1
|
Al-Jaber H, Al-Muraikhy S, Jabr A, Yousef A, Anwardeen NR, Elrayess MA, Al-Mansoori L. Comparing Methods for Induction of Insulin Resistance in Mouse 3T3-L1 Cells. Curr Diabetes Rev 2025; 21:1-12. [PMID: 38204253 DOI: 10.2174/0115733998263359231211044539] [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: 08/06/2023] [Revised: 10/26/2023] [Accepted: 10/30/2023] [Indexed: 01/12/2024]
Abstract
Cell culture plays a crucial role in addressing fundamental research questions, particularly in studying insulin resistance (IR) mechanisms. Multiple in vitro models are utilized for this purpose, but their technical distinctions and relevance to in vivo conditions remain unclear. This study aims to assess the effectiveness of existing in vitro models in inducing IR and their ability to replicate in vivo IR conditions. BACKGROUND Insulin resistance (IR) is a cellular condition linked to metabolic disorders. Despite the utility of cell culture in IR research, questions persist regarding the suitability of various models. This study seeks to evaluate these models' efficiency in inducing IR and their ability to mimic in vivo conditions. Insights gained from this research could enhance our understanding of model strengths and limitations, potentially advancing strategies to combat IR and related disorders. OBJECTIVE 1- Investigate the technical differences between existing cell culture models used to study molecular mediators of insulin resistance (IR). 2- Compare the effectiveness of present in vitro models in inducing insulin resistance (IR). 3- Assess the relevance of the existing cell culture models in simulating the in vivo conditions and environment that provoke the induction of insulin resistance (IR). METHODS AND MATERIAL In vitro, eight sets of 3T3-L1 cells were cultured until they reached 90% confluence. Subsequently, adipogenic differentiation was induced using a differentiation cocktail (media). These cells were then divided into four groups, with four subjected to normal conditions and the other four to hypoxic conditions. Throughout the differentiation process, each cell group was exposed to specific factors known to induce insulin resistance (IR). These factors included 2.5 nM tumor necrosis factor-alpha (TNFα), 20 ng/ml interleukin-6 (IL-6), 10 micromole 4-hydroxynonenal (4HNE), and high insulin (HI) at a concentration of 100 nM. To assess cell proliferation, DAPI staining was employed, and the expression of genes associated with various metabolic pathways affected by insulin resistance was investigated using Real-Time PCR. Additionally, insulin signaling was examined using the Bio-plex Pro cell signaling Akt panel. RESULTS We induced insulin resistance in 3T3-L1 cells using IL-6, TNFα, 4HNE, and high insulin in both hypoxic and normoxic conditions. Hypoxia increased HIF1a gene expression by approximately 30% (P<0.01). TNFα reduced cell proliferation by 10-20%, and chronic TNFα treatment significantly decreased mature adipocytes due to its cytotoxicity. We assessed the impact of insulin resistance (IR) on metabolic pathways, focusing on genes linked to branched-chain amino acid metabolism, detoxification, and chemotaxis. Notably, ALDH6A1 and MCCC1 genes, related to amino acid metabolism, were significantly affected under hypoxic conditions. TNFα treatment notably influenced MCP-1 and MCP-2 genes linked to chemotaxis, with remarkable increases in MCP-1 levels and MCP-2 expression primarily under hypoxia. Detoxification-related genes showed minimal impact, except for a significant increase in MAOA expression under acute hypoxic conditions with TNFα treatment. Additional genes displayed varying effects, warranting further investigation. To investigate insulin signaling's influence in vitro by IRinducing factors, we assessed phospho-protein levels. Our results reveal a significant p-Akt induction with chronic high insulin (10%) and acute TNFα (12%) treatment under hypoxia (both P<0.05). Other insulin resistance-related phospho-proteins (GSK3B, mTOR, PTEN) increased with IL-6, 4HNE, TNFα, and high insulin under hypoxia, while p-IRS1 levels remained unaffected. CONCLUSION In summary, different in vitro models using inflammatory, oxidative stress, and high insulin conditions under hypoxic conditions can capture various aspects of in vivo adipose tissue insulin resistance (IR). Among these models, acute TNFα treatment may offer the most robust approach for inducing IR in 3T3-L1 cells.
Collapse
Affiliation(s)
- Hend Al-Jaber
- Biomedical Research Center, Qatar University, Doha, Qatar
| | | | - Aldana Jabr
- Biomedical Sciences Department, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Aisha Yousef
- Biomedical Sciences Department, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | | | | | | |
Collapse
|
2
|
Zhang Y, Wang R, Liu T, Wang R. Exercise as a Therapeutic Strategy for Obesity: Central and Peripheral Mechanisms. Metabolites 2024; 14:589. [PMID: 39590824 PMCID: PMC11596326 DOI: 10.3390/metabo14110589] [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: 09/29/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Obesity is a complex, multifactorial condition involving excessive fat accumulation due to an imbalance between energy intake and expenditure, with its global prevalence steadily rising. This condition significantly increases the risk of chronic diseases, including sarcopenia, type 2 diabetes, and cardiovascular diseases, highlighting the need for effective interventions. Exercise has emerged as a potent non-pharmacological approach to combat obesity, targeting both central and peripheral mechanisms that regulate metabolism, energy expenditure, and neurological functions. In the central nervous system, exercise influences appetite, mood, and cognitive functions by modulating the reward system and regulating appetite-controlling hormones to manage energy intake. Concurrently, exercise promotes thermogenesis in adipose tissue and regulates endocrine path-ways and key metabolic organs, such as skeletal muscle and the liver, to enhance fat oxidation and support energy balance. Despite advances in understanding exercise's role in obesity, the precise interaction between the neurobiological and peripheral metabolic pathways remains underexplored, particularly in public health strategies. A better understanding of these interactions could inform more comprehensive obesity management approaches by addressing both central nervous system influences on behavior and peripheral metabolic regulation. This review synthesizes recent insights into these roles, highlighting potential therapeutic strategies targeting both systems for more effective obesity interventions.
Collapse
Affiliation(s)
- Yiyin Zhang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (Y.Z.); (R.W.)
| | - Ruwen Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (Y.Z.); (R.W.)
| | - Tiemin Liu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Ru Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (Y.Z.); (R.W.)
| |
Collapse
|
3
|
Abeysekera MV, Ni D, Gilbert L, Hibbert E, Nanan R. Linking the reversal of gestational insulin resistance to postpartum depression. BMC Med 2024; 22:433. [PMID: 39379948 PMCID: PMC11462660 DOI: 10.1186/s12916-024-03659-7] [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: 03/12/2024] [Accepted: 09/26/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Postpartum depression (PPD) constitutes a significant mental health disorder affecting almost one fifth of pregnancies globally. Despite extensive research, the precise etiological mechanisms underlying PPD remain elusive. However, several risk factors like genetic predisposition, hormonal fluctuations, and stress-related environmental and psychosocial triggers have been found to be implicated in its development. MAIN: Recently, an increased risk of PPD has been reported to be associated with gestational diabetes mellitus (GDM), which is characterized by the disruption of glucose metabolism, primarily attributed to the emergence of insulin resistance (IR). While IR during pregnancy seems to be an evolutionary adaptative mechanism to handle the profound metabolic alterations during pregnancy, its subsequent resolution following delivery necessitates a reconfiguration of the metabolic landscape in both peripheral tissues and the central nervous system (CNS). Considering the pivotal roles of energy metabolism, particularly glucose metabolism, in CNS functions, we propose a novel model that such pronounced changes in IR and the associated glucose metabolism seen postpartum might account for PPD development. This concept is based on the profound influences from insulin and glucose metabolism on brain functions, potentially via modulating neurotransmitter actions of dopamine and serotonin. Their sudden postpartum disruption is likely to be linked to mood changes, as observed in PPD. CONCLUSIONS The detailed pathogenesis of PPD might be multifactorial and still remains to be fully elucidated. Nevertheless, our hypothesis might account in part for an additional etiological factor to PPD development. If our concept is validated, it can provide guidance for future PPD prevention, diagnosis, and intervention.
Collapse
Affiliation(s)
| | - Duan Ni
- Nepean Hospital, Nepean Blue Mountains Local Health District, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Leah Gilbert
- Nepean Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Emily Hibbert
- Nepean Hospital, Nepean Blue Mountains Local Health District, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Nepean Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Ralph Nanan
- Nepean Hospital, Nepean Blue Mountains Local Health District, Sydney, NSW, Australia.
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia.
- Nepean Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
4
|
de Farias Fraga G, da Silva Rodrigues F, Jantsch J, Silva Dias V, Milczarski V, Wickert F, Pereira Medeiros C, Eller S, Gatto Barschak A, Giovenardi M, Padilha Guedes R. Omega-3 Attenuates Disrupted Neurotransmission and Partially Protects Metabolic Dysfunction Caused by Obesity in Wistar Rats. Neurochem Res 2024; 49:2763-2773. [PMID: 38960951 DOI: 10.1007/s11064-024-04201-0] [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: 12/22/2023] [Revised: 05/02/2024] [Accepted: 06/19/2024] [Indexed: 07/05/2024]
Abstract
Omega-3 (n3) is a polyunsaturated fatty acid well known for its anti-inflammatory and neuroprotective properties. Obesity is linked to chronic inflammation that disrupts metabolism, the intestine physiology and the central nervous system functioning. This study aims to determine if n3 supplementation can interfere with the effects of obesity on the mitochondrial activity, intestinal barrier, and neurotransmitter levels in the brain of Wistar rats that received cafeteria diet (CAF). We examined adipose tissue, skeletal muscle, plasma, intestine, and the cerebral cortex of four groups: CT (control diet), CTn3 (control diet with n3 supplementation), CAF, and CAFn3 (CAF and n3). Diets were offered for 13 weeks, with n3 supplementation in the final 5 weeks. Adipose tissue Electron Transport Chain complexes I, II, and III showed higher activity in CAF groups, as did complexes III and IV in skeletal muscle. Acetate levels in plasma were reduced in CAF groups, and Lipopolysaccharide (LPS) was higher in the CAF group but reduced in CAFn3 group. Claudin-5 in the intestine was lower in CAF groups, with no n3 supplementation effect. In the cerebral cortex, dopamine levels were decreased with CAF, which was reversed by n3. DOPAC, a dopamine metabolite, also showed a supplementation effect, and HVA, a diet effect. Serotonin levels increased in the CAF group that received supplementation. Therefore, we demonstrate disturbances in mitochondria, plasma, intestine and brain of rats submitted to CAF and the potential benefit of n3 supplementation in endotoxemia and neurotransmitter levels.
Collapse
Affiliation(s)
- Gabriel de Farias Fraga
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Fernanda da Silva Rodrigues
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Jeferson Jantsch
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Victor Silva Dias
- Biomedical Science Undergraduate Program, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Vitória Milczarski
- Biomedical Science Undergraduate Program, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Fernanda Wickert
- Biomedical Science Undergraduate Program, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Camila Pereira Medeiros
- Biomedical Science Undergraduate Program, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Sarah Eller
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Alethéa Gatto Barschak
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Marcia Giovenardi
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Renata Padilha Guedes
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil.
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil.
| |
Collapse
|
5
|
Khawagi WY, Al-Kuraishy HM, Hussein NR, Al-Gareeb AI, Atef E, Elhussieny O, Alexiou A, Papadakis M, Jabir MS, Alshehri AA, Saad HM, Batiha GES. Depression and type 2 diabetes: A causal relationship and mechanistic pathway. Diabetes Obes Metab 2024; 26:3031-3044. [PMID: 38802993 DOI: 10.1111/dom.15630] [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: 03/09/2024] [Revised: 04/16/2024] [Accepted: 04/16/2024] [Indexed: 05/29/2024]
Abstract
Depression is a mood disorder that may increase risk for the development of insulin resistance (IR) and type 2 diabetes (T2D), and vice versa. However, the mechanistic pathway linking depression and T2D is not fully elucidated. The aim of this narrative review, therefore, was to discuss the possible link between depression and T2D. The coexistence of T2D and depression is twice as great compared to the occurrence of either condition independently. Hyperglycaemia and dyslipidaemia promote the incidence of depression by enhancing inflammation and reducing brain serotonin (5-hydroxytryptamine [5HT]). Dysregulation of insulin signalling in T2D impairs brain 5HT signalling, leading to the development of depression. Furthermore, depression is associated with the development of hyperglycaemia and poor glycaemic control. Psychological stress and depression promote the development of T2D. In conclusion, T2D could be a potential risk factor for the development of depression through the induction of inflammatory reactions and oxidative stress that affect brain neurotransmission. In addition, chronic stress in depression may induce the development of T2D through dysregulation of the hypothalamic-pituitary-adrenal axis and increase circulating cortisol levels, which triggers IR and T2D.
Collapse
Affiliation(s)
- Wael Y Khawagi
- Department of Clinical Pharmacy, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Nawar R Hussein
- College of Pharmacy, Pharmacology Department, Al-Farahidi University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Esraa Atef
- Respiratory Therapy Department, Mohammed Al-Mana College for Medical Sciences, Dammam, Saudi Arabia
| | - Omnya Elhussieny
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matruh, Egypt
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University Chandigarh-Ludhiana Highway, Mohali, India
- Department of Research and Development, Funogen, Athens, Greece
- Department of Research and Development, AFNP Med, Wien, Austria
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, New South Wales, Australia
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Wuppertal, Germany
| | - Majid S Jabir
- Applied Science Department, University of Technology, Baghdad, Iraq
| | - Abdullah A Alshehri
- Department of Clinical Pharmacy, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matruh, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| |
Collapse
|
6
|
Jaykumar AB, Binns D, Taylor CA, Anselmo A, Birnbaum SG, Huber KM, Cobb MH. WNKs regulate mouse behavior and alter central nervous system glucose uptake and insulin signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.09.598125. [PMID: 38915673 PMCID: PMC11195145 DOI: 10.1101/2024.06.09.598125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Certain areas of the brain involved in episodic memory and behavior, such as the hippocampus, express high levels of insulin receptors and glucose transporter-4 (GLUT4) and are responsive to insulin. Insulin and neuronal glucose metabolism improve cognitive functions and regulate mood in humans. Insulin-dependent GLUT4 trafficking has been extensively studied in muscle and adipose tissue, but little work has demonstrated either how it is controlled in insulin-responsive brain regions or its mechanistic connection to cognitive functions. In this study, we demonstrate that inhibition of WNK (With-No-lysine (K)) kinases improves learning and memory in mice. Neuronal inhibition of WNK enhances in vivo hippocampal glucose uptake. Inhibition of WNK enhances insulin signaling output and insulin-dependent GLUT4 trafficking to the plasma membrane in mice primary neuronal cultures and hippocampal slices. Therefore, we propose that the extent of neuronal WNK kinase activity has an important influence on learning, memory and anxiety-related behaviors, in part, by modulation of neuronal insulin signaling.
Collapse
Affiliation(s)
- Ankita B. Jaykumar
- Departments of Pharmacology, UT Southwestern Medical Center, Dallas, USA
| | - Derk Binns
- Departments of Pharmacology, UT Southwestern Medical Center, Dallas, USA
| | - Clinton A. Taylor
- Departments of Pharmacology, UT Southwestern Medical Center, Dallas, USA
| | - Anthony Anselmo
- Departments of Pharmacology, UT Southwestern Medical Center, Dallas, USA
| | - Shari G. Birnbaum
- Departments of Peter O’Donnell Jr. Brain Institute and Psychiatry, UT Southwestern Medical Center, Dallas, USA
| | | | - Melanie H. Cobb
- Departments of Pharmacology, UT Southwestern Medical Center, Dallas, USA
| |
Collapse
|
7
|
Singh K, Das S, Sutradhar S, Howard J, Ray K. Insulin signaling accelerates the anterograde movement of Rab4 vesicles in axons through Klp98A/KIF16B recruitment via Vps34-PI3Kinase. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.24.590898. [PMID: 38895253 PMCID: PMC11185528 DOI: 10.1101/2024.04.24.590898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Rab4 GTPase organizes endosomal sorting essential for maintaining the balance between recycling and degradative pathways. Rab4 localizes to many cargos whose transport in neurons is critical for regulating neurotransmission and neuronal health. Furthermore, elevated Rab4 levels in the CNS are associated with synaptic atrophy and neurodegeneration in Drosophila and humans, respectively. However, how the transport of Rab4-associated vesicles is regulated in neurons remains unknown. Using in vivo time-lapse imaging of Drosophila larvae, we show that activation of insulin signaling via Dilp2 and dInR increases the anterograde velocity, run length, and flux of Rab4 vesicles in the axons. Molecularly, we show that activation of neuronal insulin signaling further activates Vps34, elevates the levels of PI(3)P on Rab4-associated vesicles, recruits Klp98A (a PI(3)P-binding kinesin-3 motor) and activates their anterograde transport. Together, these observations delineate the role of insulin signaling in regulating axonal transport and synaptic homeostasis.
Collapse
Affiliation(s)
- Kamaldeep Singh
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai - 400005, India
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT - 06520, United States
| | - Semanti Das
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai - 400005, India
| | - Sabyasachi Sutradhar
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT - 06520, United States
| | - Jonathon Howard
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT - 06520, United States
| | - Krishanu Ray
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai - 400005, India
- National Brain Research Centre, Manesar, Haryana – 122051, India
| |
Collapse
|
8
|
Avraham Y, Shapira-Furman T, Saklani R, Van Heukelom B, Carmel M, Vorobiev L, Lipsker L, Zwas DR, Berry EM, Domb AJ. Sustained insulin treatment restoring metabolic status, body weight, and cognition in an anorexia nervosa-like animal model in mice. Behav Brain Res 2024; 466:115001. [PMID: 38642861 DOI: 10.1016/j.bbr.2024.115001] [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: 11/02/2023] [Revised: 04/12/2024] [Accepted: 04/13/2024] [Indexed: 04/22/2024]
Abstract
INTRODUCTION Anorexia Nervosa (AN) is a psycho-socio-biological disease characterized by severe weight loss as result of dieting and hyperactivity. Effective treatments are scarce, despite its significant prevalence and mortality. AN patients show lower basal insulin levels and increased metabolic clearance, leading to weight loss, cognitive deficits, and hormonal imbalances. Low-dose polymer insulin could potentially reverse these effects by restoring brain function, reducing fear of weight gain, encouraging food intake, and restoring fat depots. This study evaluates an insulin delivery system designed for sustained release and AN treatment. METHODS AN-like model was established through dietary restriction (DR). On days 1-25, mice were on DR, and on days 26-31 they were on ad libitum regimen. An insulin-loaded delivery system was administered subcutaneously (1% w/w insulin). The impact of insulin treatment on gene expression in the hippocampus (cognition, regulation of stress, neurogenesis) and hypothalamus (eating behavior, mood) was assessed. Behavioral assays were conducted to evaluate motor activity and cognitive function. RESULTS The delivery system demonstrated sustained insulin release, maintaining therapeutic plasma levels. Diet restriction mice treated with the insulin delivery system showed body weight restoration. Gene expression analysis revealed enhanced expression of CB1 and CB2 genes associated with improved eating behavior and cognition, while POMC expression was reduced. Insulin-polymer treatment restored cognitive function and decreased hyperactivity in the AN-like model. CONCLUSION The PSA-RA-based insulin delivery system effectively restores metabolic balance, body weight, and cognitive function in the AN model. Its ability to steadily release insulin makes it a promising candidate for AN treatment."
Collapse
Affiliation(s)
- Yosefa Avraham
- Department of Metabolism and Human Nutrition, Braun School of Public Health, Hadassah-Hebrew University Medical School, Jerusalem 91120, Israel.
| | - Tovi Shapira-Furman
- Institute of Drug Research, School of Pharmacy Hadassah-Hebrew University Medical School, Jerusalem 91120, Israel
| | - Ravi Saklani
- Institute of Drug Research, School of Pharmacy Hadassah-Hebrew University Medical School, Jerusalem 91120, Israel
| | - Bob Van Heukelom
- Department of Neurology, Gelderse Vallei Hospital, 6716 RP, the Netherlands
| | - Moshe Carmel
- Department of Metabolism and Human Nutrition, Braun School of Public Health, Hadassah-Hebrew University Medical School, Jerusalem 91120, Israel
| | - Lia Vorobiev
- Department of Metabolism and Human Nutrition, Braun School of Public Health, Hadassah-Hebrew University Medical School, Jerusalem 91120, Israel
| | - Leah Lipsker
- Department of Metabolism and Human Nutrition, Braun School of Public Health, Hadassah-Hebrew University Medical School, Jerusalem 91120, Israel
| | - Donna R Zwas
- Linda Joy Pollin Cardiovascular Wellness Center for Women, Heart Institute, Hadassah University Medical Center, Jerusalem, Israel
| | - Elliot M Berry
- Department of Metabolism and Human Nutrition, Braun School of Public Health, Hadassah-Hebrew University Medical School, Jerusalem 91120, Israel
| | - Abraham J Domb
- Institute of Drug Research, School of Pharmacy Hadassah-Hebrew University Medical School, Jerusalem 91120, Israel
| |
Collapse
|
9
|
Caldarelli M, Rio P, Marrone A, Ocarino F, Chiantore M, Candelli M, Gasbarrini A, Gambassi G, Cianci R. Gut-Brain Axis: Focus on Sex Differences in Neuroinflammation. Int J Mol Sci 2024; 25:5377. [PMID: 38791415 PMCID: PMC11120930 DOI: 10.3390/ijms25105377] [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: 03/30/2024] [Revised: 05/11/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
In recent years, there has been a growing interest in the concept of the "gut-brain axis". In addition to well-studied diseases associated with an imbalance in gut microbiota, such as cancer, chronic inflammation, and cardiovascular diseases, research is now exploring the potential role of gut microbial dysbiosis in the onset and development of brain-related diseases. When the function of the intestinal barrier is altered by dysbiosis, the aberrant immune system response interacts with the nervous system, leading to a state of "neuroinflammation". The gut microbiota-brain axis is mediated by inflammatory and immunological mechanisms, neurotransmitters, and neuroendocrine pathways. This narrative review aims to illustrate the molecular basis of neuroinflammation and elaborate on the concept of the gut-brain axis by virtue of analyzing the various metabolites produced by the gut microbiome and how they might impact the nervous system. Additionally, the current review will highlight how sex influences these molecular mechanisms. In fact, sex hormones impact the brain-gut microbiota axis at different levels, such as the central nervous system, the enteric nervous one, and enteroendocrine cells. A deeper understanding of the gut-brain axis in human health and disease is crucial to guide diagnoses, treatments, and preventive interventions.
Collapse
Affiliation(s)
- Mario Caldarelli
- Department of Translational Medicine and Surgery, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Pierluigi Rio
- Department of Translational Medicine and Surgery, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Andrea Marrone
- Department of Translational Medicine and Surgery, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Francesca Ocarino
- Department of Translational Medicine and Surgery, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Monica Chiantore
- Department of Translational Medicine and Surgery, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Marcello Candelli
- Department of Emergency, Anesthesiological and Reanimation Sciences, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Department of Translational Medicine and Surgery, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Giovanni Gambassi
- Department of Translational Medicine and Surgery, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Rossella Cianci
- Department of Translational Medicine and Surgery, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| |
Collapse
|
10
|
Basatinya AM, Sajedianfard J, Nazifi S, Hosseinzadeh S. The analgesic effects of insulin and its disorders in streptozotocin-induced short-term diabetes. Physiol Rep 2024; 12:e16009. [PMID: 38639646 PMCID: PMC11027902 DOI: 10.14814/phy2.16009] [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: 12/29/2023] [Revised: 04/01/2024] [Accepted: 04/01/2024] [Indexed: 04/20/2024] Open
Abstract
Evidence suggests that insulin resistance plays an important role in developing diabetes complications. The association between insulin resistance and pain perception is less well understood. This study aimed to investigate the effects of peripheral insulin deficiency on pain pathways in the brain. Diabetes was induced in 60 male rats with streptozotocin (STZ). Insulin was injected into the left ventricle of the brain by intracerebroventricular (ICV) injection, then pain was induced by subcutaneous injection of 2.5% formalin. Samples were collected at 4 weeks after STZ injection. Dopamine (DA), serotonin, reactive oxygen species (ROS), and mitochondrial glutathione (mGSH) were measured by ELISA, and gene factors were assessed by RT-qPCR. In diabetic rats, the levels of DA, serotonin, and mGSH decreased in the nuclei of the thalamus, raphe magnus, and periaqueductal gray, and the levels of ROS increased. In addition, the levels of expression of the neuron-specific enolase and receptor for advanced glycation end genes increased, but the expression of glial fibrillary acidic protein expression was reduced. These results support the findings that insulin has an analgesic effect in non-diabetic rats, as demonstrated by the formalin test. ICV injection of insulin reduces pain sensation, but this was not observed in diabetic rats, which may be due to cell damage ameliorated by insulin.
Collapse
Affiliation(s)
| | - Javad Sajedianfard
- Department of Basic Sciences, School of Veterinary MedicineShiraz UniversityShirazIran
| | - Saeed Nazifi
- Department of Clinical Science, School of Veterinary MedicineShiraz UniversityShirazIran
| | - Saied Hosseinzadeh
- Department of Food Hygiene and Public Health, School of Veterinary MedicineShiraz UniversityShirazIran
| |
Collapse
|
11
|
Wilbrecht L, Lin WC, Callahan K, Bateson M, Myers K, Ross R. Experimental biology can inform our understanding of food insecurity. J Exp Biol 2024; 227:jeb246215. [PMID: 38449329 PMCID: PMC10949070 DOI: 10.1242/jeb.246215] [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] [Indexed: 03/08/2024]
Abstract
Food insecurity is a major public health issue. Millions of households worldwide have intermittent and unpredictable access to food and this experience is associated with greater risk for a host of negative health outcomes. While food insecurity is a contemporary concern, we can understand its effects better if we acknowledge that there are ancient biological programs that evolved to respond to the experience of food scarcity and uncertainty, and they may be particularly sensitive to food insecurity during development. Support for this conjecture comes from common findings in several recent animal studies that have modeled insecurity by manipulating predictability of food access in various ways. Using different experimental paradigms in different species, these studies have shown that experience of insecure access to food can lead to changes in weight, motivation and cognition. Some of these studies account for changes in weight through changes in metabolism, while others observe increases in feeding and motivation to work for food. It has been proposed that weight gain is an adaptive response to the experience of food insecurity as 'insurance' in an uncertain future, while changes in motivation and cognition may reflect strategic adjustments in foraging behavior. Animal studies also offer the opportunity to make in-depth controlled studies of mechanisms and behavior. So far, there is evidence that the experience of food insecurity can impact metabolic efficiency, reproductive capacity and dopamine neuron synapses. Further work on behavior, the central and peripheral nervous system, the gut and liver, along with variation in age of exposure, will be needed to better understand the full body impacts of food insecurity at different stages of development.
Collapse
Affiliation(s)
- Linda Wilbrecht
- Department of Psychology, University of California, Berkeley, Berkeley, CA 94720-1650, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Wan Chen Lin
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Kathryn Callahan
- Psychiatric Research Institute of Montefiore and Einstein, Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - Melissa Bateson
- Bioscience Institute, University of Newcastle, Newcastle upon Tyne, NE2 4HH, UK
| | - Kevin Myers
- Department of Psychology and Programs in Animal Behavior and Neuroscience, Bucknell University, Lewisburg, PA 17837, USA
| | - Rachel Ross
- Psychiatric Research Institute of Montefiore and Einstein, Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
- Department of Psychiatry, Montefiore Medical Center, Bronx, New York, NY 10467, USA
| |
Collapse
|
12
|
Nicolazzi L, Gilbert L, Horsch A, Quansah DY, Puder JJ. Trajectories and associations of symptoms of mental health and well-being with insulin resistance and metabolic health in women with gestational diabetes. Psychoneuroendocrinology 2024; 160:106919. [PMID: 38091918 DOI: 10.1016/j.psyneuen.2023.106919] [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: 07/26/2023] [Revised: 11/17/2023] [Accepted: 12/05/2023] [Indexed: 01/02/2024]
Abstract
BACKGROUND Gestational diabetes mellitus (GDM) is characterized by increased insulin resistance and carries perinatal and long-term risks for the mother and her offspring. There is a link between perinatal depression or anxiety and GDM. Mental health problems are associated with higher insulin resistance and could explain the underlying association between GDM and depression or anxiety symptoms. We investigated the trajectories and associations between symptoms of mental health and well-being with insulin resistance and metabolic health in women with GDM. METHODS This study included the control group (n = 106) of a randomized controlled trial in women with GDM that were followed-up during pregnancy and up to 1-year postpartum. We measured symptoms of mental health (Edinburgh Postnatal Depression Scale (EPDS), Anxiety subscale of the Hospital Anxiety and Depression Scale (HADS-A), well-being (The World Health Organization Well-Being Index (WHO-5)) and metabolic health, including insulin resistance variables (HOMA-insulin resistance (IR) and Matsuda Index of insulin sensitivity) as well as weight during pregnancy and in the postpartum. RESULTS Participants' pre pregnancy weight and BMI were 69.7 kg ± 16.1 and 25.9 kg/m2 ± 5.5 respectively. HOMA-IR was higher during pregnancy compared to 6-8 weeks postpartum and increased between 6-8 weeks and 1-year postpartum (all p < 0.05). Matsuda index decreased between 6-8 weeks and 1-year postpartum (p < 0.001). EPDS scores decreased between pregnancy and both 6-8 weeks and 1-year postpartum (all p < 0.05). HADS-A scores did not change between pregnancy and the postpartum. WHO-5 scores improved significantly from pregnancy and both 6-8 weeks and 1-year postpartum (p < 0.001). Correlation coefficients within outcome at the three different time points were high for metabolic measures and ranged between 0.94 and 0.96 for weight, from 0.77 to 0.89 for HOMA-IR and 0.64 for the Matsuda index (all p < 0.001). Mental health and well-being variables were moderately correlated in all three time points including r = 0.36-0.55 for the EPDS (p < 0.001), r = 0.58 for HADS (p < 0.001), and r = 0.43-0.52 for the WHO-5 (p < 0.01). After adjustment for age and pre-pregnancy BMI, Matsuda index was negatively associated with EPDS scores and positively associated to WHO-5 scores at 6-8 weeks postpartum. No other association between insulin resistance and mental health or well-being outcomes were found. CONCLUSION While insulin resistance fluctuated with values being lowest in the early postpartum and increasing thereafter, both depression and well-being scores decreased between pregnancy and the postpartum and did not change in the postpartum period. Intraindividual variability was larger for mental health and well-being than for metabolic health outcomes at different time points, indicating a higher plasticity for mental health and well-being outcomes that could be acted upon. We found only few associations between mental health and well-being and metabolic health outcomes.
Collapse
Affiliation(s)
- Ludmila Nicolazzi
- Department of Medicine, Internal Medicine service, Lausanne University Hospital, Lausanne, Switzerland.
| | - Leah Gilbert
- Obstetric service, Department Woman-Mother-Child, Lausanne University Hospital, Lausanne, Switzerland.
| | - Antje Horsch
- Institute of Higher Education and Research in Healthcare (IUFRS), University of Lausanne, Switzerland; Neonatalogy Service, Department Woman-Mother-Child, Lausanne University Hospital, Lausanne, Switzerland.
| | - Dan Yedu Quansah
- Obstetric service, Department Woman-Mother-Child, Lausanne University Hospital, Lausanne, Switzerland.
| | - Jardena J Puder
- Obstetric service, Department Woman-Mother-Child, Lausanne University Hospital, Lausanne, Switzerland.
| |
Collapse
|
13
|
Vaganova AN, Shemyakova TS, Lenskaia KV, Rodionov RN, Steenblock C, Gainetdinov RR. Trace Amine-Associated Receptors and Monoamine-Mediated Regulation of Insulin Secretion in Pancreatic Islets. Biomolecules 2023; 13:1618. [PMID: 38002300 PMCID: PMC10669413 DOI: 10.3390/biom13111618] [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: 09/27/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
Currently, metabolic syndrome treatment includes predominantly pharmacological symptom relief and complex lifestyle changes. Trace amines and their receptor systems modulate signaling pathways of dopamine, norepinephrine, and serotonin, which are involved in the pathogenesis of this disorder. Trace amine-associated receptor 1 (TAAR1) is expressed in endocrine organs, and it was revealed that TAAR1 may regulate insulin secretion in pancreatic islet β-cells. For instance, accumulating data demonstrate the positive effect of TAAR1 agonists on the dynamics of metabolic syndrome progression and MetS-associated disease development. The role of other TAARs (TAAR2, TAAR5, TAAR6, TAAR8, and TAAR9) in the islet's function is much less studied. In this review, we summarize the evidence of TAARs' contribution to the metabolic syndrome pathogenesis and regulation of insulin secretion in pancreatic islets. Additionally, by the analysis of public transcriptomic data, we demonstrate that TAAR1 and other TAAR receptors are expressed in the pancreatic islets. We also explore associations between the expression of TAARs mRNA and other genes in studied samples and demonstrate the deregulation of TAARs' functional associations in patients with metabolic diseases compared to healthy donors.
Collapse
Affiliation(s)
- Anastasia N. Vaganova
- Institute of Translational Biomedicine, St. Petersburg State University, 199034 St. Petersburg, Russia; (A.N.V.); (T.S.S.)
- St. Petersburg State University Hospital, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Taisiia S. Shemyakova
- Institute of Translational Biomedicine, St. Petersburg State University, 199034 St. Petersburg, Russia; (A.N.V.); (T.S.S.)
| | - Karina V. Lenskaia
- Department of Medicine, St. Petersburg State University, Universitetskaya nab. 7/9, 199034 St. Petersburg, Russia;
| | - Roman N. Rodionov
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (R.N.R.); (C.S.)
| | - Charlotte Steenblock
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (R.N.R.); (C.S.)
| | - Raul R. Gainetdinov
- Institute of Translational Biomedicine, St. Petersburg State University, 199034 St. Petersburg, Russia; (A.N.V.); (T.S.S.)
- St. Petersburg State University Hospital, St. Petersburg State University, 199034 St. Petersburg, Russia
| |
Collapse
|
14
|
Al-Kuraishy HM, Jabir MS, Albuhadily AK, Al-Gareeb AI, Rafeeq MF. The link between metabolic syndrome and Alzheimer disease: A mutual relationship and long rigorous investigation. Ageing Res Rev 2023; 91:102084. [PMID: 37802319 DOI: 10.1016/j.arr.2023.102084] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/01/2023] [Accepted: 10/03/2023] [Indexed: 10/08/2023]
Abstract
It has been illustrated that metabolic syndrome (MetS) is associated with Alzheimer disease (AD) neuropathology. Components of MetS including central obesity, hypertension, insulin resistance (IR), and dyslipidemia adversely affect the pathogenesis of AD by different mechanisms including activation of renin-angiotensin system (RAS), inflammatory signaling pathways, neuroinflammation, brain IR, mitochondrial dysfunction, and oxidative stress. MetS exacerbates AD neuropathology, and targeting of molecular pathways in MetS by pharmacological approach could a novel therapeutic strategy in the management of AD in high risk group. However, the underlying mechanisms of these pathways in AD neuropathology are not completely clarified. Therefore, this review aims to elucidate the association between MetS and AD regarding the oxidative and inflammatory mechanistic pathways.
Collapse
Affiliation(s)
- Haydar M Al-Kuraishy
- Department of Clinical pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Majid S Jabir
- Department of Applied science, University of technology, Iraq.
| | - Ali K Albuhadily
- Department of Clinical pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of Clinical pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | | |
Collapse
|
15
|
Koning E, McDonald A, Bambokian A, Gomes FA, Vorstman J, Berk M, Fabe J, McIntyre RS, Milev R, Mansur RB, Brietzke E. The concept of "metabolic jet lag" in the pathophysiology of bipolar disorder: implications for research and clinical care. CNS Spectr 2023; 28:571-580. [PMID: 36503605 DOI: 10.1017/s1092852922001195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Bipolar disorder (BD) is a potentially chronic mental disorder marked by recurrent depressive and manic episodes, circadian rhythm disruption, and changes in energetic metabolism. "Metabolic jet lag" refers to a state of shift in circadian patterns of energy homeostasis, affecting neuroendocrine, immune, and adipose tissue function, expressed through behavioral changes such as irregularities in sleep and appetite. Risk factors include genetic variation, mitochondrial dysfunction, lifestyle factors, poor gut microbiome health and abnormalities in hunger, satiety, and hedonistic function. Evidence suggests metabolic jet lag is a core component of BD pathophysiology, as individuals with BD frequently exhibit irregular eating rhythms and circadian desynchronization of their energetic metabolism, which is associated with unfavorable clinical outcomes. Although current diagnostic criteria lack any assessment of eating rhythms, technological advancements including mobile phone applications and ecological momentary assessment allow for the reliable tracking of biological rhythms. Overall, methodological refinement of metabolic jet lag assessment will increase knowledge in this field and stimulate the development of interventions targeting metabolic rhythms, such as time-restricted eating.
Collapse
Affiliation(s)
- Elena Koning
- Centre for Neurosciences Studies (CNS), Queen's University, Kingston, ON, Canada
| | - Alexandra McDonald
- Centre for Neurosciences Studies (CNS), Queen's University, Kingston, ON, Canada
| | - Alexander Bambokian
- Centre for Neurosciences Studies (CNS), Queen's University, Kingston, ON, Canada
| | - Fabiano A Gomes
- Department of Psychiatry, Queen's University School of Medicine, Kingston, ON, Canada
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Jacob Vorstman
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Michael Berk
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Jennifer Fabe
- Department of Neurology, McMaster Children's Hospital, Hamilton, ON, Canada
| | - Roger S McIntyre
- Department of Psychiatry and Pharmacology, University of Toronto, The Brain and Cognition Discovery Foundation, Toronto, Canada
| | - Roumen Milev
- Centre for Neurosciences Studies (CNS), Queen's University, Kingston, ON, Canada
- Department of Psychiatry, Queen's University School of Medicine, Kingston, ON, Canada
- Department of Psychiatry, Providence Care Hospital, Kingston, ON, Canada
| | - Rodrigo B Mansur
- Department of Psychiatry and Pharmacology, University of Toronto, The Brain and Cognition Discovery Foundation, Toronto, Canada
| | - Elisa Brietzke
- Centre for Neurosciences Studies (CNS), Queen's University, Kingston, ON, Canada
- Department of Psychiatry, Queen's University School of Medicine, Kingston, ON, Canada
| |
Collapse
|
16
|
Liu T, Peng MM, Wong FHC, Leung DKY, Zhang W, Wong GHY, Lum TYS. Differential Associations Between Depressive Symptom-Domains With Anxiety, Loneliness, and Cognition in a Sample of Community Older Chinese Adults: A Multiple Indicators Multiple Causes Approach. Innov Aging 2023; 7:igad075. [PMID: 37727600 PMCID: PMC10506173 DOI: 10.1093/geroni/igad075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Indexed: 09/21/2023] Open
Abstract
Background and Objectives Depressive symptoms are common in older adults, and often co-occur with other mental health problems. However, knowledge about depressive symptom-domains and their associations with other conditions is limited. This study examined depressive symptom-domains and associations with anxiety, cognition, and loneliness. Research Design and Methods A sample of 3,795 participants aged 60 years and older were recruited from the community in Hong Kong. They were assessed for depressive symptoms (Patient Health Questionnaire-9 [PHQ-9]), anxiety (Generalized Anxiety Disorder 7-item), loneliness (UCLA 3-item), and cognition (Montreal Cognitive Assessment 5-Minute Protocol). Summary descriptive statistics were calculated, followed by confirmatory factor analysis of PHQ-9. Multiple Indicators Multiple Causes analysis was used to examine the associations between mental health conditions in the general sample and subgroups based on depressive symptom severity. Results A 4-factor model based on the Research Domain Criteria showed the best model fit of PHQ-9 (χ2/df = 10.63, Root-Mean-Square Error of Approximation = 0.05, Comparative Fit Index = 0.96, Tucker-Lewis Index = 0.93). After adjusting for demographics, 4 depressive symptom-domains were differentially associated with anxiety, loneliness, and cognition across different depression severity groups. The Negative Valance Systems and Internalizing domain (NVS-I; guilt and self-harm) were consistently associated with anxiety (β = 0.45, 0.44) and loneliness (β = 0.11, 0.27) regardless of depression severity (at risk/mild vs moderate and more severe, respectively, all p < .001). Discussion and Implications The consistent associations between the NVS-I domain of depression with anxiety and loneliness warrant attention. Simultaneous considerations of depressive symptom-domains and symptom severity are needed for designing more personalized care. Clinical Trials Registration Number NCT03593889.
Collapse
Affiliation(s)
- Tianyin Liu
- Department of Social Work and Social Administration, The University of Hong Kong, Hong Kong, China
| | - Man-Man Peng
- Institute of Advanced Studies in Humanities and Social Sciences, Beijing Normal University, Zhuhai, China
| | - Frankie H C Wong
- Philip Merrill College of Journalism, University of Maryland, College Park, Maryland, USA
| | - Dara K Y Leung
- Sau Po Centre on Ageing, The University of Hong Kong, Hong Kong, China
| | - Wen Zhang
- School of Nursing and Health Studies, Hong Kong Metropolitan University, Hong Kong, China
| | - Gloria H Y Wong
- Department of Social Work and Social Administration, The University of Hong Kong, Hong Kong, China
| | - Terry Y S Lum
- Department of Social Work and Social Administration, The University of Hong Kong, Hong Kong, China
- Sau Po Centre on Ageing, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
17
|
Yeo J. Influence of food-derived bioactives on gut microbiota compositions and their metabolites by focusing on neurotransmitters. Food Sci Biotechnol 2023; 32:1019-1027. [PMID: 37215258 PMCID: PMC10195957 DOI: 10.1007/s10068-023-01293-2] [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: 12/31/2022] [Revised: 02/21/2023] [Accepted: 03/03/2023] [Indexed: 04/03/2023] Open
Abstract
The behavior of gut microbiota is closely involved in sustaining balanced immune and metabolic homeostasis, and the dysbiosis of gut microbiota can lead to severe disease. Foods and dietary patterns are the primary drivers in shaping/designing gut microbiota compositions and their metabolites across the lifetime. This indicates the importance of functional molecules present in the food matrix in the life of gut microbiota and their influence on the host's biological system. In this contribution, the effects of different dietary choices and bioactive compounds (i.e., phenolics, vitamins, carotenoids) on gut microbiome compositions and their metabolites are comprehensively discussed by focusing on neurotransmitters. This study may provide useful information that fills a gap in understanding the role of the gut microbiota and its alterations as affected by foods and food-derived bioactives.
Collapse
Affiliation(s)
- JuDong Yeo
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul Campus, Seoul, 05029 Republic of Korea
| |
Collapse
|
18
|
Silva Júnior AED, Bueno NB. Letter to the editor: Further evidence that food addiction is associated with type 2 diabetes mellitus. Clin Nutr 2023; 42:1159-1160. [PMID: 37202339 DOI: 10.1016/j.clnu.2023.05.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/10/2023] [Indexed: 05/20/2023]
Affiliation(s)
- André Eduardo da Silva Júnior
- Postgraduate Program in Nutrition, Escola Paulista de Medicina, Universidade Federal de São Paulo, SP, Brazil; Laboratório de Nutrição e Metabolismo (LANUM), Faculdade de Nutrição, Universidade Federal de Alagoas, Maceió, AL, Brazil.
| | - Nassib Bezerra Bueno
- Postgraduate Program in Nutrition, Escola Paulista de Medicina, Universidade Federal de São Paulo, SP, Brazil; Laboratório de Nutrição e Metabolismo (LANUM), Faculdade de Nutrição, Universidade Federal de Alagoas, Maceió, AL, Brazil
| |
Collapse
|
19
|
Alrouji M, Al-Kuraishy HM, Al-Buhadily AK, Al-Gareeb AI, Elekhnawy E, Batiha GES. DPP-4 inhibitors and type 2 diabetes mellitus in Parkinson's disease: a mutual relationship. Pharmacol Rep 2023:10.1007/s43440-023-00500-5. [PMID: 37269487 DOI: 10.1007/s43440-023-00500-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 05/17/2023] [Accepted: 05/19/2023] [Indexed: 06/05/2023]
Abstract
Parkinson's disease (PD) usually occurs due to the degeneration of dopaminergic neurons in the substantia nigra (SN). Management of PD is restricted to symptomatic improvement. Consequently, a novel treatment for managing motor and non-motor symptoms in PD is necessary. Abundant findings support the protection of dipeptidyl peptidase 4 (DPP-4) inhibitors in PD. Consequently, this study aims to reveal the mechanism of DPP-4 inhibitors in managing PD. DPP-4 inhibitors are oral anti-diabetic agents approved for managing type 2 diabetes mellitus (T2DM). T2DM is linked with an increased chance of the occurrence of PD. Extended usage of DPP-4 inhibitors in T2DM patients may attenuate the development of PD by inhibiting inflammatory and apoptotic pathways. Thus, DPP-4 inhibitors like sitagliptin could be a promising treatment against PD neuropathology via anti-inflammatory, antioxidant, and anti-apoptotic impacts. DPP-4 inhibitors, by increasing endogenous GLP-1, can also reduce memory impairment in PD. In conclusion, the direct effects of DPP-4 inhibitors or indirect effects through increasing circulating GLP-1 levels could be an effective therapeutic strategy in treating PD patients through modulation of neuroinflammation, oxidative stress, mitochondrial dysfunction, and neurogenesis.
Collapse
Affiliation(s)
- Mohammed Alrouji
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Shaqra, 11961, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, Baghdad, Iraq
| | - Ali K Al-Buhadily
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, Baghdad, Iraq
| | - Engy Elekhnawy
- Pharmaceutical Microbiology Department, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AL Beheira, Egypt.
| |
Collapse
|
20
|
Ding K, Zeng J, Zhang X, Wang Y, Liang F, Wang L, Guo T, Moore JB, Li R. Changes in Plant-based Dietary Quality and Subsequent Risk of Cognitive Impairment among Older Chinese Adults: A National Community-based Cohort Study. Am J Clin Nutr 2023:S0002-9165(23)57911-9. [PMID: 37187294 DOI: 10.1016/j.ajcnut.2023.05.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 05/08/2023] [Accepted: 05/11/2023] [Indexed: 05/17/2023] Open
Abstract
BACKGROUND It is unclear how changes in plant-based dietary quality are linked to subsequent risk of cognitive impairment. This study aims to evaluate this relationship using data from the Chinese Longitudinal Healthy Longevity Survey (CLHLS). METHODS A total of 6662 participants free of cognitive impairment in 2008 were included and followed up to 2018. Plant-based dietary quality was assessed by three indices: overall plant-based diet index (PDI), healthful plant-based diet index (hPDI), and unhealthful plant-based diet index (uPDI). Changes in plant-based diet quality from 2008 to 2011 were classified into quintiles. We assessed incident cognitive impairment (from 2011 to 2018) by using the Mini-Mental State Examination (MMSE). Cox proportional-hazards models were performed. RESULTS We recorded 1571 incident cases of cognitive impairment during a median of 10 years of follow-up. Compared with participants whose plant-based diet had no change or was relatively stable over three years, the full-adjusted hazard ratios (HRs) with 95% confidence intervals (CI) for cognitive impairment were 0.77 (0.64, 0.93), 0.72 (0.60, 0.86), and 1.50 (1.27, 1.77) among participants with a large increase in PDI, hPDI, and uPDI, respectively. The HRs with 95% CI were 1.22 (1.02, 1.44), 1.30 (1.11, 1.54), and 0.80 (0.67, 0.96) among participants with a large decrease in PDI, hPDI, and uPDI, respectively. Every 10-point increase in PDI and hPDI was associated with 26% and 30% lower risk of cognitive impairment, while every 10-point increase in uPDI was associated with 36% higher risk. CONCLUSIONS Older adults with increased adherence to an overall plant-based diet and a healthful plant-based diet over three years have a lower risk of cognitive impairment, while those with increased adherence to an unhealthy plant-based diet had a higher risk of cognitive impairment.
Collapse
Affiliation(s)
- Kai Ding
- School of Public Health, Wuhan University, Wuhan 430071, China
| | - Jing Zeng
- School of Public Health, Wuhan University, Wuhan 430071, China
| | - Xinge Zhang
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong Faculty of Medicine, Hong Kong Special Administrative Region, China
| | - Yechuang Wang
- School of Public Health, Wuhan University, Wuhan 430071, China
| | - Fang Liang
- School of Public Health, Wuhan University, Wuhan 430071, China
| | - Lei Wang
- School of Public Health, Wuhan University, Wuhan 430071, China
| | - Taotao Guo
- School of Public Health, Wuhan University, Wuhan 430071, China
| | - Justin B Moore
- Department of Epidemiology & Prevention, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Rui Li
- School of Public Health, Wuhan University, Wuhan 430071, China; Center for Nurturing Care Research, School of Nursing; Center for Healthy Aging, School of Nursing, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
21
|
Römer SS, Bliokas V, Teo JT, Thomas SJ. Food addiction, hormones and blood biomarkers in humans: A systematic literature review. Appetite 2023; 183:106475. [PMID: 36716820 DOI: 10.1016/j.appet.2023.106475] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 01/15/2023] [Accepted: 01/23/2023] [Indexed: 01/29/2023]
Abstract
BACKGROUND Food addiction may play a role in rising obesity rates in connection with obesogenic environments and processed food availability, however the concept of food addiction remains controversial. While animal studies show evidence for addictive processes in relation to processed foods, most human studies are psychologically focussed and there is a need to better understand evidence for biological mechanisms of food addiction in humans. Several key hormones are implicated in models of food addiction, due to their key roles in feeding, energy metabolism, stress and addictive behaviours. This systematic literature review examines evidence for relationships between food addiction, hormones and other blood biomarkers. METHODS A series of literature searches was performed in Scopus, PsychInfo, MedLine, ProQuest, CINAHL and Web of Science. A total of 3111 articles were found, of which 1045 were duplicates. Articles were included if they contained a psychometric measurement of food addiction, such as the Yale Food Addiction Scale, as well as addressed the association between FA and hormones or blood biomarkers in humans. Articles were assessed for eligibility by two independent reviewers. RESULTS Sixteen studies were identified that examined relationships between food addiction and blood biomarkers, published between 2015 and 2021. Significant findings were reported for leptin, ghrelin, cortisol, insulin and glucose, oxytocin, cholesterol, plasma dopamine, thyroid stimulating hormone (TSH), haemoglobin A1c (HbA1c), triglyceride (TG), amylin, tumour necrosis factor alpha (TNF- α) and cholecystokinin (CCK). Methodological issues included small sample sizes and variation in obesity status, sex and mental health-related comorbidities. Due to methodological limitations, definite connections between FA, hormones and other blood biomarkers cannot yet be determined. CONCLUSION This systematic review identified preliminary evidence linking FA symptoms to hormones and other blood biomarkers related to feeding, addiction, and stress. However, due to the small number of studies and methodological limitations, further research is needed to evaluate biopsychosocial models of FA and to resolve controversies.
Collapse
Affiliation(s)
- Stephanie Sophie Römer
- School of Psychology, Faculty of the Arts, Social Sciences and Humanities, University of Wollongong, Australia.
| | - Vida Bliokas
- School of Psychology, Faculty of the Arts, Social Sciences and Humanities, University of Wollongong, Australia; Illawarra Health and Medical Research Institute, University of Wollongong, 2522, Australia.
| | - Jillian Terese Teo
- School of Psychology, Faculty of the Arts, Social Sciences and Humanities, University of Wollongong, Australia.
| | - Susan J Thomas
- Illawarra Health and Medical Research Institute, University of Wollongong, 2522, Australia; Graduate School of Medicine, Faculty of Science, Medicine and Health, University of Wollongong, Australia.
| |
Collapse
|
22
|
Marano G, Mazza M, Lisci FM, Ciliberto M, Traversi G, Kotzalidis GD, De Berardis D, Laterza L, Sani G, Gasbarrini A, Gaetani E. The Microbiota-Gut-Brain Axis: Psychoneuroimmunological Insights. Nutrients 2023; 15:nu15061496. [PMID: 36986226 PMCID: PMC10059722 DOI: 10.3390/nu15061496] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/18/2023] [Accepted: 03/19/2023] [Indexed: 03/30/2023] Open
Abstract
There is growing interest in the role that the intestinal microbiota and the related autoimmune processes may have in the genesis and presentation of some psychiatric diseases. An alteration in the communication of the microbiota-gut-brain axis, which constitutes a communicative model between the central nervous system (CNS) and the gastro-enteric tract, has been identified as one of the possible causes of some psychiatric diseases. The purpose of this narrative review is to describe evidence supporting a role of the gut microbiota in psychiatric diseases and the impact of diet on microbiota and mental health. Change in the composition of the gut microbiota could determine an increase in the permeability of the intestinal barrier, leading to a cytokine storm. This could trigger a systemic inflammatory activation and immune response: this series of events could have repercussions on the release of some neurotransmitters, altering the activity of the hypothalamic-pituitary-adrenal axis, and reducing the presence of trophic brain factors. Although gut microbiota and psychiatric disorders seem to be connected, more effort is needed to understand the potential causative mechanisms underlying the interactions between these systems.
Collapse
Affiliation(s)
- Giuseppe Marano
- Department of Geriatrics, Neuroscience and Orthopedics, Institute of Psychiatry and Psychology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Marianna Mazza
- Department of Geriatrics, Neuroscience and Orthopedics, Institute of Psychiatry and Psychology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Francesco Maria Lisci
- Department of Geriatrics, Neuroscience and Orthopedics, Institute of Psychiatry and Psychology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Michele Ciliberto
- Department of Geriatrics, Neuroscience and Orthopedics, Institute of Psychiatry and Psychology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Gianandrea Traversi
- Unit of Medical Genetics, Department of Laboratory Medicine, Fatebenefratelli Isola Tiberina-Gemelli Isola, 00168 Rome, Italy
| | - Georgios Demetrios Kotzalidis
- Department of Geriatrics, Neuroscience and Orthopedics, Institute of Psychiatry and Psychology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Sant'Andrea Hospital, Sapienza University of Rome, 00189 Rome, Italy
| | | | - Lucrezia Laterza
- CEMAD Digestive Diseases Center, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Gabriele Sani
- Department of Geriatrics, Neuroscience and Orthopedics, Institute of Psychiatry and Psychology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Internal Medicine and Gastroenterology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, 00168 Rome, Italy
| | - Eleonora Gaetani
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
23
|
Actions and Consequences of Insulin in the Striatum. Biomolecules 2023; 13:biom13030518. [PMID: 36979453 PMCID: PMC10046598 DOI: 10.3390/biom13030518] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 02/26/2023] [Accepted: 02/27/2023] [Indexed: 03/14/2023] Open
Abstract
Insulin crosses the blood–brain barrier to enter the brain from the periphery. In the brain, insulin has well-established actions in the hypothalamus, as well as at the level of mesolimbic dopamine neurons in the midbrain. Notably, insulin also acts in the striatum, which shows abundant expression of insulin receptors (InsRs) throughout. These receptors are found on interneurons and striatal projections neurons, as well as on glial cells and dopamine axons. A striking functional consequence of insulin elevation in the striatum is promoting an increase in stimulated dopamine release. This boosting of dopamine release involves InsRs on cholinergic interneurons, and requires activation of nicotinic acetylcholine receptors on dopamine axons. Opposing this dopamine-enhancing effect, insulin also increases dopamine uptake through the action of insulin at InsRs on dopamine axons. Insulin acts on other striatal cells as well, including striatal projection neurons and astrocytes that also influence dopaminergic transmission and striatal function. Linking these cellular findings to behavior, striatal insulin signaling is required for the development of flavor–nutrient learning, implicating insulin as a reward signal in the brain. In this review, we discuss these and other actions of insulin in the striatum, including how they are influenced by diet and other physio-logical states.
Collapse
|
24
|
Zhang T, Rao Q, Lin K, He Y, Cai J, Yang M, Xu Y, Hou L, Lin Y, Liu H. CYP2C19-rs4986893 confers risk to major depressive disorder and bipolar disorder in the Han Chinese population whereas ABCB1-rs1045642 acts as a protective factor. BMC Psychiatry 2023; 23:69. [PMID: 36698099 PMCID: PMC9875448 DOI: 10.1186/s12888-022-04514-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 12/30/2022] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Genetic risks may predispose individuals to major mood disorders differently. This study investigated the gene polymorphisms of previously reported candidate genes for major depressive disorder (MDD) and bipolar disorder (BPD) in the Han Chinese population. METHODS Twenty loci of 13 candidate genes were detected by MALDI-TOF mass spectrometry in 439 patients with MDD, 600 patients with BPD, and 464 healthy controls. The distribution of genotypes in alleles, Hardy-Weinberg equilibrium, and genetic association were analyzed using the PLINK software. The linkage of disequilibrium and haplotype analyses were performed using the Haploview software. RESULTS Out of the 20 loci analyzed, CYP2C19-rs4986893, ABCB1-rs1045642, and SCN2A-rs17183814 passed Bonferroni correction; their statistical powers were > 55%. The minor allele frequencies (MAF) of CYP2C19-rs4986893 in the MDD group (0.0547) and BPD group (0.0533) were higher than that of the control group (0.0259, P < 0.05), leading to the odds ratios (ORs) of MDD (2.178) and BPD (2.122), respectively. In contrast, the lower MAFs of ABCB1-rs1045642 were observed in both MDD (0.3599, OR = 0.726) and BPD (0.3700, OR = 0.758) groups than controls (0.4364, P < 0.05). The MDD group had a higher MAF of SCN2A-rs17183814 than controls (0.1743 vs. 0.1207, OR = 1.538, P < 0.05). Moreover, a G-A haplotype composed by CYP2C19-rs4986893 and -rs4244285 was associated with BPD (OR = 1.361, P < 0.01), and the A-G haplotype increased the risks to both MDD (OR = 2.306, P < 0.01) and BPD (OR = 2.332, P < 0.001). The CYP2C19 intermediate metabolizer and poor metabolizer (IM&PM) status was related to the raised risk of both MDD (OR = 1.547, P < 0.01) and BPD (OR = 1.808, P < 0.001). CONCLUSION Our data indicate that the impaired CYP2C19 metabolism caused by the haplotypes integrated by CYP2C19 alleles might confer the risk to MDD and BPD, whereas the ABCB1-rs1045642 T allele serves as a protective factor.
Collapse
Affiliation(s)
- Ting Zhang
- grid.410737.60000 0000 8653 1072Clinical Laboratory, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qingmin Rao
- grid.410737.60000 0000 8653 1072Clinical Laboratory, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kangguang Lin
- grid.410737.60000 0000 8653 1072Affective Disorder Department, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yongyin He
- grid.410737.60000 0000 8653 1072Clinical Laboratory, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jintai Cai
- grid.410737.60000 0000 8653 1072Clinical Laboratory, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mengxin Yang
- grid.410737.60000 0000 8653 1072Clinical Laboratory, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ying Xu
- grid.410737.60000 0000 8653 1072Clinical Laboratory, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Le Hou
- grid.410737.60000 0000 8653 1072Department of Neurology, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yulong Lin
- Clinical Laboratory, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Haiying Liu
- Clinical Laboratory, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
25
|
Miri S, Yeo J, Abubaker S, Hammami R. Neuromicrobiology, an emerging neurometabolic facet of the gut microbiome? Front Microbiol 2023; 14:1098412. [PMID: 36733917 PMCID: PMC9886687 DOI: 10.3389/fmicb.2023.1098412] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 01/03/2023] [Indexed: 01/18/2023] Open
Abstract
The concept of the gut microbiome is emerging as a metabolic interactome influenced by diet, xenobiotics, genetics, and other environmental factors that affect the host's absorption of nutrients, metabolism, and immune system. Beyond nutrient digestion and production, the gut microbiome also functions as personalized polypharmacy, where bioactive metabolites that our microbes excrete or conjugate may reach systemic circulation and impact all organs, including the brain. Appreciable evidence shows that gut microbiota produce diverse neuroactive metabolites, particularly neurotransmitters (and their precursors), stimulating the local nervous system (i.e., enteric and vagus nerves) and affecting brain function and cognition. Several studies have demonstrated correlations between the gut microbiome and the central nervous system sparking an exciting new research field, neuromicrobiology. Microbiome-targeted interventions are seen as promising adjunctive treatments (pre-, pro-, post-, and synbiotics), but the mechanisms underlying host-microbiome interactions have yet to be established, thus preventing informed evidence-based therapeutic applications. In this paper, we review the current state of knowledge for each of the major classes of microbial neuroactive metabolites, emphasizing their biological effects on the microbiome, gut environment, and brain. Also, we discuss the biosynthesis, absorption, and transport of gut microbiota-derived neuroactive metabolites to the brain and their implication in mental disorders.
Collapse
Affiliation(s)
- Saba Miri
- School of Nutrition Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - JuDong Yeo
- School of Nutrition Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Sarah Abubaker
- School of Nutrition Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Riadh Hammami
- School of Nutrition Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
26
|
Channer B, Matt SM, Nickoloff-Bybel EA, Pappa V, Agarwal Y, Wickman J, Gaskill PJ. Dopamine, Immunity, and Disease. Pharmacol Rev 2023; 75:62-158. [PMID: 36757901 PMCID: PMC9832385 DOI: 10.1124/pharmrev.122.000618] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 12/14/2022] Open
Abstract
The neurotransmitter dopamine is a key factor in central nervous system (CNS) function, regulating many processes including reward, movement, and cognition. Dopamine also regulates critical functions in peripheral organs, such as blood pressure, renal activity, and intestinal motility. Beyond these functions, a growing body of evidence indicates that dopamine is an important immunoregulatory factor. Most types of immune cells express dopamine receptors and other dopaminergic proteins, and many immune cells take up, produce, store, and/or release dopamine, suggesting that dopaminergic immunomodulation is important for immune function. Targeting these pathways could be a promising avenue for the treatment of inflammation and disease, but despite increasing research in this area, data on the specific effects of dopamine on many immune cells and disease processes remain inconsistent and poorly understood. Therefore, this review integrates the current knowledge of the role of dopamine in immune cell function and inflammatory signaling across systems. We also discuss the current understanding of dopaminergic regulation of immune signaling in the CNS and peripheral tissues, highlighting the role of dopaminergic immunomodulation in diseases such as Parkinson's disease, several neuropsychiatric conditions, neurologic human immunodeficiency virus, inflammatory bowel disease, rheumatoid arthritis, and others. Careful consideration is given to the influence of experimental design on results, and we note a number of areas in need of further research. Overall, this review integrates our knowledge of dopaminergic immunology at the cellular, tissue, and disease level and prompts the development of therapeutics and strategies targeted toward ameliorating disease through dopaminergic regulation of immunity. SIGNIFICANCE STATEMENT: Canonically, dopamine is recognized as a neurotransmitter involved in the regulation of movement, cognition, and reward. However, dopamine also acts as an immune modulator in the central nervous system and periphery. This review comprehensively assesses the current knowledge of dopaminergic immunomodulation and the role of dopamine in disease pathogenesis at the cellular and tissue level. This will provide broad access to this information across fields, identify areas in need of further investigation, and drive the development of dopaminergic therapeutic strategies.
Collapse
Affiliation(s)
- Breana Channer
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Stephanie M Matt
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Emily A Nickoloff-Bybel
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Vasiliki Pappa
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Yash Agarwal
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Jason Wickman
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Peter J Gaskill
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| |
Collapse
|
27
|
Dakic TB, Markelic MB, Ruzicic AA, Jevdjovic TV, Lakic IV, Djordjevic JD, Vujovic PZ. Hypothalamic insulin expression remains unaltered after short-term fasting in female rats. Endocrine 2022; 78:476-483. [PMID: 36301508 DOI: 10.1007/s12020-022-03235-0] [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: 09/02/2022] [Accepted: 10/15/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE Our previous study showed that 6-h fasting increased insulin expression in the hypothalamus of male rats. We, therefore, wanted to examine if this phenomenon occurs in female rats and whether it depended on the estrus cycle phase. METHODS Female rats in proestrus or diestrus were either exposed to 6-h fasting or had ad libitum access to food. The serum, cerebrospinal fluid, and hypothalamic insulin levels were determined using radioimmunoassay. The hypothalamic insulin mRNA expression was measured by RT-qPCR, while the hypothalamic insulin distribution was assessed immunohistochemically. RESULTS Albeit the short-term fasting lowered circulating insulin, both hypothalamic insulin mRNA expression and hypothalamic insulin content remained unaltered. As for the hypothalamic insulin distribution, strong insulin immunopositivity was noted primarily in ependymal cells lining the upper part of the third ventricle and some neurons mainly located within the periventricular nucleus. The pattern of insulin distribution was similar between the controls and the females exposed to fasting regardless of the estrous cycle phase. CONCLUSION The findings of this study indicate that the control of insulin expression in the hypothalamus differs from that in the pancreatic beta cells during short-term fasting. Furthermore, they also imply that the regulation of insulin expression in the female hypothalamus is different from males but independent of the estrus cycle phase.
Collapse
Affiliation(s)
- Tamara B Dakic
- Department for Comparative Physiology and Ecophysiology, Institute for Physiology and Biochemistry Ivan Djaja, University of Belgrade-Faculty of Biology, Studentski trg 16, 11000, Belgrade, Serbia.
| | - Milica B Markelic
- Department of Cell and Tissue Biology, Institute for Zoology, University of Belgrade-Faculty of Biology, Studentski trg 16, 11000, Belgrade, Serbia
| | - Aleksandra A Ruzicic
- Department for Comparative Physiology and Ecophysiology, Institute for Physiology and Biochemistry Ivan Djaja, University of Belgrade-Faculty of Biology, Studentski trg 16, 11000, Belgrade, Serbia
| | - Tanja V Jevdjovic
- Department for Comparative Physiology and Ecophysiology, Institute for Physiology and Biochemistry Ivan Djaja, University of Belgrade-Faculty of Biology, Studentski trg 16, 11000, Belgrade, Serbia
| | - Iva V Lakic
- Department for Comparative Physiology and Ecophysiology, Institute for Physiology and Biochemistry Ivan Djaja, University of Belgrade-Faculty of Biology, Studentski trg 16, 11000, Belgrade, Serbia
| | - Jelena D Djordjevic
- Department for Comparative Physiology and Ecophysiology, Institute for Physiology and Biochemistry Ivan Djaja, University of Belgrade-Faculty of Biology, Studentski trg 16, 11000, Belgrade, Serbia
| | - Predrag Z Vujovic
- Department for Comparative Physiology and Ecophysiology, Institute for Physiology and Biochemistry Ivan Djaja, University of Belgrade-Faculty of Biology, Studentski trg 16, 11000, Belgrade, Serbia
| |
Collapse
|
28
|
Kulhanek D, Abrahante Llorens JE, Buckley L, Tkac I, Rao R, Paulsen ME. Female and male C57BL/6J offspring exposed to maternal obesogenic diet develop altered hypothalamic energy metabolism in adulthood. Am J Physiol Endocrinol Metab 2022; 323:E448-E466. [PMID: 36342228 PMCID: PMC9639756 DOI: 10.1152/ajpendo.00100.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 08/12/2022] [Accepted: 09/12/2022] [Indexed: 11/22/2022]
Abstract
Maternal obesity is exceedingly common and strongly linked to offspring obesity and metabolic disease. Hypothalamic function is critical to obesity development. Hypothalamic mechanisms causing obesity following exposure to maternal obesity have not been elucidated. Therefore, we studied a cohort of C57BL/6J dams, treated with a control or high-fat-high-sugar diet, and their adult offspring to explore potential hypothalamic mechanisms to explain the link between maternal and offspring obesity. Dams treated with obesogenic diet were heavier with mild insulin resistance, which is reflective of the most common metabolic disease in pregnancy. Adult offspring exposed to maternal obesogenic diet had no change in body weight but significant increase in fat mass, decreased glucose tolerance, decreased insulin sensitivity, elevated plasma leptin, and elevated plasma thyroid-stimulating hormone. In addition, offspring exposed to maternal obesity had decreased energy intake and activity without change in basal metabolic rate. Hypothalamic neurochemical profile and transcriptome demonstrated decreased neuronal activity and inhibition of oxidative phosphorylation. Collectively, these results indicate that maternal obesity without diabetes is associated with adiposity and decreased hypothalamic energy production in offspring. We hypothesize that altered hypothalamic function significantly contributes to obesity development. Future studies focused on neuroprotective strategies aimed to improve hypothalamic function may decrease obesity development.NEW & NOTEWORTHY Offspring exposed to maternal diet-induced obesity demonstrate a phenotype consistent with energy excess. Contrary to previous studies, the observed energy phenotype was not associated with hyperphagia or decreased basal metabolic rate but rather decreased hypothalamic neuronal activity and energy production. This was supported by neurochemical changes in the hypothalamus as well as inhibition of hypothalamic oxidative phosphorylation pathway. These results highlight the potential for neuroprotective interventions in the prevention of obesity with fetal origins.
Collapse
Affiliation(s)
- Debra Kulhanek
- Division of Neonatology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota
| | | | - Lauren Buckley
- Division of Neonatology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Ivan Tkac
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Raghavendra Rao
- Division of Neonatology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Megan E Paulsen
- Division of Neonatology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota
- Minnesota Institute for the Developing Brain, Minneapolis, Minnesota
| |
Collapse
|
29
|
Kullmann S, Veit R. Zentralnervöse Prozesse bei der Prävention von Typ-2-Diabetes – Ferdinand-Bertram-Preis 2022 – eine Kurzübersicht der Preisträgerin Stephanie Kullmann. DIABETOL STOFFWECHS 2022. [DOI: 10.1055/a-1925-2652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
30
|
Kullmann S, Goj T, Veit R, Fritsche L, Wagner L, Schneeweiss P, Hoene M, Hoffmann C, Machann J, Niess A, Preissl H, Birkenfeld AL, Peter A, Häring HU, Fritsche A, Moller A, Weigert C, Heni M. Exercise restores brain insulin sensitivity in sedentary adults who are overweight and obese. JCI Insight 2022; 7:161498. [PMID: 36134657 DOI: 10.1172/jci.insight.161498] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/04/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUNDInsulin resistance of the brain can unfavorably affect long-term weight maintenance and body fat distribution. Little is known if and how brain insulin sensitivity can be restored in humans. We aimed to evaluate the effects of an exercise intervention on insulin sensitivity of the brain and how this relates to exercise-induced changes in whole-body metabolism and behavior.METHODSIn this clinical trial, sedentary participants who were overweight and obese underwent an 8-week supervised aerobic training intervention. Brain insulin sensitivity was assessed in 21 participants (14 women, 7 men; age range 21-59 years; BMI range 27.5-45.5 kg/m2) using functional MRI, combined with intranasal administration of insulin, before and after the intervention.RESULTSThe exercise program resulted in enhanced brain insulin action to the level of a person of healthy weight, demonstrated by increased insulin-induced striatal activity and strengthened hippocampal functional connectivity. Improved brain insulin action correlated with increased mitochondrial respiration in skeletal muscle, reductions in visceral fat and hunger, as well as improved cognition. Mediation analyses suggest that improved brain insulin responsiveness helps mediate the peripheral exercise effects leading to healthier body fat distribution and reduced perception of hunger.CONCLUSIONOur study demonstrates that an 8-week exercise intervention in sedentary individuals can restore insulin action in the brain. Hence, the ameliorating benefits of exercise toward brain insulin resistance may provide an objective therapeutic target in humans in the challenge to reduce diabetes risk factors.TRIAL REGISTRATIONClinicalTrials.gov (NCT03151590).FUNDINGBMBF/DZD 01GI0925.
Collapse
Affiliation(s)
- Stephanie Kullmann
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Thomas Goj
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Institute for Clinical Chemistry and Pathobiochemistry and
| | - Ralf Veit
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Louise Fritsche
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Lore Wagner
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Patrick Schneeweiss
- Department of Sports Medicine, University Hospital Tübingen, Germany.,Interfaculty Research Institute for Sport and Physical Activity, University of Tübingen, Tübingen, Germany
| | - Miriam Hoene
- Institute for Clinical Chemistry and Pathobiochemistry and
| | | | - Jürgen Machann
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Department of Radiology, Section on Experimental Radiology, University Hospital Tübingen, Germany
| | - Andreas Niess
- Department of Sports Medicine, University Hospital Tübingen, Germany.,Interfaculty Research Institute for Sport and Physical Activity, University of Tübingen, Tübingen, Germany
| | - Hubert Preissl
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany.,Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Andreas L Birkenfeld
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Andreas Peter
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Institute for Clinical Chemistry and Pathobiochemistry and
| | - Hans-Ulrich Häring
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Andreas Fritsche
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Anja Moller
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Cora Weigert
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Institute for Clinical Chemistry and Pathobiochemistry and
| | - Martin Heni
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany.,Institute for Clinical Chemistry and Pathobiochemistry and.,Division of Endocrinology and Diabetology, Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| |
Collapse
|
31
|
Zhou R, He M, Fan J, Li R, Zuo Y, Li B, Gao G, Sun T. The role of hypothalamic endoplasmic reticulum stress in schizophrenia and antipsychotic-induced weight gain: A narrative review. Front Neurosci 2022; 16:947295. [PMID: 36188456 PMCID: PMC9523121 DOI: 10.3389/fnins.2022.947295] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/29/2022] [Indexed: 11/21/2022] Open
Abstract
Schizophrenia (SCZ) is a serious mental illness that affects 1% of people worldwide. SCZ is associated with a higher risk of developing metabolic disorders such as obesity. Antipsychotics are the main treatment for SCZ, but their side effects include significant weight gain/obesity. Despite extensive research, the underlying mechanisms by which SCZ and antipsychotic treatment induce weight gain/obesity remain unclear. Hypothalamic endoplasmic reticulum (ER) stress is one of the most important pathways that modulates inflammation, neuronal function, and energy balance. This review aimed to investigate the role of hypothalamic ER stress in SCZ and antipsychotic-induced weight gain/obesity. Preliminary evidence indicates that SCZ is associated with reduced dopamine D2 receptor (DRD2) signaling, which significantly regulates the ER stress pathway, suggesting the importance of ER stress in SCZ and its related metabolic disorders. Antipsychotics such as olanzapine activate ER stress in hypothalamic neurons. These effects may induce decreased proopiomelanocortin (POMC) processing, increased neuropeptide Y (NPY) and agouti-related protein (AgRP) expression, autophagy, and leptin and insulin resistance, resulting in hyperphagia, decreased energy expenditure, and central inflammation, thereby causing weight gain. By activating ER stress, antipsychotics such as olanzapine activate hypothalamic astrocytes and Toll-like receptor 4 signaling, thereby causing inflammation and weight gain/obesity. Moreover, evidence suggests that antipsychotic-induced ER stress may be related to their antagonistic effects on neurotransmitter receptors such as DRD2 and the histamine H1 receptor. Taken together, ER stress inhibitors could be a potential effective intervention against SCZ and antipsychotic-induced weight gain and inflammation.
Collapse
Affiliation(s)
- Ruqin Zhou
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
| | - Meng He
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
- *Correspondence: Meng He,
| | - Jun Fan
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
| | - Ruoxi Li
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yufeng Zuo
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
| | - Benben Li
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
| | - Guanbin Gao
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, China
- Guanbin Gao,
| | - Taolei Sun
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
- Taolei Sun,
| |
Collapse
|
32
|
Kumar Palepu MS, Dandekar MP. Remodeling of microbiota gut-brain axis using psychobiotics in depression. Eur J Pharmacol 2022; 931:175171. [PMID: 35926568 DOI: 10.1016/j.ejphar.2022.175171] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/14/2022] [Accepted: 07/21/2022] [Indexed: 12/11/2022]
Abstract
Depression is a multifaceted psychiatric disorder mainly orchestrated by dysfunction of neuroendocrine, neurochemical, immune, and metabolic systems. The interconnection of gut microbiota perturbation with the central nervous system disorders has been well documented in recent times. Indeed, alteration of commensal intestinal microflora is noted in several psychiatric disorders such as anxiety and depression, which are presumed to be routed through the enteric nervous system, autonomic nervous system, endocrine, and immune system. This review summarises the new mechanisms underlying the crosstalk between gut microbiota and brain involved in the management of depression. Depression-induced changes in the commensal intestinal microbiota are majorly linked with the disruption of gut integrity, hyperinflammation, and modulation of short-chain fatty acids, neurotransmitters, kynurenine metabolites, endocannabinoids, brain-derived neurotropic factors, hypothalamic-pituitary-adrenal axis, and gut peptides. The restoration of gut microbiota with prebiotics, probiotics, postbiotics, synbiotics, and fermented foods (psychobiotics) has gained a considerable attention for the management of depression. Recent evidence also propose the role of gut microbiota in the process of treatment-resistant depression. Thus, remodeling of the microbiota-gut-brain axis using psychobiotics appears to be a promising therapeutic approach for the reversal of psychiatric disorders, and it is imperative to decipher the underlying mechanisms for gut-brain crosstalk.
Collapse
Affiliation(s)
- Mani Surya Kumar Palepu
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Manoj P Dandekar
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India.
| |
Collapse
|
33
|
Insulin and Its Key Role for Mitochondrial Function/Dysfunction and Quality Control: A Shared Link between Dysmetabolism and Neurodegeneration. BIOLOGY 2022; 11:biology11060943. [PMID: 35741464 PMCID: PMC9220302 DOI: 10.3390/biology11060943] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/01/2022] [Accepted: 06/17/2022] [Indexed: 02/07/2023]
Abstract
Insulin was discovered and isolated from the beta cells of pancreatic islets of dogs and is associated with the regulation of peripheral glucose homeostasis. Insulin produced in the brain is related to synaptic plasticity and memory. Defective insulin signaling plays a role in brain dysfunction, such as neurodegenerative disease. Growing evidence suggests a link between metabolic disorders, such as diabetes and obesity, and neurodegenerative diseases, especially Alzheimer's disease (AD). This association is due to a common state of insulin resistance (IR) and mitochondrial dysfunction. This review takes a journey into the past to summarize what was known about the physiological and pathological role of insulin in peripheral tissues and the brain. Then, it will land in the present to analyze the insulin role on mitochondrial health and the effects on insulin resistance and neurodegenerative diseases that are IR-dependent. Specifically, we will focus our attention on the quality control of mitochondria (MQC), such as mitochondrial dynamics, mitochondrial biogenesis, and selective autophagy (mitophagy), in healthy and altered cases. Finally, this review will be projected toward the future by examining the most promising treatments that target the mitochondria to cure neurodegenerative diseases associated with metabolic disorders.
Collapse
|
34
|
Wagner L, Veit R, Fritsche L, Häring HU, Fritsche A, Birkenfeld AL, Heni M, Preissl H, Kullmann S. Sex differences in central insulin action: Effect of intranasal insulin on neural food cue reactivity in adults with normal weight and overweight. Int J Obes (Lond) 2022; 46:1662-1670. [PMID: 35715625 PMCID: PMC9395264 DOI: 10.1038/s41366-022-01167-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 05/30/2022] [Accepted: 06/07/2022] [Indexed: 01/17/2023]
Abstract
Background/Objectives Central insulin action influences cognitive processes, peripheral metabolism, and eating behavior. However, the contribution of obesity and sex on central insulin-mediated neural food cue processing still remains unclear. Subjects/Methods In a randomized within-participant design, including two visits, 60 participants (30 women, BMI 18–32 kg/m2, age 21–69 years) underwent a functional MRI task measuring blood oxygen level-dependent (BOLD) signal in response to visual food cues after intranasal insulin or placebo spray administration. Central insulin action was defined as the neural BOLD response to food cues after insulin compared to placebo administration. Afterwards, participants were asked to rate the food cues for desire to eat (i.e., wanting rating). For statistical analyses, participants were grouped according to BMI and sex. Results Food cue reactivity in the amygdala showed higher BOLD activation in response to central insulin compared to placebo. Furthermore, women with overweight and obesity and men of normal weight showed higher BOLD neural food cue responsivity to central insulin compared to placebo. Higher central insulin action in the insular cortex was associated with better peripheral insulin sensitivity and higher cognitive control. Moreover, central insulin action in the dorsolateral prefrontal cortex (DLPFC) revealed significant sex differences. In response to central insulin compared to placebo, men showed lower DLPFC BOLD activity, whereas women showed higher DLPFC activity in response to highly desired food cues. On behavioral level, central insulin action significantly reduced hunger, whereas the desire to eat, especially for low caloric food cues was significantly higher with central insulin than with placebo. Conclusions Obesity and sex influenced the central insulin-mediated neural BOLD activity to visual food cues in brain regions implicated in reward and cognitive control. These findings show that central insulin action regulates food response differentially in men and women, which may have consequences for metabolism and eating behavior.
Collapse
Affiliation(s)
- Lore Wagner
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany. .,German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.
| | - Ralf Veit
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Tübingen, Germany
| | - Louise Fritsche
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Tübingen, Germany
| | - Hans-Ulrich Häring
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Andreas Fritsche
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany.,Nutritional and Preventive Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Andreas L Birkenfeld
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Martin Heni
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany.,Institute for Clinical Chemistry and Pathobiochemistry, University Hospital Tübingen, Tübingen, Germany.,Department of Internal Medicine I, Division of Endocrinology and Diabetology, Ulm University Hospital, Ulm, Germany
| | - Hubert Preissl
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany.,Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Center Munich, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Stephanie Kullmann
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| |
Collapse
|
35
|
González A, Calfío C, Churruca M, Maccioni RB. Glucose metabolism and AD: evidence for a potential diabetes type 3. Alzheimers Res Ther 2022; 14:56. [PMID: 35443732 PMCID: PMC9022265 DOI: 10.1186/s13195-022-00996-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/27/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Alzheimer's disease is the most prevalent cause of dementia in the elderly. Neuronal death and synaptic dysfunctions are considered the main hallmarks of this disease. The latter could be directly associated to an impaired metabolism. In particular, glucose metabolism impairment has demonstrated to be a key regulatory element in the onset and progression of AD, which is why nowadays AD is considered the type 3 diabetes. METHODS We provide a thread regarding the influence of glucose metabolism in AD from three different perspectives: (i) as a regulator of the energy source, (ii) through several metabolic alterations, such as insulin resistance, that modify peripheral signaling pathways that influence activation of the immune system (e.g., insulin resistance, diabetes, etc.), and (iii) as modulators of various key post-translational modifications for protein aggregation, for example, influence on tau hyperphosphorylation and other important modifications, which determine its self-aggregating behavior and hence Alzheimer's pathogenesis. CONCLUSIONS In this revision, we observed a 3 edge-action in which glucose metabolism impairment is acting in the progression of AD: as blockade of energy source (e.g., mitochondrial dysfunction), through metabolic dysregulation and post-translational modifications in key proteins, such as tau. Therefore, the latter would sustain the current hypothesis that AD is, in fact, the novel diabetes type 3.
Collapse
Affiliation(s)
- Andrea González
- Laboratory of Neurosciences and Functional Medicine, International Center for Biomedicine (ICC), Avda. Vitacura 3568, D 511-512, Vitacura, Santiago, Chile
- Faculty of Sciences, University of Chile, Las Encinas 3370, Ñuñoa, Santiago, Chile
| | - Camila Calfío
- Laboratory of Neurosciences and Functional Medicine, International Center for Biomedicine (ICC), Avda. Vitacura 3568, D 511-512, Vitacura, Santiago, Chile
- Faculty of Sciences, University of Chile, Las Encinas 3370, Ñuñoa, Santiago, Chile
| | - Macarena Churruca
- Laboratory of Neurosciences and Functional Medicine, International Center for Biomedicine (ICC), Avda. Vitacura 3568, D 511-512, Vitacura, Santiago, Chile
| | - Ricardo B Maccioni
- Laboratory of Neurosciences and Functional Medicine, International Center for Biomedicine (ICC), Avda. Vitacura 3568, D 511-512, Vitacura, Santiago, Chile.
- Faculty of Sciences, University of Chile, Las Encinas 3370, Ñuñoa, Santiago, Chile.
- Department of Neurology, Faculty of Medicine East Campus Hospital Salvador, University of Chile, Salvador 486, Providencia, Santiago, Chile.
| |
Collapse
|
36
|
Compensatory Role of Insulin in the Extinction but Not Reinstatement of Morphine-Induced Conditioned Place Preference in the Streptozotocin-Induced Diabetic Rats. Neurochem Res 2022; 47:1565-1573. [DOI: 10.1007/s11064-022-03550-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/19/2022] [Accepted: 02/05/2022] [Indexed: 02/02/2023]
|
37
|
Hanssen R, Thanarajah SE, Tittgemeyer M, Brüning JC. Obesity - A Matter of Motivation? Exp Clin Endocrinol Diabetes 2022; 130:290-295. [PMID: 35181879 PMCID: PMC9286865 DOI: 10.1055/a-1749-4852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Excessive food intake and reduced physical activity have long been established as
primary causes of obesity. However, the underlying mechanisms causing this
unhealthy behavior characterized by heightened motivation for food but not for
physical effort are unclear. Despite the common unjustified stigmatization that
obesity is a result of laziness and lack of discipline, it is becoming
increasingly clear that high-fat diet feeding and obesity cause alterations in
brain circuits that are critical for the control of motivational behavior. In this mini-review, we provide a comprehensive overview of incentive motivation,
its neural encoding in the dopaminergic mesolimbic system as well as its
metabolic modulation with a focus on derangements of incentive motivation in
obesity. We further discuss the emerging field of metabolic interventions to
counteract motivational deficits and their potential clinical implications.
Collapse
Affiliation(s)
- Ruth Hanssen
- Max Planck Institute for Metabolism Research, Cologne, Germany.,Policlinic for Endocrinology, Diabetology and Preventive Medicine (PEPD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Sharmili E Thanarajah
- Max Planck Institute for Metabolism Research, Cologne, Germany.,Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Marc Tittgemeyer
- Max Planck Institute for Metabolism Research, Cologne, Germany.,Cluster of Excellence in Cellular Stress Responses in Aging-associated Diseases (CECAD), Cologne, Germany
| | - Jens C Brüning
- Max Planck Institute for Metabolism Research, Cologne, Germany.,Policlinic for Endocrinology, Diabetology and Preventive Medicine (PEPD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.,Cluster of Excellence in Cellular Stress Responses in Aging-associated Diseases (CECAD), Cologne, Germany
| |
Collapse
|
38
|
Avraham Y, Van Heukelom B, Zolotarev O, Magen I, Vorobiev L, Zwas DR. Insulin normalized brain metabolic status on a Model of Anorexia Nervosa in Mice. Physiol Behav 2022; 249:113738. [PMID: 35182554 DOI: 10.1016/j.physbeh.2022.113738] [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: 10/22/2021] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Anorexia nervosa is a psycho-socio-biological disease, characterized by self-starvation and distorted perception of body weight. Patients often over-exercise. Insulin is an anabolic hormone that increases food intake and restores body fat and is present in low levels in anorexia nervosa patients: thus may have therapeutic potential in treating anorexia nervosa. AIMS to explore whether low levels insulin administration may result in recovery of cerebral function and restoration of metabolic disorder providing a treatment option for anorexia nervosa. METHODS Female Sabra mice maintained on DR of 2.0 hours per day for 32 days, in cages with or without wheel attached to an electronic counter (activity wheel). They were then permitted to eat ad libitum for additional 15 days. On the second week, mice were injected ip with 0.5U/kg long acting Insulin(Lantus) or saline and cognitive function was evaluated. Insulin administered three times a week during days 8-32. Mice euthanized on day 48 and cerebral levels of monoamines, 2-AG and expression of genes associated with metabolic status were evaluated. RESULTS Activity wheel mice decreased body weight, 2-AG, dopamine levels and 5-HT1A and increased Camkk2 and SIRT1 gene expression compared to mice without it. Insulin increased body weight, decreased revolutions, enhanced NPY and normalized Camkk2, SIRT-1, BDNF, elevated 2-AG and improved cognition in the wheel group. CONCLUSION low dose insulin administration to animal model of anorexia associated with exercise, led to alterations and normalization in brain metabolic status and improved cognition. Insulin should be further explored as potential novel treatment for anorexia nervosa.
Collapse
Affiliation(s)
- Yosefa Avraham
- Linda Joy Pollin Cardiovascular Wellness Center for Women, Heart Institute, Hadassah University Medical Center, Jerusalem, Israel 91120.
| | - Bob Van Heukelom
- Department of Neurology Gelderse Vallei Hospital, Ede Netherlands 6716 RP Ede
| | - Olga Zolotarev
- Department of Metabolism and Human Nutrition, Braun School of Public Health, Hadassah-Hebrew University Medical School, Jerusalem, Israel 91120
| | - Iddo Magen
- Department of Metabolism and Human Nutrition, Braun School of Public Health, Hadassah-Hebrew University Medical School, Jerusalem, Israel 91120
| | - Lia Vorobiev
- Department of Metabolism and Human Nutrition, Braun School of Public Health, Hadassah-Hebrew University Medical School, Jerusalem, Israel 91120
| | - Donna R Zwas
- Linda Joy Pollin Cardiovascular Wellness Center for Women, Heart Institute, Hadassah University Medical Center, Jerusalem, Israel 91120
| |
Collapse
|
39
|
Heuberger LS, Gobbi S, Weber SC, Graf G, Tobler PN, Asarian L, Geary N, Roth M, Leeners B. Is It Worth It? Obesity Affects Snack Food Valuation Across the Menstrual Cycle. Front Neurosci 2022; 16:800976. [PMID: 35250448 PMCID: PMC8889102 DOI: 10.3389/fnins.2022.800976] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 01/21/2022] [Indexed: 11/30/2022] Open
Abstract
Background The importance of menstrual cycle physiology in appetite and obesity is poorly understood. We investigated the effects of body mass index (BMI), menstrual cycle phase and sweet and salty taste on monetary valuation of snack foods. Methods We recruited 72 women and after the application of in- and exclusion criteria 31 participants with healthy weight and 25 with obesity remained. The participants completed a willingness to pay (WTP) task to measure subjective value of 30 snack food items in the pre-ovulatory and mid-luteal cycle phases. Results Generalized linear mixed model (GLMM) analysis revealed that BMI, cycle phase and snack taste interacted to influence WTP (−0.15 [−0.22, −0.03], p = 0.002). Hence, WTP was inversely related to BMI, but the strength of the relation depended on cycle phase and taste. The WTP of participants with healthy weight for salty taste changed across cycle phase but the WTP for sweet taste was not affected by cycle phase. Moreover, the cycle effect for the salty snacks ceased in participants with obesity. Conclusion The inverse effect of BMI on WTP valuation of snack foods contrasts with the positive effect of BMI on pleasantness ratings for milkshakes by the same women that we previously reported. This indicates that the two measures reflect different aspects of food-related valuative processing in obesity. Furthermore, the WTP data suggest that the selection of salty snacks may differ from that of sweet snacks in the pre-ovulatory phase of the menstrual cycle for individuals of healthy weight. The cycle phase does not seem to affect food valuation of participants with obesity. These findings are relevant to understanding and treating obesity in women.
Collapse
Affiliation(s)
| | - Susanna Gobbi
- Zurich Center for Neuroeconomics, University of Zurich, Zurich, Switzerland
| | - Susanna C. Weber
- Zurich Center for Neuroeconomics, University of Zurich, Zurich, Switzerland
| | - Gwendolyn Graf
- Department of Reproductive Endocrinology, University Hospital of Zurich, Zurich, Switzerland
| | - Philippe N. Tobler
- Swiss Federal Institute of Technology, Zurich, Switzerland
- Zurich Center for Neuroeconomics, University of Zurich, Zurich, Switzerland
| | - Lori Asarian
- Department of Medicine, University of Vermont, Burlington, VT, United States
| | | | - Mareike Roth
- Department of Reproductive Endocrinology, University Hospital of Zurich, Zurich, Switzerland
| | - Brigitte Leeners
- Department of Reproductive Endocrinology, University Hospital of Zurich, Zurich, Switzerland
- Department of Reproductive Endocrinology, University of Zurich, Zurich, Switzerland
- *Correspondence: Brigitte Leeners,
| |
Collapse
|
40
|
Chen M, Ruan G, Chen L, Ying S, Li G, Xu F, Xiao Z, Tian Y, Lv L, Ping Y, Cheng Y, Wei Y. Neurotransmitter and Intestinal Interactions: Focus on the Microbiota-Gut-Brain Axis in Irritable Bowel Syndrome. Front Endocrinol (Lausanne) 2022; 13:817100. [PMID: 35250873 PMCID: PMC8888441 DOI: 10.3389/fendo.2022.817100] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 01/05/2022] [Indexed: 12/12/2022] Open
Abstract
Irritable bowel syndrome (IBS) is a functional gastrointestinal disorder of unknown etiology. IBS is caused by a disruption in the gut-brain axis. Given the importance of the gut microbiota in maintaining local and systemic homeostasis of immunity, endocrine, and other physiological processes, the microbiota-gut-brain axis has been proposed as a key regulator in IBS. Neurotransmitters have been shown to affect blood flow regulation, intestinal motility, nutrient absorption, the gastrointestinal immune system, and the microbiota in recent studies. It has the potential role to play a function in the pathophysiology of the gastrointestinal and neurological systems. Transmitters and their receptors, including 5-hydroxytryptamine, dopamine, γ-aminobutyric acid, and histamine, play an important role in IBS, especially in visceral sensitivity and gastrointestinal motility. Studies in this field have shed light on revealing the mechanism by which neurotransmitters act in the pathogenesis of IBS and discovering new therapeutic strategies based on traditional pharmacological approaches that target the nervous system or novel therapies that target the microbiota.
Collapse
Affiliation(s)
- Minjia Chen
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- Department of Pathogenic Biology and Immunology, School of Basic Medicine, Ningxia Medical University, Yinchuan, China
| | - Guangcong Ruan
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Lu Chen
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Senhong Ying
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Guanhu Li
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Fenghua Xu
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhifeng Xiao
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yuting Tian
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Linling Lv
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yi Ping
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yi Cheng
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- *Correspondence: Yanling Wei, ; Yi Cheng,
| | - Yanling Wei
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- *Correspondence: Yanling Wei, ; Yi Cheng,
| |
Collapse
|
41
|
Blázquez E, Hurtado-Carneiro V, LeBaut-Ayuso Y, Velázquez E, García-García L, Gómez-Oliver F, Ruiz-Albusac J, Ávila J, Pozo MÁ. Significance of Brain Glucose Hypometabolism, Altered Insulin Signal Transduction, and Insulin Resistance in Several Neurological Diseases. Front Endocrinol (Lausanne) 2022; 13:873301. [PMID: 35615716 PMCID: PMC9125423 DOI: 10.3389/fendo.2022.873301] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/23/2022] [Indexed: 12/14/2022] Open
Abstract
Several neurological diseases share pathological alterations, even though they differ in their etiology. Neuroinflammation, altered brain glucose metabolism, oxidative stress, mitochondrial dysfunction and amyloidosis are biological events found in those neurological disorders. Altered insulin-mediated signaling and brain glucose hypometabolism are characteristic signs observed in the brains of patients with certain neurological diseases, but also others such as type 2 diabetes mellitus and vascular diseases. Thus, significant reductions in insulin receptor autophosphorylation and Akt kinase activity, and increased GSK-3 activity and insulin resistance, have been reported in these neurological diseases as contributing to the decline in cognitive function. Supporting this relationship is the fact that nasal and hippocampal insulin administration has been found to improve cognitive function. Additionally, brain glucose hypometabolism precedes the unmistakable clinical manifestations of some of these diseases by years, which may become a useful early biomarker. Deficiencies in the major pathways of oxidative energy metabolism have been reported in patients with several of these neurological diseases, which supports the hypothesis of their metabolic background. This review remarks on the significance of insulin and brain glucose metabolism alterations as keystone common pathogenic substrates for certain neurological diseases, highlighting new potential targets.
Collapse
Affiliation(s)
- Enrique Blázquez
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Complutense University, Madrid, Spain
- *Correspondence: Enrique Blázquez,
| | | | - Yannick LeBaut-Ayuso
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Esther Velázquez
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Luis García-García
- Pluridisciplinary Institute, Complutense University, IdISSC, Madrid, Spain
- Department of Pharmacology, Pharmacognosy and Botany, Faculty of Pharmacy, Complutense University, Madrid, Spain
| | - Francisca Gómez-Oliver
- Pluridisciplinary Institute, Complutense University, IdISSC, Madrid, Spain
- Department of Pharmacology, Pharmacognosy and Botany, Faculty of Pharmacy, Complutense University, Madrid, Spain
| | - Juan Miguel Ruiz-Albusac
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Jesús Ávila
- Center of Molecular Biology “Severo Ochoa”, CSIC-UAM, Madrid, Spain
| | - Miguel Ángel Pozo
- Department of Physiology, Faculty of Medicine, Complutense University, Madrid, Spain
- Pluridisciplinary Institute, Complutense University, IdISSC, Madrid, Spain
| |
Collapse
|
42
|
Wilkialis L, Rodrigues NB, Cha DS, Siegel A, Majeed A, Lui LMW, Tamura JK, Gill B, Teopiz K, McIntyre RS. Social Isolation, Loneliness and Generalized Anxiety: Implications and Associations during the COVID-19 Quarantine. Brain Sci 2021; 11:1620. [PMID: 34942920 PMCID: PMC8699379 DOI: 10.3390/brainsci11121620] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 12/15/2022] Open
Abstract
The COVID-19 pandemic has resulted in a predominantly global quarantine response that has been associated with social isolation, loneliness, and anxiety. The foregoing experiences have been amply documented to have profound impacts on health, morbidity, and mortality. This narrative review uses the extant neurobiological and theoretical literature to explore the association between social isolation, loneliness, and anxiety in the context of quarantine during the COVID-19 pandemic. Emerging evidence suggests that distinct health issues (e.g., a sedentary lifestyle, a diminished overall sense of well-being) are associated with social isolation and loneliness. The health implications of social isolation and loneliness during quarantine have a heterogenous and comorbid nature and, as a result, form a link to anxiety. The limbic system plays a role in fear and anxiety response; the bed nucleus of the stria terminalis, amygdala, HPA axis, hippocampus, prefrontal cortex, insula, and locus coeruleus have an impact in a prolonged anxious state. In the conclusion, possible solutions are considered and remarks are made on future areas of exploration.
Collapse
Affiliation(s)
- Linas Wilkialis
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON M5T 2S8, Canada; (L.W.); (N.B.R.); (D.S.C.); (A.S.); (A.M.); (L.M.W.L.); (J.K.T.); (B.G.); (K.T.)
| | - Nelson B. Rodrigues
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON M5T 2S8, Canada; (L.W.); (N.B.R.); (D.S.C.); (A.S.); (A.M.); (L.M.W.L.); (J.K.T.); (B.G.); (K.T.)
| | - Danielle S. Cha
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON M5T 2S8, Canada; (L.W.); (N.B.R.); (D.S.C.); (A.S.); (A.M.); (L.M.W.L.); (J.K.T.); (B.G.); (K.T.)
| | - Ashley Siegel
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON M5T 2S8, Canada; (L.W.); (N.B.R.); (D.S.C.); (A.S.); (A.M.); (L.M.W.L.); (J.K.T.); (B.G.); (K.T.)
| | - Amna Majeed
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON M5T 2S8, Canada; (L.W.); (N.B.R.); (D.S.C.); (A.S.); (A.M.); (L.M.W.L.); (J.K.T.); (B.G.); (K.T.)
| | - Leanna M. W. Lui
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON M5T 2S8, Canada; (L.W.); (N.B.R.); (D.S.C.); (A.S.); (A.M.); (L.M.W.L.); (J.K.T.); (B.G.); (K.T.)
| | - Jocelyn K. Tamura
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON M5T 2S8, Canada; (L.W.); (N.B.R.); (D.S.C.); (A.S.); (A.M.); (L.M.W.L.); (J.K.T.); (B.G.); (K.T.)
| | - Barjot Gill
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON M5T 2S8, Canada; (L.W.); (N.B.R.); (D.S.C.); (A.S.); (A.M.); (L.M.W.L.); (J.K.T.); (B.G.); (K.T.)
| | - Kayla Teopiz
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON M5T 2S8, Canada; (L.W.); (N.B.R.); (D.S.C.); (A.S.); (A.M.); (L.M.W.L.); (J.K.T.); (B.G.); (K.T.)
| | - Roger S. McIntyre
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON M5T 2S8, Canada; (L.W.); (N.B.R.); (D.S.C.); (A.S.); (A.M.); (L.M.W.L.); (J.K.T.); (B.G.); (K.T.)
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Pharmacology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON M5T 1R8, Canada
- Brain and Cognition Discovery Foundation, Toronto, ON M4W 3W4, Canada
| |
Collapse
|
43
|
Li Y, Ren L, Fu H, Yang B, Tian J, Li Q, Liu Z, Liu S. Crosstalk between dopamine and insulin signaling in growth control of the oyster. Gen Comp Endocrinol 2021; 313:113895. [PMID: 34480943 DOI: 10.1016/j.ygcen.2021.113895] [Citation(s) in RCA: 3] [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: 03/05/2021] [Revised: 07/20/2021] [Accepted: 08/29/2021] [Indexed: 12/26/2022]
Abstract
Neuroendocrine hormones such as dopamine and insulin/insulin-like peptides play indispensable roles in growth regulation of animals, while the interplay between dopamine and insulin signaling pathways remains largely unknown in invertebrates. In the present study, we showed that tyrosine hydroxylase (TH), the rate-limiting enzyme of dopamine synthesis, was highly expressed in all tissues of the fast-growing oysters, and gradually increased with the development, which indicated the potential role of dopamine in growth regulation. Incubated with dopamine hydrochloride and insulin-like peptide recombinant proteins in vitro induced the expression of TH, suggesting a mutual regulatory relationship between insulin and dopamine signaling. Fasting and re-feeding experiments confirmed the role of TH in food intake regulation, also provide a clue about the potential regulatory relationship between the FoxO and TH. Further luciferase assay experiment confirmed that FoxO was involved in transcriptional regulation of TH gene through binding to its specific promoter region. This work provided insights into the crosstalk between dopamine and insulin signaling in growth control of mollusks.
Collapse
Affiliation(s)
- Yongjing Li
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, and College of Fisheries, Ocean University of China, Qingdao 266003, China
| | - Liting Ren
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, and College of Fisheries, Ocean University of China, Qingdao 266003, China
| | - Huiru Fu
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, and College of Fisheries, Ocean University of China, Qingdao 266003, China
| | - Ben Yang
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, and College of Fisheries, Ocean University of China, Qingdao 266003, China
| | - Jing Tian
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, and College of Fisheries, Ocean University of China, Qingdao 266003, China
| | - Qi Li
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, and College of Fisheries, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| | - Zhanjiang Liu
- Department of Biology, College of Art and Sciences, Syracuse University, Syracuse, NY 13244, USA
| | - Shikai Liu
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, and College of Fisheries, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China.
| |
Collapse
|
44
|
Kullmann S, Blum D, Jaghutriz BA, Gassenmaier C, Bender B, Häring HU, Reischl G, Preissl H, la Fougère C, Fritsche A, Reimold M, Heni M. Central Insulin Modulates Dopamine Signaling in the Human Striatum. J Clin Endocrinol Metab 2021; 106:2949-2961. [PMID: 34131733 DOI: 10.1210/clinem/dgab410] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Indexed: 01/17/2023]
Abstract
OBJECTIVE Activity in the dopaminergic pathways of the brain is highly sensitive to body weight and metabolic states. Animal studies show that dopamine neurons are important targets for the metabolic hormone insulin with abolished effects in the insulin-resistant state, leading to increases in body weight and food intake. In humans, the influence of central acting insulin on dopamine and effects of their interplay are still elusive. RESEARCH DESIGN AND METHODS We investigated whether central administered insulin influences dopaminergic activity in striatal regions and whole-brain neural activity. Using a positron emission tomography (PET)/magnetic resonance imaging (MRI) hybrid scanner, we simultaneously performed [11C]-raclopride-PET and resting-state functional MRI in 10 healthy normal-weight men after application of intranasal insulin or placebo on 2 separate days in a randomized, placebo-controlled, blinded, crossover trial. RESULTS In response to central insulin compared with placebo administration, we observed greater [11C]-raclopride binding potential in the bilateral ventral and dorsal striatum. This suggests an insulin-induced reduction in synaptic dopamine levels. Resting-state striatal activity was lower 15 and 30 minutes after nasal insulin compared with placebo. Functional connectivity of the mesocorticolimbic circuitry associated with differences in dopamine levels: individuals with a stronger insulin-induced effect on dopamine levels showed a stronger increase in functional connectivity 45 minutes after intranasal insulin. CONCLUSIONS This study indicates that central insulin modulates dopaminergic tone in the striatum, which may affect regional brain activity and connectivity. Our results deepen the understanding of the insulin-dopamine interaction and the complex network that underlies the regulation of whole-body metabolism.
Collapse
Affiliation(s)
- Stephanie Kullmann
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Dominik Blum
- Department of Nuclear Medicine and Clinical Molecular Imaging, Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Benjamin Assad Jaghutriz
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Christoph Gassenmaier
- Department of Internal Medicine, Division of Hematology, Oncology, Clinical Immunology and Rheumatology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Benjamin Bender
- Department of Diagnostic and Interventional Neuroradiology, University Hospital Tübingen, Tübingen, Germany
| | - Hans-Ulrich Häring
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Gerald Reischl
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Hubert Preissl
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Department of Pharmacy and Biochemistry, Institute of Pharmaceutical Sciences, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Christian la Fougère
- Department of Nuclear Medicine and Clinical Molecular Imaging, Eberhard-Karls-University Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Germany
| | - Andreas Fritsche
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Matthias Reimold
- Department of Nuclear Medicine and Clinical Molecular Imaging, Eberhard-Karls-University Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Germany
| | - Martin Heni
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
45
|
Rodrigues KC, Bortolatto CF, da Motta KP, de Oliveira RL, Paltian JJ, Krüger R, Roman SS, Boeira SP, Alves D, Wilhelm EA, Luchese C. The neurotherapeutic role of a selenium-functionalized quinoline in hypothalamic obese rats. Psychopharmacology (Berl) 2021; 238:1937-1951. [PMID: 33740091 DOI: 10.1007/s00213-021-05821-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 03/08/2021] [Indexed: 12/30/2022]
Abstract
RATIONALE Obesity is considered one of the major global health problems and increases the risk of several medical complications, such as diabetes and mental illnesses. OBJECTIVE The present study investigated the effect of 7-chloro-4-(phenylselanyl) quinoline (4-PSQ) on obesity parameters, behavioral and neurochemical alterations in hypothalamic obese rats. METHODS Male Wistar rats received subcutaneous neonatal injections of monosodium glutamate (MSG, 4g/kg) or saline. After the Lee Index evaluation, rats were divided into groups and treated with 4-PSQ (5 mg/kg, intragastric route) or canola oil once a day (post-natal days (PND) 60→76). Open-field, elevated plus-maze, forced swim task, object recognition/location memory, and stepdown inhibitory avoidance tasks were conducted from PND 66 to 74. On PND 76, rats were euthanized and epididymal fat, blood, cerebral cortex, andhippocampus were removed. Blood biochemical parameters and cortical/hippocampal acetylcholinesterase (AChE) and Na /K -ATPase activities were assessed. RESULTS MSG increased the Lee Index characterizing the chemically induced hypothalamic obesity model. 4-PSQ reversed the increases of epididymal fat, blood glucose, and triglyceride levels caused by MSG exposure. 4-PSQ attenuated anxiety-like and depression-like behaviors induced by neonatal administrations of MSG. Memory deficits found in MSG-obese rats were reversed by treatment with 4-PSQ. Neurochemical alterations produced by MSG evidenced by stimulation ofNa+/K+-ATPase and AChE activities in the cerebral cortex and hippocampus of rats were normalized by 4-PSQ treatment. CONCLUSIONS In brief, 4-PSQ therapy improved hypothalamic obesity-related parameters, as well as psychiatric symptoms, cognitive impairment, and neurochemical alterations found in obese rats.
Collapse
Affiliation(s)
- Karline C Rodrigues
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Pesquisa em Farmacologia Bioquímica (LaFarBio), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), Campus Capão do Leão, Pelotas, RS, CEP 96010-900, Brazil
| | - Cristiani F Bortolatto
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Bioquímica e Neurofarmacologia Molecular (LABIONEM), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), Pelotas, RS, CEP 96010-900, Brazil
| | - Ketlyn P da Motta
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Pesquisa em Farmacologia Bioquímica (LaFarBio), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), Campus Capão do Leão, Pelotas, RS, CEP 96010-900, Brazil
| | - Renata L de Oliveira
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Pesquisa em Farmacologia Bioquímica (LaFarBio), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), Campus Capão do Leão, Pelotas, RS, CEP 96010-900, Brazil
| | - Jaini J Paltian
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Pesquisa em Farmacologia Bioquímica (LaFarBio), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), Campus Capão do Leão, Pelotas, RS, CEP 96010-900, Brazil
| | - Roberta Krüger
- Programa de Pós-graduação em Química, Laboratório de Síntese Orgânica Limpa - LASOL, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), P.O. Box 354, Pelotas, RS, 96010-900, Brazil
| | - Silvane S Roman
- Universidade Regional Integrada, Campus Erechim, Erechim, RS, CEP 99700-000, Brazil
| | - Silvana P Boeira
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas, LaftamBio Pampa, Universidade Federal do Pampa, Itaqui, RS, CEP 97650-000, Brazil
| | - Diego Alves
- Programa de Pós-graduação em Química, Laboratório de Síntese Orgânica Limpa - LASOL, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), P.O. Box 354, Pelotas, RS, 96010-900, Brazil
| | - Ethel Antunes Wilhelm
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Pesquisa em Farmacologia Bioquímica (LaFarBio), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), Campus Capão do Leão, Pelotas, RS, CEP 96010-900, Brazil.
| | - Cristiane Luchese
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Pesquisa em Farmacologia Bioquímica (LaFarBio), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), Campus Capão do Leão, Pelotas, RS, CEP 96010-900, Brazil.
| |
Collapse
|
46
|
Activation of Hippocampal IR/IRS-1 Signaling Contributes to the Treatment with Zuogui Jiangtang Jieyu Decoction on the Diabetes-Related Depression. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6688723. [PMID: 34149862 PMCID: PMC8195672 DOI: 10.1155/2021/6688723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 05/25/2021] [Indexed: 11/17/2022]
Abstract
Background Zuogui Jiangtang Jieyu decoction (ZJJ) is mainly used for the treatment of diabetes-related depression in current clinical applications and research. This study aims to investigate whether the brain IR/IRS-1 signaling pathway is involved in the therapeutic effect of ZJJ on depression-like behavior in diabetic rats. Methods Sprague–Dawley rats were fed with high-fat diet and subjected to streptozotocin injection to establish the diabetes animal model. After treatment with different doses of ZJJ (20.530 g/kg or 10.265 g/kg) for 4 weeks, the blood glucose level and peripheral insulin resistance were measured. The forced-swimming test (FST) and Morris water maze test (MWMT) were applied for the mood and cognitive function assessment. Then, the Western blot method was used to analyze the protein levels of insulin receptor (IR), insulin receptor substrate-1 (IRS-1), phosphatidylonositol-3-kinase (PI3K), and protein kinase B (PKB, also as known as AKT) in the hippocampus of diabetic rats. Meanwhile, the immunofluorescence method was performed to analyze the above proteins' expression in the neuron and astrocyte. At last, the levels of glycogen, lactate, and ATP were tested by the ELISA method. Additionally, the insulin-sensitive glucose transporter 4 (GLUT4) and the lactate transporter monocarboxylate transporter 4 (MCT4) were analyzed by the Western blot method. Results ZJJ administration significantly decreased the level of blood glucose and improved the peripheral insulin resistance in diabetic rats. Besides, ZJJ attenuated the depression-like behavior and the cognitive dysfunction in rats with diabetes. Furthermore, we found the upregulation of protein expression of phospho-IR, phospho-IRS-1, phospho-PI3K, and phospho-AKT in the hippocampus of diabetic rats after being treated with ZJJ. Moreover, the above proteins are increased not only in the neuron but also in the astrocyte after ZJJ administration. In addition, ZJJ increased the content of ATP, glycogen, and lactate, as well as the expression of GLUT4 and MCT4 in the hippocampus of diabetic rats. Conclusions These findings suggest that ZJJ improves the depression-like behavior of diabetic rats by activating the IR/IRS-1 signaling pathway in both hippocampal neuron and astrocyte. And the brain IR/IRS-1 signaling pathway plays an important role in astrocyte-neuron metabolic coupling, providing a potential mechanism by which the IR/IRS-1 signaling pathway may contribute to the treatment of ZJJ on diabetes-related depression.
Collapse
|
47
|
Edem EE, Nathaniel BU, Nebo KE, Obisesan AO, Olabiyi AA, Akinluyi ET, Ishola AO. Lactobacillus plantarum mitigates sexual-reproductive deficits by modulating insulin receptor expression in the hypothalamic-pituitary-testicular axis of hyperinsulinemic mice. Drug Metab Pers Ther 2021; 36:321-336. [PMID: 34002580 DOI: 10.1515/dmpt-2021-1000195] [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] [Received: 01/01/2021] [Accepted: 04/05/2021] [Indexed: 01/15/2023]
Abstract
OBJECTIVES Hyperinsulinemia increases the risk factor of diabetes and infertility at a manifold. Lactobacillus plantarum has several medical significances with limited reports. Hence, this study assessed the effect of L. plantarum on sexual-reproductive functions and distribution of insulin receptors in the hypothalamic-pituitary-testicular axis of hyperinsulinemic mice. METHODS Forty male adult mice were divided into five groups as follows: control, high-fat diet (HFD) + streptozotocin (STZ), therapeutic, co-administration group type 1 (CO-AD) and probiotics. They were either simultaneously exposed to an HFD and L. plantarum treatment for 28 days with a dose of STZ injection to induce hyperinsulinemia on day 28 or treated with L. plantarum for 14 days, and following induction of hyperinsulinemia. Mice were subjected to a sexual behavioural test and thereafter sacrificed under euthanasia condition. Blood, brain and testes were collected for biochemical and immunohistochemical assays. RESULTS Treatment with L. plantarum ameliorated reproductive hormones activity disruption, sexual behavioural defects, antioxidant imbalance, insulin dysregulation and lipid metabolism dysfunction following exposure to HFD + STZ when compared to the hyperinsulinemic untreated mice. CONCLUSIONS Taken together, data from this study reveal that L. plantarum abrogated hyperinsulinemia-induced male sexual and reproductive deficits by modulating antioxidant status, lipid metabolism and insulin signalling in the hypothalamic-pituitary-testicular axis of mice.
Collapse
Affiliation(s)
- Edem Ekpenyong Edem
- Department of Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
| | - Blessing Uyo Nathaniel
- Department of Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
| | - Kate Eberechukwu Nebo
- Department of Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
| | - Abiola Oluwatosin Obisesan
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
| | - Ayodeji Augustine Olabiyi
- Department of Medical Biochemistry, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
| | - Elizabeth Toyin Akinluyi
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
| | - Azeez Olakunle Ishola
- Department of Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
| |
Collapse
|
48
|
Muth AK, Park SQ. The impact of dietary macronutrient intake on cognitive function and the brain. Clin Nutr 2021; 40:3999-4010. [PMID: 34139473 DOI: 10.1016/j.clnu.2021.04.043] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 04/19/2021] [Accepted: 04/23/2021] [Indexed: 12/20/2022]
Abstract
Macronutrients - carbohydrates, fats, and proteins - supply the nutrients required for optimal functioning. Inadequate intake compromises both physical and brain health. We synthesized research on macronutrients from whole meals on cognitive function in healthy adults and identified underlying mechanisms. Intake of simple carbohydrates ('sugars') is consistently associated with decreased global cognition whereas consumption of complex carbohydrates correlates with successful brain aging and improved memory both in the short- and long-term. Saturated fatty acid intake correlates with decreased memory and learning scores whereas omega-3 intake correlates positively with memory scores. Protein intake boosts executive function and working memory when task-demands are high. Individual differences affecting the macronutrient-cognition relationship are age, physical activity, and glucose metabolism. Neural correlates reflect findings on cognitive functions: cortical thickness and cerebral amyloid burden correlate with sugar intake, inflammatory status and cerebral glucose metabolism correlate with fatty acid intake. Key mechanisms by which dietary macronutrients affect the brain and cognition include glucose and insulin metabolism, neurotransmitter actions, and cerebral oxidation and inflammation. In conclusion, macronutrient intake affects cognitive function both acutely and in the long-term, involving peripheral and central mechanisms. A healthy diet supports brain integrity and functionality, whereas inadequate nutrition compromises it. Studying diet can be key to nutritional recommendations, thereby improving the landscape of mental health and healthy brain aging.
Collapse
Affiliation(s)
- Anne-Katrin Muth
- Department of Decision Neuroscience and Nutrition, German Institute of Human Nutrition (DIfE), Potsdam-Rehbrücke, Germany; Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Neuroscience Research Center, 10117, Berlin, Germany.
| | - Soyoung Q Park
- Department of Decision Neuroscience and Nutrition, German Institute of Human Nutrition (DIfE), Potsdam-Rehbrücke, Germany; Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Neuroscience Research Center, 10117, Berlin, Germany; Deutsches Zentrum für Diabetes, Neuherberg, Germany.
| |
Collapse
|
49
|
Wilkialis L, Rodrigues N, Majeed A, Lee Y, Lipsitz O, Gill H, Tamura J, Nasri F, Lui LMW, Siegel A, Mansur RB, Rosenblat JD, McIntyre RS. Loneliness-based impaired reward system pathway: Theoretical and clinical analysis and application. Psychiatry Res 2021; 298:113800. [PMID: 33618235 DOI: 10.1016/j.psychres.2021.113800] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 02/07/2021] [Indexed: 12/26/2022]
Abstract
Loneliness is a key determinant in the etiology of mental health disorders such as depression and has profound impacts on health, quality of life, and economic productivity. This narrative review uses extant neurobiology and evolutionary literature to propose a construct through which loneliness may induce depression in adulthood via the reward system (including symptom and treatment aspects). Early childhood (distal) factors were found to be important in influencing adult (proximal) factors, which lead to the formulation of the construct. Due to the heterogenous and comorbid nature of depression, a new subtype known as 'reward depression' was distinguished along with distinct symptoms to aid practitioners when assessing patient treatment options. Furthermore, an evolutionary perspective was applied to the current impaired reward construct to discuss how the ancestral purpose and environment (in terms of reward) clashes with the modern one. Finally, theoretical treatment and prevention ideas were examined and discussed, leading into future work that needs to build upon and confirm the outlined construct.
Collapse
Affiliation(s)
- Linas Wilkialis
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Nelson Rodrigues
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Amna Majeed
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Yena Lee
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Orly Lipsitz
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Hartej Gill
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Jocelyn Tamura
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Flora Nasri
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Leanna M W Lui
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Ashley Siegel
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Rodrigo B Mansur
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Joshua D Rosenblat
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Department of Pharmacology, University of Toronto, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Roger S McIntyre
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Department of Pharmacology, University of Toronto, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Brain and Cognition Discovery Foundation, Toronto, ON, Canada.
| |
Collapse
|
50
|
Beddows CA, Dodd GT. Insulin on the brain: The role of central insulin signalling in energy and glucose homeostasis. J Neuroendocrinol 2021; 33:e12947. [PMID: 33687120 DOI: 10.1111/jne.12947] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/26/2021] [Accepted: 01/28/2021] [Indexed: 12/26/2022]
Abstract
Insulin signals to the brain where it coordinates multiple physiological processes underlying energy and glucose homeostasis. This review explores where and how insulin interacts within the brain parenchyma, how brain insulin signalling functions to coordinate energy and glucose homeostasis and how this contributes to the pathogenesis of metabolic disease.
Collapse
Affiliation(s)
- Cait A Beddows
- Department of Anatomy and Physiology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, Australia
| | - Garron T Dodd
- Department of Anatomy and Physiology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|