1
|
Mentis AFA, Dalamaga M. Rare biochemical & genetic conditions: clues for broader mechanistic insights. Cell Mol Life Sci 2025; 82:156. [PMID: 40210765 PMCID: PMC11985829 DOI: 10.1007/s00018-025-05652-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 04/12/2025]
Abstract
Rare disorders often represent a molecular deviation from hi-fidelity genomic integrity networks and are often perceived as too difficult or unimportant for further mechanistic studies. Here, we synthesize evidence demonstrating how valuable knowledge of biochemical pathways related to rare disorders can be for biomedicine. To this end, we describe several rare congenital lipid, protein, organic acid, and glycan metabolism disorders and discuss how rare phenotypes (such as "extreme responders") and case reports (such as the lenalidomide cases) have provided clues for drug discovery or repurposing. We also discuss how rare disorders such as Gaucher disease and ultra-rare genetic syndromes can provide insights into cancer and mTOR-driven metabolism, respectively. Our discussion highlights the continued value of biochemical pathways and studies in understanding human pathophysiology and drug discovery even in the genomics era.
Collapse
Affiliation(s)
- Alexios-Fotios A Mentis
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
2
|
Fu Y, Cheng HW. The Influence of Cecal Microbiota Transplantation on Chicken Injurious Behavior: Perspective in Human Neuropsychiatric Research. Biomolecules 2024; 14:1017. [PMID: 39199404 PMCID: PMC11352350 DOI: 10.3390/biom14081017] [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: 06/18/2024] [Revised: 07/26/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
Numerous studies have evidenced that neuropsychiatric disorders (mental illness and emotional disturbances) with aggression (or violence) pose a significant challenge to public health and contribute to a substantial economic burden worldwide. Especially, social disorganization (or social inequality) associated with childhood adversity has long-lasting effects on mental health, increasing the risk of developing neuropsychiatric disorders. Intestinal bacteria, functionally as an endocrine organ and a second brain, release various immunomodulators and bioactive compounds directly or indirectly regulating a host's physiological and behavioral homeostasis. Under various social challenges, stress-induced dysbiosis increases gut permeability causes serial reactions: releasing neurotoxic compounds, leading to neuroinflammation and neuronal injury, and eventually neuropsychiatric disorders associated with aggressive, violent, or impulsive behavior in humans and various animals via a complex bidirectional communication of the microbiota-gut-brain (MGB) axis. The dysregulation of the MGB axis has also been recognized as one of the reasons for the prevalence of social stress-induced injurious behaviors (feather pecking, aggression, and cannibalistic pecking) in chickens. However, existing knowledge of preventing and treating these disorders in both humans and chickens is not well understood. In previous studies, we developed a non-mammal model in an abnormal behavioral investigation by rationalizing the effects of gut microbiota on injurious behaviors in chickens. Based on our earlier success, the perspective article outlines the possibility of reducing stress-induced injurious behaviors in chickens through modifying gut microbiota via cecal microbiota transplantation, with the potential for providing a biotherapeutic rationale for preventing injurious behaviors among individuals with mental disorders via restoring gut microbiota diversity and function.
Collapse
Affiliation(s)
- Yuechi Fu
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA;
| | - Heng-Wei Cheng
- Livestock Behavior Research Unit, USDA-ARS, West Lafayette, IN 47907, USA
| |
Collapse
|
3
|
Ritz NL, Bastiaanssen TFS, Cowan CSM, Smith L, Theune N, Brocka M, Myers EM, Moloney RD, Moloney GM, Shkoporov AN, Draper LA, Hill C, Dinan TG, Slattery DA, Cryan JF. Social fear extinction susceptibility is associated with Microbiota-Gut-Brain axis alterations. Brain Behav Immun 2024; 120:315-326. [PMID: 38852762 DOI: 10.1016/j.bbi.2024.06.009] [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: 11/21/2023] [Revised: 06/06/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024] Open
Abstract
Social anxiety disorder is a common psychiatric condition that severely affects quality of life of individuals and is a significant societal burden. Although many risk factors for social anxiety exist, it is currently unknown how social fear sensitivity manifests biologically. Furthermore, since some individuals are resilient and others are susceptible to social fear, it is important to interrogate the mechanisms underpinning individual response to social fear situations. The microbiota-gut-brain axis has been associated with social behaviour, has recently been linked with social anxiety disorder, and may serve as a therapeutic target for modulation. Here, we assess the potential of this axis to be linked with social fear extinction processes in a murine model of social anxiety disorder. To this end, we correlated differential social fear responses with microbiota composition, central gene expression, and immune responses. Our data provide evidence that microbiota variability is strongly correlated with alterations in social fear behaviour. Moreover, we identified altered gene candidates by amygdalar transcriptomics that are linked with social fear sensitivity. These include genes associated with social behaviour (Armcx1, Fam69b, Kcnj9, Maoa, Serinc5, Slc6a17, Spata2, and Syngr1), inflammation and immunity (Cars, Ckmt1, Klf5, Maoa, Map3k12, Pex5, Serinc5, Sidt1, Spata2), and microbe-host interaction (Klf5, Map3k12, Serinc5, Sidt1). Together, these data provide further evidence for a role of the microbiota-gut-brain axis in social fear responses.
Collapse
Affiliation(s)
- Nathaniel L Ritz
- APC Microbiome Ireland, University College Cork, Cork T12YT20, Ireland; Dept. of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland
| | - Thomaz F S Bastiaanssen
- APC Microbiome Ireland, University College Cork, Cork T12YT20, Ireland; Dept. of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland
| | - Caitlin S M Cowan
- APC Microbiome Ireland, University College Cork, Cork T12YT20, Ireland; Dept. of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland
| | - Linda Smith
- APC Microbiome Ireland, University College Cork, Cork T12YT20, Ireland; School of Microbiology, University College Cork, Cork, T12K8AF, Ireland
| | - Nigel Theune
- APC Microbiome Ireland, University College Cork, Cork T12YT20, Ireland; Dept. of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland
| | - Marta Brocka
- APC Microbiome Ireland, University College Cork, Cork T12YT20, Ireland; Dept. of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland
| | - Eibhlís M Myers
- APC Microbiome Ireland, University College Cork, Cork T12YT20, Ireland; Dept. of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland
| | - Rachel D Moloney
- APC Microbiome Ireland, University College Cork, Cork T12YT20, Ireland; Dept. of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland
| | - Gerard M Moloney
- APC Microbiome Ireland, University College Cork, Cork T12YT20, Ireland; Dept. of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland
| | - Andrey N Shkoporov
- APC Microbiome Ireland, University College Cork, Cork T12YT20, Ireland; School of Microbiology, University College Cork, Cork, T12K8AF, Ireland
| | - Lorraine A Draper
- APC Microbiome Ireland, University College Cork, Cork T12YT20, Ireland; School of Microbiology, University College Cork, Cork, T12K8AF, Ireland
| | - Colin Hill
- APC Microbiome Ireland, University College Cork, Cork T12YT20, Ireland; School of Microbiology, University College Cork, Cork, T12K8AF, Ireland
| | - Timothy G Dinan
- APC Microbiome Ireland, University College Cork, Cork T12YT20, Ireland; Dept. of Psychiatry and Neurobehavioural Science, University College Cork, Cork T12YT20, Ireland
| | - David A Slattery
- Dept. of Psychiatry, Psychosomatics and Psychotherapy, University Hospital Frankfurt, Frankfurt 60528, Germany
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork T12YT20, Ireland; Dept. of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland.
| |
Collapse
|
4
|
Furukawa M, Tada H, Raju R, Wang J, Yokoi H, Yamada M, Shikama Y, Saito T, Saido TC, Matsushita K. Effects of tooth loss on behavioral and psychological symptoms of dementia in app knock-in mice. J Oral Biosci 2024; 66:329-338. [PMID: 38521152 DOI: 10.1016/j.job.2024.03.005] [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: 10/26/2023] [Revised: 03/07/2024] [Accepted: 03/11/2024] [Indexed: 03/25/2024]
Abstract
OBJECTIVES Many patients with Alzheimer's disease (AD) experience behavioral and psychological symptoms of dementia (BPSD), which significantly affect their quality of life. It is known that 5-Hydroxytryptamine (5-HT) plays a crucial role in the development of BPSD. While the relationship between tooth loss and AD symptoms has been acknowledged, the aspect of aggression has not been focused on until now. Despite the established importance of 5-HT in BPSD, how tooth loss is related to the exacerbation of AD symptoms, especially in terms of aggression, remains largely unexplored. Although nutritional status is known to influence the progression of dementia, the specific effect of tooth loss on peripheral symptoms, notably aggression, is not well understood. METHODS In our study, we conducted maxillary molar extractions in aged C57BL/6J and AppNL-G-F mice and observed their condition over a 3-month period. During this time, we documented significant behavioral and genetic differences between mice in the control groups and mice that underwent tooth extraction. Notably, mice that underwent tooth extraction exhibited a considerable decline in cognitive function and increased in aggression 3 months after tooth extraction compared with the control groups (C57BL/6J and AppNL-G-Fmice). RESULTS Our findings suggest that molar loss may lead to reduced 5-HT levels in the hippocampus, possibly mediated by the trigeminal nerve, contributing to the development of aggression and BPSD in AD. CONCLUSION This study sheds light on the intricate relationships between oral health, 5-HT, and AD symptoms, offering valuable insights into potential therapeutic avenues for managing BPSD in patients with dementia.
Collapse
Affiliation(s)
- Masae Furukawa
- Department of Oral Disease Research, Geroscience Research Center, National Center for Geriatrics and Gerontology, 7-430, Morioka-cho, Obu City, Aichi 474-8511, Japan.
| | - Hirobumi Tada
- Department of Nutrition, Faculty of Wellness, Shigakkan University, 55 Nadakayama, Yokone-cho, Obu City, Aichi 474-8651, Japan; Department of Integrative Physiology, Geroscience Research Center, National Center for Geriatrics and Gerontology, 7-430, Morioka-cho, Obu City, Aichi 474-8511, Japan.
| | - Resmi Raju
- Department of Oral Disease Research, Geroscience Research Center, National Center for Geriatrics and Gerontology, 7-430, Morioka-cho, Obu City, Aichi 474-8511, Japan.
| | - Jingshu Wang
- Department of Oral Disease Research, Geroscience Research Center, National Center for Geriatrics and Gerontology, 7-430, Morioka-cho, Obu City, Aichi 474-8511, Japan.
| | - Haruna Yokoi
- Department of Oral Disease Research, Geroscience Research Center, National Center for Geriatrics and Gerontology, 7-430, Morioka-cho, Obu City, Aichi 474-8511, Japan; Department of Geriatric Oral Science, Graduate School of Dentistry, Tohoku University, 4-1, Seiryo-machi, Aoba-ku, Sendai City, Miyagi 980-8575, Japan.
| | - Mitsuyoshi Yamada
- Department of Oral Disease Research, Geroscience Research Center, National Center for Geriatrics and Gerontology, 7-430, Morioka-cho, Obu City, Aichi 474-8511, Japan; Department of Operative Dentistry, School of Dentistry, Aichi Gakuin University, 2-11 Suemori-dori, Chikusa-ku, Nagoya City, Aichi 464-8651, Japan.
| | - Yosuke Shikama
- Department of Oral Disease Research, Geroscience Research Center, National Center for Geriatrics and Gerontology, 7-430, Morioka-cho, Obu City, Aichi 474-8511, Japan; Department of Geriatric Oral Science, Graduate School of Dentistry, Tohoku University, 4-1, Seiryo-machi, Aoba-ku, Sendai City, Miyagi 980-8575, Japan.
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya City, Aichi 467-0001, Japan; Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1, Hirosawa, Wako City, Saitama 351-0198, Japan.
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1, Hirosawa, Wako City, Saitama 351-0198, Japan.
| | - Kenji Matsushita
- Department of Oral Disease Research, Geroscience Research Center, National Center for Geriatrics and Gerontology, 7-430, Morioka-cho, Obu City, Aichi 474-8511, Japan; Department of Geriatric Oral Science, Graduate School of Dentistry, Tohoku University, 4-1, Seiryo-machi, Aoba-ku, Sendai City, Miyagi 980-8575, Japan.
| |
Collapse
|
5
|
Mousavi H, Rimaz M, Zeynizadeh B. Practical Three-Component Regioselective Synthesis of Drug-Like 3-Aryl(or heteroaryl)-5,6-dihydrobenzo[ h]cinnolines as Potential Non-Covalent Multi-Targeting Inhibitors To Combat Neurodegenerative Diseases. ACS Chem Neurosci 2024; 15:1828-1881. [PMID: 38647433 DOI: 10.1021/acschemneuro.4c00055] [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] [Indexed: 04/25/2024] Open
Abstract
Neurodegenerative diseases (NDs) are one of the prominent health challenges facing contemporary society, and many efforts have been made to overcome and (or) control it. In this research paper, we described a practical one-pot two-step three-component reaction between 3,4-dihydronaphthalen-1(2H)-one (1), aryl(or heteroaryl)glyoxal monohydrates (2a-h), and hydrazine monohydrate (NH2NH2•H2O) for the regioselective preparation of some 3-aryl(or heteroaryl)-5,6-dihydrobenzo[h]cinnoline derivatives (3a-h). After synthesis and characterization of the mentioned cinnolines (3a-h), the in silico multi-targeting inhibitory properties of these heterocyclic scaffolds have been investigated upon various Homo sapiens-type enzymes, including hMAO-A, hMAO-B, hAChE, hBChE, hBACE-1, hBACE-2, hNQO-1, hNQO-2, hnNOS, hiNOS, hPARP-1, hPARP-2, hLRRK-2(G2019S), hGSK-3β, hp38α MAPK, hJNK-3, hOGA, hNMDA receptor, hnSMase-2, hIDO-1, hCOMT, hLIMK-1, hLIMK-2, hRIPK-1, hUCH-L1, hPARK-7, and hDHODH, which have confirmed their functions and roles in the neurodegenerative diseases (NDs), based on molecular docking studies, and the obtained results were compared with a wide range of approved drugs and well-known (with IC50, EC50, etc.) compounds. In addition, in silico ADMET prediction analysis was performed to examine the prospective drug properties of the synthesized heterocyclic compounds (3a-h). The obtained results from the molecular docking studies and ADMET-related data demonstrated that these series of 3-aryl(or heteroaryl)-5,6-dihydrobenzo[h]cinnolines (3a-h), especially hit ones, can really be turned into the potent core of new drugs for the treatment of neurodegenerative diseases (NDs), and/or due to the having some reactionable locations, they are able to have further organic reactions (such as cross-coupling reactions), and expansion of these compounds (for example, with using other types of aryl(or heteroaryl)glyoxal monohydrates) makes a new avenue for designing novel and efficient drugs for this purpose.
Collapse
Affiliation(s)
- Hossein Mousavi
- Department of Organic Chemistry, Faculty of Chemistry, Urmia University, Urmia 5756151818, Iran
| | - Mehdi Rimaz
- Department of Chemistry, Payame Noor University, P.O. Box 19395-3697, Tehran 19395-3697, Iran
| | - Behzad Zeynizadeh
- Department of Organic Chemistry, Faculty of Chemistry, Urmia University, Urmia 5756151818, Iran
| |
Collapse
|
6
|
Neri L, Marziani B, Sebastiani P, Del Beato T, Colanardi A, Legge MP, Aureli A. Aggressiveness in Italian Children with ADHD: MAOA Gene Polymorphism Involvement. Diseases 2024; 12:70. [PMID: 38667528 PMCID: PMC11049508 DOI: 10.3390/diseases12040070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
ADHD is a neurodevelopmental disorder that children and adults can develop. A complex interplay of genetic and environmental factors may underlie interindividual variability in ADHD and potentially related aggressive behavior. Using high-resolution molecular biology techniques, we investigated the impact of some MAOA and SLC6A4 variations on ADHD and aggressive behavior in a group of 80 Italian children with ADHD and in 80 healthy controls. We found that homozygous genotypes of MAOA rs6323 and rs1137070 were associated with an increased risk of ADHD (p = 0.02 and p = 0.03, respectively), whereas the heterozygous genotypes (GT of rs6323 and CT of rs1137030) (p = 0.0002 and p = 0.0006) were strongly linked to a lower risk of developing this disorder. In patients with aggressive behavior, we highlighted only a weak negative association of both MAOA polymorphisms (heterozygous genotypes) with aggressiveness, suggesting that these genotypes may be protective towards specific changes in behavior (p = 0.05). Interestingly, an increase in the GG genotype of rs6323 (p = 0.01) and a decrease in GT genotype (p = 0.0005) was also found in patients without aggressive behavior compared to controls. Regarding 5HTT gene genotyping, no allele and genotype differences have been detected among patients and controls. Our work shows that defining a genetic profile of ADHD may help in the early detection of patients who are more vulnerable to ADHD and/or antisocial and aggressive behavior and to design precision-targeted therapies.
Collapse
Affiliation(s)
- Ludovico Neri
- Neurology and Psychiatry Unit for Children and Adolescents, San Salvatore Hospital, via L. Natali, 1, Coppito, 67100 L’Aquila, Italy; (L.N.); (M.P.L.)
| | - Beatrice Marziani
- Emergency Medicine Department, Sant’Anna University Hospital, Via A. Moro, 8, Cona, 44124 Ferrara, Italy;
| | - Pierluigi Sebastiani
- CNR Institute of Translational Pharmacology, Via Carducci 32, 67100 L’Aquila, Italy; (P.S.); (T.D.B.); (A.C.)
| | - Tiziana Del Beato
- CNR Institute of Translational Pharmacology, Via Carducci 32, 67100 L’Aquila, Italy; (P.S.); (T.D.B.); (A.C.)
| | - Alessia Colanardi
- CNR Institute of Translational Pharmacology, Via Carducci 32, 67100 L’Aquila, Italy; (P.S.); (T.D.B.); (A.C.)
| | - Maria Pia Legge
- Neurology and Psychiatry Unit for Children and Adolescents, San Salvatore Hospital, via L. Natali, 1, Coppito, 67100 L’Aquila, Italy; (L.N.); (M.P.L.)
| | - Anna Aureli
- CNR Institute of Translational Pharmacology, Via Carducci 32, 67100 L’Aquila, Italy; (P.S.); (T.D.B.); (A.C.)
| |
Collapse
|
7
|
Liu L, Wang Y, Hong L, Bragazzi NL, Dai H, Chen H. Obstructive Sleep Apnea and Hypertensive Heart Disease: From Pathophysiology to Therapeutics. Rev Cardiovasc Med 2023; 24:342. [PMID: 39077075 PMCID: PMC11272867 DOI: 10.31083/j.rcm2412342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/29/2023] [Accepted: 10/12/2023] [Indexed: 07/31/2024] Open
Abstract
Obstructive sleep apnea (OSA) is characterized by recurrent episodes of complete or partial obstruction of the upper airway that lead to intermittent hypoxemia, negative intrathoracic pressure, hypercapnia, and sleep disturbances. While OSA is recognized as a significant risk factor for cardiovascular disease, it's relationship with hypertensive heart disease (HHD) remains underappreciated. HHD is a condition characterized by the pathological hypertrophy of the left ventricle, a consequence of elevated arterial hypertension. Interestingly, both OSA and HHD share similar underlying mechanisms including hypertension, left ventricular hypertrophy, myocardial fibrosis, oxidative stress, and inflammation, which ultimately contribute to the progression of HHD. This review aims to shed light on the potential role of OSA in HHD pathogenesis, summarizing current OSA treatment options. It is hoped that this review will encourage a renewed clinical focus on HHD and underscore the need for further OSA research, particularly in the context of screening and treating HHD patients.
Collapse
Affiliation(s)
- Lu Liu
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310000 Hangzhou, Zhejiang, China
| | - Yi Wang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310000 Hangzhou, Zhejiang, China
| | - Liqiong Hong
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310000 Hangzhou, Zhejiang, China
| | - Nicola Luigi Bragazzi
- Human Nutrition Unit, Department of Food and Drugs, Medical School, University of Parma, 43125 Parma, Italy
| | - Haijiang Dai
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310000 Hangzhou, Zhejiang, China
| | - Huimin Chen
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310000 Hangzhou, Zhejiang, China
| |
Collapse
|
8
|
de Bartolomeis A, De Simone G, De Prisco M, Barone A, Napoli R, Beguinot F, Billeci M, Fornaro M. Insulin effects on core neurotransmitter pathways involved in schizophrenia neurobiology: a meta-analysis of preclinical studies. Implications for the treatment. Mol Psychiatry 2023; 28:2811-2825. [PMID: 37085712 PMCID: PMC10615753 DOI: 10.1038/s41380-023-02065-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 03/28/2023] [Accepted: 03/31/2023] [Indexed: 04/23/2023]
Abstract
Impairment of insulin action and metabolic dysregulation have traditionally been associated with schizophrenia, although the molecular basis of such association remains still elusive. The present meta-analysis aims to assess the impact of insulin action manipulations (i.e., hyperinsulinemia, hypoinsulinemia, systemic or brain insulin resistance) on glutamatergic, dopaminergic, γ-aminobutyric acid (GABA)ergic, and serotonergic pathways in the central nervous system. More than one hundred outcomes, including transcript or protein levels, kinetic parameters, and other components of the neurotransmitter pathways, were collected from cultured cells, animals, or humans, and meta-analyzed by applying a random-effects model and adopting Hedges'g to compare means. Two hundred fifteen studies met the inclusion criteria, of which 180 entered the quantitative synthesis. Significant impairments in key regulators of synaptic plasticity processes were detected as the result of insulin handlings. Specifically, protein levels of N-methyl-D-aspartate receptor (NMDAR) subunits including type 2A (NR2A) (Hedges' g = -0.95, 95%C.I. = -1.50, -0.39; p = 0.001; I2 = 47.46%) and 2B (NR2B) (Hedges'g = -0.69, 95%C.I. = -1.35, -0.02; p = 0.043; I2 = 62.09%), and Postsynaptic density protein 95 (PSD-95) (Hedges'g = -0.91, 95%C.I. = -1.51, -0.32; p = 0.003; I2 = 77.81%) were found reduced in insulin-resistant animal models. Moreover, insulin-resistant animals showed significantly impaired dopamine transporter activity, whereas the dopamine D2 receptor mRNA expression (Hedges'g = 3.259; 95%C.I. = 0.497, 6.020; p = 0.021; I2 = 90.61%) increased under insulin deficiency conditions. Insulin action modulated glutamate and GABA release, as well as several enzymes involved in GABA and serotonin synthesis. These results suggest that brain neurotransmitter systems are susceptible to insulin signaling abnormalities, resembling the discrete psychotic disorders' neurobiology and possibly contributing to the development of neurobiological hallmarks of treatment-resistant schizophrenia.
Collapse
Affiliation(s)
- Andrea de Bartolomeis
- Section of Psychiatry, Laboratory of Molecular and Translational Psychiatry, Unit of Treatment-Resistant Psychiatric Disorders, Department of Neuroscience, Reproductive Sciences and Odontostomatology University of Naples "Federico II", School of Medicine, Via Pansini 5, 80131, Naples, Italy.
| | - Giuseppe De Simone
- Section of Psychiatry, Laboratory of Molecular and Translational Psychiatry, Unit of Treatment-Resistant Psychiatric Disorders, Department of Neuroscience, Reproductive Sciences and Odontostomatology University of Naples "Federico II", School of Medicine, Via Pansini 5, 80131, Naples, Italy
| | - Michele De Prisco
- Section of Psychiatry, Laboratory of Molecular and Translational Psychiatry, Unit of Treatment-Resistant Psychiatric Disorders, Department of Neuroscience, Reproductive Sciences and Odontostomatology University of Naples "Federico II", School of Medicine, Via Pansini 5, 80131, Naples, Italy
- Bipolar and Depressive Disorders Unit, Institute of Neuroscience, Hospital Clinic, University of Barcelona, IDIBAPS, CIBERSAM, 170 Villarroel st, 12-0, 08036, Barcelona, Catalonia, Spain
| | - Annarita Barone
- Section of Psychiatry, Laboratory of Molecular and Translational Psychiatry, Unit of Treatment-Resistant Psychiatric Disorders, Department of Neuroscience, Reproductive Sciences and Odontostomatology University of Naples "Federico II", School of Medicine, Via Pansini 5, 80131, Naples, Italy
| | - Raffaele Napoli
- Department of Translational Medical Sciences, University of Naples "Federico II", Via S. Pansini 5, 80131, Naples, Italy
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Francesco Beguinot
- Department of Translational Medical Sciences, University of Naples "Federico II", Via S. Pansini 5, 80131, Naples, Italy
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Martina Billeci
- Section of Psychiatry, Laboratory of Molecular and Translational Psychiatry, Unit of Treatment-Resistant Psychiatric Disorders, Department of Neuroscience, Reproductive Sciences and Odontostomatology University of Naples "Federico II", School of Medicine, Via Pansini 5, 80131, Naples, Italy
| | - Michele Fornaro
- Section of Psychiatry, Laboratory of Molecular and Translational Psychiatry, Unit of Treatment-Resistant Psychiatric Disorders, Department of Neuroscience, Reproductive Sciences and Odontostomatology University of Naples "Federico II", School of Medicine, Via Pansini 5, 80131, Naples, Italy
| |
Collapse
|
9
|
Sesso G, Masi G. Pharmacological strategies for the management of the antisocial personality disorder. Expert Rev Clin Pharmacol 2023; 16:181-194. [PMID: 36787887 DOI: 10.1080/17512433.2023.2181159] [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: 11/21/2022] [Accepted: 02/09/2023] [Indexed: 02/16/2023]
Abstract
INTRODUCTION Antisocial personality disorder (AsPD) is a pervasive pattern of violation of others' rights, related to the concept of psychopathy. AsPD is stable over time from adolescence, with evidence of conduct disorder (CD) before 15 years. DSM-5 included a specifier 'with limited prosocial emotions' (LPE), which characterizes adolescents with higher developmental vulnerability to develop AsPD. Despite being relatively frequent with considerable societal impact, AsPD is a difficult-to-treat condition with high comorbidity rates and poor evidence for effective pharmacological interventions. AREAS COVERED We conducted a narrative review and searched PubMed up to September 2022. We included RCTs and naturalistic studies evaluating pharmacological interventions on AsPD in adults, including those with comorbid substance use disorder or psychopathic traits. Evidence in youths with CD, callous-unemotional (CU) traits and aggression were also reviewed, exploring the role of CU traits as moderators of response. EXPERT OPINION Psychosocial interventions are the first option, with possible improvement of CU traits, beyond behavioral and affective symptoms, particularly if implemented early during development. Limited information, based on low-quality studies, supports the pharmacological options. Second-generation antipsychotics, lithium, anti-epileptic drugs, and stimulants are first-line medications, according to different target symptoms. Developmental pathways including ADHD suggest a specific role of psychostimulants.
Collapse
Affiliation(s)
- Gianluca Sesso
- Social and Affective Neuroscience Group, Molecular Mind Lab, IMT School for Advanced Studies Lucca, Lucca, Italy
- Department of Child and Adolescent Psychiatry and Psychopharmacology, IRCCS Stella Maris Foundation, Pisa, Italy
| | - Gabriele Masi
- Department of Child and Adolescent Psychiatry and Psychopharmacology, IRCCS Stella Maris Foundation, Pisa, Italy
| |
Collapse
|
10
|
Xenodochidis C, Staneva D, Vasileva B, Draganova M, Miloshev G, Georgieva M, Zagorchev P. The Photobiomodulation of MAO-A Affects the Contractile Activity of Smooth Muscle Gastric Tissues. Biomolecules 2022; 13:biom13010032. [PMID: 36671417 PMCID: PMC9855794 DOI: 10.3390/biom13010032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
Nowadays, the utilized electromagnetic radiation (ER) in modalities such as photobiomodulation (PBM) finds broader applications in medical practice due to the promising results suggested by numerous reports. To date, the published data do not allow for the in-depth elucidation of the molecular mechanisms through which ER impacts the human organism. Furthermore, there is a total lack of evidence justifying the relation between the enzymatic activity of monoamine oxidase A (MAO-A) and the effect of 5-hydroxytryptamine (5-HT) on the spontaneous contractile activity of smooth muscle gastric tissues exposed to various light sources. We found that exposure of these tissues to lamps, emitting light with wavelengths of 254 nm and 350 nm, lasers, emitting light with 532 nm and 808 nm, and light-emitting diodes (LEDs) with ER at a wavelength of 660 nm, increased the 5-HT effect on the contractility. On the other hand, LEDs at 365 nm and 470 nm reduced it. The analysis of MAO-A enzymatic activity after exposure to the employed light emitters endorsed these findings. Furthermore, MAOA gene expression studies confirmed the possibility of its optogenetic regulation. Therefore, we concluded that the utilized emitters could alternate the functions of significant neuromediators by modulating the activity and gene transcription levels of enzymes that degrade them. Our investigations will help to disclose the selective conditions upon which PBM can effectively treat gastrointestinal and neurological disorders.
Collapse
Affiliation(s)
- Charilaos Xenodochidis
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
- Laboratory of Molecular Genetics, Institute of Molecular Biology “Acad. Roumen Tsanev”, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| | - Dessislava Staneva
- Laboratory of Molecular Genetics, Institute of Molecular Biology “Acad. Roumen Tsanev”, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| | - Bela Vasileva
- Laboratory of Molecular Genetics, Institute of Molecular Biology “Acad. Roumen Tsanev”, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| | - Milena Draganova
- Department of Medical Biology, Medical Faculty, Medical University-Plovdiv, 15A Vasil Aprilov Blvd., 4002 Plovdiv, Bulgaria
- Research Institute, Medical University-Plovdiv, 15A Vasil Aprilov Blvd., 4002 Plovdiv, Bulgaria
| | - George Miloshev
- Laboratory of Molecular Genetics, Institute of Molecular Biology “Acad. Roumen Tsanev”, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| | - Milena Georgieva
- Laboratory of Molecular Genetics, Institute of Molecular Biology “Acad. Roumen Tsanev”, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
- Correspondence: ; Tel.: +359-896-833-604
| | - Plamen Zagorchev
- Research Institute, Medical University-Plovdiv, 15A Vasil Aprilov Blvd., 4002 Plovdiv, Bulgaria
- Department of Biophysics, Faculty of Pharmacy, Medical University-Plovdiv, 15A Vasil Aprilov Blvd., 4002 Plovdiv, Bulgaria
| |
Collapse
|
11
|
GATTACA is still pertinent 25 years later. Nat Genet 2022; 54:1758-1760. [PMID: 36424501 DOI: 10.1038/s41588-022-01242-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
12
|
Advances in Molecular Pathology of Obstructive Sleep Apnea. Molecules 2022; 27:molecules27238422. [PMID: 36500515 PMCID: PMC9739159 DOI: 10.3390/molecules27238422] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/22/2022] [Accepted: 11/24/2022] [Indexed: 12/03/2022] Open
Abstract
Obstructive sleep apnea (OSA) is a common syndrome that features a complex etiology and set of mechanisms. Here we summarized the molecular pathogenesis of OSA, especially the prospective mechanism of upper? airway dilator fatigue and the current breakthroughs. Additionally, we also introduced the molecular mechanism of OSA in terms of related studies on the main signaling pathways and epigenetics alterations, such as microRNA, long non-coding RNA, and DNA methylation. We also reviewed small molecular compounds, which are potential targets for gene regulations in the future, that are involved in the regulation of OSA. This review will be beneficial to point the way for OSA research within the next decade.
Collapse
|
13
|
Mbiydzenyuy NE, Hemmings SMJ, Qulu L. Prenatal maternal stress and offspring aggressive behavior: Intergenerational and transgenerational inheritance. Front Behav Neurosci 2022; 16:977416. [PMID: 36212196 PMCID: PMC9539686 DOI: 10.3389/fnbeh.2022.977416] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
Even though studies have shown that prenatal maternal stress is associated with increased reactivity of the HPA axis, the association between prenatal maternal stress and fetal glucocorticoid exposure is complex and most likely dependent on unidentified and poorly understood variables including nature and timing of prenatal insults. The precise mechanisms in which prenatal maternal stress influence neuroendocrine signaling between the maternal-placental-fetal interface are still unclear. The aim of this review article is to bring comprehensive basic concepts about prenatal maternal stress and mechanisms of transmission of maternal stress to the fetus. This review covers recent studies showing associations between maternal stress and alterations in offspring aggressive behavior, as well as the possible pathways for the “transmission” of maternal stress to the fetus: (1) maternal-fetal HPA axis dysregulation; (2) intrauterine environment disruption due to variations in uterine artery flow; (3) epigenetic modifications of genes implicated in aggressive behavior. Here, we present evidence for the phenomenon of intergenerational and transgenerational transmission, to better understands the mechanism(s) of transmission from parent to offspring. We discuss studies showing associations between maternal stress and alterations in offspring taking note of neuroendocrine, brain architecture and epigenetic changes that may suggest risk for aggressive behavior. We highlight animal and human studies that focus on intergenerational transmission following exposure to stress from a biological mechanistic point of view, and maternal stress-induced epigenetic modifications that have potential to impact on aggressive behavior in later generations.
Collapse
Affiliation(s)
- Ngala Elvis Mbiydzenyuy
- Department of Basic Science, School of Medicine, Copperbelt University, Ndola, Zambia
- Division of Medical Physiology, Biomedical Science Research Institute, Stellenbosch University, Cape Town, South Africa
- *Correspondence: Ngala Elvis Mbiydzenyuy,
| | - Sian Megan Joanna Hemmings
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Lihle Qulu
- Division of Medical Physiology, Biomedical Science Research Institute, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
14
|
The regulatory role of AP-2β in monoaminergic neurotransmitter systems: insights on its signalling pathway, linked disorders and theragnostic potential. Cell Biosci 2022; 12:151. [PMID: 36076256 PMCID: PMC9461128 DOI: 10.1186/s13578-022-00891-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/28/2022] [Indexed: 11/10/2022] Open
Abstract
AbstractMonoaminergic neurotransmitter systems play a central role in neuronal function and behaviour. Dysregulation of these systems gives rise to neuropsychiatric and neurodegenerative disorders with high prevalence and societal burden, collectively termed monoamine neurotransmitter disorders (MNDs). Despite extensive research, the transcriptional regulation of monoaminergic neurotransmitter systems is not fully explored. Interestingly, certain drugs that act on these systems have been shown to modulate central levels of the transcription factor AP-2 beta (AP-2β, gene: TFAP2Β). AP-2β regulates multiple key genes within these systems and thereby its levels correlate with monoamine neurotransmitters measures; yet, its signalling pathways are not well understood. Moreover, although dysregulation of TFAP2Β has been associated with MNDs, the underlying mechanisms for these associations remain elusive. In this context, this review addresses AP-2β, considering its basic structural aspects, regulation and signalling pathways in the controlling of monoaminergic neurotransmitter systems, and possible mechanisms underpinning associated MNDS. It also underscores the significance of AP-2β as a potential diagnostic biomarker and its potential and limitations as a therapeutic target for specific MNDs as well as possible pharmaceutical interventions for targeting it. In essence, this review emphasizes the role of AP-2β as a key regulator of the monoaminergic neurotransmitter systems and its importance for understanding the pathogenesis and improving the management of MNDs.
Collapse
|
15
|
Brown J, Li Z, Wang X, Kim YJ, Wang YC, Zuo Y, Hong W, Wang P, Li B, Yang L. Nanoformulation improves antitumor efficacy of MAOI immune checkpoint blockade therapy without causing aggression-related side effects. Front Pharmacol 2022; 13:970324. [PMID: 36120311 PMCID: PMC9475110 DOI: 10.3389/fphar.2022.970324] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
MAOIs, a well-established class of antidepressant that operate through the inhibition of monoamine oxidase to increase available serotonin, have recently been identified as a surprisingly effective candidate for the circumvention of tumor-induced immune suppression due to their abilities to enhance antitumor T cell activity through autocrine serotonin signaling and depolarize alternatively activated tumor-associated macrophages through a reduction in reactive oxygen species production. However, this impressive class of antidepressants-turned-cancer-drugs can induce aggressive behavioral side effects when administered in immunotherapeutic doses. In this study, we investigated the possibility of avoiding these neurological side effects while simultaneously improving antitumor activity by establishing crosslinked multilamellar liposomal vesicles (cMLVs) containing the MAOI phenelzine (PLZ). Our results showed that cMLV-PLZ treatment increases antitumor efficacy in a B16-OVA mouse melanoma model compared to treatment with free phenelzine. We also found that nanoformulation resulted in the complete elimination of MAOI-related aggression. These findings suggest a promising direction for the future of MAOIs repurposed for cancer immunotherapies.
Collapse
Affiliation(s)
- James Brown
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA, United States
| | - Zhe Li
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA, United States
| | - Xi Wang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA, United States
| | - Yu Jeong Kim
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA, United States
| | - Yu-Chen Wang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA, United States
| | - Yanning Zuo
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA, United States
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, United States
| | - Weizhe Hong
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA, United States
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, United States
| | - Pin Wang
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA, United States
| | - Bo Li
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA, United States
- *Correspondence: Bo Li, ; Lili Yang,
| | - Lili Yang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA, United States
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, United States
- Jonsson Comprehensive Cancer Center, The David Geffen School of Medicine, University of California, Los Angeles, CA, United States
- Molecular Biology Institute, University of California, Los Angeles, CA, United States
- *Correspondence: Bo Li, ; Lili Yang,
| |
Collapse
|
16
|
The neurobiology of antisocial behavior in adolescence current knowledge and relevance for youth forensic clinical practice. Curr Opin Psychol 2022; 47:101356. [DOI: 10.1016/j.copsyc.2022.101356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 03/28/2022] [Accepted: 05/04/2022] [Indexed: 11/23/2022]
|
17
|
Mentis AFA, Dalamaga M, Lu C, Polissiou MG. Saffron for "toning down" COVID-19-related cytokine storm: Hype or hope? A mini-review of current evidence. Metabol Open 2021; 11:100111. [PMID: 34312610 PMCID: PMC8294713 DOI: 10.1016/j.metop.2021.100111] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/13/2021] [Accepted: 07/15/2021] [Indexed: 12/12/2022] Open
Abstract
AIM To assess the potential role of saffron in downregulating inflammation and cytokine storm during COVID-19. MAIN FINDINGS Three main compounds of saffron, i.e., crocetin esters, picrocrocin, and safranal, present strong antioxidant and anti-inflammatory action for several disease states (e.g., Alzheimer's, cancer, and depression) but have also been studied in COVID-19. In particular, based on our comprehensive review of both in vitro and in silico studies, saffron's essential oils and other constituents appear to have both immunomodulatory and anti-asthmatic actions; these actions can be particularly helpful to treat patients with respiratory symptoms due to COVID-19. Moreover, crocin appears to reduce the COVID-19-related cytokine cascade and downregulate angiotensin-converting enzyme 2 (ACE2) gene expression. Last, in silico studies suggest that saffron's astragalin and crocin could have inhibitory actions on SARS-CoV-2 protease and spike protein, respectively. CONCLUSION Saffron represents a promising substance for toning down cytokine storm during COVID-19, as well as a potential preventive treatment for COVID-19. However, appropriate randomized clinical trials, especially those using biomarkers as surrogates to assess inflammatory status, should be designed in order to assess the clinical efficacy of saffron and allow its use as an adjunct treatment modality, particularly in resource-poor settings where access to drugs may be limited.
Collapse
Affiliation(s)
- Alexios-Fotios A Mentis
- University Research Institute of Maternal and Child Health & Precision Medicine, Athens, Greece
- UNESCO Chair on Adolescent Health Care, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Maria Dalamaga
- Department of Biological Chemistry, School of Medicine, National and Kapodistrian University of Athens, 75 Mikras Asias, 11527, Athens, Greece
| | - Cuncun Lu
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Moschos G Polissiou
- Laboratory of Chemistry, Department of Food Science and Human Nutrition, School of Food, Biotechnology and Development, Agricultural University of Athens, Athens, Greece
| |
Collapse
|