1
|
Castranova D, Kenton MI, Kraus A, Dell CW, Park JS, Venero Galanternik M, Park G, Lumbantobing DN, Dye L, Marvel M, Iben J, Taimatsu K, Pham V, Willms RJ, Blevens L, Robertson TF, Hou Y, Huttenlocher A, Foley E, Parenti LR, Frazer JK, Narayan K, Weinstein BM. The axillary lymphoid organ is an external, experimentally accessible immune organ in the zebrafish. J Exp Med 2025; 222:e20241435. [PMID: 40167600 PMCID: PMC11960710 DOI: 10.1084/jem.20241435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 01/13/2025] [Accepted: 03/03/2025] [Indexed: 04/02/2025] Open
Abstract
Lymph nodes and other secondary lymphoid organs play critical roles in immune surveillance and immune activation in mammals, but the deep internal locations of these organs make it challenging to image and study them in living animals. Here, we describe a previously uncharacterized external immune organ in the zebrafish ideally suited for studying immune cell dynamics in vivo, the axillary lymphoid organ (ALO). This small, translucent organ has an outer cortex teeming with immune cells, an inner medulla with a mesh-like network of fibroblastic reticular cells along which immune cells migrate, and a network of lymphatic vessels draining to a large adjacent lymph sac. Noninvasive high-resolution imaging of transgenically marked immune cells can be carried out in ALOs of living animals, which are readily accessible to external treatment. This newly discovered tissue provides a superb model for dynamic live imaging of immune cells and their interaction with pathogens and surrounding tissues, including blood and lymphatic vessels.
Collapse
Affiliation(s)
- Daniel Castranova
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Madeleine I. Kenton
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Aurora Kraus
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Christopher W. Dell
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Jong S. Park
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Marina Venero Galanternik
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Gilseung Park
- Section of Pediatric Hematology-Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Daniel N. Lumbantobing
- Division of Fishes, Department of Vertebrate Zoology, National Museum of Natural History, Smithsonian Institution, Washington, DC, USA
| | - Louis Dye
- Microscopy and Imaging Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Miranda Marvel
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - James Iben
- Molecular Genomics Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Kiyohito Taimatsu
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Van Pham
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Reegan J. Willms
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Lucas Blevens
- Section of Pediatric Hematology-Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Tanner F. Robertson
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA
| | - Yiran Hou
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA
| | - Anna Huttenlocher
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA
| | - Edan Foley
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Lynne R. Parenti
- Division of Fishes, Department of Vertebrate Zoology, National Museum of Natural History, Smithsonian Institution, Washington, DC, USA
| | - J. Kimble Frazer
- Section of Pediatric Hematology-Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kedar Narayan
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Brant M. Weinstein
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
2
|
Jing Z, Yinhang W, Jian C, Zhanbo Q, Xinyue W, Shuwen H. Interaction between gut microbiota and T cell immunity in colorectal cancer. Autoimmun Rev 2025; 24:103807. [PMID: 40139455 DOI: 10.1016/j.autrev.2025.103807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 02/26/2025] [Accepted: 03/22/2025] [Indexed: 03/29/2025]
Abstract
This review delves into the complex and multi-layered mechanisms that govern the interaction between gut microbiota and T cells in the context of colorectal cancer (CRC), revealing a novel "microbiota-immune regulatory landscape" within the tumor microenvironment. As CRC progresses, the gut microbiota experiences a significant transformation in both its composition and metabolic patterns. On one hand, specific microbial entities within the gut microbiota can directly engage with T cells, functioning as "immunological triggers" that shape T-cell behavior. Simultaneously, microbial metabolites, such as short-chain fatty acids and bile acids, serve as "molecular regulators" that intricately govern T-cell function and differentiation, fine-tuning the immune response. On the other hand, the quorum-sensing mechanism, a recently recognized communication network among bacteria, also plays a pivotal role in orchestrating T-cell immunity. Additionally, the gut microbiota forms an intriguing connection with the neuro-immune regulatory axis, a largely unexplored "territory" in CRC research. Regarding treatment strategies, a diverse array of intervention approaches-including dietary modifications, the utilization of probiotics, bacteriophages, and targeted antibiotic therapies-offer promising prospects for restoring the equilibrium of the gut microbiota, thereby acting as "ecosystem renovators" that impede tumor initiation and progression. Nevertheless, the current research landscape in this field is fraught with challenges. These include significant variations in microbial composition, dietary preferences, and tumor microenvironments among individuals, a lack of large-scale cohort studies, and insufficient research that integrates tumor mutation analysis, gut microbiota investigations, and immune microenvironment evaluations. This review emphasizes the necessity for future research efforts to seamlessly incorporate multiple factors and utilize bioinformatics analysis to construct a more comprehensive "interactive map" of the gut microbiota-T cell relationship in CRC. The aim is to establish a solid theoretical basis for the development of highly effective and personalized treatment regimens, ultimately transforming the therapeutic approach to CRC.
Collapse
Affiliation(s)
- Zhuang Jing
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Zhejiang-France United Laboratory of Integrated Traditional Chinese and Modern Medicine in Colorectal Cancer, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China
| | - Wu Yinhang
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Zhejiang-France United Laboratory of Integrated Traditional Chinese and Modern Medicine in Colorectal Cancer, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China
| | - Chu Jian
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Zhejiang-France United Laboratory of Integrated Traditional Chinese and Modern Medicine in Colorectal Cancer, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China
| | - Qu Zhanbo
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Zhejiang-France United Laboratory of Integrated Traditional Chinese and Modern Medicine in Colorectal Cancer, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China
| | - Wu Xinyue
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Zhejiang-France United Laboratory of Integrated Traditional Chinese and Modern Medicine in Colorectal Cancer, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China
| | - Han Shuwen
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Zhejiang-France United Laboratory of Integrated Traditional Chinese and Modern Medicine in Colorectal Cancer, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; ASIR (Institute - Association of intelligent systems and robotics), 14B rue Henri Sainte Claire Deville, 92500 Rueil-Malmaison, France.
| |
Collapse
|
3
|
Kim BS, Artis D. The sensory neuroimmune frontier. Immunity 2025; 58:1033-1039. [PMID: 40324378 DOI: 10.1016/j.immuni.2025.03.018] [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: 02/09/2025] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 05/07/2025]
Abstract
Sensing and recognition are key properties of both the immune and nervous systems. In the immune system, pattern recognition or antigen-specific receptors represent classic motifs in innate and adaptive immunity, respectively. In the nervous system, there is a major anatomic division between how we sense stimuli from within the body (vagal sensory nervous system) and the outside world (somatosensory nervous system). However, in the last 5 years, there has been an explosion of discoveries revealing interactions between the immune and the sensory nervous systems that govern an array of physiologic and pathologic processes including allergy, infection, autoimmunity, regeneration, cancer, and beyond. Herein, we highlight recent advances that demonstrate how peripheral sensory neuroimmunology has emerged as a powerful field that provides new insights into classic immunologic processes including immune hypersensitivity, inflammation, and tissue homeostasis.
Collapse
Affiliation(s)
- Brian S Kim
- Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Allen Discovery Center for Neuroimmune Interactions, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA.
| | - David Artis
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA; Friedman Center for Nutrition and Inflammation, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA; Division of Gastroenterology and Hepatology, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA; Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA; Department of Chemistry and Chemical Biology, Boyce Thompson Institute, Cornell University, Ithaca, NY 10065, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA; Allen Discovery Center for Neuroimmune Interactions, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
4
|
Crosson T, Bretaud N, Ugolini S. Role of specialized sensory neuron subtypes in modulating peripheral immune responses. Immunity 2025; 58:1161-1174. [PMID: 40324383 DOI: 10.1016/j.immuni.2025.04.008] [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] [Received: 02/14/2025] [Revised: 04/07/2025] [Accepted: 04/07/2025] [Indexed: 05/07/2025]
Abstract
The immune and sensory nervous systems detect diverse threats, from tissue damage to infection, and coordinate protective responses to restore homeostasis. Like immune cells, sensory neurons exhibit remarkable heterogeneity, with advanced genetic models revealing that distinct subsets differentially regulate immune responses. Here, we review how various immune signals engage distinct subtypes of sensory neurons to mediate inflammatory pain, itch, relief, protective behavioral adaptations, and autonomic reflexes. We also highlight how specialized sensory neuron populations modulate immune function through the release of neuropeptides, neurokines, or glutamate. This functional specialization enables precise immunomodulation adapted to the kinetics and nature of immune responses, positioning sensory neurons as key regulators of host defense and tissue homeostasis.
Collapse
Affiliation(s)
- Théo Crosson
- Aix-Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Ninon Bretaud
- Aix-Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Sophie Ugolini
- Aix-Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France.
| |
Collapse
|
5
|
da Silva AM, de Amorim Ferreira M, Schran RG, Lückemeyer DD, Prudente AS, Ferreira J. Investigation of the participation of the TRPV1 receptor in the irritant effect of dithranol in mice. Eur J Pharmacol 2025; 994:177291. [PMID: 39870229 DOI: 10.1016/j.ejphar.2025.177291] [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: 08/09/2024] [Revised: 11/19/2024] [Accepted: 01/21/2025] [Indexed: 01/29/2025]
Abstract
Dithranol is one of the most effective topical medications for treating plaque psoriasis. However, its clinical use is limited by irritative adverse reactions to the skin, such as oedema, erythema, and pruritus, caused by poorly understood mechanisms. Because TRPV1 activation mediates skin irritation caused by several drugs, we conducted blind and randomised experiments in male and female C57BL/6 mice to elucidate the role of TRPV1 in dithranol-induced irritation. Dithranol (0.01%-0.5%) or vehicle was applied topically to the right ear of the animals. Oedema, erythema, and pruritus were monitored from 2 h to 6 days after application. Treatment with 0.5% dithranol caused oedema and erythema, but not pruritus, starting at 6 h, reaching its highest point at 1 day, and persisting up to 6 days after treatment, mainly in male mice. The 0.1% dose induced erythema but not oedema. Interestingly, topical application of 1% capsaicin was shown to defunctionalise TRPV1-positive fibres and did not influence early irritation caused by dithranol (2 h-2 days). However, it increased the late phase of irritation (5-6 days). Similarly, salicylate did not reduce the early irritation caused by dithranol but also increased the late phase. Antagonism by SB366791 and 4-tert-butylcyclohexanol did not alter skin irritation. Our results suggest that, contrary to our initial hypothesis, TRPV1 appears to act protectively against skin irritation caused by dithranol, particularly in the late stage.
Collapse
Affiliation(s)
- Ana Merian da Silva
- Graduate Program in Pharmacology, Federal University of Santa Catarina (UFSC), 88037-000, Florianópolis, SC, Brazil
| | - Marcella de Amorim Ferreira
- Graduate Program in Pharmacology, Federal University of Santa Catarina (UFSC), 88037-000, Florianópolis, SC, Brazil
| | - Roberta Giusti Schran
- Graduate Program in Pharmacology, Federal University of Santa Catarina (UFSC), 88037-000, Florianópolis, SC, Brazil
| | - Debora Denardin Lückemeyer
- Graduate Program in Pharmacology, Federal University of Santa Catarina (UFSC), 88037-000, Florianópolis, SC, Brazil; Pain Research Center, Department of Anesthesiology, University of Cincinnati, College of Medicine, Cincinnati, OH, 45267, USA
| | - Arthur Silveira Prudente
- Graduate Program in Pharmacology, Federal University of Santa Catarina (UFSC), 88037-000, Florianópolis, SC, Brazil; Pain Research Center, Department of Anesthesiology, University of Cincinnati, College of Medicine, Cincinnati, OH, 45267, USA
| | - Juliano Ferreira
- Graduate Program in Pharmacology, Federal University of Santa Catarina (UFSC), 88037-000, Florianópolis, SC, Brazil.
| |
Collapse
|
6
|
Zhai J, Li Y, Liu J, Dai C. Neuroimmune interactions: The bridge between inflammatory bowel disease and the gut microbiota. Clin Transl Med 2025; 15:e70329. [PMID: 40400119 PMCID: PMC12095209 DOI: 10.1002/ctm2.70329] [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: 11/08/2024] [Revised: 04/16/2025] [Accepted: 04/21/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND The multidimensional regulatory mechanism of the gut-brain-immune axis in the context of inflammatory bowel disease (IBD) has garnered significant attention, particularly regarding how intestinal microbiota finely regulates immune responses through immune cells and sensory neurons. MAIN BODY Metabolites produced by intestinal microbiota influence the phenotype switching of immune cells via complex signalling pathways, thereby modulating their anti-inflammatory and pro-inflammatory functions during intestinal inflammation. Furthermore, sensory neurons exhibit heightened sensitivity to microbial-derived signals, which is essential for preserving intestinal balance and controlling pathological inflammation by integrating peripheral environmental signals with local immune responses. The dynamic equilibrium between immune cells and the neuroimmunoregulation mediated by sensory neurons collectively sustains immune homeostasis within the intestine. However, this coordination mechanism is markedly disrupted under the pathological conditions associated with IBD. CONCLUSION An in-depth exploration of the interactions among immune cells, gut microbiota and sensory neurons may yield significant insights into the pathological mechanisms underlying IBD and guide the creation of new treatment approaches. KEY POINTS The gut microbiota regulates the gut-brain-immune axis, modulating neuroimmune interactions in IBD. Microbiota-derived metabolites influence immune cells, thereby affecting neurons. Neurons secrete mediators, enabling bidirectional neuroimmune communication essential for intestinal homeostasis. Disruptions contribute to IBD, offering therapeutic targets.
Collapse
Affiliation(s)
- Jinxia Zhai
- Department of GastroenterologyFirst Affiliated Hospital, China Medical UniversityShenyang CityLiaoning ProvinceChina
| | - Yingjie Li
- Department of GastroenterologyFirst Affiliated Hospital, Jinzhou Medical UniversityJinzhou CityLiaoning ProvinceChina
| | - Jiameng Liu
- Department of GastroenterologyFirst Affiliated Hospital, China Medical UniversityShenyang CityLiaoning ProvinceChina
| | - Cong Dai
- Department of GastroenterologyFirst Affiliated Hospital, China Medical UniversityShenyang CityLiaoning ProvinceChina
| |
Collapse
|
7
|
Waizman DA, Brown-Soler I, Martin AL, Ma Y, Zhou K, Israni-Winger K, Zhang C, Medzhitov R, Launay P, Michieletto MF, Henao-Mejia J, Palm NW, Craft J, Eisenstein A, Wang A. Skin damage signals mediate allergic sensitization to spatially unlinked antigen. Sci Immunol 2025; 10:eadn0688. [PMID: 40184440 PMCID: PMC12100540 DOI: 10.1126/sciimmunol.adn0688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 06/10/2024] [Accepted: 03/13/2025] [Indexed: 04/06/2025]
Abstract
Our current understanding of immunity to pathogens suggests that anatomic coupling of antigens with danger signals is a required feature for the formation of immune memory. However, in the context of pathogen-independent inflammation, the stringency of this anatomical coupling is unclear. Here, we demonstrate that multiple modes of skin injury were sufficient to induce a humoral response to antigens introduced in the gut. Skin damage induced a narrow subset of endocrine cytokines that were necessary and sufficient for the priming of antigens introduced at various distal tissues. Thus, in addition to "local priming" of antigen entering through damaged skin, there also exists another paradigm of "remote priming" where anatomical coupling is not essential because of the dissemination of damage-associated intermediaries. Our findings have implications for understanding the fundamental mechanisms of the formation of humoral memory with wide implications for diseases such as food allergy and in vaccinology.
Collapse
Affiliation(s)
- Daniel A. Waizman
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Isabela Brown-Soler
- Department of Dermatology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Anjelica L. Martin
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Yifan Ma
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Kenneth Zhou
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | | | - Cuiling Zhang
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Ruslan Medzhitov
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Pierre Launay
- Centre de Recherche sur l’Inflammation, INSERM UMR1149, CNRS EMR8252, Université Paris Cité, Paris, France
| | - Michaël F. Michieletto
- Institute for Immunology and Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jorge Henao-Mejia
- Institute for Immunology and Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Mark Foundation Center for Immunotherapy, Immune Signaling, and Radiation, Perelman School of Medicine, and Division of Protective Immunity, Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Noah W. Palm
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Joe Craft
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Internal Medicine (Rheumatology, Allergy, and Immunology), Yale School of Medicine, New Haven, CT 06510, USA
| | - Anna Eisenstein
- Department of Dermatology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Andrew Wang
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Internal Medicine (Rheumatology, Allergy, and Immunology), Yale School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
8
|
Han C, Zhu X, Sokol CL. Neuroimmune Circuits in Allergic Diseases. Annu Rev Immunol 2025; 43:367-394. [PMID: 39977604 DOI: 10.1146/annurev-immunol-082423-032154] [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: 02/22/2025]
Abstract
Communication between the nervous and immune systems is evolutionarily conserved. From primitive eukaryotes to higher mammals, neuroimmune communication utilizes multiple complex and complementary mechanisms to trigger effective but balanced responses to environmental dangers such as allergens and tissue damage. These responses result from a tight integration of the nervous and immune systems, and accumulating evidence suggests that this bidirectional communication is crucial in modulating the initiation and development of allergic inflammation. In this review, we discuss the basic mechanisms of neuroimmune communication, with a focus on the recent advances underlying the importance of such communication in the allergic immune response. We examine neuronal sensing of allergens, how neuropeptides and neurotransmitters regulate allergic immune cell functions, and how inflammatory factors derived from immune cells coordinate complex peripheral and central nervous system responses. Furthermore, we highlight how fundamental aspects of host biology, from aging to circadian rhythm, might affect these pathways. Appreciating neuroimmune communications as an evolutionarily conserved and functionally integrated system that is fundamentally involved in type 2 immunity will provide new insights into allergic inflammation and reveal exciting opportunities for the management of acute and chronic allergic diseases.
Collapse
Affiliation(s)
- Cai Han
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA;
| | - Xueping Zhu
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA;
| | - Caroline L Sokol
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA;
| |
Collapse
|
9
|
Li S, Xie J, Xiang J, Yan R, Liu J, Fan Q, Lu L, Wu J, Liu J, Xue Y, Fu T, Li Z. Corneal Sensory Nerve Injury Disrupts Lacrimal Gland Function by Altering Circadian Rhythms in Mice. Invest Ophthalmol Vis Sci 2025; 66:40. [PMID: 40238116 PMCID: PMC12011127 DOI: 10.1167/iovs.66.4.40] [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: 01/26/2025] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
Purpose To investigate the impact of corneal sensory nerve injury on lacrimal gland function, focusing on mechanisms involving the superior salivatory nucleus (SSN), circadian rhythm disruption, immune microenvironment alterations, and the potential for neural regeneration. Methods A murine model of corneal sensory nerve injury was used to assess lacrimal gland function, with tear secretion measured using the phenol red thread test. Transcriptomic analysis of lacrimal glands examined circadian rhythm and immune-related gene expression. Basic fibroblast growth factor (bFGF) was used to promote corneal nerve regeneration, and its effects on tear secretion and nerve repair were evaluated. Results Corneal nerve injury resulted in a 35% reduction in tear secretion and significantly impaired SSN activity, as evidenced by a 31% decrease in c-FOS-positive neurons in choline acetyltransferase (ChAT)-expressing neurons. Transcriptomic analysis revealed significant downregulation of immune-related pathways, including Toll-like receptor (TLR), NOD-like receptor (NLR), and T-cell receptor signaling. Circadian rhythm gene expression exhibited phase shifts, with a 2.13-hour delay in peak expression and a substantial change in the number and types of rhythmic genes, which were enriched in different signaling pathways. The bFGF treatment restored tear secretion by 22% and promoted nerve regeneration, although nerve fiber density remained 74% lower than that of controls. Conclusions Corneal sensory nerve injury disrupts both central and peripheral circadian clock functions in the lacrimal gland, leading to reduced tear secretion and immune dysregulation. These findings highlight the novel role of circadian rhythms and neural-immune interactions in lacrimal gland dysfunction. Neural regeneration strategies, such as bFGF, offer therapeutic potential for dry eye syndrome, providing new directions for clinical intervention.
Collapse
Affiliation(s)
- Senmao Li
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- International Ocular Surface Research Center, Institute of Ophthalmology and Key Laboratory for Regenerative Medicine, Jinan University Medical School, Guangzhou, China
| | - Jingbin Xie
- International Ocular Surface Research Center, Institute of Ophthalmology and Key Laboratory for Regenerative Medicine, Jinan University Medical School, Guangzhou, China
| | - Jiayan Xiang
- International Ocular Surface Research Center, Institute of Ophthalmology and Key Laboratory for Regenerative Medicine, Jinan University Medical School, Guangzhou, China
| | - Ruyu Yan
- International Ocular Surface Research Center, Institute of Ophthalmology and Key Laboratory for Regenerative Medicine, Jinan University Medical School, Guangzhou, China
| | - Jiangman Liu
- International Ocular Surface Research Center, Institute of Ophthalmology and Key Laboratory for Regenerative Medicine, Jinan University Medical School, Guangzhou, China
| | - Qiwei Fan
- International Ocular Surface Research Center, Institute of Ophthalmology and Key Laboratory for Regenerative Medicine, Jinan University Medical School, Guangzhou, China
| | - Liyuan Lu
- International Ocular Surface Research Center, Institute of Ophthalmology and Key Laboratory for Regenerative Medicine, Jinan University Medical School, Guangzhou, China
| | - Jiaxin Wu
- International Ocular Surface Research Center, Institute of Ophthalmology and Key Laboratory for Regenerative Medicine, Jinan University Medical School, Guangzhou, China
| | - Jun Liu
- International Ocular Surface Research Center, Institute of Ophthalmology and Key Laboratory for Regenerative Medicine, Jinan University Medical School, Guangzhou, China
| | - Yunxia Xue
- International Ocular Surface Research Center, Institute of Ophthalmology and Key Laboratory for Regenerative Medicine, Jinan University Medical School, Guangzhou, China
| | - Ting Fu
- International Ocular Surface Research Center, Institute of Ophthalmology and Key Laboratory for Regenerative Medicine, Jinan University Medical School, Guangzhou, China
| | - Zhijie Li
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- International Ocular Surface Research Center, Institute of Ophthalmology and Key Laboratory for Regenerative Medicine, Jinan University Medical School, Guangzhou, China
| |
Collapse
|
10
|
Fang X, Chen Y, Ding H, Huang C, Hu H, Zhang C, Lin Y, Wang Q, Hu X, Lin Y, Chen Y, Zhang N, Yuan X, Huang Y, Li W, Niu S, Lin J, Yang B, Yuan T, Zhang W. Staphylococcus Aureus Tames Nociceptive Neurons to Suppress Synovial Macrophage Responses for Sustained Infection in Septic Arthritis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409251. [PMID: 39960341 PMCID: PMC11984863 DOI: 10.1002/advs.202409251] [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: 08/06/2024] [Revised: 01/28/2025] [Indexed: 04/12/2025]
Abstract
The interaction between the nervous system and immune system during chronic bacterial infection remains unclear. Here, it is reported that Staphylococcus aureus (S. aureus) infection induces calcitonin gene-related peptide (CGRP) secretion from intra-articular transient receptor potential cation channel subfamily V member 1 positive (TRPV1+) nociceptive nerves through its pore-forming toxin (PFT) α-hemolysin. The released CGRP then inhibits the production of chemotactic cytokines by CX3CR1+ tissue-resident synovial lining macrophages via receptor activity modifying protein 1 (RAMP1) receptors at the onset of septic arthritis. During the subsequent chronic course of infection, the continuous release of CGRP triggered by pain has a lasting effect on the antimicrobial capabilities of macrophages, thereby promoting bacterial survival and joint damage. This evidence suggests a critical role for neuroimmune regulation in S. aureus-induced chronic septic arthritis. CGRP receptor antagonism may reduce joint destruction, thus providing a new option for treating bone and joint infections.
Collapse
Affiliation(s)
- Xinyu Fang
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhou350000China
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Fujian Medical UniversityFuzhou350000China
- Fujian Provincial Institute of Orthopedicsthe First Affiliated Hospital, Fujian Medical UniversityFuzhou350000China
| | - Yang Chen
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhou350000China
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Fujian Medical UniversityFuzhou350000China
- Fujian Provincial Institute of Orthopedicsthe First Affiliated Hospital, Fujian Medical UniversityFuzhou350000China
| | - Haiqi Ding
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhou350000China
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Fujian Medical UniversityFuzhou350000China
- Fujian Provincial Institute of Orthopedicsthe First Affiliated Hospital, Fujian Medical UniversityFuzhou350000China
| | - Changyu Huang
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhou350000China
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Fujian Medical UniversityFuzhou350000China
- Fujian Provincial Institute of Orthopedicsthe First Affiliated Hospital, Fujian Medical UniversityFuzhou350000China
| | - Hongxin Hu
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhou350000China
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Fujian Medical UniversityFuzhou350000China
- Fujian Provincial Institute of Orthopedicsthe First Affiliated Hospital, Fujian Medical UniversityFuzhou350000China
| | - Chaofan Zhang
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhou350000China
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Fujian Medical UniversityFuzhou350000China
- Fujian Provincial Institute of Orthopedicsthe First Affiliated Hospital, Fujian Medical UniversityFuzhou350000China
| | - Yunzhi Lin
- Department of Stomatology, The First Affiliated HospitalFujian Medical UniversityFuzhou350000China
| | - Qijin Wang
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhou350000China
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Fujian Medical UniversityFuzhou350000China
- Fujian Provincial Institute of Orthopedicsthe First Affiliated Hospital, Fujian Medical UniversityFuzhou350000China
| | - Xueni Hu
- Department of Laboratory MedicineThe First Affiliated Hospital of Fujian Medical UniversityFuzhou350000China
| | - Yiming Lin
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhou350000China
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Fujian Medical UniversityFuzhou350000China
- Fujian Provincial Institute of Orthopedicsthe First Affiliated Hospital, Fujian Medical UniversityFuzhou350000China
| | - Yongfa Chen
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhou350000China
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Fujian Medical UniversityFuzhou350000China
- Fujian Provincial Institute of Orthopedicsthe First Affiliated Hospital, Fujian Medical UniversityFuzhou350000China
| | - Nanxin Zhang
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhou350000China
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Fujian Medical UniversityFuzhou350000China
- Fujian Provincial Institute of Orthopedicsthe First Affiliated Hospital, Fujian Medical UniversityFuzhou350000China
| | - Xuhui Yuan
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhou350000China
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Fujian Medical UniversityFuzhou350000China
- Fujian Provincial Institute of Orthopedicsthe First Affiliated Hospital, Fujian Medical UniversityFuzhou350000China
| | - Ying Huang
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhou350000China
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Fujian Medical UniversityFuzhou350000China
- Fujian Provincial Institute of Orthopedicsthe First Affiliated Hospital, Fujian Medical UniversityFuzhou350000China
| | - Wenbo Li
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhou350000China
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Fujian Medical UniversityFuzhou350000China
- Fujian Provincial Institute of Orthopedicsthe First Affiliated Hospital, Fujian Medical UniversityFuzhou350000China
| | - Susheng Niu
- Key Laboratory of Orthopedics & Traumatology of Traditional Chinese Medicine and Rehabilitation Ministry of EducationFujian University of Traditional Chinese MedicineFuzhou350000China
| | - Jianhua Lin
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhou350000China
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Fujian Medical UniversityFuzhou350000China
- Fujian Provincial Institute of Orthopedicsthe First Affiliated Hospital, Fujian Medical UniversityFuzhou350000China
| | - Bin Yang
- Department of Laboratory MedicineThe First Affiliated Hospital of Fujian Medical UniversityFuzhou350000China
| | - Tifei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health CenterShanghai Jiaotong University School of Medicine and School of PsychologyShanghai200000China
- Co‐innovation Center of NeuroregenerationNantong UniversityNantongJiangsu226019China
| | - Wenming Zhang
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhou350000China
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Fujian Medical UniversityFuzhou350000China
- Fujian Provincial Institute of Orthopedicsthe First Affiliated Hospital, Fujian Medical UniversityFuzhou350000China
| |
Collapse
|
11
|
Erdogan O, Hu XQ, Chiu IM. Sensory neurons on guard: roles in pathogen defense and host immunity. Curr Opin Immunol 2025; 93:102541. [PMID: 40015178 PMCID: PMC11884989 DOI: 10.1016/j.coi.2025.102541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 02/11/2025] [Accepted: 02/19/2025] [Indexed: 03/01/2025]
Abstract
The nervous system, like the immune system, constantly interfaces with the environment, encountering threats, including pathogens. Recent discoveries reveal an emerging role for sensory neurons in host defense and immunity. Sensory neurons detect infections either by directly sensing microbial signals or through immune mediators. Beyond pathogen detection, they modulate immune responses and local inflammation by interacting with immune cells, influencing inflammation and pathogen clearance. Additionally, sensory neurons trigger protective reflexes - such as pain, coughing, sneezing, and itching - that can help expel pathogens but may also facilitate their spread. Sensory neurons may also encode and shape long-term immunity. Understanding the roles of neurons in pathogen defense could offer new insights into infectious diseases and highlight therapeutic opportunities for immune modulation.
Collapse
Affiliation(s)
- Ozge Erdogan
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA; Department of Restorative Dentistry and Biomaterial Sciences, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Xiao-Qian Hu
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA; Department of Rehabilitation Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Isaac M Chiu
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
12
|
Lozo KW, Aktipis A, Alcock J. Neuroimmune Pain and Its Manipulation by Pathogens. Evol Appl 2025; 18:e70098. [PMID: 40270922 PMCID: PMC12015744 DOI: 10.1111/eva.70098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 03/08/2025] [Indexed: 04/25/2025] Open
Abstract
Recent studies highlight extensive crosstalk that exists between sensory neurons responsible for pain and the immune system. Cutaneous pain neurons detect harmful microbes, recruit immune cells, and produce anticipatory immunity in nearby tissues. These complementary systems generally protect hosts from infections. At the same time, neuroimmune pain is vulnerable to manipulation. Some pathogens evade immunity activated by nociceptors by producing opioid analogs and by interfering with sensory nerve function. Other organisms manipulate neuroimmune pain by increasing it. Hosts may gain protection from interference by adjusting pain sensitivity. Nociceptive sensitization follows expectations of signal detection theory and the smoke detector principle, allowing pain to be more easily triggered in response to microbial threats and damage. However, pain sensitization at the spinal level and cortical responses to pain are themselves the target of manipulation by parasites and other organisms. Here we review examples of parasites, bacteria, and other medically important organisms that interfere with pain signaling and describe their implications for public health, infectious disease, and the treatment of pain.
Collapse
Affiliation(s)
- Kevin W. Lozo
- University of Pittsburgh Medical CenterPittsburghPennsylvaniaUSA
| | - Athena Aktipis
- Department of PsychologyArizona State UniversityTempeArizonaUSA
- Center for Evolution and MedicineArizona State UniversityTempeArizonaUSA
| | - Joe Alcock
- Department of Emergency MedicineUniversity of New MexicoAlbuquerqueNew MexicoUSA
| |
Collapse
|
13
|
Tamari M, Ver Heul AM. Neuroimmune mechanisms of type 2 inflammation in the skin and lung. Allergol Int 2025; 74:177-186. [PMID: 40064568 DOI: 10.1016/j.alit.2025.02.001] [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: 01/20/2025] [Revised: 02/10/2025] [Accepted: 02/12/2025] [Indexed: 04/01/2025] Open
Abstract
Type 2 inflammation has a major role in barrier tissues such as the skin and airways and underlies common conditions including atopic dermatitis (AD) and asthma. Cytokines including interleukin 4 (IL-4), IL-5, and IL-13 are key immune signatures of type 2 inflammation and are the targets of multiple specific therapeutics for allergic diseases. Despite shared core immune mechanisms, the distinct structures and functions of the skin and airways lead to unique therapeutic responses. It is increasingly recognized that the nervous system has a major role in sensing and directing inflammatory processes. Indeed, crosstalk between type 2 immune activation and somatosensory functions mediates tissue-specific signatures such as itching in the skin. However, neuroimmune interactions are shaped by distinct neuronal and immune landscapes, and differ between the skin and airways. In the skin, dorsal root ganglia-derived neurons mediate pruritus via type 2 cytokines and neurogenic inflammation by mast cell or basophil activation. Conversely, vagal ganglia-derived neurons regulate airway immune responses by releasing neuropeptides/neurotransmitters such as calcitonin gene-related peptides, neuromedin U, acetylcholine, and noradrenaline. Sensory neuron-derived vasoactive intestinal peptide forms a feedback loop with IL-5, amplifying eosinophilic inflammation in the airways, a mechanism that is absent in the skin. These differences influence the efficacy of cytokine-targeted therapies. For instance, IL-4/IL-13-targeted therapies like dupilumab demonstrate efficacy in AD and allergic airway diseases, whereas IL-5-targeted therapies are effective in eosinophilic asthma but not AD. Understanding these neuroimmune interactions underscores the need for tailored therapeutic approaches to address allergic diseases where barrier tissues are involved.
Collapse
Affiliation(s)
- Masato Tamari
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan.
| | - Aaron M Ver Heul
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, USA.
| |
Collapse
|
14
|
Wang Q, Chen Y, Ding H, Cai Y, Yuan X, Lv J, Huang J, Huang J, Zhang C, Hong Z, Li H, Huang Y, Lin J, Yuan L, Lin L, Yu S, Zhang C, Lin J, Li W, Chang C, Yang B, Zhang W, Fang X. Optogenetic activation of mechanical nociceptions to enhance implant osseointegration. Nat Commun 2025; 16:3093. [PMID: 40164597 PMCID: PMC11958704 DOI: 10.1038/s41467-025-58336-x] [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: 09/22/2024] [Accepted: 03/18/2025] [Indexed: 04/02/2025] Open
Abstract
Orthopedic implants with high elastic modulus often suffer from poor osseointegration due to stress shielding, a phenomenon that suppresses the expression of intracellular mechanotransduction molecules (IMM) such as focal adhesion kinase (FAK). We find that reduced FAK expression under stress shielding is also mediated by decreased calcitonin gene-related peptide (CGRP) released from Piezo2+ mechanosensitive nerves surrounding the implant. To activate these nerves minimally invasively, we develop a fully implantable, wirelessly rechargeable optogenetic device. In mice engineered to express light-sensitive channels in Piezo2+ neurons, targeted stimulation of the L2-3 dorsal root ganglia (DRG) enhances localized CGRP release near the implant. This CGRP elevation activates the Protein Kinase A (PKA)/FAK signaling pathway in bone marrow mesenchymal stem cells (BMSCs), thereby enhancing osteogenesis and improving osseointegration. Here we show that bioelectronic modulation of mechanosensitive nerves offers a strategy to address implant failure, bridging neuroregulation and bone bioengineering.
Collapse
Affiliation(s)
- Qijin Wang
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Yang Chen
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Haiqi Ding
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Yuanqing Cai
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xuhui Yuan
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Jianhua Lv
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Jiagu Huang
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Jiexin Huang
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Chaofan Zhang
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Zihao Hong
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Hongyan Li
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Ying Huang
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Jiamin Lin
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Lin Yuan
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Lan Lin
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Shaolin Yu
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Canhong Zhang
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Jianhua Lin
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Wenbo Li
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Cheng Chang
- Institute of New Materials, Guangdong Academy of Sciences, Guangdong-Hong Kong Joint Laboratory of Modern Surface Engineering Technology, Guangdong Provincial Key Laboratory of Modern Surface Engineering Technology, Guangzhou, Guangdong, PR China
| | - Bin Yang
- Department of Laboratory Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.
- Department of Laboratory Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
| | - Wenming Zhang
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
| | - Xinyu Fang
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
15
|
MacDonald DI, Jayabalan M, Seaman JT, Balaji R, Nickolls AR, Chesler AT. Pain persists in mice lacking both Substance P and CGRPα signaling. eLife 2025; 13:RP93754. [PMID: 40100256 PMCID: PMC11919252 DOI: 10.7554/elife.93754] [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] [Indexed: 03/20/2025] Open
Abstract
The neuropeptides Substance P and CGRPα have long been thought important for pain sensation. Both peptides and their receptors are expressed at high levels in pain-responsive neurons from the periphery to the brain making them attractive therapeutic targets. However, drugs targeting these pathways individually did not relieve pain in clinical trials. Since Substance P and CGRPα are extensively co-expressed, we hypothesized that their simultaneous inhibition would be required for effective analgesia. We therefore generated Tac1 and Calca double knockout (DKO) mice and assessed their behavior using a wide range of pain-relevant assays. As expected, Substance P and CGRPα peptides were undetectable throughout the nervous system of DKO mice. To our surprise, these animals displayed largely intact responses to mechanical, thermal, chemical, and visceral pain stimuli, as well as itch. Moreover, chronic inflammatory pain and neurogenic inflammation were unaffected by loss of the two peptides. Finally, neuropathic pain evoked by nerve injury or chemotherapy treatment was also preserved in peptide-deficient mice. Thus, our results demonstrate that even in combination, Substance P and CGRPα are not required for the transmission of acute and chronic pain.
Collapse
Affiliation(s)
- Donald Iain MacDonald
- National Center for Complementary and Integrative Health, National Institutes of HealthBethesdaUnited States
| | - Monessha Jayabalan
- National Center for Complementary and Integrative Health, National Institutes of HealthBethesdaUnited States
| | - Jonathan T Seaman
- National Center for Complementary and Integrative Health, National Institutes of HealthBethesdaUnited States
| | - Rakshita Balaji
- National Center for Complementary and Integrative Health, National Institutes of HealthBethesdaUnited States
| | - Alec R Nickolls
- National Center for Complementary and Integrative Health, National Institutes of HealthBethesdaUnited States
| | - Alexander Theodore Chesler
- National Center for Complementary and Integrative Health, National Institutes of HealthBethesdaUnited States
- National Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaUnited States
| |
Collapse
|
16
|
Hołdrowicz A, Żebrowska A. Molecular Link Between Psoriasis and Depression-Update on Pathophysiology. Int J Mol Sci 2025; 26:2467. [PMID: 40141110 PMCID: PMC11942400 DOI: 10.3390/ijms26062467] [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: 02/11/2025] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
Psoriasis disease is a chronic, systemic condition. Various epidemiological studies have indicated a connection between psoriasis and psychiatric diseases. It is obvious that easily visible psoriatic skin lesions cause stigmatization of patients and impact noticeably their life quality, increasing the risk of anxiety and depressive disorders. More and more attention is recently being paid to the common pathogenesis of psoriasis and depression. The underlying cause of psoriasis is chronic inflammation, and depression is also increasingly recognized as a result of neuroinflammation. Therefore, the complexity of the processes underlying both disease entities implies the need to observe psoriatic patients in terms of possible comorbidities, such as mental disorders, regardless of the severity of skin lesions and social stigmatization. This study aims to present an update on the common pathophysiology of both diseases.
Collapse
Affiliation(s)
| | - Agnieszka Żebrowska
- Department of Dermatology and Venereology, Medical University of Lodz, 90-647 Lodz, Poland;
| |
Collapse
|
17
|
Zhou X, Ma Z, Cheng Q, Jiang N, Li J, Zhan T, Yuan N, Chen Y, Wang L, Wang J, Li Q, Jia W, Xie B, Zhao Y, Zhang B, Yang B, Dai C, Wei L, Liu J, Chen Z, Lan P. Sour neuronal signalling attenuates macrophage-mediated liver injury. J Hepatol 2025:S0168-8278(25)00135-7. [PMID: 40058705 DOI: 10.1016/j.jhep.2025.02.026] [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: 11/05/2024] [Revised: 02/01/2025] [Accepted: 02/12/2025] [Indexed: 05/01/2025]
Abstract
BACKGROUND & AIMS Liver injury, a common pathophysiological basis of various liver diseases, is associated with inflammation. Hepatic nerves regulate inflammation. However, the specific signals that trigger inflammation and methods to treat inflammation by targeting nerves remain unknown. METHODS First, we constructed an animal model to detect the effect of sour stimuli on liver ischaemia-reperfusion injury (IRI) in mice. Next, we analysed the altered gene expression of neurons during liver IRI by single-cell sequencing. In addition, we explored the effect of sour stimuli on liver IRI in mice. Finally, we designed clinical trials to explore the effect of sour stimuli on liver IRI during hepatectomy. RESULTS In this study, single-cell sequencing data from the liver and celiac ganglion showed that TAFA2 was induced in neurones during liver IRI, whereas sour stimuli decreased TAFA2 production and liver injury. In vivo studies showed that TAFA2 ablation and specific knockdown in neurones reduce liver injury. Using FLAG-tagged TAFA2, we found that TAFA2 interacted with chemokine C-C-motif receptor 2 (CCR2) and promoted macrophage activation, consistent with RNA sequencing data showing that TAFA2 induced the expression of inflammatory genes in wild-type macrophages, but not in Ccr2 knockout macrophages. Moreover, patients exposed to sour stimuli exhibited less severe liver IRI during hepatectomy. CONCLUSIONS Our results reveal a neuroimmune interaction in which neurone-derived TAFA2 recruits CCR2+ macrophages to the liver and triggers liver injury, which is at least partly reduced by nerve signalling in response to sour stimuli, i.e. consumption of acidic substances. Our findings provide new insights into the brain-liver axis and potential therapeutic approaches for liver injury. IMPACT AND IMPLICATIONS In this study, we demonstrated that sour stimuli, which are related to consumption of acidic foods, are at least partly responsible for reducing human and mouse liver ischaemia-reperfusion injury (IRI), and we confirmed the important role of the brain-liver axis in liver IRI. In this study, we found that the brain-liver axis increases liver IRI through the secretion of TAFA2 protein. TAFA2 mediated liver IRI through the recruitment and activation of macrophages via the receptor CCR2. Additionally, TAFA2 was shown to induce a proinflammatory transcriptional profile in macrophages. Our findings provide new insights into the brain-liver axis and uncover a potential therapeutic strategy to reduce IRI. CLINICAL TRIAL NUMBER This clinical trial was registered with the Chinese Clinical Trial Registry (ChiCTR2400088096).
Collapse
Affiliation(s)
- Xi Zhou
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, People's Republic of China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, 430030 Wuhan, People's Republic of China
| | - Zhibo Ma
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, People's Republic of China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, 430030 Wuhan, People's Republic of China
| | - Qi Cheng
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Na Jiang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, People's Republic of China
| | - Junbo Li
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, People's Republic of China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, 430030 Wuhan, People's Republic of China
| | - Tianao Zhan
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Naonao Yuan
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, People's Republic of China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, 430030 Wuhan, People's Republic of China
| | - Yanyu Chen
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, People's Republic of China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, 430030 Wuhan, People's Republic of China
| | - Lu Wang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, People's Republic of China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, 430030 Wuhan, People's Republic of China
| | - Jingzeng Wang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, People's Republic of China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, 430030 Wuhan, People's Republic of China
| | - Qingwen Li
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, People's Republic of China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, 430030 Wuhan, People's Republic of China
| | - Wenlong Jia
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Bowen Xie
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, People's Republic of China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, 430030 Wuhan, People's Republic of China
| | - Yuanyuan Zhao
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, People's Republic of China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, 430030 Wuhan, People's Republic of China
| | - Bo Zhang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, People's Republic of China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, 430030 Wuhan, People's Republic of China
| | - Bo Yang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, People's Republic of China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, 430030 Wuhan, People's Republic of China
| | - Chen Dai
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, People's Republic of China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, 430030 Wuhan, People's Republic of China
| | - Lai Wei
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, People's Republic of China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, 430030 Wuhan, People's Republic of China
| | - Jing Liu
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, People's Republic of China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, 430030 Wuhan, People's Republic of China.
| | - Zhishui Chen
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, People's Republic of China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, 430030 Wuhan, People's Republic of China.
| | - Peixiang Lan
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, People's Republic of China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, 430030 Wuhan, People's Republic of China.
| |
Collapse
|
18
|
Meerschaert KA, Chiu IM. The gut-brain axis and pain signalling mechanisms in the gastrointestinal tract. Nat Rev Gastroenterol Hepatol 2025; 22:206-221. [PMID: 39578592 DOI: 10.1038/s41575-024-01017-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/25/2024] [Indexed: 11/24/2024]
Abstract
Visceral pain is a major clinical problem and one of the most common reasons patients with gastrointestinal disorders seek medical help. Peripheral sensory neurons that innervate the gut can detect noxious stimuli and send signals to the central nervous system that are perceived as pain. There is a bidirectional communication network between the gastrointestinal tract and the nervous system that mediates pain through the gut-brain axis. Sensory neurons detect mechanical and chemical stimuli within the intestinal tissues, and receive signals from immune cells, epithelial cells and the gut microbiota, which results in peripheral sensitization and visceral pain. This Review focuses on molecular communication between these non-neuronal cell types and neurons in visceral pain. These bidirectional interactions can be dysregulated during gastrointestinal diseases to exacerbate visceral pain. We outline the anatomical pathways involved in pain processing in the gut and how cell-cell communication is integrated into this gut-brain axis. Understanding how bidirectional communication between the gut and nervous system is altered during disease could provide new therapeutic targets for treating visceral pain.
Collapse
Affiliation(s)
| | - Isaac M Chiu
- Department of Immunology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
19
|
Liu R, Buttaci DR, Sokol CL. Neurogenic inflammation and itch in barrier tissues. Semin Immunol 2025; 77:101928. [PMID: 39798211 PMCID: PMC11893243 DOI: 10.1016/j.smim.2024.101928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 12/31/2024] [Accepted: 12/31/2024] [Indexed: 01/15/2025]
Abstract
Once regarded as distinct systems, the nervous system and the immune system are now recognized for their complex interactions within the barrier tissues. The neuroimmune circuitry comprises a dual-network system that detects external and internal disturbances, providing critical information to tailor a context-specific response to various threats to tissue integrity, such as wounding or exposure to noxious and harmful stimuli like pathogens, toxins, or allergens. Using the skin as an example of a barrier tissue with the polarized sensory neuronal responses of itch and pain, we explore the molecular pathways driving neuronal activation and the effects of this activation on the immune response. We then apply these findings to other barrier tissues, to find common pathways controlling neuroimmune responses in the barriers.
Collapse
Affiliation(s)
- Rebecca Liu
- Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy & Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Dean R Buttaci
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy & Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Caroline L Sokol
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy & Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
20
|
Keenan EL, Granstein RD. Proinflammatory cytokines and neuropeptides in psoriasis, depression, and anxiety. Acta Physiol (Oxf) 2025; 241:e70019. [PMID: 39960105 DOI: 10.1111/apha.70019] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 01/29/2025] [Accepted: 02/01/2025] [Indexed: 05/09/2025]
Abstract
Psoriasis vulgaris has established associations with psychiatric conditions such as depression, anxiety, and chronic stress. This review aims to evaluate current theories and evidence regarding the role of proinflammatory cytokines and neuropeptides in connecting systemic inflammation, psychological stress, and inflammatory skin diseases, namely psoriasis. A literature review was conducted to analyze studies that explore the connections between psoriasis, psychiatric conditions, and biological mediators, including inflammatory cytokines [interferon (IFN)-γ, interleukin (IL)-1, IL-2, IL-6, IL-12, tumor necrosis factor (TNF)-α, IL-22, IL-17], neuropeptides [calcitonin gene-related peptide (CGRP), substance P (SP), and vasoactive intestinal peptide (VIP)], as well as the hypothalamic-pituitary-adrenal (HPA) axis. Existing literature indicates that psychiatric state can influence cutaneous conditions through immune, neural, and endocrine mediators. The elevated rates of anxiety and depression observed in psoriasis patients are likely due to both the inflammatory process itself and the chronic stress associated with disease management, highlighting the importance of managing stress, and addressing mental health to improve clinical outcomes. While the literature suggests proinflammatory cytokines and neuropeptides may be key links between systemic inflammation, psoriasis, and psychiatric comorbidities, further research is necessary to continue to elucidate physiological mechanisms and explore the potential for new treatment modalities.
Collapse
Affiliation(s)
- Emily L Keenan
- Israel Englander Department of Dermatology, Weill Cornell Medicine, New York, New York, USA
| | - Richard D Granstein
- Israel Englander Department of Dermatology, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
21
|
Jones MR, Jones J, Pandu P, Liu C, Carey CD, Falo LD, Albers KM. Neurturin GF Enhances the Acute Cytokine Response of Inflamed Skin. J Invest Dermatol 2025; 145:583-592. [PMID: 39122143 DOI: 10.1016/j.jid.2024.07.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/24/2024] [Accepted: 07/12/2024] [Indexed: 08/12/2024]
Abstract
Epidermal keratinocytes, immune cells, and sensory nerves all contribute to immune balance and skin homeostasis. Keratinocyte's release of GFs, neuromodulators, and immune activators is particularly important because each can evoke local (skin) and systemic (ie, immune and neural) responses that can initiate and exacerbate skin pathophysiology. From studies of skin and neural GFs, we hypothesized that neurturin (Nrtn), a member of the GDNF family that is expressed in the skin, has particular importance in this process. In this study, we examine how elevation of Nrtn in skin keratinocytes impacts early cytokine expression in response to complete Freund's adjuvant-mediated inflammation. Nrtn-overexpressing mice and wild-type mice injected with Nrtn exhibit an enhanced level of TNFα and IL-1β cytokines in the skin, a response previously shown to support healing. In vitro assays suggest that one source of the Nrtn-induced TNFα increase is keratinocytes, which are shown to express Nrtn and mRNAs encoding the Nrtn receptors GFRα2, Ret, ITGB1, and NCAM. These findings support the contribution of keratinocyte-derived Nrtn as an autocrine/paracrine factor that acts as a first-line defense molecule that regulates the initial cytokine response to inflammatory challenge.
Collapse
Affiliation(s)
- Marsha Ritter Jones
- Department of Anesthesia & Perioperative Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - James Jones
- Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Prathyusha Pandu
- Rutgers University School of Medicine, East Brunswick, New Jersey, USA
| | - Chunyan Liu
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Cara D Carey
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Louis D Falo
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Kathryn M Albers
- Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
22
|
Kotlyar J, Granstein RD. Neuroimmunology of psoriasis: Possible roles for calcitonin gene-related peptide in its pathogenesis. Brain Behav Immun Health 2025; 44:100958. [PMID: 40008232 PMCID: PMC11851231 DOI: 10.1016/j.bbih.2025.100958] [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: 09/24/2024] [Revised: 01/21/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
The nervous system has a complex interplay with the immune system, especially at barrier sites such as the skin. This allows it to play a role in a variety of cutaneous inflammatory disorders such as psoriasis, exerting effects on various immune cells via effector molecules such as neuropeptides. In this review, we discuss the role of calcitonin gene-related peptide in modulating the immune system and inflammation, with a focus on psoriasis.
Collapse
Affiliation(s)
- Joshua Kotlyar
- Israel Englander Department of Dermatology, Weill Cornell Medicine, 1305 York Avenue, WGC9, New York, NY, 10021, USA
- SUNY Downstate Health Sciences University College of Medicine, 450 Clarkson Avenue, Brooklyn, NY, 11203, USA
| | - Richard D. Granstein
- Israel Englander Department of Dermatology, Weill Cornell Medicine, 1305 York Avenue, WGC9, New York, NY, 10021, USA
| |
Collapse
|
23
|
Zhou S, Li Z, Li K, Ye Y, Liang H, Wang N, Liu W, Jiang J, Chiang MYM, Chen A, Xiang X, Lei M. Contact Cooling-Induced ELOVL4 Enhances Skin Wound Healing by Promoting the Inflammation-to-Proliferation Phase Transition. Int J Biol Sci 2025; 21:2067-2082. [PMID: 40083712 PMCID: PMC11900824 DOI: 10.7150/ijbs.107871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/07/2025] [Indexed: 03/16/2025] Open
Abstract
Empirical evidence indicates that the rate of wound healing varies through different seasons, where it is higher in spring and fall but lower in summer and winter, suggesting adequate temperatures may promote wound healing via an unknown mechanism. Here we show that adequate temperature facilitates wound healing by inducing the expression of Elongation of Very Long Chain Fatty Acid Elongase 4 (ELOVL4) that curtails the inflammation phase. Using skin injury and skin organoids models, bulk- and single-cell RNA-sequencing and spatial transcriptomics analysis, and in vivo functional perturbations, we first demonstrate that adjusting skin surface temperature to 20°C by contact cooling markedly increases the rate of wound healing via upregulating ELOVL4 in the injured epidermis. We then reveal docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) as the key products of ELOVL4 that independently control wound healing by dampening the expression of pro-inflammatory cytokines such as tumor necrosis factor α (TNFα). This chain of physiological events enhances wound healing via its timely exit of the inflammatory phase and entry into the proliferation phase of tissue repair. Our findings highlight the skin's adaptability to different temperatures and link the evolutionarily conserved mechanism of long-chain fatty acid synthesis to wound repair while demonstrating the potential application of contact cooling therapy in wound healing.
Collapse
Affiliation(s)
- Siyi Zhou
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Zeming Li
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Ke Li
- Shenzhen Accompany Technology Cooperation, Ltd., Shenzhen 518000, China
| | - Yuanli Ye
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Huan Liang
- Shenzhen Accompany Technology Cooperation, Ltd., Shenzhen 518000, China
| | - Nian'Ou Wang
- Shenzhen Accompany Technology Cooperation, Ltd., Shenzhen 518000, China
| | - Weiwei Liu
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Jingwei Jiang
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Martin Y M Chiang
- Materials Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
| | - Aijun Chen
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Xiao Xiang
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Mingxing Lei
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| |
Collapse
|
24
|
Rasquel-Oliveira FS, Ribeiro JM, Martelossi-Cebinelli G, Costa FB, Nakazato G, Casagrande R, Verri WA. Staphylococcus aureus in Inflammation and Pain: Update on Pathologic Mechanisms. Pathogens 2025; 14:185. [PMID: 40005560 PMCID: PMC11858194 DOI: 10.3390/pathogens14020185] [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: 01/03/2025] [Revised: 01/23/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Staphylococcus aureus (S. aureus) is a Gram-positive bacterium of significant clinical importance, known for its versatility and ability to cause a wide array of infections, such as osteoarticular, pulmonary, cardiovascular, device-related, and hospital-acquired infections. This review describes the most recent evidence of the pathogenic potential of S. aureus, which is commonly part of the human microbiota but can lead to severe infections. The prevalence of pathogenic S. aureus in hospital and community settings contributes to substantial morbidity and mortality, particularly in individuals with compromised immune systems. The immunopathogenesis of S. aureus infections involves intricate interactions with the host immune and non-immune cells, characterized by various virulence factors that facilitate adherence, invasion, and evasion of the host's defenses. This review highlights the complexity of S. aureus infections, ranging from mild to life-threatening conditions, and underscores the growing public health concern posed by multidrug-resistant strains, including methicillin-resistant S. aureus (MRSA). This article aims to provide an updated perspective on S. aureus-related infections, highlighting the main diseases linked to this pathogen, how the different cell types, virulence factors, and signaling molecules are involved in the immunopathogenesis, and the future perspectives to overcome the current challenges to treat the affected individuals.
Collapse
Affiliation(s)
- Fernanda S. Rasquel-Oliveira
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| | - Jhonatan Macedo Ribeiro
- Department of Microbiology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil (G.N.)
| | - Geovana Martelossi-Cebinelli
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| | - Fernanda Barbosa Costa
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| | - Gerson Nakazato
- Department of Microbiology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil (G.N.)
| | - Rubia Casagrande
- Department of Pharmaceutical Sciences, Center of Health Science, Londrina State University, Londrina 86038-440, PR, Brazil
| | - Waldiceu A. Verri
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| |
Collapse
|
25
|
Inclan-Rico JM, Stephenson A, Napuri CM, Rossi HL, Hung LY, Pastore CF, Luo W, Herbert DR. TRPV1+ neurons promote cutaneous immunity against Schistosoma mansoni. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.06.636930. [PMID: 39975236 PMCID: PMC11839022 DOI: 10.1101/2025.02.06.636930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Immunity against skin-invasive pathogens requires mechanisms that rapidly detect, repel or immobilize the infectious agent. While bacteria often cause painful cutaneous reactions, host skin invasion by the human parasitic helminth Schistosoma mansoni often goes unnoticed. This study investigated the role of pain-sensing skin afferents that express the ion channel Transient Receptor Potential Vanilloid 1 (TRPV1) in the detection and initiation of skin immunity against S. mansoni . Data show that mice infected with S. mansoni have reduced behavioral responses to painful stimuli and sensory neurons exposed from infected mice have significantly less calcium influx and neuropeptide release in response to the TRPV1 agonist capsaicin. Using both gain- and loss-of-function approaches, data show that TRPV1+ neurons are critical regulators of S. mansoni survival during migration from the skin into the pulmonary tract. Moreover, TRPV1+ neurons were both necessary and sufficient to promote proliferation and cytokine production from dermal γδ T cells as well as neutrophil and monocyte skin accumulation post-infection. These results suggest a model in which S. mansoni may have evolved to inhibit TRPV1+ neuron activation as a countermeasure that limits IL-17-mediated inflammation, facilitating systemic dissemination and chronic parasitism. One sentence summary The parasitic helminth Schistosoma mansoni averts IL-17-dependent protective immunity by suppressing skin-innervating TRPV1+ neurons.
Collapse
|
26
|
Zhang Y, Wang P, Neng L, Sharma K, Kachelmeier A, Shi X. Monocyte-derived macrophage recruitment mediated by TRPV1 is required for eardrum wound healing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.03.635565. [PMID: 39975094 PMCID: PMC11838451 DOI: 10.1101/2025.02.03.635565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The tympanic membrane (TM), or eardrum, is a thin, sensitive tissue critical for hearing by vibrating and transmitting sound waves to the inner ear. TM perforation and development of otitis media and conductive hearing loss are commonly seen in the clinic. In this study, we demonstrate the role of TRPV1 signaling mediated macrophage recruitment and angiogenesis in TM repair. By creating a wounded TM mouse model with a perforation in the anteroinferior region of the pars tensa - a region in humans often damaged in traumatic injury, we observed a massive accumulation of macrophages in the vicinity of the acutely wounded TM. Using 5-Ethynyl-2'-deoxyuridine pause labeling and a chimeric bone marrow transplant model, we found that most of the recruited macrophages did not originate from local tissue-resident macrophages but rather from blood-circulating monocytes. Parallel to macrophage recruitment, angiogenesis was observed near the wound on day 3 after perforation and further progressed by day 7. The angiogenic process was strongly associated with the recruited macrophages, as macrophage depletion resulted in a notable reduction in angiogenesis. At the transcriptional level, we found that macrophages facilitate angiogenesis through several signaling pathways. Additionally, we identified direct intercellular communication between macrophages and endothelial cells mediated by phosphoprotein 1 signaling. Furthermore, Gene Ontology analysis of bulk RNA sequencing data from TMs revealed that the macrophage recruitment is associated with neuroinflammatory responses. Using a fluorescence reporter mouse driven by TRPV1, we discovered that the TM contains rich sensory nerve fibers expressing TRPV1. A genetic mutation in the Trpv1 gene resulted in a marked decrease in the expression of neuroinflammatory genes, such as Tac1 . This decrease subsequently resulted in reduced macrophage recruitment, impaired angiogenesis, and delayed wound healing. Together, these findings highlight the crucial role of TRPV1 signaling in monocyte migration and macrophage-related angiogenesis, both of which are crucial for facilitating healing of the TM. These results also open new opportunities for clinical interventions. Targeting TRPV1 signaling could enhance TM immunity, improve blood circulation, promote the repair of damaged TM, and ultimately prevent middle ear infections.
Collapse
|
27
|
Natura G, Vazquez E, Richter F, Segond von Banchet G, Ebbinghaus M, Ebersberger A, König C, Maltritz J, Gajda M, Schmidt-Hieber C, Schaible HG. Antinociceptive interactions between excitatory interferon-γ and interleukin-17 in sensory neurons. Brain Behav Immun 2025; 124:55-73. [PMID: 39566665 DOI: 10.1016/j.bbi.2024.11.021] [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: 01/12/2024] [Revised: 11/12/2024] [Accepted: 11/17/2024] [Indexed: 11/22/2024] Open
Abstract
Interferon-γ (IFNγ) and interleukin-17 (IL-17) are master regulators of innate and adaptive immunity. Here we asked whether these cytokines also regulate pain. Both cytokines increased the excitability of isolated small- to medium-sized sensory neurons, suggesting a pronociceptive effect. However, in vivo IL-17 was pronociceptive, whereas IFNγ was antinociceptive. Co-administration of IFNγ and IL-17 in vivo resulted in antinociception. Pre-incubation with IFNγ also eliminated the increase in excitability by interleukin-17A in isolated sensory neurons, demonstrating that the excitatory membrane effects of IFNγ can interfere with the excitatory membrane effects of IL-17, resulting in neuronal inhibition. IFNγ increased TTX-sensitive Na+ currents, while IL-17 increased TTX-resistant Na+ currents. Blocking TTX-sensitive Na+ currents eliminated the inhibition of the IL-17 effect by IFNγ. We propose a novel form of inhibition in sensory neurons that allows the intrinsically excitatory IFNγ to attenuate pro-nociceptive effects of cytokines such as IL-17 through interactions with voltage-gated Na+ currents.
Collapse
Affiliation(s)
- Gabriel Natura
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany
| | - Enrique Vazquez
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany
| | - Frank Richter
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany
| | - Gisela Segond von Banchet
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany
| | - Matthias Ebbinghaus
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany
| | - Andrea Ebersberger
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany
| | - Christian König
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany
| | - Jakob Maltritz
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany
| | - Mieczyslaw Gajda
- Institute of Legal Medicine, Section Pathology, Jena University Hospital, Friedrich-Schiller-University, 07747 Jena, Germany
| | - Christoph Schmidt-Hieber
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany
| | - Hans-Georg Schaible
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany.
| |
Collapse
|
28
|
Ver Heul A. Scratching more than an itch. Science 2025; 387:473-474. [PMID: 39883786 DOI: 10.1126/science.adv1573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
Enhanced antibacterial skin inflammation is an adaptation of the itch-scratch cycle.
Collapse
Affiliation(s)
- Aaron Ver Heul
- Divison of Allergy and Immunology, John T. Milliken Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| |
Collapse
|
29
|
Liu AW, Zhang YR, Chen CS, Edwards TN, Ozyaman S, Ramcke T, McKendrick LM, Weiss ES, Gillis JE, Laughlin CR, Randhawa SK, Phelps CM, Kurihara K, Kang HM, Nguyen SLN, Kim J, Sheahan TD, Ross SE, Meisel M, Sumpter TL, Kaplan DH. Scratching promotes allergic inflammation and host defense via neurogenic mast cell activation. Science 2025; 387:eadn9390. [PMID: 39883751 PMCID: PMC11983162 DOI: 10.1126/science.adn9390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 09/22/2024] [Accepted: 12/03/2024] [Indexed: 02/01/2025]
Abstract
Itch is a dominant symptom in dermatitis, and scratching promotes cutaneous inflammation, thereby worsening disease. However, the mechanisms through which scratching exacerbates inflammation and whether scratching provides benefit to the host are largely unknown. We found that scratching was required for skin inflammation in mouse models dependent on FcεRI-mediated mast cell activation. Scratching-induced inflammation required pain-sensing nociceptors, the neuropeptide substance P, and the mast cell receptor MrgprB2. Scratching also increased cutaneous inflammation and augmented host defense to superficial Staphylococcus aureus infection. Thus, through the activation of nociceptor-driven neuroinflammation, scratching both exacerbated allergic skin disease and provided protection from S. aureus, reconciling the seemingly paradoxical role of scratching as a pathological process and evolutionary adaptation.
Collapse
Affiliation(s)
- Andrew W. Liu
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Youran R. Zhang
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chien-Sin Chen
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tara N. Edwards
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sumeyye Ozyaman
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Torben Ramcke
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lindsay M. McKendrick
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Eric S. Weiss
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jacob E. Gillis
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Colin R. Laughlin
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | - Kazuo Kurihara
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hannah M. Kang
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sydney-Lam N. Nguyen
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jiwon Kim
- Department of Anesthesiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tayler D. Sheahan
- Department of Anesthesiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sarah E. Ross
- Department of Anesthesiology, University of Pittsburgh, Pittsburgh, PA, USA
- Pittsburgh Center for Pain Research, Pittsburgh, PA, USA
| | - Marlies Meisel
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Tina L. Sumpter
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daniel H. Kaplan
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
30
|
Xu P, Shao R, Zhu P, Fei J, He Y. The Role of TRPV1/CGRP Pathway Activated by Prevotella melaninogenica in Pathogenesis of Oral Lichen Planus. Int J Mol Sci 2025; 26:662. [PMID: 39859376 PMCID: PMC11766222 DOI: 10.3390/ijms26020662] [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: 11/13/2024] [Revised: 01/06/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
The distinctive clinicopathologic characteristics of OLP indicated that both microbial dysbiosis and neurogenic inflammation may be jointly involved in its progression, and transient receptor potential vanilloid receptor-1 (TRPV1) may be a crucial element. The purpose of this study was to explore how TRPV1 mediated P. melaninogenica-induced inflammation. Meanwhile, we aimed to unravel how IL-36γ dysregulated the barrier function in oral keratinocytes. Here, the expression of TRPV1, calcitonin gene-related peptide (CGRP), and its receptor receptor activity-modifying protein 1 (RAMP1) in OLP patients were detected. Prevotella melaninogenica (P. melaninogenica) was used to build a mouse model of oral chronic inflammation. Normal human oral keratinocytes (NHOKs) stimulated by P. melaninogenica were used to examine TRPV1 activation and CGRP release. To investigate the effect of exogenous CGRP on Interleukin-36 gamma (IL-36γ) expression in NHOKs and bacterial viability, P. melaninogenica and NHOKs were treated with it, respectively. Recombinant IL-36γ protein was used to probe its regulation of oral epithelial barrier function. TRPV1, CGRP, and RAMP1 were substantially expressed in OLP. P. melaninogenica increased TRPV1 expression in mice and caused the release of CGRP and an increase in pro-inflammatory cytokines via activating TRPV1 in NHOKs. Blockade of TRPV1 suppressed P. melaninogenica-induced inflammation. CGRP boosted the production of IL-36γ released by NHOKs, resulting in lower expression of zonula occludens-1 (ZO-1). Also, CGRP can decrease the viability of P. melaninogenica. Together, these findings provide fresh insight into the vital role performed by P. melaninogenica-induced functional changes in oral epithelial cells and neurons in an intricate OLP inflammatory process.
Collapse
Affiliation(s)
- Pan Xu
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Oral Mucosal Diseases, Shanghai Tongji Stomatological Hospital and Dental School, Tongji University, Shanghai 200072, China; (P.X.); (R.S.); (P.Z.)
| | - Ruru Shao
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Oral Mucosal Diseases, Shanghai Tongji Stomatological Hospital and Dental School, Tongji University, Shanghai 200072, China; (P.X.); (R.S.); (P.Z.)
| | - Pingyi Zhu
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Oral Mucosal Diseases, Shanghai Tongji Stomatological Hospital and Dental School, Tongji University, Shanghai 200072, China; (P.X.); (R.S.); (P.Z.)
| | - Jian Fei
- School of Life Science and Technology, Tongji University, Shanghai 200072, China
| | - Yuan He
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Oral Mucosal Diseases, Shanghai Tongji Stomatological Hospital and Dental School, Tongji University, Shanghai 200072, China; (P.X.); (R.S.); (P.Z.)
| |
Collapse
|
31
|
Liu H, Kong X, Zeng Y, Chen J, Chen Z, Liu L, Ma Q, Liu X, Lu S. From pain to meningitis: bacteria hijack nociceptors to promote meningitis. Front Immunol 2025; 15:1515177. [PMID: 39877376 PMCID: PMC11772308 DOI: 10.3389/fimmu.2024.1515177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 12/30/2024] [Indexed: 01/31/2025] Open
Abstract
Bacterial meningitis is a severe and life-threatening infection of the central nervous system (CNS), primarily caused by Streptococcus pneumoniae and Neisseria meningitidis. This condition carries a high risk of mortality and severe neurological sequelae, such as cognitive impairment and epilepsy. Pain, a central feature of meningitis, results from the activation of nociceptor sensory neurons by inflammatory mediators or bacterial toxins. These nociceptors, abundantly present in the meninges, trigger neuroimmune signaling pathways that influence the host immune response. However, the mechanisms by which bacteria hijack these nociceptors to promote CNS invasion and exacerbate the disease remain poorly understood. This review examines the interactions between bacteria and meningeal nociceptors, focusing on their direct and indirect activation via ion channels, such as transient receptor potential vanilloid-1 (TRPV1) and transient receptor potential ankyrin 1 (TRPA1), or through the release of neuropeptides like calcitonin gene-related peptide (CGRP). These interactions suppress immune defenses by inhibiting macrophage activity and neutrophil recruitment, thus facilitating bacterial survival and invasion of the CNS. Understanding this neuroimmune axis may open potential therapeutic targets for treating bacterial meningitis by enhancing host defenses and mitigating pain. Further research using advanced methodologies is essential to clarify the role of nociceptor-mediated immune modulation in this disease.
Collapse
Affiliation(s)
- Huazhen Liu
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Shenzhen, Guangdong, China
| | - Xingxing Kong
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Shenzhen, Guangdong, China
| | - Yuqin Zeng
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Shenzhen, Guangdong, China
| | - Jinyun Chen
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Shenzhen, Guangdong, China
| | - Zhanpeng Chen
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Shenzhen, Guangdong, China
| | - Lanlan Liu
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Shenzhen, Guangdong, China
| | - Quan Ma
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Shenzhen, Guangdong, China
| | - Xuhui Liu
- Department of Tuberculosis, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Shuihua Lu
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
32
|
Wang YH, Yang X, Liu CC, Wang X, Yu KD. Unraveling the peripheral nervous System's role in tumor: A Double-edged Sword. Cancer Lett 2025; 611:217451. [PMID: 39793755 DOI: 10.1016/j.canlet.2025.217451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 01/01/2025] [Accepted: 01/07/2025] [Indexed: 01/13/2025]
Abstract
The peripheral nervous system (PNS) includes all nerves outside the brain and spinal cord, comprising various cells like neurons and glial cells, such as schwann and satellite cells. The PNS is increasingly recognized for its bidirectional interactions with tumors, exhibiting both pro- and anti-tumor effects. Our review delves into the complex mechanisms underlying these interactions, highlighting recent findings that challenge the conventional understanding of PNS's role in tumorigenesis. We emphasize the contradictory results in the literature and propose novel perspectives on how these discrepancies can be resolved. By focusing on the PNS's influence on tumor initiation, progression, and microenvironment remodeling, we provide a comprehensive analysis that goes beyond the structural description of the PNS. Our review suggests that a deeper comprehension of the PNS-tumor crosstalk is pivotal for developing targeted anticancer strategies. We conclude by emphasizing the need for future research to unravel the intricate dynamics of the PNS in cancer, which may lead to innovative diagnostic tools and therapeutic approaches.
Collapse
Affiliation(s)
- Yan-Hao Wang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center and Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China; Key Laboratory of Breast Cancer in Shanghai, Shanghai, 200032, PR China
| | - Xuan Yang
- Department of General Surgery, Shanxi Provincial People's Hospital, Taiyuan, 030000, PR China
| | - Cui-Cui Liu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center and Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China; Key Laboratory of Breast Cancer in Shanghai, Shanghai, 200032, PR China
| | - Xin Wang
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China
| | - Ke-Da Yu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center and Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China; Key Laboratory of Breast Cancer in Shanghai, Shanghai, 200032, PR China.
| |
Collapse
|
33
|
Palacios A, Kumar A, Caliwag FMC, Becerril-Garcia MA. Neonatal Immunity to Candida: Current Understanding and Contributions of Murine Models. Crit Rev Immunol 2025; 45:63-76. [PMID: 39976518 DOI: 10.1615/critrevimmunol.2024055053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
Neonatal candidiasis poses significant clinical challenges due to its potential for severe morbidity and mortality in vulnerable infants. Due to their underdeveloped immune system, neonates are at a higher risk for infections caused by Candida species. They can vary from mild to severe, including penetrating deep tissues, bloodstream spread, and dissemination to organs. The immune system of newborns is marked by a limited innate immune response, with lower levels of pro-inflammatory cytokines. Adaptive immunity, important for lasting protection, also experiences delayed maturation with weakened Th1 and Th17 responses. These shortcomings result in a higher vulnerability to Candida infections during infancy. Murine models have been crucial in understanding the reasons behind this susceptibility. These models assist in examining how different immune elements, like neutrophils, macrophages, and T cells, and their interactions are involved in Candida infections. Moreover, they offer an understanding of how early-life exposure to Candida affects immune responses and may aid in developing possible therapeutic plans. In this article we review current results from research to provide a thorough summary and critical insights into neonatal immune response to Candida, highlighting the importance of using murine models in this field of study. Understanding these immune dynamics is essential for creating specific treatments and preventive strategies to prevent newborns from Candida infections, ultimately improving neonatal health outcomes.
Collapse
Affiliation(s)
| | - Ajay Kumar
- Isra University Faculty of Medicine and Allied Medical Sciences, Hyderabad, Sindh Pakistan
| | | | - Miguel A Becerril-Garcia
- Universidad Autónoma de Nuevo León School of Medicine, Department of Microbiology, Monterrey, Nuevo León México
| |
Collapse
|
34
|
Crosson T, Bhat S, Wang JC, Salaun C, Fontaine E, Roversi K, Herzog H, Rafei M, Blunck R, Talbot S. Cytokines reprogram airway sensory neurons in asthma. Cell Rep 2024; 43:115045. [PMID: 39661516 DOI: 10.1016/j.celrep.2024.115045] [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: 07/30/2024] [Revised: 10/01/2024] [Accepted: 11/18/2024] [Indexed: 12/13/2024] Open
Abstract
Nociceptor neurons play a crucial role in maintaining the body's homeostasis by detecting and responding to potential environmental dangers. However, this function can be detrimental during allergic reactions, as vagal nociceptors contribute to immune cell infiltration, bronchial hypersensitivity, and mucus imbalance in addition to causing pain and coughing. Despite this, the specific mechanisms by which nociceptors acquire pro-inflammatory characteristics during allergic reactions are not yet fully understood. In this study, we investigate the changes in the molecular profile of airway nociceptor neurons during allergic airway inflammation and identify the signals driving such reprogramming. Using retrograde tracing and lineage reporting, we identify a specific class of inflammatory vagal nociceptor neurons that exclusively innervate the airways. In the ovalbumin mouse model of allergic airway inflammation, these neurons undergo significant reprogramming characterized by the upregulation of the neuropeptide Y (NPY) receptor Npy1r. A screening of cytokines and neurotrophins reveals that interleukin 1β (IL-1β), IL-13, and brain-derived neurotrophic factor (BDNF) drive part of this reprogramming. IL-13 triggers Npy1r overexpression in nociceptors via the JAK/STAT6 pathway. In parallel, NPY is released into the bronchoalveolar fluid of asthmatic mice, which limits the excitability of nociceptor neurons. Single-cell RNA sequencing of lung immune cells reveals that a cell-specific knockout of NPY1R in nociceptor neurons in asthmatic mice altered T cell infiltration. Opposite findings are observed in asthmatic mice in which nociceptor neurons are chemically ablated. In summary, allergic airway inflammation reprograms airway nociceptor neurons to acquire a pro-inflammatory phenotype, while a compensatory mechanism involving NPY1R limits the activity of nociceptor neurons.
Collapse
Affiliation(s)
- Theo Crosson
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
| | - Shreyas Bhat
- Centre Interdisciplinaire sur le Cerveau et l'Apprentissage, Université de Montréal, Montreal, QC, Canada; Département de Physique, Université de Montréal, Montreal, QC, Canada
| | - Jo-Chiao Wang
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
| | - Clara Salaun
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
| | - Eleanne Fontaine
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
| | - Katiane Roversi
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
| | - Herbert Herzog
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Moutih Rafei
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
| | - Rikard Blunck
- Centre Interdisciplinaire sur le Cerveau et l'Apprentissage, Université de Montréal, Montreal, QC, Canada; Département de Physique, Université de Montréal, Montreal, QC, Canada
| | - Sebastien Talbot
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Sweden; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada.
| |
Collapse
|
35
|
Zhang YR, Keshari S, Kurihara K, Liu J, McKendrick LM, Chen CS, Yang Y, Falo LD, Das J, Sumpter TL, Kaplan DH. Agonism of the glutamate receptor GluK2 suppresses dermal mast cell activation and cutaneous inflammation. Sci Transl Med 2024; 16:eadq9133. [PMID: 39661706 DOI: 10.1126/scitranslmed.adq9133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 09/17/2024] [Accepted: 11/12/2024] [Indexed: 12/13/2024]
Abstract
Activation of dermal mast cells through the Mas-related G protein-coupled receptor B2 receptor (MrgprB2 in mice and MrgprX2 in humans) is a key component of numerous inflammatory skin diseases, including dermatitis and rosacea. Sensory neurons actively suppress mast cell activation through the regulated release of glutamate, resulting in reduced expression of Mrgprb2 as well as genes associated with proteins found in mast cell granules. To determine whether exogenous glutamate receptor agonism could suppress mast cell function, we determined that mast cells have relatively selective expression of the glutamate receptor ionotropic, kainate 2 (GluK2). A GluK2-specific agonist, SYM2081, effectively inhibited mast cell degranulation in response to MrgprB2 agonism in both murine mast cells and human skin explants in vitro as well as in vivo after both intradermal and topical administration of SYM2081 to mice. Analyses of transcriptomic datasets from SYM2081-treated mast cells using standard differential expression approaches and an interpretable machine learning technique revealed a previously unrecognized cellular program coordinately regulated by GluK2 agonism. GluK2 agonism suppressed the expression of Mrgprb2 and genes associated with mast cell proliferation. Suppression of mast cell proliferation by SYM2081 exposure was confirmed on the basis of reduced Ki-67 expression and BrdU incorporation in vitro and in vivo. Last, pretreatment with SYM2081 reduced skin inflammation in murine models of dermatitis and rosacea. Thus, agonism of GluK2 represents a promising approach to suppress mast cell activation and may prove beneficial as therapy for inflammatory diseases in which mast cell activation is pathogenic.
Collapse
Affiliation(s)
- Youran R Zhang
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Swapnil Keshari
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Joint CMU-Pitt Ph.D. Program in Computational Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Kazuo Kurihara
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - James Liu
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Lindsay M McKendrick
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Chien-Sin Chen
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Yufan Yang
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA 15261, USA
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Louis D Falo
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Jishnu Das
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Tina L Sumpter
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Daniel H Kaplan
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
36
|
Hasegawa T, Lee CYC, Hotchen AJ, Fleming A, Singh R, Suzuki K, Yuzaki M, Watanabe M, Birch MA, McCaskie AW, Lénárt N, Tóth K, Dénes Á, Liu Z, Ginhoux F, Richoz N, Clatworthy MR. Macrophages and nociceptor neurons form a sentinel unit around fenestrated capillaries to defend the synovium from circulating immune challenge. Nat Immunol 2024; 25:2270-2283. [PMID: 39587345 PMCID: PMC11588661 DOI: 10.1038/s41590-024-02011-8] [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: 01/04/2024] [Accepted: 10/09/2024] [Indexed: 11/27/2024]
Abstract
A wide variety of systemic pathologies, including infectious and autoimmune diseases, are accompanied by joint pain or inflammation, often mediated by circulating immune complexes (ICs). How such stimuli access joints and trigger inflammation is unclear. Whole-mount synovial imaging revealed PV1+ fenestrated capillaries at the periphery of the synovium in the lining-sublining interface. Circulating ICs extravasated from these PV1+ capillaries, and nociceptor neurons and three distinct macrophage subsets formed a sentinel unit around them. Macrophages showed subset-specific responses to systemic IC challenge; LYVE1+CX3CR1+ macrophages orchestrated neutrophil recruitment and activated calcitonin gene-related peptide+ (CGRP+) nociceptor neurons via interleukin-1β. In contrast, major histocompatibility complex class II+CD11c+ (MHCII+CD11c+) and MHCII+CD11c- interstitial macrophages formed tight clusters around PV1+ capillaries in response to systemic immune stimuli, a feature enhanced by nociceptor-derived CGRP. Altogether, we identify the anatomical location of synovial PV1+ capillaries and subset-specific macrophage-nociceptor cross-talk that forms a blood-joint barrier protecting the synovium from circulating immune challenges.
Collapse
Affiliation(s)
- Tetsuo Hasegawa
- Molecular Immunity Unit, University of Cambridge, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, Cambridge, UK.
- Cambridge Institute for Therapeutic Immunology and Infectious Diseases, University of Cambridge, Cambridge, UK.
| | - Colin Y C Lee
- Molecular Immunity Unit, University of Cambridge, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
- Cellular Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Andrew J Hotchen
- Molecular Immunity Unit, University of Cambridge, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
- Division of Trauma and Orthopaedic Surgery, Department of Surgery, University of Cambridge, Cambridge, UK
| | - Aaron Fleming
- Molecular Immunity Unit, University of Cambridge, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Rahul Singh
- Molecular Immunity Unit, University of Cambridge, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Kunimichi Suzuki
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Michisuke Yuzaki
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Mark A Birch
- Division of Trauma and Orthopaedic Surgery, Department of Surgery, University of Cambridge, Cambridge, UK
| | - Andrew W McCaskie
- Division of Trauma and Orthopaedic Surgery, Department of Surgery, University of Cambridge, Cambridge, UK
| | - Nikolett Lénárt
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Krisztina Tóth
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Ádám Dénes
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Zhaoyuan Liu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | - Nathan Richoz
- Molecular Immunity Unit, University of Cambridge, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Menna R Clatworthy
- Molecular Immunity Unit, University of Cambridge, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, Cambridge, UK.
- Cellular Genetics, Wellcome Sanger Institute, Hinxton, UK.
| |
Collapse
|
37
|
Fang X, Ding H, Chen Y, Wang Q, Yuan X, Zhang C, Huang J, Huang J, Lv J, Hu H, Huang C, Hu X, Lin Y, Zhang N, Zhou W, Huang Y, Li W, Niu S, Wu Z, Lin J, Yang B, Yuan T, Zhang W. Wireless Optogenetic Targeting Nociceptors Helps Host Cells Win the Competitive Colonization in Implant-Associated Infections. SMALL METHODS 2024; 8:e2400216. [PMID: 39087367 DOI: 10.1002/smtd.202400216] [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: 03/01/2024] [Revised: 06/20/2024] [Indexed: 08/02/2024]
Abstract
The role of nociceptive nerves in modulating immune responses to harmful stimuli via pain or itch induction remains controversial. Compared to conventional surgery, various implant surgeries are more prone to infections even with low bacterial loads. In this study, an optogenetic technique is introduced for selectively activating peripheral nociceptive nerves using a fully implantable, wirelessly rechargeable optogenetic device. By targeting nociceptors in the limbs of awake, freely moving mice, it is found that activation induces anticipatory immunity in the innervated territory and enhances the adhesion of various host cells to the implant surface. This effect mediates acute immune cell-mediated killing of Staphylococcus aureus on implants and enables the host to win "implant surface competition" against Staphylococcus aureus. This finding provides new strategies for preventing and treating implant-associated infections.
Collapse
Affiliation(s)
- Xinyu Fang
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
| | - Haiqi Ding
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
| | - Yang Chen
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
| | - Qijin Wang
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
- Department of Orthopedics, Affiliated Mindong Hospital of Fujian Medical University, Fu'an, 355000, China
| | - Xuhui Yuan
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
| | - Chaofan Zhang
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
| | - Jiagu Huang
- Department of Orthopedic Surgery, Ningde municipal Hospital, Ningde, 352000, China
| | - Jiexin Huang
- Department of Orthopedic Surgery, Nanping First Hospital, Nanping, 353000, China
| | - Jianhua Lv
- Department of Orthopedic Surgery, Affiliated Hospital of Putian University, Putian, 351100, China
| | - Hongxin Hu
- Department of Orthopedic Surgery, Affiliated Hospital of Putian University, Putian, 351100, China
| | - Changyu Huang
- Department of Orthopedic Surgery, Quanzhou Orthopedic-traumatological Hospital, Quanzhou, 362000, China
| | - Xueni Hu
- Department of Laboratory Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, China
- Department of Laboratory Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
| | - Yiming Lin
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
| | - Nanxin Zhang
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
| | - Wei Zhou
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
| | - Ying Huang
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
| | - Wenbo Li
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
| | - Susheng Niu
- Key Laboratory of Orthopedics & Traumatology of Traditional Chinese Medicine and Rehabilitation Ministry of Education, Fujian university of Traditional Chinese Medicine, Fuzhou, 350000, China
| | - Zhaoyang Wu
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
| | - Jianhua Lin
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
| | - Bin Yang
- Department of Laboratory Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, China
- Department of Laboratory Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
| | - Tifei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai, 200000, China
| | - Wenming Zhang
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
| |
Collapse
|
38
|
Ji X, Zhou J, Zhou Z, Liu Z, Yan L, Li Y, Guo H, Su W, Wang H, Ni D. Recovering skin-nerve interaction by nanoscale metal-organic framework for diabetic ulcers healing. Bioact Mater 2024; 42:112-123. [PMID: 39280583 PMCID: PMC11402068 DOI: 10.1016/j.bioactmat.2024.08.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/05/2024] [Accepted: 08/23/2024] [Indexed: 09/18/2024] Open
Abstract
Skin-nerve interaction plays an important role in promoting wound healing. However, in diabetic ulcers (DUs), the diabetic periphery neuropathy and excessive levels of reactive oxygen species (ROS) block skin-nerve interaction and further impede the DUs healing. Herein, we developed a nanoscale metal-organic framework loaded with nerve growth factor (NGF/Ce-UiO-66, denoted NGF/CU) for the treatment of DUs. The Ce-UiO-66 (CU) was applied as an antioxidant to scavenge ROS and reduce the inflammatory response while the NGF aided in the recovery of cutaneous nerves to further promote DUs healing. Both in vitro and in vivo experiments revealed the effective ability of NGF/CU for DUs healing. Subsequent RNA sequencing analysis revealed the mechanism that NGF/CU can improve wound healing by inhibiting the NF-κB signaling pathway and recovering the neuroendocrine system of the skin. This strategy of nerve regulation will provide more ideas for the treatment of DUs and other organ injuries.
Collapse
Affiliation(s)
- Xiuru Ji
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin 2nd Rd, Shanghai, 200025, PR China
| | - Jingwei Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639, Zhizaoju Rd, Shanghai, 200011, PR China
| | - Zengding Zhou
- Department of Burn and Plastic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin 2nd Rd, Shanghai, 200025, PR China
| | - Zeyang Liu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin 2nd Rd, Shanghai, 200025, PR China
| | - Li Yan
- Department of Dermatology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin 2nd Rd, Shanghai, 200025, PR China
| | - Yuhan Li
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin 2nd Rd, Shanghai, 200025, PR China
| | - Haiyan Guo
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin 2nd Rd, Shanghai, 200025, PR China
| | - Weijie Su
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639, Zhizaoju Rd, Shanghai, 200011, PR China
| | - Han Wang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin 2nd Rd, Shanghai, 200025, PR China
| | - Dalong Ni
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin 2nd Rd, Shanghai, 200025, PR China
| |
Collapse
|
39
|
Wang Z, Song K, Kim BS, Manion J. Sensory neuroimmune interactions at the barrier. Mucosal Immunol 2024; 17:1151-1160. [PMID: 39374664 DOI: 10.1016/j.mucimm.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/24/2024] [Accepted: 10/01/2024] [Indexed: 10/09/2024]
Abstract
Epithelial barriers such as the skin, lung, and gut, in addition to having unique physiologic functions, are designed to preserve tissue homeostasis upon challenge with a variety of allergens, irritants, or pathogens. Both the innate and adaptive immune systems play a critical role in responding to epithelial cues triggered by environmental stimuli. However, the mechanisms by which organs sense and coordinate complex epithelial, stromal, and immune responses have remained a mystery. Our increasing understanding of the anatomic and functional characteristics of the sensory nervous system is greatly advancing a new field of peripheral neuroimmunology and subsequently changing our understanding of mucosal immunology. Herein, we detail how sensory biology is informing mucosal neuroimmunology, even beyond neuroimmune interactions seen within the central and autonomic nervous systems.
Collapse
Affiliation(s)
- Zhen Wang
- Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Allen Discovery Center for Neuroimmune Interactions, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
| | - Keaton Song
- Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Allen Discovery Center for Neuroimmune Interactions, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Brian S Kim
- Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Allen Discovery Center for Neuroimmune Interactions, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA.
| | - John Manion
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; Department of Urology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Surgery, Harvard Medical School, Boston, MA 02115, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
40
|
Song K, Kim BS. The peripheral neuroimmune system. J Leukoc Biol 2024; 116:1291-1300. [PMID: 39422243 PMCID: PMC11599120 DOI: 10.1093/jleuko/qiae230] [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: 07/23/2024] [Revised: 09/18/2024] [Accepted: 10/16/2024] [Indexed: 10/19/2024] Open
Abstract
Historically, the nervous and immune systems were studied as separate entities. The nervous system relays signals between the body and the brain by processing sensory inputs and executing motor outputs, whereas the immune system provides protection against injury and infection through inflammation. However, recent developments have demonstrated that these systems mount tightly integrated responses. In particular, the peripheral nervous system acts in concert with the immune system to control reflexes that maintain and restore homeostasis. Notwithstanding their homeostatic mechanisms, dysregulation of these neuroimmune interactions may underlie various pathological conditions. Understanding how these two distinct systems communicate is an emerging field of peripheral neuroimmunology that promises to reveal new insights into tissue physiology and identify novel targets to treat disease.
Collapse
Affiliation(s)
- Keaton Song
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, New York, NY 10029, USA
- Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, 787 11th Ave, New York, NY 10019, USA
- Allen Discovery Center for Neuroimmune Interactions, Icahn School of Medicine at Mount Sinai, 787 11th Ave, New York, NY 10019, USA
| | - Brian S Kim
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, New York, NY 10029, USA
- Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, 787 11th Ave, New York, NY 10019, USA
- Allen Discovery Center for Neuroimmune Interactions, Icahn School of Medicine at Mount Sinai, 787 11th Ave, New York, NY 10019, USA
| |
Collapse
|
41
|
Peng J, Khoury MJ, Hsu YC. Stem cells feel the pain. Dev Cell 2024; 59:2805-2806. [PMID: 39500285 DOI: 10.1016/j.devcel.2024.09.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 05/16/2025]
Abstract
Pain is a sensation that signals the presence of inflammation or injury. In this issue of Developmental Cell, Ben-Shaanan et al.1 show that beyond its sensory function, pain can activate hair follicle stem cells (HFSCs) by controlling their niche.
Collapse
Affiliation(s)
- Jingyu Peng
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Mark J Khoury
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Ya-Chieh Hsu
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
42
|
Ben-Shaanan TL, Knöpper K, Duan L, Liu R, Taglinao H, Xu Y, An J, Plikus MV, Cyster JG. Dermal TRPV1 innervations engage a macrophage- and fibroblast-containing pathway to activate hair growth in mice. Dev Cell 2024; 59:2818-2833.e7. [PMID: 38851191 PMCID: PMC11537826 DOI: 10.1016/j.devcel.2024.05.019] [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: 10/03/2023] [Revised: 02/25/2024] [Accepted: 05/15/2024] [Indexed: 06/10/2024]
Abstract
Pain, detected by nociceptors, is an integral part of injury, yet whether and how it can impact tissue physiology and recovery remain understudied. Here, we applied chemogenetics in mice to locally activate dermal TRPV1 innervations in naive skin and found that it triggered new regenerative cycling by dormant hair follicles (HFs). This was preceded by rapid apoptosis of dermal macrophages, mediated by the neuropeptide calcitonin gene-related peptide (CGRP). TRPV1 activation also triggered a macrophage-dependent induction of osteopontin (Spp1)-expressing dermal fibroblasts. The neuropeptide CGRP and the extracellular matrix protein Spp1 were required for the nociceptor-triggered hair growth. Finally, we showed that epidermal abrasion injury induced Spp1-expressing dermal fibroblasts and hair growth via a TRPV1 neuron and CGRP-dependent mechanism. Collectively, these data demonstrated a role for TRPV1 nociceptors in orchestrating a macrophage and fibroblast-supported mechanism to promote hair growth and enabling the efficient restoration of this mechano- and thermo-protective barrier after wounding.
Collapse
Affiliation(s)
- Tamar L Ben-Shaanan
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Konrad Knöpper
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Lihui Duan
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ruiqi Liu
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Hanna Taglinao
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ying Xu
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jinping An
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Jason G Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
43
|
Inclan-Rico JM, Napuri CM, Lin C, Hung LY, Ferguson AA, Liu X, Wu Q, Pastore CF, Stephenson A, Femoe UM, Musaigwa F, Rossi HL, Freedman BD, Reed DR, Macháček T, Horák P, Abdus-Saboor I, Luo W, Herbert DR. MrgprA3 neurons drive cutaneous immunity against helminths through selective control of myeloid-derived IL-33. Nat Immunol 2024; 25:2068-2084. [PMID: 39354200 PMCID: PMC12032830 DOI: 10.1038/s41590-024-01982-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 09/11/2024] [Indexed: 10/03/2024]
Abstract
Skin uses interdependent cellular networks for barrier integrity and host immunity, but most underlying mechanisms remain obscure. Herein, we demonstrate that the human parasitic helminth Schistosoma mansoni inhibited pruritus evoked by itch-sensing afferents bearing the Mas-related G-protein-coupled receptor A3 (MrgprA3) in mice. MrgprA3 neurons controlled interleukin (IL)-17+ γδ T cell expansion, epidermal hyperplasia and host resistance against S. mansoni through shaping cytokine expression in cutaneous antigen-presenting cells. MrgprA3 neuron activation downregulated IL-33 but induced IL-1β and tumor necrosis factor in macrophages and type 2 conventional dendritic cells partially through the neuropeptide calcitonin gene-related peptide. Macrophages exposed to MrgprA3-derived secretions or bearing cell-intrinsic IL-33 deletion showed increased chromatin accessibility at multiple inflammatory cytokine loci, promoting IL-17/IL-23-dependent changes to the epidermis and anti-helminth resistance. This study reveals a previously unrecognized intercellular communication mechanism wherein itch-inducing MrgprA3 neurons initiate host immunity against skin-invasive parasites by directing cytokine expression patterns in myeloid antigen-presenting cell subsets.
Collapse
Affiliation(s)
- Juan M Inclan-Rico
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Camila M Napuri
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cailu Lin
- Monell Chemical Senses Center, Philadelphia, PA, USA
| | - Li-Yin Hung
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Annabel A Ferguson
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xiaohong Liu
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Qinxue Wu
- Department of Neuroscience, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher F Pastore
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Adriana Stephenson
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ulrich M Femoe
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Fungai Musaigwa
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Heather L Rossi
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bruce D Freedman
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Tomáš Macháček
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Petr Horák
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Ishmail Abdus-Saboor
- Department of Biological Sciences, Zuckerman Mind, Brain, Behavior Institute, Columbia University, New York, NY, USA
| | - Wenqin Luo
- Department of Neuroscience, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Regenerative Medicine, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - De'Broski R Herbert
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
44
|
You X, Niu L, Song X, Fu J, Miao Y, Diao F, Wu C, Zhuang P, Zhang Y. Linking severe traumatic brain injury to pulmonary Infections: Translocation of intestinal bacteria mediated by nociceptor neurons. Brain Behav Immun 2024; 122:604-616. [PMID: 39187048 DOI: 10.1016/j.bbi.2024.08.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 08/12/2024] [Accepted: 08/22/2024] [Indexed: 08/28/2024] Open
Abstract
The prevalence of bacterial infections significantly increases among patients with severe traumatic brain injury (STBI), leading to a notable rise in mortality rates. While immune dysfunctions are linked to the incidence of pneumonia, our observations indicate that endogenous pathogens manifest in the lungs post-STBI due to the migration of gut commensal bacteria. This translocation involves gut-innervating nociceptor sensory neurons, which are crucial for host defense. Following STBI, the expression of transient receptor potential vanilloid 1 (TRPV1) in dorsal root ganglion (DRG) neurons significantly decreases, despite an initial brief increase. The timing of TRPV1 defects coincides with the occurrence of pulmonary infections post-STBI. This alteration in TRPV1+ neurons diminishes their ability to signal bacterial injuries, weakens defense mechanisms against intestinal bacteria, and increases susceptibility to pulmonary infections via bacterial translocation. Experimental evidence demonstrates that pulmonary infections can be successfully replicated through the chemical ablation and gene interference of TRPV1+ nociceptors, and that these infections can be mitigated by TRPV1 activation, thereby confirming the crucial role of nociceptor neurons in controlling intestinal bacterial migration. Furthermore, TRPV1+ nociceptors regulate the immune response of microfold cells by releasing calcitonin gene-related peptide (CGRP), thereby influencing the translocation of gut bacteria to the lungs. Our study elucidates how changes in nociceptive neurons post-STBI impact intestinal pathogen defense. This new understanding of endogenous risk factors within STBI pathology offers novel insights for preventing and treating pulmonary infections.
Collapse
Affiliation(s)
- Xinyu You
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300381, China
| | - Lin Niu
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xuejiao Song
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jiafeng Fu
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yulu Miao
- Department of Pharmacology, Basic Medical Sciences Center, School of Basic Medical Science, Shanxi Medical University, Taiyuan 030001, China
| | - Fengyin Diao
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Chongming Wu
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Pengwei Zhuang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300381, China.
| | - Yanjun Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300381, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China.
| |
Collapse
|
45
|
Alexander SN, Green AR, Debner EK, Ramos Freitas LE, Abdelhadi HMK, Szabo-Pardi TA, Burton MD. The influence of sex on neuroimmune communication, pain, and physiology. Biol Sex Differ 2024; 15:82. [PMID: 39439003 PMCID: PMC11494817 DOI: 10.1186/s13293-024-00660-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 10/02/2024] [Indexed: 10/25/2024] Open
Abstract
With the National Institutes of Health's mandate to consider sex as a biological variable (SABV), there has been a significant increase of studies utilizing both sexes. Historically, we have known that biological sex and hormones influence immunological processes and now studies focusing on interactions between the immune, endocrine, and nervous systems are revealing sex differences that influence pain behavior and various molecular and biochemical processes. Neuroendocrine-immune interactions represent a key integrative discipline that will reveal critical processes in each field as it pertains to novel mechanisms in sex differences and necessary therapeutics. Here we appraise preclinical and clinical literature to discuss these interactions and key pathways that drive cell- and sex-specific differences in immunity, pain, and physiology.
Collapse
Affiliation(s)
- Shevon N Alexander
- Neuroimmunology and Behavior Laboratory, Department of Neuroscience, School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, 800 W. Campbell Road, BSB 10.537, Richardson, TX, 75080, USA
| | - Audrey R Green
- Neuroimmunology and Behavior Laboratory, Department of Neuroscience, School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, 800 W. Campbell Road, BSB 10.537, Richardson, TX, 75080, USA
| | - Emily K Debner
- Neuroimmunology and Behavior Laboratory, Department of Neuroscience, School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, 800 W. Campbell Road, BSB 10.537, Richardson, TX, 75080, USA
| | - Lindsey E Ramos Freitas
- Neuroimmunology and Behavior Laboratory, Department of Neuroscience, School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, 800 W. Campbell Road, BSB 10.537, Richardson, TX, 75080, USA
| | - Hanna M K Abdelhadi
- Neuroimmunology and Behavior Laboratory, Department of Neuroscience, School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, 800 W. Campbell Road, BSB 10.537, Richardson, TX, 75080, USA
| | - Thomas A Szabo-Pardi
- Neuroimmunology and Behavior Laboratory, Department of Neuroscience, School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, 800 W. Campbell Road, BSB 10.537, Richardson, TX, 75080, USA
| | - Michael D Burton
- Neuroimmunology and Behavior Laboratory, Department of Neuroscience, School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, 800 W. Campbell Road, BSB 10.537, Richardson, TX, 75080, USA.
| |
Collapse
|
46
|
Subramanian M, Thaiss CA. Interoceptive inference and prediction in food-related disorders. Genes Dev 2024; 38:808-813. [PMID: 39362780 PMCID: PMC11535158 DOI: 10.1101/gad.352301.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2024]
Abstract
The brain's capacity to predict and anticipate changes in internal and external environments is fundamental to initiating efficient adaptive responses, behaviors, and reflexes that minimize disruptions to physiology. In the context of feeding control, the brain predicts and anticipates responses to the consumption of dietary substances, thus driving adaptive behaviors in the form of food choices, physiological preparation for meals, and engagement of defensive mechanisms. Here, we provide an integrative perspective on the multisensory computation between exteroceptive and interoceptive cues that guides feeding strategy and may result in food-related disorders.
Collapse
Affiliation(s)
- Madhav Subramanian
- Microbiology Department, Institute for Immunology, Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Christoph A Thaiss
- Microbiology Department, Institute for Immunology, Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
47
|
Zorrilla E, Della Pietra A, Russo AF. Interplay between cannabinoids and the neuroimmune system in migraine. J Headache Pain 2024; 25:178. [PMID: 39407099 PMCID: PMC11481476 DOI: 10.1186/s10194-024-01883-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024] Open
Abstract
Migraine is a common and complex neurological disorder that has a high impact on quality of life. Recent advances with drugs that target the neuropeptide calcitonin gene-related peptide (CGRP) have helped, but treatment options remain insufficient. CGRP is released from trigeminal sensory fibers and contributes to peripheral sensitization, perhaps in part due to actions on immune cells in the trigeminovascular system. In this review, we will discuss the potential of cannabinoid targeting of immune cells as an innovative therapeutic target for migraine treatment. We will cover endogenous endocannabinoids, plant-derived phytocannabinoids and synthetically derived cannabinoids. The focus will be on six types of immune cells known to express multiple cannabinoid receptors: macrophages, monocytes, mast cells, dendritic cells, B cells, and T cells. These cells also contain receptors for CGRP and as such, cannabinoids might potentially modulate the efficacy of current CGRP-targeting drugs. Unfortunately, to date most studies on cannabinoids and immune cells have relied on cell cultures and only a single preclinical study has tested cannabinoid actions on immune cells in a migraine model. Encouragingly, in that study a synthetically created stable chiral analog of an endocannabinoid reduced meningeal mast cell degranulation. Likewise, clinical trials evaluating the safety and efficacy of cannabinoid-based therapies for migraine patients have been limited but are encouraging. Thus, the field is at its infancy and there are significant gaps in our understanding of the impact of cannabinoids on immune cells in migraine. Future research exploring the interactions between cannabinoids and immune cells could lead to more targeted and effective migraine treatments.
Collapse
Affiliation(s)
- Erik Zorrilla
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, 52242, USA
| | - Adriana Della Pietra
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, 52242, USA
| | - Andrew F Russo
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, 52242, USA.
- Department of Neurology, University of Iowa, Iowa City, IA, 52242, USA.
- Veterans Affairs Healthcare System, Iowa City, IA, 52246, USA.
| |
Collapse
|
48
|
Murakami D, Kono M, Sakatani H, Iyo T, Hijiya M, Shiga T, Kinoshita T, Sumioka T, Okada Y, Saika S, Koizumi Y, Hotomi M. Inhibition of transient receptor potential vanilloid 1 reduces shedding and transmission during Streptococcus pneumoniae co-infection with influenza. Infect Immun 2024; 92:e0014624. [PMID: 39109830 PMCID: PMC11475660 DOI: 10.1128/iai.00146-24] [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: 04/02/2024] [Accepted: 07/04/2024] [Indexed: 10/16/2024] Open
Abstract
Transmission is the first step for a microorganism to establish colonization in the respiratory tract and subsequent development of infectious disease. Streptococcus pneumoniae is a leading pathogen that colonizes the mucosal surfaces of the human upper respiratory tract and causes subsequent transmission and invasive infections especially in co-infection with influenza A virus. Host factors contributing to respiratory contagion are poorly understood. Transient receptor potential vanilloid (TRPV) channels have various roles in response to microoorganism. Inhibition of TRPV exacerbates invasive infection by Streptococcus pneumoniae, but it is unclear how TRPV channels influence pneumococcal transmission. Here, we describe the effect of inhibition of TRPV1 on pneumococcal transmission. We adopted a TRPV1-deficient infant mouse model of pneumococcal transmission during co-infection with influenza A virus. We also analyzed the expression of nasal mucin or pro-inflammatory cytokines. TRPV1 deficiency attenuated pneumococcal transmission and shedding during co-infection with influenza A virus. TRPV1 deficiency suppressed the expression of nasal mucin. In addition, there were increases in the expression of tumor necrosis factor-α and type I interferon, followed by the suppressed replication of influenza A virus in TRPV1-deficient mice. Inhibition of TRPV1 was shown to attenuate pneumococcal transmission by reducing shedding through the suppression of nasal mucin during co-infection with influenza A virus. Inhibition of TRPV1 suppressed nasal mucin by modulation of pro-inflammatory responses and regulation of replication of influenza A virus. TRPV1 could be a new target in preventive strategy against pneumococcal transmission.
Collapse
Affiliation(s)
- Daichi Murakami
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Masamitsu Kono
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Hideki Sakatani
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Takuro Iyo
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Masayoshi Hijiya
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Tatsuya Shiga
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Tetsuya Kinoshita
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Takayoshi Sumioka
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| | - Yuka Okada
- Department of Ophthalmology, Wakayama Medical University Kihoku Hospital, Wakayama, Japan
| | - Shizuya Saika
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| | - Yusuke Koizumi
- Department of Clinical Infectious Diseases, Infection Control and Prevention, Wakayama Medical University, Wakayama, Japan
| | - Muneki Hotomi
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
49
|
Hanč P, Messou MA, Ajit J, von Andrian UH. Setting the tone: nociceptors as conductors of immune responses. Trends Immunol 2024; 45:783-798. [PMID: 39307581 PMCID: PMC11493364 DOI: 10.1016/j.it.2024.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 10/13/2024]
Abstract
Nociceptors have emerged as master regulators of immune responses in both homeostatic and pathologic settings; however, their seemingly contradictory effects on the functions of different immune cell subsets have been a source of confusion. Nevertheless, work by many groups in recent years has begun to identify patterns of the modalities and consequences of nociceptor-immune system communication. Here, we review recent findings of how nociceptors affect immunity and propose an integrated concept whereby nociceptors are neither inherently pro- nor anti-inflammatory. Rather, we propose that nociceptors have the role of a rheostat that, in a context-dependent manner, favors tissue homeostasis and fine-tunes immunity by preventing excessive histotoxic inflammation, promoting tissue repair, and potentiating anticipatory and adaptive immune responses.
Collapse
Affiliation(s)
- Pavel Hanč
- Department of Immunology, Harvard Medical School, Boston, MA, USA; The Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Marie-Angèle Messou
- Department of Immunology, Harvard Medical School, Boston, MA, USA; The Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Jainu Ajit
- Department of Immunology, Harvard Medical School, Boston, MA, USA; The Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Ulrich H von Andrian
- Department of Immunology, Harvard Medical School, Boston, MA, USA; The Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA.
| |
Collapse
|
50
|
Chen J, Sun W, Zhu Y, Zhao F, Deng S, Tian M, Wang Y, Gong Y. TRPV1: The key bridge in neuroimmune interactions. JOURNAL OF INTENSIVE MEDICINE 2024; 4:442-452. [PMID: 39310069 PMCID: PMC11411435 DOI: 10.1016/j.jointm.2024.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/07/2024] [Accepted: 01/23/2024] [Indexed: 09/25/2024]
Abstract
The nervous and immune systems are crucial in fighting infections and inflammation and in maintaining immune homeostasis. The immune and nervous systems are independent, yet tightly integrated and coordinated organizations. Numerous molecules and receptors play key roles in enabling communication between the two systems. Transient receptor potential vanilloid subfamily member 1 (TRPV1) is a non-selective cation channel, recently shown to be widely expressed in the neuroimmune axis and implicated in neuropathic pain, autoimmune disorders, and immune cell function. TRPV1 is a key bridge in neuroimmune interactions, allowing for smooth and convenient communication between the two systems. Here, we discuss the coordinated cross-talking between the immune and nervous systems and the functional role and the functioning manner of the TRPV1 involved. We suggest that TRPV1 provides new insights into the collaborative relationship between the nervous and immune systems, highlighting exciting opportunities for advanced therapeutic approaches to treating neurogenic inflammation and immune-mediated diseases.
Collapse
Affiliation(s)
- Jianwei Chen
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Wenqian Sun
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Youjia Zhu
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Feng Zhao
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Shuixiang Deng
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Mi Tian
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yao Wang
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Ye Gong
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| |
Collapse
|