1
|
Qin YJ, Zhang YC, Lin Y, Hong Y, Sun X, Xu F, Chen C. Autonomic nervous system imbalance in diabetic mouse choroids. Tissue Cell 2025; 94:102798. [PMID: 39970774 DOI: 10.1016/j.tice.2025.102798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 02/04/2025] [Accepted: 02/10/2025] [Indexed: 02/21/2025]
Abstract
PURPOSE We investigated neurohomeostasis and its possible underlying mechanisms in the choroids of diabetic mice. METHODS Streptozotocin-induced diabetic mice were used in this study. Choroidal nerve fiber alterations were observed via transmission electron microscopy (TEM). The levels of epinephrine and acetylcholine in the choroid were determined via ultra-high-performance liquid chromatography-tandem mass spectrometry. Tyrosine hydroxylase (TH), choline acetyl transferase (ChAT), and neuronal nitric oxide synthase (nNOS), vasoactive intestinal peptide (VIP), neuropeptide Y (NPY), and calcitonin gene-related peptide (CGRP) levels were evaluated by western blot or quantitative real-time polymerase chain reaction. In addition, immunofluorescence staining were used to analyze the changes in key enzymes (TH, dopamine beta-hydroxylase (DβH), and CGRP) in the superior cervical sympathetic ganglia (SCG) and trigeminal ganglia (TG). RESULTS TEM revealed a loose myelin sheath around nerve fibers, axon atrophy, and cytoplasmic vacuoles in Schwann cells in diabetic choroids. Lower epinephrine levels were observed in diabetic mice. Although no difference was found in acetylcholine level, the epinephrine-to-acetylcholine ratio was greatly decreased. Decreased TH and increased ChAT, nNOS, VIP, NPY, and CGRP were found in diabetic choroids. Fewer TH-positive and DβH-positive neuronal cells were found in the SCGs of diabetic mice. More CGRP-positive and fewer TH-positive neuronal cells were observed in the TG of diabetic mice. CONCLUSIONS Altered markers of the sympathetic, parasympathetic and sensory systems were present in diabetic mouse choroids. An imbalanced choroidal nervous system might explain the initiation of choroidal neovascularization in diabetic patients.
Collapse
Affiliation(s)
- Yuan-Jun Qin
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China; Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences & Department of Ophthalmology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi Zhuang Autonomous Region, China.
| | - Yong-Chao Zhang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China; Research Institute of Plastic and Aesthetic Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian Province, China.
| | - Yunru Lin
- Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences & Department of Ophthalmology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi Zhuang Autonomous Region, China; Zhuhai Aier Eye Hospital, Zhuhai, Guangdong Province, China.
| | - Yiyi Hong
- Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences & Department of Ophthalmology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi Zhuang Autonomous Region, China.
| | - Xufang Sun
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China.
| | - Fan Xu
- Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences & Department of Ophthalmology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi Zhuang Autonomous Region, China.
| | - Changzheng Chen
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China.
| |
Collapse
|
2
|
Ha WS, Chu MK. Altered immunity in migraine: a comprehensive scoping review. J Headache Pain 2024; 25:95. [PMID: 38844851 PMCID: PMC11157828 DOI: 10.1186/s10194-024-01800-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 05/30/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND The pathogenesis of migraine remains unclear; however, a large body of evidence supports the hypothesis that immunological mechanisms play a key role. Therefore, we aimed to review current studies on altered immunity in individuals with migraine during and outside attacks. METHODS We searched the PubMed database to investigate immunological changes in patients with migraine. We then added other relevant articles on altered immunity in migraine to our search. RESULTS Database screening identified 1,102 articles, of which 41 were selected. We added another 104 relevant articles. We found studies reporting elevated interictal levels of some proinflammatory cytokines, including IL-6 and TNF-α. Anti-inflammatory cytokines showed various findings, such as increased TGF-β and decreased IL-10. Other changes in humoral immunity included increased levels of chemokines, adhesion molecules, and matrix metalloproteinases; activation of the complement system; and increased IgM and IgA. Changes in cellular immunity included an increase in T helper cells, decreased cytotoxic T cells, decreased regulatory T cells, and an increase in a subset of natural killer cells. A significant comorbidity of autoimmune and allergic diseases with migraine was observed. CONCLUSIONS Our review summarizes the findings regarding altered humoral and cellular immunological findings in human migraine. We highlight the possible involvement of immunological mechanisms in the pathogenesis of migraine. However, further studies are needed to expand our knowledge of the exact role of immunological mechanisms in migraine pathogenesis.
Collapse
Affiliation(s)
- Woo-Seok Ha
- Department of Neurology, Severance Hospital, Yonsei University College of Medicine, 50-1, Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Min Kyung Chu
- Department of Neurology, Severance Hospital, Yonsei University College of Medicine, 50-1, Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
3
|
Kulalert W, Wells AC, Link VM, Lim AI, Bouladoux N, Nagai M, Harrison OJ, Kamenyeva O, Kabat J, Enamorado M, Chiu IM, Belkaid Y. The neuroimmune CGRP-RAMP1 axis tunes cutaneous adaptive immunity to the microbiota. Proc Natl Acad Sci U S A 2024; 121:e2322574121. [PMID: 38451947 PMCID: PMC10945812 DOI: 10.1073/pnas.2322574121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 01/22/2024] [Indexed: 03/09/2024] Open
Abstract
The somatosensory nervous system surveils external stimuli at barrier tissues, regulating innate immune cells under infection and inflammation. The roles of sensory neurons in controlling the adaptive immune system, and more specifically immunity to the microbiota, however, remain elusive. Here, we identified a mechanism for direct neuroimmune communication between commensal-specific T lymphocytes and somatosensory neurons mediated by the neuropeptide calcitonin gene-related peptide (CGRP) in the skin. Intravital imaging revealed that commensal-specific T cells are in close proximity to cutaneous nerve fibers in vivo. Correspondingly, we observed upregulation of the receptor for the neuropeptide CGRP, RAMP1, in CD8+ T lymphocytes induced by skin commensal colonization. The neuroimmune CGRP-RAMP1 signaling axis functions in commensal-specific T cells to constrain Type 17 responses and moderate the activation status of microbiota-reactive lymphocytes at homeostasis. As such, modulation of neuroimmune CGRP-RAMP1 signaling in commensal-specific T cells shapes the overall activation status of the skin epithelium, thereby impacting the outcome of responses to insults such as wounding. The ability of somatosensory neurons to control adaptive immunity to the microbiota via the CGRP-RAMP1 axis underscores the various layers of regulation and multisystem coordination required for optimal microbiota-reactive T cell functions under steady state and pathology.
Collapse
Affiliation(s)
- Warakorn Kulalert
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Alexandria C. Wells
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Verena M. Link
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Ai Ing Lim
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Nicolas Bouladoux
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
- National Institute of Allergy and Infectious Diseases Microbiome Program, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Motoyoshi Nagai
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Oliver J. Harrison
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Olena Kamenyeva
- Biological Imaging Section, Research Technology Branch, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Juraj Kabat
- Biological Imaging Section, Research Technology Branch, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Michel Enamorado
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
- Kimberly and Eric J. Waldman Department of Dermatology, Mark Lebwohl Center for Neuroinflammation and Sensation, Marc and Jennifer Lipschultz Precision Immunology Institute, and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY10029
| | - Isaac M. Chiu
- Department of Immunology, Harvard Medical School, Boston, MA02115
| | - Yasmine Belkaid
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
- National Institute of Allergy and Infectious Diseases Microbiome Program, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
- Unite Metaorganisme, Immunology Department, Pasteur Institute, 75015 Paris, France
| |
Collapse
|
4
|
Kulalert W, Wells AC, Link VM, Lim AI, Bouladoux N, Nagai M, Harrison OJ, Kamenyeva O, Kabat J, Enamorado M, Chiu IM, Belkaid Y. The neuroimmune CGRP-RAMP1 axis tunes cutaneous adaptive immunity to the microbiota. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.26.573358. [PMID: 38234748 PMCID: PMC10793430 DOI: 10.1101/2023.12.26.573358] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
The somatosensory nervous system surveils external stimuli at barrier tissues, regulating innate immune cells under infection and inflammation. The roles of sensory neurons in controlling the adaptive immune system, and more specifically immunity to the microbiota, however, remain elusive. Here, we identified a novel mechanism for direct neuroimmune communication between commensal-specific T lymphocytes and somatosensory neurons mediated by the neuropeptide Calcitonin Gene-Related Peptide (CGRP) in the skin. Intravital imaging revealed that commensal-specific T cells are in close proximity to cutaneous nerve fibers in vivo . Correspondingly, we observed upregulation of the receptor for the neuropeptide CGRP, RAMP1, in CD8 + T lymphocytes induced by skin commensal colonization. Neuroimmune CGRP-RAMP1 signaling axis functions in commensal-specific T cells to constrain Type 17 responses and moderate the activation status of microbiota-reactive lymphocytes at homeostasis. As such, modulation of neuroimmune CGRP-RAMP1 signaling in commensal-specific T cells shapes the overall activation status of the skin epithelium, thereby impacting the outcome of responses to insults such as wounding. The ability of somatosensory neurons to control adaptive immunity to the microbiota via the CGRP-RAMP1 axis underscores the various layers of regulation and multisystem coordination required for optimal microbiota-reactive T cell functions under steady state and pathology. Significance statement Multisystem coordination at barrier surfaces is critical for optimal tissue functions and integrity, in response to microbial and environmental cues. In this study, we identified a novel neuroimmune crosstalk mechanism between the sensory nervous system and the adaptive immune response to the microbiota, mediated by the neuropeptide CGRP and its receptor RAMP1 on skin microbiota-induced T lymphocytes. The neuroimmune CGPR-RAMP1 axis constrains adaptive immunity to the microbiota and overall limits the activation status of the skin epithelium, impacting tissue responses to wounding. Our study opens the door to a new avenue to modulate adaptive immunity to the microbiota utilizing neuromodulators, allowing for a more integrative and tailored approach to harnessing microbiota-induced T cells to promote barrier tissue protection and repair.
Collapse
|
5
|
Kruk L, Mamtimin M, Braun A, Anders HJ, Andrassy J, Gudermann T, Mammadova-Bach E. Inflammatory Networks in Renal Cell Carcinoma. Cancers (Basel) 2023; 15:cancers15082212. [PMID: 37190141 DOI: 10.3390/cancers15082212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/23/2023] [Accepted: 04/04/2023] [Indexed: 05/17/2023] Open
Abstract
Cancer-associated inflammation has been established as a hallmark feature of almost all solid cancers. Tumor-extrinsic and intrinsic signaling pathways regulate the process of cancer-associated inflammation. Tumor-extrinsic inflammation is triggered by many factors, including infection, obesity, autoimmune disorders, and exposure to toxic and radioactive substances. Intrinsic inflammation can be induced by genomic mutation, genome instability and epigenetic remodeling in cancer cells that promote immunosuppressive traits, inducing the recruitment and activation of inflammatory immune cells. In RCC, many cancer cell-intrinsic alterations are assembled, upregulating inflammatory pathways, which enhance chemokine release and neoantigen expression. Furthermore, immune cells activate the endothelium and induce metabolic shifts, thereby amplifying both the paracrine and autocrine inflammatory loops to promote RCC tumor growth and progression. Together with tumor-extrinsic inflammatory factors, tumor-intrinsic signaling pathways trigger a Janus-faced tumor microenvironment, thereby simultaneously promoting or inhibiting tumor growth. For therapeutic success, it is important to understand the pathomechanisms of cancer-associated inflammation, which promote cancer progression. In this review, we describe the molecular mechanisms of cancer-associated inflammation that influence cancer and immune cell functions, thereby increasing tumor malignancy and anti-cancer resistance. We also discuss the potential of anti-inflammatory treatments, which may provide clinical benefits in RCCs and possible avenues for therapy and future research.
Collapse
Affiliation(s)
- Linus Kruk
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilian-University, 80336 Munich, Germany
- Division of Nephrology, Department of Medicine IV, Hospital of the Ludwig-Maximilian-University, 80336 Munich, Germany
| | - Medina Mamtimin
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilian-University, 80336 Munich, Germany
- Division of Nephrology, Department of Medicine IV, Hospital of the Ludwig-Maximilian-University, 80336 Munich, Germany
| | - Attila Braun
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilian-University, 80336 Munich, Germany
| | - Hans-Joachim Anders
- Division of Nephrology, Department of Medicine IV, Hospital of the Ludwig-Maximilian-University, 80336 Munich, Germany
| | - Joachim Andrassy
- Division of General, Visceral, Vascular and Transplant Surgery, Hospital of LMU, 81377 Munich, Germany
| | - Thomas Gudermann
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilian-University, 80336 Munich, Germany
- German Center for Lung Research (DZL), 80336 Munich, Germany
| | - Elmina Mammadova-Bach
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilian-University, 80336 Munich, Germany
- Division of Nephrology, Department of Medicine IV, Hospital of the Ludwig-Maximilian-University, 80336 Munich, Germany
| |
Collapse
|
6
|
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
|
7
|
Staurengo-Ferrari L, Deng L, Chiu IM. Interactions between nociceptor sensory neurons and microbial pathogens in pain. Pain 2022; 163:S57-S68. [PMID: 36252233 PMCID: PMC9586460 DOI: 10.1097/j.pain.0000000000002721] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/21/2022] [Indexed: 02/07/2023]
Affiliation(s)
- Larissa Staurengo-Ferrari
- Harvard Medical School, Blavatnik Institute, Department of Immunology, Boston, Massachusetts, United States of America
| | - Liwen Deng
- Harvard Medical School, Blavatnik Institute, Department of Immunology, Boston, Massachusetts, United States of America
| | - Isaac M. Chiu
- Harvard Medical School, Blavatnik Institute, Department of Immunology, Boston, Massachusetts, United States of America
| |
Collapse
|
8
|
Asiedu K. Role of ocular surface neurobiology in neuronal-mediated inflammation in dry eye disease. Neuropeptides 2022; 95:102266. [PMID: 35728484 DOI: 10.1016/j.npep.2022.102266] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 01/18/2023]
Abstract
Inflammation is the consequence of dry eye disease regardless of its etiology. Several injurious or harmless processes to the ocular surface neurons promote ocular surface neurogenic inflammation, leading to the vicious cycle of dry eye disease. These processes include the regular release of neuromediators during the conduction of ocular surface sensations, hyperosmolarity-induced ocular surface neuronal damage, neuro-regenerative activities, and neuronal-mediated dendritic cell activities. Neurogenic inflammation appears to be the main culprit, instigating the self-perpetuating inflammation observed in patients with dry eye disease.
Collapse
Affiliation(s)
- Kofi Asiedu
- School of Optometry & Vision Science, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia.
| |
Collapse
|
9
|
Du Y, Chen J, Shen L, Wang B. TRP channels in inflammatory bowel disease: potential therapeutic targets. Biochem Pharmacol 2022; 203:115195. [DOI: 10.1016/j.bcp.2022.115195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 12/23/2022]
|
10
|
Flayer CH, Sokol CL. Sensory neurons control the functions of dendritic cells to guide allergic immunity. Curr Opin Immunol 2022; 74:85-91. [PMID: 34808584 PMCID: PMC8901476 DOI: 10.1016/j.coi.2021.10.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 10/20/2021] [Accepted: 10/24/2021] [Indexed: 02/03/2023]
Abstract
Dendritic cells of the innate immune system and sensory neurons of the peripheral nervous system are embedded in barrier tissues and gather information about an organisms' environment. While the mechanisms by which dendritic cells recognize and initiate adaptive immune responses to pathogens is well defined, how they sense allergens is poorly understood. Indeed, allergens induce dendritic cell maturation and migration in vivo, but not in vitro. How are adaptive immune responses to allergens initiated if dendritic cells do not directly sense allergens? Sensory neurons release neuropeptides within minutes of allergen exposure. Recent evidence demonstrated that while neuropeptides modify dendritic cell function during pathogen responses, they are required for dendritic cell function during allergic responses. These emerging studies suggest that sensory neurons do not just pass information along to the central nervous system, but also to dendritic cells, particularly during the initiation of adaptive immunity to allergens.
Collapse
Affiliation(s)
| | - Caroline L Sokol
- Corresponding author: , 149 13th St Room 8103, Charlestown, MA 02129
| |
Collapse
|
11
|
Affiliation(s)
- Liwen Deng
- Harvard Medical School, Blavatnik Institute, Department of Immunology, Boston, Massachusetts, United States of America
| | - Isaac M. Chiu
- Harvard Medical School, Blavatnik Institute, Department of Immunology, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
12
|
Meerschaert KA, Adelman PC, Friedman RL, Albers KM, Koerber HR, Davis BM. Unique Molecular Characteristics of Visceral Afferents Arising from Different Levels of the Neuraxis: Location of Afferent Somata Predicts Function and Stimulus Detection Modalities. J Neurosci 2020; 40:7216-7228. [PMID: 32817244 PMCID: PMC7534907 DOI: 10.1523/jneurosci.1426-20.2020] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/30/2020] [Accepted: 08/07/2020] [Indexed: 02/07/2023] Open
Abstract
Viscera receive innervation from sensory ganglia located adjacent to multiple levels of the brainstem and spinal cord. Here we examined whether molecular profiling could be used to identify functional clusters of colon afferents from thoracolumbar (TL), lumbosacral (LS), and nodose ganglia (NG) in male and female mice. Profiling of TL and LS bladder afferents was also performed. Visceral afferents were back-labeled using retrograde tracers injected into proximal and distal regions of colon or bladder, followed by single-cell qRT-PCR and analysis via an automated hierarchical clustering method. Genes were chosen for assay (32 for bladder; 48 for colon) based on their established role in stimulus detection, regulation of sensitivity/function, or neuroimmune interaction. A total of 132 colon afferents (from NG, TL, and LS ganglia) and 128 bladder afferents (from TL and LS ganglia) were analyzed. Retrograde labeling from the colon showed that NG and TL afferents innervate proximal and distal regions of the colon, whereas 98% of LS afferents only project to distal regions. There were clusters of colon and bladder afferents, defined by mRNA profiling, that localized to either TL or LS ganglia. Mixed TL/LS clustering also was found. In addition, transcriptionally, NG colon afferents were almost completely segregated from colon TL and LS neurons. Furthermore, colon and bladder afferents expressed genes at similar levels, although different gene combinations defined the clusters. These results indicate that genes implicated in both homeostatic regulation and conscious sensations are found at all anatomic levels, suggesting that afferents from different portions of the neuraxis have overlapping functions.SIGNIFICANCE STATEMENT Visceral organs are innervated by sensory neurons whose cell bodies are located in multiple ganglia associated with the brainstem and spinal cord. For the colon, this overlapping innervation is proposed to facilitate visceral sensation and homeostasis, where sensation and pain are mediated by spinal afferents and fear and anxiety (the affective aspects of visceral pain) are the domain of nodose afferents. The transcriptomic analysis performed here reveals that genes implicated in both homeostatic regulation and pain are found in afferents across all ganglia types, suggesting that conscious sensation and homeostatic regulation are the result of convergence, and not segregation, of sensory input.
Collapse
Affiliation(s)
- Kimberly A Meerschaert
- Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | | | - Robert L Friedman
- Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Kathryn M Albers
- Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - H Richard Koerber
- Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Brian M Davis
- Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| |
Collapse
|
13
|
Tsuru S, Ito Y, Matsuda H, Hosono K, Inoue T, Nakamoto S, Kurashige C, Mishima T, Tsujikawa K, Okamoto H, Majima M. RAMP1 signaling in immune cells regulates inflammation-associated lymphangiogenesis. J Transl Med 2020; 100:738-750. [PMID: 31911634 DOI: 10.1038/s41374-019-0364-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 12/15/2019] [Accepted: 12/17/2019] [Indexed: 02/07/2023] Open
Abstract
Calcitonin gene-related peptide (CGRP) regulates inflammation via signaling through receptor activity-modifying protein (RAMP) 1. Here, we investigated the role of RAMP1 signaling in growth of lymphatic vessels during inflammation. Lymphangiogenesis in the diaphragm of RAMP1-deficient (-/-) mice or their wild-type (WT) counterparts was induced by repeated intraperitoneal injection of lipopolysaccharide (LPS). Compared with WT mice, LPS-induced lymphangiogenesis in RAMP1-/- mice was suppressed. This was accompanied by the reduced expression of vascular endothelial growth factor (VEGF)-C and VEGF-D. The number of CD4+ cells in diaphragm tissue from WT mice was greater than RAMP1-/- mice. Removing CD4+ cells attenuated lymphangiogenesis and expression of VEGF-C and VEGF-D. CD4+ cells isolated from RAMP1-/- mice exhibited reduced expression of VEGF-C and VEGF-D. The number of CD11b+ cells from RAMP1-/- mice was higher than WT mice and was associated with the upregulated expression of genes related to pro-inflammatory macrophage phenotype and downregulation of reparative macrophage phenotype-related expression. When fluorescein isothiocyanate (FITC)-dextran was injected into the peritoneal cavity, the amount of residual FITC-dextran in WT mice was lower than that in RAMP1-/- mice. The present results suggest that RAMP1 signaling in immune cells plays a critical role in inflammation-related lymphangiogenesis; therefore, it represents a novel target for controlling lymphangiogenesis.
Collapse
Affiliation(s)
- Seri Tsuru
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Kanagawa, 252-0374, Japan.,Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan.,Department of Anesthesiology, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan
| | - Yoshiya Ito
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Kanagawa, 252-0374, Japan.,Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan
| | - Hiromi Matsuda
- Department of Anesthesiology, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan
| | - Kanako Hosono
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Kanagawa, 252-0374, Japan.,Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan
| | - Tomoyoshi Inoue
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Kanagawa, 252-0374, Japan
| | - Shuji Nakamoto
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Kanagawa, 252-0374, Japan
| | - Chie Kurashige
- Department of Anesthesiology, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan
| | - Toshiaki Mishima
- Department of Cardiovascular Surgery, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan
| | - Kazutake Tsujikawa
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Hirotsugu Okamoto
- Department of Anesthesiology, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan
| | - Masataka Majima
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Kanagawa, 252-0374, Japan. .,Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan.
| |
Collapse
|
14
|
Chen Y, Mu J, Zhu M, Mukherjee A, Zhang H. Transient Receptor Potential Channels and Inflammatory Bowel Disease. Front Immunol 2020; 11:180. [PMID: 32153564 PMCID: PMC7044176 DOI: 10.3389/fimmu.2020.00180] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 01/23/2020] [Indexed: 02/05/2023] Open
Abstract
The transient receptor potential (TRP) cation channels are present in abundance across the gastrointestinal (GI) tract, serving as detectors for a variety of stimuli and secondary transducers for G-protein coupled receptors. The activation of TRP channels triggers neurogenic inflammation with related neuropeptides and initiates immune reactions by extra-neuronally regulating immune cells, contributing to the GI homeostasis. However, under pathological conditions, such as inflammatory bowel disease (IBD), TRP channels are involved in intestinal inflammation. An increasing number of human and animal studies have indicated that TRP channels are correlated to the visceral hypersensitivity (VHS) and immune pathogenesis in IBD, leading to an exacerbation or amelioration of the VHS or intestinal inflammation. Thus, TRP channels are a promising target for novel therapeutic methods for IBD. In this review, we comprehensively summarize the functions of TRP channels, especially their potential roles in immunity and IBD. Additionally, we discuss the contradictory findings of prior studies and offer new insights with regard to future research.
Collapse
Affiliation(s)
- Yiding Chen
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
- Centre for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Jingxi Mu
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
- Centre for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Min Zhu
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
- Centre for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu, China
| | | | - Hu Zhang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
- Centre for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
15
|
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
|
16
|
CGRP Induces Differential Regulation of Cytokines from Satellite Glial Cells in Trigeminal Ganglia and Orofacial Nociception. Int J Mol Sci 2019; 20:ijms20030711. [PMID: 30736422 PMCID: PMC6386987 DOI: 10.3390/ijms20030711] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 01/29/2019] [Accepted: 02/04/2019] [Indexed: 01/29/2023] Open
Abstract
Neuron-glia interactions contribute to pain initiation and sustainment. Intra-ganglionic (IG) secretion of calcitonin gene-related peptide (CGRP) in the trigeminal ganglion (TG) modulates pain transmission through neuron-glia signaling, contributing to various orofacial pain conditions. The present study aimed to investigate the role of satellite glial cells (SGC) in TG in causing cytokine-related orofacial nociception in response to IG administration of CGRP. For that purpose, CGRP alone (10 μL of 10−5 M), Minocycline (5 μL containing 10 μg) followed by CGRP with one hour gap (Min + CGRP) were administered directly inside the TG in independent experiments. Rats were evaluated for thermal hyperalgesia at 6 and 24 h post-injection using an operant orofacial pain assessment device (OPAD) at three temperatures (37, 45 and 10 °C). Quantitative real-time PCR was performed to evaluate the mRNA expression of IL-1β, IL-6, TNF-α, IL-1 receptor antagonist (IL-1RA), sodium channel 1.7 (NaV 1.7, for assessment of neuronal activation) and glial fibrillary acidic protein (GFAP, a marker of glial activation). The cytokines released in culture media from purified glial cells were evaluated using antibody cytokine array. IG CGRP caused heat hyperalgesia between 6–24 h (paired-t test, p < 0.05). Between 1 to 6 h the mRNA and protein expressions of GFAP was increased in parallel with an increase in the mRNA expression of pro-inflammatory cytokines IL-1β and anti-inflammatory cytokine IL-1RA and NaV1.7 (one-way ANOVA followed by Dunnett’s post hoc test, p < 0.05). To investigate whether glial inhibition is useful to prevent nociception symptoms, Minocycline (glial inhibitor) was administered IG 1 h before CGRP injection. Minocycline reversed CGRP-induced thermal nociception, glial activity, and down-regulated IL-1β and IL-6 cytokines significantly at 6 h (t-test, p < 0.05). Purified glial cells in culture showed an increase in release of 20 cytokines after stimulation with CGRP. Our findings demonstrate that SGCs in the sensory ganglia contribute to the occurrence of pain via cytokine expression and that glial inhibition can effectively control the development of nociception.
Collapse
|
17
|
Neuropeptides SP and CGRP Diminish the Moraxella catarrhalis Outer Membrane Vesicle- (OMV-) Triggered Inflammatory Response of Human A549 Epithelial Cells and Neutrophils. Mediators Inflamm 2018; 2018:4847205. [PMID: 30174554 PMCID: PMC6098883 DOI: 10.1155/2018/4847205] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 06/07/2018] [Accepted: 06/30/2018] [Indexed: 12/30/2022] Open
Abstract
Neuropeptides such as substance P (SP) and calcitonin gene-related peptide (CGRP) play both pro- and anti-inflammatory activities and are produced during infection and inflammation. Moraxella catarrhalis is one of the leading infectious agents responsible for inflammatory exacerbation in chronic obstructive pulmonary disease (COPD). Since the airway inflammation in COPD is connected with activation of both epithelial cells and accumulated neutrophils, in this study we determined the in vitro effects of neuropeptides on the inflammatory potential of these cells in response to M. catarrhalis outer membrane vesicle (OMV) stimulant. The various OMV-mediated proinflammatory effects were demonstrated. Next, using hBD-2-pGL4[luc2] plasmid with luciferase reporter gene, SP and CGRP were shown to inhibit the IL-1β-dependent expression of potent neutrophil chemoattractant, hBD-2 defensin, in transfected A549 epithelial cells (type II alveolar cells) upon OMV stimulation. Both neuropeptides exerted antiapoptotic activity through rescuing a significant fraction of A549 cells from OMV-induced cell death and apoptosis. Finally, CGRP caused an impairment of specific but not azurophilic granule exocytosis from neutrophils as shown by evaluation of gelatinase-associated lipocalin (NGAL) or CD66b expression and elastase release, respectively. Concluding, these findings suggest that SP and CGRP mediate the dampening of proinflammatory action triggered by M. catarrhalis OMVs towards cells engaged in lung inflammation in vitro.
Collapse
|
18
|
Watson G, Ronai ZA, Lau E. ATF2, a paradigm of the multifaceted regulation of transcription factors in biology and disease. Pharmacol Res 2017; 119:347-357. [PMID: 28212892 PMCID: PMC5457671 DOI: 10.1016/j.phrs.2017.02.004] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 02/01/2017] [Accepted: 02/02/2017] [Indexed: 01/16/2023]
Abstract
Stringent transcriptional regulation is crucial for normal cellular biology and organismal development. Perturbations in the proper regulation of transcription factors can result in numerous pathologies, including cancer. Thus, understanding how transcription factors are regulated and how they are dysregulated in disease states is key to the therapeutic targeting of these factors and/or the pathways that they regulate. Activating transcription factor 2 (ATF2) has been studied in a number of developmental and pathological conditions. Recent findings have shed light on the transcriptional, post-transcriptional, and post-translational regulatory mechanisms that influence ATF2 function, and thus, the transcriptional programs coordinated by ATF2. Given our current knowledge of its multiple levels of regulation and function, ATF2 represents a paradigm for the mechanistic complexity that can regulate transcription factor function. Thus, increasing our understanding of the regulation and function of ATF2 will provide insights into fundamental regulatory mechanisms that influence how cells integrate extracellular and intracellular signals into a genomic response through transcription factors. Characterization of ATF2 dysfunction in the context of pathological conditions, particularly in cancer biology and response to therapy, will be important in understanding how pathways controlled by ATF2 or other transcription factors might be therapeutically exploited. In this review, we provide an overview of the currently known upstream regulators and downstream targets of ATF2.
Collapse
Affiliation(s)
- Gregory Watson
- Department of Tumor Biology and Program in Chemical Biology and Molecular Medicine, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Ze'ev A Ronai
- Tumor Initiation and Maintenance Program, Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA; Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, 3109601, Israel
| | - Eric Lau
- Department of Tumor Biology and Program in Chemical Biology and Molecular Medicine, H. Lee Moffitt Cancer Center, Tampa, FL, USA.
| |
Collapse
|
19
|
Lee HW, Jie HB, Bollyky PL, Sarracino D, Kim TS, Wilson BS. Role of dendritic cell maturation factors produced by human invariant NKT cells in immune tolerance. J Leukoc Biol 2016; 101:989-1003. [PMID: 27837018 DOI: 10.1189/jlb.1a0416-164rrr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 10/04/2016] [Accepted: 10/27/2016] [Indexed: 01/12/2023] Open
Abstract
In this study, we used the culture supernatant of iNKT cells to identify human myeloid DC maturation factors produced by human CD4+ iNKT cells. S100A8 had a strong maturation effect. Notably, the recombinant S100A8 protein displayed properties of DC maturation functioning, and the induction of DC differentiation by both the purified and the recombinant protein were blocked by anti-S100A8 and anti-TLR-4 mAbs. DC differentiation induced by anti-major histocompatibility complex class II/CD1d Ab, S100A8, or both was qualitatively indistinguishable from that induced by the coculture of DCs and iNKT cells or via culture supplementation with supernatants from activated CD4+ iNKT cells. S100A8 also induced CD4+/CD25+/Foxp3+ Treg cells from naïve T cells. S100A8 may contribute to DC differentiation by elevating transcription factors or activating transcription factor-2, heat shock factor-1, or both, in mature DCs. S100A8 is a novel candidate iNKT cell-dependent DC maturation factor.
Collapse
Affiliation(s)
- Hyeong-Woo Lee
- Departments of Tropical Medicine and Parasitology, Inha University School of Medicine, Incheon, Republic of Korea.,Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, Florida
| | - Hyun Bae Jie
- OncoMed Pharmaceuticals, Inc., Redwood City, California
| | - Paul L Bollyky
- Division of Infectious Diseases, Stanford University Medical Center, Stanford, California; and
| | - David Sarracino
- Thermo Fisher Scientific Biomarkers Research Initiatives in Mass Spectrometry (BRIMS) Center, Cambridge, Massachusetts
| | - Tong-Soo Kim
- Departments of Tropical Medicine and Parasitology, Inha University School of Medicine, Incheon, Republic of Korea;
| | - Brian S Wilson
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, Florida;
| |
Collapse
|
20
|
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: 674] [Impact Index Per Article: 74.9] [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
|
21
|
Allais L, De Smet R, Verschuere S, Talavera K, Cuvelier CA, Maes T. Transient Receptor Potential Channels in Intestinal Inflammation: What Is the Impact of Cigarette Smoking? Pathobiology 2016; 84:1-15. [DOI: 10.1159/000446568] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 05/02/2016] [Indexed: 11/19/2022] Open
|
22
|
TRPM8 on mucosal sensory nerves regulates colitogenic responses by innate immune cells via CGRP. Mucosal Immunol 2015; 8:491-504. [PMID: 25269705 PMCID: PMC4382463 DOI: 10.1038/mi.2014.82] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 08/07/2014] [Indexed: 02/04/2023]
Abstract
TRPM8 is the molecular sensor for cold; however, the physiological role of TRPM8+ neurons at mucosal surfaces is unclear. Here we evaluated the distribution and peptidergic properties of TRPM8+ fibers in naive and inflamed colons, as well as their role in mucosal inflammation. We found that Trpm8(-/-) mice were hypersusceptible to dextran sodium sulfate (DSS)-induced colitis, and that Trpm8(-/-) CD11c+ DCs (dendritic cells) showed hyperinflammatory responses to toll-like receptor (TLR) stimulation. This was phenocopied in calcitonin gene-related peptide (CGRP) receptor-deficient mice, but not in substance P receptor-deficient mice, suggesting a functional link between TRPM8 and CGRP. The DSS phenotype of CGRP receptor-deficient mice could be adoptively transferred to wild-type (WT) mice, suggesting that CGRP suppresses the colitogenic activity of bone marrow-derived cells. TRPM8+ mucosal fibers expressed CGRP in human and mouse colon. Furthermore, neuronal CGRP contents were increased in colons from naive and DSS-treated Trpm8(-/-) mice, suggesting deficient CGRP release in the absence of TRPM8 triggering. Finally, treatment of Trpm8(-/-) mice with CGRP reversed their hyperinflammatory phenotype. These results suggest that TRPM8 signaling in mucosal sensory neurons is indispensable for the regulation of innate inflammatory responses via the neuropeptide CGRP.
Collapse
|
23
|
Yang W, Xv M, Yang WC, Wang N, Zhang XZ, Li WZ. Exogenous α-calcitonin gene-related peptide attenuates lipopolysaccharide-induced acute lung injury in rats. Mol Med Rep 2015; 12:2181-8. [PMID: 25892548 DOI: 10.3892/mmr.2015.3620] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 10/24/2014] [Indexed: 11/06/2022] Open
Abstract
α-Calcitonin gene-related peptide (α-CGRP) is a 37 amino-acid neuropeptide that is primarily released from C-type sensory neurons. α-CGRP exerts multiple modulatory effects on immune responses and visceral organ function, but the role of exogenous α-CGRP in lipopolysaccharide (LPS)-induced acute lung injury (ALI) has remained to be elucidated. Forty-eight rats were randomized to receive continuous intraperitoneal infusion of α-CGRP (0.4 μg/kg/min) or normal saline for 30 min, followed by intratracheal injection of 0.5 mg/kg LPS or saline. There were four groups of animals: The saline-saline (S-S) group; the saline-α-CGRP (S-C) group; the LPS-saline (L-S) group and the LPS-α-CGRP (L-C) group. Mean arterial pressure and arterial blood gases were assessed prior to α-CGRP and LPS administration and every hour following LPS treatment. After 4 h, bronchoalveolar lavage was performed and used to assess total cell count and levels of tumor necrosis factor-α, interleukin-1β, intracellular cell adhesion molecule 1 and macrophage inflammatory protein 2. Lung tissue was also collected for assessing wet-to-dry (W/D) ratio, histology and Evans blue (EB) dye extravasation. Pulmonary α-CGRP concentration and α-CGRP receptor expression were also examined, and inducible cyclic adenosine monophosphate early repressor (ICER) and TNF-α mRNA expression levels were measured. Treatment with exogenous α-CGRP improved oxygenation during LPS-induced ALI. Correspondingly, histological injury, total cell count, inflammatory cytokine levels, W/D ratio and EB dye extravasation were also significantly reduced. α-CGRP receptor 1 expression was noted in pulmonary endothelial cells and alveolar macrophages and α-CGRP receptor expression levels were decreased during ALI, whereas pulmonary α-CGRP expression was continuously increased. Furthermore, exogenous α-CGRP induced upregulation of ICER during LPS-induced ALI. In conclusion, exogenous α-CGRP improved oxygenation and ameliorated lung damage in LPS-induced ALI, and these effects were associated with the upregulation of ICER.
Collapse
Affiliation(s)
- Wang Yang
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Meng Xv
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Wan Chao Yang
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Nan Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Xue Zhong Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Wen Zhi Li
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
24
|
Körner C, Kuchenbuch T, Pfeil U, Jung K, Padberg W, Kummer W, Mühlfeld C, Grau V. Low-dose adrenomedullin-2/intermedin(8-47) reduces pulmonary ischemia/reperfusion injury. Peptides 2014; 62:49-54. [PMID: 25290159 DOI: 10.1016/j.peptides.2014.09.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 09/25/2014] [Accepted: 09/25/2014] [Indexed: 02/07/2023]
Abstract
Adrenomedullin-2/intermedin stabilizes the pulmonary microvascular barrier challenged by application of thrombin ex vivo and by experimental ventilation in vivo. Here, we test the hypothesis that adrenomedullin-2/intermedin(8-47) protects mouse lungs from ischemia/reperfusion injury in vivo. C57BL/6 mice were anesthetized, intubated, ventilated, and heparinized. Blood vessels and the main bronchus of the left lung were clamped for 90min. Thereafter, lungs were reperfused for 120min. Five min before clamping and before reperfusion, mice obtained intravenous injections of adrenomedullin-2/intermedin(8-47). After reperfusion, mice were sacrificed and bronchoalveolar lavage of the left and the right lung was performed separately. The integrity of the blood-air barrier was investigated by electron microscopy using stereological methods. In response to ischemia/reperfusion injury, intraalveolar leukocytes accumulated in the ischemic lung. Two applications of 10ng/kg body weight adrenomedullin-2/intermedin(8-47) dramatically reduced leukocyte infiltration to about 15% (p≤0.001). Also the proportion of the subpopulation of neutrophil granulocytes decreased (12% vs 5%, p=0.013). Electron microscopy revealed a protection of the blood-air barrier by adrenomedullin-2/intermedin(8-47). Adrenomedullin-2/intermedin(8-47) ameliorates early ischemia/reperfusion injury in mouse lungs by protecting the integrity of the blood-air barrier and by potently reducing leukocyte influx into the alveolar space. Adrenomedullin-2/intermedin(8-47) might be of therapeutic interest in lung transplantation and cardiopulmonary bypass.
Collapse
Affiliation(s)
- Christian Körner
- Laboratory of Experimental Surgery, Department of General and Thoracic Surgery, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany.
| | - Tim Kuchenbuch
- Laboratory of Experimental Surgery, Department of General and Thoracic Surgery, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Uwe Pfeil
- Institute for Anatomy and Cell Biology, Justus-Liebig-University Giessen, UGMLC, Member of the DZL, Giessen, Germany
| | - Kristina Jung
- Institute for Anatomy and Cell Biology, Justus-Liebig-University Giessen, UGMLC, Member of the DZL, Giessen, Germany
| | - Winfried Padberg
- Laboratory of Experimental Surgery, Department of General and Thoracic Surgery, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Wolfgang Kummer
- Institute for Anatomy and Cell Biology, Justus-Liebig-University Giessen, UGMLC, Member of the DZL, Giessen, Germany
| | - Christian Mühlfeld
- Institute for Anatomy and Cell Biology, Justus-Liebig-University Giessen, UGMLC, Member of the DZL, Giessen, Germany; Institute of Functional and Applied Anatomy, Hannover Medical School, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the DZL, Hannover, Germany
| | - Veronika Grau
- Laboratory of Experimental Surgery, Department of General and Thoracic Surgery, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| |
Collapse
|
25
|
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
|
26
|
Lou G, Ma X, Fu X, Meng Z, Zhang W, Wang YD, Van Ness C, Yu D, Xu R, Huang W. GPBAR1/TGR5 mediates bile acid-induced cytokine expression in murine Kupffer cells. PLoS One 2014; 9:e93567. [PMID: 24755711 PMCID: PMC3995640 DOI: 10.1371/journal.pone.0093567] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 03/06/2014] [Indexed: 12/21/2022] Open
Abstract
GPBAR1/TGR5 is a novel plasma membrane-bound G protein–coupled bile acid (BA) receptor. BAs are known to induce the expression of inflammatory cytokines in the liver with unknown mechanism. Here we show that without other external stimuli, TGR5 activation alone induced the expression of interleukin 1β (IL-1β) and tumor necrosis factor-α (TNF-α) in murine macrophage cell line RAW264.7 or murine Kupffer cells. The TGR5-mediated increase of pro-inflammatory cytokine expression was suppressed by JNK inhibition. Moreover, the induced pro-inflammatory cytokine expression in mouse liver by 1% cholic acid (CA) diet was blunted in JNK−/− mice. TGR5 activation by its ligands enhanced the phosphorylation levels, DNA-binding and trans-activities of c-Jun and ATF2 transcription factors. Finally, the induced pro-inflammatory cytokine expression in Kupffer cells by TGR5 activation correlated with the suppression of Cholesterol 7α-hydroxylase (Cyp7a1) expression in murine hepatocytes. These results suggest that TGR5 mediates the BA-induced pro-inflammatory cytokine production in murine Kupffer cells through JNK-dependent pathway. This novel role of TGR5 may correlate to the suppression of Cyp7a1 expression in hepatocytes and contribute to the delicate BA feedback regulation.
Collapse
Affiliation(s)
- Guiyu Lou
- Department of Biochemistry and Molecular Biology, Third Military Medical University, Chongqing, China; Division of Molecular Diabetes Research, Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Xiaoxiao Ma
- Division of Molecular Diabetes Research, Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America; Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Xianghui Fu
- Division of Molecular Diabetes Research, Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Zhipeng Meng
- Division of Molecular Diabetes Research, Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America; Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Wenyu Zhang
- Division of Molecular Diabetes Research, Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Yan-Dong Wang
- Division of Molecular Diabetes Research, Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Carl Van Ness
- Division of Molecular Diabetes Research, Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Donna Yu
- Division of Molecular Diabetes Research, Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Rongzhen Xu
- Department of Hematology and Cancer Institute, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wendong Huang
- Division of Molecular Diabetes Research, Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| |
Collapse
|
27
|
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
|
28
|
Mascarenhas DD, ElAyadi A, Singh BK, Prasai A, Hegde SD, Herndon DN, Finnerty CC. Nephrilin peptide modulates a neuroimmune stress response in rodent models of burn trauma and sepsis. INTERNATIONAL JOURNAL OF BURNS AND TRAUMA 2013; 3:190-200. [PMID: 24273694 PMCID: PMC3828741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 09/15/2013] [Indexed: 06/02/2023]
Abstract
Sepsis occurs three times more often in burns than in other types of trauma, suggesting an overlap or synergy between underlying immune mechanisms in burn trauma and sepsis. Nephrilin peptide, a designed inhibitor of mTORC2, has previously been shown to modulate a neuroimmune stress response in rodent models of xenobiotic and metabolic stress. Here we investigate the effect of nephrilin peptide administration in different rodent models of burn trauma and sepsis. In a rat scald burn model, daily subcutaneous bolus injection of 4 mg/kg nephrilin significantly reduced the elevation of kidney tissue substance P, S100A9 gene expression, PMN infiltration and plasma inflammatory markers in the acute phase, while suppressing plasma CCL2 and insulin C-peptide, kidney p66shc-S36 phosphorylation and PKC-beta and CGRP in dorsal root ganglia at 14 days (chronic phase). In the mouse cecal ligation and puncture model of sepsis, nephrilin fully protected mice from mortality between surgery and day 7, compared to 67% mortality in saline-treated animals, while significantly reducing elevated CCL2 in plasma. mTORC2 may modulate important neuroimmune responses in both burn trauma and sepsis.
Collapse
Affiliation(s)
- Desmond D Mascarenhas
- Mayflower Organization for Research & EducationSunnyvale, CA 94085, USA
- Protigen Inc.Sunnyvale, CA 94085, USA
| | - Amina ElAyadi
- Department of Surgery, The University of Texas Medical Branch, and Shriners Hospitals for ChildrenGalveston, TX 77550, USA
| | | | - Anesh Prasai
- Department of Surgery, The University of Texas Medical Branch, and Shriners Hospitals for ChildrenGalveston, TX 77550, USA
| | - Sachin D Hegde
- Department of Surgery, The University of Texas Medical Branch, and Shriners Hospitals for ChildrenGalveston, TX 77550, USA
| | - David N Herndon
- Department of Surgery, The University of Texas Medical Branch, and Shriners Hospitals for ChildrenGalveston, TX 77550, USA
| | - Celeste C Finnerty
- Department of Surgery, The University of Texas Medical Branch, and Shriners Hospitals for ChildrenGalveston, TX 77550, USA
- The Institute for Translational Sciences and The Sealy Center for Molecular Medicine, The University of Texas Medical BranchGalveston TX 77550, USA
| |
Collapse
|
29
|
CHIKIN VV, ZNAMENSKAYA LF, KATUNINA OR, LVOV AN, FRIGO NV, INOYATOVA LA. Modern methods and approaches to studying the role of neurotransmitters in the pathogenesis of chronic inflammatory skin diseases accompanied with itching. VESTNIK DERMATOLOGII I VENEROLOGII 2012. [DOI: 10.25208/vdv726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
|
30
|
Lau E, Ronai ZA. ATF2 - at the crossroad of nuclear and cytosolic functions. J Cell Sci 2012; 125:2815-24. [PMID: 22685333 DOI: 10.1242/jcs.095000] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
An increasing number of transcription factors have been shown to elicit oncogenic and tumor suppressor activities, depending on the tissue and cell context. Activating transcription factor 2 (ATF2; also known as cAMP-dependent transcription factor ATF-2) has oncogenic activities in melanoma and tumor suppressor activities in non-malignant skin tumors and breast cancer. Recent work has shown that the opposing functions of ATF2 are associated with its subcellular localization. In the nucleus, ATF2 contributes to global transcription and the DNA damage response, in addition to specific transcriptional activities that are related to cell development, proliferation and death. ATF2 can also translocate to the cytosol, primarily following exposure to severe genotoxic stress, where it impairs mitochondrial membrane potential and promotes mitochondrial-based cell death. Notably, phosphorylation of ATF2 by the epsilon isoform of protein kinase C (PKCε) is the master switch that controls its subcellular localization and function. Here, we summarize our current understanding of the regulation and function of ATF2 in both subcellular compartments. This mechanism of control of a non-genetically modified transcription factor represents a novel paradigm for 'oncogene addiction'.
Collapse
Affiliation(s)
- Eric Lau
- Signal Transduction Program, Sanford-Burnham Medical Research Institute, 10901 N. Torrey Pines Rd, La Jolla, CA 92130, USA.
| | | |
Collapse
|
31
|
Gais P, Reim D, Jusek G, Rossmann-Bloeck T, Weighardt H, Pfeffer K, Altmayr F, Janssen KP, Holzmann B. Cutting edge: Divergent cell-specific functions of MyD88 for inflammatory responses and organ injury in septic peritonitis. THE JOURNAL OF IMMUNOLOGY 2012; 188:5833-7. [PMID: 22586041 DOI: 10.4049/jimmunol.1200038] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Although global MyD88 deficiency attenuates lethal inflammation in sepsis, cell-specific functions of MyD88 remain largely unknown. Using mice with selective expression of MyD88 in myeloid cells (Myd88(MYEL)), we show that, during polymicrobial septic peritonitis, both myeloid and nonmyeloid cells contribute to systemic inflammation, whereas myeloid cell MyD88 was sufficient to fully establish the peritoneal cytokine response. Importantly, Myd88(MYEL) mice developed markedly aggravated liver injury that was linked to impaired upregulation of cellular inhibitor of apoptosis protein 2 and an excessive production of TNF-α. Upregulation of inducible cAMP early repressor (ICER), a known transcriptional repressor of the Tnfa gene, was impaired in Myd88(MYEL) mice. Moreover, Myd88(MYEL) mice showed enhanced transcription of the Tnfa gene and an excessive production of CCL3, which is also negatively regulated by ICER, but they had normal levels of CXCL1, which is expressed in an ICER-independent manner. Together, these findings suggest a novel protective role for nonmyeloid cell MyD88 in attenuating liver injury during septic peritonitis.
Collapse
Affiliation(s)
- Petra Gais
- Chirurgische Klinik und Poliklinik, Technische Universität München, 81675 Munich, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Jusek G, Reim D, Tsujikawa K, Holzmann B. Deficiency of the CGRP receptor component RAMP1 attenuates immunosuppression during the early phase of septic peritonitis. Immunobiology 2012; 217:761-7. [PMID: 22656887 DOI: 10.1016/j.imbio.2012.04.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 04/27/2012] [Indexed: 01/15/2023]
Abstract
The neuropeptide CGRP contributes to the control of excessive cytokine production in endotoxemia models. However, the function of CGRP in sepsis caused by infection with viable pathogens is unknown. Here, we show that mice deficient for the CGRP receptor component RAMP1 have an improved anti-bacterial defense during the early, but not late, phase of polymicrobial septic peritonitis. The protective effect of Ramp1-deficiency was associated with reduced levels of IL-10 in plasma and peritoneal lavage fluid. Consistent with these findings, CGRP markedly increased IL-10 production of peritoneal and bone marrow-derived macrophages in response to short term stimulation with LPS in vitro. In addition, the lack of an intact CGRP receptor resulted in an increased recruitment and activation of neutrophils and caused an enhanced release of defensin-α1 in the peritoneal cavity. Considered together, our results identify the neuropeptide CGRP as a crucial immunosuppressive mediator impairing host defense during the early, but not late, phase of septic peritonitis.
Collapse
Affiliation(s)
- Gabriela Jusek
- Department of Surgery, Technical University Munich, Ismaninger Strasse 22, 81675 Munich, Germany
| | | | | | | |
Collapse
|
33
|
Holzmann B. Modulation of immune responses by the neuropeptide CGRP. Amino Acids 2011; 45:1-7. [PMID: 22113645 DOI: 10.1007/s00726-011-1161-2] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Accepted: 11/14/2011] [Indexed: 01/15/2023]
Abstract
The peripheral nervous system is connected with lymphoid organs through sensory nerves that mediate pain reflexes and may influence immune responses through the release of neuropeptides such as calcitonin gene-related peptide (CGRP). Local and systemic levels of CGRP increase rapidly during inflammatory responses. CGRP inhibits effector functions of various immune cells and dampens inflammation by distinct pathways involving the amplification of IL-10 production and/or the induction of the transcriptional repressor inducible cAMP early repressor (ICER). Thus, available evidence suggests that, in neuro-immunological interactions, CGRP mediates a potent peptidergic anti-inflammatory pathway.
Collapse
Affiliation(s)
- Bernhard Holzmann
- Department of Surgery, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675 München, Germany.
| |
Collapse
|
34
|
Rochlitzer S, Veres TZ, Kühne K, Prenzler F, Pilzner C, Knothe S, Winkler C, Lauenstein HD, Willart M, Hammad H, Müller M, Krug N, Lambrecht BN, Braun A. The neuropeptide calcitonin gene-related peptide affects allergic airway inflammation by modulating dendritic cell function. Clin Exp Allergy 2011; 41:1609-21. [PMID: 21752117 DOI: 10.1111/j.1365-2222.2011.03822.x] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND The neuropeptide calcitonin gene-related peptide (CGRP) is released in the lung by sensory nerves during allergic airway responses. Pulmonary dendritic cells (DC) orchestrating the allergic inflammation could be affected by CGRP. OBJECTIVE To determine the immunomodulatory effects of CGRP on DC function and its impact on the induction of allergic airway inflammation. METHODS CGRP receptor expression on lung DC was determined by RT-PCR and immunofluorescence staining. The functional consequences of CGRP receptor triggering were evaluated in vitro using bone marrow-derived DC. DC maturation and the induction of ovalbumin (OVA)-specific T cell responses were analysed by flow cytometry. The in vivo relevance of the observed DC modulation was assessed in a DC-transfer model of experimental asthma. Mice were sensitized by an intrapharyngeal transfer of OVA-pulsed DC and challenged with OVA aerosol. The impact of CGRP pretreatment of DC on airway inflammation was characterized by analysing differential cell counts and cytokines in bronchoalveolar lavage fluid (BALF), lung histology and cytokine responses in mediastinal lymph nodes. RESULTS RT-PCR, immunofluorescence and cAMP assay demonstrated the expression of functionally active CGRP receptors in lung DC. RT-PCR revealed a transcriptional CGRP receptor down-regulation during airway inflammation. CGRP specifically inhibited the maturation of in vitro generated DC. Maturation was restored by blocking with the specific antagonist CGRP(8-37) . Consequently, CGRP-pretreated DC reduced the activation and proliferation of antigen-specific T cells and induced increased the numbers of T regulatory cells. The transfer of CGRP-pretreated DC diminished allergic airway inflammation in vivo, shown by reduced eosinophil numbers and increased levels of IL-10 in BALF. CONCLUSIONS AND CLINICAL RELEVANCE CGRP inhibits DC maturation and allergen-specific T cell responses, which affects the outcome of the allergic airway inflammation in vivo. This suggests an additional mechanism by which nerve-derived mediators interfere with local immune responses. Thus, CGRP as an anti-inflammatory mediator could represent a new therapeutic tool in asthma therapy.
Collapse
Affiliation(s)
- S Rochlitzer
- Department of Airway Immunology, Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Nguyen TT, Johnsen IB, Knetter CF, Drabløs F, Fitzgerald KA, Lien E, Anthonsen MW. Differential gene expression downstream of Toll-like receptors (TLRs): role of c-Src and activating transcription factor 3 (ATF3). J Biol Chem 2010; 285:17011-9. [PMID: 20351107 DOI: 10.1074/jbc.m109.068817] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Interferon regulatory factors (IRFs) are crucial for transcription during innate immune responses. We have previously shown that the tyrosine kinase c-Src enhances IRF-3-dependent transcription in response to viral double-stranded RNA. In this study, we show that c-Src has distinct roles in Toll-like receptor (TLR)-mediated activation of IRF-5 and IRF-3. Surprisingly, c-Src inhibition markedly enhanced IRF-5 activation after treatment with unmethylated CpG, while suppressing IRF-3 activation. Also, CpG-elicited interleukin-6 mRNA production was increased, whereas IP10 mRNA synthesis was reduced in cells deficient in c-Src. Interestingly, c-Src regulated TLR-stimulated induction of activating transcription factor 3 (ATF3), a transcriptional repressor. Depletion of ATF3 by small interfering RNA markedly enhanced interleukin-6 production after CpG treatment, whereas IP10 production was reduced. These results demonstrate functional specificity for c-Src in TLR-stimulated responses and suggest that c-Src modulation and ATF3 activity may contribute to differential regulation of IRF-3- versus IRF-5-mediated gene expression.
Collapse
Affiliation(s)
- Thuy Thanh Nguyen
- Department of Laboratory Medicine, Children's and Women's Health, Norwegian University of Science and Technology, N-7006 Trondheim, Norway
| | | | | | | | | | | | | |
Collapse
|