1
|
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] [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
|
2
|
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
|
3
|
Shibao T, Hase H, Mizokami K, Usui A, Kitae K, Ueda Y, Jingushi K, Tsujikawa K. CGRPβ suppresses the pathogenesis of ulcerative colitis via the immunoproteasome. Sci Rep 2025; 15:7224. [PMID: 40021701 PMCID: PMC11871240 DOI: 10.1038/s41598-025-91933-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 02/24/2025] [Indexed: 03/03/2025] Open
Abstract
Various factors have been implicated in the pathogenesis of ulcerative colitis (UC), with immune system failure being the most important one. Calcitonin gene-related peptide (CGRP), a neuropeptide with two isoforms, CGRPα and CGRPβ, has been reported to regulate the immune system. In this study, we investigated the role of CGRP isoforms in UC pathogenesis. We induced UC-like symptoms in CGRPα and CGRPβ knockout (KO) mice using dextran sulphate sodium. Compared to wild-type and CGRPα KO mice, CGRPβ-deficient mice exhibited severe symptoms with increased blood in the stool and diarrhoea. Proteome analysis revealed significant up-regulation of immune-related proteins and immunoproteasome components in CGRPβ-deficient mice, suggesting that an enhanced immune response contributes to the severity of this disease. Treatment with ONX-0914, an immunoproteasome inhibitor, markedly improved these symptoms, highlighting the role of the immunoproteasome in exacerbating UC. This study provides the first evidence that CGRPβ protects against UC by modulating immune responses, particularly those mediated by the immunoproteasome. Our findings suggest that functional differences in CGRP isoforms may influence the severity and management of UC. This insight into the neuro-immune mechanism of UC opens avenues for novel therapies that address both the neural and immune aspects of this disease.
Collapse
Affiliation(s)
- Tatsuya Shibao
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, 565-0871, Osaka, Japan
| | - Hiroaki Hase
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, 565-0871, Osaka, Japan.
| | - Kodai Mizokami
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, 565-0871, Osaka, Japan
| | - Atsushi Usui
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, 565-0871, Osaka, Japan
| | - Kaori Kitae
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, 565-0871, Osaka, Japan
| | - Yuko Ueda
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, 565-0871, Osaka, Japan
| | - Kentaro Jingushi
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, 565-0871, Osaka, Japan
| | - Kazutake Tsujikawa
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, 565-0871, Osaka, Japan
| |
Collapse
|
4
|
Yang YL, Yao N, Ge SQ, Song B, Xu H, Li Z, Li XF, Li J. N-(4-methoxyphenyl) quinoline-8-sulfonamide reduces the inflammatory response of fibroblast-like synoviocytes by targeting receptor (calcitonin) activity modifying protein 1 in rheumatoid arthritis. Eur J Pharmacol 2024; 984:177064. [PMID: 39427859 DOI: 10.1016/j.ejphar.2024.177064] [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: 07/12/2024] [Revised: 10/06/2024] [Accepted: 10/17/2024] [Indexed: 10/22/2024]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is characterized by chronic inflammation of the synovium of joints. Fibroblast-like synoviocytes (FLS) play an important role in RA pathogenesis. We aimed to investigate the effect of N-(4-methoxyphenyl) quinoline-8-sulfonamide (QS-3g) on the inflammatory response of FLS and explore the potential underlying mechanisms. METHODS We screened and found that QS-3g exhibits the best anti-inflammatory response against FLS using the CCK8 assay. To investigate the therapeutic effects of QS-3g on K/BxN STA mice, we used H&E staining, immunofluorescence, immunohistochemistry, micro-CT, and other techniques. Additional investigations, including RNA-seq, molecular docking, and CETSA, revealed that QS-3g binds to RAMP1. RESULTS Among a series of 8-quinoline sulfonyl amide derivatives, QS-3g reduced the inflammatory response in TNF-α stimulated FLS, such as the release of interleukin (IL)-1β and IL-6. H&E staining and micro-CT showed that QS-3g inhibited synovial hypertrophy, inflammatory cell infiltration, and bone destruction. RNA-seq and CETSA analyses revealed the targeted inhibition of RAMP1 by QS-3g. Inhibition of RAMP1 expression could reduce IL-6 and IL-1β levels. Compared with RAMP1-si, combined administration of QS-3g and RAMP1-si reduced the TNF-α-induced inflammation in TNF-α stimulated FLS without statistically significant differences. Finally, the results of in vitro experiments showed that QS-3g could restore the balance of Gαi/Gαs by inhibiting Gαi and activating Gαs and up-regulate the expression of cAMP protein, thus inhibiting the RAMP1-mediated inflammatory response in FLS. CONCLUSION QS-3g could inhibit RAMP1 activity and mediate the Gαs/Gαi-cAMP pathway to reduce FLS inflammatory response. Therefore, QS-3g may serve as a novel anti-inflammatory compound for treating RA.
Collapse
Affiliation(s)
- Ying-Li Yang
- Inflammation and Immune Mediated Disease Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Department of Pharmacy, Obstetrics and Gynecology Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Ning Yao
- Inflammation and Immune Mediated Disease Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Shang-Qing Ge
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - Biao Song
- Department of Pharmacy, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Han Xu
- Inflammation and Immune Mediated Disease Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Department of Hematopathology, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Zeng Li
- Inflammation and Immune Mediated Disease Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Xiao-Feng Li
- Inflammation and Immune Mediated Disease Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China.
| | - Jun Li
- Inflammation and Immune Mediated Disease Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
5
|
Yamashita A, Ito Y, Osada M, Matsuda H, Hosono K, Tsujikawa K, Okamoto H, Amano H. RAMP1 Signaling Mitigates Acute Lung Injury by Distinctively Regulating Alveolar and Monocyte-Derived Macrophages. Int J Mol Sci 2024; 25:10107. [PMID: 39337592 PMCID: PMC11432488 DOI: 10.3390/ijms251810107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a life-threatening lung injury that induces cytokine hypersecretion. Receptor activity-modifying protein (RAMP) 1, a subunit of the calcitonin gene-related peptide (CGRP) receptor, regulates the production of cytokines. This study examined the role of RAMP1 signaling during lipopolysaccharide (LPS)-induced acute lung injury (ALI). LPS administration to wild-type (WT) mice depleted alveolar macrophages (AMs) and recruited monocyte-derived macrophages (MDMs) and neutrophils. RAMP1-deficient (RAMP1-/-) mice exhibited higher lung injury scores, cytokine levels, and cytokine-producing neutrophil infiltration. RAMP1-deficient AMs produced more cytokines in response to LPS than WT AMs. Adoptive transfer of RAMP1-deficient AMs to RAMP1-/- mice increased cytokine levels and neutrophil accumulation compared to the transfer of WT AMs. RAMP1-/- mice had reduced MDM recruitment and lower pro-inflammatory and reparative macrophage profiles. Cultured bone marrow (BM)-derived RAMP1-deficient macrophages stimulated with LPS showed decreased expression of pro-inflammatory and pro-repairing genes. CGRP administration to WT mice reduced cytokine production and neutrophil accumulation. These findings indicate that RAMP1 signaling mitigates LPS-induced ALI by inactivating AMs and promoting inflammatory and repair activities of MDMs. Targeting RAMP1 signaling presents a potential therapeutic approach for the treatment of ARDS.
Collapse
Affiliation(s)
- Atsushi Yamashita
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara 252-0373, Japan; (A.Y.); (Y.I.); (K.H.)
- Department of Anesthesiology, Kitasato University School of Medicine, Sagamihara 252-0374, Japan (H.O.)
| | - Yoshiya Ito
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara 252-0373, Japan; (A.Y.); (Y.I.); (K.H.)
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara 252-0374, Japan
| | - Mayuko Osada
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara 252-0373, Japan; (A.Y.); (Y.I.); (K.H.)
- Department of Emergency and Critical Care Medicine, Kitasato University School of Medicine, Sagamihara 252-0374, Japan
| | - Hiromi Matsuda
- Department of Anesthesiology, Kitasato University School of Medicine, Sagamihara 252-0374, Japan (H.O.)
| | - Kanako Hosono
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara 252-0373, Japan; (A.Y.); (Y.I.); (K.H.)
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara 252-0374, Japan
| | - Kazutake Tsujikawa
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan;
| | - Hirotsugu Okamoto
- Department of Anesthesiology, Kitasato University School of Medicine, Sagamihara 252-0374, Japan (H.O.)
| | - Hideki Amano
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara 252-0373, Japan; (A.Y.); (Y.I.); (K.H.)
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara 252-0374, Japan
| |
Collapse
|
6
|
Joshi PR, Adhikari S, Onah C, Carrier C, Judd A, Mack M, Baral P. Lung-innervating nociceptor sensory neurons promote pneumonic sepsis during carbapenem-resistant Klebsiella pneumoniae lung infection. SCIENCE ADVANCES 2024; 10:eadl6162. [PMID: 39241063 PMCID: PMC11378917 DOI: 10.1126/sciadv.adl6162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 07/30/2024] [Indexed: 09/08/2024]
Abstract
Carbapenem-resistant Klebsiella pneumoniae (CRKP) causes Gram-negative lung infections and fatal pneumonic sepsis for which limited therapeutic options are available. The lungs are densely innervated by nociceptor sensory neurons that mediate breathing, cough, and bronchoconstriction. The role of nociceptors in defense against Gram-negative lung pathogens is unknown. Here, we found that lung-innervating nociceptors promote CRKP pneumonia and pneumonic sepsis. Ablation of nociceptors in mice increased lung CRKP clearance, suppressed trans-alveolar dissemination of CRKP, and protected mice from hypothermia and death. Furthermore, ablation of nociceptors enhanced the recruitment of neutrophils and Ly6Chi monocytes and cytokine induction. Depletion of Ly6Chi monocytes, but not of neutrophils, abrogated lung and extrapulmonary CRKP clearance in ablated mice, suggesting that Ly6Chi monocytes are a critical cellular population to regulate pneumonic sepsis. Further, neuropeptide calcitonin gene-related peptide suppressed the induction of reactive oxygen species in Ly6Chi monocytes and their CRKP-killing abilities. Targeting nociceptor signaling could be a therapeutic approach for treating multidrug-resistant Gram-negative infection and pneumonic sepsis.
Collapse
Affiliation(s)
- Prabhu Raj Joshi
- Section of Microbiology and Immunology, Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| | - Sandeep Adhikari
- Section of Microbiology and Immunology, Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| | - Chinemerem Onah
- Section of Microbiology and Immunology, Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| | - Camille Carrier
- Section of Microbiology and Immunology, Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| | - Abigail Judd
- Section of Microbiology and Immunology, Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| | - Matthias Mack
- Department of Nephrology, Regensburg University Medical Center, Regensburg 93042, Germany
| | - Pankaj Baral
- Section of Microbiology and Immunology, Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| |
Collapse
|
7
|
Niu Q, Wang M, Liu XS. The evolving landscape of IL-10, IL-22 and IL-26 in pleurisy especially in tuberculous pleurisy. Respir Res 2024; 25:275. [PMID: 39003443 PMCID: PMC11245850 DOI: 10.1186/s12931-024-02896-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 06/29/2024] [Indexed: 07/15/2024] Open
Abstract
Pleurisy can be categorized as primary or secondary, arising from immunological, tumorous, or microbial conditions. It often results in lung structure damage and the development of various respiratory issues. Among the different types, tuberculous pleurisy has emerged as a prominent focus for both clinical and scientific investigations. The IL-10 family, known for its anti-inflammatory properties in the human immune system, is increasingly being studied for its involvement in the pathogenesis of pleurisy. This review aims to present a detailed overview of the intricate role of IL-10 family members (specifically IL-10, IL-22, and IL-26) in human and animal pleuritic diseases or relevant animal models. These insights could serve as valuable guidance and references for further studies on pleurisy and potential therapeutic strategies.
Collapse
Affiliation(s)
- Qian Niu
- Department of Respiratory and Critical Care Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Meng Wang
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Pathology, Baoji Gaoxin Hospital, Baoji, 721000, China
| | - Xian-Sheng Liu
- Department of Respiratory and Critical Care Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China.
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
8
|
Maximiano TKE, Carneiro JA, Fattori V, Verri WA. TRPV1: Receptor structure, activation, modulation and role in neuro-immune interactions and pain. Cell Calcium 2024; 119:102870. [PMID: 38531262 DOI: 10.1016/j.ceca.2024.102870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 03/06/2024] [Accepted: 03/06/2024] [Indexed: 03/28/2024]
Abstract
In the 1990s, the identification of a non-selective ion channel, especially responsive to capsaicin, revolutionized the studies of somatosensation and pain that were to follow. The TRPV1 channel is expressed mainly in neuronal cells, more specifically, in sensory neurons responsible for the perception of noxious stimuli. However, its presence has also been detected in other non-neuronal cells, such as immune cells, β- pancreatic cells, muscle cells and adipocytes. Activation of the channel occurs in response to a wide range of stimuli, such as noxious heat, low pH, gasses, toxins, endocannabinoids, lipid-derived endovanilloid, and chemical agents, such as capsaicin and resiniferatoxin. This activation results in an influx of cations through the channel pore, especially calcium. Intracellular calcium triggers different responses in sensory neurons. Dephosphorylation of the TRPV1 channel leads to its desensitization, which disrupts its function, while its phosphorylation increases the channel's sensitization and contributes to the channel's rehabilitation after desensitization. Kinases, phosphoinositides, and calmodulin are the main signaling pathways responsible for the channel's regulation. Thus, in this review we provide an overview of TRPV1 discovery, its tissue expression as well as on the mechanisms by which TRPV1 activation (directly or indirectly) induces pain in different disease models.
Collapse
Affiliation(s)
- Thaila Kawane Euflazio Maximiano
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
| | - Jessica Aparecida Carneiro
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
| | - Victor Fattori
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital-Harvard Medical School, Karp Research Building, 300 Longwood Ave, 02115, Boston, Massachusetts, United States.
| | - Waldiceu A Verri
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil.
| |
Collapse
|
9
|
Lu YZ, Nayer B, Singh SK, Alshoubaki YK, Yuan E, Park AJ, Maruyama K, Akira S, Martino MM. CGRP sensory neurons promote tissue healing via neutrophils and macrophages. Nature 2024; 628:604-611. [PMID: 38538784 PMCID: PMC11023938 DOI: 10.1038/s41586-024-07237-y] [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: 08/04/2023] [Accepted: 02/26/2024] [Indexed: 04/06/2024]
Abstract
The immune system has a critical role in orchestrating tissue healing. As a result, regenerative strategies that control immune components have proved effective1,2. This is particularly relevant when immune dysregulation that results from conditions such as diabetes or advanced age impairs tissue healing following injury2,3. Nociceptive sensory neurons have a crucial role as immunoregulators and exert both protective and harmful effects depending on the context4-12. However, how neuro-immune interactions affect tissue repair and regeneration following acute injury is unclear. Here we show that ablation of the NaV1.8 nociceptor impairs skin wound repair and muscle regeneration after acute tissue injury. Nociceptor endings grow into injured skin and muscle tissues and signal to immune cells through the neuropeptide calcitonin gene-related peptide (CGRP) during the healing process. CGRP acts via receptor activity-modifying protein 1 (RAMP1) on neutrophils, monocytes and macrophages to inhibit recruitment, accelerate death, enhance efferocytosis and polarize macrophages towards a pro-repair phenotype. The effects of CGRP on neutrophils and macrophages are mediated via thrombospondin-1 release and its subsequent autocrine and/or paracrine effects. In mice without nociceptors and diabetic mice with peripheral neuropathies, delivery of an engineered version of CGRP accelerated wound healing and promoted muscle regeneration. Harnessing neuro-immune interactions has potential to treat non-healing tissues in which dysregulated neuro-immune interactions impair tissue healing.
Collapse
Affiliation(s)
- Yen-Zhen Lu
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia
| | - Bhavana Nayer
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia
| | - Shailendra Kumar Singh
- Laboratory of Host Defense, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Yasmin K Alshoubaki
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia
| | - Elle Yuan
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia
| | - Anthony J Park
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia
| | - Kenta Maruyama
- Laboratory of Host Defense, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Department of Pharmacology, School of Medicine, Aichi Medical University, Aichi, Japan
| | - Shizuo Akira
- Laboratory of Host Defense, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Mikaël M Martino
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia.
- Laboratory of Host Defense, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Osaka, Japan.
- Victorian Heart Institute, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
10
|
Hanč P, Messou MA, Wang Y, von Andrian UH. Control of myeloid cell functions by nociceptors. Front Immunol 2023; 14:1127571. [PMID: 37006298 PMCID: PMC10064072 DOI: 10.3389/fimmu.2023.1127571] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/23/2023] [Indexed: 03/19/2023] Open
Abstract
The immune system has evolved to protect the host from infectious agents, parasites, and tumor growth, and to ensure the maintenance of homeostasis. Similarly, the primary function of the somatosensory branch of the peripheral nervous system is to collect and interpret sensory information about the environment, allowing the organism to react to or avoid situations that could otherwise have deleterious effects. Consequently, a teleological argument can be made that it is of advantage for the two systems to cooperate and form an “integrated defense system” that benefits from the unique strengths of both subsystems. Indeed, nociceptors, sensory neurons that detect noxious stimuli and elicit the sensation of pain or itch, exhibit potent immunomodulatory capabilities. Depending on the context and the cellular identity of their communication partners, nociceptors can play both pro- or anti-inflammatory roles, promote tissue repair or aggravate inflammatory damage, improve resistance to pathogens or impair their clearance. In light of such variability, it is not surprising that the full extent of interactions between nociceptors and the immune system remains to be established. Nonetheless, the field of peripheral neuroimmunology is advancing at a rapid pace, and general rules that appear to govern the outcomes of such neuroimmune interactions are beginning to emerge. Thus, in this review, we summarize our current understanding of the interaction between nociceptors and, specifically, the myeloid cells of the innate immune system, while pointing out some of the outstanding questions and unresolved controversies in the field. We focus on such interactions within the densely innervated barrier tissues, which can serve as points of entry for infectious agents and, where known, highlight the molecular mechanisms underlying these interactions.
Collapse
Affiliation(s)
- Pavel Hanč
- Department of Immunology, Harvard Medical School, Boston, MA, United States
- The Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
- *Correspondence: Pavel Hanč, ; Ulrich H. von Andrian,
| | - Marie-Angèle Messou
- Department of Immunology, Harvard Medical School, Boston, MA, United States
- The Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
| | - Yidi Wang
- Department of Immunology, Harvard Medical School, Boston, MA, United States
- The Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
| | - Ulrich H. von Andrian
- Department of Immunology, Harvard Medical School, Boston, MA, United States
- The Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
- *Correspondence: Pavel Hanč, ; Ulrich H. von Andrian,
| |
Collapse
|
11
|
Pinho-Ribeiro FA, Deng L, Neel DV, Erdogan O, Basu H, Yang D, Choi S, Walker AJ, Carneiro-Nascimento S, He K, Wu G, Stevens B, Doran KS, Levy D, Chiu IM. Bacteria hijack a meningeal neuroimmune axis to facilitate brain invasion. Nature 2023; 615:472-481. [PMID: 36859544 PMCID: PMC10593113 DOI: 10.1038/s41586-023-05753-x] [Citation(s) in RCA: 76] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 01/23/2023] [Indexed: 03/03/2023]
Abstract
The meninges are densely innervated by nociceptive sensory neurons that mediate pain and headache1,2. Bacterial meningitis causes life-threatening infections of the meninges and central nervous system, affecting more than 2.5 million people a year3-5. How pain and neuroimmune interactions impact meningeal antibacterial host defences are unclear. Here we show that Nav1.8+ nociceptors signal to immune cells in the meninges through the neuropeptide calcitonin gene-related peptide (CGRP) during infection. This neuroimmune axis inhibits host defences and exacerbates bacterial meningitis. Nociceptor neuron ablation reduced meningeal and brain invasion by two bacterial pathogens: Streptococcus pneumoniae and Streptococcus agalactiae. S. pneumoniae activated nociceptors through its pore-forming toxin pneumolysin to release CGRP from nerve terminals. CGRP acted through receptor activity modifying protein 1 (RAMP1) on meningeal macrophages to polarize their transcriptional responses, suppressing macrophage chemokine expression, neutrophil recruitment and dural antimicrobial defences. Macrophage-specific RAMP1 deficiency or pharmacological blockade of RAMP1 enhanced immune responses and bacterial clearance in the meninges and brain. Therefore, bacteria hijack CGRP-RAMP1 signalling in meningeal macrophages to facilitate brain invasion. Targeting this neuroimmune axis in the meninges can enhance host defences and potentially produce treatments for bacterial meningitis.
Collapse
Affiliation(s)
- Felipe A Pinho-Ribeiro
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Division of Dermatology, John T. Milliken Department of Medicine, Washington University School of Medicine in St Louis, St Louis, MO, USA
| | - Liwen Deng
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Dylan V Neel
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Ozge Erdogan
- Department of Restorative Dentistry and Biomaterial Sciences, Harvard School of Dental Medicine, Boston, MA, USA
| | - Himanish Basu
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Daping Yang
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Samantha Choi
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Alec J Walker
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Simone Carneiro-Nascimento
- Departments of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Kathleen He
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Glendon Wu
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Beth Stevens
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA
| | - Kelly S Doran
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Dan Levy
- Harvard Medical School, Boston, MA, USA
- Departments of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Isaac M Chiu
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
12
|
Sensory nerves promote corneal inflammation resolution via CGRP mediated transformation of macrophages to the M2 phenotype through the PI3K/AKT signaling pathway. Int Immunopharmacol 2021; 102:108426. [PMID: 34906854 DOI: 10.1016/j.intimp.2021.108426] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/24/2021] [Accepted: 11/28/2021] [Indexed: 02/07/2023]
Abstract
OBJECTIVES To explore the role of the corneal sensory nerves in Pseudomonas aeruginosa (P. aeruginosa) keratitis, the synergistic effect between the sensory neurons and macrophages in calcitonin gene-related peptide (CGRP) release, and the functional mechanisms of CGRP-mediated transformation of macrophages to the M2 phenotype. METHODS Corneal nerve loss, macrophage recruitment, and CGRP expression were evaluated. To explore the synergistic effect between the sensory neurons and macrophages, RAW 264.7 cells were challenged with lipopolysaccharide (LPS), then trigeminal ganglion (TG) sensory neurons were isolated and co-incubated with macrophages, and CGRP expression was tested. To investigate the biological function of cornea neuron-initiated immune responses mediated by CGRP, BIBN 4096BS was used to inhibit CGRP in vivo and α-CGRP was used to simulate CGRP in vitro. The expressions of inflammatory cytokines (IL-1β, IL-6, TNF-α, and IL-10), M1 (CD80/CD86), M2 (CD163/CD206) macrophage markers, and signal transducers (PI3K/AKT) were detected. RESULTS P. aeruginosa infection induced corneal nerve loss, macrophage recruitment, and CGRP up-expression. CGRP was co-localized with macrophages. Co-culture showed that sensory neurons and macrophages can mediate CGRP release. More CGRP was released when the two types of cells were combined to respond to LPS. BIBN 4096BS promoted pro-inflammatory cytokines and inhibited the anti-inflammatory cytokines and signal transducers, while, α-CGRP inhibited the pro-inflammatory cytokines and M1 markers and promoted the anti-inflammatory cytokine, M2 markers, and signal transducers. CONCLUSIONS P. aeruginosa infection induces corneal sensory neuron activation, macrophage recruitment, and CGRP up-expression. The synergistic effect between the sensory neurons and macrophages promotes CGRP release. CGRP inhibits corneal inflammation and promotes the transformation of macrophages to the M2 phenotype through the PI3K/AKT signaling pathway.
Collapse
|
13
|
Lehmann C, Aali M, Zhou J, Holbein B. Comparison of Treatment Effects of Different Iron Chelators in Experimental Models of Sepsis. Life (Basel) 2021; 11:life11010057. [PMID: 33466819 PMCID: PMC7830599 DOI: 10.3390/life11010057] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/09/2021] [Accepted: 01/11/2021] [Indexed: 01/19/2023] Open
Abstract
Growing evidence indicates that dysregulated iron metabolism with altered and excess iron availability in some body compartments plays a significant role in the course of infection and sepsis in humans. Given that all bacterial pathogens require iron for growth, that iron withdrawal is a normal component of innate host defenses and that bacterial pathogens have acquired increasing levels of antibiotic resistance, targeting infection and sepsis through use of appropriate iron chelators has potential to provide new therapeutics. We have directly compared the effects of three Food and Drug Administration (FDA)-approved chelators (deferoxamine—DFO; deferiprone—DFP; and deferasirox—DFX), as were developed for treating hematological iron overload conditions, to DIBI, a novel purpose-designed, anti-infective and anti-inflammatory water-soluble hydroxypyridinone containing iron-selective copolymers. Two murine sepsis models, endotoxemia and polymicrobial abdominal sepsis, were utilized to help differentiate anti-inflammatory versus anti-infective activities of the chelators. Leukocyte adhesion, as measured by intravital microscopy, was observed in both models, with DIBI providing the most effective reduction and DFX the poorest. Inflammation in the abdominal sepsis model, assessed by cytokine measurements, indicated exacerbation by DFX and DFO for plasma Interleukin (IL)-6 and reductions to near-control levels for DIBI and DFP. Peritoneal infection burden was reduced 10-fold by DIBI while DFX and DFP provided no reductions. Overall, the results, together with those from other studies, revealed serious limitations for each of the three hematological chelators, i.e., as potentially repurposed for treating infection/sepsis. In contrast, DIBI provided therapeutic benefits, consistent with various in vitro and in vivo results from other studies, supporting the potential for its use in treating sepsis.
Collapse
Affiliation(s)
- Christian Lehmann
- Department of Anesthesia, Pain Management and Perioperative Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada;
- Correspondence:
| | - Maral Aali
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS B3H 4R2, Canada;
| | - Juan Zhou
- Department of Anesthesia, Pain Management and Perioperative Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada;
| | - Bruce Holbein
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada;
| |
Collapse
|
14
|
Xie D, Xu Y, Yang Y, Hua Z, Li J, Fu G, Wu Q. Sensory denervation increases potential of bisphosphonates to induce osteonecrosis via disproportionate expression of calcitonin gene-related peptide and substance P. Ann N Y Acad Sci 2020; 1487:56-73. [PMID: 33301204 DOI: 10.1111/nyas.14540] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/30/2020] [Accepted: 11/03/2020] [Indexed: 12/17/2022]
Abstract
Bisphosphonate-related osteonecrosis of the jaw (BRONJ) is a serious side effect of systematic administration of bisphosphonates (BPs). Sensory innervation is crucial for bone healing. We established inferior alveolar nerve injury (IANI) and inferior alveolar nerve transection (IANT) models characterized by disorganized periosteum, increased osteoclasts, and unbalanced neuropeptide expression. Zoledronate injection disrupted neuropeptide expression in the IANI and IANT models by decreasing calcitonin gene-related peptide (CGRP) and increasing substance P (SP); associated with this, BRONJ prevalence was significantly higher in the IANT model, followed by the IANI model and the sham control. CGRP treatment significantly reduced BRONJ occurrence, whereas SP administration had the opposite effect. In vitro, RAW 264.7 cells were treated with BPs and then CGRP and/or SP to study changes in zoledronate toxicity; combined application of CGRP and SP decreased zoledronate toxicity, whereas CGRP or SP applied alone showed no effects. These results demonstrate that sensory denervation facilitates the occurrence of BRONJ and that CGRP used therapeutically may prevent BRONJ progression, provided that SP is also present. Further studies are necessary to determine the optimal ratio of CGRP to SP for promoting bone healing and to uncover the mechanism by which CGRP and SP cooperate.
Collapse
Affiliation(s)
- Dongni Xie
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Yamei Xu
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Yao Yang
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Ziyi Hua
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Jiao Li
- Department of Oral Implantology, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Gang Fu
- Department of Oral Implantology, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Qingqing Wu
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
15
|
Fattori V, Ferraz CR, Rasquel-Oliveira FS, Verri WA. Neuroimmune communication in infection and pain: Friends or foes? Immunol Lett 2020; 229:32-43. [PMID: 33248166 DOI: 10.1016/j.imlet.2020.11.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 10/02/2020] [Accepted: 11/20/2020] [Indexed: 12/13/2022]
Abstract
Clinically, a variety of micro-organisms cause painful infections. Before seen as bystanders in the context of infections, recent studies have demonstrated that, as immune cells, nociceptors can sense pathogen-derived products. Nociceptors and immune cells, therefore, have evolved to communicate with each other to control inflammatory and host responses against pathogens in a complementary way. This interaction is named as neuroimmune communication (or axon-axon immune reflex) and initiates after the release of neuropeptides, such as CGRP and VIP by neurons. By this neurogenic response, nociceptors orchestrate the activity of innate and adaptive immune cells in a context-dependent manner. In this review, we focus on how nociceptors sense pathogen-derived products to shape the host response. We also highlight the new concept involving the resolution of inflammation, which is related to an active and time-dependent biosynthetic shift from pro-inflammatory to pro-resolution mediators, the so-called specialized pro-resolving lipid mediators (SPMs). At very low doses, SPMs act on specific receptors to silence nociceptors, limit pain and neurogenic responses, and resolve infections. Furthermore, stimulation of the vagus nerve induces SPMs production to regulate immune responses in infections. Therefore, harnessing the current understanding of neuro-immune communication and neurogenic responses might provide the bases for reprogramming host responses against infections through well balanced and effective immune response and inflammation resolution.
Collapse
Affiliation(s)
- Victor Fattori
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, PR, Brazil; Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Camila R Ferraz
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, PR, Brazil
| | - Fernanda S Rasquel-Oliveira
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, PR, Brazil
| | - Waldiceu A Verri
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, PR, Brazil.
| |
Collapse
|
16
|
Wallrapp A, Burkett PR, Riesenfeld SJ, Kim SJ, Christian E, Abdulnour REE, Thakore PI, Schnell A, Lambden C, Herbst RH, Khan P, Tsujikawa K, Xavier RJ, Chiu IM, Levy BD, Regev A, Kuchroo VK. Calcitonin Gene-Related Peptide Negatively Regulates Alarmin-Driven Type 2 Innate Lymphoid Cell Responses. Immunity 2019; 51:709-723.e6. [PMID: 31604686 PMCID: PMC7076585 DOI: 10.1016/j.immuni.2019.09.005] [Citation(s) in RCA: 169] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 06/19/2019] [Accepted: 09/09/2019] [Indexed: 12/22/2022]
Abstract
Neuroimmune interactions have emerged as critical modulators of allergic inflammation, and type 2 innate lymphoid cells (ILC2s) are an important cell type for mediating these interactions. Here, we show that ILC2s expressed both the neuropeptide calcitonin gene-related peptide (CGRP) and its receptor. CGRP potently inhibited alarmin-driven type 2 cytokine production and proliferation by lung ILC2s both in vitro and in vivo. CGRP induced marked changes in ILC2 expression programs in vivo and in vitro, attenuating alarmin-driven proliferative and effector responses. A distinct subset of ILCs scored highly for a CGRP-specific gene signature after in vivo alarmin stimulation, suggesting CGRP regulated this response. Finally, we observed increased ILC2 proliferation and type 2 cytokine production as well as exaggerated responses to alarmins in mice lacking the CGRP receptor. Together, these data indicate that endogenous CGRP is a critical negative regulator of ILC2 responses in vivo.
Collapse
Affiliation(s)
- Antonia Wallrapp
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Patrick R Burkett
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Samantha J Riesenfeld
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Se-Jin Kim
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Elena Christian
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Raja-Elie E Abdulnour
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Pratiksha I Thakore
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Alexandra Schnell
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Conner Lambden
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Rebecca H Herbst
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Pavana Khan
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Kazutake Tsujikawa
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan
| | - Ramnik J Xavier
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Isaac M Chiu
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Bruce D Levy
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Aviv Regev
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Howard Hughes Medical Institute and Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.
| | - Vijay K Kuchroo
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
17
|
Zhao Q, Wang J, Yin C, Zhang P, Zhang J, Shi M, Shen K, Xiao Y, Zhao Y, Yang X, Zhang Y. Near-Infrared Light-Sensitive Nano Neuro-Immune Blocker Capsule Relieves Pain and Enhances the Innate Immune Response for Necrotizing Infection. NANO LETTERS 2019; 19:5904-5914. [PMID: 31389707 DOI: 10.1021/acs.nanolett.9b01459] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Sensory neurons promote profound suppressive effects on neutrophils during Streptococcus pyogenes infection and contribute to the pathogenesis of necrotizing infection ("flesh-eating disease"). Thus, the development of new antibacterial agents for necrotizing infection is promising because of the clear streptococcal neuro-immune communication. Herein, based on the immune escape membrane exterior and competitive membrane functions of the glioma cell membrane, a novel nano neuro-immune blocker capsule was designed to prevent neuronal activation and improve neutrophil immune responses for necrotizing infection. These nano neuro-immune blockers could neutralize streptolysin S, suppress neuron pain conduction and calcitonin gene-related peptide release, and recruit neutrophils to the infection site, providing a strong therapeutic effect against necrotizing infection. Furthermore, nano neuro-immune blockers could serve as an effective inflammatory regulator and antibacterial agent via photothermal effects under near-infrared irradiation. In the Streptococcus pyogenes-induced necrotizing fasciitis mouse model, nano neuro-immune blockers showed significant therapeutic efficacy by ameliorating sensitivity to pain and promoting the antibacterial effect of neutrophils.
Collapse
Affiliation(s)
- Qin Zhao
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology , Wuhan University , Wuhan 430079 , China
| | - Jinyang Wang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology , Wuhan University , Wuhan 430079 , China
| | - Chengcheng Yin
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology , Wuhan University , Wuhan 430079 , China
| | - Peng Zhang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology , Wuhan University , Wuhan 430079 , China
| | - Jinglun Zhang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology , Wuhan University , Wuhan 430079 , China
| | - Miusi Shi
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology , Wuhan University , Wuhan 430079 , China
| | - Kailun Shen
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology , Wuhan University , Wuhan 430079 , China
| | - Yin Xiao
- Institute of Health and Biomedical Innovation , Queensland University of Technology , Kelvin Grove , Queensland 4059 , Australia
| | - Yanbing Zhao
- National Engineering Research Center for Nanomedicine, Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, College of Life Science and Technology , Huazhong University of Science and Technology , Wuhan 430074 , China
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, College of Life Science and Technology , Huazhong University of Science and Technology , Wuhan 430074 , China
| | - Yufeng Zhang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology , Wuhan University , Wuhan 430079 , China
- Medical Research Institute, School of Medicine , Wuhan University , Wuhan , 430071 , China
| |
Collapse
|
18
|
Borkum JM. CGRP and Brain Functioning: Cautions for Migraine Treatment. Headache 2019; 59:1339-1357. [DOI: 10.1111/head.13591] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Jonathan M. Borkum
- Department of Psychology University of Maine Orono ME USA
- Health Psych Maine Waterville ME USA
| |
Collapse
|
19
|
Abstract
Pain is a hallmark of tissue injury, inflammatory diseases, pathogen invasion and neuropathy. It is mediated by nociceptor sensory neurons that innervate the skin, joints, bones, muscles and mucosal tissues and protects organisms from noxious stimuli. Nociceptors are sensitized by inflammatory mediators produced by the immune system, including cytokines, lipid mediators and growth factors, and can also directly detect pathogens and their secreted products to produce pain during infection. Upon activation, nociceptors release neuropeptides from their terminals that potently shape the function of innate and adaptive immune cells. For some pathogens, neuron-immune interactions enhance host protection from infection, but for other pathogens, neuron-immune signalling pathways can be exploited to facilitate pathogen survival. Here, we discuss the role of nociceptor interactions with the immune system in pain and infection and how understanding these pathways could produce new approaches to treat infectious diseases and chronic pain.
Collapse
|
20
|
Jochheim LS, Odysseos G, Hidalgo-Sastre A, Zhong S, Staufer LM, Kroiss M, Kabacaoglu D, Lange S, Engleitner T, Hartmann D, Hüser N, Steiger K, Schmid RM, Holzmann B, von Figura G. The neuropeptide receptor subunit RAMP1 constrains the innate immune response during acute pancreatitis in mice. Pancreatology 2019; 19:541-547. [PMID: 31109903 DOI: 10.1016/j.pan.2019.05.455] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 05/02/2019] [Accepted: 05/10/2019] [Indexed: 12/11/2022]
Abstract
OBJECTIVES The importance of the Calcitonin-gene-related-peptide-pathway (CGRP) as neuronal modulator of innate immune responses in mice has been previously demonstrated. The CGRP-receptor is composed of two subunits: the receptor-activity-modifying-protein-1 (RAMP1) and the calcitonin-receptor-like-receptor (CLR). CGRP can influence immune cells and their capacity of producing inflammatory cytokines. Using a RAMP1 knockout-mouse (RAMP1-/-) we examined the role of the CGRP-receptor in the acute-phase of cerulein-induced pancreatitis. METHODS Hourly cerulein-injections for a period of 8 h in RAMP1-/- and wild-type mice were performed. To compare severity and extent of inflammation in RAMP1-/- and wild-type mice, histological analyses were done and cytokine levels were assessed using qRT-PCR 8 h, 24 h, 2 days, and 7 days post-cerulein-treatment. Furthermore, serum activities of LDH and lipase were determined. RESULTS After 8 h RAMP1-/- mice showed a higher pancreas-to-body-weight-ratio, increased tissue edema and immune cell infiltration with higher amount of F4/80-positive cells as compared to wild-type mice. Overall infiltration of immune cells at 24 h was increased in RAMP1-/- mice and composed predominantly of MPO-positive neutrophils. In addition, after 24 h RAMP1-/- mice presented a higher pancreas-to-body-weight-ratio, higher expression of Ccl3, Il6, and Il1b and increased number of cleaved caspase 3 positive cells. Serum lipase correlated with the extent of tissue damage in RAMP1-/- compared to wild-type mice 24 h post-cerulein treatment. CONCLUSION Mice lacking RAMP1 showed increased inflammation, tissue edema, and pancreas injury particularly in the early phase of acute pancreatitis. This study highlights the essential role of CGRP for dampening the innate immune response in acute pancreatitis.
Collapse
Affiliation(s)
- Leonie S Jochheim
- Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Department of Internal Medicine II, Munich, Germany
| | - Georgios Odysseos
- Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Department of Internal Medicine II, Munich, Germany
| | - Ana Hidalgo-Sastre
- Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Department of Internal Medicine II, Munich, Germany
| | - Suyang Zhong
- Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Department of Internal Medicine II, Munich, Germany
| | - Lina M Staufer
- Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Department of Internal Medicine II, Munich, Germany
| | - Markus Kroiss
- Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Department of Internal Medicine II, Munich, Germany
| | - Derya Kabacaoglu
- Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Department of Internal Medicine II, Munich, Germany
| | - Sebastian Lange
- Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Department of Internal Medicine II, Munich, Germany; Technical University of Munich, School of Medicine, Institute of Molecular Oncology and Functional Genomics, Munich, Germany
| | - Thomas Engleitner
- Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Department of Internal Medicine II, Munich, Germany; Technical University of Munich, School of Medicine, Institute of Molecular Oncology and Functional Genomics, Munich, Germany
| | - Daniel Hartmann
- Technical University Munich, School of Medicine, Klinikum Rechts der Isar, Department of Surgery, Munich, Germany
| | - Norbert Hüser
- Technical University Munich, School of Medicine, Klinikum Rechts der Isar, Department of Surgery, Munich, Germany
| | - Katja Steiger
- Technical University Munich, School of Medicine, Department of Pathology, Munich, Germany
| | - Roland M Schmid
- Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Department of Internal Medicine II, Munich, Germany
| | - Bernhard Holzmann
- Technical University Munich, School of Medicine, Klinikum Rechts der Isar, Department of Surgery, Munich, Germany
| | - Guido von Figura
- Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Department of Internal Medicine II, Munich, Germany.
| |
Collapse
|
21
|
The cAMP Pathway Amplifies Early MyD88-Dependent and Type I Interferon-Independent LPS-Induced Interleukin-10 Expression in Mouse Macrophages. Mediators Inflamm 2019; 2019:3451461. [PMID: 31148944 PMCID: PMC6501241 DOI: 10.1155/2019/3451461] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 01/05/2019] [Accepted: 02/11/2019] [Indexed: 12/20/2022] Open
Abstract
Interleukin-10 (IL-10) is a key anti-inflammatory cytokine, secreted by macrophages and other immune cells to attenuate inflammation. Autocrine type I interferons (IFNs) largely mediate the delayed expression of IL-10 by LPS-stimulated macrophages. We have previously shown that IL-10 is synergistically expressed in macrophages following a costimulus of a TLR agonist and cAMP. We now show that the cAMP pathway directly upregulates IL-10 transcription and plays an important permissive and synergistic role in early, but not late, LPS-stimulated IL-10 mRNA and protein expression in mouse macrophages and in a mouse septic shock model. Our results suggest that the loss of synergism is not due to desensitization of the cAMP inducing signal, and it is not mediated by a positive crosstalk between the cAMP and type I IFN pathways. First, cAMP elevation in LPS-treated cells decreased the secretion of type I IFN. Second, autocrine/paracrine type I IFNs induce IL-10 promoter reporter activity only additively, but not synergistically, with the cAMP pathway. IL-10 promoter reporter activity was synergistically induced by cAMP elevation in macrophages stimulated by an agonist of either TLR4, TLR2/6, or TLR7, receptors which signal via MyD88, but not by an agonist of TLR3 which signals independently of MyD88. Moreover, MyD88 knockout largely reduced the synergistic IL-10 expression, indicating that MyD88 is required for the synergism displayed by LPS with cAMP. This report delineates the temporal regulation of early cAMP-accelerated vs. late type I IFN-dependent IL-10 transcription in LPS-stimulated murine macrophages that can limit inflammation at its onset.
Collapse
|
22
|
Pinho-Ribeiro FA, Verri WA, Chiu IM. Nociceptor Sensory Neuron-Immune Interactions in Pain and Inflammation. Trends Immunol 2016; 38:5-19. [PMID: 27793571 DOI: 10.1016/j.it.2016.10.001] [Citation(s) in RCA: 665] [Impact Index Per Article: 73.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 10/02/2016] [Accepted: 10/03/2016] [Indexed: 12/12/2022]
Abstract
Nociceptor sensory neurons protect organisms from danger by eliciting pain and driving avoidance. Pain also accompanies many types of inflammation and injury. It is increasingly clear that active crosstalk occurs between nociceptor neurons and the immune system to regulate pain, host defense, and inflammatory diseases. Immune cells at peripheral nerve terminals and within the spinal cord release mediators that modulate mechanical and thermal sensitivity. In turn, nociceptor neurons release neuropeptides and neurotransmitters from nerve terminals that regulate vascular, innate, and adaptive immune cell responses. Therefore, the dialog between nociceptor neurons and the immune system is a fundamental aspect of inflammation, both acute and chronic. A better understanding of these interactions could produce approaches to treat chronic pain and inflammatory diseases.
Collapse
Affiliation(s)
- Felipe A Pinho-Ribeiro
- Department of Microbiology and Immunobiology, Division of Immunology, Harvard Medical School, Boston, MA 02115, USA; Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, PR 10011, Brazil
| | - Waldiceu A Verri
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, PR 10011, Brazil
| | - Isaac M Chiu
- Department of Microbiology and Immunobiology, Division of Immunology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
23
|
Abstract
It is now recognized that G protein-coupled receptors (GPCRs), once considered largely independent functional units, have a far more diverse molecular architecture. Receptor activity-modifying proteins (RAMPs) provide an important example of proteins that interact with GPCRs to modify their function. RAMPs are able to act as pharmacological switches and chaperones, and they can regulate signaling and/or trafficking in a receptor-dependent manner. This review covers recent discoveries in the RAMP field and summarizes the known GPCR partners and functions of RAMPs. We also discuss the first peptide-bound structures of RAMP-GPCR complexes, which give insight into the molecular mechanisms that enable RAMPs to alter the pharmacology and signaling of GPCRs.
Collapse
Affiliation(s)
- Debbie L Hay
- School of Biological Sciences and Maurice Wilkins Center, University of Auckland, Auckland 1142, New Zealand;
| | - Augen A Pioszak
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104;
| |
Collapse
|
24
|
McMahon SB, Russa FL, Bennett DLH. Crosstalk between the nociceptive and immune systems in host defence and disease. Nat Rev Neurosci 2015; 16:389-402. [DOI: 10.1038/nrn3946] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
25
|
Baliu-Piqué M, Jusek G, Holzmann B. Neuroimmunological communication via CGRP promotes the development of a regulatory phenotype in TLR4-stimulated macrophages. Eur J Immunol 2014; 44:3708-16. [PMID: 25316186 DOI: 10.1002/eji.201444553] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 09/04/2014] [Accepted: 10/09/2014] [Indexed: 12/21/2022]
Abstract
Environmental signals shape the phenotype and function of activated macrophages. Here, we show that the neuropeptide calcitonin gene-related peptide (CGRP), which is released from sensory nerves, modulates the phenotype of TLR4-activated murine macrophages by enhancing expression of the regulatory macrophage markers IL-10, sphingosine kinase 1 (SPHK1), and LIGHT (lymphotoxin-like, exhibits inducible expression and competes with HSV glycoprotein D for herpesvirus entry mediator, a receptor expressed by T lymphocytes). In contrast, CGRP inhibits production of cytokines characteristic of inflammatory macrophages and does not affect expression of wound-healing macrophage markers upon TLR4 engagement. In IL-4-stimulated macrophages, CGRP increased LIGHT expression, but failed to induce IL-10 and SPHK1. The stimulatory effect of CGRP on IL-10 production required activation of protein kinase A and was linked to prolonged phosphorylation of CREB and sustained nuclear accumulation of CRTC2 and CRTC3 (where CRTC is CREB-regulated transcriptional cofactor). CGRP enhanced expression of regulatory macrophage markers during the early, but not late, phase of LPS-stimulation and this effect was independent of autocrine type-I IFN activity. In contrast, autocrine type-I IFN activity and treatment of macrophages with IFN-β promoted late-phase IL-10 production, but had only minor influence on LIGHT and SPHK1 expression. Together, the results identify neuroimmunological communication through CGRP as a novel costimulatory pathway promoting the development of a regulatory phenotype of TLR4-stimulated macrophages. CGRP appears to act through a mechanism that involves sustained activation of CREB-dependent gene transcription.
Collapse
|
26
|
Assas BM, Miyan JA, Pennock JL. Cross-talk between neural and immune receptors provides a potential mechanism of homeostatic regulation in the gut mucosa. Mucosal Immunol 2014; 7:1283-9. [PMID: 25183366 DOI: 10.1038/mi.2014.80] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 07/25/2014] [Indexed: 02/07/2023]
Abstract
The relationship between elements of the immune system and the nervous system in the presence of bacteria has been addressed recently. In particular, the sensory vanilloid receptor 1 (transient receptor potential cation channel subfamily V member 1 (TRPV1)) and the neuropeptide calcitonin gene-related peptide (CGRP) have been found to modulate cytokine response to lipopolysaccharide (LPS) independently of adaptive immunity. In this review we discuss mucosal homeostasis in the gastrointestinal tract where bacterial concentration is high. We propose that the Gram-negative bacterial receptor Toll-like receptor 4 (TLR4) can activate TRPV1 via intracellular signaling, and thereby induce the subsequent release of anti-inflammatory CGRP to maintain mucosal homeostasis.
Collapse
Affiliation(s)
- B M Assas
- 1] Faculty of Applied Medical Sciences, King Abdul Aziz University, Jeddah, Saudi Arabia [2] Faculty of Medicine and Human Sciences, University of Manchester, Manchester, UK
| | - J A Miyan
- Faculty of Life Sciences, University of Manchester, Manchester, UK
| | - J L Pennock
- Institute of Inflammation and Repair, Faculty of Medicine and Human Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
27
|
Russell FA, King R, Smillie SJ, Kodji X, Brain SD. Calcitonin gene-related peptide: physiology and pathophysiology. Physiol Rev 2014; 94:1099-142. [PMID: 25287861 PMCID: PMC4187032 DOI: 10.1152/physrev.00034.2013] [Citation(s) in RCA: 824] [Impact Index Per Article: 74.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Calcitonin gene-related peptide (CGRP) is a 37-amino acid neuropeptide. Discovered 30 years ago, it is produced as a consequence of alternative RNA processing of the calcitonin gene. CGRP has two major forms (α and β). It belongs to a group of peptides that all act on an unusual receptor family. These receptors consist of calcitonin receptor-like receptor (CLR) linked to an essential receptor activity modifying protein (RAMP) that is necessary for full functionality. CGRP is a highly potent vasodilator and, partly as a consequence, possesses protective mechanisms that are important for physiological and pathological conditions involving the cardiovascular system and wound healing. CGRP is primarily released from sensory nerves and thus is implicated in pain pathways. The proven ability of CGRP antagonists to alleviate migraine has been of most interest in terms of drug development, and knowledge to date concerning this potential therapeutic area is discussed. Other areas covered, where there is less information known on CGRP, include arthritis, skin conditions, diabetes, and obesity. It is concluded that CGRP is an important peptide in mammalian biology, but it is too early at present to know if new medicines for disease treatment will emerge from our knowledge concerning this molecule.
Collapse
Affiliation(s)
- F A Russell
- Cardiovascular Division, BHF Centre of Research Excellence & Centre of Integrative Biomedicine, King's College London, Waterloo Campus, London SE1 9NH, United Kingdom
| | - R King
- Cardiovascular Division, BHF Centre of Research Excellence & Centre of Integrative Biomedicine, King's College London, Waterloo Campus, London SE1 9NH, United Kingdom
| | - S-J Smillie
- Cardiovascular Division, BHF Centre of Research Excellence & Centre of Integrative Biomedicine, King's College London, Waterloo Campus, London SE1 9NH, United Kingdom
| | - X Kodji
- Cardiovascular Division, BHF Centre of Research Excellence & Centre of Integrative Biomedicine, King's College London, Waterloo Campus, London SE1 9NH, United Kingdom
| | - S D Brain
- Cardiovascular Division, BHF Centre of Research Excellence & Centre of Integrative Biomedicine, King's College London, Waterloo Campus, London SE1 9NH, United Kingdom
| |
Collapse
|
28
|
da Silva AO, Damaceno Alves A, Almeida DATD, Balogun SO, de Oliveira RG, Aires Aguiar A, Soares IM, Marson-Ascêncio PG, Ascêncio SD, de Oliveira Martins DT. Evaluation of anti-inflammatory and mechanism of action of extract of Macrosiphonia longiflora (Desf.) Müll. Arg. JOURNAL OF ETHNOPHARMACOLOGY 2014; 154:319-329. [PMID: 24681039 DOI: 10.1016/j.jep.2014.03.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 01/31/2014] [Accepted: 03/06/2014] [Indexed: 06/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Macrosiphonia longiflora (Desf.) Müll. Arg (Apocynaceae), popularly known as 'velame' and 'velame branco', is a native subshrub that grows in the Brazilian Cerrado. This plant is widely used in traditional medicine in the form of decoction and infusion, particularly as anti-inflammatory, depurative, anti-rheumatic, antisyphilitic and antiulcer remedy. There is no available information in the literature that has addressed its pharmacological activity and phytochemical analysis. AIM OF THE STUDY This study aimed to evaluate the anti-inflammatory pharmacological profile of the hydroethanolic extract of Macrosiphonia longiflora, using in vivo and in vitro acute inflammation experimental models, as well as investigate the roles of cytokines and nitric oxide in its mechanism of action, and including phytochemical analysis constitution of its hydroethanolic extract. MATERIALS AND METHODS Hydroethanolic (70%) extract of Macrosiphonia longiflora (HEMl) was prepared by maceration. The preliminary phytochemical analysis was performed according to procedures described in the literature. Selected secondary metabolites detected were quantified by spectrophotometry and high performance liquid chromatography (HPLC). Its cytotoxic potential in Chinese hamster ovary (CHO-k1) epithelial cell lines was evaluated using Alamar Blue. in vivo anti-inflammatory activity was evaluated with carrageenan- and dextran-induced paw edemas, carrageenan-induced pleurisy in rats and lipopolysaccharide (LPS)-induced peritonitis in mice. The in vitro anti-inflammatory activity was evaluated using RAW 264.7 cells stimulated with LPS and interferon (INF)-γ. Effects of HEMl on the inflammatory cytokines (IL-1β, IL-10, IL-17, INF-γ and TNF-α) concentrations in the peritoneal lavage were evaluated using commercial ELISA kits, while the Griess method was employed to determine nitric oxide (NO) concentrations in the peritoneal lavage, as well as in the supernatants of RAW 264.7 cells. RESULTS Preliminary phytochemical analysis, revealed the presence of phenolics compounds, terpenoids, alkaloids and flavonoids. Spectrophotometric analysis revealed the presence of relatively high content of phenolics and flavonoids in HEMl. HPLC analysis confirmed the presence of the quantified compounds and demonstrated the presence of ellagic acid in the detected matrix of compounds. HEMl appeared to be non-cytotoxic. It effectively inhibited (p<0.05) paw edema induced by carrageenan and dextran. Furthermore, HEMl also significantly reduced exudates volume and leukocyte migration in the carrageenan-induced pleurisy and LPS-induced peritonitis, neutrophils counts in LPS-induced peritonitis. HEMl also acts by effectively inhibiting the following inflammatory cytokines: IL-1β and IL-10 levels in the peritoneal lavage, but had no effect on IL-17 level in the peritonitis model. In addition, HEMl had no effect on the levels of tumor necrosis factor alpha (TNF-α) present in the peritoneal lavage and cells supernatants. The concentration of NO, as assessed by measurement of nitrite (NO2(-)), showed that pretreatment with HEMl reduced NO significantly in the peritoneal lavage and in RAW 264.7 cells co-stimulated with LPS and INF-γ. CONCLUSION The results obtained in this study indicate that HEMl possesses very low cytotoxic potential. In addition, it demonstrated a potent anti-inflammatory activity in both the in vivo and in vitro models of acute inflammation. The anti-inflammatory effect is partly related to the inhibition of IL-1β, IL-10, and nitric oxide releases, but independent of TNF-α and IL-17 modulation. Phytochemical analysis revealed the predominant presence of the flavonoids (naringin, rutin, myricetin, morin, quercetin, (±)-naringenin, and luteolin) and phenols (ellagic acid), which are possibly involved in the anti-inflammatory effect of HEMl. The current study provided supportive evidence for the popular use of HEMl in the treatment of inflammatory conditions, and shed more light on the possible roles of the inflammatory cytokines in its mechanisms of action as anti-inflammatory agent.
Collapse
Affiliation(s)
- Anísio Onório da Silva
- Department of Basic Sciences in Health, Faculty of Medicine, Federal University of Mato Grosso, Av. Fernando Correa da Costa, n. 2367, Cuiabá 78060-900, Mato Grosso, Brazil
| | - Aurea Damaceno Alves
- Department of Basic Sciences in Health, Faculty of Medicine, Federal University of Mato Grosso, Av. Fernando Correa da Costa, n. 2367, Cuiabá 78060-900, Mato Grosso, Brazil
| | - Danielle Ayr Tavares de Almeida
- Department of Basic Sciences in Health, Faculty of Medicine, Federal University of Mato Grosso, Av. Fernando Correa da Costa, n. 2367, Cuiabá 78060-900, Mato Grosso, Brazil
| | - Sikiru Olaitan Balogun
- Department of Basic Sciences in Health, Faculty of Medicine, Federal University of Mato Grosso, Av. Fernando Correa da Costa, n. 2367, Cuiabá 78060-900, Mato Grosso, Brazil
| | - Ruberlei Godinho de Oliveira
- Department of Basic Sciences in Health, Faculty of Medicine, Federal University of Mato Grosso, Av. Fernando Correa da Costa, n. 2367, Cuiabá 78060-900, Mato Grosso, Brazil
| | - Aline Aires Aguiar
- Research Laboratory of Natural Products (LPPN), Federal University of Tocantins, Av. NS15, Palmas, Tocantins 77020-210, Brazil
| | - Ilsamar Mendes Soares
- Research Laboratory of Natural Products (LPPN), Federal University of Tocantins, Av. NS15, Palmas, Tocantins 77020-210, Brazil
| | - Poliana Guerino Marson-Ascêncio
- Research Laboratory of Natural Products (LPPN), Federal University of Tocantins, Av. NS15, Palmas, Tocantins 77020-210, Brazil
| | - Sérgio Donizeti Ascêncio
- Research Laboratory of Natural Products (LPPN), Federal University of Tocantins, Av. NS15, Palmas, Tocantins 77020-210, Brazil
| | - Domingos Tabajara de Oliveira Martins
- Department of Basic Sciences in Health, Faculty of Medicine, Federal University of Mato Grosso, Av. Fernando Correa da Costa, n. 2367, Cuiabá 78060-900, Mato Grosso, Brazil.
| |
Collapse
|
29
|
Assas BM, Pennock JI, Miyan JA. Calcitonin gene-related peptide is a key neurotransmitter in the neuro-immune axis. Front Neurosci 2014; 8:23. [PMID: 24592205 PMCID: PMC3924554 DOI: 10.3389/fnins.2014.00023] [Citation(s) in RCA: 148] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 01/27/2014] [Indexed: 12/26/2022] Open
Abstract
The question of how the neural and immune systems interact in host defense is important, integrating a system that senses the whole body with one that protects. Understanding the mechanisms and routes of control could produce novel and powerful ways of promoting and enhancing normal functions as well as preventing or treating abnormal functions. Fragmentation of biological research into specialities has resulted in some failures in recognizing and understanding interactions across different systems and this is most striking across immunology, hematology, and neuroscience. This reductionist approach does not allow understanding of the in vivo orchestrated response generated through integration of all systems. However, many factors make the understanding of multisystem cross-talk in response to a threat difficult, for instance the nervous and immune systems share communication molecules and receptors for a wide range of physiological signals. But, it is clear that physical, hard-wired connections exist between the two systems, with the key link involving sensory, unmyelinated nerve fibers (c fibers) containing the neuropeptide calcitonin gene-related peptide (CGRP), and modified macrophages, mast cells and other immune and host defense cells in various locations throughout the body. In this review we will therefore focus on the induction of CGRP and its key role in the neuroimmune axis.
Collapse
Affiliation(s)
- Bakri M Assas
- Translational Medicine, Faculty of Medical and Human Sciences, The University of Manchester Manchester, UK ; Department of Immunology, Faculty of Applied Sciences, King Abdulaziz University Jeddah, Saudi Arabia
| | - Joanne I Pennock
- Translational Medicine, Faculty of Medical and Human Sciences, The University of Manchester Manchester, UK
| | - Jaleel A Miyan
- Neurosciences, Faculty of Life Sciences, The University of Manchester Manchester, UK
| |
Collapse
|
30
|
Lee JK, Jung JS, Park SH, Sim YB, Suh HW. Deficiency of alpha-calcitonin gene-related peptide induces inflammatory responses and lethality in sepsis. Cytokine 2013; 64:548-54. [PMID: 24021706 DOI: 10.1016/j.cyto.2013.07.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Revised: 07/08/2013] [Accepted: 07/30/2013] [Indexed: 01/15/2023]
Abstract
In the present study, we examined the role of alpha-calcitonin gene-related peptide (αCGRP) on expression of neuropeptides in the brain, inflammatory responses, and survival rate in septic shock condition. We examined expression of neuropeptides such as αCGRP, proopiomelanocortin (POMC), corticotrophin releasing hormone (CRH), and proenkephalin (ProENK) in the hippocampus and hypothalamus in C57BL/6 (WT) or αCGRP-/- (KO) mice subjected to sepsis. Cecal ligation and puncture (CLP) or lipopolysaccharide/D-galactosamine (LPS/D-GalN) treatment showed significant increases of hippocampal and hypothalamic αCGRP, POMC, CRH, and ProENK mRNA levels in WT mice, but not ProENK mRNA in the hypothalamus at 6h after on-set of sepsis. However, enhanced mRNA levels of POMC, CRH, and ProENK genes were not increased in the hippocampus and hypothalamus of CLP-subjected KO mice at 6h following sepsis. KO mice treated with LPS/D-GalN displayed a significant enhancement of plasma corticosterone, aspartate aminotransferase, and alanine aminotransferase levels compared to LPS/D-GalN treated WT mice at 12h after induction of sepsis. In addition, plasma levels of pro-inflammatory cytokines, such as IL-1β and TNF-α, were also further increased in KO mice compared to WT mice at 24h after CLP or LPS/D-GalN treatment. Interestingly, mRNA expressions of IL-6 and IL-10, anti-inflammatory cytokines, were synergistically enhanced in liver and lymph node of KO mice compared to WT mice at 6h after CLP. However, plasma level of IL-10 but not IL-6 was significantly decreased in KO mice compared to WT mice at 24h after CLP or LPS/D-GalN challenge. The survival rate of KO mice was significantly reduced compared to WT mice following mild (1 punch) and moderate (2 punch) CLP and LPS/D-GalN administration. Taken together, our findings suggest that the activation of αCGRP may induce other neuropeptides associated with immunomodulation at CNS level and modulate immune responses as enhancing anti-inflammatory cytokines and reducing pro-inflammatory cytokines during the sepsis.
Collapse
Affiliation(s)
- Jin-Koo Lee
- Department of Pharmacology, College of Medicine, Institute of Bio-Science Technology, Dankook University, Cheonan 330-714, Republic of Korea; Translational Research Center, Institute of Bio-Science Technology, Dankook University, Cheonan 330-714, Republic of Korea
| | | | | | | | | |
Collapse
|