] [Indexed: 12/25/2022]
Abstract
Selecting the first antipsychotic agent for the acute phase of a first episode of psychosis (FEP) is a critical task that may impact on the long-term outcome. Despite that, there is a lack of research comparing head-to-head different second-generation antipsychotics at this stage. The aim of this study was to compare the effectiveness of aripiprazole and risperidone in the treatment of the acute phase after a FEP. For that purpose, from February 2011 to October 2018, a prospective, randomized, open-label study was undertaken. Two hundred-sixty-six first-episode, drug-naïve patients were randomly assigned to aripiprazole (n = 136), or risperidone (n = 130) and followed-up for 6-weeks. The primary effectiveness measure was all-cause treatment discontinuation. In addition, an analysis based on intention-to-treat principle was conducted to assess clinical efficacy. The overall dropout rate at 6-week reached 19.5%. Effectiveness measures were similar between both treatment groups as treatment discontinuation rates (χ2 = 1.863; p = 0.172) and mean time until all-cause discontinuation (log rank = 1.421; p = 0.233) showed no statistically significant differences. In terms of clinical efficacy, risperidone proved a statistically significant better performance according to BPRS mean change between baseline and 6-week total score (t = 3.187; p = 0.002). Patients under risperidone treatment were significantly more likely to suffer sex-related adverse events. In conclusion, no differences regarding effectiveness were found between aripiprazole and risperidone for the acute-phase treatment of FEP. Despite the importance of efficacy during this phase of treatment, selecting the most effective treatment for the long-term outcome, requires addressing safety and patient´s preferences.
Collapse
Affiliation(s)
- Marcos Gómez-Revuelta
- University Hospital Marqués de Valdecilla-IDIVAL, Department of Psychiatry, School of Medicine, University of Cantabria, Avda. Valdecilla n 25, 39008, SANTANDER, Cantabria, Santander, Spain.
| | - José María Pelayo-Terán
- University Hospital Marqués de Valdecilla-IDIVAL, Department of Psychiatry, School of Medicine, University of Cantabria, Avda. Valdecilla n 25, 39008, SANTANDER, Cantabria, Santander, Spain; CIBERSAM, Centro Investigación Biomédica en Red Salud Mental, Madrid, Spain; Servicio de Psiquiatría y Salud Mental, Hospital El Bierzo, GASBI, Servicio de Salud de Castilla y León (SACYL), Ponferrada (León), Spain
| | - Javier Vázquez-Bourgon
- University Hospital Marqués de Valdecilla-IDIVAL, Department of Psychiatry, School of Medicine, University of Cantabria, Avda. Valdecilla n 25, 39008, SANTANDER, Cantabria, Santander, Spain; CIBERSAM, Centro Investigación Biomédica en Red Salud Mental, Madrid, Spain
| | - Víctor Ortiz-García de la Foz
- University Hospital Marqués de Valdecilla-IDIVAL, Department of Psychiatry, School of Medicine, University of Cantabria, Avda. Valdecilla n 25, 39008, SANTANDER, Cantabria, Santander, Spain; CIBERSAM, Centro Investigación Biomédica en Red Salud Mental, Madrid, Spain
| | - Jacqueline Mayoral-van Son
- University Hospital Marqués de Valdecilla-IDIVAL, Department of Psychiatry, School of Medicine, University of Cantabria, Avda. Valdecilla n 25, 39008, SANTANDER, Cantabria, Santander, Spain; CIBERSAM, Centro Investigación Biomédica en Red Salud Mental, Madrid, Spain; Hospital Universitario Virgen del Rocío, Department of Psychiatry, Universidad de Sevilla, Sevilla, Spain. Instituto de Investigacion Sanitaria de Sevilla, IBiS Spain
| | - Rosa Ayesa-Arriola
- University Hospital Marqués de Valdecilla-IDIVAL, Department of Psychiatry, School of Medicine, University of Cantabria, Avda. Valdecilla n 25, 39008, SANTANDER, Cantabria, Santander, Spain; CIBERSAM, Centro Investigación Biomédica en Red Salud Mental, Madrid, Spain
| | - Benedicto Crespo-Facorro
- University Hospital Marqués de Valdecilla-IDIVAL, Department of Psychiatry, School of Medicine, University of Cantabria, Avda. Valdecilla n 25, 39008, SANTANDER, Cantabria, Santander, Spain; CIBERSAM, Centro Investigación Biomédica en Red Salud Mental, Madrid, Spain; Hospital Universitario Virgen del Rocío, Department of Psychiatry, Universidad de Sevilla, Sevilla, Spain. Instituto de Investigacion Sanitaria de Sevilla, IBiS Spain
| |
Collapse
|
29
|
Boczek T, Mackiewicz J, Sobolczyk M, Wawrzyniak J, Lisek M, Ferenc B, Guo F, Zylinska L. The Role of G Protein-Coupled Receptors (GPCRs) and Calcium Signaling in Schizophrenia. Focus on GPCRs Activated by Neurotransmitters and Chemokines. Cells 2021; 10:cells10051228. [PMID: 34067760 PMCID: PMC8155952 DOI: 10.3390/cells10051228] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/12/2021] [Accepted: 05/14/2021] [Indexed: 01/13/2023] Open
Abstract
Schizophrenia is a common debilitating disease characterized by continuous or relapsing episodes of psychosis. Although the molecular mechanisms underlying this psychiatric illness remain incompletely understood, a growing body of clinical, pharmacological, and genetic evidence suggests that G protein-coupled receptors (GPCRs) play a critical role in disease development, progression, and treatment. This pivotal role is further highlighted by the fact that GPCRs are the most common targets for antipsychotic drugs. The GPCRs activation evokes slow synaptic transmission through several downstream pathways, many of them engaging intracellular Ca2+ mobilization. Dysfunctions of the neurotransmitter systems involving the action of GPCRs in the frontal and limbic-related regions are likely to underly the complex picture that includes the whole spectrum of positive and negative schizophrenia symptoms. Therefore, the progress in our understanding of GPCRs function in the control of brain cognitive functions is expected to open new avenues for selective drug development. In this paper, we review and synthesize the recent data regarding the contribution of neurotransmitter-GPCRs signaling to schizophrenia symptomology.
Collapse
Affiliation(s)
- Tomasz Boczek
- Department of Molecular Neurochemistry, Faculty of Health Sciences, Medical University of Lodz, 92215 Lodz, Poland; (T.B.); (J.M.); (M.S.); (J.W.); (M.L.); (B.F.)
| | - Joanna Mackiewicz
- Department of Molecular Neurochemistry, Faculty of Health Sciences, Medical University of Lodz, 92215 Lodz, Poland; (T.B.); (J.M.); (M.S.); (J.W.); (M.L.); (B.F.)
| | - Marta Sobolczyk
- Department of Molecular Neurochemistry, Faculty of Health Sciences, Medical University of Lodz, 92215 Lodz, Poland; (T.B.); (J.M.); (M.S.); (J.W.); (M.L.); (B.F.)
| | - Julia Wawrzyniak
- Department of Molecular Neurochemistry, Faculty of Health Sciences, Medical University of Lodz, 92215 Lodz, Poland; (T.B.); (J.M.); (M.S.); (J.W.); (M.L.); (B.F.)
| | - Malwina Lisek
- Department of Molecular Neurochemistry, Faculty of Health Sciences, Medical University of Lodz, 92215 Lodz, Poland; (T.B.); (J.M.); (M.S.); (J.W.); (M.L.); (B.F.)
| | - Bozena Ferenc
- Department of Molecular Neurochemistry, Faculty of Health Sciences, Medical University of Lodz, 92215 Lodz, Poland; (T.B.); (J.M.); (M.S.); (J.W.); (M.L.); (B.F.)
| | - Feng Guo
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang 110122, China;
| | - Ludmila Zylinska
- Department of Molecular Neurochemistry, Faculty of Health Sciences, Medical University of Lodz, 92215 Lodz, Poland; (T.B.); (J.M.); (M.S.); (J.W.); (M.L.); (B.F.)
- Correspondence:
| |
Collapse
|
30
|
Foster DJ, Bryant ZK, Conn PJ. Targeting muscarinic receptors to treat schizophrenia. Behav Brain Res 2021; 405:113201. [PMID: 33647377 PMCID: PMC8006961 DOI: 10.1016/j.bbr.2021.113201] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 02/02/2021] [Accepted: 02/18/2021] [Indexed: 11/23/2022]
Abstract
Schizophrenia is a severe neuropsychiatric disorder characterized by a diverse range of symptoms that can have profound impacts on the lives of patients. Currently available antipsychotics target dopamine receptors, and while they are useful for ameliorating the positive symptoms of the disorder, this approach often does not significantly improve negative and cognitive symptoms. Excitingly, preclinical and clinical research suggests that targeting specific muscarinic acetylcholine receptor subtypes could provide more comprehensive symptomatic relief with the potential to ameliorate numerous symptom domains. Mechanistic studies reveal that M1, M4, and M5 receptor subtypes can modulate the specific brain circuits and physiology that are disrupted in schizophrenia and are thought to underlie positive, negative, and cognitive symptoms. Novel therapeutic strategies for targeting these receptors are now advancing in clinical and preclinical development and expand upon the promise of these new treatment strategies to potentially provide more comprehensive relief than currently available antipsychotics.
Collapse
Affiliation(s)
- Daniel J Foster
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, United States; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, 37232, United States
| | - Zoey K Bryant
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, United States; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, 37232, United States
| | - P Jeffrey Conn
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, United States; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, 37232, United States.
| |
Collapse
|
31
|
Gomes FV, Grace AA. Beyond Dopamine Receptor Antagonism: New Targets for Schizophrenia Treatment and Prevention. Int J Mol Sci 2021; 22:4467. [PMID: 33922888 PMCID: PMC8123139 DOI: 10.3390/ijms22094467] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/20/2021] [Accepted: 04/22/2021] [Indexed: 02/06/2023] Open
Abstract
Treatment of schizophrenia (SCZ) historically relies on the use of antipsychotic drugs to treat psychosis, with all of the currently available antipsychotics acting through the antagonism of dopamine D2 receptors. Although antipsychotics reduce psychotic symptoms in many patients, they induce numerous undesirable effects and are not effective against negative and cognitive symptoms. These highlight the need to develop new drugs to treat SCZ. An advanced understanding of the circuitry of SCZ has pointed to pathological origins in the excitation/inhibition balance in regions such as the hippocampus, and restoring function in this region, particularly as a means to compensate for parvalbumin (PV) interneuron loss and resultant hippocampal hyperactivity, may be a more efficacious approach to relieve a broad range of SCZ symptoms. Other targets, such as cholinergic receptors and the trace amine-associated receptor 1 (TAAR1), have also shown some promise for the treatment of SCZ. Importantly, assessing efficacy of novel compounds must take into consideration treatment history of the patient, as preclinical studies suggest prior antipsychotic treatment may interfere with the efficacy of these novel agents. However, while novel therapeutic targets may be more effective in treating SCZ, a more effective approach would be to prevent the transition to SCZ in susceptible individuals. A focus on stress, which has been shown to be a predisposing factor in risk for SCZ, is a possible avenue that has shown promise in preclinical studies. Therefore, therapeutic approaches based on our current understanding of the circuitry of SCZ and its etiology are likely to enable development of more effective therapeutic interventions for this complex disorder.
Collapse
Affiliation(s)
- Felipe V. Gomes
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 01000-000, Brazil;
| | - Anthony A. Grace
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| |
Collapse
|
32
|
Robichon K, Sondhauss S, Jordan TW, Keyzers RA, Connor B, La Flamme AC. Localisation of clozapine during experimental autoimmune encephalomyelitis and its impact on dopamine and its receptors. Sci Rep 2021; 11:2966. [PMID: 33536582 PMCID: PMC7858600 DOI: 10.1038/s41598-021-82667-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 01/14/2021] [Indexed: 01/11/2023] Open
Abstract
Multiple sclerosis is a disease characterised by axonal demyelination in the central nervous system (CNS). The atypical antipsychotic drug clozapine attenuates experimental autoimmune encephalomyelitis (EAE), a mouse model used to study multiple sclerosis, but the precise mechanism is unknown and could include both peripheral and CNS-mediated effects. To better understand where clozapine exerts its protective effects, we investigated the tissue distribution and localisation of clozapine using matrix-assisted laser desorption ionization imaging mass spectrometry and liquid chromatography-mass spectrometry. We found that clozapine was detectable in the brain and enriched in specific brain regions (cortex, thalamus and olfactory bulb), but the distribution was not altered by EAE. Furthermore, although not altered in other organs, clozapine levels were significantly elevated in serum during EAE. Because clozapine antagonises dopamine receptors, we analysed dopamine levels in serum and brain as well as dopamine receptor expression on brain-resident and infiltrating immune cells. While neither clozapine nor EAE significantly affected dopamine levels, we observed a significant downregulation of dopamine receptors 1 and 5 and up-regulation of dopamine receptor 2 on microglia and CD4+-infiltrating T cells during EAE. Together these findings provide insight into how neuroinflammation, as modelled by EAE, alters the distribution and downstream effects of clozapine.
Collapse
Affiliation(s)
- Katharina Robichon
- School of Biological Sciences, Victoria University of Wellington, P.O. Box 600, Wellington, 6140, New Zealand
- Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Sven Sondhauss
- School of Biological Sciences, Victoria University of Wellington, P.O. Box 600, Wellington, 6140, New Zealand
- Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - T William Jordan
- School of Biological Sciences, Victoria University of Wellington, P.O. Box 600, Wellington, 6140, New Zealand
- Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Robert A Keyzers
- Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
- School of Chemical and Physical Sciences, Victoria University of Wellington, Wellington, 6140, New Zealand
| | - Bronwen Connor
- Department of Pharmacology and Clinical Pharmacology, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Anne C La Flamme
- School of Biological Sciences, Victoria University of Wellington, P.O. Box 600, Wellington, 6140, New Zealand.
- Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand.
- Malaghan Institute of Medical Research, Wellington, New Zealand.
| |
Collapse
|
33
|
Tsartsalis S, Tournier BB, Gloria Y, Millet P, Ginovart N. Effect of 5-HT2A receptor antagonism on levels of D2/3 receptor occupancy and adverse behavioral side-effects induced by haloperidol: a SPECT imaging study in the rat. Transl Psychiatry 2021; 11:51. [PMID: 33446643 PMCID: PMC7809418 DOI: 10.1038/s41398-020-01179-5] [Citation(s) in RCA: 3] [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: 07/25/2020] [Revised: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 12/30/2022] Open
Abstract
Several studies suggested that 5-HT2A receptor (5-HT2AR) blockade may provide a more favorable efficacy and side-effect profile to antipsychotic treatment. We hypothesized that a combined haloperidol (a D2/3 receptor (D2/3R) antagonist) and MDL-100,907 (a 5-HT2AR antagonist) treatment would reverse the side effects and the neurochemical alterations induced by haloperidol alone and would potentialize its efficacy. We thus chronically treated male Mdr1a knock-out rats with several doses of haloperidol alone or in combination with a saturating dose of a MDL-100,907. Receptor occupancy at clinically relevant levels was validated with a dual-radiotracer in-vivo SPECT imaging of D2/3R and 5-HT2AR occupancy. Experimental tests of efficacy (dizocilpine-disrupted prepulse inhibition (PPI) of the startle reflex) and side effects (catalepsy, vacuous chewing movements) were performed. Finally, a second dual-radiotracer in-vivo SPECT scan assessed the neurochemical changes induced by the chronic treatments. Chronic haloperidol failed to reverse PPI disruption induced by dizocilpine, whilst administration of MDL-100,907 along with haloperidol was associated with a reversal of the effect of dizocilpine. Haloperidol at 0.5 mg/kg/day and at 1 mg/kg/day induced catalepsy that was significantly alleviated (by ~50%) by co-treatment with MDL-100,907 but only at 0.5 mg/kg/day dose of haloperidol. Chronic haloperidol treatment, event at doses as low as 0.1 mg/kg/day induced a significant upregulation of the D2/3R in the striatum (by over 40% in the nucleus accumbens and over 20% in the caudate-putamen nuclei), that was not reversed by MDL-100,907. Finally, an upregulation of 5-HT2AR after chronic haloperidol treatment at a moderate dose only (0.25 mg/kg/day) was demonstrated in frontal cortical regions and the ventral tegmental area. Overall, a partial contribution of a 5-HT2AR antagonism to the efficacy and side-effect profile of antipsychotic agents is suggested.
Collapse
Affiliation(s)
- Stergios Tsartsalis
- Division of Adult Psychiatry, Department of Psychiatry, Geneva University Hospitals, Geneva, Switzerland. .,Division of Psychiatric Specialties, Department of Psychiatry, Geneva University Hospitals, Geneva, Switzerland.
| | - Benjamin B. Tournier
- grid.150338.c0000 0001 0721 9812Division of Adult Psychiatry, Department of Psychiatry, Geneva University Hospitals, Geneva, Switzerland
| | - Yesica Gloria
- grid.150338.c0000 0001 0721 9812Division of Adult Psychiatry, Department of Psychiatry, Geneva University Hospitals, Geneva, Switzerland
| | - Philippe Millet
- grid.150338.c0000 0001 0721 9812Division of Adult Psychiatry, Department of Psychiatry, Geneva University Hospitals, Geneva, Switzerland ,grid.8591.50000 0001 2322 4988Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Nathalie Ginovart
- grid.8591.50000 0001 2322 4988Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland ,grid.8591.50000 0001 2322 4988Department of Basic Neurosciences, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
34
|
Sonnenschein SF, Grace AA. Emerging therapeutic targets for schizophrenia: a framework for novel treatment strategies for psychosis. Expert Opin Ther Targets 2021; 25:15-26. [PMID: 33170748 PMCID: PMC7855878 DOI: 10.1080/14728222.2021.1849144] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 11/05/2020] [Indexed: 01/10/2023]
Abstract
Introduction: Antipsychotic drugs are central to the treatment of schizophrenia, but their limitations necessitate improved treatment strategies. Multiple lines of research have implicated glutamatergic dysfunction in the hippocampus as an early source of pathophysiology in schizophrenia. Novel compounds have been designed to treat glutamatergic dysfunction, but they have produced inconsistent results in clinical trials. Areas covered: This review discusses how the hippocampus is thought to drive psychotic symptoms through its influence on the dopamine system. It offers the reader an evaluation of proposed treatment strategies including direct modulation of GABA or glutamate neurotransmission or reducing the deleterious impact of stress on circuit development. Finally, we offer a perspective on aspects of future research that will advance our knowledge and may create new therapeutic opportunities. PubMed was searched for relevant literature between 2010 and 2020 and related studies. Expert opinion: Targeting aberrant excitatory-inhibitory neurotransmission in the hippocampus and its related circuits has the potential to alleviate symptoms and reduce the risk of transition to psychosis if implemented as an early intervention. Longitudinal multimodal brain imaging combined with mechanistic theories generated from animal models can be used to better understand the progression of hippocampal-dopamine circuit dysfunction and heterogeneity in treatment response.
Collapse
Affiliation(s)
| | - Anthony A. Grace
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
35
|
Kätzel D, Wolff AR, Bygrave AM, Bannerman DM. Hippocampal Hyperactivity as a Druggable Circuit-Level Origin of Aberrant Salience in Schizophrenia. Front Pharmacol 2020; 11:486811. [PMID: 33178010 PMCID: PMC7596262 DOI: 10.3389/fphar.2020.486811] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 09/18/2020] [Indexed: 01/21/2023] Open
Abstract
The development of current neuroleptics was largely aiming to decrease excessive dopaminergic signaling in the striatum. However, the notion that abnormal dopamine creates psychotic symptoms by causing an aberrant assignment of salience that drives maladaptive learning chronically during disease development suggests a therapeutic value of early interventions that correct salience-related neural processing. The mesolimbic dopaminergic output is modulated by several interconnected brain-wide circuits centrally involving the hippocampus and key relays like the ventral and associative striatum, ventral pallidum, amygdala, bed nucleus of the stria terminalis, nucleus reuniens, lateral and medial septum, prefrontal and cingulate cortex, among others. Unraveling the causal relationships between these circuits using modern neuroscience techniques holds promise for identifying novel cellular-and ultimately molecular-treatment targets for reducing transition to psychosis and symptoms of schizophrenia. Imaging studies in humans have implicated a hyperactivity of the hippocampus as a robust and early endophenotype in schizophrenia. Experiments in rodents, in turn, suggested that the activity of its output region-the ventral subiculum-may modulate dopamine release from ventral tegmental area (VTA) neurons in the ventral striatum. Even though these observations suggested a novel circuit-level target for anti-psychotic action, no therapy has yet been developed along this rationale. Recently evaluated treatment strategies-at least in part-target excess glutamatergic activity, e.g. N-acetyl-cysteine (NAC), levetiracetam, and mGluR2/3 modulators. We here review the evidence for the central implication of the hippocampus-VTA axis in schizophrenia-related pathology, discuss its symptom-related implications with a particular focus on aberrant assignment of salience, and evaluate some of its short-comings and prospects for drug discovery.
Collapse
Affiliation(s)
- Dennis Kätzel
- Institute for Applied Physiology, Ulm University, Ulm, Germany
| | - Amy R. Wolff
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Alexei M. Bygrave
- Department of Neuroscience, Johns Hopkins University, Baltimore, MD, United States
| | - David M. Bannerman
- Department of Experimental Psychology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
36
|
Kantrowitz JT, Grinband J, Goff DC, Lahti AC, Marder SR, Kegeles LS, Girgis RR, Sobeih T, Wall MM, Choo TH, Green MF, Yang YS, Lee J, Horga G, Krystal JH, Potter WZ, Javitt DC, Lieberman JA. Proof of mechanism and target engagement of glutamatergic drugs for the treatment of schizophrenia: RCTs of pomaglumetad and TS-134 on ketamine-induced psychotic symptoms and pharmacoBOLD in healthy volunteers. Neuropsychopharmacology 2020; 45:1842-1850. [PMID: 32403118 PMCID: PMC7608251 DOI: 10.1038/s41386-020-0706-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 04/12/2020] [Accepted: 05/06/2020] [Indexed: 12/15/2022]
Abstract
Glutamate neurotransmission is a prioritized target for antipsychotic drug development. Two metabotropic glutamate receptor 2/3 (mGluR2/3) agonists (pomaglumetad [POMA] and TS-134) were assessed in two Phase Ib proof of mechanism studies of comparable designs and using identical clinical assessments and pharmacoBOLD methodology. POMA was examined in a randomized controlled trial under double-blind conditions for 10-days at doses of 80 or 320 mg/d POMA versus placebo (1:1:1 ratio). The TS-134 trial was a randomized, single-blind, 6-day study of 20 or 60 mg/d TS-134 versus placebo (5:5:2 ratio). Primary outcomes were ketamine-induced changes in pharmacoBOLD in the dorsal anterior cingulate cortex (dACC) and symptoms reflected on the Brief Psychiatric Rating Scale (BPRS). Both trials were conducted contemporaneously. 95 healthy volunteers were randomized to POMA and 63 to TS-134. High-dose POMA significantly reduced ketamine-induced BPRS total symptoms within and between-groups (p < 0.01, d = -0.41; p = 0.04, d = -0.44, respectively), but neither POMA dose significantly suppressed ketamine-induced dACC pharmacoBOLD. In contrast, low-dose TS-134 led to moderate to large within and between group reductions in both BPRS positive symptoms (p = 0.02, d = -0.36; p = 0.008, d = -0.82, respectively) and dACC pharmacoBOLD (p = 0.004, d = -0.56; p = 0.079, d = -0.50, respectively) using pooled across-study placebo data. High-dose POMA exerted significant effects on clinical symptoms, but not on target engagement, suggesting a higher dose may yet be needed, while the low dose of TS-134 showed evidence of symptom reduction and target engagement. These results support further investigation of mGluR2/3 and other glutamate-targeted treatments for schizophrenia.
Collapse
Affiliation(s)
- Joshua T. Kantrowitz
- grid.21729.3f0000000419368729Columbia University, New York, NY USA ,grid.413734.60000 0000 8499 1112New York State Psychiatric Institute, New York, NY USA ,grid.250263.00000 0001 2189 4777Nathan Kline Institute, Orangeburg, NY USA
| | - Jack Grinband
- grid.21729.3f0000000419368729Columbia University, New York, NY USA ,grid.413734.60000 0000 8499 1112New York State Psychiatric Institute, New York, NY USA
| | - Donald C. Goff
- grid.250263.00000 0001 2189 4777Nathan Kline Institute, Orangeburg, NY USA ,grid.240324.30000 0001 2109 4251NYU Langone Medical Center, New York, NY USA
| | - Adrienne C. Lahti
- grid.265892.20000000106344187University of Alabama at Birmingham, Birmingham, AL USA
| | | | - Lawrence S. Kegeles
- grid.21729.3f0000000419368729Columbia University, New York, NY USA ,grid.413734.60000 0000 8499 1112New York State Psychiatric Institute, New York, NY USA
| | - Ragy R. Girgis
- grid.21729.3f0000000419368729Columbia University, New York, NY USA ,grid.413734.60000 0000 8499 1112New York State Psychiatric Institute, New York, NY USA
| | - Tarek Sobeih
- grid.250263.00000 0001 2189 4777Nathan Kline Institute, Orangeburg, NY USA
| | - Melanie M. Wall
- grid.21729.3f0000000419368729Columbia University, New York, NY USA ,grid.413734.60000 0000 8499 1112New York State Psychiatric Institute, New York, NY USA
| | - Tse-Hwei Choo
- grid.21729.3f0000000419368729Columbia University, New York, NY USA ,grid.413734.60000 0000 8499 1112New York State Psychiatric Institute, New York, NY USA
| | | | - Yvonne S. Yang
- grid.19006.3e0000 0000 9632 6718UCLA, Los Angeles, CA USA
| | - Junghee Lee
- grid.19006.3e0000 0000 9632 6718UCLA, Los Angeles, CA USA
| | - Guillermo Horga
- grid.21729.3f0000000419368729Columbia University, New York, NY USA ,grid.413734.60000 0000 8499 1112New York State Psychiatric Institute, New York, NY USA
| | - John H. Krystal
- grid.47100.320000000419368710Yale University School of Medicine, New Haven, CT USA
| | - William Z. Potter
- grid.94365.3d0000 0001 2297 5165National Institutes of Health, Bethesda, MD USA
| | - Daniel C. Javitt
- grid.21729.3f0000000419368729Columbia University, New York, NY USA ,grid.413734.60000 0000 8499 1112New York State Psychiatric Institute, New York, NY USA ,grid.250263.00000 0001 2189 4777Nathan Kline Institute, Orangeburg, NY USA
| | - Jeffrey A. Lieberman
- grid.21729.3f0000000419368729Columbia University, New York, NY USA ,grid.413734.60000 0000 8499 1112New York State Psychiatric Institute, New York, NY USA
| |
Collapse
|
37
|
Leicht G, Andreou C, Nafe T, Nägele F, Rauh J, Curic S, Schauer P, Schöttle D, Steinmann S, Mulert C. Alterations of oscillatory neuronal activity during reward processing in schizophrenia. J Psychiatr Res 2020; 129:80-87. [PMID: 32619750 DOI: 10.1016/j.jpsychires.2020.05.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/29/2020] [Accepted: 05/29/2020] [Indexed: 11/30/2022]
Abstract
OBJECTIVES Reward system dysfunctions are considered to be a pathophysiological mechanism in schizophrenia. Electrophysiological studies of reward system functions have identified frequency-specific brain networks for the processing of positive (high-beta frequency) and negative (theta frequency) events. Remarkably, midbrain dopaminergic signalling also includes theta and high-beta frequency modes, which have been assumed to reflect tonic and phasic dopamine responses, respectively. The aim of the present study was to identify alterations of oscillatory responses to reward feedback in patients with schizophrenia. METHODS Seventeen patients with schizophrenia and 18 healthy controls performed a gambling task during recording of 64-channel electroencephalography. The theta and high-beta band total power were investigated in response to feedback events depending on feedback valence (loss or gain) and magnitude (5 vs. 25 points). RESULTS Both the increase of theta oscillatory activity in response to loss feedback (compared to gain feedback) and the increase of high-beta oscillatory activity in response to gain feedback (compared to loss feedback) were reduced in patients. The difference in high-beta responses to gain versus loss feedback in patients was associated with the severity of negative symptoms. CONCLUSIONS Our findings are consistent with current models of reward system dysfunction in schizophrenia, and indicate deficits in both cortical tonic and subcortical phasic dopamine activity, consistent with the complex dopaminergic abnormalities in schizophrenia.
Collapse
Affiliation(s)
- Gregor Leicht
- Department of Psychiatry and Psychotherapy, Psychiatry Neuroimaging Branch, University Medical Center Hamburg-Eppendorf, Martinistr. 52, D-20246, Hamburg, Germany.
| | - Christina Andreou
- Department of Psychiatry and Psychotherapy, Psychiatry Neuroimaging Branch, University Medical Center Hamburg-Eppendorf, Martinistr. 52, D-20246, Hamburg, Germany; Department of Psychiatry and Psychotherapy, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Till Nafe
- Department of Psychiatry and Psychotherapy, Psychiatry Neuroimaging Branch, University Medical Center Hamburg-Eppendorf, Martinistr. 52, D-20246, Hamburg, Germany
| | - Felix Nägele
- Department of Psychiatry and Psychotherapy, Psychiatry Neuroimaging Branch, University Medical Center Hamburg-Eppendorf, Martinistr. 52, D-20246, Hamburg, Germany
| | - Jonas Rauh
- Department of Psychiatry and Psychotherapy, Psychiatry Neuroimaging Branch, University Medical Center Hamburg-Eppendorf, Martinistr. 52, D-20246, Hamburg, Germany
| | - Stjepan Curic
- Department of Psychiatry and Psychotherapy, Psychiatry Neuroimaging Branch, University Medical Center Hamburg-Eppendorf, Martinistr. 52, D-20246, Hamburg, Germany
| | - Paul Schauer
- Department of Psychiatry and Psychotherapy, Psychiatry Neuroimaging Branch, University Medical Center Hamburg-Eppendorf, Martinistr. 52, D-20246, Hamburg, Germany
| | - Daniel Schöttle
- Department of Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Saskia Steinmann
- Department of Psychiatry and Psychotherapy, Psychiatry Neuroimaging Branch, University Medical Center Hamburg-Eppendorf, Martinistr. 52, D-20246, Hamburg, Germany
| | - Christoph Mulert
- Department of Psychiatry and Psychotherapy, Psychiatry Neuroimaging Branch, University Medical Center Hamburg-Eppendorf, Martinistr. 52, D-20246, Hamburg, Germany; Center of Psychiatry, Justus-Liebig University, Klinikstr. 36, 35385, Giessen, Germany
| |
Collapse
|
38
|
Poddar I, Callahan PM, Hernandez CM, Pillai A, Yang X, Bartlett MG, Terry AV. Oral quetiapine treatment results in time-dependent alterations of recognition memory and brain-derived neurotrophic factor-related signaling molecules in the hippocampus of rats. Pharmacol Biochem Behav 2020; 197:172999. [PMID: 32702397 DOI: 10.1016/j.pbb.2020.172999] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/13/2020] [Accepted: 07/13/2020] [Indexed: 11/16/2022]
Abstract
Antipsychotic drugs (APDs) have a variety of important therapeutic applications for neuropsychiatric disorders. However, they are routinely prescribed off-label across all age categories, a controversial practice given their potential for producing metabolic and extrapyramidal side effects. Evidence also suggests that chronic treatment with some APDs may lead to impairments in cognition and decreases in brain volume, although these findings are controversial. The purpose of the studies described here was to evaluate one of the most commonly prescribed APDs, quetiapine, for chronic effects on recognition memory, brain-derived neurotrophic factor (BDNF), its precursor proBDNF, as well as relevant downstream signaling molecules that are known to influence neuronal plasticity and cognition. Multiple cohorts of adult rats were treated with quetiapine (25.0 mg/kg/day) for 30 or 90 days in their drinking water then evaluated for drug effects on motor function in a catalepsy assessment, recognition memory in a spontaneous novel object recognition (NOR) task, and BDNF-related signaling molecules in the post mortem hippocampus via Western Blot. The results indicated that oral quetiapine at a dose that did not induce catalepsy, led to time-dependent impairments in NOR performance, increases in the proBDNF/BDNF ratio, and decreases in Akt and CREB phosphorylation in the hippocampus. These results indicate that chronic treatment with quetiapine has the potential to adversely affect recognition memory and neurotrophin-related signaling molecules that support synaptic plasticity and cognitive function. Given the widespread use this APD across multiple conditions and patient populations, such long-term effects observed in animals should be considered.
Collapse
Affiliation(s)
- Indrani Poddar
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - Patrick M Callahan
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America; Small Animal Behavior Core, Augusta University, Augusta, GA 30912, United States of America
| | - Caterina M Hernandez
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - Anilkumar Pillai
- Department of Psychiatry and Health Behavior, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - Xiangkun Yang
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia College of Pharmacy, Athens, GA 30602, United States of America
| | - Michael G Bartlett
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia College of Pharmacy, Athens, GA 30602, United States of America
| | - Alvin V Terry
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America; Small Animal Behavior Core, Augusta University, Augusta, GA 30912, United States of America.
| |
Collapse
|
39
|
Issy AC, Pedrazzi JFC, van Oosten ABS, Checheto T, Silva RR, Noël F, Del-Bel E. Effects of Doxycycline in Swiss Mice Predictive Models of Schizophrenia. Neurotox Res 2020; 38:1049-1060. [PMID: 32929685 DOI: 10.1007/s12640-020-00268-z] [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: 12/26/2019] [Revised: 08/03/2020] [Accepted: 08/04/2020] [Indexed: 01/14/2023]
Abstract
Schizophrenia patients show very complex symptoms in several psychopathological domains. Some of these symptoms remain poorly treated. Therefore, continued effort is needed to find novel pharmacological strategies for improving schizophrenia symptoms. Recently, minocycline, a second-generation tetracycline, has been suggested as an adjunctive treatment for schizophrenia. The antipsychotic-like effect of doxycycline, a minocycline analog, was investigated here. We found that both minocycline and doxycycline prevented amphetamine-induced prepulse inhibition (PPI) disruption. However, neither of them blocked MK801-induced effects, albeit doxycycline had a modest impact against ketamine-induced effects. Neither c-Fos nor nNOS expression, which was evaluated in limbic regions, were modified after acute or sub-chronic treatment with doxycycline. Therefore, apomorphine inducing either PPI disruption and climbing behavior was not prevented by doxycycline. This result discards a direct blockade of D2-like receptors, also suggested by the lack of doxycycline cataleptic-induced effect. Contrasting, doxycycline prevented SKF 38393-induced effects, suggesting a preferential doxycycline action at D1-like rather than D2-like receptors. However, doxycycline did not bind to the orthosteric sites of D1, D2, D3, D4, 5-HT2A, 5-HT1A, and A2A receptors suggesting no direct modulation of these receptors. Our data corroborate the antipsychotic-like effect of doxycycline. However, these effects are probably not mediated by doxycycline direct interaction with classical receptors enrolled in the antipsychotic effect.
Collapse
Affiliation(s)
- Ana Carolina Issy
- Dental School of Ribeirão Preto, Department of Basic and Oral Biology, University of São Paulo (USP), Ribeirão Preto, SP, Brazil.,Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), USP, São Paulo, Brazil.,Medical School of Ribeirão Preto, Department of Neuroscience and Behavior Sciences, USP, Ribeirão Preto, SP, Brazil
| | - João Francisco C Pedrazzi
- Dental School of Ribeirão Preto, Department of Basic and Oral Biology, University of São Paulo (USP), Ribeirão Preto, SP, Brazil.,Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), USP, São Paulo, Brazil.,Medical School of Ribeirão Preto, Department of Neuroscience and Behavior Sciences, USP, Ribeirão Preto, SP, Brazil
| | - Anna Beatriz Saito van Oosten
- Dental School of Ribeirão Preto, Department of Basic and Oral Biology, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Thiago Checheto
- Dental School of Ribeirão Preto, Department of Basic and Oral Biology, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Rafaela R Silva
- Laboratory of Biochemical and Molecular Pharmacology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - François Noël
- Laboratory of Biochemical and Molecular Pharmacology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Elaine Del-Bel
- Dental School of Ribeirão Preto, Department of Basic and Oral Biology, University of São Paulo (USP), Ribeirão Preto, SP, Brazil. .,Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), USP, São Paulo, Brazil. .,Medical School of Ribeirão Preto, Department of Neuroscience and Behavior Sciences, USP, Ribeirão Preto, SP, Brazil. .,Medical School of Ribeirão Preto, Department of Physiology, USP, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
40
|
Abstract
Schizophrenia is a major mental illness associated with profound disability. Current treatments for schizophrenia (antipsychotics) all have a similar mechanism of action and are primarily dopamine type 2 receptor (D2R) antagonists. Antipsychotics are not fully effective for the majority of schizophrenia patients, suggesting the need for alternative approaches. The primary focus of this review is to assess the evidence for the role of the serotonin type 2A receptor (5-HT2AR) in schizophrenia. Topics include an overview of 5-HT2AR physiology and pathophysiology in schizophrenia, 5-HT2AR interaction with other neurotransmitter systems, including the glutamatergic system, a review of the 5-HT2AR/d-lysergic acid diethylamide (LSD) model of schizophrenia, a contrast of the 5-HT2AR and glutamatergic models of schizophrenia, and finally, a review of Food and Drug Administration (FDA)-approved and investigational 5-HT2AR-modulating compounds. Recent studies with lumeteperone, pimavanserin, and roluperidone are highlighted.
Collapse
|
41
|
Afonso AC, Pacheco FD, Canever L, Wessler PG, Mastella GA, Godoi AK, Hubbe I, Bischoff LM, Bialecki AVS, Zugno AI. Schizophrenia-like behavior is not altered by melatonin supplementation in rodents. AN ACAD BRAS CIENC 2020; 92:e20190981. [PMID: 32844989 DOI: 10.1590/0001-3765202020190981] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 12/06/2019] [Indexed: 11/22/2022] Open
Abstract
An emerging area in schizophrenia research focuses on the impact of immunomodulatory drugs such as melatonin, which have played important roles in many biological systems and functions, and appears to be promising. The objective was to evaluate the effect of melatonin on behavioral parameters in an animal model of schizophrenia. For this, Wistar rats were divided and used in two different protocols. In the prevention protocol, the animals received 1 or 10mg/kg of melatonin or water for 14 days, and between the 8th and 14th day they received ketamine or saline. In the reversal protocol, the opposite occurred. On the 14th day, the animals underwent behavioral tests: locomotor activity and prepulse inhibition task. In both protocols, the results revealed that ketamine had effects on locomotor activity and prepulse inhibition, confirming the validity of ketamine construction as a good animal model of schizophrenia. However, at least at the doses used, melatonin was not able to reverse/prevent ketamine damage. More studies are necessary to evaluate the role of melatonin as an adjuvant treatment in psychiatric disorders.
Collapse
Affiliation(s)
- Arlindo C Afonso
- Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Felipe D Pacheco
- Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Lara Canever
- Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Patricia G Wessler
- Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Gustavo A Mastella
- Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Amanda K Godoi
- Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Isabela Hubbe
- Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Laura M Bischoff
- Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Alex Victor S Bialecki
- Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Alexandra I Zugno
- Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| |
Collapse
|
42
|
Boloc D, Rodríguez N, Torres T, García-Cerro S, Parellada M, Saiz-Ruiz J, Cuesta MJ, Bernardo M, Gassó P, Lafuente A, Mas S, Arnaiz JA. Identifying key transcription factors for pharmacogenetic studies of antipsychotics induced extrapyramidal symptoms. Psychopharmacology (Berl) 2020; 237:2151-2159. [PMID: 32382784 DOI: 10.1007/s00213-020-05526-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 04/13/2020] [Indexed: 12/14/2022]
Abstract
INTRODUCTION We explore the transcription factors involved in the molecular mechanism of antipsychotic (AP)-induced acute extrapyramidalsymptoms (EPS) in order to identify new candidate genes for pharmacogenetic studies. METHODS Protein-protein interaction (PPI) networks previously created from three pharmacogenomic models (in vitro, animal, and peripheral blood inhumans) were used to, by means of several bioinformatic tools; identify key transcription factors (TFs) that regulate each network. Once the TFs wereidentified, SNPs disrupting the binding sites (TFBS) of these TFs in the genes of each network were selected for genotyping. Finally, SNP-basedassociations with EPS were analyzed in a sample of 356 psychiatric patients receiving AP. RESULTS Our analysis identified 33 TFs expressed in the striatum, and 125 SNPs disrupting TFBS in 50 genes of our initial networks. Two SNPs (rs938112,rs2987902) in two genes (LSMAP and ABL1) were significantly associated with AP induced EPS (p < 0.001). These SNPs disrupt TFBS regulated byPOU2F1. CONCLUSION Our results highlight the possible role of the disruption of TFBS by SNPs in the pharmacological response to AP.
Collapse
Affiliation(s)
- Daniel Boloc
- Department of Medicine, University of Barcelona, Barcelona, Spain
| | | | - Teresa Torres
- Dept. Clinical Foundations, Pharmacology Unit, University of Barcelona, Barcelona, Spain
| | - Susana García-Cerro
- Dept. Clinical Foundations, Pharmacology Unit, University of Barcelona, Barcelona, Spain
| | - Mara Parellada
- Child and Adolescent Psychiatry Department, Hospital General Universitario Gregorio Marañón, School of Medicine, Universidad Complutense, IiSGM, Madrid, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, Madrid, Spain
| | - Jeronimo Saiz-Ruiz
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, Madrid, Spain
- Hospital Ramon y Cajal, Universidad de Alcala, IRYCIS, Madrid, Spain
| | - Manuel J Cuesta
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, Madrid, Spain
- Department of Psychiatry, Complejo Hospitalario de Navarra. Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Miquel Bernardo
- Department of Medicine, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, Madrid, Spain
- Barcelona Clínic Schizophrenia Unit, Hospital Clínic de Barcelona, Barcelona, Spain
- Spain The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Patricia Gassó
- Dept. Clinical Foundations, Pharmacology Unit, University of Barcelona, Barcelona, Spain
- Spain The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Amalia Lafuente
- Dept. Clinical Foundations, Pharmacology Unit, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, Madrid, Spain
- Spain The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Sergi Mas
- Dept. Clinical Foundations, Pharmacology Unit, University of Barcelona, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, Madrid, Spain.
- Spain The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain.
| | - Joan Albert Arnaiz
- Dept. Clinical Foundations, Pharmacology Unit, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
43
|
Young JW, Roberts BZ, Breier M, Swerdlow NR. Amphetamine improves rat 5-choice continuous performance test (5C-CPT) irrespective of concurrent low-dose haloperidol treatment. Psychopharmacology (Berl) 2020; 237:1959-1972. [PMID: 32318751 DOI: 10.1007/s00213-020-05511-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 03/26/2020] [Indexed: 12/22/2022]
Abstract
RATIONALE Cognitive dysfunction mediates functional impairment in patients with schizophrenia, necessitating the timely development of pro-cognitive therapeutics. An important initial step in this process is to establish what, if any, pro-cognitive agents and associated mechanisms can be identified using cross-species translational paradigms. For example, attentional deficits-a core feature of schizophrenia-can be measured across species using the 5-choice continuous performance test (5C-CPT). The psychostimulant, amphetamine, improves human and rodent 5C-CPT performance. OBJECTIVE Here, we tested whether amphetamine would similarly improve 5C-CPT performance in the presence of dopamine D2 receptor blockade, since pro-cognitive treatments in schizophrenia would virtually always be used in conjunction with D2 receptor antagonists. METHODS We established the dose-response effects of amphetamine (0, 0.1, 0.3, or 1.0 mg/kg) and haloperidol (0, 3.2, 10, or 32 μg/kg) on 5C-CPT performance in Long Evans rats, and then tested an amphetamine (0.3 mg/kg) × haloperidol (10 μg/kg) interaction; the low dose was chosen because higher doses exerted deleterious non-specific effects on performance. RESULTS Amphetamine improved 5C-CPT performance in poorly performing rats by increasing target detection, independent of haloperidol pretreatment. CONCLUSIONS The pro-attentional effects of amphetamine were most likely mediated by dopamine release at D1-family receptors, since they persisted in the presence of acute D2 blockade. Alternative explanations for these findings are also discussed, as are their potential implications for future pro-cognitive therapeutics in schizophrenia.
Collapse
Affiliation(s)
- Jared W Young
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA, 92093-0804, USA. .,Research Service, VA San Diego Healthcare System, San Diego, CA, USA.
| | - Benjamin Z Roberts
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA, 92093-0804, USA
| | - Michelle Breier
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA, 92093-0804, USA
| | - Neal R Swerdlow
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA, 92093-0804, USA
| |
Collapse
|
44
|
Kantrowitz JT, Javitt DC, Freedman R, Sehatpour P, Kegeles LS, Carlson M, Sobeih T, Wall MM, Choo TH, Vail B, Grinband J, Lieberman JA. Double blind, two dose, randomized, placebo-controlled, cross-over clinical trial of the positive allosteric modulator at the alpha7 nicotinic cholinergic receptor AVL-3288 in schizophrenia patients. Neuropsychopharmacology 2020; 45:1339-1345. [PMID: 32015461 PMCID: PMC7298033 DOI: 10.1038/s41386-020-0628-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 01/06/2020] [Accepted: 01/26/2020] [Indexed: 11/09/2022]
Abstract
Despite their theoretical rationale, nicotinic alpha-7 acetylcholine (nα7) receptor agonists, have largely failed to demonstrate efficacy in placebo-controlled trials in schizophrenia. AVL-3288 is a nα7 positive allosteric modulator (PAM), which is only active in the presence of the endogenous ligand (acetylcholine), and thus theoretically less likely to cause receptor desensitization. We evaluated the efficacy of AVL-3288 in a Phase 1b, randomized, double-blind, placebo-controlled, triple cross-over study. Twenty-four non-smoking, medicated, outpatients with schizophrenia or schizoaffective disorder and a Repeatable Battery for the Assessment of Neuropsychological Status (RBANS) ≥62 were randomized. Each subject received 5 days of AVL-3288 (10, 30 mg) and placebo across three separate treatment weeks. The primary outcome measure was the RBANS total scale score, with auditory P50 evoked potential suppression the key target engagement biomarker. Secondary outcome measures include task-based fMRI (RISE task), mismatch negativity, the Scale for the Assessment of Negative Symptoms of Schizophrenia (SANS) and the Brief Psychiatric Rating Scale (BPRS). Twenty-four subjects were randomized and treated without any clinically significant treatment emergent adverse effects. Baseline RBANS (82 ± 17) and BPRS (41 ± 13) scores were consistent with moderate impairment. Primary outcomes were negative, with non-significant worsening for both active groups vs. placebo in the P50 and minimal between group changes on the RBANS. In conclusion, the results did not indicate efficacy of the compound, consistent with most prior results for the nα7 target.
Collapse
Affiliation(s)
- Joshua T. Kantrowitz
- 0000000419368729grid.21729.3fColumbia University, New York, USA ,0000 0000 8499 1112grid.413734.6New York State Psychiatric Institute, New York, USA ,0000 0001 2189 4777grid.250263.0Nathan Kline Institute, Orangeburg, USA
| | - Daniel C. Javitt
- 0000000419368729grid.21729.3fColumbia University, New York, USA ,0000 0000 8499 1112grid.413734.6New York State Psychiatric Institute, New York, USA ,0000 0001 2189 4777grid.250263.0Nathan Kline Institute, Orangeburg, USA
| | | | - Pejman Sehatpour
- 0000000419368729grid.21729.3fColumbia University, New York, USA ,0000 0000 8499 1112grid.413734.6New York State Psychiatric Institute, New York, USA ,0000 0001 2189 4777grid.250263.0Nathan Kline Institute, Orangeburg, USA
| | - Lawrence S. Kegeles
- 0000000419368729grid.21729.3fColumbia University, New York, USA ,0000 0000 8499 1112grid.413734.6New York State Psychiatric Institute, New York, USA
| | - Marlene Carlson
- 0000000419368729grid.21729.3fColumbia University, New York, USA ,0000 0000 8499 1112grid.413734.6New York State Psychiatric Institute, New York, USA
| | - Tarek Sobeih
- 0000 0001 2189 4777grid.250263.0Nathan Kline Institute, Orangeburg, USA
| | - Melanie M. Wall
- 0000000419368729grid.21729.3fColumbia University, New York, USA ,0000 0000 8499 1112grid.413734.6New York State Psychiatric Institute, New York, USA
| | - Tse-Hwei Choo
- 0000000419368729grid.21729.3fColumbia University, New York, USA ,0000 0000 8499 1112grid.413734.6New York State Psychiatric Institute, New York, USA
| | - Blair Vail
- 0000 0000 8499 1112grid.413734.6New York State Psychiatric Institute, New York, USA
| | - Jack Grinband
- 0000000419368729grid.21729.3fColumbia University, New York, USA ,0000 0000 8499 1112grid.413734.6New York State Psychiatric Institute, New York, USA
| | - Jeffrey A. Lieberman
- 0000000419368729grid.21729.3fColumbia University, New York, USA ,0000 0000 8499 1112grid.413734.6New York State Psychiatric Institute, New York, USA
| |
Collapse
|
45
|
Sonnenschein SF, Gomes FV, Grace AA. Dysregulation of Midbrain Dopamine System and the Pathophysiology of Schizophrenia. Front Psychiatry 2020; 11:613. [PMID: 32719622 PMCID: PMC7350524 DOI: 10.3389/fpsyt.2020.00613] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 06/12/2020] [Indexed: 11/25/2022] Open
Abstract
Dysregulation of the dopamine system is central to many models of the pathophysiology of psychosis in schizophrenia. However, emerging evidence suggests that this dysregulation is driven by the disruption of upstream circuits that provide afferent control of midbrain dopamine neurons. Furthermore, stress can profoundly disrupt this regulatory circuit, particularly when it is presented at critical vulnerable prepubertal time points. This review will discuss the dopamine system and the circuits that regulate it, focusing on the hippocampus, medial prefrontal cortex, thalamic nuclei, and medial septum, and the impact of stress. A greater understanding of the regulation of the dopamine system and its disruption in schizophrenia may provide a more complete neurobiological framework to interpret clinical findings and develop novel treatments.
Collapse
Affiliation(s)
- Susan F. Sonnenschein
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Felipe V. Gomes
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Anthony A. Grace
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
46
|
Go J, Ryu YK, Park HY, Choi DH, Choi YK, Hwang DY, Lee CH, Kim KS. NQO1 regulates pharmaco-behavioral effects of d-amphetamine in striatal dopaminergic system in mice. Neuropharmacology 2020; 170:108039. [PMID: 32165217 DOI: 10.1016/j.neuropharm.2020.108039] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 02/11/2020] [Accepted: 03/04/2020] [Indexed: 11/18/2022]
Abstract
The NAD(P)H:quinone oxidoreductase 1 (NQO1) gene encodes a cytosolic flavoenzyme that catalyzes the two-electron reduction of quinones to hydroquinones. A polymorphic form of NQO1 is associated with mood disorders such as schizophrenia. However, the role of NQO1 in dopaminergic system has not yet been elucidated. To determine the role of NQO1 in the dopaminergic system, we investigated pharmaco-behavioral effects of d-amphetamine using NQO1-deficienct mice. According to our comparative study involving NQO1+/+ and NQO1-/- mice, NQO1 deficiency increased d-amphetamine-induced psychomotor activity and psychological dependency compared to wild-type mice. Basal and d-amphetamine-induced dopamine levels were also enhanced by NQO1 deficiency. In NQO1-/- mice, neural activation induced by d-amphetamine was higher in dorsolateral striatum, but not in dorsomedial and ventral striata. Although protein level of CaMKIIα, which is a key player in amphetamine-induced dopamine efflux, was decreased in striata of NQO1-/- mice, phosphorylation of CaMKIIα was markedly enhanced in NQO1-/- mice compared to wild-type mice. Interestingly, experiments with pharmacological antagonist showed that D2 antagonist-induced suppression of locomotion required activation of NQO1. Moreover, the rewarding effect in response to D1 agonist was increased by NQO1 deficiency. These results suggest that striatal NQO1 is of considerable interest to understand the mechanism of dopaminergic regulation of psychiatric disorders.
Collapse
Affiliation(s)
- Jun Go
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea; Department of Biomaterials Science, College of Natural Resources and Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang, 50463, Republic of Korea
| | - Young-Kyoung Ryu
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea; College of Biosciences & Biotechnology, Chung-Nam National University, Daejeon, 34134, Republic of Korea
| | - Hye-Yeon Park
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea; Department of Brain & Cognitive Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Dong-Hee Choi
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Young-Keun Choi
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Dae Youn Hwang
- Department of Biomaterials Science, College of Natural Resources and Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang, 50463, Republic of Korea
| | - Chul-Ho Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea; Department of Functional Genomics, University of Science and Technology, Daejeon, 34113, Republic of Korea.
| | - Kyoung-Shim Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea; Department of Functional Genomics, University of Science and Technology, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
47
|
Venkataramaiah C. Modulations in the ATPases during ketamine-induced schizophrenia and regulatory effect of "3-(3, 4-dimethoxy phenyl) -1- (4-methoxyphenyl) prop-2-en-1-one": an in vivo and in silico studies. J Recept Signal Transduct Res 2020; 40:148-156. [PMID: 32009493 DOI: 10.1080/10799893.2020.1720242] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Schizophrenia is a devastating illness and displays a wide range of psychotic symptoms. Accumulating evidence indicate impairment of bioenergetic pathways including energy storage and usage in the pathogenesis of schizophrenia. Although well-established synthetic drugs are being used for the management of schizophrenia, most of them have several adverse effects. Hence, natural products derived from medicinal plants represent a continuous major source for ethnomedicine-derived pharmaceuticals for different neurological disorders including schizophrenia. In the present study, we have investigated the neuroprotective effect of the novel bioactive compound i.e. "3-(3,4-dimethoxy phenyl) -1- (4-methoxyphenyl) prop-2-en-1-one" of Celastrus paniculata against ketamine-induced schizophrenia with particular reference to the activities of ATPase using in vivo and in silico methods. Ketamine-induced schizophrenia caused significant reduction in the activities of all three ATPases (Na+/K+, Ca2+ and Mg2+) in different regions of brain which reflects the decreased turnover of ATP, presumably due to the inhibition of oxidoreductase system and uncoupling of the same from the electron transport system. On par with the reference compound, clozapine, the activity levels of all three ATPases were restored to normal after pretreatment with the compound suggesting recovery of energy loss that was occurred during ketamine-induced schizophrenia. Besides, the compound has shown strong interaction and exhibited highest binding energies against all the three ATPases with a lowest inhibition constant value than the clozapine. The results of the present study clearly imply that the compound exhibit significant neuroprotective and antischizophrenic effect by modulating bioenergietic pathways that were altered during induced schizophrenia.
Collapse
Affiliation(s)
- Chintha Venkataramaiah
- Division of Molecular Biology, Department of Zoology, Sri Venkateswara University, Tirupati, India
| |
Collapse
|
48
|
Poddar I, Callahan PM, Hernandez CM, Pillai A, Yang X, Bartlett MG, Terry AV. Chronic oral treatment with risperidone impairs recognition memory and alters brain-derived neurotrophic factor and related signaling molecules in rats. Pharmacol Biochem Behav 2020; 189:172853. [PMID: 31945381 DOI: 10.1016/j.pbb.2020.172853] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 01/09/2020] [Accepted: 01/12/2020] [Indexed: 01/09/2023]
Abstract
Antipsychotic drugs (APDs) are essential for the treatment of schizophrenia and other neuropsychiatric illnesses such as bipolar disease. However, they are also extensively prescribed off-label for many other conditions, a practice that is controversial given their potential for long-term side effects. There is clinical and preclinical evidence that chronic treatment with some APDs may lead to impairments in cognition and decreases in brain volume, although the molecular mechanisms of these effects are unknown. The purpose of the rodent studies described here was to evaluate a commonly prescribed APD, risperidone, for chronic effects on recognition memory, brain-derived neurotrophic factor (BDNF), its precursor proBDNF, as well as relevant downstream signaling molecules that are known to influence neuronal plasticity and cognition. Multiple cohorts of adult rats were treated with risperidone (2.5 mg/kg/day) or vehicle (dilute acetic acid solution) in their drinking water for 30 or 90 days. Subjects were then evaluated for drug effects on recognition memory in a spontaneous novel object recognition task and protein levels of BDNF-related signaling molecules in the hippocampus and prefrontal cortex. The results indicated that depending on the treatment period, a therapeutically relevant daily dose of risperidone impaired recognition memory and increased the proBDNF/BDNF ratio in the hippocampus and prefrontal cortex. Risperidone treatment also led to a decrease in Akt and CREB phosphorylation in the prefrontal cortex. These results indicate that chronic treatment with a commonly prescribed APD, risperidone, has the potential to adversely affect recognition memory and neurotrophin-related signaling molecules that support synaptic plasticity and cognitive function.
Collapse
Affiliation(s)
- Indrani Poddar
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - Patrick M Callahan
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America.; Small Animal Behavior Core, Augusta University, Augusta, GA 30912, United States of America
| | - Caterina M Hernandez
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - Anilkumar Pillai
- Department of Psychiatry and Health Behavior, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - Xiangkun Yang
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia College of Pharmacy, Athens, GA 30607, United States of America
| | - Michael G Bartlett
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia College of Pharmacy, Athens, GA 30607, United States of America
| | - Alvin V Terry
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America.; Small Animal Behavior Core, Augusta University, Augusta, GA 30912, United States of America.
| |
Collapse
|
49
|
Fyfe TJ, Kellam B, Sykes DA, Capuano B, Scammells PJ, Lane JR, Charlton SJ, Mistry SN. Structure-Kinetic Profiling of Haloperidol Analogues at the Human Dopamine D 2 Receptor. J Med Chem 2019; 62:9488-9520. [PMID: 31580666 DOI: 10.1021/acs.jmedchem.9b00864] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Haloperidol is a typical antipsychotic drug (APD) associated with an increased risk of extrapyramidal side effects (EPSs) and hyperprolactinemia relative to atypical APDs such as clozapine. Both drugs are dopamine D2 receptor (D2R) antagonists, with contrasting kinetic profiles. Haloperidol displays fast association/slow dissociation at the D2R, whereas clozapine exhibits relatively slow association/fast dissociation. Recently, we have provided evidence that slow dissociation from the D2R predicts hyperprolactinemia, whereas fast association predicts EPS. Unfortunately, clozapine can cause severe side effects independent of its D2R action. Our results suggest an optimal kinetic profile for D2R antagonist APDs that avoids EPS. To begin exploring this hypothesis, we conducted a structure-kinetic relationship study of haloperidol and revealed that subtle structural modifications dramatically change binding kinetic rate constants, affording compounds with a clozapine-like kinetic profile. Thus, optimization of these kinetic parameters may allow development of novel APDs based on the haloperidol scaffold with improved side-effect profiles.
Collapse
Affiliation(s)
- Tim J Fyfe
- School of Pharmacy, Centre for Biomolecular Sciences , University of Nottingham , Nottingham NG7 2RD , U.K
| | - Barrie Kellam
- School of Pharmacy, Centre for Biomolecular Sciences , University of Nottingham , Nottingham NG7 2RD , U.K
| | - David A Sykes
- School of Life Sciences, Queen's Medical Centre , University of Nottingham , Nottingham NG7 2UH , U.K.,Centre of Membrane Protein and Receptors , University of Nottingham , Nottingham NG7 2UH , U.K
| | | | | | - J Robert Lane
- School of Life Sciences, Queen's Medical Centre , University of Nottingham , Nottingham NG7 2UH , U.K.,Centre of Membrane Protein and Receptors , University of Nottingham , Nottingham NG7 2UH , U.K
| | - Steven J Charlton
- School of Life Sciences, Queen's Medical Centre , University of Nottingham , Nottingham NG7 2UH , U.K.,Centre of Membrane Protein and Receptors , University of Nottingham , Nottingham NG7 2UH , U.K.,Excellerate Bioscience Ltd., BioCity , Nottingham NG1 1GF , U.K
| | - Shailesh N Mistry
- School of Pharmacy, Centre for Biomolecular Sciences , University of Nottingham , Nottingham NG7 2RD , U.K
| |
Collapse
|
50
|
Insights on current and novel antipsychotic mechanisms from the MAM model of schizophrenia. Neuropharmacology 2019; 163:107632. [PMID: 31077730 DOI: 10.1016/j.neuropharm.2019.05.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 04/25/2019] [Accepted: 05/07/2019] [Indexed: 12/18/2022]
Abstract
Current antipsychotic drugs (APDs) act on D2 receptors, and preclinical studies demonstrate that repeated D2 antagonist administration downregulates spontaneously active DA neurons by producing overexcitation-induced inactivation of firing (depolarization block). Animal models of schizophrenia based on the gestational MAM administration produces offspring with adult phenotypes consistent with schizophrenia, including ventral hippocampal hyperactivity and a DA neuron overactivity. The MAM model reveals that APDs act differently in a hyperdopamineregic system compared to a normal one, including rapid onset of depolarization block in response to acute D2 antagonist administration and downregulation of DA neuron population activity following acute and repeated D2 partial agonist administration, none of which are observed in normal rats. Novel target compounds have been developed based on the theory that glutamatergic dysfunction is central to schizophrenia pathology. Despite showing promise in preclinical research, none of the novel drugs succeeded in clinical trials. However, preclinical research is generally performed in normal, drug-naïve rats, whereas models with disease-relevant pathology and prior APD exposure may improve the predictive validity of preclinical research. Indeed, in MAM rats, chronic D2 antagonist treatment leads to persistent DA supersensitivity that interferes with the response to drugs that target upstream pathology. Moreover, MAM rats revealed that the peri-pubertal period is a stress-sensitive window that can be targeted to prevent the development of MAM pathology in adulthood. Neurodevelopmental models, such as the MAM model, can thus be used to test potential pharmacotherapies that may be able to treat schizophrenia in early stages of the disease. This article is part of the issue entitled 'Special Issue on Antipsychotics'.
Collapse
|