1
|
Smiley CE, Pate BS, Bouknight SJ, Wood SK. Individual differences in behavioral responses to predator odor predict subsequent stress reactivity in female rats. Stress 2025; 28:2479739. [PMID: 40181610 PMCID: PMC12081064 DOI: 10.1080/10253890.2025.2479739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 03/10/2025] [Indexed: 04/05/2025] Open
Abstract
Stress-induced neuropsychiatric disorders are among the most prevalent medical conditions and have widespread effects on both patients and society. Females experience over twice the rates of stress-related anxiety and depression when compared to males and often exhibit worse symptomatology and treatment outcomes. However, preclinical experiments exploring the neurobiological mechanisms of stress susceptibility in females have been traditionally understudied. Previous data from our lab has determined that females are selectively vulnerable to the consequences of vicarious witness stress, and these experiments were designed to determine specific behavioral and physiological factors that could predict which groups would be more susceptible to the effects of stress. Adult, female, Sprague-Dawley rats were first exposed to a ferret predator odor to determine baseline individual differences in behavioral responses. Rats were stratified by the duration of freezing behavior exhibited in response to the ferret odor and equally balanced into non-stressed controls and vicarious witness stress exposed groups. These female rats were then assessed on a battery of behavioral tasks including sucrose preference, elevated plus maze, acoustic startle, and the ferret odor and witness stress cue exposures to determine if baseline differences in stress responding can predict the behavioral response to future stress and stress cues. High freezing in response to the ferret odor was associated with behavioral sensitization to witness stress and hypervigilant responses to stress cues that was accompanied by exaggerated neuroimmune responses. These experiments establish a powerful behavioral predictor of stress susceptibility in females and begin to address neurobiological correlates that underlie this response.
Collapse
Affiliation(s)
- Cora E. Smiley
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC 29209
- WJB Dorn Veterans Administration Medical Center, Columbia, SC 29209
| | - Brittany S. Pate
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC 29209
- University of South Carolina, Department of Exercise Science, Columbia, SC 29209
| | - Samantha J. Bouknight
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC 29209
| | - Susan K. Wood
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC 29209
- WJB Dorn Veterans Administration Medical Center, Columbia, SC 29209
- USC Institute for Cardiovascular Disease Research, Columbia, SC, 29209
| |
Collapse
|
2
|
Liu JH, Liu KY, Zhao X, Zhou X, Jiang Y. Cardiovascular toxicities associated with chimeric antigen receptor T-cell therapy. Front Pharmacol 2025; 16:1578157. [PMID: 40406483 PMCID: PMC12094984 DOI: 10.3389/fphar.2025.1578157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 04/08/2025] [Indexed: 05/26/2025] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has emerged as a groundbreaking immunotherapeutic approach, particularly for oncohematological patients who are refractory to conventional treatments. As clinical trials expand the applications of CAR T-cell therapy beyond hematologic malignancies, a critical understanding of its associated toxicities, particularly cardiovascular complications, becomes imperative. This review synthesizes current literature on the interplay between cytokine release syndrome (CRS) and cardiotoxicity related to CAR T-cell therapy, emphasizing the potential severity of these adverse events. While significant progress has been made in managing CRS, the cardiac manifestations-ranging from mild events to life-threatening complications-remain underreported in pivotal studies. We explore the incidence and nature of cardiotoxicity in real-world and clinical trial settings, identify risk factors contributing to cardiovascular events, and propose guidelines for pre-therapy evaluations, post-infusion monitoring, and management strategies. By highlighting the urgent need for heightened awareness and proactive care, this review aims to enhance patient safety and optimize outcomes in the evolving landscape of CAR T-cell therapy.
Collapse
Affiliation(s)
- Jia-Hui Liu
- Cancer Center, The First Hospital of Jilin University, Changchun, China
- Cancer Research Institute of Jilin University, The First Hospital of Jilin University, Changchun, China
- International Center of Future Science, Jilin University, Changchun, China
| | - Kun-Yao Liu
- Cancer Research Institute of Jilin University, The First Hospital of Jilin University, Changchun, China
| | - Xiang Zhao
- Cancer Research Institute of Jilin University, The First Hospital of Jilin University, Changchun, China
| | - Xin Zhou
- Cancer Center, The First Hospital of Jilin University, Changchun, China
- Cancer Research Institute of Jilin University, The First Hospital of Jilin University, Changchun, China
- International Center of Future Science, Jilin University, Changchun, China
| | - Yichuan Jiang
- Department of Pharmacy, China-Japan Union Hospital of Jilin University, Changchun, China
- Department of Clinical Pharmacy, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
3
|
Nie J, Zhou L, Tian W, Liu X, Yang L, Yang X, Zhang Y, Wei S, Wang DW, Wei J. Deep insight into cytokine storm: from pathogenesis to treatment. Signal Transduct Target Ther 2025; 10:112. [PMID: 40234407 PMCID: PMC12000524 DOI: 10.1038/s41392-025-02178-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/22/2024] [Accepted: 02/12/2025] [Indexed: 04/17/2025] Open
Abstract
Cytokine storm (CS) is a severe systemic inflammatory syndrome characterized by the excessive activation of immune cells and a significant increase in circulating levels of cytokines. This pathological process is implicated in the development of life-threatening conditions such as fulminant myocarditis (FM), acute respiratory distress syndrome (ARDS), primary or secondary hemophagocytic lymphohistiocytosis (HLH), cytokine release syndrome (CRS) associated with chimeric antigen receptor-modified T (CAR-T) therapy, and grade III to IV acute graft-versus-host disease following allogeneic hematopoietic stem cell transplantation. The significant involvement of the JAK-STAT pathway, Toll-like receptors, neutrophil extracellular traps, NLRP3 inflammasome, and other signaling pathways has been recognized in the pathogenesis of CS. Therapies targeting these pathways have been developed or are currently being investigated. While novel drugs have demonstrated promising therapeutic efficacy in mitigating CS, the overall mortality rate of CS resulting from underlying diseases remains high. In the clinical setting, the management of CS typically necessitates a multidisciplinary team strategy encompassing the removal of abnormal inflammatory or immune system activation, the preservation of vital organ function, the treatment of the underlying disease, and the provision of life supportive therapy. This review provides a comprehensive overview of the key signaling pathways and associated cytokines implicated in CS, elucidates the impact of dysregulated immune cell activation, and delineates the resultant organ injury associated with CS. In addition, we offer insights and current literature on the management of CS in cases of FM, ARDS, systemic inflammatory response syndrome, treatment-induced CRS, HLH, and other related conditions.
Collapse
Grants
- 82070217, 81873427 National Natural Science Foundation of China (National Science Foundation of China)
- 82100401 National Natural Science Foundation of China (National Science Foundation of China)
- 81772477, 81201848, 82473220 National Natural Science Foundation of China (National Science Foundation of China)
- 82330010,81630010,81790624 National Natural Science Foundation of China (National Science Foundation of China)
- National High Technology Research and Development Program of China, Grant number: 2021YFA1101500.
- The Hubei Provincial Natural Science Foundation (No.2024AFB050)
- Project of Shanxi Bethune Hospital, Grant Numbber: 2023xg02); Fundamental Research Program of Shanxi Province, Grant Numbber: 202303021211224
- The Key Scientific Research Project of COVID-19 Infection Emergency Treatment of Shanxi Bethune Hospital (2023xg01), 2023 COVID-19 Research Project of Shanxi Provincial Health Commission (No.2023XG001, No. 2023XG005), Four “Batches” Innovation Project of Invigorating Medical through Science and Technology of Shanxi Province (2023XM003), Cancer special Fund research project of Shanxi Bethune Hospital (No. 2020-ZL04), and External Expert Workshop Fund Program of Shanxi Provincial Health Commission(Proteomics Shanxi studio for Huanghe professor)
- Fundamental Research Program of Shanxi Province(No.202303021221192); 2023 COVID-19 Emergency Project of Shanxi Health Commission (Nos.2023XG001,2023XG005)
Collapse
Affiliation(s)
- Jiali Nie
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Ling Zhou
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Branch of National Clinical Research Center for Infectious Diseases, Wuhan Pulmonary Hospital (Wuhan Tuberculosis Prevention and Control Institute), Wuhan, China
| | - Weiwei Tian
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Xiansheng Liu
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Branch of National Clinical Research Center for Infectious Diseases, Wuhan Pulmonary Hospital (Wuhan Tuberculosis Prevention and Control Institute), Wuhan, China
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Liping Yang
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Xingcheng Yang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yicheng Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuang Wei
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Branch of National Clinical Research Center for Infectious Diseases, Wuhan Pulmonary Hospital (Wuhan Tuberculosis Prevention and Control Institute), Wuhan, China.
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China.
| | - Jia Wei
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
4
|
Kränkel N. The hidden costs of fast travel: aircraft noise and diabetes risk. Eur J Prev Cardiol 2025; 32:315-316. [PMID: 40037400 DOI: 10.1093/eurjpc/zwae394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Affiliation(s)
- Nicolle Kränkel
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Hindenburgdamm 30, 12203 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
- Friede Springer-Cardiovascular Prevention Center @ Charité, Hindenburgdamm 30, 12203 Berlin, Germany
| |
Collapse
|
5
|
Kozlowska L, Viegas S, Scheepers PTJ, Duca RC, Godderis L, Martins C, Ciura K, Jagiello K, João Silva M, Mahiout S, Mārtiņsone I, Matisāne L, Nieuwenhuyse AV, Puzyn T, Sijko-Szpanska M, Verdonck J, Santonen T. HBM4EU E-waste study - An untargeted metabolomics approach to characterize metabolic changes during E-waste recycling. ENVIRONMENT INTERNATIONAL 2025; 196:109281. [PMID: 39842165 DOI: 10.1016/j.envint.2025.109281] [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: 08/20/2024] [Revised: 12/05/2024] [Accepted: 01/13/2025] [Indexed: 01/24/2025]
Abstract
E-waste contains hazardous chemicals that may be a direct health risk for workers involved in recycling. We conducted an untargeted metabolomics analysis of urine samples collected from male e-waste processing workers to explore metabolic changes associated with chemical exposures in e-waste recycling in Belgium, Finland, Latvia, Luxembourg, the Netherlands, Poland, and Portugal. Questionnaire data and urine samples were obtained from workers involved in the processing of e-waste (sorting, dismantling, shredding, pre-processing, metal, and non-metal processing), as well as from controls with no known occupational exposure. Pre- and post-shift urine samples were collected and analysed using ultrahigh-performance liquid chromatography-mass spectrometry (UPLC-MS). A total of 32 endogenous urinary metabolites were annotated with a Variable Importance in Projection (VIP) above 2, indicating that e-waste recycling is mainly associated with changes in steroid hormone and neurotransmitter metabolism, energy metabolism, bile acid biosynthesis, and inflammation. The highest VIP was observed for dopamine-o-quinone, which is linked to Parkinson's disease. These and other changes in metabolism in workers employed in the processing of e-waste need further verification in targeted studies.
Collapse
Affiliation(s)
- Lucyna Kozlowska
- Laboratory of Human Metabolism Research, Warsaw University of Life Sciences, Warsaw, Poland.
| | - Susana Viegas
- NOVA National School of Public Health, Public Health Research Centre, Comprehensive Health Research Center, CHRC, REAL, CCAL, NOVA University Lisbon, Lisbon, Portugal
| | - Paul T J Scheepers
- Radboud Institute for Biological and Environmental Science, Radboud University, Nijmegen, the Netherlands
| | - Radu C Duca
- Labotoire National de Santé (LNS), Unit Environmental Hygiene and Human Biological Monitoring, Department of Health Protection, Dudelange, Luxembourg; Environment and Health, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
| | - Lode Godderis
- Environment and Health, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium; Idewe, External Service for Prevention and Protection at Work, Heverlee, Belgium
| | - Carla Martins
- NOVA National School of Public Health, Public Health Research Centre, Comprehensive Health Research Center, CHRC, REAL, CCAL, NOVA University Lisbon, Lisbon, Portugal
| | - Krzesimir Ciura
- Medical University of Gdansk, Gdansk, Poland; QSAR Laboratory Ltd., Gdansk, Poland; Department of Environmental Chemistry and Radiochemistry, Faculty of Chemistry, University of Gdansk, Gdansk, Poland
| | - Karolina Jagiello
- QSAR Laboratory Ltd., Gdansk, Poland; Department of Environmental Chemistry and Radiochemistry, Faculty of Chemistry, University of Gdansk, Gdansk, Poland
| | - Maria João Silva
- Department of Human Genetics, National Institute of Health Dr. Ricardo Jorge, Lisbon, Portugal; ToxOmics, NOVA Medical School, NOVA University of Lisbon, Lisbon, Portugal
| | - Selma Mahiout
- Finnish Institute of Occupational Health, Helsinki, Finland
| | - Inese Mārtiņsone
- Institute of Occupational Health and Environmental Safety, Riga Stradins University, Riga, Latvia
| | - Linda Matisāne
- Institute of Occupational Health and Environmental Safety, Riga Stradins University, Riga, Latvia
| | - An van Nieuwenhuyse
- Labotoire National de Santé (LNS), Unit Environmental Hygiene and Human Biological Monitoring, Department of Health Protection, Dudelange, Luxembourg; Environment and Health, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
| | - Tomasz Puzyn
- QSAR Laboratory Ltd., Gdansk, Poland; Department of Environmental Chemistry and Radiochemistry, Faculty of Chemistry, University of Gdansk, Gdansk, Poland
| | - Monika Sijko-Szpanska
- Laboratory of Human Metabolism Research, Warsaw University of Life Sciences, Warsaw, Poland
| | - Jelle Verdonck
- Environment and Health, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
| | - Tiina Santonen
- Finnish Institute of Occupational Health, Helsinki, Finland
| |
Collapse
|
6
|
Son JW, Kang P, Min SS, Seol GH. Differential effects of clary sage ( Salvia sclarea L.) oil and linalyl acetate on depression levels in diabetic foot ulcer patients with T2DM: a randomized blinded controlled trial. Front Med (Lausanne) 2025; 12:1523441. [PMID: 39911870 PMCID: PMC11794106 DOI: 10.3389/fmed.2025.1523441] [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: 11/06/2024] [Accepted: 01/07/2025] [Indexed: 02/07/2025] Open
Abstract
Background Diabetic foot ulcer (DFU) is a significant global health concern due to its substantial burden on individuals and healthcare systems. In addition to its physical impact, DFU has emotional effects on patients. This study analyzed the effects of inhaling clary sage (CS; Salvia sclarea L.) or linalyl acetate (LA) on depression and psychological factors in patients with DFU. Methods This study, performed at the Diabetic Wound Center, enrolled DFU patients, categorized as having mild or moderate to severe depression based on visual analog scale (VAS) for depression scores. Patients were randomized to inhalation of 5% CS oil, 5% LA or almond oil (control) by natural breathing. Blood pressure, heart rate, Depression-VAS, Anxiety-VAS, and Stress-VAS were measured before and after inhalation. Results This study enrolled 72 patients with DFU, including 43 with mild and 29 with moderate to severe depression. Of the 43 patients with mild depression, 14, 14, and 15 were randomized to inhalation of CS, LA, and almond oil, respectively. Of the 29 patients with moderate to severe depression, 9, 11, and 9 were randomized to inhalation of CS, LA, and almond oil, respectively. Compared with inhalation of almond oil, inhalation of CS oil significantly reduced stress (p < 0.05) and (p < 0.01) in patients with mild depression, whereas inhalation of LA significantly reduced anxiety-VAS (p < 0.05) and depression-VAS (p < 0.05) in patients with moderate to severe depression. Conclusion Inhalation of CS oil may have the potential to alleviate stress and anxiety in DFU patients with mild depression, whereas inhalation of LA may have the potential to alleviate anxiety and depression in DFU patients with moderate to severe depression. These findings suggest that adjunct therapy in DFU patients should be individualized according to the degree of depression. Clinical trial registration http://cris.nih.go.kr/, identifier KCT0009722.
Collapse
Affiliation(s)
- Ji Won Son
- Department of Basic Nursing Science, College of Nursing, Korea University, Seoul, Republic of Korea
| | - Purum Kang
- Department of Nursing, College of Nursing, Woosuk University, Jeonju, Republic of Korea
| | - Sun Seek Min
- Department of Physiology and Biophysics, Eulji University School of Medicine, Daejeon, Republic of Korea
| | - Geun Hee Seol
- Department of Basic Nursing Science, College of Nursing, Korea University, Seoul, Republic of Korea
- BK21 FOUR Program of Transdisciplinary Major in Learning Health Systems, Graduate School, Korea University, Seoul, Republic of Korea
| |
Collapse
|
7
|
Wellens J, Sabino J, Vanuytsel T, Tack J, Vermeire S. Recent advances in clinical practice: mastering the challenge-managing IBS symptoms in IBD. Gut 2025; 74:312-321. [PMID: 39532478 DOI: 10.1136/gutjnl-2024-333565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 10/19/2024] [Indexed: 11/16/2024]
Abstract
Many patients with IBD report persisting symptoms, despite resolution of the inflammatory process. Although by definition, a diagnosis of IBS cannot be made, the prevalence of 'IBS in IBD' surpasses the rate of IBS in the global population by fivefold. Because IBS-like symptoms are associated with a decreased quality of life and increased healthcare utilisation in IBD, diagnosis and treatment are necessary. In this review, we summarise the current knowledge on IBS-like symptoms in IBD. A pathophysiological common ground is present, which includes genetic susceptibility, environmental triggers, gut microbial dysbiosis, increased intestinal permeability, visceral hypersensitivity and involvement of brain-gut interaction. When symptoms persist after resolution of inflammation, other GI diseases should be excluded based on the chief complaint, considering any possible psychological co-morbidity early in the diagnostic work-up. Subsequent treatment should be initiated that is evidence-based and often multimodal, including classical and non-classical pharmacological agents as well as lifestyle and microbiota-based approaches, spanning the breadth of the gut, brain and its interaction. Treatment goals in this substantial part of the IBD population should be adapted to not only focus on treating the inflammation but taking care of the patient.
Collapse
Affiliation(s)
- Judith Wellens
- Gastroenterology and Hepatology, KU Leuven University Hospitals Leuven Gasthuisberg Campus Hospital Pharmacy, Leuven, Belgium
- Chronic Diseases, Metabolism and Ageing, Translational Research in GastroIntestinal Disorders, KU Leuven, Leuven, Belgium
| | - João Sabino
- Gastroenterology and Hepatology, KU Leuven University Hospitals Leuven Gasthuisberg Campus Hospital Pharmacy, Leuven, Belgium
- Chronic Diseases, Metabolism and Ageing, Translational Research in GastroIntestinal Disorders, KU Leuven, Leuven, Belgium
| | - Tim Vanuytsel
- Gastroenterology and Hepatology, KU Leuven University Hospitals Leuven Gasthuisberg Campus Hospital Pharmacy, Leuven, Belgium
- Chronic Diseases, Metabolism and Ageing, Translational Research in GastroIntestinal Disorders, KU Leuven, Leuven, Belgium
| | - Jan Tack
- Gastroenterology and Hepatology, KU Leuven University Hospitals Leuven Gasthuisberg Campus Hospital Pharmacy, Leuven, Belgium
- Chronic Diseases, Metabolism and Ageing, Translational Research in GastroIntestinal Disorders, KU Leuven, Leuven, Belgium
| | - Séverine Vermeire
- Gastroenterology and Hepatology, KU Leuven University Hospitals Leuven Gasthuisberg Campus Hospital Pharmacy, Leuven, Belgium
- Chronic Diseases, Metabolism and Ageing, Translational Research in GastroIntestinal Disorders, KU Leuven, Leuven, Belgium
| |
Collapse
|
8
|
Ishino F, Itoh J, Matsuzawa A, Irie M, Suzuki T, Hiraoka Y, Yoshikawa M, Kaneko-Ishino T. RTL4, a Retrovirus-Derived Gene Implicated in Autism Spectrum Disorder, Is a Microglial Gene That Responds to Noradrenaline in the Postnatal Brain. Int J Mol Sci 2024; 25:13738. [PMID: 39769499 PMCID: PMC11678650 DOI: 10.3390/ijms252413738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/09/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Retrotransposon Gag-like 4 (RTL4), a gene acquired from a retrovirus, is a causative gene in autism spectrum disorder. Its knockout mice exhibit increased impulsivity, impaired short-term spatial memory, failure to adapt to novel environments, and delayed noradrenaline (NA) recovery in the frontal cortex. However, due to its very low expression in the brain, it remains unknown which brain cells express RTL4 and its dynamics in relation to NA. We addressed these issues using knock-in mice carrying endogenous Rtl4 fused to Venus, which encodes a fluorescent protein. The RTL4-Venus fusion protein was detected as a secreted protein in the midbrain, hypothalamus, hippocampus and amygdala in the postnatal brain. Its signal intensity was high during critical periods of neonatal adaptation to novel environments. It was upregulated by various stimuli, including isoproterenol administration, whereas it was decreased by anesthesia but was maintained by milnacipran administration, suggesting its highly sensitive response to stressors, possible dependence on the arousal state and involvement in the NA reuptake process. In vitro mixed glial culture experiments demonstrated that Rtl4 is a microglial gene and suggested that RTL4 secretion responds rapidly to isoproterenol. Microglial RTL4 plays an important role in the NA response and possibly in the development of the NAergic neuronal network in the brain.
Collapse
Affiliation(s)
- Fumitoshi Ishino
- Department of Epigenetics, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan; (A.M.); (M.I.)
| | - Johbu Itoh
- Department of Neurology, School of Medicine, Tokai University School of Medicine, Isehara 259-1193, Japan;
| | - Ayumi Matsuzawa
- Department of Epigenetics, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan; (A.M.); (M.I.)
| | - Masahito Irie
- Department of Epigenetics, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan; (A.M.); (M.I.)
- Faculty of Nursing, Tokai University School of Medicine, Isehara 259-1193, Japan
| | - Toru Suzuki
- Laboratory of Genome Editing for Biomedical Research, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan; (T.S.); (Y.H.)
| | - Yuichi Hiraoka
- Laboratory of Genome Editing for Biomedical Research, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan; (T.S.); (Y.H.)
- Laboratory of Molecular Neuroscience, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan
| | - Masanobu Yoshikawa
- Department of Clinical Pharmacology, Tokai University School of Medicine, Isehara 259-1193, Japan;
| | - Tomoko Kaneko-Ishino
- Faculty of Nursing, Tokai University School of Medicine, Isehara 259-1193, Japan
| |
Collapse
|
9
|
Riggott C, Ford AC, Gracie DJ. Review article: The role of the gut-brain axis in inflammatory bowel disease and its therapeutic implications. Aliment Pharmacol Ther 2024; 60:1200-1214. [PMID: 39367676 DOI: 10.1111/apt.18192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/01/2024] [Accepted: 07/10/2024] [Indexed: 10/06/2024]
Abstract
BACKGROUND Treatments targeting the gut-brain axis (GBA) are effective at reducing symptom burden in irritable bowel syndrome (IBS). The prevalence of common mental disorders and IBS-type symptom reporting is significantly higher in inflammatory bowel disease (IBD) than would be expected, suggesting potential GBA effects in this setting. Manipulation of the GBA may offer novel treatment strategies in selected patients with IBD. We present a narrative review of the bi-directional effects of the GBA in IBD and explore the potential for GBA-targeted therapies in this setting. METHODS We searched MEDLINE, EMBASE, EMBASE Classic, PsychINFO, and the Cochrane Central Register of Controlled Trials for relevant articles published by March 2024. RESULTS The bi-directional relationship between psychological well-being and adverse longitudinal disease activity outcomes, and the high prevalence of IBS-type symptom reporting highlight the presence of GBA-mediated effects in IBD. Treatments targeting gut-brain interactions including brain-gut behavioural treatments, neuromodulators, and dietary interventions appear to be useful adjunctive treatments in a subset of patients. CONCLUSIONS Psychological morbidity is prevalent in patients with IBD. The relationship between longitudinal disease activity outcomes, IBS-type symptom reporting, and poor psychological health is mediated via the GBA. Proactive management of psychological health should be integrated into routine care. Further clinical trials of GBA-targeted therapies, conducted in selected groups of patients with co-existent common mental disorders, or those who report IBS-type symptoms, are required to inform effective integrated models of care in the future.
Collapse
Affiliation(s)
- Christy Riggott
- Leeds Gastroenterology Institute, St. James's University Hospital, Leeds, UK
| | - Alexander C Ford
- Leeds Gastroenterology Institute, St. James's University Hospital, Leeds, UK
- Leeds Institute of Medical Research at St. James's, University of Leeds, Leeds, UK
| | - David J Gracie
- Leeds Gastroenterology Institute, St. James's University Hospital, Leeds, UK
| |
Collapse
|
10
|
Smiley CE, Pate BS, Bouknight SJ, Harrington EN, Jasnow AM, Wood SK. The functional role of locus coeruleus microglia in the female stress response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.10.575076. [PMID: 38260568 PMCID: PMC10802589 DOI: 10.1101/2024.01.10.575076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Neuropsychiatric disorders that result from stress exposure are highly associated with central inflammation. Our previous work established that females selectively exhibit heightened proinflammatory cytokine production within the noradrenergic locus coeruleus (LC) along with a hypervigilant behavioral phenotype in response to witnessing social stress, and ablation of microglia using pharmacological techniques prevents this behavioral response. These studies were designed to further investigate the impact of stress-induced neuroimmune signaling on the long-term behavioral and neuronal consequences of social stress exposure in females using chemogenetics. We first characterized the use of an AAV-CD68-Gi-DREADD virus targeted to microglia within the LC and confirmed viral transduction, selectivity, and efficacy. Clozapine-n-oxide (CNO) was used for the suppression of microglial reactivity during acute and chronic exposure to vicarious/witness social defeat in female rats. Chemogenetic-mediated inhibition of microglial reactivity during stress blunted the neuroimmune response to stress and prevented both acute and long-term hypervigilant behavioral responses. Further, a history of microglial suppression during stress prevented the heightened LC activity typically observed in response to stress cues. These studies are among the first to use a chemogenetic approach to inhibit microglia within the female brain in vivo and establish LC inflammation as a key mechanism underlying the behavioral and neuronal responses to social stress in females.
Collapse
Affiliation(s)
- Cora E. Smiley
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC 29209
- WJB Dorn Veterans Administration Medical Center, Columbia, SC 29209
| | - Brittany S. Pate
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC 29209
- University of South Carolina, Department of Exercise Science, Columbia, SC 29209
| | - Samantha J. Bouknight
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC 29209
| | - Evelynn N. Harrington
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC 29209
- WJB Dorn Veterans Administration Medical Center, Columbia, SC 29209
| | - Aaron M. Jasnow
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC 29209
| | - Susan K. Wood
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC 29209
- WJB Dorn Veterans Administration Medical Center, Columbia, SC 29209
| |
Collapse
|
11
|
Likhvantsev VV, Berikashvili LB, Yadgarov MY, Yakovlev AA, Kuzovlev AN. The Tri-Steps Model of Critical Conditions in Intensive Care: Introducing a New Paradigm for Chronic Critical Illness. J Clin Med 2024; 13:3683. [PMID: 38999249 PMCID: PMC11242724 DOI: 10.3390/jcm13133683] [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: 05/03/2024] [Revised: 06/15/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024] Open
Abstract
Background: The prevailing model for understanding chronic critical illness is a biphasic model, suggesting phases of acute and chronic critical conditions. A major challenge within this model is the difficulty in determining the timing of the process chronicity. It is likely that the triad of symptoms (inflammation, catabolism, and immunosuppression [ICIS]) could be associated with this particular point. We aimed to explore the impact of the symptom triad (inflammation, catabolism, immunosuppression) on the outcomes of patients hospitalized in intensive care units (ICUs). Methods: The eICU-CRD database with 200,859 ICU admissions was analyzed. Adult patients with the ICIS triad, identified by elevated CRP (>20 mg/L), reduced albumin (<30 g/L), and low lymphocyte counts (<0.8 × 109/L), were included. The cumulative risk of developing ICIS was assessed using the Nelson-Aalen estimator. Results: This retrospective cohort study included 894 patients (485 males, 54%), with 60 (6.7%) developing ICIS. The cumulative risk of ICIS by day 21 was 22.5%, with incidence peaks on days 2-3 and 10-12 after ICU admission. Patients with the ICIS triad had a 2.5-fold higher mortality risk (p = 0.009) and double the likelihood of using vasopressors (p = 0.008). The triad onset day did not significantly affect mortality (p = 0.104). Patients with ICIS also experienced extended hospital (p = 0.041) and ICU stays (p < 0.001). Conclusions: The symptom triad (inflammation, catabolism, immunosuppression) during hospitalization increases mortality risk by 2.5 times (p = 0.009) and reflects the chronicity of the critical condition. Identifying two incidence peaks allows the proposal of a new Tri-steps model of chronic critical illness with acute, extended, and chronic phases.
Collapse
Affiliation(s)
- Valery V Likhvantsev
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
| | - Levan B Berikashvili
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
| | - Mikhail Ya Yadgarov
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
| | - Alexey A Yakovlev
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
| | - Artem N Kuzovlev
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
| |
Collapse
|
12
|
Bodo CR, Salcudean A, Nirestean A, Lukacs E, Lica MM, Muntean DL, Anculia RC, Popovici RA, Neda Stepan O, Enătescu VR, Strete EG. Association between Chronic Misophonia-Induced Stress and Gastrointestinal Pathology in Children-A Hypothesis. CHILDREN (BASEL, SWITZERLAND) 2024; 11:699. [PMID: 38929278 PMCID: PMC11201990 DOI: 10.3390/children11060699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/30/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024]
Abstract
Misophonia is a neurophysiological disorder with behavioral implications, is complex and multifactorial in origin, and is characterized by an atypical and disproportionate emotional response to specific sounds or associated visual stimuli. Triggers include human-generated sounds, mainly sounds related to feeding and breathing processes, and repetitive mechanical sounds. In response to the triggering stimulus, the patient experiences immediate, high-intensity, disproportionate physical and emotional reactions that affect their quality of life and social functioning. The symptoms of misophonia can occur at any age, but onset in childhood or adolescence is most common. Affected children live in a constant state of anxiety, suffer continuous physical and emotional discomfort, and are thus exposed to significant chronic stress. Chronic stress, especially during childhood, has consequences on the main biological systems through the dysregulation of the hypothalamic-pituitary-adrenal axis, including the gastrointestinal tract. Here, we provide arguments for a positive correlation between misophonic pathology and gastrointestinal symptoms, and this hypothesis may be the starting point for further longitudinal studies that could investigate the correlations between these childhood vulnerabilities caused by misophonia and their effect on the gastrointestinal system. Further research to study this hypothesis is essential to ensure correct and timely diagnosis and optimal psychological and pharmacological support.
Collapse
Affiliation(s)
- Cristina Raluca Bodo
- Department of Ethics and Social Sciences, George Emil Palade University of Medicine and Pharmacy, Sciences and Technology of Târgu Mureș, 540136 Târgu Mureș, Romania; (C.R.B.); (M.M.L.)
| | - Andreea Salcudean
- Department of Ethics and Social Sciences, George Emil Palade University of Medicine and Pharmacy, Sciences and Technology of Târgu Mureș, 540136 Târgu Mureș, Romania; (C.R.B.); (M.M.L.)
| | - Aurel Nirestean
- Department of Psychiatry, George Emil Palade University of Medicine and Pharmacy, Sciences and Technology of Târgu Mureș, 540136 Târgu Mureș, Romania; (A.N.); (E.L.); (E.G.S.)
| | - Emese Lukacs
- Department of Psychiatry, George Emil Palade University of Medicine and Pharmacy, Sciences and Technology of Târgu Mureș, 540136 Târgu Mureș, Romania; (A.N.); (E.L.); (E.G.S.)
| | - Maria Melania Lica
- Department of Ethics and Social Sciences, George Emil Palade University of Medicine and Pharmacy, Sciences and Technology of Târgu Mureș, 540136 Târgu Mureș, Romania; (C.R.B.); (M.M.L.)
| | - Daniela Lucia Muntean
- Department of Analytical Chemistry and Drug Analysis, Faculty of Pharmacy, George Emil Palade University of Medicine and Pharmacy, Sciences and Technology of Târgu Mureș, 540136 Târgu Mureș, Romania;
| | | | - Ramona Amina Popovici
- Department of Dental Preventive Medicine, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Oana Neda Stepan
- Department VIII-Neurosciences, Discipline of Psychiatry, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania; (O.N.S.); (V.R.E.)
| | - Virgil Radu Enătescu
- Department VIII-Neurosciences, Discipline of Psychiatry, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania; (O.N.S.); (V.R.E.)
| | - Elena Gabriela Strete
- Department of Psychiatry, George Emil Palade University of Medicine and Pharmacy, Sciences and Technology of Târgu Mureș, 540136 Târgu Mureș, Romania; (A.N.); (E.L.); (E.G.S.)
| |
Collapse
|
13
|
Chen X, Gu J, Zhang X. Brain-Heart Axis and the Inflammatory Response: Connecting Stroke and Cardiac Dysfunction. Cardiology 2024; 149:369-382. [PMID: 38574466 PMCID: PMC11309082 DOI: 10.1159/000538409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 03/12/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND In recent years, the mechanistic interaction between the brain and heart has been explored in detail, which explains the effects of brain injuries on the heart and those of cardiac dysfunction on the brain. Brain injuries are the predominant cause of post-stroke deaths, and cardiac dysfunction is the second leading cause of mortality after stroke onset. SUMMARY Several studies have reported the association between brain injuries and cardiac dysfunction. Therefore, it is necessary to study the influence on the heart post-stroke to understand the underlying mechanisms of stroke and cardiac dysfunction. This review focuses on the mechanisms and the effects of cardiac dysfunction after the onset of stroke (ischemic or hemorrhagic stroke). KEY MESSAGES The role of the site of stroke and the underlying mechanisms of the brain-heart axis after stroke onset, including the hypothalamic-pituitary-adrenal axis, inflammatory and immune responses, brain-multi-organ axis, are discussed.
Collapse
Affiliation(s)
- Xiaosheng Chen
- Department of Neurosurgery, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| | - Jiajie Gu
- Department of Neurosurgery, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| | - Xiaojia Zhang
- Department of Neurosurgery, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
14
|
Wang S, Zhou S, Han Z, Yu B, Xu Y, Lin Y, Chen Y, Jin Z, Li Y, Cao Q, Xu Y, Zhang Q, Wang YC. From gut to brain: understanding the role of microbiota in inflammatory bowel disease. Front Immunol 2024; 15:1384270. [PMID: 38576620 PMCID: PMC10991805 DOI: 10.3389/fimmu.2024.1384270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 03/12/2024] [Indexed: 04/06/2024] Open
Abstract
With the proposal of the "biological-psychological-social" model, clinical decision-makers and researchers have paid more attention to the bidirectional interactive effects between psychological factors and diseases. The brain-gut-microbiota axis, as an important pathway for communication between the brain and the gut, plays an important role in the occurrence and development of inflammatory bowel disease. This article reviews the mechanism by which psychological disorders mediate inflammatory bowel disease by affecting the brain-gut-microbiota axis. Research progress on inflammatory bowel disease causing "comorbidities of mind and body" through the microbiota-gut-brain axis is also described. In addition, to meet the needs of individualized treatment, this article describes some nontraditional and easily overlooked treatment strategies that have led to new ideas for "psychosomatic treatment".
Collapse
Affiliation(s)
- Siyu Wang
- Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Department of Gastroenterology, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Shuwei Zhou
- Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Zhongyu Han
- Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Bin Yu
- Department of Gastroenterology, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Yin Xu
- Department of Gastroenterology, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Yumeng Lin
- Eye School of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yutong Chen
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zi Jin
- Department of Anesthesiology and Pain Rehabilitation, Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | - Yalong Li
- Anorectal Department, The Third Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming, China
| | - Qinhan Cao
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine (TCM), Chengdu, China
| | - Yunying Xu
- Clinical Medical School, Affiliated Hospital of Chengdu University, Chengdu, China
| | - Qiang Zhang
- Department of Gastroenterology, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Yuan-Cheng Wang
- Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
15
|
Wei H, Deng M, Ding R, Wei L, Yuan H. Macrophage β2-AR activation amplifies inflammation in wound healing by upregulating Trem1 via the cAMP/PKA/CREB pathway. Int Immunopharmacol 2024; 128:111463. [PMID: 38190789 DOI: 10.1016/j.intimp.2023.111463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/22/2023] [Accepted: 12/27/2023] [Indexed: 01/10/2024]
Abstract
BACKGROUND Inflammation is an important part of the wound healing process. The stress hormone epinephrine has been demonstrated to modulate the inflammatory response via its interaction with β2-adrenergic receptor (β2-AR). However, the precise molecular mechanism through which β2-AR exerts its influence on inflammation during the wound healing process remains an unresolved question. METHODS Transcriptome datasets of wound and macrophages from the GEO database were reanalyzed using bioinformatics. The role of β2-AR in wound healing was explored by a mouse hind paw plantar wound model, and histological analyses were performed to assess wound healing. In vivo and in vitro assays were performed to elucidate the role of β2-AR on the inflammatory response. Triggering receptor expressed on myeloid cells 1 (Trem1) was knocked down with siRNA on RAW cells and western blot and qPCR assays were performed. RESULTS Trem1 was upregulated within 24 h of wounding, and macrophage β2-AR activation also upregulated Trem1. In vivo experiments demonstrated that β2-AR agonists impaired wound healing, accompanied by upregulation of Trem1 and activation of cAMP/PKA/CREB pathway, as well as by a high level of pro-inflammatory cytokine production. In vitro experiments showed that macrophage β2-AR activation amplified LPS-induced inflammation, and knockdown of Trem1 reversed this effect. Using activator and inhibitor of cAMP, macrophage β2-AR activation was confirmed to upregulate Trem1 via the cAMP/PKA/CREB pathway. CONCLUSION Our study found that β2-AR agonists increase Trem1 expression in wounds, accompanied by amplification of the inflammatory response, impairing wound healing. β2-AR activation in RAW cells induces Trem1 upregulation via the cAMP/PKA/CREB pathway and amplifies LPS-induced inflammatory responses.
Collapse
Affiliation(s)
- Huawei Wei
- Department of Anesthesiology, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Mengqiu Deng
- Department of Anesthesiology, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Ruifeng Ding
- Department of Anesthesiology, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Liangtian Wei
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Hongbin Yuan
- Department of Anesthesiology, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China.
| |
Collapse
|
16
|
Dong TS, Mayer E. Advances in Brain-Gut-Microbiome Interactions: A Comprehensive Update on Signaling Mechanisms, Disorders, and Therapeutic Implications. Cell Mol Gastroenterol Hepatol 2024; 18:1-13. [PMID: 38336171 PMCID: PMC11126987 DOI: 10.1016/j.jcmgh.2024.01.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024]
Abstract
The complex, bidirectional interactions between the brain, the gut, and the gut microbes are best referred to as the brain gut microbiome system. Animal and clinical studies have identified specific signaling mechanisms within this system, with gut microbes communicating to the brain through neuronal, endocrine, and immune pathways. The brain, in turn, modulates the composition and function of the gut microbiota through the autonomic nervous system, regulating gut motility, secretion, permeability, and the release of hormones impacting microbial gene expression. Perturbations at any level of these interactions can disrupt the intricate balance, potentially contributing to the pathogenesis of intestinal, metabolic, neurologic, and psychiatric disorders. Understanding these interactions and their underlying mechanisms holds promise for identifying biomarkers, as well as novel therapeutic targets, and for developing more effective treatment strategies for these complex disorders. Continued research will advance our knowledge of this system, with the potential for improved understanding and management of a wide range of disorders. This review provides an update on the current state of knowledge regarding this system, with a focus on recent advancements and emerging research areas.
Collapse
Affiliation(s)
- Tien S Dong
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California Los Angeles, Los Angeles, California; Goodman-Luskin Microbiome Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California; The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Emeran Mayer
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California Los Angeles, Los Angeles, California; Goodman-Luskin Microbiome Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California; The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.
| |
Collapse
|
17
|
Kwon J, Kim DY, Cho KJ, Hashimoto M, Matsuoka K, Kamijo T, Wang Z, Karnup S, Robertson AM, Tyagi P, Yoshimura N. Pathophysiology of Overactive Bladder and Pharmacologic Treatments Including β3-Adrenoceptor Agonists -Basic Research Perspectives. Int Neurourol J 2024; 28:12-33. [PMID: 38461853 DOI: 10.5213/inj.2448002.001] [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: 12/31/2023] [Accepted: 01/10/2024] [Indexed: 03/12/2024] Open
Abstract
Overactive bladder (OAB) is a symptom-based syndrome defined by urinary urgency, frequency, and nocturia with or without urge incontinence. The causative pathology is diverse; including bladder outlet obstruction (BOO), bladder ischemia, aging, metabolic syndrome, psychological stress, affective disorder, urinary microbiome, localized and systemic inflammatory responses, etc. Several hypotheses have been suggested as mechanisms of OAB generation; among them, neurogenic, myogenic, and urothelial mechanisms are well-known hypotheses. Also, a series of local signals called autonomous myogenic contraction, micromotion, or afferent noises, which can occur during bladder filling, may be induced by the leak of acetylcholine (ACh) or urothelial release of adenosine triphosphate (ATP). They can be transmitted to the central nervous system through afferent fibers to trigger coordinated urgency-related detrusor contractions. Antimuscarinics, commonly known to induce smooth muscle relaxation by competitive blockage of muscarinic receptors in the parasympathetic postganglionic nerve, have a minimal effect on detrusor contraction within therapeutic doses. In fact, they have a predominant role in preventing signals in the afferent nerve transmission process. β3-adrenergic receptor (AR) agonists inhibit afferent signals by predominant inhibition of mechanosensitive Aδ-fibers in the normal bladder. However, in pathologic conditions such as spinal cord injury, it seems to inhibit capsaicin-sensitive C-fibers. Particularly, mirabegron, a β3-agonist, prevents ACh release in the BOO-induced detrusor overactivity model by parasympathetic prejunctional mechanisms. A recent study also revealed that vibegron may have 2 mechanisms of action: inhibition of ACh from cholinergic efferent nerves in the detrusor and afferent inhibition via urothelial β3-AR.
Collapse
Affiliation(s)
- Joonbeom Kwon
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Leaders Urology Clinic, Daegu, Korea
| | - Duk Yoon Kim
- Department of Urology, Catholic University of Daegu School of Medicine, Daegu, Korea
| | - Kang Jun Cho
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Urology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Mamoru Hashimoto
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kanako Matsuoka
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Tadanobu Kamijo
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Zhou Wang
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sergei Karnup
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Anne M Robertson
- Department of Mechanical Engineering and Materials Science, University of Pittsburgh School of Bioengineering, Pittsburgh, PA, USA
| | - Pradeep Tyagi
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Naoki Yoshimura
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
18
|
Weickert TW, Jacomb I, Lenroot R, Lappin J, Weinberg D, Brooks WS, Brown D, Pellen D, Kindler J, Mohan A, Wakefield D, Lloyd AR, Stanton C, O'Donnell M, Liu D, Galletly C, Shannon Weickert C. Adjunctive canakinumab reduces peripheral inflammation markers and improves positive symptoms in people with schizophrenia and inflammation: A randomized control trial. Brain Behav Immun 2024; 115:191-200. [PMID: 37848096 DOI: 10.1016/j.bbi.2023.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 10/11/2023] [Accepted: 10/14/2023] [Indexed: 10/19/2023] Open
Abstract
BACKGROUND Clinical trials of anti-inflammatories in schizophrenia do not show clear and replicable benefits, possibly because patients were not recruited based on elevated inflammation status. Interleukin 1-beta (IL-1β) mRNA and protein levels are increased in serum, plasma, cerebrospinal fluid, and brain of some chronically ill patients with schizophrenia, first episode psychosis, and clinical high-risk individuals. Canakinumab, an approved anti-IL-1β monoclonal antibody, interferes with the bioactivity of IL-1β and interrupts downstream signaling. However, the extent to which canakinumab reduces peripheral inflammation markers, such as, high sensitivity C-reactive protein (hsCRP) and symptom severity in schizophrenia patients with inflammation is unknown. TRIAL DESIGN We conducted a randomized, placebo-controlled, double-blind, parallel groups, 8-week trial of canakinumab in chronically ill patients with schizophrenia who had elevated peripheral inflammation. METHODS Twenty-seven patients with schizophrenia or schizoaffective disorder and elevated peripheral inflammation markers (IL-1β, IL-6, hsCRP and/or neutrophil to lymphocyte ratio: NLR) were randomized to a one-time, subcutaneous injection of canakinumab (150 mg) or placebo (normal saline) as an adjunctive antipsychotic treatment. Peripheral blood hsCRP, NLR, IL-1β, IL-6, IL-8 levels were measured at baseline (pre injection) and at 1-, 4- and 8-weeks post injection. Symptom severity was assessed at baseline and 4- and 8-weeks post injection. RESULTS Canakinumab significantly reduced peripheral hsCRP over time, F(3, 75) = 5.16, p = 0.003. Significant hsCRP reductions relative to baseline were detected only in the canakinumab group at weeks 1, 4 and 8 (p's = 0.0003, 0.000002, and 0.004, respectively). There were no significant hsCRP changes in the placebo group. Positive symptom severity scores were significantly reduced at week 8 (p = 0.02) in the canakinumab group and week 4 (p = 0.02) in the placebo group. The change in CRP between week 8 and baseline (b = 1.9, p = 0.0002) and between week 4 and baseline (b = 6.0, p = 0.001) were highly significant predictors of week 8 change in PANSS Positive Symptom severity scores. There were no significant changes in negative symptoms, general psychopathology or cognition in either group. Canakinumab was well tolerated and only 7 % discontinued. CONCLUSIONS Canakinumab quickly reduces peripheral hsCRP serum levels in patients with schizophrenia and inflammation; after 8 weeks of canakinumab treatment, the reductions in hsCRP are related to reduced positive symptom severity. Future studies should consider increased doses or longer-term treatment to confirm the potential benefits of adjunctive canakinumab in schizophrenia. Australian and New Zealand Clinical Trials Registry number: ACTRN12615000635561.
Collapse
Affiliation(s)
- Thomas W Weickert
- Neuroscience Research Australia, Sydney, New South Wales, Australia; School of Psychiatry and Mental Health, University of New South Wales, Sydney, New South Wales, Australia.
| | - Isabella Jacomb
- Neuroscience Research Australia, Sydney, New South Wales, Australia
| | - Rhoshel Lenroot
- Neuroscience Research Australia, Sydney, New South Wales, Australia; School of Psychiatry and Mental Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Julia Lappin
- School of Psychiatry and Mental Health, University of New South Wales, Sydney, New South Wales, Australia
| | | | - William S Brooks
- Neuroscience Research Australia, Sydney, New South Wales, Australia; School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - David Brown
- NSW Health Pathology-ICPMR, Centre for Immunology and Allergy Research, Westmead Institute for Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Daniel Pellen
- Neuroscience Research Australia, Sydney, New South Wales, Australia
| | - Jochen Kindler
- Neuroscience Research Australia, Sydney, New South Wales, Australia; University Hospital of Child and Adolescent Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
| | - Adith Mohan
- School of Psychiatry and Mental Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Denis Wakefield
- School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Andrew R Lloyd
- Viral Immunology Systems Program, Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Clive Stanton
- Neuroscience Research Australia, Sydney, New South Wales, Australia; Prince of Wales Hospital, Sydney, New South Wales, Australia
| | - Maryanne O'Donnell
- Neuroscience Research Australia, Sydney, New South Wales, Australia; School of Psychiatry and Mental Health, University of New South Wales, Sydney, New South Wales, Australia; Prince of Wales Hospital, Sydney, New South Wales, Australia
| | - Dennis Liu
- Discipline of Psychiatry, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Northern Adelaide Locah Health Network, Adelaide, South Australia, Australia
| | - Cherrie Galletly
- Discipline of Psychiatry, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Northern Adelaide Locah Health Network, Adelaide, South Australia, Australia
| | - Cynthia Shannon Weickert
- Neuroscience Research Australia, Sydney, New South Wales, Australia; School of Psychiatry and Mental Health, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
19
|
Zheng H, Zhang C, Zhang J, Duan L. "Sentinel or accomplice": gut microbiota and microglia crosstalk in disorders of gut-brain interaction. Protein Cell 2023; 14:726-742. [PMID: 37074139 PMCID: PMC10599645 DOI: 10.1093/procel/pwad020] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/06/2023] [Indexed: 04/20/2023] Open
Abstract
Abnormal brain-gut interaction is considered the core pathological mechanism behind the disorders of gut-brain interaction (DGBI), in which the intestinal microbiota plays an important role. Microglia are the "sentinels" of the central nervous system (CNS), which participate in tissue damage caused by traumatic brain injury, resist central infection and participate in neurogenesis, and are involved in the occurrence of various neurological diseases. With in-depth research on DGBI, we could find an interaction between the intestinal microbiota and microglia and that they are jointly involved in the occurrence of DGBI, especially in individuals with comorbidities of mental disorders, such as irritable bowel syndrome (IBS). This bidirectional regulation of microbiota and microglia provides a new direction for the treatment of DGBI. In this review, we focus on the role and underlying mechanism of the interaction between gut microbiota and microglia in DGBI, especially IBS, and the corresponding clinical application prospects and highlight its potential to treat DGBI in individuals with psychiatric comorbidities.
Collapse
Affiliation(s)
- Haonan Zheng
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing 100191, China
| | - Cunzheng Zhang
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing 100191, China
| | - Jindong Zhang
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing 100191, China
| | - Liping Duan
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing 100191, China
| |
Collapse
|
20
|
Zhang A, Liu Y, Wang X, Xu H, Fang C, Yuan L, Wang K, Zheng J, Qi Y, Chen S, Zhang J, Shao A. Clinical Potential of Immunotherapies in Subarachnoid Hemorrhage Treatment: Mechanistic Dissection of Innate and Adaptive Immune Responses. Aging Dis 2023; 14:1533-1554. [PMID: 37196120 PMCID: PMC10529760 DOI: 10.14336/ad.2023.0126] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 01/26/2023] [Indexed: 05/19/2023] Open
Abstract
Subarachnoid hemorrhage (SAH), classified as a medical emergency, is a devastating and severe subtype of stroke. SAH induces an immune response, which further triggers brain injury; however, the underlying mechanisms need to be further elucidated. The current research is predominantly focused on the production of specific subtypes of immune cells, especially innate immune cells, post-SAH onset. Increasing evidence suggests the critical role of immune responses in SAH pathophysiology; however, studies on the role and clinical significance of adaptive immunity post-SAH are limited. In this present study, we briefly review the mechanistic dissection of innate and adaptive immune responses post-SAH. Additionally, we summarized the experimental studies and clinical trials of immunotherapies for SAH treatment, which may form the basis for the development of improved therapeutic approaches for the clinical management of SAH in the future.
Collapse
Affiliation(s)
- Anke Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China.
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China.
| | - Yibo Liu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China.
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China.
| | - Xiaoyu Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China.
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China.
| | - Houshi Xu
- Department of Neurosurgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Chaoyou Fang
- Department of Neurosurgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Ling Yuan
- Department of Neurosurgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - KaiKai Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China.
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China.
| | - Jingwei Zheng
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China.
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China.
| | - Yangjian Qi
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China.
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China.
| | - Sheng Chen
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China.
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China.
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China.
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China.
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China.
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
21
|
Waschmann M, Stuart A, Trieschmann K, Lin HC, Hunter AK. Assessing the Impact of the COVID-19 Pandemic on the Severity of Pediatric Inflammatory Bowel Disease Admissions and New Diagnoses. CROHN'S & COLITIS 360 2023; 5:otad062. [PMID: 37941600 PMCID: PMC10629215 DOI: 10.1093/crocol/otad062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Indexed: 11/10/2023] Open
Abstract
Introduction The COVID-19 pandemic has introduced new challenges to the diagnosis and management of pediatric inflammatory bowel disease (IBD). Many patients have had only limited access to their providers through telemedicine, and many chose to delay nonemergent treatment. Methods A retrospective chart review of patients with IBD seen by the Pediatric Gastroenterology Division at Doernbecher Children's Hospital from January 2018 to August 2021 was conducted. The study cohort was divided into 2 groups: those presenting before the onset of the COVID-19 pandemic (January 1, 2018 to February 28, 2020) and those presenting during the pandemic (March 1, 2020 to August 1, 2021). Variables collected included: age, sex, race, ethnicity, IBD type, insurance type, location of residence. Primary outcome measures selected focused on disease severity, initial type of treatment, or surgical intervention offered. A subgroup analysis of the new diagnosis patients was performed. Data were analyzed using independent t-tests, chi-squared analysis, and Wilcoxon rank sum tests. Results Two hundred and eleven patients met inclusion criteria, 107 (72 new diagnoses, 35 admissions) within the pre-COVID epoch and 104 (67 new diagnoses, 37 admissions) within the during-COVID epoch. Patients in the during-COVID epoch had higher fecal calprotectin level and were more likely to be started on a biologic as initial treatment. Patients admitted during COVID for IBD flare were more likely to require surgical intervention. Subgroup analysis of newly diagnosed patients revealed higher incidence of comorbid depression and anxiety. Conclusions Our review identified increased disease severity in newly diagnosed pediatric patients with IBD as well as pediatric patients admitted for flare during COVID. Increases in anxiety and depression rates during COVID may have contributed to worsened disease severity.
Collapse
Affiliation(s)
- Malika Waschmann
- Department of Pediatrics, University of Washington, Seattle Children’s Hospital, Seattle, WA, USA
| | - Ariana Stuart
- School of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Kimberly Trieschmann
- Department of Pediatrics, University of Washington, Seattle Children’s Hospital, Seattle, WA, USA
- Division of Pediatric Gastroenterology, Doernbecher Children’s Hospital, Portland, OR, USA
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
| | - Henry C Lin
- Division of Pediatric Gastroenterology, Doernbecher Children’s Hospital, Portland, OR, USA
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
| | - Anna K Hunter
- Division of Pediatric Gastroenterology, Doernbecher Children’s Hospital, Portland, OR, USA
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
22
|
Tong RL, Kahn UN, Grafe LA, Hitti FL, Fried NT, Corbett BF. Stress circuitry: mechanisms behind nervous and immune system communication that influence behavior. Front Psychiatry 2023; 14:1240783. [PMID: 37706039 PMCID: PMC10495591 DOI: 10.3389/fpsyt.2023.1240783] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/16/2023] [Indexed: 09/15/2023] Open
Abstract
Inflammatory processes are increased by stress and contribute to the pathology of mood disorders. Stress is thought to primarily induce inflammation through peripheral and central noradrenergic neurotransmission. In healthy individuals, these pro-inflammatory effects are countered by glucocorticoid signaling, which is also activated by stress. In chronically stressed individuals, the anti-inflammatory effects of glucocorticoids are impaired, allowing pro-inflammatory effects to go unchecked. Mechanisms underlying this glucocorticoid resistance are well understood, but the precise circuits and molecular mechanisms by which stress increases inflammation are not as well known. In this narrative review, we summarize the mechanisms by which chronic stress increases inflammation and contributes to the onset and development of stress-related mood disorders. We focus on the neural substrates and molecular mechanisms, especially those regulated by noradrenergic signaling, that increase inflammatory processes in stressed individuals. We also discuss key knowledge gaps in our understanding of the communication between nervous and immune systems during stress and considerations for future therapeutic strategies. Here we highlight the mechanisms by which noradrenergic signaling contributes to inflammatory processes during stress and how this inflammation can contribute to the pathology of stress-related mood disorders. Understanding the mechanisms underlying crosstalk between the nervous and immune systems may lead to novel therapeutic strategies for mood disorders and/or provide important considerations for treating immune-related diseases in individuals suffering from stress-related disorders.
Collapse
Affiliation(s)
- Rose L. Tong
- Corbett Laboratory, Department of Biology, Rutgers University, Camden, NJ, United States
| | - Ubaidah N. Kahn
- Fried Laboratory, Department of Biology, Rutgers University, Camden, NJ, United States
| | - Laura A. Grafe
- Grafe Laboratory, Department of Psychology, Bryn Mawr College, Bryn Mawr, PA, United States
| | - Frederick L. Hitti
- Hitti Laboratory, Department of Neurological Surgery and Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Nathan T. Fried
- Fried Laboratory, Department of Biology, Rutgers University, Camden, NJ, United States
| | - Brian F. Corbett
- Corbett Laboratory, Department of Biology, Rutgers University, Camden, NJ, United States
| |
Collapse
|
23
|
Chaudhry TS, Senapati SG, Gadam S, Mannam HPSS, Voruganti HV, Abbasi Z, Abhinav T, Challa AB, Pallipamu N, Bheemisetty N, Arunachalam SP. The Impact of Microbiota on the Gut-Brain Axis: Examining the Complex Interplay and Implications. J Clin Med 2023; 12:5231. [PMID: 37629273 PMCID: PMC10455396 DOI: 10.3390/jcm12165231] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/30/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
The association and interaction between the central nervous system (CNS) and enteric nervous system (ENS) is well established. Essentially ENS is the second brain, as we call it. We tried to understand the structure and function, to throw light on the functional aspect of neurons, and address various disease manifestations. We summarized how various neurological disorders influence the gut via the enteric nervous system and/or bring anatomical or physiological changes in the enteric nervous system or the gut and vice versa. It is known that stress has an effect on Gastrointestinal (GI) motility and causes mucosal erosions. In our literature review, we found that stress can also affect sensory perception in the central nervous system. Interestingly, we found that mutations in the neurohormone, serotonin (5-HT), would result in dysfunctional organ development and further affect mood and behavior. We focused on the developmental aspects of neurons and cognition and their relation to nutritional absorption via the gastrointestinal tract, the development of neurodegenerative disorders in relation to the alteration in gut microbiota, and contrariwise associations between CNS disorders and ENS. This paper further summarizes the synergetic relation between gastrointestinal and neuropsychological manifestations and emphasizes the need to include behavioral therapies in management plans.
Collapse
Affiliation(s)
| | | | - Srikanth Gadam
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (S.G.); (N.P.)
| | - Hari Priya Sri Sai Mannam
- GIH Artificial Intelligence Laboratory (GAIL), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (H.P.S.S.M.); (H.V.V.); (Z.A.); (T.A.); (N.B.)
| | - Hima Varsha Voruganti
- GIH Artificial Intelligence Laboratory (GAIL), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (H.P.S.S.M.); (H.V.V.); (Z.A.); (T.A.); (N.B.)
| | - Zainab Abbasi
- GIH Artificial Intelligence Laboratory (GAIL), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (H.P.S.S.M.); (H.V.V.); (Z.A.); (T.A.); (N.B.)
| | - Tushar Abhinav
- GIH Artificial Intelligence Laboratory (GAIL), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (H.P.S.S.M.); (H.V.V.); (Z.A.); (T.A.); (N.B.)
| | | | - Namratha Pallipamu
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (S.G.); (N.P.)
| | - Niharika Bheemisetty
- GIH Artificial Intelligence Laboratory (GAIL), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (H.P.S.S.M.); (H.V.V.); (Z.A.); (T.A.); (N.B.)
| | - Shivaram P. Arunachalam
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (S.G.); (N.P.)
- GIH Artificial Intelligence Laboratory (GAIL), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (H.P.S.S.M.); (H.V.V.); (Z.A.); (T.A.); (N.B.)
- Microwave Engineering and Imaging Laboratory (MEIL), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
24
|
Simon CB, Bishop MD, Wallace MR, Staud R, DelRocco N, Wu SS, Dai Y, Borsa PA, Greenfield WH, Fillingim RB, George SZ. Circulating Inflammatory Biomarkers Predict Pain Change Following Exercise-Induced Shoulder Injury: Findings From the Biopsychosocial Influence on Shoulder Pain Preclinical Trial. THE JOURNAL OF PAIN 2023; 24:1465-1477. [PMID: 37178095 PMCID: PMC10523877 DOI: 10.1016/j.jpain.2023.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/03/2023] [Accepted: 04/02/2023] [Indexed: 05/15/2023]
Abstract
Shoulder pain is a highly prevalent musculoskeletal condition that frequently leads to suboptimal clinical outcomes. This study tested the extent to which circulating inflammatory biomarkers are associated with reports of shoulder pain and upper-extremity disability for a high-risk genetic by psychological subgroup (catechol-O-methyltransferase [COMT] variation by pain catastrophizing [PCS]). Pain-free adults meeting high-risk COMT × PCS subgroup criteria completed an exercise-induced muscle injury protocol. Thirteen biomarkers were collected and analyzed from plasma 48 hours after muscle injury. Shoulder pain intensity and disability (Quick-DASH) were reported at 48 and 96 hours to calculate change scores. Using an extreme sampling technique, 88 participants were included in this analysis. After controlling for age, sex, and BMI, there were moderate positive associations between higher c-reactive protein (CRP; βˆ = .62; 95% confidence interval [CI] = -.03, 1.26), interleukin-6 (IL-6; βˆ = 3.13; CI = -.11, 6.38), and interleukin-10 (IL-10; βˆ = 2.51; CI = -.30, 5.32); and greater pain reduction from 48 to 96 hours post exercise muscle injury. Using an exploratory multivariable model to predict pain changes from 48 to 96 hours, we found participants with higher IL-10 were less likely to experience a high increase in pain (βˆ = -10.77; CI = -21.25, -2.69). Study findings suggest CRP, IL-6, and IL-10 are related to shoulder pain change for a preclinical high-risk COMT × PCS subgroup. Future studies will translate to clinical shoulder pain and decipher the complex and seemingly pleiotropic interplay between inflammatory biomarkers and shoulder pain change. PERSPECTIVE: In a preclinical high-risk COMT × PCS subgroup, 3 circulating inflammatory biomarkers (CRP, IL-6, and IL-10) were moderately associated with pain improvement following exercise-induced muscle injury.
Collapse
Affiliation(s)
- Corey B Simon
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, North Carolina; Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina.
| | - Mark D Bishop
- Department of Physical Therapy, University of Florida, Gainesville, Florida
| | - Margaret R Wallace
- Department of Molecular Genetics and Microbiology, UF Genetics Institute, University of Florida, Gainesville, Florida
| | - Roland Staud
- Department of Medicine, University of Florida, Gainesville, Florida
| | - Natalie DelRocco
- Department of Biostatistics, University of Florida, Gainesville, Florida
| | - Samuel S Wu
- Department of Biostatistics, University of Florida, Gainesville, Florida
| | - Yunfeng Dai
- Department of Biostatistics, University of Florida, Gainesville, Florida
| | - Paul A Borsa
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida
| | | | - Roger B Fillingim
- Pain Research & Intervention Center of Excellence, University of Florida, Gainesville, Florida; Department of Community Dentistry and Behavioral Science, University of Florida, Gainesville, Florida
| | - Steven Z George
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, North Carolina; Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina
| |
Collapse
|
25
|
Rodríguez-Palma EJ, Velazquez-Lagunas I, Salinas-Abarca AB, Vidal-Cantú GC, Escoto-Rosales MJ, Castañeda-Corral G, Fernández-Guasti A, Granados-Soto V. Spinal alarmin HMGB1 and the activation of TLR4 lead to chronic stress-induced nociceptive hypersensitivity in rodents. Eur J Pharmacol 2023:175804. [PMID: 37244377 DOI: 10.1016/j.ejphar.2023.175804] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/11/2023] [Accepted: 05/22/2023] [Indexed: 05/29/2023]
Abstract
Chronic stress affects millions of people around the world, and it can trigger different behavioral disorders like nociceptive hypersensitivity and anxiety, among others. However, the mechanisms underlaying these chronic stress-induced behavioral disorders have not been yet elucidated. This study was designed to understand the role of high-mobility group box-1 (HMGB1) and toll-like receptor 4 (TLR4) in chronic stress-induced nociceptive hypersensitivity. Chronic restraint stress induced bilateral tactile allodynia, anxiety-like behaviors, phosphorylation of ERK and p38MAPK and activation of spinal microglia. Moreover, chronic stress enhanced HMGB1 and TLR4 protein expression at the dorsal root ganglion, but not at the spinal cord. Intrathecal injection of HMGB1 or TLR4 antagonists reduced tactile allodynia and anxiety-like behaviors induced by chronic stress. Additionally, deletion of TLR4 diminished the establishment of chronic stress-induced tactile allodynia in male and female mice. Lastly, the antiallodynic effect of HMGB1 and TLR4 antagonists were similar in stressed male and female rats and mice. Our results suggest that chronic restraint stress induces nociceptive hypersensitivity, anxiety-like behaviors, and up-regulation of spinal HMGB1 and TLR4 expression. Blockade of HMGB1 and TLR4 reverses chronic restraint stress-induced nociceptive hypersensitivity and anxiety-like behaviors and restores altered HMGB1 and TLR4 expression. The antiallodynic effects of HMGB1 and TLR4 blockers in this model are sex independent. TLR4 could be a potential pharmacological target for the treatment of the nociceptive hypersensitivity associated with widespread chronic pain.
Collapse
Affiliation(s)
- Erick J Rodríguez-Palma
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| | - Isabel Velazquez-Lagunas
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| | - Ana Belen Salinas-Abarca
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| | - Guadalupe C Vidal-Cantú
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| | - María J Escoto-Rosales
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| | | | | | - Vinicio Granados-Soto
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico.
| |
Collapse
|
26
|
Hopkins S, Kelley T, Roller R, Thompson RS, Colagiovanni DB, Chupka K, Fleshner M. Oral CBD-rich hemp extract modulates sterile inflammation in female and male rats. Front Physiol 2023; 14:1112906. [PMID: 37275221 PMCID: PMC10234154 DOI: 10.3389/fphys.2023.1112906] [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: 11/30/2022] [Accepted: 04/26/2023] [Indexed: 06/07/2023] Open
Abstract
Introduction: Cannabidiol (CBD) extract from the cannabis plant has biomedical and nutraceutical potential. Unlike tetrahydrocannabinol (THC), CBD products produce few psychoactive effects and pose little risk for abuse. There is emerging preclinical and clinical evidence that CBD is stress modulatory and may have anti-inflammatory properties. People across the United States legally ingest CBD-rich hemp extracts to manage mental and physical health problems, including stress and inflammation. Preclinical studies have revealed potential mechanisms for these effects; however, the impact of this prior work is diminished because many studies: 1) tested synthetic CBD rather than CBD-rich hemp extracts containing terpenes and/or other cannabinoids thought to enhance therapeutic benefits; 2) administered CBD via injection into the peritoneal cavity or the brain instead of oral ingestion; and 3) failed to examine potential sex differences. To address these gaps in the literature, the following study tested the hypothesis that the voluntary oral ingestion of CBD-rich hemp extract will attenuate the impact of stressor exposure on plasma and tissue inflammatory and stress proteins in females and males. Methods: Adult male and female Sprague Dawley rats (10-15/group) were randomly assigned to be given cereal coated with either vehicle (coconut oil) or CBD-rich hemp extract (L-M0717, CBDrx/Functional Remedies, 20.0 mg/kg). After 7 days, rats were exposed to a well-established acute model of stress (100, 1.5 mA, 5-s, intermittent tail shocks, 90 min total duration) or remained in home cages as non-stressed controls. Results: Stressor exposure induced a robust stress response, i.e., increased plasma corticosterone and blood glucose, and decreased spleen weight (a surrogate measure of sympathetic nervous system activation). Overall, stress-induced increases in inflammatory and stress proteins were lower in females than males, and oral CBD-rich hemp extract constrained these responses in adipose tissue (AT) and mesenteric lymph nodes (MLN). Consistent with previous reports, females had higher levels of stress-evoked corticosterone compared to males, which may have contributed to the constrained inflammatory response measured in females. Discussion: Results from this study suggest that features of the acute stress response are impacted by oral ingestion of CBD-rich hemp extract in female and male rats, and the pattern of changes may be sex and tissue dependent.
Collapse
Affiliation(s)
- Shelby Hopkins
- Department of Integrative Physiology, University of Colorado at Boulder, Boulder, CO, United States
- Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, United States
| | - Tel Kelley
- Department of Integrative Physiology, University of Colorado at Boulder, Boulder, CO, United States
| | - Rachel Roller
- Department of Integrative Physiology, University of Colorado at Boulder, Boulder, CO, United States
| | - Robert S. Thompson
- Department of Integrative Physiology, University of Colorado at Boulder, Boulder, CO, United States
- Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, United States
| | | | - Kris Chupka
- Next Frontier Biosciences, Westminster, CO, United States
| | - Monika Fleshner
- Department of Integrative Physiology, University of Colorado at Boulder, Boulder, CO, United States
- Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, United States
| |
Collapse
|
27
|
Nguyen TV, Yamanaka K, Tomita K, Zubcevic J, Gouraud SSS, Waki H. Impact of exercise on brain-bone marrow interactions in chronic stress: potential mechanisms preventing stress-induced hypertension. Physiol Genomics 2023; 55:222-234. [PMID: 36939204 PMCID: PMC10151049 DOI: 10.1152/physiolgenomics.00168.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/15/2023] [Accepted: 03/15/2023] [Indexed: 03/21/2023] Open
Abstract
We examined the effect of chronic restraint stress and the counteractive effects of daily exercise on the molecular basis of the brain-bone marrow (BM) interactions, by especially focusing on the paraventricular nucleus (PVN) of the hypothalamus. Male Wistar rats were assigned into control, restraint stress, and stress + daily spontaneous exercise (SE) groups. BM and hypothalamic gene expression profiles were examined through the undertaking of RT-PCR and microarrays, respectively. The inflammatory blood cell population was investigated through flow cytometry. Through the use of immunohistochemistry, we examined the presence of BM-derived C-C chemokine receptor type 2 (CCR2)-expressing microglial cells in the rat PVN. The gene expression levels of BM inflammatory factors such as those of interleukin 1 beta and CCR2, and the inflammatory blood cell population were found to be significantly higher in both restrained groups compared with control group. Interestingly, chronic restraint stress alone activated the recruitment of BM-derived CCR2-expressing microglial cells into the PVN, whereas daily spontaneous exercise prevented it. A notable finding was that restraint stress upregulated relative gene expression of hypothalamic matrix metalloproteinase 3 (MMP3), which increases the permeability of the blood-brain barrier (BBB), and that exercise managed to normalize it. Moreover, relative expression of some hypothalamic genes directly involved in the facilitation of cell migration was downregulated by daily exercise. Our findings suggest that daily spontaneous exercise can reduce the numbers of BM-derived CCR2-expressing microglial cells into the PVN through the prevention of stress-induced changes in the hypothalamic gene expression.NEW & NOTEWORTHY Chronic restraint stress can upregulate MMP3 gene expression in the rat hypothalamus, whereas daily spontaneous exercise can prevent this stress-induced effect. Stress-induced BM-derived inflammatory cell recruitment into the rat PVN can be prevented by daily spontaneous exercise. Stress-induced increase of hypothalamic MMP3 gene expression may be responsible for BBB injury, thereby allowing for BM-derived inflammatory cells to be recruited and to accumulate in the rat PVN, and to be subsequently involved in the onset of stress-induced hypertension.
Collapse
Affiliation(s)
- Thu Van Nguyen
- Department of Physiology, Graduate School of Health and Sports Science, Juntendo University, Chiba, Japan
- Department of Military Occupational Medicine, Vietnam Military Medical University, Hanoi, Vietnam
| | - Ko Yamanaka
- Department of Physiology, Graduate School of Health and Sports Science, Juntendo University, Chiba, Japan
| | - Keisuke Tomita
- Department of Physiology, Graduate School of Health and Sports Science, Juntendo University, Chiba, Japan
| | - Jasenka Zubcevic
- Department of Physiology and Pharmacology, University of Toledo, Toledo, Ohio, United States
| | - Sabine S S Gouraud
- College of Liberal Arts, International Christian University, Tokyo, Japan
| | - Hidefumi Waki
- Department of Physiology, Graduate School of Health and Sports Science, Juntendo University, Chiba, Japan
| |
Collapse
|
28
|
Banday UZ, Nazir A, Fatima M, Swaleh SB, Al-Asadi M, Usmani N. Heavy metal-induced genotoxic, physiological, and biochemical responses in Schizothorax esocinus (Heckel 1838) inhabiting the Dal Lake, India, and phytoremediation by indwelling plants. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:65688-65701. [PMID: 37086321 DOI: 10.1007/s11356-023-27064-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 04/12/2023] [Indexed: 05/03/2023]
Abstract
Heavy metals have an immense impact on aquatic ecosystems, and their toxic effects are transferred to the inhabiting organisms. Experiments were conducted to investigate the health of snow trout Schizothorax esocinus inhabiting Dal Lake. Heavy metals (Cd > Ni > Cu > Cr) were found to accumulate in the major immune organs of the fish (head kidney, liver, spleen, thymus) which led to change in the overall physiology. The head kidney, liver, and spleen of a fish contain high amount of these metals. The least accumulation of these metals was found in the blood, whereas Cd and Ni were completely absent in the integument. Hepatic marker enzymes (aspartate aminotransferase (AST), alanine aminotransferase (ALT), and alkaline phosphatase (ALP)) were normal, while the renal marker enzyme creatine kinase showed marked difference in its value. The cortisol level was normal, while immunoglobulin M showed elevated level representing active immunity. At a cellular level, the histopathology of immune organs showed marked damage. Metallothionein (MT) and glutathione peroxidase (GPX) genes showed variable expression pattern in the immune organs with the head kidney showing the highest expression of both the genes, and blood showed the least. We observed that the aquatic plants (Nelumbo nucifera and Trapa natans) inhabiting the lake played an important role in phytoremediation. An integrated approach involving biochemical, hematological, genotoxic, and histopathological studies can provide a valuable information to understand fish adaptive patterns and monitor water quality.
Collapse
Affiliation(s)
- Umarah Zahoor Banday
- Aquatic Toxicology Laboratory, Department of Zoology, Aligarh Muslim University, Aligarh, 202002, India.
| | - Aafaq Nazir
- Interdisciplinary Centre for Water Research, Indian Institute of Science, Bengaluru, 560 012, India
- Institute of Oceanography, National Taiwan University, Taipei, 10617, Taiwan
| | - Muizzah Fatima
- Aquatic Toxicology Laboratory, Department of Zoology, Aligarh Muslim University, Aligarh, 202002, India
| | - Sadiya Binte Swaleh
- Aquatic Toxicology Laboratory, Department of Zoology, Aligarh Muslim University, Aligarh, 202002, India
| | - Moneeb Al-Asadi
- Aquatic Toxicology Laboratory, Department of Zoology, Aligarh Muslim University, Aligarh, 202002, India
| | - Nazura Usmani
- Aquatic Toxicology Laboratory, Department of Zoology, Aligarh Muslim University, Aligarh, 202002, India
| |
Collapse
|
29
|
Beldowska A, Barszcz M, Dunislawska A. State of the art in research on the gut-liver and gut-brain axis in poultry. J Anim Sci Biotechnol 2023; 14:37. [PMID: 37038205 PMCID: PMC10088153 DOI: 10.1186/s40104-023-00853-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 02/12/2023] [Indexed: 04/12/2023] Open
Abstract
The relationship between the intestines and their microbiota, the liver, and the neuronal system is called the gut-liver-brain axis. This relationship has been studied and observed for a relatively short time but is considered in the development of research focused on, e.g., liver diseases and intestinal dysbiosis. The role of the gut microbiota in this relationship is crucial, as it acts on poultry's performance and feed utilization, affecting meat and egg quality. The correct composition of the intestinal microbiota makes it possible to determine the essential metabolic pathways and biological processes of the individual components of the microbiota, allowing further speculation of the role of microbial populations on internal organs such as the liver and brain in the organism. The gut microbiota forms a complex, dense axis with the autonomic and enteric nervous systems. The symbiotic relationship between the liver and gut microbiota is based on immune, metabolic and neuroendocrine regulation, and stabilization. On the other hand, the gut-brain axis is a bidirectional interaction and information transfer system between the gastrointestinal tract and the central nervous system. The following paper will discuss the current state of knowledge of the gut-liver-brain axis of poultry, including factors that may affect this complex relationship.
Collapse
Affiliation(s)
- Aleksandra Beldowska
- Department of Animal Biotechnology and Genetics, Bydgoszcz University of Science and Technology, Mazowiecka 28, Bydgoszcz, 85-084, Poland
| | - Marcin Barszcz
- Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3, Jabłonna, 05-110, Poland
| | - Aleksandra Dunislawska
- Department of Animal Biotechnology and Genetics, Bydgoszcz University of Science and Technology, Mazowiecka 28, Bydgoszcz, 85-084, Poland.
| |
Collapse
|
30
|
Virtanen MI, Brinchmann MF, Patel DM, Iversen MH. Chronic stress negatively impacts wound healing, welfare, and stress regulation in internally tagged Atlantic salmon (Salmo salar). Front Physiol 2023; 14:1147235. [PMID: 37078022 PMCID: PMC10106625 DOI: 10.3389/fphys.2023.1147235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/09/2023] [Indexed: 04/05/2023] Open
Abstract
The desire to understand fish welfare better has led to the development of live monitoring sensor tags embedded within individuals for long periods. Improving and understanding welfare must not come at the cost of impaired welfare due to a tag’s presence and implantation process. When welfare is compromised, the individual will experience negative emotions such as fear, pain, and distress, impacting the stress response. In this study, Atlantic salmon (Salmo salar) underwent surgical implantation of a dummy tag. Additionally, half of this group was introduced to daily crowding stress. Both groups and an untagged group were followed for 8 weeks using triplicate tanks per group. Sampling took place once a week, and where stress was given, it was conducted 24 h before sampling. Stress-related measurements were taken to understand if tagging caused chronic stress and explore the chronic stress response and its impact on wound healing. Primary stress response hormones measured included CRH, dopamine, adrenocorticotropic hormone, and cortisol. Secondary stress response parameters measured included glucose, lactate, magnesium, calcium, chloride, and osmolality. Tertiary stress response parameters measured included weight, length, and five fins for fin erosion. Wound healing was calculated by taking the incision length and width, the inflammation length and width, and the inside wound length and width. The wound healing process showed that stressed fish have a larger and longer-lasting inflammation period and a slower wound healing process, as seen from the inside wound. The tagging of Atlantic salmon did not cause chronic stress. In contrast, daily stress led to an allostatic overload type two response. ACTH was elevated in the plasma after 4 weeks, and cortisol followed elevation after 6 weeks, highlighting a breakdown of the stress regulation. Fin erosion was elevated alongside cortisol increase in the stressed group. This data suggests that tagging previously unstressed fish in a controlled environment does not negatively affect welfare regarding stress responses. It also indicates that stress delays wound healing and increases the inflammatory response, highlighting how continued stress causes a breakdown in some stress responses. Ultimately, the tagging of Atlantic salmon can be successful under certain conditions where proper healing is observed, tag retention is high, and chronic stress is not present, which could allow for the possible measurement of welfare indicators via smart-tags.
Collapse
|
31
|
Bartocci B, Dal Buono A, Gabbiadini R, Busacca A, Quadarella A, Repici A, Mencaglia E, Gasparini L, Armuzzi A. Mental Illnesses in Inflammatory Bowel Diseases: mens sana in corpore sano. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:medicina59040682. [PMID: 37109640 PMCID: PMC10145199 DOI: 10.3390/medicina59040682] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/25/2023] [Accepted: 03/28/2023] [Indexed: 04/29/2023]
Abstract
Background and aims: Inflammatory bowel diseases (IBD) are chronic disorders associated with a reduced quality of life, and patients often also suffer from psychiatric comorbidities. Overall, both mood and cognitive disorders are prevalent in chronic organic diseases, especially in the case of a strong immune component, such as rheumatoid arthritis, multiple sclerosis, and cancer. Divergent data regarding the true incidence and prevalence of mental disorders in patients with IBD are available. We aimed to review the current evidence on the topic and the burden of mental illness in IBD patients, the role of the brain-gut axis in their co-existence, and its implication in an integrated clinical management. Methods: PubMed was searched to identify relevant studies investigating the gut-brain interactions and the incidence and prevalence of psychiatric disorders, especially of depression, anxiety, and cognitive dysfunction in the IBD population. Results: Among IBD patients, there is a high prevalence of psychiatric comorbidities, especially of anxiety and depression. Approximately 20-30% of IBD patients are affected by mood disorders and/or present with anxiety symptoms. Furthermore, it has been observed that the prevalence of mental illnesses increases in patients with active intestinal disease. Psychiatric comorbidities continue to be under-diagnosed in IBD patients and remain an unresolved issue in the management of these patients. Conclusions: Psychiatric illnesses co-occurring in IBD patients deserve acknowledgment from IBD specialists. These comorbidities highly impact the management of IBD patients and should be studied as an adjunctive therapeutic target.
Collapse
Affiliation(s)
- Bianca Bartocci
- IBD Center, Humanitas Research Hospital-IRCCS, Via Manzoni 56, Rozzano, 20089 Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| | - Arianna Dal Buono
- IBD Center, Humanitas Research Hospital-IRCCS, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Roberto Gabbiadini
- IBD Center, Humanitas Research Hospital-IRCCS, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Anita Busacca
- IBD Center, Humanitas Research Hospital-IRCCS, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Alessandro Quadarella
- IBD Center, Humanitas Research Hospital-IRCCS, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Alessandro Repici
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| | - Emanuela Mencaglia
- Medical Oncology and Haematology Unit, Humanitas Cancer Center, Humanitas Research Hospital IRCCS, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Linda Gasparini
- Child Neuropsychiatry Unit, Niguarda Hospital, 20162 Milan, Italy
| | - Alessandro Armuzzi
- IBD Center, Humanitas Research Hospital-IRCCS, Via Manzoni 56, Rozzano, 20089 Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| |
Collapse
|
32
|
Pilipović I, Stojić-Vukanić Z, Leposavić G. Adrenoceptors as potential target for add-on immunomodulatory therapy in multiple sclerosis. Pharmacol Ther 2023; 243:108358. [PMID: 36804434 DOI: 10.1016/j.pharmthera.2023.108358] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 02/03/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023]
Abstract
This review summarizes recent findings related to the role of the sympathetic nervous system (SNS) in pathogenesis of multiple sclerosis (MS) and its commonly used experimental model - experimental autoimmune encephalomyelitis (EAE). They indicate that noradrenaline, the key end-point mediator of the SNS, acting through β-adrenoceptor, has a contributory role in the early stages of MS/EAE development. This stage is characterized by the SNS hyperactivity (increased release of noradrenaline) reflecting the net effect of different factors, such as the disease-associated inflammation, stress, vitamin D hypovitaminosis, Epstein-Barr virus infection and dysbiosis. Thus, the administration of propranolol, a non-selective β-adrenoceptor blocker, readily crossing the blood-brain barrier, to experimental rats before the autoimmune challenge and in the early (preclinical/prodromal) phase of the disease mitigates EAE severity. This phenomenon has been ascribed to the alleviation of neuroinflammation (due to attenuation of primarily microglial activation/proinflammatory functions) and the diminution of the magnitude of the primary CD4+ T-cell autoimmune response (the effect associated with impaired autoantigen uptake by antigen presenting cells and their migration into draining lymph nodes). The former is partly related to breaking of the catecholamine-dependent self-amplifying microglial feed-forward loop and the positive feedback loop between microglia and the SNS, leading to down-regulation of the SNS hyperactivity and its enhancing influence on microglial activation/proinflammatory functions and the magnitude of autoimmune response. The effects of propranolol are shown to be more prominent in male EAE animals, the phenomenon important as males (like men) are likely to develop clinically more severe disease. Thus, these findings could serve as a firm scientific background for formulation of a new sex-specific immune-intervention strategy for the early phases of MS (characterized by the SNS hyperactivity) exploiting anti-(neuro)inflammatory and immunomodulatory properties of propranolol and other relatively cheap and safe adrenergic drugs with similar therapeutic profile.
Collapse
Affiliation(s)
- Ivan Pilipović
- Institute of Virology, Vaccines and Sera "Torlak", Belgrade, Serbia
| | - Zorica Stojić-Vukanić
- University of Belgrade-Faculty of Pharmacy, Department of Microbiology and Immunology, Belgrade, Serbia
| | - Gordana Leposavić
- University of Belgrade-Faculty of Pharmacy, Department of Pathobiology, Belgrade, Serbia.
| |
Collapse
|
33
|
Chess-Williams R, Sellers DJ. Pathophysiological Mechanisms Involved in Overactive Bladder/Detrusor Overactivity. CURRENT BLADDER DYSFUNCTION REPORTS 2023. [DOI: 10.1007/s11884-023-00690-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
Abstract
Purpose of Review
To examine the latest published findings on the pathophysiological mechanisms involved in the development of overactive bladder (OAB) and detrusor overactivity (DO), and to identify common pathways linked to the risk factors associated with these conditions.
Recent Findings
Evidence is accumulating, both clinical and experimental, that many of the factors linked to the development of OAB/DO, including ageing, bladder outlet obstruction, psychological stress, and obesity are associated with reduced bladder blood flow. This induces local tissue inflammation with cytokine release and enhanced oxidative stress, ultimately resulting in altered detrusor sensitivity, detrusor hypertrophy and fibrosis, together with afferent hypersensitivity. These mechanisms would explain the symptoms of urgency and frequency observed in OAB patients. Although not a characteristic of OAB, undetected low level bacterial infections of the bladder have been proposed to explain the OAB symptoms in patients resistant to standard treatments. In this condition, inflammatory responses without reductions in perfusion activate the inflammatory pathways.
Summary
Evidence is mounting that poor bladder perfusion and local inflammatory responses are central mechanisms involved in the development of OAB/DO. As our understanding of these pathophysiological mechanisms advances, new avenues for drug development will be identified and ultimately treatment may become more individualized depending on the particular pathway involved and the drugs available.
Collapse
|
34
|
Minné D, Marnewick JL, Engel-Hills P. Early Chronic Stress Induced Changes within the Locus Coeruleus in Sporadic Alzheimer's Disease. Curr Alzheimer Res 2023; 20:301-317. [PMID: 37872793 DOI: 10.2174/1567205020666230811092956] [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: 02/27/2023] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 10/25/2023]
Abstract
Chronic exposure to stress throughout the lifespan has been the focus of many studies on Alzheimer's disease (AD) because of the similarities between the biological mechanisms involved in chronic stress and the pathophysiology of AD. In fact, the earliest abnormality associated with the disease is the presence of phosphorylated tau protein in locus coeruleus neurons, a brain structure highly responsive to stress and perceived threat. Here, we introduce allostatic load as a useful concept for understanding many of the complex, interacting neuropathological changes involved in the AD degenerative process. In response to chronic stress, aberrant tau proteins that begin to accumulate within the locus coeruleus decades prior to symptom onset appear to represent a primary pathological event in the AD cascade, triggering a wide range of interacting brain changes involving neuronal excitotoxicity, endocrine alterations, inflammation, oxidative stress, and amyloid plaque exacerbation. While it is acknowledged that stress will not necessarily be the major precipitating factor in all cases, early tau-induced changes within the locus coeruleus-norepinephrine pathway suggests that a therapeutic window might exist for preventative measures aimed at managing stress and restoring balance within the HPA axis.
Collapse
Affiliation(s)
- Donné Minné
- Applied Microbial & Health Biotechnology Institute, Cape Peninsula University of Technology, Cape Town, 7535, South Africa
- Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Cape Town, 7535, South Africa
| | - Jeanine L Marnewick
- Applied Microbial & Health Biotechnology Institute, Cape Peninsula University of Technology, Cape Town, 7535, South Africa
| | - Penelope Engel-Hills
- Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Cape Town, 7535, South Africa
| |
Collapse
|
35
|
Balestrieri P, Cicala M, Ribolsi M. Psychological distress in inflammatory bowel disease. Expert Rev Gastroenterol Hepatol 2023; 17:539-553. [PMID: 37254523 DOI: 10.1080/17474124.2023.2209723] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 04/28/2023] [Indexed: 06/01/2023]
Abstract
INTRODUCTION Mental health disorders are common in inflammatory bowel disease (IBD) and affect patients' quality of life, impacting on disease outcomes and health care-related costs. AREAS COVERED Even if psychological issues in IBD patients are highly burdened in terms of quality of life, psychiatric comorbidities still receive less attention into routine care than the physical symptoms of the disease. The present review provides an overview of recent literature, focusing on the association between perceived stress and IBD outcomes. For this purpose, the epidemiology of more common psychological comorbidities in IBD and their potential effect on the onset and disease course have been examined. Moreover, therapeutic interventions in the management of these patients have also been evaluated. EXPERT OPINION Screening of patients at high risk of psychological issues is currently an unmet, clinical need in the management of IBD. Under-diagnosed and under-treated mental health disorders in IBD patients may impact outcomes, leading to increased disability and health-care utilization and associated costs. A patient-tailored, integrated model of care in the management of IBD is required to optimize disease outcomes and improve patients' quality of life.
Collapse
Affiliation(s)
- Paola Balestrieri
- Unit of Gastroenterology and Digestive Endoscopy, Campus Bio Medico University, Rome, Italy
| | - Michele Cicala
- Unit of Gastroenterology and Digestive Endoscopy, Campus Bio Medico University, Rome, Italy
| | - Mentore Ribolsi
- Unit of Gastroenterology and Digestive Endoscopy, Campus Bio Medico University, Rome, Italy
| |
Collapse
|
36
|
Inflammatory Response and Immune Regulation in Brain-Heart Interaction after Stroke. Cardiovasc Ther 2022; 2022:2406122. [PMID: 36474712 PMCID: PMC9683992 DOI: 10.1155/2022/2406122] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 10/25/2022] [Indexed: 11/17/2022] Open
Abstract
Cerebrocardiac syndrome (CCS) is one of the secondary myocardial injuries after stroke. Cerebrocardiac syndrome may result in a poor prognosis with high mortality. Understanding the mechanism of the brain-heart interaction may be crucial for clinical treatment of pathological changes in CCS. Accumulating evidence suggests that the inflammatory response is involved in the brain-heart interaction after stroke. Systemic inflammatory response syndrome (SIRS) evoked by stroke may injure myocardial cells directly, in which the interplay between inflammatory response, oxidative stress, cardiac sympathetic/parasympathetic dysfunction, and splenic immunoregulation may be also the key pathophysiology factor. This review article summarizes the current understanding of inflammatory response and immune regulation in brain-heart interaction after stroke.
Collapse
|
37
|
Schramm E, Waisman A. Microglia as Central Protagonists in the Chronic Stress Response. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2022; 9:9/6/e200023. [PMID: 36357946 PMCID: PMC9453699 DOI: 10.1212/nxi.0000000000200023] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 07/06/2022] [Indexed: 05/15/2023]
Abstract
Chronic stress is a major risk factor for developing psychiatric conditions. In addition to elevating the levels of stress hormones released in the body, chronic stress activates the immune system, resulting in increased levels of proinflammatory cytokines and innate immune cells in the circulation of rodents and humans. Furthermore, exposure to chronic stress alters the phenotype of microglia, a population of innate immune cells that reside in the CNS parenchyma. In rodent models, chronic stress activates microglia in defined brain regions and induces changes in their phenotype and functional properties. In this review, we discussed how microglia are activated in stressful situations. Furthermore, we described how microglia affect the CNS environment during chronic stress, through the production of cytokines, the induction of reactive oxygen species, and phagocytosis. We suggested that, due to their strategic location as immune cells within the CNS, microglia are important players in the induction of psychopathologies after chronic stress.
Collapse
Affiliation(s)
- Eva Schramm
- From the Institute for Molecular Medicine (E.S., A.W.), University Medical Center, Johannes Gutenberg University Mainz; Focus Program Translational Neurosciences (A.W.), University Medical Center of the Johannes Gutenberg-University Mainz; and Research Center for Immunotherapy (A.W.), University Medical Center of the Johannes Gutenberg-University Mainz, Germany
| | - Ari Waisman
- From the Institute for Molecular Medicine (E.S., A.W.), University Medical Center, Johannes Gutenberg University Mainz; Focus Program Translational Neurosciences (A.W.), University Medical Center of the Johannes Gutenberg-University Mainz; and Research Center for Immunotherapy (A.W.), University Medical Center of the Johannes Gutenberg-University Mainz, Germany.
| |
Collapse
|
38
|
Smiley CE, Wood SK. Stress- and drug-induced neuroimmune signaling as a therapeutic target for comorbid anxiety and substance use disorders. Pharmacol Ther 2022; 239:108212. [PMID: 35580690 DOI: 10.1016/j.pharmthera.2022.108212] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/10/2022] [Accepted: 05/10/2022] [Indexed: 10/18/2022]
Abstract
Stress and substance use disorders remain two of the most highly prevalent psychiatric conditions and are often comorbid. While individually these conditions have a debilitating impact on the patient and a high cost to society, the symptomology and treatment outcomes are further exacerbated when they occur together. As such, there are few effective treatment options for these patients, and recent investigation has sought to determine the neural processes underlying the co-occurrence of these disorders to identify novel treatment targets. One such mechanism that has been linked to stress- and addiction-related conditions is neuroimmune signaling. Increases in inflammatory factors across the brain have been heavily implicated in the etiology of these disorders, and this review seeks to determine the nature of this relationship. According to the "dual-hit" hypothesis, also referred to as neuroimmune priming, prior exposure to either stress or drugs of abuse can sensitize the neuroimmune system to be hyperresponsive when exposed to these insults in the future. This review completes an examination of the literature surrounding stress-induced increases in inflammation across clinical and preclinical studies along with a summarization of the evidence regarding drug-induced alterations in inflammatory factors. These changes in neuroimmune profiles are also discussed within the context of their impact on the neural circuitry responsible for stress responsiveness and addictive behaviors. Further, this review explores the connection between neuroimmune signaling and susceptibility to these conditions and highlights the anti-inflammatory pharmacotherapies that may be used for the treatment of stress and substance use disorders.
Collapse
Affiliation(s)
- Cora E Smiley
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC 29209, United States of America; WJB Dorn Veterans Administration Medical Center, Columbia, SC 29209, United States of America.
| | - Susan K Wood
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC 29209, United States of America; WJB Dorn Veterans Administration Medical Center, Columbia, SC 29209, United States of America.
| |
Collapse
|
39
|
Abstract
Mental health disorders, particularly depression and anxiety, affect a significant number of the global population. Several pathophysiological pathways for these disorders have been identified, including the hypothalamic-pituitary-adrenal axis, autonomic nervous system, and the immune system. In addition, life events, environmental factors, and lifestyle affect the onset, progression, and recurrence of mental health disorders. These may all overlap with periodontal and/or peri-implant disease. Mental health disorders are associated with more severe periodontal disease and, in some cases, poorer healing outcomes to nonsurgical periodontal therapy. They can result in behavior modification, such as poor oral hygiene practices, tobacco smoking, and alcohol abuse, which are also risk factors for periodontal disease and, therefore, may have a contributory effect. Stress has immunomodulatory effects regulating immune cell numbers and function, as well as proinflammatory cytokine production. Stress markers such as cortisol and catecholamines may modulate periodontal bacterial growth and the expression of virulence factors. Stress and some mental health disorders are accompanied by a low-grade chronic inflammation that may be involved in their relationship with periodontal disease and vice versa. Although the gut microbiome interacting with the central nervous system (gut-brain axis) is thought to play a significant role in mental illness, less is understood about the role of the oral microbiome. The evidence for mental health disorders on implant outcomes is lacking, but may mainly be through behaviourial changes. Through lack of compliance withoral hygiene and maintenance visits, peri-implant health can be affected. Increased smoking and risk of periodontal disease may also affect implant outcomes. Selective serotonin reuptake inhibitors have been linked with higher implant failure. They have an anabolic effect on bone, reducing turnover, which could account for the increased loss.
Collapse
Affiliation(s)
- Jake Ball
- Centre for Rural Dentistry and Oral HealthCharles Sturt UniversityOrangeNew South WalesAustralia
| | - Ivan Darby
- Periodontics, Melbourne Dental SchoolThe University of MelbourneMelbourneVictoriaAustralia
| |
Collapse
|
40
|
Sood A, Chaudhari PR, Tiwari P, Shah S, Vaidya VA. Acute immobilization stress evokes sexually dimorphic peripheral and hippocampal neuroimmune responses in adult rats. Neurosci Lett 2022; 789:136871. [PMID: 36108934 DOI: 10.1016/j.neulet.2022.136871] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 09/04/2022] [Accepted: 09/08/2022] [Indexed: 10/14/2022]
Abstract
Stress perception and response vary across sexes and may contribute to the sex differences in susceptibility to psychopathology. Stress also engages the immune system and baseline immune system markers are known to be sexually dimorphic. Here, we investigated if the neuroimmune consequences following a single episode of acute immobilization stress (AIS) are sexually dimorphic in male and female Sprague-Dawley rats. We analyzed immune parameters in the periphery, and markers of neuroinflammation in the hippocampus, a key target of stress effects in the brain. We observed sexual dimorphism in the pattern of regulation of peripheral cytokines following stress, with males showing a significant increase in the levels of specific cytokines compared to females. Hippocampal cytokine and neuroinflammation-associated gene expression level analysis did not reveal any sexually dimorphic effects of AIS. However, we noted lower baseline expression levels for specific cytokines and many of the genes analyzed in the hippocampus of control females compared to control males. Finally, we assessed the levels of components of the NLRP3 inflammasome in the hippocampus and observed increased NLRP3 protein levels at baseline in females. We further noted that while males showed an increase in NLRP3 levels following AIS, females failed to show a similar change. Together, our results highlight a sexual dimorphism in neuroimmune consequences following AIS, both in the periphery and within the hippocampus, with males displaying robust proinflammatory changes and similar changes not observed in females. Our study underlines the importance of investigating the effect of sex on neuroimmune consequences following acute stress.
Collapse
Affiliation(s)
- Ankit Sood
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| | - Pratik R Chaudhari
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| | - Praachi Tiwari
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| | - Sneha Shah
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| | - Vidita A Vaidya
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India.
| |
Collapse
|
41
|
Kim IB, Lee JH, Park SC. The Relationship between Stress, Inflammation, and Depression. Biomedicines 2022; 10:1929. [PMID: 36009476 PMCID: PMC9405608 DOI: 10.3390/biomedicines10081929] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/29/2022] [Accepted: 07/31/2022] [Indexed: 12/18/2022] Open
Abstract
A narrative review about the relationship between stress, inflammation, and depression is made as follows: Chronic stress leads to various stress-related diseases such as depression. Although most human diseases are related to stress exposure, the common pathways between stress and pathophysiological processes of different disorders are still debatable. Chronic inflammation is a crucial component of chronic diseases, including depression. Both experimental and clinical studies have demonstrated that an increase in the levels of pro-inflammatory cytokines and stress hormones, such as glucocorticoids, substantially contributes to the behavioral alterations associated with depression. Evidence suggests that inflammation plays a key role in the pathology of stress-related diseases; however, this link has not yet been completely explored. In this study, we aimed to determine the role of inflammation in stress-induced diseases and whether a common pathway for depression exists. Recent studies support pharmacological and non-pharmacological treatment approaches significantly associated with ameliorating depression-related inflammation. In addition, major depression can be associated with an activated immune system, whereas antidepressants can exert immunomodulatory effects. Moreover, non-pharmacological treatments for major depression (i.e., exercise) may be mediated by anti-inflammatory actions. This narrative review highlights the mechanisms underlying inflammation and provides new insights into the prevention and treatment of stress-related diseases, particularly depression.
Collapse
Affiliation(s)
- Il-Bin Kim
- Department of Psychiatry, Hanyang University Guri Hospital, Guri 11923, Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Jae-Hon Lee
- Department of Psychiatry, Schulich of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
| | - Seon-Cheol Park
- Department of Psychiatry, Hanyang University Guri Hospital, Guri 11923, Korea
- Department of Psychiatry, Hanyang University College of Medicine, Seoul 04763, Korea
| |
Collapse
|
42
|
Shin YK, Kwon S, Hsieh YS, Han AY, Seol GH. Linalyl acetate restores colon contractility and blood pressure in repeatedly stressed-ulcerative colitis rats. Environ Health Prev Med 2022; 27:27. [PMID: 35753805 PMCID: PMC9283910 DOI: 10.1265/ehpm.22-00041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Background Ulcerative colitis (UC) is related to stress, but few studies have evaluated the influence of stress on factors affecting colon contractility in rats with UC. Also, there have been no studies investigating beneficial effects of linalyl acetate (LA), the major component of lavender essential oil, in repeatedly stressed-ulcerative colitis rats. Therefore, we investigated the differences in factors affecting colon contractility of UC rats with or without repeated restraint stress (RRS) and the effects of LA on these parameters in repeatedly stressed-UC rats. Methods Rats were assigned to following groups: control, RRS, UC, RRS+UC, and RRS+UC treated with LA or sulfasalazine. To induce UC, rats were administered 2% dextran sodium sulfate (DSS) water on days 1–5, followed by tap water on days 6–15 and DSS water on days 16–20. RRS was induced by immobilizing rats for 2 hr/day on days 1–20. LA or sulfasalazine were daily administered on days 16–20. Results Disease activity index (DAI) was markedly increased in RRS+UC. Serum interleukin-6 levels and acetylcholine-induced colon contraction were higher in RRS+UC than in control, RRS and UC. Colon nitrite levels also significantly increased in RRS+UC compared to the control and RRS. Blood pressure (BP) was higher in RRS+UC than in the control and UC. Both LA and sulfasalazine was effective in decreasing DAI, colon nitrite levels, acetylcholine-induced colon contraction in RRS+UC. Sulfasalazine significantly reduced serum IL-6 levels in RRS+UC with decreasing tendency in RRS+UC treated by LA. Only LA significantly reduced BP in RRS+UC. Conclusions Our findings emphasize the importance of stress management in UC patients. Also, LA may be beneficially used in repeatedly stressed-UC patients with high BP.
Collapse
Affiliation(s)
- You Kyoung Shin
- Department of Basic Nursing Science, School of Nursing, Korea University
| | - Soonho Kwon
- Department of Basic Nursing Science, School of Nursing, Korea University
| | - Yu Shan Hsieh
- Department of Basic Nursing Science, School of Nursing, Korea University.,Department of Nursing, School of Nursing, National Taipei University of Nursing and Health Sciences
| | - A Young Han
- Department of Basic Nursing Science, School of Nursing, Korea University.,Department of Nursing, School of Nursing, Sunchon National University
| | - Geun Hee Seol
- Department of Basic Nursing Science, School of Nursing, Korea University.,BK21 FOUR Program of Transdisciplinary Major in Learning Health Systems, Graduate School, Korea University
| |
Collapse
|
43
|
Iban-Arias R, Sebastian-Valverde M, Wu H, Lyu W, Wu Q, Simon J, Pasinetti GM. Role of Polyphenol-Derived Phenolic Acid in Mitigation of Inflammasome-Mediated Anxiety and Depression. Biomedicines 2022; 10:1264. [PMID: 35740286 PMCID: PMC9219614 DOI: 10.3390/biomedicines10061264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/23/2022] [Accepted: 05/26/2022] [Indexed: 02/01/2023] Open
Abstract
Overexposure to mental stress throughout life is a significant risk factor for the development of neuropsychiatric disorders, including depression and anxiety. The immune system can initiate a physiological response, releasing stress hormones and pro-inflammatory cytokines, in response to stressors. These effects can overcome allostatic physiological mechanisms and generate a pro-inflammatory environment with deleterious effects if occurring chronically. Previous studies in our lab have identified key anti-inflammatory properties of a bioavailable polyphenolic preparation BDPP and its ability to mitigate stress responses via the attenuation of NLRP3 inflammasome-dependent responses. Inflammasome activation is part of the first line of defense against stimuli of different natures, provides a rapid response, and, therefore, is of capital importance within the innate immunity response. malvidin-3-O-glucoside (MG), a natural anthocyanin present in high proportions in grapes, has been reported to exhibit anti-inflammatory effects, but its mechanisms remain poorly understood. This study aims to elucidate the therapeutic potential of MG on inflammasome-induced inflammation in vitro and in a mouse model of chronic unpredictable stress (CUS). Here, it is shown that MG is an anti-pyroptotic phenolic metabolite that targets NLRP3, NLRC4, and AIM2 inflammasomes, subsequently reducing caspase-1 and IL-1β protein levels in murine primary cortical microglia and the brain, as its beneficial effect to counteract anxiety and depression is also demonstrated. The present study supports the role of MG to mitigate bacterial-mediated inflammation (lipopolysaccharide or LPS) in vitro and CUS-induced behavior impairment in vivo to address stress-induced inflammasome-mediated innate response.
Collapse
Affiliation(s)
- Ruth Iban-Arias
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (R.I.-A.); (M.S.-V.); (H.W.)
| | - Maria Sebastian-Valverde
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (R.I.-A.); (M.S.-V.); (H.W.)
| | - Henry Wu
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (R.I.-A.); (M.S.-V.); (H.W.)
| | - Weiting Lyu
- New Use Agriculture and Natural Plant Products Program, Department of Plant Biology, Center for Agricultural Food Ecosystems, Institute of Food, Nutrition & Health, SEBS, Rutgers University, 59 Dudley Road, New Brunswick, NJ 08901, USA; (W.L.); (Q.W.); (J.S.)
- Department of Medicinal Chemistry, Ernest Mario School of Pharmacy, Rutgers University, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA
| | - Qingli Wu
- New Use Agriculture and Natural Plant Products Program, Department of Plant Biology, Center for Agricultural Food Ecosystems, Institute of Food, Nutrition & Health, SEBS, Rutgers University, 59 Dudley Road, New Brunswick, NJ 08901, USA; (W.L.); (Q.W.); (J.S.)
| | - Jim Simon
- New Use Agriculture and Natural Plant Products Program, Department of Plant Biology, Center for Agricultural Food Ecosystems, Institute of Food, Nutrition & Health, SEBS, Rutgers University, 59 Dudley Road, New Brunswick, NJ 08901, USA; (W.L.); (Q.W.); (J.S.)
| | - Giulio Maria Pasinetti
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (R.I.-A.); (M.S.-V.); (H.W.)
| |
Collapse
|
44
|
Hollis F, Pope BS, Gorman-Sandler E, Wood SK. Neuroinflammation and Mitochondrial Dysfunction Link Social Stress to Depression. Curr Top Behav Neurosci 2022; 54:59-93. [PMID: 35184261 DOI: 10.1007/7854_2021_300] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Major depressive disorder is a debilitating mental illness and a leading cause of global disease burden. While many etiological factors have been identified, social stress is a highly prevalent causative factor for the onset of depression. Unfortunately, rates of depression continue to increase around the world, and the recent COVID-19 pandemic has further exacerbated this mental health crisis. Though several therapeutic strategies are available, nearly 50% of patients who receive treatment never reach remission. The exact mechanisms by which social stress exposure promotes the development of depression are unclear, making it challenging to develop novel and more effective therapeutics. However, accumulating evidence points to a role for stress-induced neuroinflammation, particularly in treatment-resistant patients. Moreover, recent evidence has expanded the concept of the pathogenesis of depression to mitochondrial dysfunction, suggesting that the combined effects of social stress on mitochondria and inflammation may synergize to facilitate stress-related depression. In this chapter, we review evidence for neuroinflammation and mitochondrial dysfunction in the pathogenesis of social stress-induced depression and discuss these in the context of novel therapeutic targets for the treatment of depression.
Collapse
Affiliation(s)
- Fiona Hollis
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Brittany S Pope
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
- Department of Exercise Science, University of South Carolina Arnold School of Public Health, Columbia, SC, USA
| | - Erin Gorman-Sandler
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Susan K Wood
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA.
| |
Collapse
|
45
|
Wang J, Zhou Y, Li K, Li X, Guo M, Peng M. A Noradrenergic Lesion Attenuates Surgery-Induced Cognitive Impairment in Rats by Suppressing Neuroinflammation. Front Mol Neurosci 2021; 14:752838. [PMID: 34916906 PMCID: PMC8671038 DOI: 10.3389/fnmol.2021.752838] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/25/2021] [Indexed: 11/22/2022] Open
Abstract
Postoperative cognitive dysfunction (POCD) is a common postoperative neurocognitive complication in elderly patients. However, the specific pathogenesis is unknown, and it has been demonstrated that neuroinflammation plays a key role in POCD. Recently, increasing evidence has proven that the locus coeruleus noradrenergic (LCNE) system participates in regulating neuroinflammation in some neurodegenerative disorders. We hypothesize that LCNE plays an important role in the neuroinflammation of POCD. In this study, 400 μg of N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP-4) was injected intracerebroventricularly into each rat 7 days before anesthesia/surgery to deplete the locus coeruleus (LC) noradrenaline (NE). We applied a simple laparotomy and brief upper mesenteric artery clamping surgery as the rat POCD model. The open field test, novel objection and novel location (NL) recognition, and Morris water maze (MWM) were performed to assess postoperative cognition. High-performance liquid chromatography (HPLC) was used to measure the level of NE in plasma and brain tissues, and immunofluorescence staining was applied to evaluate the activation of microglia and astrocytes. We also used enzyme-linked immune-sorbent assay (ELISA) to assess the levels of inflammatory cytokines and brain-derived neurotrophic factor (BDNF). Pretreatment with DSP-4 decreased the levels of systemic and central NE, increased the level of interleukin-6 (IL-6) in the plasma at 6 h after the surgery, decreased the concentration of IL-6 in the prefrontal cortex and hippocampus, and decreased the level of interleukin-1β (IL-1β) in the plasma, prefrontal cortex, and hippocampus at 1 week postoperatively. In addition, DSP-4 treatment attenuated hippocampal-dependent learning and memory impairment in rats with POCD, with a downregulation of the activation of microglia and astrocytes in the prefrontal cortex and hippocampus. In conclusion, these findings provide evidence of the effects of LCNE in modulating neuroinflammation in rats with POCD and provide a new perspective in the prevention and treatment of POCD.
Collapse
Affiliation(s)
- Jiayu Wang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ying Zhou
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ke Li
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiaofeng Li
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Meimei Guo
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Mian Peng
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
46
|
Safarzadeh Kozani P, Safarzadeh Kozani P, Rahbarizadeh F. Optimizing the Clinical Impact of CAR-T Cell Therapy in B-Cell Acute Lymphoblastic Leukemia: Looking Back While Moving Forward. Front Immunol 2021; 12:765097. [PMID: 34777381 PMCID: PMC8581403 DOI: 10.3389/fimmu.2021.765097] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/07/2021] [Indexed: 12/13/2022] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy has been successful in creating extraordinary clinical outcomes in the treatment of hematologic malignancies including relapsed or refractory (R/R) B-cell acute lymphoblastic leukemia (B-ALL). With several FDA approvals, CAR-T therapy is recognized as an alternative treatment option for particular patients with certain conditions of B-ALL, diffuse large B-cell lymphoma, mantle cell lymphoma, follicular lymphoma, or multiple myeloma. However, CAR-T therapy for B-ALL can be surrounded by challenges such as various adverse events including the life-threatening cytokine release syndrome (CRS) and neurotoxicity, B-cell aplasia-associated hypogammaglobulinemia and agammaglobulinemia, and the alloreactivity of allogeneic CAR-Ts. Furthermore, recent advances such as improvements in media design, the reduction of ex vivo culturing duration, and other phenotype-determining factors can still create room for a more effective CAR-T therapy in R/R B-ALL. Herein, we review preclinical and clinical strategies with a focus on novel studies aiming to address the mentioned hurdles and stepping further towards a milestone in CAR-T therapy of B-ALL.
Collapse
Affiliation(s)
- Pouya Safarzadeh Kozani
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran.,Student Research Committee, Medical Biotechnology Research Center, School of Nursing, Midwifery, and Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Pooria Safarzadeh Kozani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Rahbarizadeh
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.,Research and Development Center of Biotechnology, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
47
|
Cao C, Chowdhury VS, Cline MA, Gilbert ER. The Microbiota-Gut-Brain Axis During Heat Stress in Chickens: A Review. Front Physiol 2021; 12:752265. [PMID: 34744792 PMCID: PMC8563997 DOI: 10.3389/fphys.2021.752265] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/29/2021] [Indexed: 12/20/2022] Open
Abstract
Heat stress is a global issue for the poultry industries with substantial annual economic losses and threats to bird health and welfare. When chickens are exposed to high ambient temperatures, like other species they undergo multiple physiological alterations, including behavioral changes, such as cessation of feeding, initiation of a stress signaling cascade, and intestinal immune, and inflammatory responses. The brain and gut are connected and participate in bidirectional communication via the nervous and humoral systems, this network collectively known as the gut-brain axis. Moreover, heat stress not only induces hyperthermia and oxidative stress at the gut epithelium, leading to impaired permeability and then susceptibility to infection and inflammation, but also alters the composition and abundance of the microbiome. The gut microflora, primarily via bacterially derived metabolites and hormones and neurotransmitters, also communicate via similar pathways to regulate host metabolic homeostasis, health, and behavior. Thus, it stands to reason that reshaping the composition of the gut microbiota will impact intestinal health and modulate host brain circuits via multiple reinforcing and complementary mechanisms. In this review, we describe the structure and function of the microbiota-gut-brain axis, with an emphasis on physiological changes that occur in heat-stressed poultry.
Collapse
Affiliation(s)
- Chang Cao
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Vishwajit S Chowdhury
- Laboratory of Stress Physiology and Metabolism, Faculty of Arts and Science, Kyushu University, Fukuoka, Japan
| | - Mark A Cline
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Elizabeth R Gilbert
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| |
Collapse
|
48
|
Woodburn SC, Bollinger JL, Wohleb ES. The semantics of microglia activation: neuroinflammation, homeostasis, and stress. J Neuroinflammation 2021; 18:258. [PMID: 34742308 PMCID: PMC8571840 DOI: 10.1186/s12974-021-02309-6] [Citation(s) in RCA: 376] [Impact Index Per Article: 94.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 10/28/2021] [Indexed: 02/08/2023] Open
Abstract
Microglia are emerging as critical regulators of neuronal function and behavior in nearly every area of neuroscience. Initial reports focused on classical immune functions of microglia in pathological contexts, however, immunological concepts from these studies have been applied to describe neuro-immune interactions in the absence of disease, injury, or infection. Indeed, terms such as 'microglia activation' or 'neuroinflammation' are used ubiquitously to describe changes in neuro-immune function in disparate contexts; particularly in stress research, where these terms prompt undue comparisons to pathological conditions. This creates a barrier for investigators new to neuro-immunology and ultimately hinders our understanding of stress effects on microglia. As more studies seek to understand the role of microglia in neurobiology and behavior, it is increasingly important to develop standard methods to study and define microglial phenotype and function. In this review, we summarize primary research on the role of microglia in pathological and physiological contexts. Further, we propose a framework to better describe changes in microglia1 phenotype and function in chronic stress. This approach will enable more precise characterization of microglia in different contexts, which should facilitate development of microglia-directed therapeutics in psychiatric and neurological disease.
Collapse
Affiliation(s)
- Samuel C Woodburn
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Justin L Bollinger
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Eric S Wohleb
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
49
|
Mansouri V, Yazdanpanah N, Rezaei N. The immunologic aspects of cytokine release syndrome and graft versus host disease following CAR T cell therapy. Int Rev Immunol 2021; 41:649-668. [PMID: 34607523 DOI: 10.1080/08830185.2021.1984449] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Chimeric antigen receptor (CAR) T cells are the pioneers of cancer immunotherapy, which to this date have several FDA-approved products. They have been substantially improved since their first introduction in 1993 and have shown promising results regardless of their inevitable side effects. Cytokine release syndrome (CRS), the most common toxicity after CAR T cell treatment, is affiliated to a systemic inflammation through surge of cytokines, mainly IL-6, IL-1, and INF-γ. Furthermore, difference between histocompatibility antigens activates the graft versus host disease (GvHD) effect of the allogenic CAR T cells against the host cells. Immunological reactions induced by CAR T cells in the form of CRS or GvHD is necessary for fostering good responses, while excess reactions can potentially threaten patient life. In this review, we first describe the history, applications, and structure of CAR T cells, followed by a comprehensive review of CRS regarding its definition, management, and immunological aspects. Finally, we discuss about the clinical aspects of CRS and GvHD after CAR T cell therapy and how to harness anti-tumoral effects, while mitigating the adverse effects.
Collapse
Affiliation(s)
- Vahid Mansouri
- Gene Therapy Research Center, Digestive Diseases Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Niloufar Yazdanpanah
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
50
|
Choudhary M, Chen Y, Friedman O, Cuk N, Ben-Shlomo A. Pheochromocytoma Crisis Presenting With ARDS Successfully Treated With ECMO-Assisted Adrenalectomy. AACE Clin Case Rep 2021; 7:310-314. [PMID: 34522771 PMCID: PMC8426613 DOI: 10.1016/j.aace.2021.03.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 01/19/2023] Open
Abstract
Objective Pheochromocytoma (PCC) crisis caused by acute catecholamine release from an adrenal PCC or extra-adrenal paraganglioma can be difficult to diagnose and may require an unconventional management strategy to achieve good outcomes. We describe a case of PCC crisis presenting with acute respiratory distress syndrome (ARDS) that resolved with stabilization on veno-venous (VV) extracorporeal membrane oxygenation (ECMO) during adrenalectomy. Case Description A 30-year-old man with a history of severe alcohol use disorder and a prior hospital admission for alcohol withdrawal syndrome presented with sudden-onset hemoptysis, altered mental status, and severe dyspnea that rapidly deteriorated to ARDS requiring ECMO support. He demonstrated hemodynamic collapse after cannulation for VV-ECMO and stabilized after conversion to veno-arterial-ECMO, but ARDS persisted and he developed acute renal failure. Computed tomography without contrast done as part of work-up for a presumed infection revealed a 6.9 × 6.4 cm right adrenal mass suspicious for pheochromocytoma. Plasma and random urine metanephrine levels were markedly elevated. ARDS persisted despite α- and β-adrenoreceptor blockade, and he underwent laparoscopic right adrenalectomy with VV-ECMO support. Pathology confirmed PCC with intermediate risk for malignancy. Postoperatively, he was weaned off respiratory and renal support within 10 days, showed rapid clinical improvement, and was discharged 1 month later. Conclusion This case highlights diagnostic and management challenges associated with patients with PCC crisis presenting with ARDS. A multidisciplinary team approach is critical to identifying appropriate treatment strategies.
Collapse
Affiliation(s)
- Manita Choudhary
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Yufei Chen
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California
| | - Oren Friedman
- Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Natasha Cuk
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Anat Ben-Shlomo
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| |
Collapse
|