1
|
Tanaka M, Szabó Á, Vécsei L. Redefining Roles: A Paradigm Shift in Tryptophan-Kynurenine Metabolism for Innovative Clinical Applications. Int J Mol Sci 2024; 25:12767. [PMID: 39684480 DOI: 10.3390/ijms252312767] [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: 10/14/2024] [Revised: 11/16/2024] [Accepted: 11/23/2024] [Indexed: 12/18/2024] Open
Abstract
The tryptophan-kynurenine (KYN) pathway has long been recognized for its essential role in generating metabolites that influence various physiological processes. Traditionally, these metabolites have been categorized into distinct, often opposing groups, such as pro-oxidant versus antioxidant, excitotoxic/neurotoxic versus neuroprotective. This dichotomous framework has shaped much of the research on conditions like neurodegenerative and neuropsychiatric disorders, as well as cancer, where metabolic imbalances are a key feature. The effects are significantly influenced by various factors, including the concentration of metabolites and the particular cellular milieu in which they are generated. A molecule that acts as neuroprotective at low concentrations may exhibit neurotoxic effects at elevated levels. The oxidative equilibrium of the surrounding environment can alter the function of KYN from an antioxidant to a pro-oxidant. This narrative review offers a comprehensive examination and analysis of the contemporary understanding of KYN metabolites, emphasizing their multifaceted biological functions and their relevance in numerous physiological and pathological processes. This underscores the pressing necessity for a paradigm shift in the comprehension of KYN metabolism. Understanding the context-dependent roles of KYN metabolites is vital for novel therapies in conditions like Alzheimer's disease, multiple sclerosis, and cancer. Comprehensive pathway modulation, including balancing inflammatory signals and enzyme regulation, offers promising avenues for targeted, effective treatments.
Collapse
Affiliation(s)
- Masaru Tanaka
- HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Danube Neuroscience Research Laboratory, Tisza Lajos krt. 113, H-6725 Szeged, Hungary
| | - Ágnes Szabó
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
- Doctoral School of Clinical Medicine, University of Szeged, Korányi fasor 6, H-6720 Szeged, Hungary
| | - László Vécsei
- HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Danube Neuroscience Research Laboratory, Tisza Lajos krt. 113, H-6725 Szeged, Hungary
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| |
Collapse
|
2
|
Marín-García PJ, Piles M, Sánchez JP, Pascual M, Llobat L, Pascual JJ, Hedemann MS. Untargeted urine metabolomics suggests that ascorbic acid may serve as a promising biomarker for reduced feed intake in rabbits. Sci Rep 2024; 14:29180. [PMID: 39587239 PMCID: PMC11589781 DOI: 10.1038/s41598-024-80701-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 11/21/2024] [Indexed: 11/27/2024] Open
Abstract
Feed restriction is a common nutritional practice in rabbit farming; however, decreased feed intake can also signal potential digestive disorders at an early stage. This study endeavors to investigate the impact of feed restriction on selected productive traits and the urinary metabolome of juvenile rabbits across diverse genetic backgrounds. Our objective is to identify potential biomarkers capable of detecting periods of fasting. A total of 48 growing rabbits were used from two genetic types: Prat line (selected for litter size at weaning, n = 24) and Caldes line (selected for post-weaning growth rate, n = 24). At 60 days of age, a digestibility trial was carried out. Changes in productive traits (through bioelectrical impedance analysis, live weight control, average daily gain, energy, and protein retention) were evaluated when the animals were fed ad libitum from 60 to 64 days of age and when the same animals were subjected to feed restriction (50% of maintenance energy requirements) from 70 to 74 days of age, in a split-plot trial. In addition, untargeted urine metabolomics analysis was performed at both periods (ad libitum vs. restricted). Although some differences between genetic lines were observed in the animals' performance traits (average daily gain and retention of energy and protein), no differences in the urine metabolome were found between genetic types. However, feed restriction caused notable changes in the metabolome. When the animals were subjected to feed restriction, they had higher levels of ascorbic acid (P = 0.001) and p-cresol sulphate (P = 0.058) and lower levels of pyrocatechol sulphate/hydroquinone sulphate (P < 0.001), resorcinol sulphate (P = 0.002), enterolactone sulphate (P < 0.001), enterolactone (P < 0.001), kynurenic acid (P = 0.0002), proline betaine (P < 0.001), pipecolic acid betaine (P < 0.001), xanthurenic acid (P < 0.001) and quinaldic acid (P < 0.001) than the same animals when they were fed ad libitum. This study proposes urine ascorbic acid as potential biomarker for fasting events in rabbits. As urine ascorbic acid is the sole metabolite that significantly increases in the restricted group, it offers promising indicator for early detection and targeted management of digestive disorders in rabbits.
Collapse
Affiliation(s)
- Pablo Jesús Marín-García
- Department of Animal Production and Health, Veterinary Public Health and Food Science and Technology (PASAPTA), Facultad de Veterinaria, Universidad Cardenal Herrera-CEU, CEU Universities, 46113, Valencia, Spain.
| | - Miriam Piles
- Institute of Agrifood Research and Technology (IRTA)-Animal Breeding and Genetics, Caldes de Montbui, Barcelona, Spain
| | - Juan Pablo Sánchez
- Institute of Agrifood Research and Technology (IRTA)-Animal Breeding and Genetics, Caldes de Montbui, Barcelona, Spain
| | - Mariam Pascual
- Institute of Agrifood Research and Technology (IRTA)-Animal Breeding and Genetics, Caldes de Montbui, Barcelona, Spain
| | - Lola Llobat
- Department of Animal Production and Health, Veterinary Public Health and Food Science and Technology (PASAPTA), Facultad de Veterinaria, Universidad Cardenal Herrera-CEU, CEU Universities, 46113, Valencia, Spain
| | - Juan José Pascual
- Institute for Animal Science and Technology, Universitat Politècnica de València, Camino de Vera s/n, 46022, Valencia, Spain
| | - Mette Skou Hedemann
- Department of Animal and Veterinary Sciences, Aarhus University, Blichers Alle 20, 8830, Tjele, Denmark
| |
Collapse
|
3
|
Angiari S, Carlucci T, Budui SL, Bach SD, Dusi S, Walter J, Ellmeier E, Schnabl A, Stracke A, Bordag N, Tafrali C, Demjaha R, Khalil M, Angelini G, Terrabuio E, Pietronigro EC, Zenaro E, Laudanna C, Rossi B, Constantin G. Coenzyme A fueling with pantethine limits autoreactive T cell pathogenicity in experimental neuroinflammation. J Neuroinflammation 2024; 21:287. [PMID: 39501296 PMCID: PMC11536535 DOI: 10.1186/s12974-024-03270-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 10/22/2024] [Indexed: 11/09/2024] Open
Abstract
BACKGROUND Immune cell metabolism governs the outcome of immune responses and contributes to the development of autoimmunity by controlling lymphocyte pathogenic potential. In this study, we evaluated the metabolic profile of myelin-specific murine encephalitogenic T cells, to identify novel therapeutic targets for autoimmune neuroinflammation. METHODS We performed metabolomics analysis on actively-proliferating encephalitogenic T cells to study their overall metabolic profile in comparison to resting T cells. Metabolomics, phosphoproteomics, in vitro functional assays, and in vivo studies in experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis (MS), were then implemented to evaluate the effect of metabolic targeting on autoreactive T cell pathogenicity. Finally, we confirmed the translational potential of our targeting approach in human pro-inflammatory T helper cell subsets and in T cells from MS patients. RESULTS We found that autoreactive encephalitogenic T cells display an altered coenzyme A (CoA) synthesis pathway, compared to resting T cells. CoA fueling with the CoA precursor pantethine (PTTH) affected essential immune-related processes of myelin-specific T cells, such as cell proliferation, cytokine production, and cell adhesion, both in vitro and in vivo. Accordingly, pre-clinical treatment with PTTH before disease onset inhibited the development of EAE by limiting T cell pro-inflammatory potential in vivo. Importantly, PTTH also significantly ameliorated the disease course when administered after disease onset in a therapeutic setting. Finally, PTTH reduced pro-inflammatory cytokine production by human T helper 1 (Th1) and Th17 cells and by T cells from MS patients, confirming its translational potential. CONCLUSION Our data demonstrate that CoA fueling with PTTH in pro-inflammatory and autoreactive T cells may represent a novel therapeutic approach for the treatment of autoimmune neuroinflammation.
Collapse
Affiliation(s)
- Stefano Angiari
- Otto Loewi Research Center, Division of Immunology, Medical University of Graz, Neue Stiftingtalstraße 6, 8010, Graz, Austria.
| | - Tommaso Carlucci
- Department of Medicine, Section of General Pathology, University of Verona, Strada le Grazie 8, 37134, Verona, Italy
| | - Simona L Budui
- Department of Medicine, Section of General Pathology, University of Verona, Strada le Grazie 8, 37134, Verona, Italy
| | - Simone D Bach
- Department of Medicine, Section of General Pathology, University of Verona, Strada le Grazie 8, 37134, Verona, Italy
| | - Silvia Dusi
- Department of Medicine, Section of General Pathology, University of Verona, Strada le Grazie 8, 37134, Verona, Italy
| | - Julia Walter
- Otto Loewi Research Center, Division of Immunology, Medical University of Graz, Neue Stiftingtalstraße 6, 8010, Graz, Austria
| | - Elena Ellmeier
- Otto Loewi Research Center, Division of Immunology, Medical University of Graz, Neue Stiftingtalstraße 6, 8010, Graz, Austria
| | - Alyssa Schnabl
- Otto Loewi Research Center, Division of Immunology, Medical University of Graz, Neue Stiftingtalstraße 6, 8010, Graz, Austria
| | - Anika Stracke
- Otto Loewi Research Center, Division of Immunology, Medical University of Graz, Neue Stiftingtalstraße 6, 8010, Graz, Austria
| | - Natalie Bordag
- Department of Dermatology and Venereology, Medical University of Graz, Graz, Austria
| | - Cansu Tafrali
- Department of Neurology, Medical University of Graz, Graz, Austria
| | - Rina Demjaha
- Department of Neurology, Medical University of Graz, Graz, Austria
| | - Michael Khalil
- Department of Neurology, Medical University of Graz, Graz, Austria
| | - Gabriele Angelini
- Department of Medicine, Section of General Pathology, University of Verona, Strada le Grazie 8, 37134, Verona, Italy
| | - Eleonora Terrabuio
- Department of Medicine, Section of General Pathology, University of Verona, Strada le Grazie 8, 37134, Verona, Italy
| | - Enrica C Pietronigro
- Department of Medicine, Section of General Pathology, University of Verona, Strada le Grazie 8, 37134, Verona, Italy
| | - Elena Zenaro
- Department of Medicine, Section of General Pathology, University of Verona, Strada le Grazie 8, 37134, Verona, Italy
| | - Carlo Laudanna
- Department of Medicine, Section of General Pathology, University of Verona, Strada le Grazie 8, 37134, Verona, Italy
- The Center for Biomedical Computing (CBMC), University of Verona, Verona, Italy
| | - Barbara Rossi
- Department of Medicine, Section of General Pathology, University of Verona, Strada le Grazie 8, 37134, Verona, Italy
| | - Gabriela Constantin
- Department of Medicine, Section of General Pathology, University of Verona, Strada le Grazie 8, 37134, Verona, Italy.
| |
Collapse
|
4
|
Bogielski B, Michalczyk K, Głodek P, Tempka B, Gębski W, Stygar D. Association between small intestine bacterial overgrowth and psychiatric disorders. Front Endocrinol (Lausanne) 2024; 15:1438066. [PMID: 39497810 PMCID: PMC11532184 DOI: 10.3389/fendo.2024.1438066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 10/01/2024] [Indexed: 11/07/2024] Open
Abstract
Small intestinal bacterial overgrowth (SIBO) is a gastrointestinal condition characterized by abnormal colonization of bacteria in the small intestine, leading to overgrowth and alteration, which is linked to gastrointestinal issues, potentially affecting neurological and mental health. Despite existing research, we still do not understand how SIBO affects tryptophan metabolism and psychiatric diseases. We investigated the literature for connections between SIBO, tryptophan metabolism disruptions, and psychiatric disorders like autism, schizophrenia, Alzheimer's, and Parkinson's diseases. We also explored the interaction between thyroid disorders and their influence on SIBO and psychiatric illnesses. PubMed and Google Scholar databases were searched using keywords and phrases, individual and in combinations, like "SIBO," "gut microbiota," "neurologic disorders," "mental disorders," "tryptophan," "dopamine," and "thyroid disease." We focused on original research and review papers that presented empirical studies conducted on animal models and human subjects published in English between February 1992 to February 2023. The initial 2 634 534 records were preliminary screened based on title and abstract and then subjected to full-text review to exclude publications with insufficient data on SIBO, lack of a psychiatric disorder component, or methodological limitations compromising the integrity of the findings. The analysis highlights the significance of the association between psychiatric disorders and SIBO, emphasizing the role of gut-microbial diversity in mental health. We advocate for more detailed studies, including longitudinal research, to clarify the causal relationships between SIBO, gut dysbiosis, and psychiatric disorders and for an integrated approach while treating complex psychiatric conditions.
Collapse
Affiliation(s)
| | | | | | | | | | - Dominika Stygar
- Department of Physiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Zabrze, Poland
| |
Collapse
|
5
|
Tossetta G, Fantone S, Togni L, Santarelli A, Olivieri F, Marzioni D, Rippo MR. Modulation of NRF2/KEAP1 Signaling by Phytotherapeutics in Periodontitis. Antioxidants (Basel) 2024; 13:1270. [PMID: 39456522 PMCID: PMC11504014 DOI: 10.3390/antiox13101270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Periodontitis affects up to 40% of adults over 60 years old and is a consequence of gingivitis. Periodontitis is characterized by a chronic inflammation, periodontal damage, and alveolar bone resorption. The nuclear factor erythroid 2-related factor 2 (NFE2L2 or NRF2)/Kelch-like ECH-Associated Protein 1 (KEAP1) (NRF2/KEAP1) signaling pathway plays a key role in periodontitis by modulating redox balance and inflammation of the periodontium. However, NRF2 expression is decreased in gingival tissues of patients with periodontitis while oxidative stress is significantly increased in this pathology. Oxidative stress and lipopolysaccharide (LPS) produced by gram-negative bacteria favor the production of inflammatory causing periodontal inflammation and favoring alveolar bone. In this review, we analyzed the current literature regarding the role of natural and synthetic compounds in modulating the NRF2/KEAP1 pathway in in vitro and in vivo models of periodontitis in order to evaluate new potential treatments of periodontitis that can improve the outcome of this disease.
Collapse
Affiliation(s)
- Giovanni Tossetta
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy;
| | - Sonia Fantone
- Scientific Direction, IRCCS INRCA, 60124 Ancona, Italy; (S.F.); (F.O.)
| | - Lucrezia Togni
- Department of Clinical Specialistic and Dental Sciences, Università Politecnica delle Marche, 60126 Ancona, Italy; (L.T.); (A.S.)
| | - Andrea Santarelli
- Department of Clinical Specialistic and Dental Sciences, Università Politecnica delle Marche, 60126 Ancona, Italy; (L.T.); (A.S.)
- Dentistry Clinic, National Institute of Health and Science of Aging, IRCCS INRCA, 60126 Ancona, Italy
| | - Fabiola Olivieri
- Scientific Direction, IRCCS INRCA, 60124 Ancona, Italy; (S.F.); (F.O.)
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, 60126 Ancona, Italy;
| | - Daniela Marzioni
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy;
- IRCCS INRCA, 60124 Ancona, Italy
| | - Maria Rita Rippo
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, 60126 Ancona, Italy;
| |
Collapse
|
6
|
Alves LDF, Moore JB, Kell DB. The Biology and Biochemistry of Kynurenic Acid, a Potential Nutraceutical with Multiple Biological Effects. Int J Mol Sci 2024; 25:9082. [PMID: 39201768 PMCID: PMC11354673 DOI: 10.3390/ijms25169082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 09/03/2024] Open
Abstract
Kynurenic acid (KYNA) is an antioxidant degradation product of tryptophan that has been shown to have a variety of cytoprotective, neuroprotective and neuronal signalling properties. However, mammalian transporters and receptors display micromolar binding constants; these are consistent with its typically micromolar tissue concentrations but far above its serum/plasma concentration (normally tens of nanomolar), suggesting large gaps in our knowledge of its transport and mechanisms of action, in that the main influx transporters characterized to date are equilibrative, not concentrative. In addition, it is a substrate of a known anion efflux pump (ABCC4), whose in vivo activity is largely unknown. Exogeneous addition of L-tryptophan or L-kynurenine leads to the production of KYNA but also to that of many other co-metabolites (including some such as 3-hydroxy-L-kynurenine and quinolinic acid that may be toxic). With the exception of chestnut honey, KYNA exists at relatively low levels in natural foodstuffs. However, its bioavailability is reasonable, and as the terminal element of an irreversible reaction of most tryptophan degradation pathways, it might be added exogenously without disturbing upstream metabolism significantly. Many examples, which we review, show that it has valuable bioactivity. Given the above, we review its potential utility as a nutraceutical, finding it significantly worthy of further study and development.
Collapse
Affiliation(s)
- Luana de Fátima Alves
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Building 220, Søltofts Plads, 2800 Kongens Lyngby, Denmark
| | - J. Bernadette Moore
- School of Food Science & Nutrition, University of Leeds, Leeds LS2 9JT, UK;
- Department of Biochemistry, Cell & Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown St., Liverpool L69 7ZB, UK
| | - Douglas B. Kell
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Building 220, Søltofts Plads, 2800 Kongens Lyngby, Denmark
- Department of Biochemistry, Cell & Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown St., Liverpool L69 7ZB, UK
| |
Collapse
|
7
|
Stone TW, Darlington LG, Badawy AAB, Williams RO. The Complex World of Kynurenic Acid: Reflections on Biological Issues and Therapeutic Strategy. Int J Mol Sci 2024; 25:9040. [PMID: 39201726 PMCID: PMC11354734 DOI: 10.3390/ijms25169040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 09/03/2024] Open
Abstract
It has been unequivocally established that kynurenic acid has a number of actions in a variety of cells and tissues, raising, in principle, the possibility of targeting its generation, metabolism or sites of action to manipulate those effects to a beneficial therapeutic end. However, many basic aspects of the biology of kynurenic acid remain unclear, potentially leading to some confusion and misinterpretations of data. They include questions of the source, generation, targets, enzyme expression, endogenous concentrations and sites of action. This essay is intended to raise and discuss many of these aspects as a source of reference for more balanced discussion. Those issues are followed by examples of situations in which modulating and correcting kynurenic acid production or activity could bring significant therapeutic benefit, including neurological and psychiatric conditions, inflammatory diseases and cell protection. More information is required to obtain a clear overall view of the pharmacological environment relevant to kynurenic acid, especially with respect to the active concentrations of kynurenine metabolites in vivo and changed levels in disease. The data and ideas presented here should permit a greater confidence in appreciating the sites of action and interaction of kynurenic acid under different local conditions and pathologies, enhancing our understanding of kynurenic acid itself and the many clinical conditions in which manipulating its pharmacology could be of clinical value.
Collapse
Affiliation(s)
- Trevor W. Stone
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7FY, UK;
| | - L. Gail Darlington
- Worthing Hospital, University Hospitals Sussex NHS Foundation Trust, Worthing BN11 2DH, UK
| | - Abdulla A.-B. Badawy
- Formerly School of Health Sciences, Cardiff Metropolitan University, Cardiff CF5 2YB, UK
| | - Richard O. Williams
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7FY, UK;
| |
Collapse
|
8
|
Brouwer A, Carhart‐Harris RL, Raison CL. Psychotomimetic compensation versus sensitization. Pharmacol Res Perspect 2024; 12:e1217. [PMID: 38923845 PMCID: PMC11194300 DOI: 10.1002/prp2.1217] [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: 04/11/2024] [Accepted: 05/08/2024] [Indexed: 06/28/2024] Open
Abstract
It is a paradox that psychotomimetic drugs can relieve symptoms that increase risk of and cooccur with psychosis, such as attention and motivational deficits (e.g., amphetamines), pain (e.g., cannabis) and symptoms of depression (e.g., psychedelics, dissociatives). We introduce the ideas of psychotomimetic compensation and psychotomimetic sensitization to explain this paradox. Psychotomimetic compensation refers to a short-term stressor or drug-induced compensation against stress that is facilitated by engagement of neurotransmitter/modulator systems (endocannabinoid, serotonergic, glutamatergic and dopaminergic) that mediate the effects of common psychotomimetic drugs. Psychotomimetic sensitization occurs after repeated exposure to stress and/or drugs and is evidenced by the gradual intensification and increase of psychotic-like experiences over time. Theoretical and practical implications of this model are discussed.
Collapse
Affiliation(s)
- Ari Brouwer
- Department of Human Development and Family Studies, School of Human EcologyUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Robin L. Carhart‐Harris
- Department of Neurology and PsychiatryUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Charles L. Raison
- Department of Psychiatry, School of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Vail Health Behavioral Health Innovation CenterVailColoradoUSA
- Center for the Study of Human HealthEmory UniversityAtlantaGeorgiaUSA
- Department of Spiritual HealthEmory University Woodruff Health Sciences CenterAtlantaGeorgiaUSA
| |
Collapse
|
9
|
Carrillo-Mora P, Landa-Solís C, Valle-Garcia D, Luna-Angulo A, Avilés-Arnaut H, Robles-Bañuelos B, Sánchez-Chapul L, Rangel-López E. Kynurenines and Inflammation: A Remarkable Axis for Multiple Sclerosis Treatment. Pharmaceuticals (Basel) 2024; 17:983. [PMID: 39204088 PMCID: PMC11356993 DOI: 10.3390/ph17080983] [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: 06/22/2024] [Revised: 07/20/2024] [Accepted: 07/23/2024] [Indexed: 09/03/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory autoimmune neurological disease characterized by the recurrent appearance of demyelinating lesions and progressive disability. Currently, there are multiple disease-modifying treatments, however, there is a significant need to develop new therapeutic targets, especially for the progressive forms of the disease. This review article provides an overview of the most recent studies aimed at understanding the inflammatory processes that are activated in response to the accumulation of kynurenine pathway (KP) metabolites, which exacerbate an imbalance between immune system cells (e.g., Th1, Th2, and T reg) and promote the release of pro-inflammatory interleukins that modulate different mechanisms: membrane-receptors function; nuclear factors expression; and cellular signals. Together, these alterations trigger cell death mechanisms in brain cells and promote neuron loss and axon demyelination. This hypothesis could represent a remarkable approach for disease-modifying therapies for MS. Here, we also provide a perspective on the repositioning of some already approved drugs involved in other signaling pathways, which could represent new therapeutic strategies for MS treatment.
Collapse
Affiliation(s)
- Paul Carrillo-Mora
- Clinical Neurosciences Division, National Institute of Rehabilitation “Luis Guillermo Ibarra Ibarra”, Mexico City 14389, Mexico;
| | - Carlos Landa-Solís
- Tissue Engineering, Cell Therapy, and Regenerative Medicine Unit, National Institute of Rehabilitation “Luis Guillermo Ibarra Ibarra”, Mexico City 14389, Mexico;
| | - David Valle-Garcia
- Neuroimmunology Laboratory, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico;
| | - Alexandra Luna-Angulo
- Neuromuscular Diseases Laboratory, Clinical Neurosciences Division, National Institute of Rehabilitation “Luis Guillermo Ibarra Ibarra”, Mexico City 14389, Mexico;
| | - Hamlet Avilés-Arnaut
- Faculty of Biological Sciences, Institute of Biotechnology, National Autonomous University of Nuevo Leon, Nuevo León 66455, Mexico;
| | - Benjamín Robles-Bañuelos
- Cell Reprogramming Laboratory, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico;
| | - Laura Sánchez-Chapul
- Neuromuscular Diseases Laboratory, Clinical Neurosciences Division, National Institute of Rehabilitation “Luis Guillermo Ibarra Ibarra”, Mexico City 14389, Mexico;
| | - Edgar Rangel-López
- Cell Reprogramming Laboratory, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico;
| |
Collapse
|
10
|
Hu H, Huang Y, Li A, Mi Q, Wang K, Chen L, Zhao Z, Zhang Q, Bai X, Pan H. Effects of different energy levels in low-protein diet on liver lipid metabolism in the late-phase laying hens through the gut-liver axis. J Anim Sci Biotechnol 2024; 15:98. [PMID: 38987834 PMCID: PMC11238517 DOI: 10.1186/s40104-024-01055-y] [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: 12/16/2023] [Accepted: 05/26/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND The energy/protein imbalance in a low-protein diet induces lipid metabolism disorders in late-phase laying hens. Reducing energy levels in the low-protein diet to adjust the energy-to-protein ratio may improve fat deposition, but this also decreases the laying performance of hens. This study investigated the mechanism by which different energy levels in the low-protein diet influences liver lipid metabolism in late-phase laying hens through the enterohepatic axis to guide feed optimization and nutrition strategies. A total of 288 laying hens were randomly allocated to the normal-energy and normal-protein diet group (positive control: CK) or 1 of 3 groups: low-energy and low-protein diet (LL), normal-energy and low-protein diet (NL), and high-energy and low-protein diet (HL) groups. The energy-to-protein ratios of the CK, LL, NL, and HL diets were 0.67, 0.74, 0.77, and 0.80, respectively. RESULTS Compared with the CK group, egg quality deteriorated with increasing energy intake in late-phase laying hens fed low-protein diet. Hens fed LL, NL, and HL diets had significantly higher triglyceride, total cholesterol, acetyl-CoA carboxylase, and fatty acid synthase levels, but significantly lower hepatic lipase levels compared with the CK group. Liver transcriptome sequencing revealed that genes involved in fatty acid beta-oxidation (ACOX1, HADHA, EHHADH, and ACAA1) were downregulated, whereas genes related to fatty acid synthesis (SCD, FASN, and ACACA) were upregulated in LL group compared with the CK group. Comparison of the cecal microbiome showed that in hens fed an LL diet, Lactobacillus and Desulfovibrio were enriched, whereas riboflavin metabolism was suppressed. Cecal metabolites that were most significantly affected by the LL diet included several vitamins, such as riboflavin (vitamin B2), pantethine (vitamin B5 derivative), pyridoxine (vitamin B6), and 4-pyridoxic acid. CONCLUSION A lipid metabolism disorder due to deficiencies of vitamin B2 and pantethine originating from the metabolism of the cecal microbiome may be the underlying reason for fat accumulation in the liver of late-phase laying hens fed an LL diet. Based on the present study, we propose that targeting vitamin B2 and pantethine (vitamin B5 derivative) might be an effective strategy for improving lipid metabolism in late-phase laying hens fed a low-protein diet.
Collapse
Affiliation(s)
- Hong Hu
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Ying Huang
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Anjian Li
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Qianhui Mi
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Kunping Wang
- College of Animal Science, Anhui Science and Technology University, Bengbu, 233000, China
| | - Liang Chen
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agriculture Sciences, Beijing, 100193, China
| | - Zelong Zhao
- Shanghai BIOZERON Biotechnology Co., Ltd, Shanghai, 201800, China
| | - Qiang Zhang
- WOD Poultry Research Institute, Beijing, 100193, China
| | - Xi Bai
- College of Animal Science, Anhui Science and Technology University, Bengbu, 233000, China.
| | - Hongbin Pan
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China.
| |
Collapse
|
11
|
Grishanova AY, Perepechaeva ML. Kynurenic Acid/AhR Signaling at the Junction of Inflammation and Cardiovascular Diseases. Int J Mol Sci 2024; 25:6933. [PMID: 39000041 PMCID: PMC11240928 DOI: 10.3390/ijms25136933] [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: 05/20/2024] [Revised: 06/14/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
Persistent systemic chronic inflammatory conditions are linked with many pathologies, including cardiovascular diseases (CVDs), a leading cause of death across the globe. Among various risk factors, one of the new possible contributors to CVDs is the metabolism of essential amino acid tryptophan. Proinflammatory signals promote tryptophan metabolism via the kynurenine (KYN) pathway (KP), thereby resulting in the biosynthesis of several immunomodulatory metabolites whose biological effects are associated with the development of symptoms and progression of various inflammatory diseases. Some participants in the KP are agonists of aryl hydrocarbon receptor (AhR), a central player in a signaling pathway that, along with a regulatory influence on the metabolism of environmental xenobiotics, performs a key immunomodulatory function by triggering various cellular mechanisms with the participation of endogenous ligands to alleviate inflammation. An AhR ligand with moderate affinity is the central metabolite of the KP: KYN; one of the subsequent metabolites of KYN-kynurenic acid (KYNA)-is a more potent ligand of AhR. Understanding the role of AhR pathway-related metabolites of the KP that regulate inflammatory factors in cells of the cardiovascular system is interesting and important for achieving effective treatment of CVDs. The purpose of this review was to summarize the results of studies about the participation of the KP metabolite-KYNA-and of the AhR signaling pathway in the regulation of inflammation in pathological conditions of the heart and blood vessels and about the possible interaction of KYNA with AhR signaling in some CVDs.
Collapse
Affiliation(s)
| | - Maria L. Perepechaeva
- Institute of Molecular Biology and Biophysics, Federal Research Center of Fundamental and Translational Medicine, Timakova Str. 2, Novosibirsk 630060, Russia;
| |
Collapse
|
12
|
Wei T, Mueed A, Luo T, Sun Y, Zhang B, Zheng L, Deng Z, Li J. 1,3-Dioleoyl-2-palmitoyl-glycerol and 1-oleoyl-2-palmitoyl-3-linoleoyl-glycerol: Structure-function relationship, triacylglycerols preparation, nutrition value. Food Chem 2024; 443:138560. [PMID: 38295563 DOI: 10.1016/j.foodchem.2024.138560] [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: 09/21/2023] [Revised: 12/31/2023] [Accepted: 01/21/2024] [Indexed: 02/02/2024]
Abstract
Based on multivariate statistics, this review compared major triacylglycerols (TAGs) in animal milk and human milk fat from China and other countries. Human milk fat differs from animal milk fat in that it has longer acyl chains and higher concentrations of 1,3-dioleoyl-2-palmitoyl-glycerol (O-P-O) and 1-oleoyl-2-palmitoyl-3-linoleoylglycerol (O-P-L). O-P-L is a significant and distinct TAG in human milk fat, particularly in China. 1-oleoyl-2-palmitoyl-3-linoleoylglycerol (OPL) is human milk's major triglyceride molecule of O-P-L, accounting for more than 70%. As a result, OPL has piqued the interest of Chinese academics. The synthesis process and nutritional outcomes of OPL have been studied, including changes in gut microbiota, serum lipid composition, improved fatty acid and calcium absorption, and increased total bile acid levels. However, current OPL research is limited. Therefore, this review discussed enzymatic preparation of 1,3-dioleoyl-2-palmitoyl-glycerol (OPO) and OPL and their nutritional and physiological activity to direct future research direction for sn-2 palmitate and OPL.
Collapse
Affiliation(s)
- Teng Wei
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi 330047, China.
| | - Abdul Mueed
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi 330047, China
| | - Ting Luo
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi 330047, China; International Institute of Food Innovation, Nanchang University, Nanchang 330031, China.
| | - Yong Sun
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi 330047, China; International Institute of Food Innovation, Nanchang University, Nanchang 330031, China.
| | - Bing Zhang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi 330047, China; International Institute of Food Innovation, Nanchang University, Nanchang 330031, China.
| | - Liufeng Zheng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi 330047, China; International Institute of Food Innovation, Nanchang University, Nanchang 330031, China.
| | - Zeyuan Deng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi 330047, China; International Institute of Food Innovation, Nanchang University, Nanchang 330031, China; National Center of Technology Innovation for Dairy, Hohhot, Inner Mongolia 010110, China.
| | - Jing Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi 330047, China; International Institute of Food Innovation, Nanchang University, Nanchang 330031, China; National Center of Technology Innovation for Dairy, Hohhot, Inner Mongolia 010110, China.
| |
Collapse
|
13
|
Venketasubramanian N, Yeo TT, Chen CLH. Translational Medicine in Acute Ischemic Stroke and Traumatic Brain Injury-NeuroAiD Trials, from Traditional Beliefs to Evidence-Based Therapy. Biomolecules 2024; 14:680. [PMID: 38927083 PMCID: PMC11202287 DOI: 10.3390/biom14060680] [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: 04/01/2024] [Revised: 06/08/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024] Open
Abstract
Acute ischemic stroke (AIS) and traumatic brain injury (TBI) are two severe neurological events, both being major causes of death and prolonged impairment. Their incidence continues to rise due to the global increase in the number of people at risk, representing a significant burden on those remaining impaired, their families, and society. These molecular and cellular mechanisms of both stroke and TBI present similarities that can be targeted by treatments with a multimodal mode of action, such as traditional Chinese medicine. Therefore, we performed a detailed review of the preclinical and clinical development of MLC901 (NeuroAiDTMII), a natural multi-herbal formulation targeting several biological pathways at the origin of the clinical deficits. The endogenous neurobiological processes of self-repair initiated by the brain in response to the onset of brain injury are often insufficient to achieve complete recovery of impaired functions. This review of MLC901 and its parent formulation MLC601 confirms that it amplifies the natural self-repair process of brain tissue after AIS or TBI. Following AIS and TBI where "time is brain", many patients enter the post-acute phase with their functions still impaired, a period when "the brain needs time to repair itself". The treatment goal must be to accelerate recovery as much as possible. MLC901/601 demonstrated a significant reduction by 18 months of recovery time compared to a placebo, indicating strong potential for facilitating the improvement of health outcomes and the more efficient use of healthcare resources.
Collapse
Affiliation(s)
| | - Tseng Tsai Yeo
- Division of Neurosurgery, Department of Surgery, National University Hospital, 5 Lower Kent Ridge Road, Singapore 119074, Singapore;
| | - Christopher Li Hsian Chen
- Memory Aging and Cognition Centre, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Blk MD3, 16 Medical Drive, #04-01, Singapore 117600, Singapore;
| |
Collapse
|
14
|
Yang Y, Liu X, Liu X, Xie C, Shi J. The role of the kynurenine pathway in cardiovascular disease. Front Cardiovasc Med 2024; 11:1406856. [PMID: 38883986 PMCID: PMC11176437 DOI: 10.3389/fcvm.2024.1406856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/23/2024] [Indexed: 06/18/2024] Open
Abstract
The kynurenine pathway (KP) serves as the primary route for tryptophan metabolism in most mammalian organisms, with its downstream metabolites actively involved in various physiological and pathological processes. Indoleamine 2,3-dioxygenase (IDO) and tryptophan 2,3-dioxygenase (TDO) serve as the initial and pivotal enzymes of the KP, with IDO playing important and intricate roles in cardiovascular diseases. Multiple metabolites of KP have been observed to exhibit elevated concentrations in plasma across various cardiovascular diseases, such as atherosclerosis, hypertension, and acute myocardial infarction. Multiple studies have indicated that kynurenine (KYN) may serve as a potential biomarker for several adverse cardiovascular events. Furthermore, Kynurenine and its downstream metabolites have complex roles in inflammation, exhibiting both inhibitory and stimulatory effects on inflammatory responses under different conditions. In atherosclerosis, upregulation of IDO stimulates KYN production, mediating aromatic hydrocarbon receptor (AhR)-induced exacerbation of vascular inflammation and promotion of foam cell formation. Conversely, in arterial calcification, this mediation alleviates osteogenic differentiation of vascular smooth muscle cells. Additionally, in cardiac remodeling, KYN-mediated AhR activation exacerbates pathological left ventricular hypertrophy and fibrosis. Interventions targeting components of the KP, such as IDO inhibitors, 3-hydroxyanthranilic acid, and anthranilic acid, demonstrate cardiovascular protective effects. This review outlines the mechanistic roles of KP in coronary atherosclerosis, arterial calcification, and myocardial diseases, highlighting the potential diagnostic, prognostic, and therapeutic value of KP in cardiovascular diseases, thus providing novel insights for the development and application of related drugs in future research.
Collapse
Affiliation(s)
- Yuehang Yang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing Liu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinyi Liu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chiyang Xie
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiawei Shi
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
15
|
Osuch B, Misztal T, Pałatyńska K, Tomaszewska-Zaremba D. Implications of Kynurenine Pathway Metabolism for the Immune System, Hypothalamic-Pituitary-Adrenal Axis, and Neurotransmission in Alcohol Use Disorder. Int J Mol Sci 2024; 25:4845. [PMID: 38732064 PMCID: PMC11084367 DOI: 10.3390/ijms25094845] [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: 03/14/2024] [Revised: 04/21/2024] [Accepted: 04/28/2024] [Indexed: 05/13/2024] Open
Abstract
In recent years, there has been a marked increase in interest in the role of the kynurenine pathway (KP) in mechanisms associated with addictive behavior. Numerous reports implicate KP metabolism in influencing the immune system, hypothalamic-pituitary-adrenal (HPA) axis, and neurotransmission, which underlie the behavioral patterns characteristic of addiction. An in-depth analysis of the results of these new studies highlights interesting patterns of relationships, and approaching alcohol use disorder (AUD) from a broader neuroendocrine-immune system perspective may be crucial to better understanding this complex phenomenon. In this review, we provide an up-to-date summary of information indicating the relationship between AUD and the KP, both in terms of changes in the activity of this pathway and modulation of this pathway as a possible pharmacological approach for the treatment of AUD.
Collapse
Affiliation(s)
- Bartosz Osuch
- Department of Animal Physiology, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3, 05-110 Jabłonna, Poland; (T.M.); (K.P.); (D.T.-Z.)
| | | | | | | |
Collapse
|
16
|
Chen J, Zou J, Huang P, Gao X, Lun J, Li Y, Gong Z, Cao H. KYNA Ameliorates Glutamate Toxicity of HAND by Enhancing Glutamate Uptake in A2 Astrocytes. Int J Mol Sci 2024; 25:4286. [PMID: 38673879 PMCID: PMC11050540 DOI: 10.3390/ijms25084286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/02/2024] [Accepted: 02/09/2024] [Indexed: 04/28/2024] Open
Abstract
Reactive astrocytes are key players in HIV-associated neurocognitive disorders (HAND), and different types of reactive astrocytes play opposing roles in the neuropathologic progression of HAND. A recent study by our group found that gp120 mediates A1 astrocytes (neurotoxicity), which secrete proinflammatory factors and promote HAND disease progression. Here, by comparing the expression of A2 astrocyte (neuroprotective) markers in the brains of gp120 tgm mice and gp120+/α7nAChR-/- mice, we found that inhibition of alpha 7 nicotinic acetylcholine receptor (α7nAChR) promotes A2 astrocyte generation. Notably, kynurenine acid (KYNA) is an antagonist of α7nAChR, and is able to promote the formation of A2 astrocytes, the secretion of neurotrophic factors, and the enhancement of glutamate uptake through blocking the activation of α7nAChR/NF-κB signaling. In addition, learning, memory and mood disorders were significantly improved in gp120 tgm mice by intraperitoneal injection of kynurenine (KYN) and probenecid (PROB). Meanwhile, the number of A2 astrocytes in the mouse brain was significantly increased and glutamate toxicity was reduced. Taken together, KYNA was able to promote A2 astrocyte production and neurotrophic factor secretion, reduce glutamate toxicity, and ameliorate gp120-induced neuropathological deficits. These findings contribute to our understanding of the role that reactive astrocytes play in the development of HAND pathology and provide new evidence for the treatment of HAND via the tryptophan pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Hong Cao
- Department of Microbiology, School of Public Health, Southern Medical University (Guangdong Provincial Key Laboratory of Tropical Disease Research), Guangzhou 510515, China; (J.C.)
| |
Collapse
|
17
|
Ye J, Fan H, Shi R, Song G, Wu X, Wang D, Xia B, Zhao Z, Zhao B, Liu X, Wang Y, Dai X. Dietary lipoic acid alleviates autism-like behavior induced by acrylamide in adolescent mice: the potential involvement of the gut-brain axis. Food Funct 2024; 15:3395-3410. [PMID: 38465655 DOI: 10.1039/d3fo05078e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Consuming fried foods has been associated with an increased susceptibility to mental health disorders. Nevertheless, the impact of alpha-lipoic acid (α-LA, LA) on fried food-induced autism-like behavior remains unclear. This study aimed to explore how LA affects autism-related behavior and cognitive deficits caused by acrylamide in mice, a representative food hazard found in fried foods. This improvement was accomplished by enhanced synaptic plasticity, increased neurotrophin expression, elevated calcium-binding protein D28k, and restored serotonin. Additionally, LA substantially influenced the abundance of bacteria linked to autism and depression, simultaneously boosted short-chain fatty acid (SCFA) levels in fecal samples, and induced changes in serum amino acid concentrations. In summary, these findings suggested that exposure to acrylamide in adolescent mice could induce the development of social disorders in adulthood. LA showed promise as a nutritional intervention strategy to tackle emotional disorders during adolescence.
Collapse
Affiliation(s)
- Jin Ye
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China.
| | - Hua Fan
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China.
| | - Renjie Shi
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China.
| | - Ge Song
- BGI Institute of Applied Agriculture, BGI-Shenzhen, Shenzhen, China.
| | - Xiaoning Wu
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China.
| | - Danna Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China.
| | - Bing Xia
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China.
| | - Zhenting Zhao
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China.
| | - Beita Zhao
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China.
| | - Xuebo Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China.
| | - Yutang Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China.
| | - Xiaoshuang Dai
- BGI Institute of Applied Agriculture, BGI-Shenzhen, Shenzhen, China.
| |
Collapse
|
18
|
Zhang X, Li Y, Zhu K, Li C, Zhao Q, Gu F, Xu F, Chu Z. Microbiome-Metabolomic Analysis Revealed the Immunoprotective Effects of the Extract of Vanilla planifolia Andrew (EVPA) on Immunosuppressed Mice. Foods 2024; 13:701. [PMID: 38472814 PMCID: PMC10930650 DOI: 10.3390/foods13050701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 02/16/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
This study investigated the immunoprotective effects of the extract of Vanilla planifolia Andrew (EVPA) on cyclophosphamide (Cy)-induced immunosuppression in mice. The results show that EVPA administration significantly alleviated the immune damage induced by Cy, as evidenced by an improved body weight, organ index, and colonic injury. A further analysis of microbial diversity revealed that the EVPA primarily increased the abundance of the beneficial bacteria Verrucomicrobiota, Lactobacillaceae, and Lactobacillus while decreasing Akkermansiaceae, Akkermansia, Romboutsia, and Lactococcus, thereby ameliorating the microbial dysbiosis caused by Cy. A metabolomic analysis revealed significant alterations in the microbial metabolite levels after EVPA treatment, including urobilinogen, formamidopyrimidine nucleoside triphosphate, Cer (d18:1/18:0), pantetheine, and LysoPC (15:0/0:0). These altered metabolites are associated with pathways related to sphingolipid metabolism, carbapenem biosynthesis, pantothenate and CoA biosynthesis, glycerophospholipid metabolism, and porphyrin metabolism. Furthermore, significant correlations were observed between certain microbial groups and the differential metabolites. These findings provide new insights into the immunomodulatory effects of EVPA on the intestinal microbiota and metabolism, laying the foundation for more extensive utilization.
Collapse
Affiliation(s)
- Xin Zhang
- Spice and Beverage Research Institute, Chinese Academy of Tropical Agricultural Sciences, Wanning 571533, China; (X.Z.); (Y.L.); (K.Z.); (Q.Z.); (F.G.); (Z.C.)
- School of Food Science and Engineering, Hainan University, Haikou 570228, China;
| | - Yunlong Li
- Spice and Beverage Research Institute, Chinese Academy of Tropical Agricultural Sciences, Wanning 571533, China; (X.Z.); (Y.L.); (K.Z.); (Q.Z.); (F.G.); (Z.C.)
- School of Food Science and Engineering, Hainan University, Haikou 570228, China;
| | - Kexue Zhu
- Spice and Beverage Research Institute, Chinese Academy of Tropical Agricultural Sciences, Wanning 571533, China; (X.Z.); (Y.L.); (K.Z.); (Q.Z.); (F.G.); (Z.C.)
- National Center of Important Tropical Crops Engineering and Technology Research, Wanning 571533, China
- Key Laboratory of Processing Suitability and Quality Control of the Special Tropical Crops of Hainan Province, Wanning 571533, China
| | - Chuan Li
- School of Food Science and Engineering, Hainan University, Haikou 570228, China;
| | - Qingyun Zhao
- Spice and Beverage Research Institute, Chinese Academy of Tropical Agricultural Sciences, Wanning 571533, China; (X.Z.); (Y.L.); (K.Z.); (Q.Z.); (F.G.); (Z.C.)
- Sanya Research Institute of Chinese Academy of Tropical Agricultural Sciences, Sanya 572000, China
| | - Fenglin Gu
- Spice and Beverage Research Institute, Chinese Academy of Tropical Agricultural Sciences, Wanning 571533, China; (X.Z.); (Y.L.); (K.Z.); (Q.Z.); (F.G.); (Z.C.)
- National Center of Important Tropical Crops Engineering and Technology Research, Wanning 571533, China
- Key Laboratory of Processing Suitability and Quality Control of the Special Tropical Crops of Hainan Province, Wanning 571533, China
- Sanya Research Institute of Chinese Academy of Tropical Agricultural Sciences, Sanya 572000, China
| | - Fei Xu
- Spice and Beverage Research Institute, Chinese Academy of Tropical Agricultural Sciences, Wanning 571533, China; (X.Z.); (Y.L.); (K.Z.); (Q.Z.); (F.G.); (Z.C.)
- National Center of Important Tropical Crops Engineering and Technology Research, Wanning 571533, China
- Key Laboratory of Processing Suitability and Quality Control of the Special Tropical Crops of Hainan Province, Wanning 571533, China
| | - Zhong Chu
- Spice and Beverage Research Institute, Chinese Academy of Tropical Agricultural Sciences, Wanning 571533, China; (X.Z.); (Y.L.); (K.Z.); (Q.Z.); (F.G.); (Z.C.)
- National Center of Important Tropical Crops Engineering and Technology Research, Wanning 571533, China
- Key Laboratory of Processing Suitability and Quality Control of the Special Tropical Crops of Hainan Province, Wanning 571533, China
| |
Collapse
|
19
|
Zhao Y, Jia H, Deng H, Ge C, Xing W, Yu H, Li J. Integrated microbiota and multi-omics analysis reveal the differential responses of earthworm to conventional and biodegradable microplastics in soil under biogas slurry irrigation. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 907:168191. [PMID: 37907108 DOI: 10.1016/j.scitotenv.2023.168191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 10/27/2023] [Accepted: 10/27/2023] [Indexed: 11/02/2023]
Abstract
As one of the promising alternatives of conventional plastic mulching film (C-PMF), biodegradable plastic mulching films (B-PMF) were employed in agronomy production to alleviate the environmental burden of C-PMF. However, information regarding the potential toxicity effects of biodegradable microplastics (MPs) in soil still in scarcity, and the available findings were found to be controversial. Additionally, little is known about the molecular toxicity effects of conventional and biodegradable MPs on terrestrial organisms. Thus, 5 % (w/w) biodegradable (polylactic acid, PLA) and conventional (polyvinylchloride, PVC; low-density polyvinylchloride, LDPE) MPs were employed to assess the toxicity effects on Eisenia fetida in agricultural soil with biogas slurry irrigation. In the present study, transcriptomic, metabolomic profiles and individual indexes were selected to reveal the toxicity mechanisms from molecular level to the individual response. Furthermore, dysbiosis of bacterial community in gut was also investigated for obtaining comprehensive knowledge on the MPs toxicity. At the end of the exposure, the number of survival earthworms after MPs exposure was significantly reduced. Compared with the initial body weight, PLA and LDPE increased the biomass of earthworms after MPs exposure, while no significant influence on the biomass was observed in PVC treatment. Microbacterium, Klebsiella and Chryseobacterium were significantly enriched in earthworm gut after PLA, PVC and LDPE exposure, respectively (p < 0.05). Transcriptomic and metabolomic analysis revealed that PLA exposure induced neurotransmission disorder and high energetic expenditure in earthworms. However, PVC and LDPE inhibited the nutrient absorption efficiency and activated the innate immunity responses of earthworms. The PLS-SEM results showed that the effects of MPs were dominated by the polymer types, and hence, significantly and directly influence the gut bacterial community of earthworms. This study provides a better understanding of the similarities and discrepancies in toxicity effects of biodegradable and conventional MPs from the perspectives of individual, gut bacterial community, transcriptome and metabolome.
Collapse
Affiliation(s)
- Yuanyuan Zhao
- Key Laboratory of Agro-Forestry Environmental Processes and Ecological Regulation of Hainan Province, Hainan University, Haikou 570228, China; Key Laboratory of Environmental Toxicology, Hainan University, Haikou 570228, China; Center for Eco-Environment Restoration Engineering of Hainan Province, Hainan University, Haikou 570228, China; College of Ecology and Environment, Hainan University, Renmin Road, Haikou 570228, China
| | - Huiting Jia
- Key Laboratory of Agro-Forestry Environmental Processes and Ecological Regulation of Hainan Province, Hainan University, Haikou 570228, China; Key Laboratory of Environmental Toxicology, Hainan University, Haikou 570228, China; Center for Eco-Environment Restoration Engineering of Hainan Province, Hainan University, Haikou 570228, China; College of Ecology and Environment, Hainan University, Renmin Road, Haikou 570228, China
| | - Hui Deng
- Key Laboratory of Agro-Forestry Environmental Processes and Ecological Regulation of Hainan Province, Hainan University, Haikou 570228, China; Key Laboratory of Environmental Toxicology, Hainan University, Haikou 570228, China; Center for Eco-Environment Restoration Engineering of Hainan Province, Hainan University, Haikou 570228, China; College of Ecology and Environment, Hainan University, Renmin Road, Haikou 570228, China
| | - Chengjun Ge
- Key Laboratory of Agro-Forestry Environmental Processes and Ecological Regulation of Hainan Province, Hainan University, Haikou 570228, China; Key Laboratory of Environmental Toxicology, Hainan University, Haikou 570228, China; Center for Eco-Environment Restoration Engineering of Hainan Province, Hainan University, Haikou 570228, China; College of Ecology and Environment, Hainan University, Renmin Road, Haikou 570228, China.
| | - Wenzhe Xing
- Key Laboratory of Agro-Forestry Environmental Processes and Ecological Regulation of Hainan Province, Hainan University, Haikou 570228, China; Key Laboratory of Environmental Toxicology, Hainan University, Haikou 570228, China; Center for Eco-Environment Restoration Engineering of Hainan Province, Hainan University, Haikou 570228, China; College of Ecology and Environment, Hainan University, Renmin Road, Haikou 570228, China
| | - Huamei Yu
- Key Laboratory of Agro-Forestry Environmental Processes and Ecological Regulation of Hainan Province, Hainan University, Haikou 570228, China; Key Laboratory of Environmental Toxicology, Hainan University, Haikou 570228, China; Center for Eco-Environment Restoration Engineering of Hainan Province, Hainan University, Haikou 570228, China; College of Ecology and Environment, Hainan University, Renmin Road, Haikou 570228, China.
| | - Jiatong Li
- Key Laboratory of Agro-Forestry Environmental Processes and Ecological Regulation of Hainan Province, Hainan University, Haikou 570228, China; Key Laboratory of Environmental Toxicology, Hainan University, Haikou 570228, China; Center for Eco-Environment Restoration Engineering of Hainan Province, Hainan University, Haikou 570228, China; College of Ecology and Environment, Hainan University, Renmin Road, Haikou 570228, China
| |
Collapse
|
20
|
Jóźwiak-Bębenista M, Sokołowska P, Wiktorowska-Owczarek A, Kowalczyk E, Sienkiewicz M. Ketamine - A New Antidepressant Drug with Anti-Inflammatory Properties. J Pharmacol Exp Ther 2024; 388:134-144. [PMID: 37977808 DOI: 10.1124/jpet.123.001823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/04/2023] [Accepted: 10/04/2023] [Indexed: 11/19/2023] Open
Abstract
Ketamine is a new, potent and rapid-acting antidepressant approved for therapy of treatment-resistant depression, which has a different mechanism of action than currently-available antidepressant therapies. It owes its uniquely potent antidepressant properties to a complex mechanism of action, which currently remains unclear. However, it is thought that it acts by modulating the functioning of the glutamatergic system, which plays an important role in the process of neuroplasticity associated with depression. However, preclinical and clinical studies have also found ketamine to reduce inflammation, either directly or indirectly (by activating neuroprotective branches of the kynurenine pathway), among patients exhibiting higher levels of inflammation. Inflammation and immune system activation are believed to play key roles in the development and course of depression. Therefore, the present work examines the role of the antidepressant effect of ketamine and its anti-inflammatory properties in the treatment of depression. SIGNIFICANCE STATEMENT: The present work examines the relationship between the antidepressant effect of ketamine and its anti-inflammatory properties, and the resulting benefits in treatment-resistant depression (TRD). The antidepressant mechanism of ketamine remains unclear, and there is an urgent need to develop new therapeutic strategies for treatment of depression, particularly TRD.
Collapse
Affiliation(s)
- Marta Jóźwiak-Bębenista
- Department of Pharmacology and Toxicology (M.J.-B., P.S., A.W.-O., E.K.) and Department of Pharmaceutical Microbiology and Microbiological Diagnostics (M.S.), Medical University of Lodz, Lodz, Poland
| | - Paulina Sokołowska
- Department of Pharmacology and Toxicology (M.J.-B., P.S., A.W.-O., E.K.) and Department of Pharmaceutical Microbiology and Microbiological Diagnostics (M.S.), Medical University of Lodz, Lodz, Poland
| | - Anna Wiktorowska-Owczarek
- Department of Pharmacology and Toxicology (M.J.-B., P.S., A.W.-O., E.K.) and Department of Pharmaceutical Microbiology and Microbiological Diagnostics (M.S.), Medical University of Lodz, Lodz, Poland
| | - Edward Kowalczyk
- Department of Pharmacology and Toxicology (M.J.-B., P.S., A.W.-O., E.K.) and Department of Pharmaceutical Microbiology and Microbiological Diagnostics (M.S.), Medical University of Lodz, Lodz, Poland
| | - Monika Sienkiewicz
- Department of Pharmacology and Toxicology (M.J.-B., P.S., A.W.-O., E.K.) and Department of Pharmaceutical Microbiology and Microbiological Diagnostics (M.S.), Medical University of Lodz, Lodz, Poland
| |
Collapse
|
21
|
Heng B, Pires AS, Chow S, Krishnamurthy S, Bonnell B, Bustamante S, Guillemin GJ. Stability Studies of Kynurenine Pathway Metabolites in Blood Components Define Optimal Blood Processing Conditions. Int J Tryptophan Res 2023; 16:11786469231213521. [PMID: 38106464 PMCID: PMC10725091 DOI: 10.1177/11786469231213521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/25/2023] [Indexed: 12/19/2023] Open
Abstract
The kynurenine pathway (KP) is the main pathway of tryptophan (TRP) metabolism that generates energy for multiple cellular processes. The activity of this pathway has been shown to be dysregulated in multiple human diseases. The resultant modulation of metabolites has been suggested to comprise biomarkers to track disease progression or could identify new therapeutic targets. While metabolite changes can be measured readily in blood, there is limited knowledge on the effect of blood matrices and sample processing time may have on the stability of KP metabolites. Understanding the stability of KP metabolites in blood is integral to obtaining accurate KP data to correlate with clinical pathology. Hence, the aim of this study was to assess the concentration of KP metabolites in matched whole blood, plasma and serum. The impact of pre-analytical sample processing time in the various blood matrices was also analysed. Serum and plasma had the higher concentration of KP metabolites compared to whole blood. Furthermore, concentrations of KP metabolites declined when the collected blood was processed after 24 hours storage at 4°C. Our study shows that that type of blood matrix and the time to processing have an impact on the stability of the KP metabolites. Serum or plasma are the preferred choice of matrix and the isolation of these matrices from whole blood is best performed immediately after collection for optimal analytical KP data.
Collapse
Affiliation(s)
- Benjamin Heng
- Macquarie Medical School, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Ananda Staats Pires
- Macquarie Medical School, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Sharron Chow
- Macquarie Medical School, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Shivani Krishnamurthy
- Macquarie Medical School, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Brooke Bonnell
- Macquarie Medical School, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Sonia Bustamante
- Bioanalytical Mass Spectrometry Facility, University of New South Wales, Sydney, Australia
| | - Gilles J Guillemin
- Macquarie Medical School, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
22
|
Gao Y, Guo X, Zhou Y, Du J, Lu C, Zhang L, Sun S, Wang S, Li Y. Kynurenic acid inhibits macrophage pyroptosis by suppressing ROS production via activation of the NRF2 pathway. Mol Med Rep 2023; 28:211. [PMID: 37772394 PMCID: PMC10552067 DOI: 10.3892/mmr.2023.13098] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 09/04/2023] [Indexed: 09/30/2023] Open
Abstract
Macrophage pyroptosis and related inflammatory responses play an important role in periodontitis. Kynurenic acid (KA) is hypothesized to have anti‑inflammatory potential, but whether KA can inhibit macrophage pyroptosis and the underlying mechanisms remain unclear. Lipopolysaccharide (LPS) was used to induce pyroptosis in THP‑1‑derived macrophages. KA or ML385 was used to pretreat macrophages, after which, cell viability, NOD‑like receptor protein 3 (NLRP3) inflammasome‑related protein expression, oxidative stress levels and nuclear factor erythroid 2‑related factor 2 (NRF2) expression were measured. The results showed that KA improved the LPS‑induced decrease in macrophage viability and lactate dehydrogenase release. KA prevented THP‑1 macrophage pyroptosis induced by LPS by reducing the expression of NLRP3, Gasdermin‑D, and Caspase1, and decreased the expression of inflammatory factors. KA suppressed NLRP3 inflammasome activation by inhibiting ROS overproduction and increasing Heme Oxygenase 1 and glutathione levels. Moreover, KA promoted NRF2 translocation from the cytoplasm to the nucleus. In addition, the anti‑pyroptotic and antioxidant effects of KA were reversed by ML385 inhibition of NRF2. In the present study, it was found that KA significantly suppressed macrophage pyroptosis induced by LPS. It was further demonstrated that the anti‑pyroptotic effects of KA were mediated by activation of the NRF2 pathway.
Collapse
Affiliation(s)
- Yuwei Gao
- Department of Pediatric Dentistry, Jiamusi University Affiliated Stomatological Hospital, Jiamusi, Heilongjiang 154000, P.R. China
| | - Xiaohui Guo
- Department of Pathology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Yunpeng Zhou
- Department of Urology, Tianjin Baodi Hospital, Baodi Clinical College of Tianjin Medical University, Tianjin 300203, P.R. China
| | - Jie Du
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Chengbo Lu
- Department of Cardiology, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154002, P.R. China
| | - Lei Zhang
- Department of Cardiology, People's Hospital of Taihe County, Taihe, Anhui 236600, P.R. China
| | - Siyuan Sun
- Department of Cardiology, People's Hospital of Taihe County, Taihe, Anhui 236600, P.R. China
| | - Shengfang Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Yang Li
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
23
|
Białczyk A, Wełniak A, Kamińska B, Czajkowski R. Oxidative Stress and Potential Antioxidant Therapies in Vitiligo: A Narrative Review. Mol Diagn Ther 2023; 27:723-739. [PMID: 37737953 PMCID: PMC10590312 DOI: 10.1007/s40291-023-00672-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/17/2023] [Indexed: 09/23/2023]
Abstract
Vitiligo is a chronic skin disorder characterised by the loss of melanocytes and subsequent skin depigmentation. Although many theories have been proposed in the literature, none alone explains the pathogenesis of vitiligo. Oxidative stress has been identified as a potential factor in the pathogenesis of vitiligo. A growing body of evidence suggests that antioxidant therapies may offer a promising approach to managing this condition. This review summarises the potential mechanisms of oxidative stress and the types of melanocyte death in vitiligo. We also provide a brief overview of the most commonly studied antioxidants. Melanocytes in vitiligo are thought to be damaged by an accumulation of reactive oxygen species to destroy the structural and functional integrity of their DNA, lipids, and proteins. Various causes, including exogenous and endogenous stress factors, an imbalance between prooxidants and antioxidants, disruption of antioxidant pathways, and gene polymorphisms, lead to the overproduction of reactive oxygen species. Although necroptosis, pyroptosis, ferroptosis, and oxeiptosis are newer types of cell death that may contribute to the pathophysiology of vitiligo, apoptosis remains the most studied cell death mechanism in vitiligo. According to studies, vitamin E helps to treat lipid peroxidation of the skin caused by psoralen ultra-violet A treatment. In addition, Polypodium leucotomos increased the efficacy of psoralen ultra-violet A or narrow-band ultraviolet B therapy. Our review provides valuable insights into the potential role of oxidative stress in pathogenesis and antioxidant-based supporting therapies in treating vitiligo, offering a promising avenue for further research and the development of effective treatment strategies.
Collapse
Affiliation(s)
- Aleksandra Białczyk
- Students' Scientific Club of Dermatology, Department of Dermatology and Venerology, Ludwik Rydygier Collegium Medicum, Nicolaus Copernicus University, 9 Skłodowskiej-Curie Street, 85-094, Bydgoszcz, Poland.
| | - Adam Wełniak
- Students' Scientific Club of Dermatology, Department of Dermatology and Venerology, Ludwik Rydygier Collegium Medicum, Nicolaus Copernicus University, 9 Skłodowskiej-Curie Street, 85-094, Bydgoszcz, Poland
| | - Barbara Kamińska
- Students' Scientific Club of Dermatology, Department of Dermatology and Venerology, Ludwik Rydygier Collegium Medicum, Nicolaus Copernicus University, 9 Skłodowskiej-Curie Street, 85-094, Bydgoszcz, Poland
| | - Rafał Czajkowski
- Department of Dermatology and Venerology, Ludwik Rydygier Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| |
Collapse
|
24
|
Song C, Kang T, Gao K, Shi X, Zhang M, Zhao L, Zhou L, Guo J. Preparation for mice spaceflight: Indications for training C57BL/6J mice to adapt to microgravity effect with three-dimensional clinostat on the ground. Heliyon 2023; 9:e19355. [PMID: 37662714 PMCID: PMC10472007 DOI: 10.1016/j.heliyon.2023.e19355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 08/09/2023] [Accepted: 08/20/2023] [Indexed: 09/05/2023] Open
Abstract
Like astronauts, animals need to undergo training and screening before entering space. At present, pre-launch training for mice mainly focuses on adaptation to habitat system. Training for the weightless environment of space in mice has not received much attention. Three-dimensional (3D) clinostat is a method to simulate the effects of microgravity on Earth. However, few studies have used a 3D clinostat apparatus to simulate the effects of microgravity on animal models. Therefore, we conducted a study to evaluate the feasibility and effects of long-term treatment with three-dimensional clinostat in C57BL/6 J mice. Thirty 8-week-old male C57BL/6 J mice were randomly assigned to three groups: mice in individually ventilated cages (MC group, n = 6), mice in survival boxes (SB group, n = 12), and mice in survival boxes receiving 3D clinostat treatment (CS group, n = 12). The mice showed good tolerance after 12 weeks of alternate day training. To evaluate the biological effects of simulated microgravity, the changes in serum metabolites were monitored using untargeted metabolomics, whereas bone loss was assessed using microcomputed tomography of the left femur. Compared with the metabolome of the SB group, the metabolome of the CS group showed significant differences during the first three weeks and the last three weeks. The KEGG pathways in the late stages were mainly related to the nervous system, indicating the influence of long-term microgravity on the central nervous system. Besides, a marked reduction in the trabecular number (P < 0.05) and an increasing trend of trabecular spacing (P < 0.1) were observed to occur in a time-dependent manner in the CS group compared with the SB group. These results showed that mice tolerated well in a 3D clinostat and may provide a new strategy in pre-launch training for mice and conducting relevant ground-based modeling experiments.
Collapse
Affiliation(s)
- Chenchen Song
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Taisheng Kang
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Kai Gao
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Xudong Shi
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Meng Zhang
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Lianlian Zhao
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Li Zhou
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Jianguo Guo
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| |
Collapse
|
25
|
De la Fuente Muñoz M, Román-Carmena M, Amor S, García-Villalón ÁL, Espinel AE, González-Hedström D, Granado García M. Effects of Supplementation with the Standardized Extract of Saffron (affron ®) on the Kynurenine Pathway and Melatonin Synthesis in Rats. Antioxidants (Basel) 2023; 12:1619. [PMID: 37627614 PMCID: PMC10451224 DOI: 10.3390/antiox12081619] [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: 07/17/2023] [Revised: 08/08/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
Melatonin is a hormone that regulates sleep-wake cycles and is mainly synthesized in the pineal gland from tryptophan after its conversion into serotonin. Under normal conditions, less than 5% of tryptophan is reserved for the synthesis of serotonin and melatonin. The remaining 95% is metabolized in the liver through the kynurenine pathway. Increased levels of proinflammatory cytokines and cortisol increase the metabolism of tryptophan through the kynurenine pathway and reduce its availability for the synthesis of melatonin and serotonin, which may cause alterations in mood and sleep. The standardized saffron extract (affron®) has shown beneficial effects on mood and sleep disorders in humans, but the underlying mechanisms are not well understood. Thus, the aim of this work was to study the effects of affron® supplementation on the kynurenine pathway and the synthesis of melatonin in rats. For this purpose, adult male Wistar rats were supplemented for 7 days with 150 mg/kg of affron® or vehicle (2 mL/kg water) administered by gavage one hour before sleep. Affron® supplementation reduced body weight gain and increased the circulating levels of melatonin, testosterone, and c-HDL. Moreover, animals supplemented with affron® showed decreased serum levels of kynurenine, ET-1, and c-LDL. In the pineal gland, affron® reduced Il-6 expression and increased the expression of Aanat, the key enzyme for melatonin synthesis. In the liver, affron® administration decreased the mRNA levels of the enzymes of the kynurenine pathway Ido-2, Tod-2, and Aadat, as well as the gene expression of Il-1β and Tnf-α. Finally, rats treated with affron® showed increased mRNA levels of the antioxidant enzymes Ho-1, Sod-1, Gsr, and Gpx-3, both in the liver and in the pineal gland. In conclusion, affron® supplementation reduces kynurenine levels and promotes melatonin synthesis in rats, possibly through its antioxidant and anti-inflammatory effects, making this extract a possible alternative for the treatment and/or prevention of mood and sleep disorders.
Collapse
Affiliation(s)
- Mario De la Fuente Muñoz
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (M.D.l.F.M.); (M.R.-C.); (S.A.); (Á.L.G.-V.)
| | - Marta Román-Carmena
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (M.D.l.F.M.); (M.R.-C.); (S.A.); (Á.L.G.-V.)
| | - Sara Amor
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (M.D.l.F.M.); (M.R.-C.); (S.A.); (Á.L.G.-V.)
| | - Ángel Luís García-Villalón
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (M.D.l.F.M.); (M.R.-C.); (S.A.); (Á.L.G.-V.)
| | - Alberto E. Espinel
- Pharmactive Biotech Products S.L.U., Parque Científico de Madrid, Avenida del Doctor Severo Ochoa, 37 Local 4J, 28108 Alcobendas, Spain; (A.E.E.); (D.G.-H.)
| | - Daniel González-Hedström
- Pharmactive Biotech Products S.L.U., Parque Científico de Madrid, Avenida del Doctor Severo Ochoa, 37 Local 4J, 28108 Alcobendas, Spain; (A.E.E.); (D.G.-H.)
| | - Miriam Granado García
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (M.D.l.F.M.); (M.R.-C.); (S.A.); (Á.L.G.-V.)
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
26
|
Messina A, Concerto C, Rodolico A, Petralia A, Caraci F, Signorelli MS. Is It Time for a Paradigm Shift in the Treatment of Schizophrenia? The Use of Inflammation-Reducing and Neuroprotective Drugs-A Review. Brain Sci 2023; 13:957. [PMID: 37371435 DOI: 10.3390/brainsci13060957] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/03/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Comprehending the pathogenesis of schizophrenia represents a challenge for global mental health. To date, although it is evident that alterations in dopaminergic, serotonergic, and glutamatergic neurotransmission underlie the clinical expressiveness of the disease, neuronal disconnections represent only an epiphenomenon. In recent years, several clinical studies have converged on the hypothesis of microglia hyperactivation and a consequent neuroinflammatory state as a pathogenic substrate of schizophrenia. Prenatal, perinatal, and postnatal factors can cause microglia to switch from M2 anti-inflammatory to M1 pro-inflammatory states. A continuous mild neuroinflammatory state progressively leads to neuronal loss, a reduction in dendritic spines, and myelin degeneration. The augmentation of drugs that reduce neuroinflammation to antipsychotics could be an effective therapeutic modality in managing schizophrenia. This review will consider studies in which drugs with anti-inflammatory and neuroprotective properties have been used in addition to antipsychotic treatment in patients with schizophrenia.
Collapse
Affiliation(s)
- Antonino Messina
- Department of Clinical and Experimental Medicine, Institute of Psychiatry, University of Catania, 95123 Catania, Italy
| | - Carmen Concerto
- Department of Clinical and Experimental Medicine, Institute of Psychiatry, University of Catania, 95123 Catania, Italy
| | - Alessandro Rodolico
- Department of Clinical and Experimental Medicine, Institute of Psychiatry, University of Catania, 95123 Catania, Italy
| | - Antonino Petralia
- Department of Clinical and Experimental Medicine, Institute of Psychiatry, University of Catania, 95123 Catania, Italy
| | - Filippo Caraci
- Department of Drug and Health Sciences, University of Catania, 95123 Catania, Italy
- Unit of Translational Neuropharmacology and Translational Neurosciences, Oasi Research Institute-IRCCS, 94018 Troina, Italy
| | - Maria Salvina Signorelli
- Department of Clinical and Experimental Medicine, Institute of Psychiatry, University of Catania, 95123 Catania, Italy
| |
Collapse
|
27
|
Cavestro C, Diodato D, Tiranti V, Di Meo I. Inherited Disorders of Coenzyme A Biosynthesis: Models, Mechanisms, and Treatments. Int J Mol Sci 2023; 24:ijms24065951. [PMID: 36983025 PMCID: PMC10054636 DOI: 10.3390/ijms24065951] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/09/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Coenzyme A (CoA) is a vital and ubiquitous cofactor required in a vast number of enzymatic reactions and cellular processes. To date, four rare human inborn errors of CoA biosynthesis have been described. These disorders have distinct symptoms, although all stem from variants in genes that encode enzymes involved in the same metabolic process. The first and last enzymes catalyzing the CoA biosynthetic pathway are associated with two neurological conditions, namely pantothenate kinase-associated neurodegeneration (PKAN) and COASY protein-associated neurodegeneration (CoPAN), which belong to the heterogeneous group of neurodegenerations with brain iron accumulation (NBIA), while the second and third enzymes are linked to a rapidly fatal dilated cardiomyopathy. There is still limited information about the pathogenesis of these diseases, and the knowledge gaps need to be resolved in order to develop potential therapeutic approaches. This review aims to provide a summary of CoA metabolism and functions, and a comprehensive overview of what is currently known about disorders associated with its biosynthesis, including available preclinical models, proposed pathomechanisms, and potential therapeutic approaches.
Collapse
Affiliation(s)
- Chiara Cavestro
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20126 Milan, Italy
| | - Daria Diodato
- Unit of Muscular and Neurodegenerative Disorders, Ospedale Pediatrico Bambino Gesù, 00165 Rome, Italy
| | - Valeria Tiranti
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20126 Milan, Italy
| | - Ivano Di Meo
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20126 Milan, Italy
| |
Collapse
|
28
|
Zhang T, Zhang D, Zhang Z, Tian J, An J, Zhang W, Ben Y. Alpha-lipoic acid activates AMPK to protect against oxidative stress and apoptosis in rats with diabetic peripheral neuropathy. Hormones (Athens) 2023; 22:95-105. [PMID: 36289188 DOI: 10.1007/s42000-022-00413-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 10/18/2022] [Indexed: 11/04/2022]
Abstract
PURPOSE To investigate the AMPK pathway-mediated effect of alpha-lipoic acid (ALA) on the dorsal root ganglia (DRGs) of rats with diabetic peripheral neuropathy (DPN) and to attempt to elucidate the underlying mechanism. METHODS Sprague-Dawley rats (n = 15) were randomly divided into three groups. The control group was fed a standard diet, and the other groups were fed a high-carbohydrate/high-fat diet. Diabetes was established by a single streptozotocin (STZ) (30 mg/kg) injection, and control rats were injected with an equal volume of citrate buffer. ALA (60 mg/kg/day) was administered for 12 weeks. The nerve conduction velocity (NCV) of the sciatic nerve was measured. Glutathione (GSH) and malondialdehyde (MDA) concentrations in serum were measured with the thiobarbituric acid method and biochemistry. Pathological changes in the rat DRGs were observed. AMPK, phospho-AMPK (p-AMPK), nuclear factor erythroid-2-related factor 2 (Nrf2), phospho-nuclear factor erythroid-2-related factor 2 (p-Nrf2), heme oxygenase 1 (HO-1), quinone oxidoreductase 1 (NQO1), Forkhead box O3 (FoxO3a), phospho-Forkhead box O3 (p-FoxO3a), and Bcl-2 interacting mediator of cell death (Bim) expression levels were assessed by immunohistochemistry and western blotting. RESULTS ALA improved the motor NCV (MNCV) and sensory NCV (SNCV) of rats with DPN and reduced their mechanical pain threshold. ALA increased serum GSH concentrations and decreased serum MDA concentrations. Additionally, AMPK was activated by ALA. Nrf2, p-Nrf2, HO-1, and NQO1 expression was upregulated, while FoxO3a, p-FoxO3a, and Bim expression was downregulated. ALA reduced oxidative stress and apoptosis in DRG. CONCLUSION ALA alleviates DPN and improves peripheral nerve function. ALA reduces oxidative stress by activating Nrf2 through AMPK and inhibits FoxO3a and Bim thereby reducing neuronal apoptosis.
Collapse
Affiliation(s)
- Tianya Zhang
- Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People's Republic of China
| | - Dong Zhang
- Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People's Republic of China
| | - Zhihong Zhang
- Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People's Republic of China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People's Republic of China
| | - Jiaxin Tian
- Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People's Republic of China
| | - Jingwen An
- Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People's Republic of China
| | - Wang Zhang
- Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People's Republic of China
| | - Ying Ben
- Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People's Republic of China.
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People's Republic of China.
| |
Collapse
|
29
|
van Zundert SKM, Griffioen PH, van Rossem L, Willemsen SP, de Rijke YB, van Schaik RHN, Steegers-Theunissen RPM, Mirzaian M. Simultaneous quantification of tryptophan metabolites by liquid chromatography tandem mass spectrometry during early human pregnancy. Clin Chem Lab Med 2023; 61:442-451. [PMID: 36458576 DOI: 10.1515/cclm-2022-0790] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 11/22/2022] [Indexed: 12/05/2022]
Abstract
OBJECTIVES In this study we describe the development and validation of a liquid chromatography mass spectrometry method (LC-MS/MS) to quantify five tryptophan (TRP) metabolites within the kynurenine- and serotonin pathway and apply the method to serum samples of women in the first trimester of pregnancy. A secondary aim was to investigate the correlation between body mass index (BMI) and the five analytes. METHODS A LC-MS/MS was developed for the analysis of TRP, kynurenine (KYN), 5-hydroxytryptophan (5-HTP), hydroxytryptamine (5-HT), and 5-hydroxyindole acetic acid (5-HIAA). Serum samples (n=374) were analyzed of pregnant women (median gestational age: 8 ± 2 weeks) participating in a subcohort of the Rotterdam Periconceptional Cohort (Predict study). RESULTS The LC-MS/MS method provided satisfactory separation of the five analytes (7 min run). For all analytes R2 was >0.995. Within- and between-run accuracies were 72-97% and 79-104%, and the precisions were all <15% except for the between-run precisions of the low QC-samples of 5-HTP and 5-HT (both 16%). Analyte concentrations were determined in serum samples of pregnant women (median (IQR)); TRP (µmol/L): 57.5 (13.4), KYN (µmol/L): 1.4 (0.4), 5-HTP (nmol/L): 4.1 (1.2), 5-HT (nmol/L): 615 (323.1), and 5-HIAA (nmol/L): 39.9 (17.0). BMI was negatively correlated with TRP, 5-HTP, and 5-HIAA (TRP: r=-0.18, p<0.001; 5-HTP: r=-0.13, p=0.02; natural log of 5-HIAA: r=-0.11, p=0.04), and positively with KYN (r=0.11, p=0.04). CONCLUSIONS The LC-MS/MS method is able to accurately quantify kynurenine- and serotonin pathway metabolites in pregnant women, providing an opportunity to investigate the role of the TRP metabolism in the (patho)physiology of pregnancy.
Collapse
Affiliation(s)
- Sofie K M van Zundert
- Department of Clinical Chemistry, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
- Department of Obstetrics and Gynecology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Pieter H Griffioen
- Department of Clinical Chemistry, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Lenie van Rossem
- Department of Obstetrics and Gynecology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Sten P Willemsen
- Department of Obstetrics and Gynecology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
- Department of Biostatistics, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Yolanda B de Rijke
- Department of Clinical Chemistry, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Ron H N van Schaik
- Department of Clinical Chemistry, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | | | - Mina Mirzaian
- Department of Clinical Chemistry, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
30
|
Kanemaru E, Miyazaki Y, Marutani E, Ezaka M, Goto S, Ohshima E, Bloch DB, Ichinose F. Intranasal administration of polysulfide prevents neurodegeneration in spinal cord and rescues mice from delayed paraplegia after spinal cord ischemia. Redox Biol 2023; 60:102620. [PMID: 36753926 PMCID: PMC9932672 DOI: 10.1016/j.redox.2023.102620] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/25/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Delayed paraplegia is a devastating complication of thoracoabdominal aortic surgery. Hydrogen sulfide (H2S) was reported to be protective in a mouse model of spinal cord ischemia and the beneficial effect of H2S has been attributed to polysulfides. The objective of this study was to investigate the effects of polysulfides on delayed paraplegia after spinal cord ischemia. METHODS AND RESULTS Spinal cord ischemia was induced in male and female C57BL/6J mice by clamping the aortic arch and the left subclavian artery. Glutathione trisulfide (GSSSG), glutathione (GSH), glutathione disulfide (GSSG), or vehicle alone was administered intranasally at 0, 8, 23, and 32 h after surgery. All mice treated with vehicle alone developed paraplegia within 48 h after surgery. GSSSG, but not GSH or GSSG, prevented paraplegia in 8 of 11 male mice (73%) and 6 of 8 female mice (75%). Intranasal administration of 34S-labeled GSSSG rapidly increased 34S-labeled sulfane sulfur species in the lumbar spinal cord. In mice treated with intranasal GSSSG, there were increased sulfane sulfur levels, and decreased neurodegeneration, microglia activation, and caspase-3 activation in the lumbar spinal cord. In vitro studies using murine primary cortical neurons showed that GSSSG increased intracellular levels of sulfane sulfur. GSSSG, but not GSH or GSSG, dose-dependently improved cell viability after oxygen and glucose deprivation/reoxygenation (OGD/R). Pantethine trisulfide (PTN-SSS) also increased intracellular sulfane sulfur and improved cell viability after OGD/R. Intranasal administration of PTN-SSS, but not pantethine, prevented paraplegia in 6 of 9 male mice (66%). CONCLUSIONS Intranasal administration of polysulfides rescued mice from delayed paraplegia after transient spinal cord ischemia. The neuroprotective effects of GSSSG were associated with increased levels of polysulfides and sulfane sulfur in the lumbar spinal cord. Targeted delivery of sulfane sulfur by polysulfides may prove to be a novel approach to the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Eiki Kanemaru
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA.
| | - Yusuke Miyazaki
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA.
| | - Eizo Marutani
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA.
| | - Mariko Ezaka
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA.
| | - Shunsaku Goto
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA.
| | - Etsuo Ohshima
- Corporate Strategy Department, Kyowa Hakko Bio Co., Ltd., Tokyo, 164-0001, Japan.
| | - Donald B. Bloch
- Department of Medicine, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Fumito Ichinose
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
31
|
Lipoic/Capsaicin-Related Amides: Synthesis and Biological Characterization of New TRPV1 Agonists Endowed with Protective Properties against Oxidative Stress. Int J Mol Sci 2022; 23:ijms232113580. [DOI: 10.3390/ijms232113580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
α-Lipoic acid is a sulfur-containing nutrient endowed with pleiotropic actions and a safe biological profile selected to replace the unsaturated alkyl acid of capsaicin with the aim of obtaining lipoic amides potentially active as a TRPV1 ligand and with significant antioxidant properties. Thus, nine compounds were obtained in good yields following a simple synthetic procedure and tested for their functional TRPV1 activity and radical-scavenger activity. The safe biological profile together with the protective effect against hypoxia damage as well as the in vitro antioxidant properties were also evaluated. Although less potent than capsaicin, almost all lipoic amides were found to be TRPV1 agonists and, specifically, compound 4, the lipoic analogue of capsaicin, proved to be the best ligand in terms of efficacy and potency. EPR experiments and in vitro biological assays suggested the potential protective role against oxidative stress of the tested compounds and their safe biological profile. Compounds 4, 5 and 9 significantly ameliorated the mitochondrial membrane potential caused by hypoxia condition and decreased F2-isoprostanes, known markers of oxidative stress. Thus, the experimental results encourage further investigation of the therapeutic potential of these lipoic amides.
Collapse
|
32
|
Amelioration of hydrolyzed guar gum on high-fat diet-induced obesity: Integrated hepatic transcriptome and metabolome. Carbohydr Polym 2022; 297:120051. [DOI: 10.1016/j.carbpol.2022.120051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 08/25/2022] [Accepted: 08/25/2022] [Indexed: 11/18/2022]
|
33
|
Massimini M, Bachetti B, Dalle Vedove E, Benvenga A, Di Pierro F, Bernabò N. A Set of Dysregulated Target Genes to Reduce Neuroinflammation at Molecular Level. Int J Mol Sci 2022; 23:ijms23137175. [PMID: 35806178 PMCID: PMC9266409 DOI: 10.3390/ijms23137175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/20/2022] [Accepted: 06/23/2022] [Indexed: 01/27/2023] Open
Abstract
Increasing evidence links chronic neurodegenerative diseases with neuroinflammation; it is known that neuroprotective agents are capable of modulating the inflammatory processes, that occur with the onset of neurodegeneration pathologies. Here, with the intention of providing a means for active compounds’ screening, a dysregulation of neuronal inflammatory marker genes was induced and subjected to neuroprotective active principles, with the aim of selecting a set of inflammatory marker genes linked to neurodegenerative diseases. Considering the important role of microglia in neurodegeneration, a murine co-culture of hippocampal cells and inflamed microglia cells was set up. The evaluation of differentially expressed genes and subsequent in silico analysis showed the main dysregulated genes in both cells and the principal inflammatory processes involved in the model. Among the identified genes, a well-defined set was chosen, selecting those in which a role in human neurodegenerative progression in vivo was already defined in literature, matched with the rate of prediction derived from the Principal Component Analysis (PCA) of in vitro treatment-affected genes variation. The obtained panel of dysregulated target genes, including Cxcl9 (Chemokine (C-X-C motif) ligand 9), C4b (Complement Component 4B), Stc1 (Stanniocalcin 1), Abcb1a (ATP Binding Cassette Subfamily B Member 1), Hp (Haptoglobin) and Adm (Adrenomedullin), can be considered an in vitro tool to select old and new active compounds directed to neuroinflammation.
Collapse
Affiliation(s)
- Marcella Massimini
- Faculty of Veterinary Medicine, University of Teramo, 64100 Teramo, Italy
- Correspondence:
| | - Benedetta Bachetti
- R&D Division, C.I.A.M. Srl, 63100 Ascoli Piceno, Italy; (B.B.); (E.D.V.); (A.B.)
| | - Elena Dalle Vedove
- R&D Division, C.I.A.M. Srl, 63100 Ascoli Piceno, Italy; (B.B.); (E.D.V.); (A.B.)
| | - Alessia Benvenga
- R&D Division, C.I.A.M. Srl, 63100 Ascoli Piceno, Italy; (B.B.); (E.D.V.); (A.B.)
| | - Francesco Di Pierro
- Velleja Research, 20125 Milan, Italy;
- Digestive Endoscopy Unit and Gastroenterology, Fondazione Poliambulanza, 25124 Brescia, Italy
| | - Nicola Bernabò
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy;
| |
Collapse
|
34
|
Identification of Human Brain Proteins for Bitter-Sweet Taste Perception: A Joint Proteome-Wide and Transcriptome-Wide Association Study. Nutrients 2022; 14:nu14102177. [PMID: 35631318 PMCID: PMC9143225 DOI: 10.3390/nu14102177] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/17/2022] [Accepted: 05/20/2022] [Indexed: 02/04/2023] Open
Abstract
Objective: Bitter or sweet beverage perception is associated with alterations in brain structure and function. Our aim is to analyze the genetic association between bitter or sweet beverage perception and human brain proteins. Materials and methods: In our study, 8356 and 11,518 proteins were first collected from two reference datasets of human brain proteomes, the ROS/MAP and Banner. The bitter or sweet beverage perception-related proteome-wide association studies (PWAS) were then conducted by integrating recent genome-wide association study (GWAS) data (n = 422,300) of taste perception with human brain proteomes. The human brain gene expression profiles were collected from two reference datasets, including the brain RNA-seq (CBR) and brain RNA-seq splicing (CBRS). The taste perception-related transcriptome-wide association studies (TWAS) were finally performed by integrating the same GWAS data with human brain gene expression profiles to validate the PWAS findings. Results: In PWAS, four statistically significant proteins were identified using the ROS/MAP and then replicated using the Banner reference dataset (all permutated p < 0.05), including ABCG2 for total bitter beverages and tea, CPNE1 for total bitter beverage, ACTR1B for artificially sweetened beverages, FLOT2 for alcoholic bitter beverages and total sweet beverages. In TWAS analysis, six statistically significant genes were detected by CBR and confirmed by the CBRS reference dataset (all permutated p < 0.05), including PIGG for total bitter beverages and non-alcoholic bitter beverages, C3orf18 for total bitter beverages, ZSWIM7 for non-alcoholic bitter beverages, PEX7 for coffee, PKP4 for tea and RPLP2 for grape juice. Further comparison of the PWAS and TWAS found three common statistically significant proteins/genes identified from the Banner and CBR reference datasets, including THBS4 for total bitter beverages, CA4 for non-alcoholic bitter beverages, LIAS for non-grape juices. Conclusions: Our results support the potential effect of bitter or sweet beverage perception on brain function and identify several candidate brain proteins for bitter or sweet beverage perception.
Collapse
|
35
|
Pressurized liquids vs. high intensity focused ultrasounds for the extraction of proteins from a pomegranate seed waste. INNOV FOOD SCI EMERG 2022. [DOI: 10.1016/j.ifset.2022.102958] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
36
|
Del'Arco AE, Argolo DS, Guillemin G, Costa MDFD, Costa SL, Pinheiro AM. Neurological Infection, Kynurenine Pathway, and Parasitic Infection by Neospora caninum. Front Immunol 2022; 12:714248. [PMID: 35154065 PMCID: PMC8826404 DOI: 10.3389/fimmu.2021.714248] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 12/31/2021] [Indexed: 11/30/2022] Open
Abstract
Neuroinflammation is one of the most frequently studied topics of neurosciences as it is a common feature in almost all neurological disorders. Although the primary function of neuroinflammation is to protect the nervous system from an insult, the complex and sequential response of activated glial cells can lead to neurological damage. Depending on the type of insults and the time post-insult, the inflammatory response can be neuroprotective, neurotoxic, or, depending on the glial cell types, both. There are multiple pathways activated and many bioactive intermediates are released during neuroinflammation. One of the most common one is the kynurenine pathway, catabolizing tryptophan, which is involved in immune regulation, neuroprotection, and neurotoxicity. Different models have been used to study the kynurenine pathway metabolites to understand their involvements in the development and maintenance of the inflammatory processes triggered by infections. Among them, the parasitic infection Neospora caninum could be used as a relevant model to study the role of the kynurenine pathway in the neuroinflammatory response and the subset of cells involved.
Collapse
Affiliation(s)
- Ana Elisa Del'Arco
- Laboratory of Biochemistry and Veterinary Immunology, Center of Agrarian, Environmental and Biological Sciences, Federal University of Recôncavo of Bahia (UFRB), Cruz das Almas, Brazil
| | - Deivison Silva Argolo
- Laboratory of Neurochemistry and Cellular Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia (UFBA), Bahia, Brazil
| | - Gilles Guillemin
- Neuroinflammation Group, Macquarie Medicine School, Macquarie University, Sydney, NSW, Australia
| | - Maria de Fátima Dias Costa
- Laboratory of Neurochemistry and Cellular Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia (UFBA), Bahia, Brazil
| | - Silvia Lima Costa
- Laboratory of Neurochemistry and Cellular Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia (UFBA), Bahia, Brazil
| | - Alexandre Moraes Pinheiro
- Laboratory of Biochemistry and Veterinary Immunology, Center of Agrarian, Environmental and Biological Sciences, Federal University of Recôncavo of Bahia (UFRB), Cruz das Almas, Brazil
| |
Collapse
|
37
|
Guo D, Li F, Zhao J, Zhang H, Liu B, Pan J, Zhang W, Chen W, Xu Y, Jiang S, Zhai Q. Effect of an infant formula containing sn-2 palmitate on fecal microbiota and metabolome profiles of healthy term infants: a randomized, double-blind, parallel, controlled study. Food Funct 2022; 13:2003-2018. [PMID: 35098958 DOI: 10.1039/d1fo03692k] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Different infant diets have strong effects on child development and may engender variations in fecal microbiota and metabolites. The objective of this study was to evaluate the effect of an infant formula containing sn-2 palmitate on fecal microbiota and metabolites in healthy term infants. The study involved three groups as indicated below. Investigational: the group fed a formula containing high sn-2 palmitate for 16 weeks. Control: the group fed a formula using a regular vegetable oil for 16 weeks. Breastfed: the group fed breast milk for 16 weeks. Fecal samples were collected at 8 weeks (n = 35, 37, and 35, respectively) and 16 weeks (n = 30, 32, and 30, respectively) for the control, investigational, and breastfed infants. Microbiota data were obtained using 16S rRNA sequencing. Short-chain fatty acid (SCFA) analysis was performed using GC-MS, and untargeted metabolomics was conducted using LC-MS. The effect of the formula containing sn-2 palmitate was different from that of the control formula on microbiota and metabolites. Sn-2 palmitate promoted the proliferation of Bifidobacterium and reduced the abundance of Escherichia-Shigella at 8 weeks. Furthermore, it increased α-diversity and enhanced acetate content in feces at both 8 and 16 weeks. In the investigational group infants, the abundance of DL-tryptophan, indole-3-acrylic acid, acetyl-β-methylcholine, L-methionine, and 2-hydroxyvaleric acid significantly increased at 8 weeks, while a notable increase in the abundance of 3-phenyllactic acid, palmitic acid, L-phenylalanine, and leucylproline was observed at 16 weeks. In addition, compared with that of the control infants, the intestinal microbiota and metabolites of sn-2 palmitate-supplemented infants were more similar to those of the breastfed infants. The study hopes to provide a scientific basis for the development of functional infant formulas in the future.
Collapse
Affiliation(s)
- Danying Guo
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, P. R. China. .,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Fei Li
- Developmental and Behavioral Pediatric Department & Child Primary Care Department, Brain and Behavioral Research Unit of Shanghai Institute for Pediatric Research and MOE-Shanghai Key Laboratory for Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China. .,Shanghai Institute of Pediatric Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, P. R. China. .,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, P. R. China. .,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China.,National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China.,Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, China
| | - Bryan Liu
- College of Biotechnology, East China University of Science and Technology, Shanghai, China
| | - Jiancun Pan
- Nutrition and Metabolism Research Division, Innovation Center, Heilongjiang Feihe Dairy Co., Ltd, C-16, 10A Jiuxianqiao Rd., Chaoyang, Beijing 100015, China. .,PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, Xueyuan Road 38, Haidian, Beijing 100083, China.
| | - Wei Zhang
- Nutrition and Metabolism Research Division, Innovation Center, Heilongjiang Feihe Dairy Co., Ltd, C-16, 10A Jiuxianqiao Rd., Chaoyang, Beijing 100015, China. .,PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, Xueyuan Road 38, Haidian, Beijing 100083, China.
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, P. R. China. .,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China.,National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yajun Xu
- PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, Xueyuan Road 38, Haidian, Beijing 100083, China. .,Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Xueyuan Road 38, Haidian, Beijing 100083, China.,Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Xueyuan Road 38, Haidian, Beijing 100083, China
| | - Shilong Jiang
- Nutrition and Metabolism Research Division, Innovation Center, Heilongjiang Feihe Dairy Co., Ltd, C-16, 10A Jiuxianqiao Rd., Chaoyang, Beijing 100015, China. .,PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, Xueyuan Road 38, Haidian, Beijing 100083, China.
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, P. R. China. .,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| |
Collapse
|
38
|
Rimmele TS, Li S, Andersen JV, Westi EW, Rotenberg A, Wang J, Aldana BI, Selkoe DJ, Aoki CJ, Dulla CG, Rosenberg PA. Neuronal Loss of the Glutamate Transporter GLT-1 Promotes Excitotoxic Injury in the Hippocampus. Front Cell Neurosci 2022; 15:788262. [PMID: 35035352 PMCID: PMC8752461 DOI: 10.3389/fncel.2021.788262] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/08/2021] [Indexed: 12/26/2022] Open
Abstract
GLT-1, the major glutamate transporter in the mammalian central nervous system, is expressed in presynaptic terminals that use glutamate as a neurotransmitter, in addition to astrocytes. It is widely assumed that glutamate homeostasis is regulated primarily by glutamate transporters expressed in astrocytes, leaving the function of GLT-1 in neurons relatively unexplored. We generated conditional GLT-1 knockout (KO) mouse lines to understand the cell-specific functions of GLT-1. We found that stimulus-evoked field extracellular postsynaptic potentials (fEPSPs) recorded in the CA1 region of the hippocampus were normal in the astrocytic GLT-1 KO but were reduced and often absent in the neuronal GLT-1 KO at 40 weeks. The failure of fEPSP generation in the neuronal GLT-1 KO was also observed in slices from 20 weeks old mice but not consistently from 10 weeks old mice. Using an extracellular FRET-based glutamate sensor, we found no difference in stimulus-evoked glutamate accumulation in the neuronal GLT-1 KO, suggesting a postsynaptic cause of the transmission failure. We hypothesized that excitotoxicity underlies the failure of functional recovery of slices from the neuronal GLT-1 KO. Consistent with this hypothesis, the non-competitive NMDA receptor antagonist MK801, when present in the ACSF during the recovery period following cutting of slices, promoted full restoration of fEPSP generation. The inclusion of an enzymatic glutamate scavenging system in the ACSF conferred partial protection. Excitotoxicity might be due to excess release or accumulation of excitatory amino acids, or to metabolic perturbation resulting in increased vulnerability to NMDA receptor activation. Previous studies have demonstrated a defect in the utilization of glutamate by synaptic mitochondria and aspartate production in the synGLT-1 KO in vivo, and we found evidence for similar metabolic perturbations in the slice preparation. In addition, mitochondrial cristae density was higher in synaptic mitochondria in the CA1 region in 20–25 weeks old synGLT-1 KO mice in the CA1 region, suggesting compensation for loss of axon terminal GLT-1 by increased mitochondrial efficiency. These data suggest that GLT-1 expressed in presynaptic terminals serves an important role in the regulation of vulnerability to excitotoxicity, and this regulation may be related to the metabolic role of GLT-1 expressed in glutamatergic axon terminals.
Collapse
Affiliation(s)
- Theresa S Rimmele
- Department of Neurology and the F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, United States
| | - Shaomin Li
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Jens Velde Andersen
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Emil W Westi
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Alexander Rotenberg
- Department of Neurology and the F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, United States.,Program in Neuroscience, Harvard Medical School, Boston, MA, United States
| | - Jianlin Wang
- Department of Neurology and the F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, United States
| | - Blanca Irene Aldana
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Dennis J Selkoe
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Chiye J Aoki
- Center for Neural Science, New York University, NY, United States.,Neuroscience Institute NYU Langone Medical Center, NY, United States
| | - Chris G Dulla
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
| | - Paul Allen Rosenberg
- Department of Neurology and the F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, United States.,Program in Neuroscience, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
39
|
Emerging effects of tryptophan pathway metabolites and intestinal microbiota on metabolism and intestinal function. Amino Acids 2022; 54:57-70. [PMID: 35038025 DOI: 10.1007/s00726-022-03123-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 01/03/2022] [Indexed: 12/14/2022]
Abstract
The metabolism of dietary tryptophan occurs locally in the gut primarily via host enzymes, with ~ 5% metabolized by gut microbes. Three major tryptophan metabolic pathways are serotonin (beyond the scope of this review), indole, kynurenine and related derivatives. We introduce the gut microbiome, dietary tryptophan and the potential interplay of host and bacterial enzymes in tryptophan metabolism. Examples of bacterial transformation to indole and its derivative indole-3 propionic acid demonstrate associations with human metabolic disease and gut permeability, although causality remains to be determined. This review will focus on less well-known data, suggestive of local generation and functional significance in the gut, where kynurenine is converted to kynurenic acid and xanthurenic acid via enzymatic action present in both host and bacteria. Our functional data demonstrate a limited effect on intestinal epithelial cell monolayer permeability and on healthy mouse ileum. Other data suggest a modulatory effect on the microbiome, potentially in pathophysiology. Supportive of this, we found that the expression of mRNA for three kynurenine pathway enzymes were increased in colon from high-fat-fed mice, suggesting that this host pathway is perturbed in metabolic disease. These data, along with that from bacterial genomic analysis and germ-free mice, confirms expression and functional machinery of enzymes in this pathway. Therefore, the host and microbiota may play a significant dual role in either the production or regulation of these kynurenine metabolites which, in turn, can influence both host and microbiome, especially in the context of obesity and intestinal permeability.
Collapse
|
40
|
Huang Y, Zhao M, Chen X, Zhang R, Le A, Hong M, Zhang Y, Jia L, Zang W, Jiang C, Wang J, Fan X, Wang J. Tryptophan Metabolism in Central Nervous System Diseases: Pathophysiology and Potential Therapeutic Strategies. Aging Dis 2022; 14:858-878. [PMID: 37191427 DOI: 10.14336/ad.2022.0916] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 09/16/2022] [Indexed: 11/19/2022] Open
Abstract
The metabolism of L-tryptophan (TRP) regulates homeostasis, immunity, and neuronal function. Altered TRP metabolism has been implicated in the pathophysiology of various diseases of the central nervous system. TRP is metabolized through two main pathways, the kynurenine pathway and the methoxyindole pathway. First, TRP is metabolized to kynurenine, then kynurenic acid, quinolinic acid, anthranilic acid, 3-hydroxykynurenine, and finally 3-hydroxyanthranilic acid along the kynurenine pathway. Second, TRP is metabolized to serotonin and melatonin along the methoxyindole pathway. In this review, we summarize the biological properties of key metabolites and their pathogenic functions in 12 disorders of the central nervous system: schizophrenia, bipolar disorder, major depressive disorder, spinal cord injury, traumatic brain injury, ischemic stroke, intracerebral hemorrhage, multiple sclerosis, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and Huntington's disease. Furthermore, we summarize preclinical and clinical studies, mainly since 2015, that investigated the metabolic pathway of TRP, focusing on changes in biomarkers of these neurologic disorders, their pathogenic implications, and potential therapeutic strategies targeting this metabolic pathway. This critical, comprehensive, and up-to-date review helps identify promising directions for future preclinical, clinical, and translational research on neuropsychiatric disorders.
Collapse
|
41
|
Büki A, Kekesi G, Horvath G, Vécsei L. A Potential Interface between the Kynurenine Pathway and Autonomic Imbalance in Schizophrenia. Int J Mol Sci 2021; 22:10016. [PMID: 34576179 PMCID: PMC8467675 DOI: 10.3390/ijms221810016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 02/07/2023] Open
Abstract
Schizophrenia is a neuropsychiatric disorder characterized by various symptoms including autonomic imbalance. These disturbances involve almost all autonomic functions and might contribute to poor medication compliance, worsened quality of life and increased mortality. Therefore, it has a great importance to find a potential therapeutic solution to improve the autonomic disturbances. The altered level of kynurenines (e.g., kynurenic acid), as tryptophan metabolites, is almost the most consistently found biochemical abnormality in schizophrenia. Kynurenic acid influences different types of receptors, most of them involved in the pathophysiology of schizophrenia. Only few data suggest that kynurenines might have effects on multiple autonomic functions. Publications so far have discussed the implication of kynurenines and the alteration of the autonomic nervous system in schizophrenia independently from each other. Thus, the coupling between them has not yet been addressed in schizophrenia, although their direct common points, potential interfaces indicate the consideration of their interaction. The present review gathers autonomic disturbances, the impaired kynurenine pathway in schizophrenia, and the effects of kynurenine pathway on autonomic functions. In the last part of the review, the potential interaction between the two systems in schizophrenia, and the possible therapeutic options are discussed.
Collapse
Affiliation(s)
- Alexandra Büki
- Department of Physiology, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 10., H-6720 Szeged, Hungary; (A.B.); (G.K.); (G.H.)
| | - Gabriella Kekesi
- Department of Physiology, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 10., H-6720 Szeged, Hungary; (A.B.); (G.K.); (G.H.)
| | - Gyongyi Horvath
- Department of Physiology, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 10., H-6720 Szeged, Hungary; (A.B.); (G.K.); (G.H.)
| | - László Vécsei
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6., H-6725 Szeged, Hungary
- MTA-SZTE Neuroscience Research Group, H-6725 Szeged, Hungary
- Interdisciplinary Excellence Center, Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6., H-6725 Szeged, Hungary
| |
Collapse
|
42
|
Wang CC, Hsieh PW, Kuo JR, Wang SJ. Rosmarinic Acid, a Bioactive Phenolic Compound, Inhibits Glutamate Release from Rat Cerebrocortical Synaptosomes through GABA A Receptor Activation. Biomolecules 2021; 11:1029. [PMID: 34356653 PMCID: PMC8301814 DOI: 10.3390/biom11071029] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 07/13/2021] [Accepted: 07/13/2021] [Indexed: 12/15/2022] Open
Abstract
Rosmarinic acid, a major component of rosemary, is a polyphenolic compound with potential neuroprotective effects. Asreducing the synaptic release of glutamate is crucial to achieving neuroprotectant's pharmacotherapeutic effects, the effect of rosmarinic acid on glutamate release was investigated in rat cerebrocortical nerve terminals (synaptosomes). Rosmarinic acid depressed the 4-aminopyridine (4-AP)-induced glutamate release in a concentration-dependent manner. The removal of extracellular calcium and the blockade of vesicular transporters prevented the inhibition of glutamate release by rosmarinic acid. Rosmarinic acid reduced 4-AP-induced intrasynaptosomal Ca2+ elevation. The inhibition of N-, P/Q-type Ca2+ channels and the calcium/calmodulin-dependent kinase II (CaMKII) prevented rosmarinic acid from having effects on glutamate release. Rosmarinic acid also reduced the 4-AP-induced activation of CaMKII and the subsequent phosphorylation of synapsin I, the main presynaptic target of CaMKII. In addition, immunocytochemistry confirmed the presence of GABAA receptors. GABAA receptor agonist and antagonist blocked the inhibitory effect of rosmarinic acid on 4-AP-evoked glutamate release. Docking data also revealed that rosmarinic acid formed a hydrogen bond with the amino acid residues of GABAA receptor. These results suggested that rosmarinic acid activates GABAA receptors in cerebrocortical synaptosomes to decrease Ca2+ influx and CaMKII/synapsin I pathway to inhibit the evoked glutamate release.
Collapse
Affiliation(s)
- Che-Chuan Wang
- Chi Mei Medical Center, Department of Neurosurgery, Tainan 71004, Taiwan; (C.-C.W.); (J.-R.K.)
- Biotechnology, Southern Taiwan University of Science and Technology, Tainan 71005, Taiwan
| | - Pei-Wen Hsieh
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan;
- Graduate Institute of Natural Products, School of Traditional Chinese Medicine, and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33303, Taiwan
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan 33303, Taiwan
| | - Jinn-Rung Kuo
- Chi Mei Medical Center, Department of Neurosurgery, Tainan 71004, Taiwan; (C.-C.W.); (J.-R.K.)
- Biotechnology, Southern Taiwan University of Science and Technology, Tainan 71005, Taiwan
| | - Su-Jane Wang
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan;
- School of Medicine, Fu Jen Catholic University, New Taipei City 24205, Taiwan
| |
Collapse
|
43
|
Genome-wide association study of stimulant dependence. Transl Psychiatry 2021; 11:363. [PMID: 34226506 PMCID: PMC8257618 DOI: 10.1038/s41398-021-01440-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 05/04/2021] [Accepted: 05/13/2021] [Indexed: 11/08/2022] Open
Abstract
Stimulant dependence is heritable, but specific genetic factors underlying the trait have not been identified. A genome-wide association study for stimulant dependence was performed in a discovery cohort of African- (AA) and European-ancestry (EA) subjects ascertained for genetic studies of alcohol, opioid, and cocaine use disorders. The sample comprised individuals with DSM-IV stimulant dependence (393 EA cases, 5288 EA controls; 155 AA cases, 5603 AA controls). An independent cohort from the family-based Collaborative Study on the Genetics of Alcoholism (532 EA cases, 7635 EA controls; 53 AA cases, AA 3352 controls) was used for replication. One variant in SLC25A16 (rs2394476, p = 3.42 × 10-10, odds ratio [OR] = 3.70) was GWS in AAs. Four other loci showed suggestive evidence, including KCNA4 in AAs (rs11500237, p = 2.99 × 10-7, OR = 2.31) which encodes one of the potassium voltage-gated channel protein that has been linked to several other substance use disorders, and CPVL in the combined population groups (rs1176440, p = 3.05 × 10-7, OR = 1.35), whose expression was previously shown to be upregulated in the prefrontal cortex from users of cocaine, cannabis, and phencyclidine. Analysis of the top GWAS signals revealed a significant enrichment with nicotinic acetylcholine receptor genes (adjusted p = 0.04) and significant pleiotropy between stimulant dependence and alcohol dependence in EAs (padj = 3.6 × 10-3), an anxiety disorder in EAs (padj = 2.1 × 10-4), and ADHD in both AAs (padj = 3.0 × 10-33) and EAs (padj = 6.7 × 10-35). Our results implicate novel genes and pathways as having roles in the etiology of stimulant dependence.
Collapse
|
44
|
Park DH, Park JY, Kang KS, Hwang GS. Neuroprotective Effect of Gallocatechin Gallate on Glutamate-Induced Oxidative Stress in Hippocampal HT22 Cells. Molecules 2021; 26:molecules26051387. [PMID: 33806640 PMCID: PMC7961752 DOI: 10.3390/molecules26051387] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/25/2021] [Accepted: 03/01/2021] [Indexed: 12/13/2022] Open
Abstract
Oxidative stress leads to protein degeneration or mitochondrial dysfunction, causing neuronal cell death. Glutamate is a neurotransmitter that nerve cells use to send signals. However, the excess accumulation of glutamate can cause excitotoxicity in the central nervous system. In this study, we deciphered the molecular mechanism of catechin-mediated neuroprotective effect on glutamate-induced oxidative stress in mouse hippocampal neuronal HT22 cells. Cellular antioxidant activity was determined using the 1,1-diphenyl-picryl hydrazyl (DPPH) assay and 2',7'-dichlorodihydrofluorescein diacetate (DCFDA) staining. Furthermore, the levels of intracellular calcium (Ca2+) as well as nuclear condensation and protein expression related to neuronal damage were assessed. All five catechins (epigallocatechin gallate, gallocatechin gallate (GCG), gallocatechin, epicatechin gallate, and epicatechin) showed strong antioxidant effects. Among them, GCG exhibited the highest neuroprotective effect against glutamate excitotoxicity and was used for further mechanistic studies. The glutamate-induced increase in intracellular Ca2+ was reduced after GCG treatment. Moreover, GCG reduced nuclear condensation and the phosphorylation of extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinases (JNK) involved in cell death. The neuroprotective effect of GCG against glutamate-induced oxidative stress in HT22 cells was attributed to the reduction in intracellular free radicals and Ca2+ influx and also the inhibition of phosphorylation of ERK and JNK. Furthermore, the antioxidant effect of GCG was found to be likely due to the inhibition of phosphorylation of ERK and JNK that led to the effective suppression of neurocytotoxicity caused by glutamate in HT22 cells.
Collapse
Affiliation(s)
- Do Hwi Park
- College of Korean Medicine, Gachon University, Seongnam 13120, Korea;
| | - Jun Yeon Park
- Department of Food Science and Biotechnology, Kyonggi University, Suwon 16227, Korea;
| | - Ki Sung Kang
- College of Korean Medicine, Gachon University, Seongnam 13120, Korea;
- Correspondence: (K.S.K.); (G.S.H.); Tel.: +82-31-750-5402 (K.S.K.); +82-31-750-5421 (G.S.H.); Fax: +82-31-750-6028 (K.S.K.); +82-31-750-7029 (G.S.H.)
| | - Gwi Seo Hwang
- College of Korean Medicine, Gachon University, Seongnam 13120, Korea;
- Correspondence: (K.S.K.); (G.S.H.); Tel.: +82-31-750-5402 (K.S.K.); +82-31-750-5421 (G.S.H.); Fax: +82-31-750-6028 (K.S.K.); +82-31-750-7029 (G.S.H.)
| |
Collapse
|