Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 121
Filtrer
1.
Trends Immunol ; 45(8): 574-576, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39060142

RÉSUMÉ

The immune and sensory nervous systems communicate to maintain homeostasis. Wu et al. recently demonstrated that sensory neurons innervate the mouse spleen. These neurons promote calcitonin gene-related peptide (CGRP)-dependent responses in splenic B cell germinal centers (GCs) and antigen-specific antibody production. Dietary capsaicin activates these neurons to enhance humoral immunity against influenza virus infection.


Sujet(s)
Lymphocytes B , Centre germinatif , Immunité humorale , Rate , Animaux , Rate/immunologie , Rate/innervation , Humains , Centre germinatif/immunologie , Souris , Lymphocytes B/immunologie , Cellules réceptrices sensorielles/immunologie , Cellules réceptrices sensorielles/physiologie , Peptide relié au gène de la calcitonine/métabolisme , Peptide relié au gène de la calcitonine/immunologie , Capsaïcine/pharmacologie
2.
Trends Immunol ; 45(5): 381-396, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38697871

RÉSUMÉ

Recent studies have uncovered a new role for sensory neurons in influencing mammalian host immunity, challenging conventional notions of the nervous and immune systems as separate entities. In this review we delve into this groundbreaking paradigm of neuroimmunology and discuss recent scientific evidence for the impact of sensory neurons on host responses against a wide range of pathogens and diseases, encompassing microbial infections and cancers. These valuable insights enhance our understanding of the interactions between the nervous and immune systems, and also pave the way for developing candidate innovative therapeutic interventions in immune-mediated diseases highlighting the importance of this interdisciplinary research field.


Sujet(s)
Cellules réceptrices sensorielles , Animaux , Humains , Interactions hôte-pathogène , Immunité , Tumeurs/immunologie , Tumeurs/thérapie , Neuro-immunomodulation , Cellules réceptrices sensorielles/immunologie , Cellules réceptrices sensorielles/physiologie
3.
Trends Immunol ; 45(5): 371-380, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38653601

RÉSUMÉ

Peripheral sensory neurons recognize diverse noxious stimuli, including microbial products and allergens traditionally thought to be targets of the mammalian immune system. Activation of sensory neurons by these stimuli leads to pain and itch responses as well as the release of neuropeptides that interact with their cognate receptors expressed on immune cells, such as dendritic cells (DCs). Neuronal control of immune cell function through neuropeptide release not only affects local inflammatory responses but can impact adaptive immune responses through downstream effects on T cell priming. Numerous neuropeptide receptors are expressed by DCs but only a few have been characterized, presenting opportunities for further investigation of the pathways by which cutaneous neuroimmune interactions modulate host immunity.


Sujet(s)
Cellules réceptrices sensorielles , Peau , Humains , Animaux , Cellules réceptrices sensorielles/immunologie , Cellules réceptrices sensorielles/métabolisme , Cellules réceptrices sensorielles/physiologie , Peau/immunologie , Neuropeptides/métabolisme , Neuropeptides/immunologie , Cellules dendritiques/immunologie , Neuro-immunomodulation , Récepteur aux neuropeptides/métabolisme , Récepteur aux neuropeptides/immunologie
4.
J Allergy Clin Immunol ; 154(1): 11-19, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38492673

RÉSUMÉ

Various immune cells in the skin contribute to its function as a first line of defense against infection and disease, and the skin's dense innervation by pain-sensing sensory neurons protects the host against injury or damage signals. Dendritic cells (DCs) are a heterogeneous population of cells that link the innate immune response to the adaptive response by capturing, processing, and presenting antigens to promote T-cell differentiation and activation. DCs are abundant across peripheral tissues, including the skin, where they are found in the dermis and epidermis. Langerhans cells (LCs) are a DC subset located only in the epidermis; both populations of cells can migrate to lymph nodes to contribute to broad immune responses. Dermal DCs and LCs are found in close apposition with sensory nerve fibers in the skin and express neurotransmitter receptors, allowing them to communicate directly with the peripheral nervous system. Thus, neuroimmune signaling between DCs and/or LCs and sensory neurons can modulate physiologic and pathophysiologic pathways, including immune cell regulation, host defense, allergic response, homeostasis, and wound repair. Here, we summarize the latest discoveries on DC- and LC-neuron interaction with neurons while providing an overview of gaps and areas not previously explored. Understanding the interactions between these 2 defence systems may provide key insight into developing therapeutic targets for treating diseases such as psoriasis, neuropathic pain, and lupus.


Sujet(s)
Cellules dendritiques , Cellules de Langerhans , Peau , Humains , Cellules de Langerhans/immunologie , Animaux , Peau/immunologie , Peau/innervation , Cellules dendritiques/immunologie , Cellules réceptrices sensorielles/physiologie , Cellules réceptrices sensorielles/immunologie , Communication cellulaire/immunologie , Neuro-immunomodulation
5.
Int J Mol Sci ; 22(23)2021 Dec 03.
Article de Anglais | MEDLINE | ID: mdl-34884907

RÉSUMÉ

Endometriosis (EM) is an estrogen-dependent disease characterized by the presence of epithelial, stromal, and smooth muscle cells outside the uterine cavity. It is a chronic and debilitating condition affecting ~10% of women. EM is characterized by infertility and pain, such as dysmenorrhea, chronic pelvic pain, dyspareunia, dysuria, and dyschezia. Although EM was first described in 1860, its aetiology and pathogenesis remain uncertain. Recent evidence demonstrates that the peripheral nervous system plays an important role in the pathophysiology of this disease. Sensory nerves, which surround and innervate endometriotic lesions, not only drive the chronic and debilitating pain associated with EM but also contribute to a growth phenotype by secreting neurotrophic factors and interacting with surrounding immune cells. Here we review the role that peripheral nerves play in driving and maintaining endometriotic lesions. A better understanding of the role of this system, as well as its interactions with immune cells, will unearth novel disease-relevant pathways and targets, providing new therapeutics and better-tailored treatment options.


Sujet(s)
Endométriose/immunologie , Facteurs de croissance nerveuse/métabolisme , Inflammation neurogénique/étiologie , Endométriose/complications , Femelle , Régulation de l'expression des gènes , Humains , Inflammation neurogénique/immunologie , Douleur pelvienne/étiologie , Douleur pelvienne/immunologie , Cellules réceptrices sensorielles/immunologie
6.
J Neuroimmunol ; 361: 577757, 2021 12 15.
Article de Anglais | MEDLINE | ID: mdl-34768040

RÉSUMÉ

Antibodies against FGFR3 define a subgroup of sensory neuropathy (SN). The aim of this study was to identify the epitope(s) of anti-FGFR3 autoantibodies and potential epitope-dependent clinical subtypes. Using SPOT methodology, five specific candidate epitopes, three in the juxtamembrane domain (JMD) and two in the tyrosine kinase domain (TKD), were screened with 68 anti-FGFR3-positive patients and 35 healthy controls. The identified epitopes cover 6/15 functionally relevant sites of the protein. Four patients reacted with the JMD and 11 with the TKD, partly even in a phosphorylation-state dependent manner. The epitope could not be identified in the others. Patients with antibodies recognizing TKD exhibited a more severe clinical and electrophysiological impairment than others.


Sujet(s)
Autoanticorps/immunologie , Autoantigènes/immunologie , Maladies auto-immunes du système nerveux/immunologie , Épitopes/immunologie , Protéines de tissu nerveux/immunologie , Récepteur de type 3 des facteurs de croissance fibroblastique/immunologie , Troubles sensitifs/immunologie , Adulte , Autoanticorps/sang , Autoantigènes/composition chimique , Femelle , Ganglions sensitifs des nerfs spinaux/immunologie , Humains , Mâle , Adulte d'âge moyen , Phosphorylation , Domaines protéiques , Maturation post-traductionnelle des protéines , Récepteur de type 3 des facteurs de croissance fibroblastique/composition chimique , Cellules réceptrices sensorielles/immunologie
7.
Int J Mol Sci ; 22(22)2021 Nov 16.
Article de Anglais | MEDLINE | ID: mdl-34830245

RÉSUMÉ

Although histamine is a well-known itch mediator, histamine H1-receptor blockers often lack efficacy in chronic itch. Recent molecular and cellular based studies have shown that non-histaminergic mediators, such as proteases, neuropeptides and cytokines, along with their cognate receptors, are involved in evocation and modulation of itch sensation. Many of these molecules are produced and secreted by immune cells, which act on sensory nerve fibers distributed in the skin to cause itching and sensitization. This understanding of the connections between immune cell-derived mediators and sensory nerve fibers has led to the development of new treatments for itch. This review summarizes current knowledge of immune cell-derived itch mediators and neuronal response mechanisms, and discusses therapeutic agents that target these systems.


Sujet(s)
Anti-inflammatoires/usage thérapeutique , Histamine/immunologie , Facteurs immunologiques/usage thérapeutique , Prurit/immunologie , Récepteur histaminergique H1/immunologie , Cellules réceptrices sensorielles/immunologie , Anticorps monoclonaux/usage thérapeutique , Cytokines/antagonistes et inhibiteurs , Cytokines/immunologie , Cytokines/métabolisme , Expression des gènes , Histamine/métabolisme , Antihistaminiques/usage thérapeutique , Humains , Lymphocytes/effets des médicaments et des substances chimiques , Lymphocytes/immunologie , Lymphocytes/anatomopathologie , Cellules myéloïdes/effets des médicaments et des substances chimiques , Cellules myéloïdes/immunologie , Cellules myéloïdes/anatomopathologie , Neuropeptides/antagonistes et inhibiteurs , Neuropeptides/immunologie , Neuropeptides/métabolisme , Peptide hydrolases/immunologie , Peptide hydrolases/métabolisme , Inhibiteurs de protéases/usage thérapeutique , Prurit/traitement médicamenteux , Prurit/génétique , Prurit/anatomopathologie , Récepteur histaminergique H1/génétique , Cellules réceptrices sensorielles/effets des médicaments et des substances chimiques , Cellules réceptrices sensorielles/anatomopathologie , Peau/effets des médicaments et des substances chimiques , Peau/immunologie , Peau/innervation , Peau/anatomopathologie
8.
J Neuroinflammation ; 18(1): 227, 2021 Oct 13.
Article de Anglais | MEDLINE | ID: mdl-34645458

RÉSUMÉ

BACKGROUND: Macrophages in the peripheral nervous system are key players in the repair of nerve tissue and the development of neuropathic pain due to peripheral nerve injury. However, there is a lack of information on the origin and morphological features of macrophages in sensory ganglia after peripheral nerve injury, unlike those in the brain and spinal cord. We analyzed the origin and morphological features of sensory ganglionic macrophages after nerve ligation or transection using wild-type mice and mice with bone-marrow cell transplants. METHODS: After protecting the head of C57BL/6J mice with lead caps, they were irradiated and transplanted with bone-marrow-derived cells from GFP transgenic mice. The infraorbital nerve of a branch of the trigeminal nerve of wild-type mice was ligated or the infraorbital nerve of GFP-positive bone-marrow-cell-transplanted mice was transected. After immunostaining the trigeminal ganglion, the structures of the ganglionic macrophages, neurons, and satellite glial cells were analyzed using two-dimensional or three-dimensional images. RESULTS: The number of damaged neurons in the trigeminal ganglion increased from day 1 after infraorbital nerve ligation. Ganglionic macrophages proliferated from days 3 to 5. Furthermore, the numbers of macrophages increased from days 3 to 15. Bone-marrow-derived macrophages increased on day 7 after the infraorbital nerve was transected in the trigeminal ganglion of GFP-positive bone-marrow-cell-transplanted mice but most of the ganglionic macrophages were composed of tissue-resident cells. On day 7 after infraorbital nerve ligation, ganglionic macrophages increased in volume, extended their processes between the neurons and satellite glial cells, and contacted these neurons. Most of the ganglionic macrophages showed an M2 phenotype when contact was observed, and little neuronal cell death occurred. CONCLUSION: Most of the macrophages that appear after a nerve injury are tissue-resident, and these make direct contact with damaged neurons that act in a tissue-protective manner in the M2 phenotype. These results imply that tissue-resident macrophages signal to neurons directly through physical contact.


Sujet(s)
Transplantation de moelle osseuse/méthodes , Augmentation de la taille cellulaire , Ganglions sensitifs/anatomopathologie , Macrophages/anatomopathologie , Lésions des nerfs périphériques/anatomopathologie , Cellules réceptrices sensorielles/anatomopathologie , Animaux , Ganglions sensitifs/immunologie , Macrophages/immunologie , Mâle , Souris , Souris de lignée C57BL , Souris transgéniques , Lésions des nerfs périphériques/immunologie , Lésions des nerfs périphériques/thérapie , Cellules réceptrices sensorielles/immunologie
9.
Free Radic Res ; 55(7): 757-775, 2021 Jul.
Article de Anglais | MEDLINE | ID: mdl-34238089

RÉSUMÉ

The mechanistic interactions among redox status of leukocytes, muscle, and exercise in pain regulation are still poorly understood and limit targeted treatment. Exercise benefits are numerous, including the treatment of chronic pain. However, unaccustomed exercise may be reported as undesirable as it may contribute to pain. The aim of the present review is to evaluate the relationship between oxidative metabolism and acute exercise-induced pain, and as to whether improved antioxidant capacity underpins the analgesic effects of regular exercise. Preclinical and clinical studies addressing relevant topics on mechanisms by which exercise modulates the nociceptive activity and how redox status can outline pain and analgesia are discussed, in sense of translating into refined outcomes. Emerging evidence points to the role of oxidative stress-induced signaling in sensitizing nociceptor sensory neurons. In response to acute exercise, there is an increase in oxidative metabolism, and consequently, pain. Instead, regular exercise can modulate redox status in favor of antioxidant capacity and repair mechanisms, which have consequently increased resistance to oxidative stress, damage, and pain. Data indicate that acute sessions of unaccustomed prolonged and/or intense exercise increase oxidative metabolism and regulate exercise-induced pain in the post-exercise recovery period. Further, evidence demonstrates regular exercise improves antioxidant status, indicating its therapeutic utility for chronic pain disorders. An improved comprehension of the role of redox status in exercise can provide helpful insights into immune-muscle communication during pain modulatory effects of exercise and support new therapeutic efforts and rationale for the promotion of exercise.


Sujet(s)
Analgésie/effets indésirables , Exercice physique , Muscles squelettiques/anatomopathologie , Nocicepteurs/anatomopathologie , Stress oxydatif , Douleur/anatomopathologie , Cellules réceptrices sensorielles/anatomopathologie , Humains , Muscles squelettiques/métabolisme , Nocicepteurs/immunologie , Nocicepteurs/métabolisme , Oxydoréduction , Douleur/étiologie , Douleur/métabolisme , Cellules réceptrices sensorielles/immunologie , Cellules réceptrices sensorielles/métabolisme
10.
Front Immunol ; 12: 644664, 2021.
Article de Anglais | MEDLINE | ID: mdl-34135889

RÉSUMÉ

Alphaherpesviruses (α-HV) are a large family of double-stranded DNA viruses which cause many human and animal diseases. There are three human α-HVs: Herpes Simplex Viruses (HSV-1 and HSV-2) and Varicella Zoster Virus (VZV). All α-HV have evolved multiple strategies to suppress or exploit host cell innate immune signaling pathways to aid in their infections. All α-HVs initially infect epithelial cells (primary site of infection), and later spread to infect innervating sensory neurons. As with all herpesviruses, α-HVs have both a lytic (productive) and latent (dormant) stage of infection. During the lytic stage, the virus rapidly replicates in epithelial cells before it is cleared by the immune system. In contrast, latent infection in host neurons is a life-long infection. Upon infection of mucosal epithelial cells, herpesviruses immediately employ a variety of cellular mechanisms to evade host detection during active replication. Next, infectious viral progeny bud from infected cells and fuse to neuronal axonal terminals. Here, the nucleocapsid is transported via sensory neuron axons to the ganglion cell body, where latency is established until viral reactivation. This review will primarily focus on how HSV-1 induces various innate immune responses, including host cell recognition of viral constituents by pattern-recognition receptors (PRRs), induction of IFN-mediated immune responses involving toll-like receptor (TLR) signaling pathways, and cyclic GMP-AMP synthase stimulator of interferon genes (cGAS-STING). This review focuses on these pathways along with other mechanisms including autophagy and the complement system. We will summarize and discuss recent evidence which has revealed how HSV-1 is able to manipulate and evade host antiviral innate immune responses both in neuronal (sensory neurons of the trigeminal ganglia) and non-neuronal (epithelial) cells. Understanding the innate immune response mechanisms triggered by HSV-1 infection, and the mechanisms of innate immune evasion, will impact the development of future therapeutic treatments.


Sujet(s)
Axones/immunologie , Herpès/immunologie , Herpèsvirus humain de type 1/immunologie , Échappement immunitaire , Immunité innée , Cellules réceptrices sensorielles/immunologie , Animaux , Herpès/thérapie , Humains , Transduction du signal/immunologie
11.
Nat Commun ; 12(1): 2936, 2021 05 18.
Article de Anglais | MEDLINE | ID: mdl-34006861

RÉSUMÉ

Host protection against cutaneous herpes simplex virus 1 (HSV-1) infection relies on the induction of a robust adaptive immune response. Here, we show that Nav1.8+ sensory neurons, which are involved in pain perception, control the magnitude of CD8 T cell priming and expansion in HSV-1-infected mice. The ablation of Nav1.8-expressing sensory neurons is associated with extensive skin lesions characterized by enhanced inflammatory cytokine and chemokine production. Mechanistically, Nav1.8+ sensory neurons are required for the downregulation of neutrophil infiltration in the skin after viral clearance to limit the severity of tissue damage and restore skin homeostasis, as well as for eliciting robust CD8 T cell priming in skin-draining lymph nodes by controlling dendritic cell responses. Collectively, our data reveal an important role for the sensory nervous system in regulating both innate and adaptive immune responses to viral infection, thereby opening up possibilities for new therapeutic strategies.


Sujet(s)
Lymphocytes T CD8+/immunologie , Herpès/immunologie , Herpèsvirus humain de type 1/immunologie , Douleur nociceptive/immunologie , Cellules réceptrices sensorielles/immunologie , Animaux , Lymphocytes T CD8+/métabolisme , Lymphocytes T CD8+/virologie , Cytokines/immunologie , Cytokines/métabolisme , Femelle , Herpès/génétique , Herpès/virologie , Herpèsvirus humain de type 1/physiologie , Humains , Souris de lignée C57BL , Souris knockout , Souris transgéniques , Canal sodique voltage-dépendant NAV1.8/génétique , Canal sodique voltage-dépendant NAV1.8/immunologie , Canal sodique voltage-dépendant NAV1.8/métabolisme , Infiltration par les neutrophiles/immunologie , Douleur nociceptive/génétique , Douleur nociceptive/métabolisme , Cellules réceptrices sensorielles/métabolisme , Cellules réceptrices sensorielles/virologie , Peau/immunologie , Peau/métabolisme , Peau/virologie
12.
Front Immunol ; 12: 662234, 2021.
Article de Anglais | MEDLINE | ID: mdl-34012447

RÉSUMÉ

Herpes simplex virus type 1 (HSV-1) infection is highly prevalent in humans, with approximately two-thirds of the world population living with this virus. However, only a fraction of those carrying HSV-1, which elicits lifelong infections, are symptomatic. HSV-1 mainly causes lesions in the skin and mucosae but reaches the termini of sensory neurons innervating these tissues and travels in a retrograde manner to the neuron cell body where it establishes persistent infection and remains in a latent state until reactivated by different stimuli. When productive reactivations occur, the virus travels back along axons to the primary infection site, where new rounds of replication are initiated in the skin, in recurrent or secondary infections. During this process, new neuron infections occur. Noteworthy, the mechanisms underlying viral reactivations and the exit of latency are somewhat poorly understood and may be regulated by a crosstalk between the infected neurons and components of the immune system. Here, we review and discuss the immune responses that occur at the skin during primary and recurrent infections by HSV-1, as well as at the interphase of latently-infected neurons. Moreover, we discuss the implications of neuronal signals over the priming and migration of immune cells in the context of HSV-1 infection.


Sujet(s)
Cellules épithéliales/métabolisme , Herpès/immunologie , Herpèsvirus humain de type 1/immunologie , Cellules réceptrices sensorielles/métabolisme , Dermatoses virales/immunologie , Animaux , Techniques de culture cellulaire , Cellules épithéliales/immunologie , Régulation de l'expression des gènes viraux , Herpèsvirus humain de type 1/génétique , Humains , Souris , Cellules réceptrices sensorielles/immunologie , Activation virale , Latence virale , Réplication virale
13.
Front Immunol ; 12: 660203, 2021.
Article de Anglais | MEDLINE | ID: mdl-33912189

RÉSUMÉ

Bidirectional interplay between the peripheral immune and nervous systems plays a crucial role in maintaining homeostasis and responding to noxious stimuli. This crosstalk is facilitated by a variety of cytokines, inflammatory mediators and neuropeptides. Dysregulation of this delicate physiological balance is implicated in the pathological mechanisms of various skin disorders and peripheral neuropathies. The skin is a highly complex biological structure within which peripheral sensory nerve terminals and immune cells colocalise. Herein, we provide an overview of the sensory innervation of the skin and immune cells resident to the skin. We discuss modulation of cutaneous immune response by sensory neurons and their mediators (e.g., nociceptor-derived neuropeptides), and sensory neuron regulation by cutaneous immune cells (e.g., nociceptor sensitization by immune-derived mediators). In particular, we discuss recent findings concerning neuroimmune communication in skin infections, psoriasis, allergic contact dermatitis and atopic dermatitis. We then summarize evidence of neuroimmune mechanisms in the skin in the context of peripheral neuropathic pain states, including chemotherapy-induced peripheral neuropathy, diabetic polyneuropathy, post-herpetic neuralgia, HIV-induced neuropathy, as well as entrapment and traumatic neuropathies. Finally, we highlight the future promise of emerging therapies associated with skin neuroimmune crosstalk in neuropathic pain.


Sujet(s)
Cytokines/immunologie , Médiateurs de l'inflammation/immunologie , Névralgie/immunologie , Neuro-immunomodulation/immunologie , Cellules réceptrices sensorielles/immunologie , Peau/immunologie , Animaux , Cytokines/métabolisme , Humains , Médiateurs de l'inflammation/métabolisme , Modèles immunologiques , Névralgie/métabolisme , Névralgie/physiopathologie , Nocicepteurs/immunologie , Nocicepteurs/métabolisme , Cellules réceptrices sensorielles/métabolisme , Peau/innervation , Peau/métabolisme
14.
FASEB J ; 35(3): e21320, 2021 03.
Article de Anglais | MEDLINE | ID: mdl-33660333

RÉSUMÉ

Influenza A virus (IAV) is rapidly detected in the airways by the immune system, with resident parenchymal cells and leukocytes orchestrating viral sensing and the induction of antiviral inflammatory responses. The airways are innervated by heterogeneous populations of vagal sensory neurons which also play an important role in pulmonary defense. How these neurons respond to IAV respiratory infection remains unclear. Here, we use a murine model to provide the first evidence that vagal sensory neurons undergo significant transcriptional changes following a respiratory IAV infection. RNA sequencing on vagal sensory ganglia showed that IAV infection induced the expression of many genes associated with an antiviral and pro-inflammatory response and this was accompanied by a significant increase in inflammatory cell recruitment into the vagal ganglia. Assessment of gene expression in single-vagal sensory neurons confirmed that IAV infection induced a neuronal inflammatory phenotype, which was most prominent in bronchopulmonary neurons, and also evident in some neurons innervating other organs. The altered transcriptome could be mimicked by intranasal treatment with cytokines and the lung homogenates of infected mice, in the absence of infectious virus. These data argue that IAV pulmonary infection and subsequent inflammation induces vagal sensory ganglia neuroinflammation and this may have important implications for IAV-induced morbidity.


Sujet(s)
Inflammation/immunologie , Virus de la grippe A , Poumon/innervation , Infections à Orthomyxoviridae/immunologie , Cellules réceptrices sensorielles/immunologie , Nerf vague/immunologie , Animaux , Femelle , Poumon/virologie , Mâle , Souris , Souris de lignée C57BL , Cellules réceptrices sensorielles/métabolisme , Transcription génétique , Nerf vague/métabolisme
15.
Front Immunol ; 12: 785355, 2021.
Article de Anglais | MEDLINE | ID: mdl-34975876

RÉSUMÉ

The lungs are constantly exposed to non-sterile air which carries harmful threats, such as particles and pathogens. Nonetheless, this organ is equipped with fast and efficient mechanisms to eliminate these threats from the airways as well as prevent pathogen invasion. The respiratory tract is densely innervated by sensory neurons, also known as nociceptors, which are responsible for the detection of external stimuli and initiation of physiological and immunological responses. Furthermore, expression of functional innate receptors by nociceptors have been reported; however, the influence of these receptors to the lung function and local immune response is poorly described. The COVID-19 pandemic has shown the importance of coordinated and competent pulmonary immunity for the prevention of pathogen spread as well as prevention of excessive tissue injury. New findings suggest that lung nociceptors can be a target of SARS-CoV-2 infection; what remains unclear is whether innate receptor trigger sensory neuron activation during SARS-CoV-2 infection and what is the relevance for the outcomes. Moreover, elderly individuals often present with respiratory, neurological and immunological dysfunction. Whether aging in the context of sensory nerve function and innate receptors contributes to the disorders of these systems is currently unknown. Here we discuss the expression of innate receptors by nociceptors, particularly in the lungs, and the possible impact of their activation on pulmonary immunity. We then demonstrate recent evidence that suggests lung sensory neurons as reservoirs for SARS-CoV-2 and possible viral recognition via innate receptors. Lastly, we explore the mechanisms by which lung nociceptors might contribute to disturbance in respiratory and immunological responses during the aging process.


Sujet(s)
Vieillissement/immunologie , COVID-19/immunologie , Immunité innée/immunologie , Poumon/immunologie , Nocicepteurs/immunologie , SARS-CoV-2/immunologie , Canaux cationiques TRP/immunologie , Sujet âgé , COVID-19/virologie , Humains , Poumon/innervation , Poumon/virologie , Nocicepteurs/métabolisme , Nocicepteurs/virologie , SARS-CoV-2/physiologie , Cellules réceptrices sensorielles/immunologie , Cellules réceptrices sensorielles/métabolisme , Cellules réceptrices sensorielles/virologie , Canaux cationiques TRP/métabolisme
16.
Cell ; 184(2): 441-459.e25, 2021 01 21.
Article de Anglais | MEDLINE | ID: mdl-33333021

RÉSUMÉ

Barrier tissue immune responses are regulated in part by nociceptors. Nociceptor ablation alters local immune responses at peripheral sites and within draining lymph nodes (LNs). The mechanisms and significance of nociceptor-dependent modulation of LN function are unknown. Using high-resolution imaging, viral tracing, single-cell transcriptomics, and optogenetics, we identified and functionally tested a sensory neuro-immune circuit that is responsive to lymph-borne inflammatory signals. Transcriptomics profiling revealed that multiple sensory neuron subsets, predominantly peptidergic nociceptors, innervate LNs, distinct from those innervating surrounding skin. To uncover LN-resident cells that may interact with LN-innervating sensory neurons, we generated a LN single-cell transcriptomics atlas and nominated nociceptor target populations and interaction modalities. Optogenetic stimulation of LN-innervating sensory fibers triggered rapid transcriptional changes in the predicted interacting cell types, particularly endothelium, stromal cells, and innate leukocytes. Thus, a unique population of sensory neurons monitors peripheral LNs and may locally regulate gene expression.


Sujet(s)
Immunomodulation , Noeuds lymphatiques/immunologie , Noeuds lymphatiques/innervation , Cellules réceptrices sensorielles/immunologie , Potentiels d'action , Animaux , Inflammation/anatomopathologie , Souris , Nocicepteurs/métabolisme , Optogénétique , Peptides/métabolisme , Peau/innervation , Système nerveux sympathique/physiologie , Récepteurs de type Toll/agonistes , Récepteurs de type Toll/métabolisme
17.
Immunol Lett ; 229: 32-43, 2021 01.
Article de Anglais | MEDLINE | ID: mdl-33248166

RÉSUMÉ

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.


Sujet(s)
Infections/étiologie , Infections/métabolisme , Neuro-immunomodulation , Douleur/étiologie , Animaux , Marqueurs biologiques , Communication cellulaire , Prédisposition aux maladies/immunologie , Métabolisme énergétique , Interactions hôte-parasite/immunologie , Interactions hôte-pathogène/immunologie , Humains , Système immunitaire/immunologie , Système immunitaire/innervation , Système immunitaire/métabolisme , Infections/complications , Inflammation/complications , Inflammation/étiologie , Inflammation/métabolisme , Médiateurs de l'inflammation/métabolisme , Métabolisme lipidique , Douleur/diagnostic , Douleur/métabolisme , Cellules réceptrices sensorielles/immunologie , Cellules réceptrices sensorielles/métabolisme
18.
Immunity ; 53(5): 1063-1077.e7, 2020 11 17.
Article de Anglais | MEDLINE | ID: mdl-33098765

RÉSUMÉ

Dendritic cells (DCs) of the cDC2 lineage initiate allergic immunity and in the dermis are marked by their expression of CD301b. CD301b+ dermal DCs respond to allergens encountered in vivo, but not in vitro. This suggests that another cell in the dermis may sense allergens and relay that information to activate and induce the migration of CD301b+ DCs to the draining lymph node (dLN). Using a model of cutaneous allergen exposure, we show that allergens directly activated TRPV1+ sensory neurons leading to itch and pain behaviors. Allergen-activated sensory neurons released the neuropeptide Substance P, which stimulated proximally located CD301b+ DCs through the Mas-related G-protein coupled receptor member A1 (MRGPRA1). Substance P induced CD301b+ DC migration to the dLN where they initiated T helper-2 cell differentiation. Thus, sensory neurons act as primary sensors of allergens, linking exposure to activation of allergic-skewing DCs and the initiation of an allergic immune response.


Sujet(s)
Allergènes/immunologie , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Hypersensibilité/étiologie , Hypersensibilité/métabolisme , Cellules réceptrices sensorielles/métabolisme , Substance P/biosynthèse , Animaux , Marqueurs biologiques , Mouvement cellulaire/immunologie , Femelle , Ganglions sensitifs des nerfs spinaux/cytologie , Hypersensibilité/diagnostic , Mâle , Souris , Cellules réceptrices sensorielles/immunologie
19.
J Virol ; 94(21)2020 10 14.
Article de Anglais | MEDLINE | ID: mdl-32796067

RÉSUMÉ

Neurotropic Alphaherpesvirinae subfamily members such as bovine herpesvirus 1 (BoHV-1) and herpes simplex virus 1 (HSV-1) establish and maintain lifelong latent infections in neurons. Following infection of ocular, oral, or nasal cavities, sensory neurons within trigeminal ganglia (TG) are an important site for latency. Certain external stressors can trigger reactivation from latency, in part because activation of the glucocorticoid receptor (GR) stimulates productive infection and promoters that drive expression of key viral transcriptional regulators. The Akt serine/threonine protein kinase family is linked to maintaining latency. For example, Akt3 is detected in more TG neurons during BoHV-1 latency than in reactivation and uninfected calves. Furthermore, Akt signaling correlates with maintaining HSV-1 latency in certain neuronal models of latency. Finally, an active Akt protein kinase is crucial for the ability of the HSV-1 latency-associated transcript (LAT) to inhibit apoptosis in neuronal cell lines. Consequently, we hypothesized that viral and/or cellular factors impair stress-induced transcription and reduce the incidence of reactivation triggered by low levels of stress. New studies demonstrate that Akt1 and Akt2, but not Akt3, significantly reduced GR-mediated transactivation of the BoHV-1 immediate early transcription unit 1 (IEtu1) promoter, the HSV-1 infected cell protein 0 (ICP0) promoter, and the mouse mammary tumor virus long terminal repeat (MMTV-LTR). Akt3, but not Akt1 or Akt2, significantly enhanced neurite formation in mouse neuroblastoma cells, which correlates with repairing damaged neurons. These studies suggest that unique biological properties of the three Akt family members promote the maintenance of latency in differentiated neurons.IMPORTANCE External stressful stimuli are known to increase the incidence of reactivation of Alphaherpesvirinae subfamily members. Activation of the glucocorticoid receptor (GR) by the synthetic corticosteroid dexamethasone (DEX) stimulates bovine herpesvirus 1 (BoHV-1) and herpes simplex virus 1 (HSV-1) reactivation. Furthermore, GR and dexamethasone stimulate productive infection and promoters that drive expression of viral transcriptional regulators. These observations lead us to predict that stress-induced transcription is impaired by factors abundantly expressed during latency. Interestingly, activation of the Akt family of serine/threonine protein kinases is linked to maintenance of latency. New studies reveal that Akt1 and Ak2, but not Akt3, impaired GR- and dexamethasone-mediated transactivation of the BoHV-1 immediate early transcription unit 1 and HSV-1 ICP0 promoters. Strikingly, Akt3, but not Akt1 or Akt2, stimulated neurite formation in mouse neuroblastoma cells, a requirement for neurogenesis. These studies provide insight into how Akt family members may promote the maintenance of lifelong latency.


Sujet(s)
Herpès/immunologie , Infections à Herpesviridae/immunologie , Interactions hôte-pathogène/immunologie , Protéines proto-oncogènes c-akt/immunologie , Cellules réceptrices sensorielles/virologie , Animaux , Bovins , Différenciation cellulaire , Lignée cellulaire tumorale , Herpès/génétique , Herpès/anatomopathologie , Herpès/virologie , Infections à Herpesviridae/génétique , Infections à Herpesviridae/anatomopathologie , Infections à Herpesviridae/virologie , Herpèsvirus bovin de type 1/génétique , Herpèsvirus bovin de type 1/immunologie , Herpèsvirus humain de type 1/génétique , Herpèsvirus humain de type 1/immunologie , Interactions hôte-pathogène/génétique , Humains , Protéines précoces immédiates/génétique , Protéines précoces immédiates/immunologie , Souris , Neurites/immunologie , Neurites/ultrastructure , Neurites/virologie , Régions promotrices (génétique) , Protéines proto-oncogènes c-akt/génétique , Récepteurs aux glucocorticoïdes/génétique , Récepteurs aux glucocorticoïdes/immunologie , Cellules réceptrices sensorielles/immunologie , Cellules réceptrices sensorielles/anatomopathologie , Transduction du signal , Activation de la transcription/immunologie , Ganglion trigéminal/immunologie , Ganglion trigéminal/anatomopathologie , Ganglion trigéminal/virologie , Ubiquitin-protein ligases/génétique , Ubiquitin-protein ligases/immunologie
20.
Brain Behav Immun ; 89: 559-568, 2020 10.
Article de Anglais | MEDLINE | ID: mdl-32497778

RÉSUMÉ

The SARS-CoV-2 virus infects cells of the airway and lungs in humans causing the disease COVID-19. This disease is characterized by cough, shortness of breath, and in severe cases causes pneumonia and acute respiratory distress syndrome (ARDS) which can be fatal. Bronchial alveolar lavage fluid (BALF) and plasma from mild and severe cases of COVID-19 have been profiled using protein measurements and bulk and single cell RNA sequencing. Onset of pneumonia and ARDS can be rapid in COVID-19, suggesting a potential neuronal involvement in pathology and mortality. We hypothesized that SARS-CoV-2 infection drives changes in immune cell-derived factors that then interact with receptors expressed by the sensory neuronal innervation of the lung to further promote important aspects of disease severity, including ARDS. We sought to quantify how immune cells might interact with sensory innervation of the lung in COVID-19 using published data from patients, existing RNA sequencing datasets from human dorsal root ganglion neurons and other sources, and a genome-wide ligand-receptor pair database curated for pharmacological interactions relevant for neuro-immune interactions. Our findings reveal a landscape of ligand-receptor interactions in the lung caused by SARS-CoV-2 viral infection and point to potential interventions to reduce the burden of neurogenic inflammation in COVID-19 pulmonary disease. In particular, our work highlights opportunities for clinical trials with existing or under development rheumatoid arthritis and other (e.g. CCL2, CCR5 or EGFR inhibitors) drugs to treat high risk or severe COVID-19 cases.


Sujet(s)
Liquide de lavage bronchoalvéolaire/immunologie , Infections à coronavirus/immunologie , Cytokines/immunologie , Poumon/immunologie , Poumon/innervation , Pneumopathie virale/immunologie , Récepteurs aux cytokines/immunologie , Cellules réceptrices sensorielles/immunologie , Antirhumatismaux/usage thérapeutique , Betacoronavirus , COVID-19 , Infections à coronavirus/traitement médicamenteux , Infections à coronavirus/métabolisme , Cytokines/métabolisme , Bases de données factuelles , Ganglions sensitifs des nerfs spinaux , Humains , Poumon/métabolisme , Poumon/physiopathologie , Thérapie moléculaire ciblée , Nocicepteurs/métabolisme , Pandémies , Pneumopathie virale/traitement médicamenteux , Pneumopathie virale/métabolisme , RNA-Seq , Récepteurs aux cytokines/métabolisme , /immunologie , /métabolisme , /physiopathologie , SARS-CoV-2 , Cellules réceptrices sensorielles/métabolisme , Transcriptome , Régulation positive , Traitements médicamenteux de la COVID-19
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE