1
|
Zuo H, Yang X, Wang Y, Hu B, Zhu Z, Guo Z, Weng S, He J, Xu X. Excessive activation of JAK-STAT signaling contributes to inflammation induced by acute Vibrio infection in shrimp. Virulence 2025; 16:2451169. [PMID: 39819453 PMCID: PMC11749392 DOI: 10.1080/21505594.2025.2451169] [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: 05/06/2024] [Revised: 12/12/2024] [Accepted: 01/05/2025] [Indexed: 01/19/2025] Open
Abstract
Uncontrolled immune responses resulting from overactivated cellular signaling pathways, leading to inflammation and tissue injury, are a major cause of death in pathogen-infected individuals. This phenomenon has been well studied in mammals but is less explored in invertebrates. Bacteria of the genus Vibrio are among the most harmful pathogens to humans and aquatic animals. In shrimp, Vibrio infection is generally characterized by the sudden onset of disease, with pathological signs of opaque and whitish muscle tissue. The current study shows that shrimp acutely infected with high dose of Vibrio parahaemolyticus develop inflammation-like pathological changes, leading to rapid death. Excessive activation of JAK-STAT signaling, rather than the Dorsal and Relish pathways, results in overactivation of shrimp immunity and is a major cause of inflammation induced by acute Vibrio infection. Weakening JAK-STAT signaling attenuates the inflammatory response and reduces mortality caused by acute Vibrio infection in shrimp, whereas enhancing JAK-STAT signaling can convert a normal infection into an acute one, accelerating shrimp death. Therefore, this study indicates that, similar to that in mammals, the pathogenesis of infectious diseases in invertebrates is complicated by inflammatory responses triggered by dysregulated immune signaling.
Collapse
Affiliation(s)
- Hongliang Zuo
- State Key Laboratory of Biocontrol, School of Life Sciences/Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Sun Yat-sen University, Guangzhou, P. R. China
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, P. R. China
- Provincial Observation and Research Station for Marine Ranching in Lingdingyang bay, Zhuhai, P. R. China
| | - Xiya Yang
- State Key Laboratory of Biocontrol, School of Life Sciences/Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Sun Yat-sen University, Guangzhou, P. R. China
| | - Youxi Wang
- State Key Laboratory of Biocontrol, School of Life Sciences/Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Sun Yat-sen University, Guangzhou, P. R. China
| | - Bangping Hu
- State Key Laboratory of Biocontrol, School of Life Sciences/Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Sun Yat-sen University, Guangzhou, P. R. China
| | - Zhiming Zhu
- State Key Laboratory of Biocontrol, School of Life Sciences/Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Sun Yat-sen University, Guangzhou, P. R. China
- Provincial Observation and Research Station for Marine Ranching in Lingdingyang bay, Zhuhai, P. R. China
| | - Zhixun Guo
- State Key Laboratory of Biocontrol, School of Life Sciences/Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Sun Yat-sen University, Guangzhou, P. R. China
- South China Sea Fisheries Research Institute (CAFS), Guangzhou, P. R. China
| | - Shaoping Weng
- State Key Laboratory of Biocontrol, School of Life Sciences/Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Sun Yat-sen University, Guangzhou, P. R. China
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, P. R. China
| | - Jianguo He
- State Key Laboratory of Biocontrol, School of Life Sciences/Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Sun Yat-sen University, Guangzhou, P. R. China
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, P. R. China
| | - Xiaopeng Xu
- State Key Laboratory of Biocontrol, School of Life Sciences/Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Sun Yat-sen University, Guangzhou, P. R. China
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, P. R. China
| |
Collapse
|
2
|
Wiley CR, Williams DP, Sigrist C, Brownlow BN, Markser A, Hong S, Sternberg EM, Kapuku G, Koenig J, Thayer JF. Differences in inflammation among black and white individuals: A systematic review and meta-analysis. Brain Behav Immun 2025; 127:269-286. [PMID: 40101808 DOI: 10.1016/j.bbi.2025.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 02/13/2025] [Accepted: 03/09/2025] [Indexed: 03/20/2025] Open
Abstract
IMPORTANCE Despite persisting health disparities between Black and White individuals, racial differences in inflammation have yet to be comprehensively examined. OBJECTIVE To determine if significant differences in circulating levels of inflammatory markers between Black and White populations exist. DATA SOURCES Studies were identified through systematic searches of four electronic databases in January 2022. Additional studies were identified via reference lists and e-mail contact. STUDY SELECTION Eligible studies included full-text empirical articles that consisted of Black and White individuals and reported statistics for inflammatory markers for each racial group. Of the 1368 potentially eligible studies, 84 (6.6 %) representing more than one million participants met study selection criteria. DATA EXTRACTION AND SYNTHESIS Risk of bias was assessed via meta regressions that considered relevant covariates. Data heterogeneity was tested using both the Cochrane Q-statistic and the standard I2 index. Random effects models were used to calculate estimates of effect size from standardized mean differences. MAIN OUTCOMES AND MEASURES Outcome measures included 12 inflammatory markers, including C-reactive protein (CRP), Fibrinogen, Interleukin-6 (IL-6), Tumor necrosis factor-alpha (TNF-α), and soluble intercellular adhesion molecule 1 (sICAM-1). RESULTS Several markers had robust sample sizes for analysis, including CRP (White N = 934,594; Black N = 55,234), Fibrinogen (White N = 80,880; Black N = 18,001), and IL-6 (White N = 20,269; Black N = 14,675). Initial results indicated significant effects on CRP (k = 56, pooled Hedges' g = 0.24), IL-6 (k = 33, g = 0.15), and Fibrinogen (k = 19, g = 0.49), with Black individuals showing higher levels. Results also indicated significant effects on sICAM-1 (k = 6, g = -0.46), and Interleukin-10 (k = 4, g = -0.18), with White individuals showing higher levels. Sensitivity analyses confirmed robust effects for CRP, IL-6, Fibrinogen, and sICAM-1 while also revealing significant effects on TNF-α (k = 18, g = -0.17) and Interleukin-8 (k = 5, g = -0.19), with White individuals showing higher levels of both. CONCLUSIONS AND RELEVANCE Current meta-analytic results provide evidence for marked racial differences in common circulating inflammatory markers and illustrate the complexity of the inflammatory profile differences between Black and White individuals. Review Pre-Registration: PROSPERO Identifier - CRD42022312352.
Collapse
Affiliation(s)
- Cameron R Wiley
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Department of Psychological Science, University of California, Irvine, Irvine, CA, United States.
| | - DeWayne P Williams
- Department of Psychological Science, University of California, Irvine, Irvine, CA, United States
| | - Christine Sigrist
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, United States
| | - Briana N Brownlow
- Duke University Medical Center, Duke University, Durham, NC, United States
| | - Anna Markser
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, United States
| | - Suzi Hong
- Herbert Wertheim School of Public Health, University of California, San Diego, San Diego, CA, United States; Department of Psychiatry, School of Medicine, University of California, San Diego, San Diego, CA, United States
| | - Esther M Sternberg
- Center for Integrative Medicine, The University of Arizona, Tucson, AZ, United States
| | - Gaston Kapuku
- Department of Pediatrics and Medicine, Georgia Prevention Institute, Augusta, GA, United States
| | - Julian Koenig
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, United States
| | - Julian F Thayer
- Department of Psychological Science, University of California, Irvine, Irvine, CA, United States; Department of Psychology, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
3
|
Zheng H, Liu W, Chen B, Hu S, Mo D, Guo P, Jiang X, Yang R, Wang S, Zhong H. Inflammatory chemokines in adolescents' suicide state with depressive disorders. Ann Gen Psychiatry 2025; 24:37. [PMID: 40450340 DOI: 10.1186/s12991-025-00571-z] [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: 10/24/2024] [Accepted: 05/03/2025] [Indexed: 06/03/2025] Open
Abstract
Suicide has become one of the leading causes of death in adolescents and is a direct consequence in patients with depressive disorders. Recently, neuroinflammation has been shown to play a role in the development of depression. This study examined serum chemokine levels in adolescents with depressive disorders at different suicide stages to identify chemokines that discriminate between suicidal behaviors. This study enrolled 111 adolescent patients with depressive disorders who were further subdivided into three groups based on the presence of suicidal behavior or ideation: suicide attempt group (n = 45), suicidal ideation group (n = 44), and non-suicidal depression group (n = 22), along with 23 healthy controls. Eight inflammatory chemokines were detected using the mesoscale discovery method. Patients in the suicide attempters group had higher levels of IL8, MCP-1, Eotaxin, and Eotaxin-3 than those in the non-suicide depression group. Eotaxin-2 had the greatest effect on suicidal behaviors of all factors.
Collapse
Affiliation(s)
- Hongyu Zheng
- Affiliated Psychological Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Child and Adolescents, Hefei Fourth People's Hospital, Hefei, Anhui, China
- Anhui Mental Health Center, Hefei, Anhui, China
| | - Wenyuan Liu
- Affiliated Psychological Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Child and Adolescents, Hefei Fourth People's Hospital, Hefei, Anhui, China
- Anhui Mental Health Center, Hefei, Anhui, China
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Binbin Chen
- Affiliated Psychological Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Child and Adolescents, Hefei Fourth People's Hospital, Hefei, Anhui, China
- Anhui Mental Health Center, Hefei, Anhui, China
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Shuwen Hu
- Department of Psychology, Anhui Provincial Children's Hospital, Hefei, Anhui, 230051, China
| | - Daming Mo
- Affiliated Psychological Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Child and Adolescents, Hefei Fourth People's Hospital, Hefei, Anhui, China
- Anhui Mental Health Center, Hefei, Anhui, China
| | - Pengfei Guo
- Affiliated Psychological Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Child and Adolescents, Hefei Fourth People's Hospital, Hefei, Anhui, China
- Anhui Mental Health Center, Hefei, Anhui, China
| | - Xiaolu Jiang
- Affiliated Psychological Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Child and Adolescents, Hefei Fourth People's Hospital, Hefei, Anhui, China
- Anhui Mental Health Center, Hefei, Anhui, China
| | - Rong Yang
- Affiliated Psychological Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Child and Adolescents, Hefei Fourth People's Hospital, Hefei, Anhui, China
- Anhui Mental Health Center, Hefei, Anhui, China
| | - Shuo Wang
- Affiliated Psychological Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Child and Adolescents, Hefei Fourth People's Hospital, Hefei, Anhui, China
- Anhui Mental Health Center, Hefei, Anhui, China
| | - Hui Zhong
- Affiliated Psychological Hospital of Anhui Medical University, Hefei, Anhui, China.
- Department of Child and Adolescents, Hefei Fourth People's Hospital, Hefei, Anhui, China.
- Anhui Mental Health Center, Hefei, Anhui, China.
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
4
|
Zuo H, Liao A, Shen S, Wang Y, Yang X, Weng S, He J, Xu X. microRNA-34 mediates a negative feedback loop in the JAK-STAT pathway to attenuate immune overactivation in an invertebrate. Cell Commun Signal 2025; 23:228. [PMID: 40382609 PMCID: PMC12085071 DOI: 10.1186/s12964-025-02230-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Accepted: 05/04/2025] [Indexed: 05/20/2025] Open
Abstract
The JAK-STAT pathway is an essential signaling mechanism that initiates immune responses against pathogen infections. The intrinsic homeostatic regulation of JAK-STAT signaling is critical for maintaining immune homeostasis. Previous studies have shown that aberrant activation of the invertebrate JAK-STAT pathway leads to inflammation-like symptoms. Understanding the homeostatic mechanisms of this pathway in invertebrates is of significant interest. Pacific white shrimp (Penaeus vannamei) is one of the most extensively studied invertebrates in immune system research. In this study, we demonstrate that the shrimp microRNA-34 (miR-34) attenuates JAK-STAT pathway activation by targeting JAK, thereby inhibiting STAT phosphorylation and nuclear translocation. Interestingly, miR-34 expression is directly regulated by STAT, forming a negative feedback regulatory loop in the JAK-STAT pathway. Disrupting this loop results in excessive JAK-STAT pathway activation and immune overactivation, exacerbating inflammation caused by Vibrio parahaemolyticus infection in shrimp. This study provides new insights into the regulatory mechanism of the JAK-STAT pathway and its roles in maintaining immune homeostasis in invertebrates.
Collapse
Affiliation(s)
- Hongliang Zuo
- State Key Laboratory of Biocontrol, School of Life Sciences/ Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/ China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Sun Yat-sen University, Guangzhou, P. R. China
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, P. R. China
| | - Alian Liao
- State Key Laboratory of Biocontrol, School of Life Sciences/ Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/ China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Sun Yat-sen University, Guangzhou, P. R. China
| | - Sihui Shen
- State Key Laboratory of Biocontrol, School of Life Sciences/ Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/ China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Sun Yat-sen University, Guangzhou, P. R. China
| | - Youxi Wang
- State Key Laboratory of Biocontrol, School of Life Sciences/ Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/ China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Sun Yat-sen University, Guangzhou, P. R. China
| | - Xiya Yang
- State Key Laboratory of Biocontrol, School of Life Sciences/ Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/ China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Sun Yat-sen University, Guangzhou, P. R. China
| | - Shaoping Weng
- State Key Laboratory of Biocontrol, School of Life Sciences/ Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/ China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Sun Yat-sen University, Guangzhou, P. R. China
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, P. R. China
| | - Jianguo He
- State Key Laboratory of Biocontrol, School of Life Sciences/ Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/ China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Sun Yat-sen University, Guangzhou, P. R. China.
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, P. R. China.
| | - Xiaopeng Xu
- State Key Laboratory of Biocontrol, School of Life Sciences/ Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/ China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Sun Yat-sen University, Guangzhou, P. R. China.
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, P. R. China.
| |
Collapse
|
5
|
Khoshakhlagh AH, Yazdanirad S, Gruszecka-Kosowska A, Drake CL, Emerson W. Investigation of relationship between occupational exposure to aerosol and sleep problems: A systematic review and meta-analysis. PLoS One 2025; 20:e0321515. [PMID: 40343941 PMCID: PMC12063870 DOI: 10.1371/journal.pone.0321515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 03/04/2025] [Indexed: 05/11/2025] Open
Abstract
There are various occupational and industrial activities that are associated with the production of suspended particles. Little is known about sleep disturbance caused by exposure to aerosol exposure. Presented systematic review and meta-analysis study aimed to investigate the impact of various aerosols during occupational exposure on sleep. A systematic search in Scopus, Web of Science, PubMed, Embase, and Medline databases was performed until 20 February 2024. Three sets of keywords and their possible combinations were used in the search algorithm. To evaluate the quality and risk of bias in studies, the Joanna Briggs Institute (JBI) tools and risk of bias in non-randomized studies of exposure (ROBINS-E) instruments were applied, respectively. The pooled values were also computed by meta-analysis. Based on inclusion/exclusion criteria, 23 articles were entered into the review. 9 out of 11 articles with high quality (81.82 percent), 8 out of 9 articles with moderate quality (88.89 percent), and 2 out of 3 articles with low quality (66.67 percent) indicated that aerosol exposure had a meaningful negative effect on sleep among workers in various occupations. Among articles, 69.6% (N = 16) were given a high risk of bias rating, 13.0% (N = 3) were rated as moderate risk of bias, and 17.4% (N = 4) were rated as low risk of bias. The results of the meta-analysis indicated that the pooled value of the prevalence in the cross-sectional, cohort, and case control studies was 42.35 (95%CI [34.55, 50.16]), 10.82 (95%CI [6.76, 14.87]), and 35.70 (95%CI [13.96, 57.45]), respectively. Also, the results of the meta-analysis showed that the pooled values of the odds ratio in the cross-sectional and cohort studies were 1.82 (95% CI [1.43, 2.21]) and 1.73 (95% CI [1.49, 1.96]), respectively. Totally, most studies indicated that various sources of occupational aerosol exposure significantly affected sleep among employees.
Collapse
Affiliation(s)
- Amir Hossein Khoshakhlagh
- Department of Occupational Health, School of Health, Kashan University of Medical Sciences, Kashan, Iran
| | - Saeid Yazdanirad
- Department of Occupational Health, School of Health, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Social Determinants of Health Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Agnieszka Gruszecka-Kosowska
- Department of Environmental Protection, Faculty of Geology, Geophysics, and Environmental Protection, AGH University of Krakow, Al. Mickiewicza 30, 30-059, Krakow, Poland
| | - Christopher L. Drake
- Henry Ford Health System, Sleep Disorders and Research Center, Detroit, Michigan, United States of America
| | - Wickwire Emerson
- Psychiatry and Medicine, Section Head, Sleep Medicine, University of Maryland School of Medicine, Baltimore, United States of America
| |
Collapse
|
6
|
Feng WD, Liu DN, Shang YF, Zhang WF, Xu S, Feng DH, Wang YH. Neuroimmune modulators derived from natural products: Mechanisms and potential therapies. Pharmacol Ther 2025; 269:108830. [PMID: 40015519 DOI: 10.1016/j.pharmthera.2025.108830] [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: 05/26/2024] [Revised: 12/26/2024] [Accepted: 02/23/2025] [Indexed: 03/01/2025]
Abstract
Neuroimmunology is a multidisciplinary field that investigates the interactions between the nervous and immune systems. Neuroimmune interactions persist throughout the entire lifespan, and their dysregulation can lead to the onset and development of multiple diseases. Despite significant progress over the past decades in elucidating the interaction between neuroscience and immunology, the exact mechanism underlying neuroimmune crosstalk has not yet been fully elucidated. In recent years, natural products have emerged as a promising avenue for the therapeutic implications of neuroimmune diseases. Naturally derived anti-neuroimmune disease agents, such as polyphenols, flavonoids, alkaloids, and saponins, have been extensively studied for their potential neuroimmune modulatory effects. This comprehensive review delves into the specific molecular mechanisms of bidirectional neuro-immune interactions, with particular emphasis on the role of neuro-immune units. The review synthesizes a substantial body of evidence from in vitro and in vivo experiments as well as clinical studies, highlighting the therapeutic potential of various natural products in intervening in neuroimmune disorders.
Collapse
Affiliation(s)
- Wan-Di Feng
- Beijing Key Laboratory of Innovative Drug Discovery and Polymorphic Druggability Research for Cerebrovascular Diseases, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Dong-Ni Liu
- Beijing Key Laboratory of Innovative Drug Discovery and Polymorphic Druggability Research for Cerebrovascular Diseases, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yu-Fu Shang
- Beijing Key Laboratory of Innovative Drug Discovery and Polymorphic Druggability Research for Cerebrovascular Diseases, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Wen-Fang Zhang
- Beijing Key Laboratory of Innovative Drug Discovery and Polymorphic Druggability Research for Cerebrovascular Diseases, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Shuang Xu
- Beijing Key Laboratory of Innovative Drug Discovery and Polymorphic Druggability Research for Cerebrovascular Diseases, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Dan-Hong Feng
- Beijing Key Laboratory of Innovative Drug Discovery and Polymorphic Druggability Research for Cerebrovascular Diseases, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yue-Hua Wang
- Beijing Key Laboratory of Innovative Drug Discovery and Polymorphic Druggability Research for Cerebrovascular Diseases, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
7
|
Yang X, Li B, Wu L, Cui Y. Acupuncture treatment of vascular cognitive impairment through peripheral nerve stimulation pathway: a scoping review. Front Aging Neurosci 2025; 17:1515327. [PMID: 40357233 PMCID: PMC12066784 DOI: 10.3389/fnagi.2025.1515327] [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: 10/22/2024] [Accepted: 04/14/2025] [Indexed: 05/15/2025] Open
Abstract
Objective This study aims to explore the central effects of acupuncture on vascular cognitive impairment (VCI) through peripheral nerve stimulation. Methods This scoping review followed the methodological framework proposed by Arksey and O'Malley and the PRISMA-ScR guidelines. A comprehensive search of databases, including PubMed, Web of Science, MEDLINE, and Embase, was conducted, including 79 studies on acupuncture interventions for VCI. Acupoints and their underlying anatomical structures related to peripheral nerves were summarized, and the potential pathways of acupuncture effects via different peripheral nerves were explored. Results The results showed that acupuncture, by stimulating specific acupoints on the head, face, torso, and limbs, significantly affects peripheral nerve networks, including the cervical, lumbar, and sacral plexuses, thoracic nerves, vagus nerve, trigeminal nerve and its branches. The nerve stimulation effects of acupuncture can enhance the regulation of cerebral blood flow, modulate neuroimmune responses, improve brain function, and promote neuroplasticity through multiple central nervous system pathways, ultimately improving cognitive function and treating VCI. Conclusion Acupuncture is a treatment modality that influences the central nervous system through peripheral nerve stimulation to treat VCI. A deeper understanding of the central effects induced by acupuncture-triggered neural reflexes can contribute to the improvement of existing therapies and help elucidate the scientific principles underlying acupuncture's therapeutic effects.
Collapse
Affiliation(s)
- Xinming Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Bo Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Qinghai Provincial Hospital of Traditional Chinese Medicine, Xining, China
| | - Linna Wu
- The First Clinical Medical School, Yunnan University of Chinese Medicine, Kunming, China
| | - Ying Cui
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
8
|
Saggu S, Pless A, Dew E, Ware D, Jiao K, Wang Q. Monoamine signaling and neuroinflammation: mechanistic connections and implications for neuropsychiatric disorders. Front Immunol 2025; 16:1543730. [PMID: 40356905 PMCID: PMC12066344 DOI: 10.3389/fimmu.2025.1543730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 04/04/2025] [Indexed: 05/15/2025] Open
Abstract
Monoamines, including norepinephrine, serotonin, and dopamine, orchestrate a broad spectrum of neurophysiological and homeostatic events. Recent research shows a pivotal role for monoaminergic signaling in modulating neuroinflammation by regulating proinflammatory cytokines and chemokines within the central nervous system. Importantly, this modulation is not unidirectional; released proinflammatory cytokines markedly "feedback" to influence the metabolism of monoamine neurotransmitters, impacting their synthesis, release, and reuptake. This bidirectional interplay significantly links monoaminergic pathways and neuroinflammatory responses. In this review, we summarize current knowledge of the dynamic interactions between monoamine signaling and neuroinflammation, as well as their critical implications for the pathophysiology of neuropsychiatric disorders, including Parkinson's Disease, Major Depressive Disorder, and Alzheimer's Disease. By integrating recent findings, we shed light on potential therapeutic targets within these interconnected pathways, providing insights into novel treatment strategies for these devastating disorders.
Collapse
Affiliation(s)
- Shalini Saggu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Andrew Pless
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Emily Dew
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Destany Ware
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Kai Jiao
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Qin Wang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
| |
Collapse
|
9
|
Mravec B, Szantova M. Liver Neurobiology: Regulation of Liver Functions by the Nervous System. Semin Liver Dis 2025. [PMID: 40239709 DOI: 10.1055/a-2562-2000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
The nervous system plays an important role in the regulation of liver functions during physiological as well as pathological conditions. This regulatory effect is based on the processing of signals transmitted to the brain by sensory nerves innervating the liver tissue and other visceral organs and by humoral pathways transmitting signals from peripheral tissues and organs. Based on these signals, the brain modulates metabolism, detoxification, regeneration, repair, inflammation, and other processes occurring in the liver. The nervous system thus determines the functional and morphological characteristics of the liver. Liver innervation also mediates the influence of psychosocial factors on liver functions. The aim of this review is to describe complexity of bidirectional interactions between the brain and liver and to characterize the mechanisms and pathways through which the nervous system influences liver function during physiological conditions and maintains liver and systemic homeostasis.
Collapse
Affiliation(s)
- Boris Mravec
- Department of Physiology Faculty of Medicine, Comenius University, Bratislava, Slovakia
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Maria Szantova
- 3rd Department of Internal Medicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| |
Collapse
|
10
|
Herta T, Bhattacharyya A, Hippenstiel S, Zahlten J. The role of KLF4 in phagocyte activation during infectious diseases. Front Immunol 2025; 16:1584873. [PMID: 40313940 PMCID: PMC12044337 DOI: 10.3389/fimmu.2025.1584873] [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: 02/27/2025] [Accepted: 03/31/2025] [Indexed: 05/03/2025] Open
Abstract
Phagocytes, including granulocytes (especially neutrophils), monocytes, macrophages, and dendritic cells, are essential components of the innate immune system, bridging innate and adaptive immunity. Their activation and function are tightly regulated by transcription factors that coordinate immune responses. Among these, Krüppel-like factor 4 (KLF4) has gained attention as a regulator of phagocyte differentiation, polarization, and inflammatory modulation. However, its role is highly context-dependent, exhibiting both pro- and anti-inflammatory properties based on environmental signals, cellular states, and the invading pathogen. KLF4 influences monocyte-to-macrophage differentiation and shapes macrophage polarization, promoting either inflammatory or regulatory phenotypes depending on external cues. In neutrophils, it affects reactive oxygen species production and immune activation, while in dendritic cells, it regulates monocyte-to-dendritic cell differentiation and cytokine secretion. Its diverse involvements in immune responses suggests that it contributes to maintaining a balance between effective pathogen defense and the prevention of excessive and potentially harmful inflammation. This review summarizes current knowledge on the function of KLF4 in phagocytes during infections, highlighting its regulatory mechanisms, context-dependent roles, and its impact on immune activation and resolution. Additionally, potential implications for therapeutic interventions targeting KLF4 are discussed.
Collapse
Affiliation(s)
- Toni Herta
- Department of Hepatology and Gastroenterology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health at Charité –Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, Berlin, Germany
| | - Aritra Bhattacharyya
- Department of Respiratory Medicine and Critical Care Medicine with Sleep Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Stefan Hippenstiel
- Department of Respiratory Medicine and Critical Care Medicine with Sleep Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Janine Zahlten
- Department of Respiratory Medicine and Critical Care Medicine with Sleep Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
11
|
Cheng M, Jiang Z, Yang J, Sun X, Song N, Du C, Luo Z, Zhang Z. The role of the neuroinflammation and stressors in premenstrual syndrome/premenstrual dysphoric disorder: a review. Front Endocrinol (Lausanne) 2025; 16:1561848. [PMID: 40225329 PMCID: PMC11985436 DOI: 10.3389/fendo.2025.1561848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 03/10/2025] [Indexed: 04/15/2025] Open
Abstract
Premenstrual syndrome (PMS) and premenstrual dysphoric disorder (PMDD) are prevalent emotional disorders in females, characterized by cyclic variations in physiological stress responses and emotional symptoms that correspond with the menstrual cycle. Despite extensive research, the underlying causes of these disorders remain elusive. This review delves into the neurobiological mechanisms connecting stress-induced neuroinflammation with PMS/PMDD. Additionally, it traces the conceptual development and historical context of PMS/PMDD. The review further evaluates clinical evidence on the association between PMS/PMDD and stress, along with findings from both clinical and animal studies that link these disorders to inflammatory processes. Additionally, the neurobiological pathways by which inflammatory responses may play a role in the pathogenesis of PMS/PMDD were elucidated, including their interactions with the hypothalamic-pituitary-ovary (HPO) axis, serotonin-kynurenine (5-HT-KYN) system, GABAergic system, brain-derived neurotrophic factor (BDNF), hypothalamic-pituitary-adrena(HPA)axis and. Future research is encouraged to further investigate the pathogenesis of PMS/PMDD through the perspective of neuroinflammatory responses.
Collapse
Affiliation(s)
- Ming Cheng
- Yangsheng College of Traditional Chinese Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Zhaoshu Jiang
- Yangsheng College of Traditional Chinese Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Jie Yang
- Yangsheng College of Traditional Chinese Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Xu Sun
- Research and Development Department, Qinhuangdao Shanhaiguan Pharmaceutical Co., Ltd, Qinhuangdao, Hebei, China
| | - Nan Song
- Research and Development Department, Qinhuangdao Shanhaiguan Pharmaceutical Co., Ltd, Qinhuangdao, Hebei, China
| | - Chunyu Du
- Research and Development Department, Qinhuangdao Shanhaiguan Pharmaceutical Co., Ltd, Qinhuangdao, Hebei, China
| | - Zhenliang Luo
- Yangsheng College of Traditional Chinese Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Zhen Zhang
- Yangsheng College of Traditional Chinese Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
- Research and Development Department, Qinhuangdao Shanhaiguan Pharmaceutical Co., Ltd, Qinhuangdao, Hebei, China
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
12
|
Meng Y, Li Y, Gu H, Chen Z, Cui X, Wang X. Androgen receptors in corticotropin-releasing hormone neurons mediate the sexual dimorphism in restraint-induced thymic atrophy. Proc Natl Acad Sci U S A 2025; 122:e2426107122. [PMID: 40106355 PMCID: PMC11962470 DOI: 10.1073/pnas.2426107122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 02/18/2025] [Indexed: 03/22/2025] Open
Abstract
Sexual dimorphism in immune responses is well documented, but the underlying mechanisms remain incompletely understood. Here, we identified a subset of corticotropin-releasing hormone (CRH) neurons that express androgen receptors (ARs) as key mediators of sex differences in restraint-induced immunosuppression. Mechanistically, androgens directly activate AR-positive CRH neurons, enhancing the hypothalamic-pituitary-adrenal axis activation. This results in elevated corticosterone levels in response to restraint stress, leading to increased immune cell apoptosis and immune organ atrophy in male mice. Conditional knockout of ARs in CRH neurons eliminated this sexual dimorphism, highlighting ARs in CRH neurons as pivotal regulators of sex-specific immune responses to stress.
Collapse
Affiliation(s)
- Yutong Meng
- National Institute of Biological Sciences, Beijing and Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing102206, China
| | - Yaning Li
- National Institute of Biological Sciences, Beijing and Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing102206, China
| | - Huating Gu
- Institute of Biophysics, Chinese Academy of Sciences, Beijing100101, China
| | - Ziyao Chen
- National Institute of Biological Sciences, Beijing and Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing102206, China
| | - Xiaoyang Cui
- Zhili College, Tsinghua University, Beijing100084, China
| | - Xiaodong Wang
- National Institute of Biological Sciences, Beijing and Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing102206, China
| |
Collapse
|
13
|
Dong W, Li Y, Fei Q, Li S, He X, Chai Y, Zhou J, Zong Y, Geng J, Li Z. Targeted spleen modulation: a novel strategy for next-generation disease immunotherapy. Theranostics 2025; 15:4416-4445. [PMID: 40225564 PMCID: PMC11984396 DOI: 10.7150/thno.111116] [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: 01/26/2025] [Accepted: 03/09/2025] [Indexed: 04/15/2025] Open
Abstract
The spleen, the largest lymphatic organ, comprises a diverse array of immunocytes in approximately one quarter of the body, including T cells, B cells, natural killer cells, and myeloid cells (such as dendritic cells, neutrophils, myeloid-derived suppressor cells, and macrophages). These immune cells undergo dynamic transitions and mobilization, enabling the spleen to execute a wide range of immunological functions. The spleen's structural organization and multicellular composition, along with its reservoir of lymphocytes, facilitate the capture and clearance of blood-borne antigens while also orchestrating both innate and adaptive immune responses. Additionally, the spleen plays critical roles in hematopoiesis and the removal of aged or damaged red blood cells. Despite being innervated by sympathetic (catecholaminergic) nerve fibers, the spleen lacks parasympathetic (vagal or cholinergic) innervation. The neuroimmune axis, particularly the interplay between sympathetic and parasympathetic nervous system immune circuits, significantly influences disease onset and progression. Extensive research employing physical, genetic, and pharmacological approaches has sought to directly modulate splenic immunocytes and activate neuroimmune interactions to restore immune homeostasis and counteract disease. Two primary mechanisms underlie these immunomodulatory interventions: (1) the cholinergic anti-inflammatory pathway, wherein norepinephrine released by splenic catecholaminergic fibers binds to β2-adrenergic receptors on CD4⁺ T cells, triggering acetylcholine secretion, which in turn suppresses inflammatory cytokine production in macrophages via α7 nicotinic acetylcholine receptor signaling, and (2) direct immunomodulation of splenic immunocytes, which regulates key genes and signaling pathways, alters cytokine secretion, and modulates ion flux to influence cellular functions. Among various therapeutic strategies, physical methods, particularly electrical stimulation and splenic ultrasound stimulation, have demonstrated the greatest promise for clinical applications in splenic immunomodulation and disease management.
Collapse
Affiliation(s)
- Wei Dong
- National and Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
- Department of Geriatric General Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
- Center for Tumor and Immunology, The Precision Medical Institute, Xi'an Jiaotong University, Xi'an, China
- Shaanxi Provincial Clinical Medical Research Center for Liver and Spleen Diseases, CHESS-Shaanxi consortium, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yucheng Li
- National and Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Qiaoman Fei
- National and Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
- Shaanxi Provincial Clinical Medical Research Center for Liver and Spleen Diseases, CHESS-Shaanxi consortium, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Senyang Li
- National and Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
- Shaanxi Provincial Clinical Medical Research Center for Liver and Spleen Diseases, CHESS-Shaanxi consortium, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Xinrui He
- National and Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yichao Chai
- National and Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
- Department of Geriatric General Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
- Shaanxi Provincial Clinical Medical Research Center for Liver and Spleen Diseases, CHESS-Shaanxi consortium, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Junyi Zhou
- National and Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yujin Zong
- National and Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Jing Geng
- National and Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
- Center for Tumor and Immunology, The Precision Medical Institute, Xi'an Jiaotong University, Xi'an, China
- Shaanxi Provincial Clinical Medical Research Center for Liver and Spleen Diseases, CHESS-Shaanxi consortium, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Zongfang Li
- National and Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
- Department of Geriatric General Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
- Center for Tumor and Immunology, The Precision Medical Institute, Xi'an Jiaotong University, Xi'an, China
- Shaanxi Provincial Clinical Medical Research Center for Liver and Spleen Diseases, CHESS-Shaanxi consortium, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
14
|
Van Den Noortgate M, Van Den Eede F, Coppens V, Giltay EJ, De Picker L, Morrens M. Immune-neuroendocrine crosstalk in mood and psychotic disorders: A meta-analysis and systematic review. Brain Behav Immun Health 2025; 44:100965. [PMID: 40040865 PMCID: PMC11879693 DOI: 10.1016/j.bbih.2025.100965] [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: 03/05/2024] [Revised: 02/09/2025] [Accepted: 02/10/2025] [Indexed: 03/06/2025] Open
Abstract
Background Bidirectional interactions between immune and neuroendocrine mechanisms are involved in mood and psychotic disorders, although individual studies report inconsistent and even contradictory findings on the nature of this crosstalk. Our objective was to perform an up to date systematic review and meta-analysis of the association between hypothalamic-pituitary-adrenal (HPA) axis and immune system functioning in mood and psychotic disorders. Methods We searched the Pubmed, Web of Science and Embase databases for studies reporting correlations between one or more HPA- and immune markers (IM) in patients with mood or psychotic disorders. We analyzed unchallenged correlations as well as challenge studies investigating the HPA-immune interaction through dexamethasone (DEX) and/or CRH suppression, HPA-mediated challenge of immune cell proliferation, immune challenges, or psychological stressors. Finally, genetic studies focusing on HPA x immune interrelation were evaluated. For meta-analyzable data, three primary outcome measures were defined for immune functioning, namely the pro-inflammatory index (PII) and anti-inflammatory index (AII) for the molecular IM and a composite cellular immune marker score (CCIM) for the cellular IM. Secondary analyses were performed for the individual molecular and cellular IM. Heterogeneity was evaluated with the I2 statistic. Meta-regression analyses were performed to evaluate the impact of potential covariates (publication year, gender, age, symptom severity) on the primary outcome analyses. Results 93 studies (n = 8226) were included, of which 50 (n = 5649) contained meta-analyzable data. The majority of the included studies (k = 72) investigated major depressive disorder (MDD) patients, nineteen schizophrenia spectrum disorders (SSD) and six bipolar disorder (BD). Under physiological conditions, a poor association was found between cortisol and the PII only in the unmedicated subsample of MDD (k = 8; n = 425; r = .205; z = 2.151; p = .031) and the medicated subsample of SSD (k = 4; n = 152; r = .0.237; z = 2.314; p = .021). No significant correlation was found in MDD between the AII and cortisol (k = 3; n = 1243; r = .005; z = .188; p = .851). Similar results were found for the association between immune cell numbers and cortisol in both MDD (k = 10; n = 773; r = -.005; z = -.113; p = .894) and SSD (k = 4; n = 99; r = .167; z = 1.356; p = .175). A total of 42 studies discussed post-challenge associations between immune alterations and HPA disturbances, of which 12 (n = 389; all MDD) contained meta-analyzable data and 37 entered the systematic review (n = 1783). No post-DEX correlations were found between cortisol and PII (k = 3; n = 105; r = .074; z = .355; p = .722) or CCIM (k = 5; n = 259; r = -.153; z = -1.294; p = .196). However, a significant association was found between post-DEX cortisol/ACTH and PII produced by stimulated blood cells in vitro (k = 3; n = 61; r = .508; z = 4.042; p < .001) as well as for cortisol and CCIM score in MDD after in vitro mitogen stimulation (k = 4; n = 90; r = -.309; z = -2498; p = .012). Following a psychological stressor (k = 6; n = 121), cortisol responses tended to be blunted in all included pathologies, while immune activation was comparable to healthy controls. Genetic studies (k = 7; n = 464) demonstrate altered gene expression of glucocorticoid receptors (GR) in peripheral immune cells in MDD. Heterogeneity over studies tended to be moderate to high. Discussion The main limitations are the heterogeneity of outcome measures (both HPA and IM) and small sample sizes of the included studies. We conclude that, in physiological conditions, associations between HPA-axis and molecular or cellular IM are absent or poor in both MDD and SSD and psychotropic medication may influence this crosstalk differently in both patient groups. Studies using challenge paradigms in MDD populations did reveal differences in the HPA-immune crosstalk. The normally expected decrease in lymphocytes after DEX distribution tended to be less pronounced in MDD, especially in glucocorticoid-insensitive non-suppressors. It is recommended that future studies should be properly powered and assess HPA functioning using multiple cortisol assessments. Challenge studies are probably more useful than baseline biomarker studies and cellular IM are more informative than molecular IM. It is recommended to broadly assess leucocyte function and, when possible, perform subgroup analyses based on HPA- and/or immune function.
Collapse
Affiliation(s)
- Minne Van Den Noortgate
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Filip Van Den Eede
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
- University Department of Psychiatry, Campus Antwerp University Hospital, Edegem, Belgium
| | - Violette Coppens
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Erik J. Giltay
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
- Department of Psychiatry, Leiden University Medical Center, Leiden, the Netherlands
- Department of Public Health and Primary Care, Health Campus the Hague, Leiden University Medical Center, The Hague, the Netherlands
| | - Livia De Picker
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
- Scientific Initiative of Neuropsychiatric and Psychopharmacological Studies (SINAPS), University Psychiatric Centre Campus Duffel, Duffel, Belgium
| | - Manuel Morrens
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
- Scientific Initiative of Neuropsychiatric and Psychopharmacological Studies (SINAPS), University Psychiatric Centre Campus Duffel, Duffel, Belgium
| |
Collapse
|
15
|
Soranno DE, Coopersmith CM, Brinkworth JF, Factora FNF, Muntean JH, Mythen MG, Raphael J, Shaw AD, Vachharajani V, Messer JS. A review of gut failure as a cause and consequence of critical illness. Crit Care 2025; 29:91. [PMID: 40011975 PMCID: PMC11866815 DOI: 10.1186/s13054-025-05309-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 02/05/2025] [Indexed: 02/28/2025] Open
Abstract
In critical illness, all elements of gut function are perturbed. Dysbiosis develops as the gut microbial community loses taxonomic diversity and new virulence factors appear. Intestinal permeability increases, allowing for translocation of bacteria and/or bacterial products. Epithelial function is altered at a cellular level and homeostasis of the epithelial monolayer is compromised by increased intestinal epithelial cell death and decreased proliferation. Gut immunity is impaired with simultaneous activation of maladaptive pro- and anti-inflammatory signals leading to both tissue damage and susceptibility to infections. Additionally, splanchnic vasoconstriction leads to decreased blood flow with local ischemic changes. Together, these interrelated elements of gastrointestinal dysfunction drive and then perpetuate multi-organ dysfunction syndrome. Despite the clear importance of maintaining gut homeostasis, there are very few reliable measures of gut function in critical illness. Further, while multiple therapeutic strategies have been proposed, most have not been shown to conclusively demonstrate benefit, and care is still largely supportive. The key role of the gut in critical illness was the subject of the tenth Perioperative Quality Initiative meeting, a conference to summarize the current state of the literature and identify key knowledge gaps for future study. This review is the product of that conference.
Collapse
Affiliation(s)
- Danielle E Soranno
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Craig M Coopersmith
- Department of Surgery and Emory Critical Care Center, Emory University, Atlanta, GA, USA
| | - Jessica F Brinkworth
- Department of Anthropology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Faith N F Factora
- Intensive Care and Resuscitation, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Julia H Muntean
- Intensive Care and Resuscitation, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Monty G Mythen
- Perioperative Medicine, University College London, London, England
| | - Jacob Raphael
- Anesthesiology and Perioperative Medicine, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Andrew D Shaw
- Intensive Care and Resuscitation, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Vidula Vachharajani
- Department of Pulmonary and Critical Care, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Jeannette S Messer
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
16
|
Zhao D, Sui Y, Sun Y. Causal Relationship Between Immune Cells and Hypopituitarism: Bidirectional Mendelian Randomization Study. World Neurosurg 2025; 194:123411. [PMID: 39522807 DOI: 10.1016/j.wneu.2024.10.140] [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: 10/22/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Hypopituitarism, one or more pituitary hormones inefficiently produced by the anterior pituitary or released from the posterior pituitary to adapt to the needs of the organism. Existing epidemiological data show that immune-mediated diffuse infiltration of the anterior pituitary is important in the development of hypopituitarism. However, the precise connection between immune cells and hypopituitarism remains unclear. This study aimed to elucidate the potential causal links between the 731 immune cell types and hypopituitarism risk. METHODS Based on data from a genome-wide association study, a bidirectional two-sample Mendelian randomization analysis was performed using 5 methods to explore the potential influence of immune cell phenotypes on hypopituitarism. Sensitivity analyses were conducted to examine the robustness of these findings. RESULTS Our findings support that B cells, T cells, Tregs, dendritic cells, monocytes, and myeloid cells each have a bidirectional influence on hypopituitarism. One B-cell phenotype was associated with increased hypopituitarism risk, while 2 B-cell phenotypes play a protective role in hypopituitarism. Moreover, 7 T-cell phenotypes demonstrate significant protective properties on hypopituitarism. Seven Tregs were associated with increased hypopituitarism risk. Furthermore, one monocyte was identified to be significantly associated with hypopituitarism risk. In addition, reverse Mendelian randomization analysis revealed that hypopituitarism was positively associated with 30 additional immune cell phenotypes and negatively associated with 17 immune cells. CONCLUSIONS Our investigation shed light on the intricate potential relationship between immune cells and hypopituitarism via genetic methods, underscoring the immune-mediated pathway in hypopituitarism pathogenesis, thereby offering valuable insights for future clinical investigations.
Collapse
Affiliation(s)
- Dadi Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Yuan Sui
- Department of Neurosurgery, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Yesheng Sun
- Department of Cellular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany.
| |
Collapse
|
17
|
Campos-Sánchez JC, Cabrera-Álvarez MJ, Saraiva JL. Review of Fish Neuropeptides: A Novel Perspective on Animal Welfare. J Comp Neurol 2025; 533:e70029. [PMID: 40008573 DOI: 10.1002/cne.70029] [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: 02/23/2024] [Revised: 10/09/2024] [Accepted: 01/28/2025] [Indexed: 02/27/2025]
Abstract
Neuropeptides are highly variable but widely conserved molecules, the main functions of which are the regulation and coordination of physiological processes and behaviors. They are synthesized in the nervous system and generally act on other neuronal and non-neuronal tissues or organs. In recent years, diverse neuropeptide isoforms and their receptors have been identified in different fish species, regulating functions in the neuroendocrine (e.g., corticotropin-releasing hormone and arginine vasotocin), immune (e.g., vasoactive intestinal polypeptide and somatostatin), digestive (e.g., neuropeptide Y), and reproductive (e.g., isotocin) systems, as well as in the commensal microbiota. Interestingly, all these processes carried out by neuropeptides are integrated into the nervous system and are manifested externally in the behavior and affective states of fish, thus having an impact on the modulation of these actions. In this sense, the monitoring of neuropeptides may represent a new approach to assess animal welfare, targeting both physiological and affective aspects in fish. Therefore, although there are many studies investigating the action of neuropeptides in a wide range of paradigms, especially in mammals, their study within a fish welfare framework is scarce. To the best of our knowledge, this is the first review that gathers and integrates up-to-date information on neuropeptides from an animal welfare perspective. In this review, we summarize current findings on neuropeptides in fish and discuss their possible implication in the physiological and emotional state of fish, and therefore in their welfare.
Collapse
Affiliation(s)
- Jose Carlos Campos-Sánchez
- Immunobiology for Aquaculture group, Department of Cell Biology and Histology, Faculty of Biology, Campus Regional de Excelencia Internacional "Campus Mare Nostrum", University of Murcia, Murcia, Spain
- Fish Ethology and Welfare Group, Centro de Ciências do Mar (CCMAR), Campus de Gambelas, Edificio 7, Universidade do Algarve - CCMAR/CIMAR-LA, Faro, Portugal
| | - María José Cabrera-Álvarez
- Fish Ethology and Welfare Group, Centro de Ciências do Mar (CCMAR), Campus de Gambelas, Edificio 7, Universidade do Algarve - CCMAR/CIMAR-LA, Faro, Portugal
- FishEthoGroup Association, Incubadora de Empresas da Universidade do Algarve Campus de Gambelas, pavilhão B1 8005-226, Faro, Portugal
| | - Joao L Saraiva
- Fish Ethology and Welfare Group, Centro de Ciências do Mar (CCMAR), Campus de Gambelas, Edificio 7, Universidade do Algarve - CCMAR/CIMAR-LA, Faro, Portugal
- FishEthoGroup Association, Incubadora de Empresas da Universidade do Algarve Campus de Gambelas, pavilhão B1 8005-226, Faro, Portugal
| |
Collapse
|
18
|
Onisiforou A, Charalambous EG, Zanos P. Shattering the Amyloid Illusion: The Microbial Enigma of Alzheimer's Disease Pathogenesis-From Gut Microbiota and Viruses to Brain Biofilms. Microorganisms 2025; 13:90. [PMID: 39858858 PMCID: PMC11767882 DOI: 10.3390/microorganisms13010090] [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: 12/11/2024] [Revised: 12/18/2024] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
For decades, Alzheimer's Disease (AD) research has focused on the amyloid cascade hypothesis, which identifies amyloid-beta (Aβ) as the primary driver of the disease. However, the consistent failure of Aβ-targeted therapies to demonstrate efficacy, coupled with significant safety concerns, underscores the need to rethink our approach to AD treatment. Emerging evidence points to microbial infections as environmental factors in AD pathoetiology. Although a definitive causal link remains unestablished, the collective evidence is compelling. This review explores unconventional perspectives and emerging paradigms regarding microbial involvement in AD pathogenesis, emphasizing the gut-brain axis, brain biofilms, the oral microbiome, and viral infections. Transgenic mouse models show that gut microbiota dysregulation precedes brain Aβ accumulation, emphasizing gut-brain signaling pathways. Viral infections like Herpes Simplex Virus Type 1 (HSV-1) and Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) may lead to AD by modulating host processes like the immune system. Aβ peptide's antimicrobial function as a response to microbial infection might inadvertently promote AD. We discuss potential microbiome-based therapies as promising strategies for managing and potentially preventing AD progression. Fecal microbiota transplantation (FMT) restores gut microbial balance, reduces Aβ accumulation, and improves cognition in preclinical models. Probiotics and prebiotics reduce neuroinflammation and Aβ plaques, while antiviral therapies targeting HSV-1 and vaccines like the shingles vaccine show potential to mitigate AD pathology. Developing effective treatments requires standardized methods to identify and measure microbial infections in AD patients, enabling personalized therapies that address individual microbial contributions to AD pathogenesis. Further research is needed to clarify the interactions between microbes and Aβ, explore bacterial and viral interplay, and understand their broader effects on host processes to translate these insights into clinical interventions.
Collapse
Affiliation(s)
- Anna Onisiforou
- Translational Neuropharmacology Laboratory, Department of Psychology, University of Cyprus, 75 Kallipoleos Avenue, 1678 Nicosia, Cyprus;
- Center of Applied Neuroscience, 75 Kallipoleos Avenue, 1678 Nicosia, Cyprus
| | - Eleftheria G. Charalambous
- Translational Neuropharmacology Laboratory, Department of Psychology, University of Cyprus, 75 Kallipoleos Avenue, 1678 Nicosia, Cyprus;
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, 1–2, Ellernholzstr., 17489 Greifswald, Germany
| | - Panos Zanos
- Translational Neuropharmacology Laboratory, Department of Psychology, University of Cyprus, 75 Kallipoleos Avenue, 1678 Nicosia, Cyprus;
- Center of Applied Neuroscience, 75 Kallipoleos Avenue, 1678 Nicosia, Cyprus
| |
Collapse
|
19
|
McCain KM, Mansilla G, Sheldon EL, Zimmer C, Schrey AW, Rowe M, Dor R, Kohl KD, Søraker JS, Jensen H, Mathot KJ, Vu T, Phuong HT, Jimeno B, Buchanan KL, Thiam M, Briskie J, Martin LB. Microbial surveillance versus cytokine responsiveness in native and non-native house sparrows. Biol Lett 2025; 21:20240431. [PMID: 39878139 PMCID: PMC11776021 DOI: 10.1098/rsbl.2024.0431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/24/2024] [Accepted: 11/12/2024] [Indexed: 01/31/2025] Open
Abstract
The success of introduced species often relies on flexible traits, including immune system traits. While theories predict non-natives will have weak defences due to decreased parasite pressure, effective parasite surveillance remains crucial, as infection risk is rarely zero and the evolutionary novelty of infection is elevated in non-native areas. This study examines the relationship between parasite surveillance and cytokine responsiveness in native and non-native house sparrows, hypothesizing that non-natives maintain high pathogen surveillance while avoiding costly inflammation. We made this specific prediction, as this pattern could enable invaders to effectively mitigate pathogen risk in a manner commensurate with the life-history priorities of a colonizing organism (i.e. rapid maturation and high reproductive effort). To test this hypothesis, we measured TLR-2 and TLR-4 expression, markers of pathogen surveillance and cytokine responses (changes in IL-1β and IL-10), regulators of inflammation, to a simulated bacterial infection. In non-native sparrows, we found that as TLR-4 expression increased, IL-1β and IL-10 responses decreased, a relationship not observed in native sparrows. Additionally, higher body condition predicted larger IL-1β and IL-10 responses in all birds. These findings suggest that high TLR-4 surveillance may mitigate strong inflammatory responses in non-native sparrows, with pathological and resource-based costs driving immune variation among and within populations.
Collapse
Affiliation(s)
- Kailey M. McCain
- Global Enviornmental and Genomic Health Sciences, University of South Florida, Tampa, FL33612, USA
| | - Gabby Mansilla
- Global Enviornmental and Genomic Health Sciences, University of South Florida, Tampa, FL33612, USA
| | - Elizabeth L. Sheldon
- Sorbonne Université, Villefranche Oceanography Laboratory, 181 Chem. du Lazaret, Villefranche-sur-Mer06230, France
| | - Cedric Zimmer
- Laboratoire d’Ethologie Expérimentale et Comparée, LEEC, Université Sorbonne Paris Nord, UR 4443, Villetaneuse93430, France
| | - Aaron W. Schrey
- Department of Biology, Georgia Southern University Armstrong Campus, Science Center 1505,16 11935 Abercorn Street, Savannah, GA31419, USA
| | - Melissah Rowe
- Department of Animal Ecology, Netherlands Institute of Ecology (NIOO‐KNAW), Wageningen6700 AB, The Netherlands
| | - Roi Dor
- Department of Natural Sciences, The Open University of Israel, Ra’anana, Israel
| | - Kevin D. Kohl
- Department of Biological Sciences, University of Pittsburgh, 4249 Fifth Avenue Pittsburgh, Pittsburgh, PA15260, USA
| | - Jørgen S. Søraker
- Department of Biology, Centre for Conservation Biology, Norwegian University of Science and Technology, N-7491 Trondheim, Trondheim, Norway
| | - Henrik Jensen
- Department of Biology, Centre for Conservation Biology, Norwegian University of Science and Technology, N-7491 Trondheim, Trondheim, Norway
| | - Kimberley J. Mathot
- Canada Research Chair in Integrative Ecology, Department of Biological Sciences,CW405 Biological Sciences Building, University of Alberta, EdmontonAB T6G 2E9, Canada
| | - Thinh Vu
- Department of Wildlife, Faculty of Forest Resource and Environmental Management, Vietnam National University of Forestry, Chương Mỹ, Vietnam
| | - Ho Thu Phuong
- Department of Wildlife, Faculty of Forest Resource and Environmental Management, Vietnam National University of Forestry, Chương Mỹ, Vietnam
| | - Blanca Jimeno
- Department of Biological Conservation and Ecosystem Restoration, Pyrenean Institute of Ecology (IPE-CSIC). Av. Nuestra Señora de la Victoria, 22700, Jaca, Spain
| | - Katherine L. Buchanan
- School of Life and Environmental Sciences, Deakin University Geelong, Geelong, Vic3216, Australia
| | - Massamba Thiam
- Laboratory of Zoology of Terrestrial Vertebrates, Fundamental Institute of Black Africa,Cheikh Anta Diop University of Dakar Senegal, Dakar, Senegal
| | - James Briskie
- School of Biological Sciences, University of Canterbury, Christchurch, New Zealand
| | - Lynn B. Martin
- Global Enviornmental and Genomic Health Sciences, University of South Florida, Tampa, FL33612, USA
| |
Collapse
|
20
|
Pierre-Jerome C. The peripheral nervous system: peripheral neuropathies in the diabetic foot. MYOPATHIES AND TENDINOPATHIES OF THE DIABETIC FOOT 2025:451-482. [DOI: 10.1016/b978-0-443-13328-2.00022-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
21
|
Abstract
PURPOSE OF REVIEW This editorial aims to highlight the evolving concept of protective hemodynamics in the management of critically ill patients. RECENT FINDINGS Recent literature underscores the limitations of rigid blood pressure targets, particularly in the context of critical care and perioperative management. High blood pressure targets, especially when coupled with high-dose vasopressors, can lead to poor outcomes. 'Protective hemodynamics' aims to maintain cardiovascular stability while reducing risks associated with interventions. SUMMARY The implications of adopting protective hemodynamics are profound for both clinical practice and research. Clinically, this approach can reduce iatrogenic harm and improve long-term outcomes for critically ill patients. For research, it opens new avenues for investigating individualized hemodynamic management strategies that prioritize overall patient stability and long-term health over rigid target attainment.
Collapse
Affiliation(s)
- Filippo D'Amico
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute
| | - Giovanni Landoni
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
22
|
Zhang Y, Ai H, Zhang P, Li B. C-Type Lectin S Group TcCTL4 Participates in the Immunity of Tribolium castaneum. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2024; 117:e70012. [PMID: 39689093 DOI: 10.1002/arch.70012] [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: 09/06/2024] [Revised: 10/29/2024] [Accepted: 11/18/2024] [Indexed: 12/19/2024]
Abstract
C-type lectin S (CTL-S) plays a crucial role in pathogen recognition and the activation of immune response. In comparison, the proportion of CTL-S was relatively high in insects, but the study was much smaller than the proportion observed. In this study, we cloned and characterized one CTL-S, TcCTL4, from Tribolium castaneum. Our analysis revealed that TcCTL4 was highly expressed during the early pupal stage, with expression levels exhibiting a tendency to change with developmental stages. Additionally, tissue expression analysis showed a high expression of TcCTL4 in the central nervous system (CNS). Moreover, we observed a significant increase in TcCTL4 transcripts after bacterial challenge. The RNA interference (RNAi) of TcCTL4 before bacterial treatment resulted in a significant reduction in the transcripts of immune factors (IFs) and antimicrobial peptides (AMPs), indicating that TcCTL4 may regulate AMP expression through the activation of the immune signaling pathway. Furthermore, our investigation revealed that the recombinant protein TcCTL4 (rTcCTL4) not only recognized bacteria but also agglutinated bacteria in a Ca2+-dependent manner. Enzyme activity analysis suggested that rTcCTL4 could enhance phenoloxidase activity, implying its potential involvement in the prophenoloxidase activation pathway. In conclusion, these results indicate that TcCTL4 is involved in the immunity of T. castaneum, providing valuable insights into insect CTLs.
Collapse
Affiliation(s)
- Yonglei Zhang
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
- State Key Laboratory of Medical Genomics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Huayi Ai
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Ping Zhang
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Bin Li
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| |
Collapse
|
23
|
Besedovsky H, Del Rey A. A Glucocorticoid-Mediated Immunoregulatory Circuit Integrated at Brain Levels: Our Early Studies and a Present View. Neuroimmunomodulation 2024; 31:230-245. [PMID: 39504948 DOI: 10.1159/000542401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/29/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND It was known since the 1940s that pharmacological administration of glucocorticoids can inhibit inflammatory and immune processes, and these hormones are still today among the most widely used therapeutic tools to treat diseases with immune components. However, it became clear later that endogenous glucocorticoids can either support or restrain immune processes. SUMMARY Early studies showed that (a) endogenous levels of glucocorticoids can modulate immune cell activity; (b) the immune response itself can stimulate the hypothalamus-pituitary-adrenal (HPA) axis to release glucocorticoids to levels that can exert immunoregulatory effects; (c) immune products, later identified as cytokines, mediate this effect. On these bases, the existence of a glucocorticoid-mediated immunoregulatory circuit was proposed. It was also shown that increased levels of endogenous glucocorticoids exert protective effects during infections and other diseases with immune components. However, it was found in animal models and in humans that these effects can be blunted in several immune-linked diseases by defects at several levels, for example, by glucocorticoid resistance or by adrenal insufficiency. Evidence was later provided that the glucocorticoid-mediated immunoregulatory circuit can also be activated by cytokines produced not only as consequence of immune stimulation but also following psycho/sensorial and physical stimuli. Thus, this circuit can be integrated at brain levels and, besides stimulating the HPA axis, cytokines can also affect synaptic plasticity, most likely via a tripartite synapse, with astrocytes as neuro-immune cells acting as the third component. KEY MESSAGES It is now well established that the glucocorticoid-mediated immunoregulatory circuit plays a central role in maintaining health. However, several variables can condition the efficacy of the effect of endogenous glucocorticoids. Furthermore, since cytokines and other immune products have many other neuroendocrine and metabolic effects, other neuroendocrine-immune circuits could simultaneously operate or become predominant during different pathologies. The consideration of these aspects might help to implement strategies to eventually decrease therapeutic doses of exogenous glucocorticoids.
Collapse
Affiliation(s)
- Hugo Besedovsky
- Research Group Immunophysiology, Department Neurophysiology, Institute of Physiology and Pathophysiology, Marburg, Germany
| | - Adriana Del Rey
- Research Group Immunophysiology, Department Neurophysiology, Institute of Physiology and Pathophysiology, Marburg, Germany
| |
Collapse
|
24
|
Joung JY, Lee YH, Son CG. An evolutionary perspective for integrating mechanisms of acupuncture therapy. Explore (NY) 2024; 20:103060. [PMID: 39278099 DOI: 10.1016/j.explore.2024.103060] [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: 01/19/2024] [Revised: 08/03/2024] [Accepted: 09/04/2024] [Indexed: 09/17/2024]
Abstract
This study applies an evolutionary medicine perspective to comprehend the therapeutic effects of acupuncture. It draws upon modern evolutionary theory to integrate the currently fragmented theories regarding the efficacy of acupuncture in alleviating pain and promoting healing. We explore the interaction between the nervous and immune systems in the context of survival and homeostasis, and elucidate both the local and systemic effects of acupuncture therapy on pain relief and tissue healing. The mechanisms involved are categorized into two main types: local effects, which include immune cell migration, local vasodilation, and the release of adenosine; and distal systemic effects, which involve the regulation of the descending pain control system and the autonomic nervous system, with a particular focus on the parasympathetic nervous system. In conclusion, this integrated perspective not only deepens our understanding of acupuncture within a scientific narrative but also underscores the need for further research to validate and expand our knowledge, thereby enhancing its scientific credibility and clinical applicability.
Collapse
Affiliation(s)
- Jin-Yong Joung
- Department of Internal Medicine, Daejeon Good-morning Oriental Hospital, 21, Dunsan-ro 123beon-gil, Seo-gu, Daejeon 35240, Republic of Korea; Department of Korean Medicine, Korean Medical College of Daejeon University, Daehak-ro 62, Dong-gu, Daejeon 34520, Republic of Korea
| | - Young Ho Lee
- Department of Anatomy, College of Medicine, Chungnam National University, 266, Munhwa-ro, Jung-gu, Daejeon 35015, Republic of Korea
| | - Chang-Gue Son
- Liver and Immunology Research Center, Daejeon Oriental Hospital of Daejeon University, Daedukdae-ro 176 bun-gil 75, Seo-gu, Daejeon 35235, Republic of Korea; Department of Korean Medicine, Korean Medical College of Daejeon University, Daehak-ro 62, Dong-gu, Daejeon 34520, Republic of Korea.
| |
Collapse
|
25
|
Zhang G, Wang S, Ma P, Pan J. Neurophysiology and Molecular Basis of Cognitive Behavioral Therapy for Patients with Insomnia: Implications for Non-Pharmacological Approaches. J Integr Neurosci 2024; 23:200. [PMID: 39613462 DOI: 10.31083/j.jin2311200] [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: 03/21/2024] [Revised: 06/17/2024] [Accepted: 06/25/2024] [Indexed: 12/01/2024] Open
Abstract
Cognitive behavioral therapy for insomnia (CBT-I) is a widely used psychological intervention known for its effectiveness in improving insomnia symptoms. However, the neurophysiological mechanisms underlying the cognitive-behavioral treatment of insomnia remain unclear. This narrative review aimed to elucidate the neurophysiological and molecular mechanisms of CBT-I, focusing on the fields of psychology, neurophysiology, neuroendocrinology, immunology, medical microbiology, epigenetics, neuroimaging and brain function. A comprehensive search was conducted using databases including: PubMed, Embase, PsycINFO and Web of Science, with customized search strategies tailored to each database that included controlled vocabulary and alternative synonyms. It revealed that CBT-I may have a beneficial effect on the central nervous system, boost the immune system, upregulate genes involved in interferon and antibody responses, enhance functional connectivity between the hippocampus and frontoparietal areas and increase cortical gray matter thickness. In conclusion, an integrated model is proposed that elucidates the mechanisms of CBT-I and offers a new direction for investigations into its neurophysiological mechanisms.
Collapse
Affiliation(s)
- Guimei Zhang
- Department of Psychiatry, Sleep Medicine Centre, First Affiliated Hospital of Jinan University, 510632 Guangzhou, Guangdong, China
| | - Sisi Wang
- Department of Psychiatry, Sleep Medicine Centre, First Affiliated Hospital of Jinan University, 510632 Guangzhou, Guangdong, China
| | - Ping Ma
- Department of Psychiatry, Sleep Medicine Centre, First Affiliated Hospital of Jinan University, 510632 Guangzhou, Guangdong, China
| | - Jiyang Pan
- Department of Psychiatry, Sleep Medicine Centre, First Affiliated Hospital of Jinan University, 510632 Guangzhou, Guangdong, China
| |
Collapse
|
26
|
Qiao X, Bao L, Liu G, Cui X. Nanomaterial journey in the gut: from intestinal mucosal interaction to systemic transport. NANOSCALE 2024; 16:19207-19220. [PMID: 39347780 DOI: 10.1039/d4nr02480j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Engineered nanomaterials (NMs) are commonly utilized in food additives, cosmetics, and therapeutic applications due to their advantageous properties. Consequently, humans are frequently exposed to exogenous nanomaterials through oral ingestion, thus making the intestinal mucosal system a primary site for these particles. Understanding the interactions between nanomaterials and the intestinal mucosal system is crucial for harnessing their therapeutic potential and mitigating potential health risks from unintended exposure. This review aims to elucidate recent advancements in the dual effects of nanomaterials on the intestinal mucosal system. Upon entering the gut lumen, nanomaterials will interact with diverse intestinal components, including trillions of gut microbiota, mucus layer, intestinal epithelial cells (IECs), and the intestinal immune system. Additionally, the systemic fate and transportation of nanomaterials to distal organs, such as central nervous system, are also highlighted. These interactions result in a distinct biological effect of nanomaterials on the multilayer structure of intestine, thus displaying complex journeys and outcomes of nanomaterials in the living body. This in-depth exploration of the in vivo destiny and immunological implications of nanomaterials encountering the intestine has the potential to propel advancements in oral drug delivery techniques and motivate future investigations in novel toxicology research.
Collapse
Affiliation(s)
- Xin Qiao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Lin Bao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Guanyu Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Xuejing Cui
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| |
Collapse
|
27
|
Wang C, Feng GG, Takagi J, Fujiwara Y, Sano T, Note H. Catecholamines Attenuate LPS-Induced Inflammation through β2 Adrenergic Receptor Activation- and PKA Phosphorylation-Mediated TLR4 Downregulation in Macrophages. Curr Issues Mol Biol 2024; 46:11336-11348. [PMID: 39451555 PMCID: PMC11506017 DOI: 10.3390/cimb46100675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 10/05/2024] [Accepted: 10/11/2024] [Indexed: 10/26/2024] Open
Abstract
Inflammation is a tightly regulated process involving immune receptor recognition, immune cell migration, inflammatory mediator secretion, and pathogen elimination, all essential for combating infection and restoring damaged tissue. However, excessive inflammatory responses drive various human diseases. The autonomic nervous system (ANS) is known to regulate inflammatory responses; however, the detailed mechanisms underlying this regulation remain incompletely understood. Herein, we aimed to study the anti-inflammatory effects and mechanism of action of the ANS in RAW264.7 cells. Quantitative PCR and immunoblotting assays were used to assess lipopolysaccharide (LPS)-induced tumor necrosis factor α (TNFα) expression. The anti-inflammatory effects of catecholamines (adrenaline, noradrenaline, and dopamine) and acetylcholine were examined in LPS-treated cells to identify the receptors involved. Catecholamines inhibited LPS-induced TNFα expression by activating the β2 adrenergic receptor (β2-AR). β2-AR activation in turn downregulated the expression of Toll-like receptor 4 (TLR4) by stimulating protein kinase A (PKA) phosphorylation, resulting in the suppression of TNFα levels. Collectively, our findings reveal a novel mechanism underlying the inhibitory effect of catecholamines on LPS-induced inflammatory responses, whereby β2-AR activation and PKA phosphorylation downregulate TLR4 expression in macrophages. These findings could provide valuable insights for the treatment of inflammatory diseases and anti-inflammatory drug development.
Collapse
Affiliation(s)
- Cong Wang
- Department of Gastroenterological Surgery, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute 480-1195, Aichi, Japan; (C.W.); (T.S.)
| | - Guo-Gang Feng
- Department of Anesthesiology, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute 480-1195, Aichi, Japan;
| | - Junko Takagi
- Division of Endocirnology and Metabolism, Department of Internal Medicine, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute 480-1195, Aichi, Japan;
| | - Yoshihiro Fujiwara
- Department of Anesthesiology and Pain Medicine, Fujita Health University Bantane Hospital, 3-6-10 Otobashi, Nakagawaku, Nagoya 454-8509, Aichi, Japan;
| | - Tsuyoshi Sano
- Department of Gastroenterological Surgery, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute 480-1195, Aichi, Japan; (C.W.); (T.S.)
| | - Hideaki Note
- Department of Anesthesiology, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute 480-1195, Aichi, Japan;
| |
Collapse
|
28
|
Khoury RD, Minhoto GB, de Barros PP, Junqueira JC, Gagliardi CF, do Prado RF, Valera MC. Systemic administration of propranolol reduces bone resorption and inflammation in apical periodontitis of chronically stressed rats. Int Endod J 2024; 57:1478-1491. [PMID: 39003599 DOI: 10.1111/iej.14118] [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: 02/19/2024] [Revised: 05/21/2024] [Accepted: 06/20/2024] [Indexed: 07/15/2024]
Abstract
AIM To evaluate the effect of systemic administration of propranolol on the severity of apical periodontitis (AP) in chronically stressed rats. METHODOLOGY Twenty-four 70-day-old male Wistar rats (Rattus norvegicus, albinus) were distributed into three groups (n = 8): rats with AP without stressful conditions (AP-Control), rats with AP and submitted to a chronic unpredictable stress (CUS) protocol (AP + S) and rats with AP and submitted to a CUS protocol treated with propranolol (AP + S + PRO). Stress procedures were applied daily until the end of the experiment. After 3 weeks of CUS, AP was induced in all groups by exposing the pulpal tissue of mandibular and maxillary first molars to the oral environment. Propranolol treatment was administered orally once a day for the entire period of the experiment. Rats were sacrificed at 42 days, and the blood was collected for stress biomarkers serum dosage by multiplex assay. Mandibles were removed and submitted to microtomography and histopathological analyses. Periapical tissue surrounding the upper first molar was homogenized and subjected to RT-PCR analysis to evaluate the mRNA expression of RANKL, TRAP and OPG. Parametric data were assessed using one-way ANOVA followed by Tukey's test while the nonparametric data were analysed by the Kruskal-Wallis followed by Dunn's test. Significance level was set at 5% (p < .05) for all assessed parameters. RESULTS Micro-CT revealed statistically significant differences in bone resorption which was greater in the AP + S group (p < .05), but no differences were observed between the Control and AP + S + PRO groups (p > .05). The AP + S + PRO group had a lower intensity and extent of inflammatory infiltrate compared to the AP + S group with smaller areas of bone loss (p < 0.05). The gene expression of RANKL and TRAP was significantly higher in the stressed group AP + S compared to the control group (p < .05), and a significantly higher OPG expression was observed in AP + S + PRO compared to the AP + S group (p < .05). CONCLUSIONS Oral administration of propranolol had a significant effect on the AP severity in stressed rats, suggesting an anti-inflammatory effect and a protective role on bone resorption of AP in stressed animals. Further research is necessary to fully comprehend the underlying mechanisms.
Collapse
Affiliation(s)
- Rayana Duarte Khoury
- Department of Restorative Dentistry, Endodontic Division, São Paulo State University - UNESP, Institute of Science and Technology, São Paulo, Brazil
| | - Giovanna Bignoto Minhoto
- Department of Restorative Dentistry, Endodontic Division, São Paulo State University - UNESP, Institute of Science and Technology, São Paulo, Brazil
| | | | - Juliana Campos Junqueira
- Department of Bioscience and Oral Diagnosis, Institute of Science and Technology, São Paulo State University - UNESP, Institute of Science and Technology, São Paulo, Brazil
| | - Carolina Fedel Gagliardi
- Department of Restorative Dentistry, Endodontic Division, São Paulo State University - UNESP, Institute of Science and Technology, São Paulo, Brazil
| | - Renata Falchete do Prado
- Department of Bioscience and Oral Diagnosis, Institute of Science and Technology, São Paulo State University - UNESP, Institute of Science and Technology, São Paulo, Brazil
| | - Marcia Carneiro Valera
- Department of Restorative Dentistry, Endodontic Division, São Paulo State University - UNESP, Institute of Science and Technology, São Paulo, Brazil
| |
Collapse
|
29
|
Liu Z, Wang W, Zong Y, Li M, Gao Y, Xin X, Zhu T, Wang L, Song L. Norepinephrine regulates TNF expression via the A1AR-p38 MAPK-Relish pathway in granulocytes of oyster Crassostrea gigas. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 159:105217. [PMID: 38901503 DOI: 10.1016/j.dci.2024.105217] [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: 03/20/2024] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 06/22/2024]
Abstract
Norepinephrine (NE) is involved in regulating cytokine expression and phagocytosis of immune cells in the innate immunity of vertebrates. In the present study, the modulation mechanism of NE on the biosynthesis of TNFs in oyster granulocytes was explored. The transcripts of CgTNF-1, CgTNF-2 and CgTNF-3 were highly expressed in granulocytes, and they were significantly up-regulated after LPS stimulation, while down-regulated after NE treatment. The phagocytic rate and apoptosis index of oyster granulocytes were also triggered by LPS stimulation and suppressed by NE treatment. The mRNA expressions of CgMAPK14 and CgRelish were significantly induced after NE treatment, and the translocation of CgRelish from cytoplasm to nucleus was observed. The concentration of intracellular Ca2+ in granulocytes was significantly up-regulated upon NE incubation, and this trend reverted after the treatment with DOX (specific antagonist for NE receptor, CgA1AR-1). No obvious significance was observed in intracellular cAMP concentrations in the PBS, NE and NE + DOX groups. Once CgA1AR-1 was blocked by DOX, the mRNA expressions of CgMAPK14 and CgRelish were significantly inhibited, and the translocation of CgRelish from cytoplasm to nucleus was also dramatically suppressed, while the mRNA expression of CgTNF-1 and the apoptosis index increased significantly to the same level with those in LPS group, respectively. These results collectively suggested that NE modulated TNF expression in oyster granulocyte through A1AR-p38 MAPK-Relish signaling pathway.
Collapse
Affiliation(s)
- Zhaoqun Liu
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Weilin Wang
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Yanan Zong
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Meijia Li
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Yuqian Gao
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Xiaoyu Xin
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Functional Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Ting Zhu
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Lingling Wang
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Functional Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China.
| | - Linsheng Song
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Functional Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China.
| |
Collapse
|
30
|
Fatahzadeh M, Ravi A, Thomas P, Ziccardi VB. Systemic Factors Affecting Healing in Dentistry. Dent Clin North Am 2024; 68:799-812. [PMID: 39244258 DOI: 10.1016/j.cden.2024.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2024]
Abstract
Healing process in the oral cavity is influenced by a range of systemic factors. More specifically, patient health status, medications, habits, and nutritional state play crucial roles in dental healing. Additionally, the body's immune response, inflammation, and overall well-being are key determinants in wound repair. Understanding these systemic factors is essential for dental professionals to optimize patient care, minimize complications, and achieve successful healing.
Collapse
Affiliation(s)
- Mahnaz Fatahzadeh
- Division of Oral Medicine, Department of Diagnostic Sciences, Rutgers School of Dental Medicine, 110 Bergen Street, Newark, NJ 07103, USA
| | - Anjali Ravi
- University of Pittsburgh School of Dental Medicine, 341 Darragh Street, Unit 313, Pittsburgh, PA 15213, USA.
| | - Prisly Thomas
- Diplomate American Board of Orofacial Pain, Believers Church Medical College Hospital, St. Thomas Nagar Kuttapuzha, Thiruvalla Kerala-689103, India
| | - Vincent B Ziccardi
- Department of Oral and Maxillofacial Surgery, Rutgers School of Dental Medicine, Room B854, 110 Bergen Street, Newark, NJ 07103, USA
| |
Collapse
|
31
|
Pamminger T, Basley K, Goulson D, Hughes WOH. Potential acetylcholine-based communication in honeybee haemocytes and its modulation by a neonicotinoid insecticide. PeerJ 2024; 12:e17978. [PMID: 39285925 PMCID: PMC11404474 DOI: 10.7717/peerj.17978] [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: 03/03/2018] [Accepted: 08/06/2024] [Indexed: 09/19/2024] Open
Abstract
There is growing concern that some managed and wild insect pollinator populations are in decline, potentially threatening biodiversity and sustainable food production on a global scale. In recent years, there has been increasing evidence that sub-lethal exposure to neurotoxic, neonicotinoid pesticides can negatively affect pollinator immunocompetence and could amplify the effects of diseases, likely contributing to pollinator declines. However, a direct pathway connecting neonicotinoids and immune functions remains elusive. In this study we show that haemocytes and non-neural tissues of the honeybee Apis mellifera express the building blocks of the nicotinic acetylcholine receptors that are the target of neonicotinoids. In addition, we demonstrate that the haemocytes, which form the cellular arm of the innate immune system, actively express choline acetyltransferase, a key enzyme necessary to synthesize acetylcholine. In a last step, we show that the expression of this key enzyme is affected by field-realistic doses of clothianidin, a widely used neonicotinoid. These results support a potential mechanistic framework to explain the effects of sub-lethal doses of neonicotinoids on the immune function of pollinators.
Collapse
Affiliation(s)
- Tobias Pamminger
- School of Life Sciences, University of Sussex, Brighton, UK
- Bayer AG, Monheim am Rhein, Germany
| | - Kate Basley
- School of Life Sciences, University of Sussex, Brighton, UK
| | - Dave Goulson
- School of Life Sciences, University of Sussex, Brighton, UK
| | | |
Collapse
|
32
|
Mihalić A, Železnjak J, Lisnić B, Jonjić S, Juranić Lisnić V, Brizić I. Immune surveillance of cytomegalovirus in tissues. Cell Mol Immunol 2024; 21:959-981. [PMID: 39134803 PMCID: PMC11364667 DOI: 10.1038/s41423-024-01186-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 05/14/2024] [Indexed: 09/01/2024] Open
Abstract
Cytomegalovirus (CMV), a representative member of the Betaherpesvirinae subfamily of herpesviruses, is common in the human population, but immunocompetent individuals are generally asymptomatic when infected with this virus. However, in immunocompromised individuals and immunologically immature fetuses and newborns, CMV can cause a wide range of often long-lasting morbidities and even death. CMV is not only widespread throughout the population but it is also widespread in its hosts, infecting and establishing latency in nearly all tissues and organs. Thus, understanding the pathogenesis of and immune responses to this virus is a prerequisite for developing effective prevention and treatment strategies. Multiple arms of the immune system are engaged to contain the infection, and general concepts of immune control of CMV are now reasonably well understood. Nonetheless, in recent years, tissue-specific immune responses have emerged as an essential factor for resolving CMV infection. As tissues differ in biology and function, so do immune responses to CMV and pathological processes during infection. This review discusses state-of-the-art knowledge of the immune response to CMV infection in tissues, with particular emphasis on several well-studied and most commonly affected organs.
Collapse
Affiliation(s)
- Andrea Mihalić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Jelena Železnjak
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Berislav Lisnić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Stipan Jonjić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
- Department of Biomedical Sciences, Croatian Academy of Sciences and Arts, Rijeka, Croatia
| | - Vanda Juranić Lisnić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia.
| | - Ilija Brizić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia.
| |
Collapse
|
33
|
Stone TW, Williams RO. Tryptophan metabolism as a 'reflex' feature of neuroimmune communication: Sensor and effector functions for the indoleamine-2, 3-dioxygenase kynurenine pathway. J Neurochem 2024; 168:3333-3357. [PMID: 38102897 DOI: 10.1111/jnc.16015] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/16/2023] [Accepted: 11/08/2023] [Indexed: 12/17/2023]
Abstract
Although the central nervous system (CNS) and immune system were regarded as independent entities, it is now clear that immune system cells can influence the CNS, and neuroglial activity influences the immune system. Despite the many clinical implications for this 'neuroimmune interface', its detailed operation at the molecular level remains unclear. This narrative review focuses on the metabolism of tryptophan along the kynurenine pathway, since its products have critical actions in both the nervous and immune systems, placing it in a unique position to influence neuroimmune communication. In particular, since the kynurenine pathway is activated by pro-inflammatory mediators, it is proposed that physical and psychological stressors are the stimuli of an organismal protective reflex, with kynurenine metabolites as the effector arm co-ordinating protective neural and immune system responses. After a brief review of the neuroimmune interface, the general perception of tryptophan metabolism along the kynurenine pathway is expanded to emphasize this environmentally driven perspective. The initial enzymes in the kynurenine pathway include indoleamine-2,3-dioxygenase (IDO1), which is induced by tissue damage, inflammatory mediators or microbial products, and tryptophan-2,3-dioxygenase (TDO), which is induced by stress-induced glucocorticoids. In the immune system, kynurenic acid modulates leucocyte differentiation, inflammatory balance and immune tolerance by activating aryl hydrocarbon receptors and modulates pain via the GPR35 protein. In the CNS, quinolinic acid activates N-methyl-D-aspartate (NMDA)-sensitive glutamate receptors, whereas kynurenic acid is an antagonist: the balance between glutamate, quinolinic acid and kynurenic acid is a significant regulator of CNS function and plasticity. The concept of kynurenine and its metabolites as mediators of a reflex coordinated protection against stress helps to understand the variety and breadth of their activity. It should also help to understand the pathological origin of some psychiatric and neurodegenerative diseases involving the immune system and CNS, facilitating the development of new pharmacological strategies for treatment.
Collapse
Affiliation(s)
- Trevor W Stone
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
| | - Richard O Williams
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
| |
Collapse
|
34
|
Wang D, Zhao J, Zhang J, Lv C, Bao S, Gao P, He M, Li L, Zhao H, Zhang C. Targeting TNF-α: The therapeutic potential of certolizumab pegol in the early period of cerebral ischemia reperfusion injury in mice. Int Immunopharmacol 2024; 137:112498. [PMID: 38908079 DOI: 10.1016/j.intimp.2024.112498] [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: 02/20/2024] [Revised: 06/05/2024] [Accepted: 06/13/2024] [Indexed: 06/24/2024]
Abstract
The neuroinflammatory response triggered by cerebral ischemia-reperfusion injury (CIRI) is characterized by the upsurge of pro-inflammatory cytokines, including TNF-α, IL-1β, and IL-6, which promote leukocyte infiltration and subsequent accumulation in the ischemic zone. This accumulation further intensifies inflammation and aggravates ischemic damage. Certolizumab pegol (CZP), a monoclonal antibody targeting TNF-α, is widely used in treating various inflammatory diseases. This study explored the therapeutic potential of CZP in a mouse model of CIRI, induced by middle cerebral artery occlusion (MCAO), focusing on its influence on the microglial inflammatory response. In vitro analyses revealed that CZP markedly inhibits TNF-α-stimulated inflammation in primary microglia with an EC50 of 1.743 ng/mL. In vivo, MCAO mice treated with CZP (10 μg/mouse, i.p.) for 3 days showed reduced infarct volume, partially improved neurological function, and diminished blood-brain barrierdisruption. Additionally, CZP treatment curtailed microglial activation and the release of pro-inflammatory mediators in the early stages of stroke. It also favorably modulated microglial M1/M2 polarization, rebalanced Th17/Treg cells dynamics, and inhibited Caspase-8-mediated GSDMD cleavage, preventing microglial pyroptosis. Collectively, this study described that the treatment with CZP reversed damaging process caused by CIRI, offering a promising therapeutic strategy for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Dexiao Wang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, PR China; National Local Joint Engineering Research Center of Entomoceutics, Dali, PR China
| | - Jie Zhao
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, PR China; National Local Joint Engineering Research Center of Entomoceutics, Dali, PR China
| | - Jingyu Zhang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, PR China; National Local Joint Engineering Research Center of Entomoceutics, Dali, PR China
| | - Changling Lv
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, PR China
| | - Shuangyan Bao
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, PR China; National Local Joint Engineering Research Center of Entomoceutics, Dali, PR China
| | - Pengfei Gao
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, PR China; National Local Joint Engineering Research Center of Entomoceutics, Dali, PR China
| | - Miao He
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, PR China
| | - Lijuan Li
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, PR China; School of Public Health, Dali University, Dali, PR China.
| | - Hairong Zhao
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, PR China; National Local Joint Engineering Research Center of Entomoceutics, Dali, PR China.
| | - Chenggui Zhang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, PR China; National Local Joint Engineering Research Center of Entomoceutics, Dali, PR China.
| |
Collapse
|
35
|
Zhang Y, Yan Y, Smagghe G, Yang H, Dai RH, Yang WJ. Identification and immune analysis of antimicrobial peptides from the cigarette beetle (Lasioderma serricorne). INSECT SCIENCE 2024; 31:1121-1134. [PMID: 37984503 DOI: 10.1111/1744-7917.13298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 11/22/2023]
Abstract
Antimicrobial peptides (AMPs) in insects are endogenous peptides that are effector components of the innate defense system of the insect. AMPs may serve as antimicrobial agents because of their small molecular weight and broad-spectrum antimicrobial activity. In this study, we performed transcriptome analysis of cigarette beetle (Lasioderma serricorne) larvae, parasitized by the ectoparasitic wasp, Anisopteromalus calandrae. Several AMP genes were significantly upregulated following A. calandrae parasitism, postulating the hypothesis that the parasitization enhanced the host's resistance against pathogenic microorganisms through the regulation of host AMP genes. Specifically, 3 AMP genes (LsDef1, LsDef2, and LsCole) were significantly upregulated and we studied their immune function in L. serricorne. Immune challenge and functional analysis showed that LsCole was responsible for the immune response against Gram-negative and Gram-positive bacteria, while LsDef1 and LsDef2 were involved in insect defense against Gram-positive bacteria. Purified recombinant LsCole exhibited antimicrobial activities against the Gram-negative bacterium Escherichia coli and the Gram-positive bacterium Staphylococcus aureus. LsDef2 showed an antibacterial effect against S. aureus. LsCole and LsDef2 exhibited antibiofilm activity against S. aureus. The 2 AMPs disrupted cell membranes and caused leakage of S. aureus cell contents. The results indicated that the 3 AMPs in L. serricorne are involved in the innate immunity of this pest insect. These AMPs may have potential as antimicrobial agents for bacterial infection chemotherapy. Hence, data are discussed in relation to new control strategies with greater biosafety against pest insects with use of microbial biocontrol agents in combination with RNA interference against the insect's defensive AMP genes.
Collapse
Affiliation(s)
- Yue Zhang
- Guizhou Provincial Key Laboratory for Agricultural Pest Management of the Mountainous Region, Institute of Entomology, Guizhou University, Guiyang, China
| | - Yi Yan
- Guizhou Provincial Key Laboratory for Agricultural Pest Management of the Mountainous Region, Institute of Entomology, Guizhou University, Guiyang, China
- Guizhou Provincial Key Laboratory for Rare Animal and Economic Insect of the Mountainous Region, College of Biology and Environmental Engineering, Guiyang University, Guiyang, China
| | - Guy Smagghe
- Guizhou Provincial Key Laboratory for Agricultural Pest Management of the Mountainous Region, Institute of Entomology, Guizhou University, Guiyang, China
| | - Hong Yang
- Guizhou Provincial Key Laboratory for Agricultural Pest Management of the Mountainous Region, Institute of Entomology, Guizhou University, Guiyang, China
| | - Ren-Huai Dai
- Guizhou Provincial Key Laboratory for Agricultural Pest Management of the Mountainous Region, Institute of Entomology, Guizhou University, Guiyang, China
| | - Wen-Jia Yang
- Guizhou Provincial Key Laboratory for Rare Animal and Economic Insect of the Mountainous Region, College of Biology and Environmental Engineering, Guiyang University, Guiyang, China
| |
Collapse
|
36
|
Bu LK, Jia PP, Huo WB, Pei DS. Assessment of Probiotics' Impact on Neurodevelopmental and Behavioral Responses in Zebrafish Models: Implications for Autism Spectrum Disorder Therapy. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10335-y. [PMID: 39090455 DOI: 10.1007/s12602-024-10335-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2024] [Indexed: 08/04/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder; the prevalence of which has been on the rise with unknown causes. Alterations in the gut-brain axis have been widely recognized in ASD patients, and probiotics are considered to potentially benefit the rescuing of autism-like behaviors. However, the effectiveness and mechanisms of multiple probiotics on zebrafish models are still not clearly revealed. This study aims to use the germ-free (GF) and conventionally raised (CR) AB wild-type zebrafish and the mutant Tbr1b-/- and Katnal2-/- lines as human-linked ASD animal models to evaluate the effects of multiple probiotics on mitigating developmental and behavioral defects. Results showed that the addition of probiotics increased the basic important developmental indexes, such as body length, weight, and survival rate of treated zebrafish. Moreover, the Lactobacillus plantarum and Lactobacillus rhamnosus affected the behavior of CR zebrafish by increasing their mobility, lowering the GF zebrafish manic, and mitigating transgenic zebrafish abnormal behavior. Moreover, the expression levels of key genes related to gamma-aminobutyric acid (GABA), dopamine (DA), and serotonin (5-HT) as important neuropathways to influence the appearance and development of autism-related disorders, including gad1b, tph1a, htr3a, th, and slc6a3, were significantly activated by some of the probiotics' treatment at some extent. Taken together, this study indicates the beneficial effects of different probiotics, which may provide a novel understanding of probiotic function in related diseases' therapy.
Collapse
Affiliation(s)
- Ling-Kang Bu
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China
| | - Pan-Pan Jia
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China
| | - Wen-Bo Huo
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China
| | - De-Sheng Pei
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China.
- Chongqing Miankai Biotechnology Research Institute Co., Ltd., Chongqing, 400025, China.
| |
Collapse
|
37
|
Bu Y, Burks J, Yang K, Prince J, Borna A, Coe CL, Simmons A, Tu XM, Baker D, Kimball D, Rao R, Shah V, Huang M, Schwindt P, Coleman TP, Lerman I. Non-invasive ventral cervical magnetoneurography as a proxy of in vivo lipopolysaccharide-induced inflammation. Commun Biol 2024; 7:893. [PMID: 39075164 PMCID: PMC11286963 DOI: 10.1038/s42003-024-06435-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 06/10/2024] [Indexed: 07/31/2024] Open
Abstract
Maintenance of autonomic homeostasis is continuously calibrated by sensory fibers of the vagus nerve and sympathetic chain that convey compound action potentials (CAPs) to the central nervous system. Lipopolysaccharide (LPS) intravenous challenge reliably elicits a robust inflammatory response that can resemble systemic inflammation and acute endotoxemia. Here, we administered LPS intravenously in nine healthy subjects while recording ventral cervical magnetoneurography (vcMNG)-derived CAPs at the rostral Right Nodose Ganglion (RNG) and the caudal Right Carotid Artery (RCA) with optically pumped magnetometers (OPM). We observed vcMNG RNG and RCA neural firing rates that tracked changes in TNF-α levels in the systemic circulation. Further, endotype subgroups based on high and low IL-6 responders segregate RNG CAP frequency (at 30-120 min) and based on high and low IL-10 response discriminate RCA CAP frequency (at 0-30 min). These vcMNG tools may enhance understanding and management of the neuroimmune axis that can guide personalized treatment based on an individual's distinct endophenotype.
Collapse
Affiliation(s)
- Yifeng Bu
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Jamison Burks
- Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Kun Yang
- Division of Biostatistics and Bioinformatics, University of California San Diego, La Jolla, CA, 92093, USA
| | - Jacob Prince
- Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Amir Borna
- Quantum Information Sciences, Sandia National Laboratories, Albuquerque, NM, 87123, USA
| | - Christopher L Coe
- Department of Psychology, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Alan Simmons
- Center for Stress and Mental Health (CESAMH) VA San Diego, La Jolla, CA, 92093, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Xin M Tu
- Division of Biostatistics and Bioinformatics, University of California San Diego, La Jolla, CA, 92093, USA
| | - Dewleen Baker
- Center for Stress and Mental Health (CESAMH) VA San Diego, La Jolla, CA, 92093, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Donald Kimball
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ramesh Rao
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Vishal Shah
- Quspin Laboratory Head Quarters, Boulder, CO, 80305, USA
| | - Mingxiong Huang
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, CA, 92093, USA
- Department of Radiology, University of California San Diego, La Jolla, CA, 92093, USA
| | - Peter Schwindt
- Quantum Information Sciences, Sandia National Laboratories, Albuquerque, NM, 87123, USA
| | - Todd P Coleman
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, CA, 92093, USA
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Imanuel Lerman
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, CA, 92093, USA.
- Center for Stress and Mental Health (CESAMH) VA San Diego, La Jolla, CA, 92093, USA.
- InflammaSense Incorporated Head Quarters, La Jolla, CA, 92093, USA.
- Department of Anesthesiology, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
38
|
Abdel-Aziz N, Saif-Elnasr M. Citicoline modulates inflammatory signaling pathways in the spleen of rats exposed to gamma-radiation. Immunopharmacol Immunotoxicol 2024:1-8. [PMID: 39049671 DOI: 10.1080/08923973.2024.2381759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 07/14/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND AND AIM The spleen has an essential role in immune responses regulation and is considered the biggest peripheral immune organ. Citicoline is used for various brain disorders management. This study aimed to examine the using possibility of citicoline to treat γ-radiation-induced splenic inflammation in rats. MATERIALS AND METHODS Eighteen male albino rats were classified into: Group 1 (control) animals were kept as control. Group 2 (γ-radiation) animals were total-body γ-irradiated with 6 Gy. Group 3 (γ-radiation + citicoline) rats were γ-irradiated with 6 Gy, then injected intraperitoneally with citicoline (300 mg/kg/d) 5 min after irradiation for one week. Levels of TNF-α, IL-1β, iNOS, NF-κB, JAK2, and STAT3 were determined in spleen tissue, along with histopathological examination. RESULTS Rats exposure to gamma-radiation led to elevation in splenic TNF-α, IL-1β, NF-κB, iNOS, JAK2, and STAT3 levels significantly. Treatment with citicoline after gamma-radiation exposure improved this elevation, and modulated gamma-radiation-induced histopathological alterations. CONCLUSIONS This data showed that citicoline inhibited γ-radiation-induced splenic inflammation via suppressing NF-κB and JAK2/STAT3 signaling pathways in spleen tissue.
Collapse
Affiliation(s)
- Nahed Abdel-Aziz
- Radiation Biology Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Mostafa Saif-Elnasr
- Health Radiation Research Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
39
|
Beurel E, Nemeroff CB. Early Life Adversity, Microbiome, and Inflammatory Responses. Biomolecules 2024; 14:802. [PMID: 39062516 PMCID: PMC11275239 DOI: 10.3390/biom14070802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
Early life adversity has a profound impact on physical and mental health. Because the central nervous and immune systems are not fully mature at birth and continue to mature during the postnatal period, a bidirectional interaction between the central nervous system and the immune system has been hypothesized, with traumatic stressors during childhood being pivotal in priming individuals for later adult psychopathology. Similarly, the microbiome, which regulates both neurodevelopment and immune function, also matures during childhood, rendering this interaction between the brain and the immune system even more complex. In this review, we provide evidence for the role of the immune response and the microbiome in the deleterious effects of early life adversity, both in humans and rodent models.
Collapse
Affiliation(s)
- Eléonore Beurel
- Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Charles B. Nemeroff
- Department of Psychiatry and Behavioral Sciences, Mulva Clinic for Neurosciences, University of Texas (UT) Dell Medical School, Austin, TX 78712, USA
- Mulva Clinic for Neurosciences, UT Austin Dell Medical School, Austin, TX 78712, USA
| |
Collapse
|
40
|
Haykin H, Avishai E, Krot M, Ghiringhelli M, Reshef M, Abboud Y, Melamed S, Merom S, Boshnak N, Azulay-Debby H, Ziv T, Gepstein L, Rolls A. Reward system activation improves recovery from acute myocardial infarction. NATURE CARDIOVASCULAR RESEARCH 2024; 3:841-856. [PMID: 39196183 DOI: 10.1038/s44161-024-00491-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 05/16/2024] [Indexed: 08/29/2024]
Abstract
Psychological processes have a crucial role in the recovery from acute myocardial infarction (AMI), yet the underlying mechanisms of these effects remain elusive. Here we demonstrate the impact of the reward system, a brain network associated with motivation and positive expectations, on the clinical outcomes of AMI in mice. Chemogenetic activation of dopaminergic neurons in the reward system improved the remodeling processes and vascularization after AMI, leading to enhanced cardiac performance compared to controls. These effects were mediated through several physiological mechanisms, including alterations in immune activity and reduced adrenergic input to the liver. We further demonstrate an anatomical connection between the reward system and the liver, functionally manifested by altered transcription of complement component 3, which in turn affects vascularization and recovery from AMI. These findings establish a causal connection between a motivational brain network and recovery from AMI, introducing potential therapeutic avenues for intervention.
Collapse
Affiliation(s)
- H Haykin
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Department of Neuroscience, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Department of Physiology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - E Avishai
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Department of Neuroscience, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - M Krot
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Department of Neuroscience, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - M Ghiringhelli
- Department of Physiology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - M Reshef
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Department of Neuroscience, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Y Abboud
- Department of Physiology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - S Melamed
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - S Merom
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Department of Neuroscience, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - N Boshnak
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Department of Neuroscience, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - H Azulay-Debby
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Department of Neuroscience, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - T Ziv
- Smoler Proteomics Center, Lorry I. Lokey Interdisciplinary Center for Life Sciences and Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - L Gepstein
- Department of Physiology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.
- Cardiology Department, Rambam Health Care Campus, Haifa, Israel.
| | - A Rolls
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.
- Department of Neuroscience, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
41
|
Cardoso Santos G, Alves de Jesus A, Passaglia P, Novaes Morgan HJ, Carvalho Navegantes LC, Leico Kagohara Elias L, Cárnio EC. Central angiotensin-(1-7) attenuates hypoglycemia in sepsis-like conditions via reducing systemic and hepatic inflammation. Cytokine 2024; 179:156637. [PMID: 38723454 DOI: 10.1016/j.cyto.2024.156637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/08/2024] [Accepted: 05/02/2024] [Indexed: 05/21/2024]
Abstract
Sepsis is understood as the result of initiating systemic inflammation derived from an inadequate host response against pathogens. In its acute phase, sepsis is marked by an exacerbated reaction to infection, tissue damage, organ failure, and metabolic dysfunction. Among these, hypoglycemia, characterized by disorders of the gluconeogenesis pathway, is related to one of the leading causes of mortality in septic patients. Recent research has investigated the involvement of sympathetic efferent neuroimmune pathways during systemic inflammation. These pathways can be stimulated by several centrally administered drugs, including Angiotensin-(1-7) (Ang-(1-7)). Therefore, the present study aims to evaluate the effects of central treatment with Ang-(1-7) on hypoglycemia during endotoxemia. For this, male Wistar Hannover rats underwent stereotaxic surgery for intracerebroventricular (i.c.v.) administration of Ang-(1-7) and cannulation of the jugular vein for lipopolysaccharide (LPS) injection. Our results demonstrate that LPS was capable of inducing hypoglycemia and that prior central treatment with Ang-(1-7) attenuated this effect. Our data also show that Ang-(1-7) reduced plasma concentrations of TNF-α, IL-1β, IL-6, and nitric oxide, in addition to the decrease and increase of hepatic IL-6 and IL-10 respectively, in animals subjected to systemic inflammation by LPS, resulting in the reduction of systemic and hepatic inflammation, thus attenuating the deleterious effects of LPS on phosphoenolpyruvate carboxykinase protein content. In summary, the data suggest that central treatment with Ang-(1-7) attenuates hypoglycemia induced by endotoxemia, probably through anti-inflammatory action, leading to reestablishing hepatic gluconeogenesis.
Collapse
Affiliation(s)
- Gabriel Cardoso Santos
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil.
| | - Aline Alves de Jesus
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Patrícia Passaglia
- Department of Oral and Basic Biology, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Henrique J Novaes Morgan
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | | | - Lucila Leico Kagohara Elias
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Evelin Capellari Cárnio
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil; Department of General and Specialized Nursing, Ribeirão Preto, College of Nursing,University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
42
|
Fang S, Wong CK. Anti-Inflammatory Effects of Glucagon-Like Peptide-1 Receptor Agonists via the Neuroimmune Axis. DNA Cell Biol 2024; 43:267-270. [PMID: 38579130 DOI: 10.1089/dna.2024.0057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2024] Open
Abstract
Glucagon-like peptide 1 receptor agonists (GLP-1RAs) have shown efficacy in the treatment of metabolic disease-related complications, partially attributable to their anti-inflammatory properties. However, the specific cell types and pathways involved in these effects were not fully understood. A recent study by Wong et al. demonstrated the importance of the brain GLP-1R in mediating the anti-inflammatory effects of GLP-1RAs in Toll-like receptor and sepsis-mediated inflammation. In this discussion, we review the existing literature on the action of GLP-1RAs in inflammation and explore the implications of these recent findings.
Collapse
Affiliation(s)
- Susanna Fang
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Chi Kin Wong
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| |
Collapse
|
43
|
Galin S, Keren H. The Predictive Potential of Heart Rate Variability for Depression. Neuroscience 2024; 546:88-103. [PMID: 38513761 DOI: 10.1016/j.neuroscience.2024.03.013] [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: 08/17/2023] [Revised: 02/29/2024] [Accepted: 03/16/2024] [Indexed: 03/23/2024]
Abstract
Heart rate variability (HRV),a measure of the fluctuations in the intervals between consecutive heartbeats, is an indicator of changes in the autonomic nervous system. A chronic reduction in HRV has been repeatedly linked to clinical depression. However, the chronological and mechanistic aspects of this relationship, between the neural, physiological, and psychopathological levels, remain unclear. In this review we present evidence by which changes in HRV might precede the onset of depression. We describe several pathways that can facilitate this relationship. First, we examine a theoretical model of the impact of autonomic imbalance on HRV and its role in contributing to mood dysregulation and depression. We then highlight brain regions that are regulating both HRV and emotion, suggesting these neural regions, and the Insula in particular, as potential mediators of this relationship. We also present additional possible mediating mechanisms involving the immune system and inflammation processes. Lastly, we support this model by showing evidence that modification of HRV with biofeedback leads to an improvement in some symptoms of depression. The possibility that changes in HRV precede the onset of depression is critical to put to the test, not only because it could provide insights into the mechanisms of the illness but also because it may offer a predictive anddiagnosticphysiological marker for depression. Importantly, it could also help to develop new effective clinical interventions for treating depression.
Collapse
Affiliation(s)
- Shir Galin
- Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel; Gonda Interdisciplinary Brain Research Center, Bar Ilan University, Ramat Gan, Israel
| | - Hanna Keren
- Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel; Gonda Interdisciplinary Brain Research Center, Bar Ilan University, Ramat Gan, Israel.
| |
Collapse
|
44
|
Wang Q, Liu C, Chen M, Zhao J, Wang D, Gao P, Zhang C, Zhao H. Mastoparan M promotes functional recovery in stroke mice by activating autophagy and inhibiting ferroptosis. Biomed Pharmacother 2024; 174:116560. [PMID: 38583338 DOI: 10.1016/j.biopha.2024.116560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/20/2024] [Accepted: 04/04/2024] [Indexed: 04/09/2024] Open
Abstract
Neuronal ferroptosis and autophagy are crucial in the pathogenesis of cerebral ischemia-reperfusion injury (CIRI). Mastoparan M (Mast-M), extracted from the crude venom of Vespa magnifica (Smith), comprises 14 amino acid residues. Previous studies suggested that Mast-M reduces neuronal damage following global CIRI, but its protective mechanisms remain unclear. The present study examined the effect of Mast-M on middle cerebral artery occlusion/reperfusion (MCAO/R) induced neurological deficits using Grip, Rotarod, Longa test, and TTC staining, followed by treating the mice for three days with Mast-M (20, 40, and 80 μg/kg, subcutaneously). The results demonstrate that Mast-M promotes functional recovery in mice post-ischemic stroke, evidenced by improved neurological impairment, reduced infarct volume and neuronal damage. Meanwhile, the level of iron (Fe2+) and malonyldialdehyde was decreased in the ischemic hemisphere of MCAO/R mice at 24 hours or 48 hours by Mast-M (80 μg/kg) treatment, while the expression of NRF2, x-CT, GPX4, and LC3B protein was increased. Furthermore, these findings were validated in three models-oxygen-glucose deprivation/ reoxygenation, H2O2-induced peroxidation, and erastin-induced ferroptosis-in hippocampal neuron HT22 cells or primary neurons. These data suggested that Mast-M activates autophagy as well as inhibits ferroptosis. Finally, autophagy inhibitors were introduced to determine the relationship between the autophagy and ferroptosis, indicating that Mast-M alleviates ferroptosis by activating autophagy. Taken together, this study described that Mast-M alleviates cerebral infarction, neurologic impairment, and neuronal damage by activating autophagy and inhibiting ferroptosis, presenting a potential therapeutic approach for CIRI.
Collapse
Affiliation(s)
- Qian Wang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, PR China
| | - Chaojie Liu
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, PR China
| | - Mingran Chen
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, PR China
| | - Jie Zhao
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, PR China
| | - Dexiao Wang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, PR China
| | - Pengfei Gao
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, PR China; National-Local Joint Engineering Research Center of Entomoceutics, Dali, PR China
| | - Chenggui Zhang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, PR China; National-Local Joint Engineering Research Center of Entomoceutics, Dali, PR China.
| | - Hairong Zhao
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, PR China; National-Local Joint Engineering Research Center of Entomoceutics, Dali, PR China.
| |
Collapse
|
45
|
Nusslock R, Alloy LB, Brody GH, Miller GE. Annual Research Review: Neuroimmune network model of depression: a developmental perspective. J Child Psychol Psychiatry 2024; 65:538-567. [PMID: 38426610 PMCID: PMC11090270 DOI: 10.1111/jcpp.13961] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/18/2024] [Indexed: 03/02/2024]
Abstract
Depression is a serious public health problem, and adolescence is an 'age of risk' for the onset of Major Depressive Disorder. Recently, we and others have proposed neuroimmune network models that highlight bidirectional communication between the brain and the immune system in both mental and physical health, including depression. These models draw on research indicating that the cellular actors (particularly monocytes) and signaling molecules (particularly cytokines) that orchestrate inflammation in the periphery can directly modulate the structure and function of the brain. In the brain, inflammatory activity heightens sensitivity to threats in the cortico-amygdala circuit, lowers sensitivity to rewards in the cortico-striatal circuit, and alters executive control and emotion regulation in the prefrontal cortex. When dysregulated, and particularly under conditions of chronic stress, inflammation can generate feelings of dysphoria, distress, and anhedonia. This is proposed to initiate unhealthy, self-medicating behaviors (e.g. substance use, poor diet) to manage the dysphoria, which further heighten inflammation. Over time, dysregulation in these brain circuits and the inflammatory response may compound each other to form a positive feedback loop, whereby dysregulation in one organ system exacerbates the other. We and others suggest that this neuroimmune dysregulation is a dynamic joint vulnerability for depression, particularly during adolescence. We have three goals for the present paper. First, we extend neuroimmune network models of mental and physical health to generate a developmental framework of risk for the onset of depression during adolescence. Second, we examine how a neuroimmune network perspective can help explain the high rates of comorbidity between depression and other psychiatric disorders across development, and multimorbidity between depression and stress-related medical illnesses. Finally, we consider how identifying neuroimmune pathways to depression can facilitate a 'next generation' of behavioral and biological interventions that target neuroimmune signaling to treat, and ideally prevent, depression in youth and adolescents.
Collapse
Affiliation(s)
- Robin Nusslock
- Department of Psychology, Northwestern University, Evanston IL, USA
- Institute for Policy Research, Northwestern University, Evanston IL, USA
| | - Lauren B. Alloy
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA. USA
| | - Gene H. Brody
- Center for Family Research, University of Georgia, Athens GA, USA
| | - Gregory E. Miller
- Department of Psychology, Northwestern University, Evanston IL, USA
- Institute for Policy Research, Northwestern University, Evanston IL, USA
| |
Collapse
|
46
|
Ziaka M, Exadaktylos A. Pathophysiology of acute lung injury in patients with acute brain injury: the triple-hit hypothesis. Crit Care 2024; 28:71. [PMID: 38454447 PMCID: PMC10918982 DOI: 10.1186/s13054-024-04855-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 03/01/2024] [Indexed: 03/09/2024] Open
Abstract
It has been convincingly demonstrated in recent years that isolated acute brain injury (ABI) may cause severe dysfunction of peripheral extracranial organs and systems. Of all potential target organs and systems, the lung appears to be the most vulnerable to damage after ABI. The pathophysiology of the bidirectional brain-lung interactions is multifactorial and involves inflammatory cascades, immune suppression, and dysfunction of the autonomic system. Indeed, the systemic effects of inflammatory mediators in patients with ABI create a systemic inflammatory environment ("first hit") that makes extracranial organs vulnerable to secondary procedures that enhance inflammation, such as mechanical ventilation (MV), surgery, and infections ("second hit"). Moreover, accumulating evidence supports the knowledge that gut microbiota constitutes a critical superorganism and an organ on its own, potentially modifying various physiological functions of the host. Furthermore, experimental and clinical data suggest the existence of a communication network among the brain, gastrointestinal tract, and its microbiome, which appears to regulate immune responses, gastrointestinal function, brain function, behavior, and stress responses, also named the "gut-microbiome-brain axis." Additionally, recent research evidence has highlighted a crucial interplay between the intestinal microbiota and the lungs, referred to as the "gut-lung axis," in which alterations during critical illness could result in bacterial translocation, sustained inflammation, lung injury, and pulmonary fibrosis. In the present work, we aimed to further elucidate the pathophysiology of acute lung injury (ALI) in patients with ABI by attempting to develop the "double-hit" theory, proposing the "triple-hit" hypothesis, focused on the influence of the gut-lung axis on the lung. Particularly, we propose, in addition to sympathetic hyperactivity, blast theory, and double-hit theory, that dysbiosis and intestinal dysfunction in the context of ABI alter the gut-lung axis, resulting in the development or further aggravation of existing ALI, which constitutes the "third hit."
Collapse
Affiliation(s)
- Mairi Ziaka
- Clinic for Geriatric Medicine, Center for Geriatric Medicine and Rehabilitation, Kantonsspital Baselland, Bruderholz, Switzerland.
- Department of Emergency Medicine, Inselspital, University Hospital, University of Bern, Bern, Switzerland.
| | - Aristomenis Exadaktylos
- Department of Emergency Medicine, Inselspital, University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
47
|
Nonaka F, Fukui S, Michitsuji T, Endo Y, Nishino A, Shimizu T, Umeda M, Sumiyoshi R, Koga T, Iwamoto N, Origuchi T, Ueki Y, Eiraku N, Suzuki T, Okada A, Matsuoka N, Takaoka H, Hamada H, Tsuru T, Arinobu Y, Hidaka T, Fujikawa K, Yoshitama T, Tada Y, Ohtsubo H, Ishizaki J, Asano T, Maeda T, Kawakami A, Kawashiri SY. The impact of glucocorticoid use on the outcomes of rheumatoid arthritis in a multicenter ultrasound cohort study. Int J Rheum Dis 2024; 27:e15118. [PMID: 38487995 DOI: 10.1111/1756-185x.15118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/06/2024] [Accepted: 02/28/2024] [Indexed: 03/19/2024]
Abstract
OBJECTIVE Glucocorticoids are effective in treating rheumatoid arthritis (RA) when used appropriately considering the balance of the risks and benefits, especially at low doses. We aimed to evaluate the response of biologic and targeted synthetic disease-modifying antirheumatic drugs (b/tsDMARDs) in patients having already been treated with glucocorticoids. METHODS We reviewed RA patients treated with b/tsDMARDs in a prospective multicenter ultrasound cohort study. We compared the differences in the clinical characteristics at baseline and outcomes at 12 months between the two groups having been treated with and without glucocorticoids at baseline. The differences in the clinical characteristics and the treatments were balanced by the inverse probability weighting (IPW) with the propensity score. RESULTS Of 307 patients with RA, 160 patients were treated with glucocorticoids at baseline. The median dose of glucocorticoids was equivalent to 5.0 mg/day of prednisolone. Significant differences were in age and concomitant methotrexate use, composite measures for the disease activity, and the ultrasound grayscale score at baseline. Patients treated with glucocorticoids had less frequent remissions defined by composite measures and ultrasound findings than those treated without glucocorticoids. These significant differences in the achievement of remissions remained robust even after adjusting differences in the clinical characteristics and the treatments between the two groups by IPW. CONCLUSION RA patients treated with glucocorticoids had a higher disease activity at baseline and a poorer response to treatments with b/tsDMARDs than those without glucocorticoids. The states of patients requiring glucocorticoids might be associated with the poor response to the b/tsDMARDs.
Collapse
Affiliation(s)
- Fumiaki Nonaka
- Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Island and Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Shoichi Fukui
- Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Tohru Michitsuji
- Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yushiro Endo
- Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Ayako Nishino
- Kyushu Multicenter Rheumatoid Arthritis Ultrasound Prospective Observational Cohort Study Group, Nagasaki, Japan
| | - Toshimasa Shimizu
- Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Masataka Umeda
- Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Remi Sumiyoshi
- Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Tomohiro Koga
- Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Naoki Iwamoto
- Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Tomoki Origuchi
- Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yukitaka Ueki
- Kyushu Multicenter Rheumatoid Arthritis Ultrasound Prospective Observational Cohort Study Group, Nagasaki, Japan
| | - Nobutaka Eiraku
- Kyushu Multicenter Rheumatoid Arthritis Ultrasound Prospective Observational Cohort Study Group, Nagasaki, Japan
| | - Takahisa Suzuki
- Kyushu Multicenter Rheumatoid Arthritis Ultrasound Prospective Observational Cohort Study Group, Nagasaki, Japan
| | - Akitomo Okada
- Department of Island and Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Naoki Matsuoka
- Kyushu Multicenter Rheumatoid Arthritis Ultrasound Prospective Observational Cohort Study Group, Nagasaki, Japan
| | - Hirokazu Takaoka
- Department of Island and Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hiroaki Hamada
- Kyushu Multicenter Rheumatoid Arthritis Ultrasound Prospective Observational Cohort Study Group, Nagasaki, Japan
| | - Tomomi Tsuru
- Kyushu Multicenter Rheumatoid Arthritis Ultrasound Prospective Observational Cohort Study Group, Nagasaki, Japan
| | - Yojiro Arinobu
- Kyushu Multicenter Rheumatoid Arthritis Ultrasound Prospective Observational Cohort Study Group, Nagasaki, Japan
| | - Toshihiko Hidaka
- Kyushu Multicenter Rheumatoid Arthritis Ultrasound Prospective Observational Cohort Study Group, Nagasaki, Japan
| | - Keita Fujikawa
- Kyushu Multicenter Rheumatoid Arthritis Ultrasound Prospective Observational Cohort Study Group, Nagasaki, Japan
| | - Tamami Yoshitama
- Kyushu Multicenter Rheumatoid Arthritis Ultrasound Prospective Observational Cohort Study Group, Nagasaki, Japan
| | - Yoshifumi Tada
- Kyushu Multicenter Rheumatoid Arthritis Ultrasound Prospective Observational Cohort Study Group, Nagasaki, Japan
| | - Hideo Ohtsubo
- Kyushu Multicenter Rheumatoid Arthritis Ultrasound Prospective Observational Cohort Study Group, Nagasaki, Japan
| | - Jun Ishizaki
- Department of Island and Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Tomoyuki Asano
- Department of Island and Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Takahiro Maeda
- Department of Island and Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of General Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Atsushi Kawakami
- Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Shin-Ya Kawashiri
- Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Kyushu Multicenter Rheumatoid Arthritis Ultrasound Prospective Observational Cohort Study Group, Nagasaki, Japan
- Center for Collaborative Medical Education and Development, Nagasaki University Institute of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
48
|
Wang L, Cheng M, Wang Y, Chen J, Xie F, Huang LH, Zhan C. Fasting-activated ventrolateral medulla neurons regulate T cell homing and suppress autoimmune disease in mice. Nat Neurosci 2024; 27:462-470. [PMID: 38182836 DOI: 10.1038/s41593-023-01543-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 12/01/2023] [Indexed: 01/07/2024]
Abstract
Dietary fasting markedly influences the distribution and function of immune cells and exerts potent immunosuppressive effects. However, the mechanisms through which fasting regulates immunity remain obscure. Here we report that catecholaminergic (CA) neurons in the ventrolateral medulla (VLM) are activated during fasting in mice, and we demonstrate that the activity of these CA neurons impacts the distribution of T cells and the development of autoimmune disease in an experimental autoimmune encephalomyelitis (EAE) model. Ablation of VLM CA neurons largely reversed fasting-mediated T cell redistribution. Activation of these neurons drove T cell homing to bone marrow in a CXCR4/CXCL12 axis-dependent manner, which may be mediated by a neural circuit that stimulates corticosterone secretion. Similar to fasting, the continuous activation of VLM CA neurons suppressed T cell activation, proliferation, differentiation and cytokine production in autoimmune mouse models and substantially alleviated disease symptoms. Collectively, our study demonstrates neuronal control of inflammation and T cell distribution, suggesting a neural mechanism underlying fasting-mediated immune regulation.
Collapse
Affiliation(s)
- Liang Wang
- Department of Hematology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Research Center for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Mingxiu Cheng
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
- National Institute of Biological Sciences, Beijing, China
| | - Yuchen Wang
- Department of Hematology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Research Center for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jing Chen
- School of Sport Science, Beijing Sport University, Beijing, China
| | - Famin Xie
- School of Life Sciences, Fudan University, Shanghai, China
| | - Li-Hao Huang
- Institute of Metabolism & Integrative Biology, Fudan University, Shanghai, China
| | - Cheng Zhan
- Department of Hematology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Research Center for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China.
- National Institute of Biological Sciences, Beijing, China.
| |
Collapse
|
49
|
Kodi T, Sankhe R, Gopinathan A, Nandakumar K, Kishore A. New Insights on NLRP3 Inflammasome: Mechanisms of Activation, Inhibition, and Epigenetic Regulation. J Neuroimmune Pharmacol 2024; 19:7. [PMID: 38421496 PMCID: PMC10904444 DOI: 10.1007/s11481-024-10101-5] [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: 03/07/2023] [Accepted: 11/06/2023] [Indexed: 03/02/2024]
Abstract
Inflammasomes are important modulators of inflammation. Dysregulation of inflammasomes can enhance vulnerability to conditions such as neurodegenerative diseases, autoinflammatory diseases, and metabolic disorders. Among various inflammasomes, Nucleotide-binding oligomerization domain leucine-rich repeat and pyrin domain-containing protein 3 (NLRP3) is the best-characterized inflammasome related to inflammatory and neurodegenerative diseases. NLRP3 is an intracellular sensor that recognizes pathogen-associated molecular patterns and damage-associated patterns resulting in the assembly and activation of NLRP3 inflammasome. The NLRP3 inflammasome includes sensor NLRP3, adaptor apoptosis-associated speck-like protein (ASC), and effector cysteine protease procaspase-1 that plays an imperative role in caspase-1 stimulation which further initiates a secondary inflammatory response. Regulation of NLRP3 inflammasome ameliorates NLRP3-mediated diseases. Much effort has been invested in studying the activation, and exploration of specific inhibitors and epigenetic mechanisms controlling NLRP3 inflammasome. This review gives an overview of the established NLRP3 inflammasome assembly, its brief molecular mechanistic activations as well as a current update on specific and non-specific NLRP3 inhibitors that could be used in NLRP3-mediated diseases. We also focused on the recently discovered epigenetic mechanisms mediated by DNA methylation, histone alterations, and microRNAs in regulating the activation and expression of NLRP3 inflammasome, which has resulted in a novel method of gaining insight into the mechanisms that modulate NLRP3 inflammasome activity and introducing potential therapeutic strategies for CNS disorders.
Collapse
Affiliation(s)
- Triveni Kodi
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Runali Sankhe
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Adarsh Gopinathan
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Krishnadas Nandakumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Anoop Kishore
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
50
|
Cheng J, Hu H, Ju Y, Liu J, Wang M, Liu B, Zhang Y. Gut microbiota-derived short-chain fatty acids and depression: deep insight into biological mechanisms and potential applications. Gen Psychiatr 2024; 37:e101374. [PMID: 38390241 PMCID: PMC10882305 DOI: 10.1136/gpsych-2023-101374] [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: 10/04/2023] [Accepted: 12/25/2023] [Indexed: 02/24/2024] Open
Abstract
The gut microbiota is a complex and dynamic ecosystem known as the 'second brain'. Composing the microbiota-gut-brain axis, the gut microbiota and its metabolites regulate the central nervous system through neural, endocrine and immune pathways to ensure the normal functioning of the organism, tuning individuals' health and disease status. Short-chain fatty acids (SCFAs), the main bioactive metabolites of the gut microbiota, are involved in several neuropsychiatric disorders, including depression. SCFAs have essential effects on each component of the microbiota-gut-brain axis in depression. In the present review, the roles of major SCFAs (acetate, propionate and butyrate) in the pathophysiology of depression are summarised with respect to chronic cerebral hypoperfusion, neuroinflammation, host epigenome and neuroendocrine alterations. Concluding remarks on the biological mechanisms related to gut microbiota will hopefully address the clinical value of microbiota-related treatments for depression.
Collapse
Affiliation(s)
- Junzhe Cheng
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Medicine Eight-Year Program, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Hongkun Hu
- Clinical Medicine Eight-Year Program, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yumeng Ju
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Mental Health Institute of Central South University, China National Technology Institute on Mental Disorders, Hunan Key Laboratory of Psychiatry and Mental Health, Hunan Medical Center for Mental Health, Changsha, Hunan, China
| | - Jin Liu
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Mental Health Institute of Central South University, China National Technology Institute on Mental Disorders, Hunan Key Laboratory of Psychiatry and Mental Health, Hunan Medical Center for Mental Health, Changsha, Hunan, China
| | - Mi Wang
- Department of Mental Health Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Bangshan Liu
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Mental Health Institute of Central South University, China National Technology Institute on Mental Disorders, Hunan Key Laboratory of Psychiatry and Mental Health, Hunan Medical Center for Mental Health, Changsha, Hunan, China
| | - Yan Zhang
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Mental Health Institute of Central South University, China National Technology Institute on Mental Disorders, Hunan Key Laboratory of Psychiatry and Mental Health, Hunan Medical Center for Mental Health, Changsha, Hunan, China
| |
Collapse
|