Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Neuron ; 111(17): 2709-2726.e9, 2023 09 06.
Article in English | MEDLINE | ID: mdl-37348508

ABSTRACT

Programmed death protein 1 (PD-1) and its ligand PD-L1 constitute an immune checkpoint pathway. We report that neuronal PD-1 signaling regulates learning/memory in health and disease. Mice lacking PD-1 (encoded by Pdcd1) exhibit enhanced long-term potentiation (LTP) and memory. Intraventricular administration of anti-mouse PD-1 monoclonal antibody (RMP1-14) potentiated learning and memory. Selective deletion of PD-1 in excitatory neurons (but not microglia) also enhances LTP and memory. Traumatic brain injury (TBI) impairs learning and memory, which is rescued by Pdcd1 deletion or intraventricular PD-1 blockade. Conversely, re-expression of Pdcd1 in PD-1-deficient hippocampal neurons suppresses memory and LTP. Exogenous PD-L1 suppresses learning/memory in mice and the excitability of mouse and NHP hippocampal neurons through PD-1. Notably, neuronal activation suppresses PD-L1 secretion, and PD-L1/PD-1 signaling is distinctly regulated by learning and TBI. Thus, conditions that reduce PD-L1 levels or PD-1 signaling could promote memory in both physiological and pathological conditions.


Subject(s)
B7-H1 Antigen , Brain Injuries, Traumatic , Humans , B7-H1 Antigen/metabolism , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Learning , Hippocampus/metabolism , Antibodies, Monoclonal/metabolism , Neurons/metabolism
3.
Cell Tissue Res ; 387(2): 225-247, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34859291

ABSTRACT

The fungiform papilla (FP) is a gustatory and somatosensory structure incorporating chorda tympani (CT) nerve fibers that innervate taste buds (TB) and also contain somatosensory endings for touch and temperature. Hedgehog (HH) pathway inhibition eliminates TB, but CT innervation remains in the FP. Importantly, after HH inhibition, CT neurophysiological responses to taste stimuli are eliminated, but tactile responses remain. To examine CT fibers that respond to tactile stimuli in the absence of TB, we used Phox2b-Cre; Rosa26LSL-TdTomato reporter mice to selectively label CT fibers with TdTomato. Normally CT fibers project in a compact bundle directly into TB, but after HH pathway inhibition, CT fibers reorganize and expand just under the FP epithelium where TB were. This widened expanse of CT fibers coexpresses Synapsin-1, ß-tubulin, S100, and neurofilaments. Further, GAP43 expression in these fibers suggests they are actively remodeling. Interestingly, CT fibers have complex terminals within the apical FP epithelium and in perigemmal locations in the FP apex. These extragemmal fibers remain after HH pathway inhibition. To identify tactile end organs in FP, we used a K20 antibody to label Merkel cells. In control mice, K20 was expressed in TB cells and at the base of epithelial ridges outside of FP. After HH pathway inhibition, K20 + cells remained in epithelial ridges but were eliminated in the apical FP without TB. These data suggest that the complex, extragemmal nerve endings within and disbursed under the apical FP are the mechanosensitive nerve endings of the CT that remain after HH pathway inhibition.


Subject(s)
Hedgehog Proteins , Taste Buds , Animals , Chorda Tympani Nerve/metabolism , Hedgehog Proteins/metabolism , Mice , Nerve Endings/metabolism , Taste/physiology , Taste Buds/metabolism , Tongue
4.
Oncoimmunology ; 10(1): 1997385, 2021.
Article in English | MEDLINE | ID: mdl-34858725

ABSTRACT

Type-I interferon (IFN-I) signaling is critical to maintaining antigen-presenting cell function for anti-tumor immunity. However, recent studies have suggested that IFN-I signaling may also contribute to more aggressive phenotypes, raising the possibility that IFN-I downstream signaling in cancer and myeloid cells may exert dichotomous functions.We analyzed the clinicopathologic correlation of cancer-specific IFN-I activation in 195 head and neck squamous cell carcinoma patients. We also characterized the immune impact of IFN-I receptor (IFNAR1)-deficiency in syngeneic tumor models using biochemistry, flow cytometry, and single-cell RNA-Seq. We stained HNSCC tissue microarrays with a sensitive IFN-I downstream signaling activation marker, MX1, and quantitated cancer cell-specific MX1 staining. Kaplan-Meier analysis revealed that MX1-high tumors exhibited worse survival, a phenotype that depends on the number of CD8+ intratumoral T-cells. We found that cancer-specific IFNAR1 engagement promotes cancer stemness and higher expression levels of suppressive immune checkpoint receptor ligands in cancer-derived exosomes. Notably, mice bearing Ifnar1-deficient tumors exhibited lower tumor burden, increased T-cell infiltration, reduced exhausted CD4+PD1high T-cells, and increased effector population CD8+IFN-γ+ T-cells. Then, we performed single-cell RNA-sequencing and discovered that cancer-specific IFN-I signaling not only restricts effector cells expansion but also dampens their functional fitness.The beneficial role of IFN-I activation is largely dependent on the myeloid compartment. Cancer-specific IFN-I receptor engagement promotes cancer stemness and the release of cancer-derived exosomes with high expression levels of immune checkpoint receptor ligands. Cancer-specific IFN-I activation is associated with poor immunogenicity and worse clinical outcomes in HNSCC.


Subject(s)
CD8-Positive T-Lymphocytes , Head and Neck Neoplasms , Animals , Humans , Mice , Signal Transduction , Squamous Cell Carcinoma of Head and Neck
5.
Neuron ; 109(17): 2691-2706.e5, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34473953

ABSTRACT

Although sex dimorphism is increasingly recognized as an important factor in pain, female-specific pain signaling is not well studied. Here we report that administration of IL-23 produces mechanical pain (mechanical allodynia) in female but not male mice, and chemotherapy-induced mechanical pain is selectively impaired in female mice lacking Il23 or Il23r. IL-23-induced pain is promoted by estrogen but suppressed by androgen, suggesting an involvement of sex hormones. IL-23 requires C-fiber nociceptors and TRPV1 to produce pain but does not directly activate nociceptor neurons. Notably, IL-23 requires IL-17A release from macrophages to evoke mechanical pain in females. Low-dose IL-17A directly activates nociceptors and induces mechanical pain only in females. Finally, deletion of estrogen receptor subunit α (ERα) in TRPV1+ nociceptors abolishes IL-23- and IL-17-induced pain in females. These findings demonstrate that the IL-23/IL-17A/TRPV1 axis regulates female-specific mechanical pain via neuro-immune interactions. Our study also reveals sex dimorphism at both immune and neuronal levels.


Subject(s)
Estrogen Receptor alpha/metabolism , Interleukin-17/metabolism , Interleukin-23/metabolism , Macrophages/metabolism , Nociceptive Pain/metabolism , Nociceptors/metabolism , TRPV Cation Channels/metabolism , Animals , Cells, Cultured , Female , Humans , Interleukin-17/pharmacology , Interleukin-23/pharmacology , Male , Mice , Mice, Inbred C57BL , Nerve Fibers, Unmyelinated/metabolism , Nerve Fibers, Unmyelinated/physiology , Nociceptive Pain/physiopathology , Nociceptors/drug effects , Nociceptors/physiology , Sex Factors , Signal Transduction
6.
Nat Commun ; 12(1): 4558, 2021 07 27.
Article in English | MEDLINE | ID: mdl-34315904

ABSTRACT

Patients with advanced stage cancers frequently suffer from severe pain as a result of bone metastasis and bone destruction, for which there is no efficacious treatment. Here, using multiple mouse models of bone cancer, we report that agonists of the immune regulator STING (stimulator of interferon genes) confer remarkable protection against cancer pain, bone destruction, and local tumor burden. Repeated systemic administration of STING agonists robustly attenuates bone cancer-induced pain and improves locomotor function. Interestingly, STING agonists produce acute pain relief through direct neuronal modulation. Additionally, STING agonists protect against local bone destruction and reduce local tumor burden through modulation of osteoclast and immune cell function in the tumor microenvironment, providing long-term cancer pain relief. Finally, these in vivo effects are dependent on host-intrinsic STING and IFN-I signaling. Overall, STING activation provides unique advantages in controlling bone cancer pain through distinct and synergistic actions on nociceptors, immune cells, and osteoclasts.


Subject(s)
Bone Neoplasms/complications , Cancer Pain/etiology , Cancer Pain/immunology , Membrane Proteins/metabolism , Neurons/metabolism , Analgesics/pharmacology , Animals , Bone Neoplasms/blood , Cancer Pain/blood , Cell Line, Tumor , Disease Models, Animal , Female , Femur/diagnostic imaging , Femur/drug effects , Femur/pathology , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Homeodomain Proteins/metabolism , Hyperalgesia/complications , Interferons/blood , Interferons/metabolism , Male , Mammary Neoplasms, Animal/complications , Membrane Proteins/agonists , Mice, Inbred C57BL , Neoplasm Metastasis , Neurons/drug effects , Nociception/drug effects , Osteoclasts/drug effects , Osteoclasts/pathology , Osteogenesis/drug effects , Receptor, Interferon alpha-beta/metabolism , Signal Transduction/drug effects , Tumor Burden/drug effects , Tumor Microenvironment/drug effects , Xanthones/pharmacology
7.
Pain Rep ; 6(1): e867, 2021.
Article in English | MEDLINE | ID: mdl-33981921

ABSTRACT

Many common cancers such as breast, prostate, and lung cancer metastasize to bones at advanced stages, producing severe pain and functional impairment. At present, the current pharmacotherapies available for bone cancer pain are insufficient to provide safe and efficacious pain relief. In this narrative review, we discuss the mechanisms used by cancer cells within the bone tumor microenvironment (TME) to drive bone cancer pain. In particular, we highlight the reciprocal interactions between tumor cells, bone-resorbing osteoclasts, and pain-sensing sensory neurons (nociceptors), which drive bone cancer pain. We discuss how tumor cells present within the bone TME accelerate osteoclast differentiation (osteoclastogenesis) and alter osteoclast activity and function. Furthermore, we highlight how this perturbed state of osteoclast overactivation contributes to bone cancer pain through (1) direct mechanisms, through their production of pronociceptive factors that act directly on sensory afferents; and (2) by indirect mechanisms, wherein osteoclasts drive bone resorption that weakens tumor-bearing bones and predisposes them to skeletal-related events, thereby driving bone cancer pain and functional impairment. Finally, we discuss some potential therapeutic agents, such as denosumab, bisphosphonates, and nivolumab, and discuss their respective effects on bone cancer pain, osteoclast overactivation, and tumor growth within the bone TME.

8.
Nat Commun ; 12(1): 1704, 2021 03 17.
Article in English | MEDLINE | ID: mdl-33731716

ABSTRACT

GPR37 was discovered more than two decades ago, but its biological functions remain poorly understood. Here we report a protective role of GPR37 in multiple models of infection and sepsis. Mice lacking Gpr37 exhibited increased death and/or hypothermia following challenge by lipopolysaccharide (LPS), Listeria bacteria, and the mouse malaria parasite Plasmodium berghei. Sepsis induced by LPS and Listeria in wild-type mice is protected by artesunate (ARU) and neuroprotectin D1 (NPD1), but the protective actions of these agents are lost in Gpr37-/- mice. Notably, we found that ARU binds to GPR37 in macrophages and promotes phagocytosis and clearance of pathogens. Moreover, ablation of macrophages potentiated infection, sepsis, and their sequelae, whereas adoptive transfer of NPD1- or ARU-primed macrophages reduced infection, sepsis, and pain-like behaviors. Our findings reveal physiological actions of ARU in host cells by activating macrophages and suggest that GPR37 agonists may help to treat sepsis, bacterial infections, and malaria.


Subject(s)
Macrophages/metabolism , Pain/prevention & control , Receptors, G-Protein-Coupled/metabolism , Sepsis/prevention & control , Adoptive Transfer , Animals , Artesunate/metabolism , Artesunate/pharmacology , Artesunate/therapeutic use , Disease Models, Animal , Docosahexaenoic Acids/metabolism , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/therapeutic use , Lipopolysaccharides/toxicity , Listeria monocytogenes/pathogenicity , Macrophages/drug effects , Macrophages/pathology , Macrophages/transplantation , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Docking Simulation , Pain/immunology , Pain/mortality , Phagocytosis/drug effects , Plasmodium berghei/pathogenicity , Receptors, G-Protein-Coupled/deficiency , Sepsis/immunology , Sepsis/mortality , Sepsis/therapy
9.
Nature ; 591(7849): 275-280, 2021 03.
Article in English | MEDLINE | ID: mdl-33442058

ABSTRACT

The innate immune regulator STING is a critical sensor of self- and pathogen-derived DNA. DNA sensing by STING leads to the induction of type-I interferons (IFN-I) and other cytokines, which promote immune-cell-mediated eradication of pathogens and neoplastic cells1,2. STING is also a robust driver of antitumour immunity, which has led to the development of STING activators and small-molecule agonists as adjuvants for cancer immunotherapy3. Pain, transmitted by peripheral nociceptive sensory neurons (nociceptors), also aids in host defence by alerting organisms to the presence of potentially damaging stimuli, including pathogens and cancer cells4,5. Here we demonstrate that STING is a critical regulator of nociception through IFN-I signalling in peripheral nociceptors. We show that mice lacking STING or IFN-I signalling exhibit hypersensitivity to nociceptive stimuli and heightened nociceptor excitability. Conversely, intrathecal activation of STING produces robust antinociception in mice and non-human primates. STING-mediated antinociception is governed by IFN-Is, which rapidly suppress excitability of mouse, monkey and human nociceptors. Our findings establish the STING-IFN-I signalling axis as a critical regulator of physiological nociception and a promising new target for treating chronic pain.


Subject(s)
Interferon Type I/metabolism , Membrane Proteins/metabolism , Nociception/physiology , Pain/metabolism , Sensory Receptor Cells/metabolism , Analgesia , Animals , Female , Humans , Interferon Type I/deficiency , Interferon Type I/genetics , Interferon Type I/immunology , Macaca mulatta , Male , Membrane Proteins/agonists , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Nociception/drug effects , Signal Transduction
10.
Sci Rep ; 10(1): 22117, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33335119

ABSTRACT

During development of the peripheral taste system, oral sensory neurons of the geniculate ganglion project via the chorda tympani nerve to innervate taste buds in fungiform papillae. Germline deletion of the p75 neurotrophin receptor causes dramatic axon guidance and branching deficits, leading to a loss of geniculate neurons. To determine whether the developmental functions of p75 in geniculate neurons are cell autonomous, we deleted p75 specifically in Phox2b + oral sensory neurons (Phox2b-Cre; p75fx/fx) or in neural crest-derived cells (P0-Cre; p75fx/fx) and examined geniculate neuron development. In germline p75-/- mice half of all geniculate neurons were lost. The proportion of Phox2b + neurons, as compared to Phox2b-pinna-projecting neurons, was not altered, indicating that both populations were affected similarly. Chorda tympani nerve recordings demonstrated that p75-/- mice exhibit profound deficits in responses to taste and tactile stimuli. In contrast to p75-/- mice, there was no loss of geniculate neurons in either Phox2b-Cre; p75fx/fx or P0-Cre; p75fx/fx mice. Electrophysiological analyses demonstrated that Phox2b-Cre; p75fx/fx mice had normal taste and oral tactile responses. There was a modest but significant loss of fungiform taste buds in Phox2b-Cre; p75fx/fx mice, although there was not a loss of chemosensory innervation of the remaining fungiform taste buds. Overall, these data suggest that the developmental functions of p75 are largely cell non-autonomous and require p75 expression in other cell types of the chorda tympani circuit.


Subject(s)
Geniculate Ganglion/metabolism , Receptors, Nerve Growth Factor/metabolism , Sensory Receptor Cells/metabolism , Alleles , Animals , Biomarkers , Chorda Tympani Nerve/metabolism , Fluorescent Antibody Technique , Genotype , Immunohistochemistry , Mice , Mice, Knockout , Mice, Transgenic , Receptors, Nerve Growth Factor/genetics , Taste/physiology , Touch
11.
iScience ; 23(10): 101570, 2020 Oct 23.
Article in English | MEDLINE | ID: mdl-33083737

ABSTRACT

The immune checkpoint inhibitor programmed cell death protein 1 (PD-1) plays a critical role in immune regulation. Recent studies have demonstrated functional PD-1 expression in peripheral sensory neurons, which contributes to neuronal excitability, pain, and opioid analgesia. Here we report neuronal expression and function of PD-1 in the central nervous system (CNS), including the spinal cord, thalamus, and cerebral cortex. Notably, GABA-induced currents in spinal dorsal horn neurons, thalamic neurons, and cortical neurons are suppressed by the PD-1-neutralizing immunotherapeutic Nivolumab in spinal cord slices, brain slices, and dissociated cortical neurons. Reductions in GABA-mediated currents in CNS neurons were also observed in Pd1 -/- mice without changes in GABA receptor expression. Mechanistically, Nivolumab binds spinal cord neurons and elicits ERK phosphorylation to suppress GABA currents. Finally, both GABA-mediated analgesia and anesthesia are impaired by Pd1 deficiency. Our findings reveal PD-1 as a CNS-neuronal inhibitor that regulates GABAergic signaling and GABA-mediated behaviors.

12.
Cell Tissue Res ; 382(1): 57-64, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32767110

ABSTRACT

The glial cell line-derived neurotrophic factor (GDNF) family ligands (GFLs) comprise a group of four homologous and potent growth factors that includes GDNF, neurturin (NRTN), artemin (ARTN), and persephin (PSPN). The survival, growth, and mitotic activities of the GFLs are conveyed by a single receptor tyrosine kinase, Ret. The GFLs do not bind directly to Ret in order to activate it, and instead bind with high affinity to glycerophosphatidylinositol (GPI)-anchored coreceptors called the GDNF family receptor-αs (GFRαs). Several mechanisms have recently been identified that influence the trafficking of Ret and GFRαs in and out of the plasma membrane, thereby affecting their availability for ligand binding, as well as their levels by targeting to degradative pathways. This review describes these mechanisms and their powerful effects on GFL signaling and function. We also describe the recent discovery that p75 and Ret form a signaling complex, also regulated by plasma membrane shuttling, that either enhances GFL survival signals or p75 pro-apoptotic signals, dependent on the cellular context.


Subject(s)
Cell Membrane/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Protein Transport/physiology , Humans , Signal Transduction
13.
Neurotherapeutics ; 17(3): 846-860, 2020 07.
Article in English | MEDLINE | ID: mdl-32820378

ABSTRACT

Interactions between central glial cells and neurons in the pain circuitry are critical contributors to the pathogenesis of chronic pain. In the central nervous system (CNS), two major glial cell types predominate: astrocytes and microglia. Injuries or pathological conditions which evoke pain are concurrently associated with the presence of a reactive microglia or astrocyte state, which is characterized by a variety of changes in the morphological, molecular, and functional properties of these cells. In this review, we highlight the changes that reactive microglia and astrocytes undergo following painful injuries and insults and discuss the critical and interactive role these two cell types play in the initiation and maintenance of chronic pain. Additionally, we focus on several crucial mechanisms by which microglia and astrocytes contribute to chronic pain and provide commentary on the therapeutic promise of targeting these pathways. In particular, we discuss how the inflammasome in activated microglia drives maturation and release of key pro-inflammatory cytokines, which drive pain through neuronal- and glial regulations. Moreover, we highlight several potentially-druggable hemichannels and proteases produced by reactive microglia and astrocytes in pain states and discuss how these pathways regulate distinct phases during pain pathogenesis. We also review two emerging areas in chronic pain research: 1) sexually dimorphic glial cell signaling and 2) the role of oligodendrocytes. Finally, we highlight important considerations for potential pain therapeutics targeting glial cell mediators as well as questions that remain in our conceptual understanding of glial cell activation in pain states.


Subject(s)
Analgesics/administration & dosage , Central Nervous System/drug effects , Chronic Pain/drug therapy , Drug Delivery Systems/methods , Neuroglia/drug effects , Analgesics/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Central Nervous System/metabolism , Chronic Pain/metabolism , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Humans , Microglia/drug effects , Microglia/metabolism , Neuroglia/metabolism
14.
Trends Neurosci ; 43(10): 822-838, 2020 10.
Article in English | MEDLINE | ID: mdl-32839001

ABSTRACT

Sensory neurons are activated by physical and chemical stimuli, eliciting sensations such as temperature, touch, pain, and itch. From an evolutionary perspective, sensing danger is essential for organismal survival. Upon infection and injury, immune cells respond to pathogen/damage-associated molecular patterns (PAMPs/DAMPs) through pattern recognition receptors (PRRs) such as Toll-like receptors (TLRs), and produce inflammatory mediators that activate sensory neurons through neuro-immune interactions. Sensory neurons also express TLRs and other PRRs that directly sense danger signals after injury or during infection, leading to pain, itch, or analgesia. In addition to slow-acting canonical TLR signaling, TLRs function uniquely in sensory neurons through non-canonical coupling to ion channels, enabling rapid modulation of neuronal activity. We discuss how sensory neurons utilize TLRs and other PRR pathways to detect danger signals in their environment.


Subject(s)
Receptors, Pattern Recognition , Toll-Like Receptors , Humans , Pain , Sensory Receptor Cells , Signal Transduction
15.
Neurotherapeutics ; 17(3): 886-899, 2020 07.
Article in English | MEDLINE | ID: mdl-32696274

ABSTRACT

The current crises in opioid abuse and chronic pain call for the development of nonopioid and nonpharmacological therapeutics for pain relief. Neuromodulation-based approaches, such as spinal cord stimulation, dorsal root ganglion simulation, and nerve stimulation including vagus nerve stimulation, have shown efficacy in achieving pain control in preclinical and clinical studies. However, the mechanisms by which neuromodulation alleviates pain are not fully understood. Accumulating evidence suggests that neuromodulation regulates inflammation and neuroinflammation-a localized inflammation in peripheral nerves, dorsal root ganglia/trigeminal ganglia, and spinal cord/brain-through neuro-immune interactions. Specialized proresolving mediators (SPMs) such as resolvins, protectins, maresins, and lipoxins are lipid molecules produced during the resolution phase of inflammation and exhibit multiple beneficial effects in resolving inflammation in various animal models. Recent studies suggest that SPMs inhibit inflammatory pain, postoperative pain, neuropathic pain, and cancer pain in rodent models via immune, glial, and neuronal modulations. It is noteworthy that sham surgery is sufficient to elevate resolvin levels and may serve as a model of resolution. Interestingly, it has been shown that the vagus nerve produces SPMs and vagus nerve stimulation (VNS) induces SPM production in vitro. In this review, we discuss how neuromodulation such as VNS controls pain via immunomodulation and neuro-immune interactions and highlight possible involvement of SPMs. In particular, we demonstrate that VNS via auricular electroacupuncture effectively attenuates chemotherapy-induced neuropathic pain. Furthermore, auricular stimulation is able to increase resolvin levels in mice. Thus, we propose that neuromodulation may control pain and inflammation/neuroinflammatioin via SPMs. Finally, we discuss key questions that remain unanswered in our understanding of how neuromodulation-based therapies provide short-term and long-term pain relief.


Subject(s)
Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Neuroimmunomodulation/physiology , Pain Management/methods , Pain/metabolism , Analgesics/pharmacology , Analgesics/therapeutic use , Animals , Docosahexaenoic Acids/metabolism , Docosahexaenoic Acids/therapeutic use , Humans , Lipid Metabolism/drug effects , Lipid Metabolism/physiology , Lipoxins/metabolism , Lipoxins/therapeutic use , Neuroimmunomodulation/drug effects , Vagus Nerve Stimulation/methods
16.
J Clin Invest ; 130(7): 3603-3620, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32484460

ABSTRACT

Emerging immune therapy, such as with the anti-programmed cell death-1 (anti-PD-1) monoclonal antibody nivolumab, has shown efficacy in tumor suppression. Patients with terminal cancer suffer from cancer pain as a result of bone metastasis and bone destruction, but how PD-1 blockade affects bone cancer pain remains unknown. Here, we report that mice lacking Pdcd1 (Pd1-/-) demonstrated remarkable protection against bone destruction induced by femoral inoculation of Lewis lung cancer cells. Compared with WT mice, Pd1-/- mice exhibited increased baseline pain sensitivity, but the development of bone cancer pain was compromised in Pd1-/- mice. Consistently, these beneficial effects in Pd1-/- mice were recapitulated by repeated i.v. applications of nivolumab in WT mice, even though nivolumab initially increased mechanical and thermal pain. Notably, PD-1 deficiency or nivolumab treatment inhibited osteoclastogenesis without altering tumor burden. PD-L1 and CCL2 are upregulated within the local tumor microenvironment, and PD-L1 promoted RANKL-induced osteoclastogenesis through JNK activation and CCL2 secretion. Bone cancer upregulated CCR2 in primary sensory neurons, and CCR2 antagonism effectively reduced bone cancer pain. Our findings suggest that, despite a transient increase in pain sensitivity following each treatment, anti-PD-1 immunotherapy could produce long-term benefits in preventing bone destruction and alleviating bone cancer pain by suppressing osteoclastogenesis.


Subject(s)
Bone Neoplasms , Cancer Pain , Carcinoma, Lewis Lung , Neoplasm Proteins , Nivolumab/pharmacology , Osteoclasts/metabolism , Programmed Cell Death 1 Receptor , Animals , Bone Neoplasms/drug therapy , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cancer Pain/drug therapy , Cancer Pain/genetics , Cancer Pain/metabolism , Cancer Pain/pathology , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Female , Mice , Mice, Knockout , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Osteoclasts/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism
17.
Curr Opin Neurobiol ; 62: 17-25, 2020 06.
Article in English | MEDLINE | ID: mdl-31809997

ABSTRACT

Inflammation is the body's protective reaction to injury and infection. Pain is a hallmark of inflammation and can be either protective or detrimental during acute or chronic phase. Macrophages play a chief role in the pathogenesis of pain and have bilateral communications with nociceptors, the specialized primary sensory neurons that sense pain. Macrophages 'talk to' nociceptors by releasing pro-inflammatory mediators (e.g. pro-inflammatory cytokines) that induce pain via direct activation of nociceptors. Macrophages also 'listen to' nociceptors, by which nociceptors secrete neuropeptides and chemokines which act on macrophages. Activation of toll-like receptors (TLRs) in nociceptors releases CCL2, activating macrophages and potentiating pathological pain. Emerging evidence also points to a pro-resolution role of macrophages in inflammation and pain. Macrophage GPR37 is activated by neuroprotectin D1, a specialized pro-resolving mediator (SPM) and resolves inflammatory pain via phagocytosis and production of IL-10 that inhibits nociceptors. Macrophage-nociceptor interactions are also mediated by microRNAs and microRNA-containing exosomes in chronic pain. Notably, extracellular microRNAs (e.g. let-7b and miR-711) can directly bind and activate nociceptors. Targeting macrophage-nociceptor interactions will help to control inflammation and pain.


Subject(s)
Pain , Humans , Inflammation , Macrophages , Neuroimmunomodulation , Nociceptors , Sensory Receptor Cells
18.
J Clin Invest ; 130(4): 1635-1652, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31874109

ABSTRACT

The incidence of human papillomavirus-positive (HPV+) head and neck squamous cell carcinoma (HNSCC) has surpassed that of cervical cancer and is projected to increase rapidly until 2060. The coevolution of HPV with transforming epithelial cells leads to the shutdown of host immune detection. Targeting proximal viral nucleic acid-sensing machinery is an evolutionarily conserved strategy among viruses to enable immune evasion. However, E7 from the dominant HPV subtype 16 in HNSCC shares low homology with HPV18 E7, which was shown to inhibit the STING DNA-sensing pathway. The mechanisms by which HPV16 suppresses STING remain unknown. Recently, we characterized the role of the STING/type I interferon (IFN-I) pathway in maintaining immunogenicity of HNSCC in mouse models. Here we extended those findings into the clinical domain using tissue microarrays and machine learning-enhanced profiling of STING signatures with immune subsets. We additionally showed that HPV16 E7 uses mechanisms distinct from those used by HPV18 E7 to antagonize the STING pathway. We identified NLRX1 as a critical intermediary partner to facilitate HPV16 E7-potentiated STING turnover. The depletion of NLRX1 resulted in significantly improved IFN-I-dependent T cell infiltration profiles and tumor control. Overall, we discovered a unique HPV16 viral strategy to thwart host innate immune detection that can be further exploited to restore cancer immunogenicity.


Subject(s)
Head and Neck Neoplasms/immunology , Human papillomavirus 16/immunology , Membrane Proteins/immunology , Mitochondrial Proteins/immunology , Proteolysis , Squamous Cell Carcinoma of Head and Neck/immunology , Tumor Escape , Animals , Cell Line, Tumor , Female , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/virology , Human papillomavirus 16/genetics , Humans , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , Mitochondrial Proteins/genetics , Papillomavirus E7 Proteins/genetics , Papillomavirus E7 Proteins/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/virology
19.
Neuron ; 103(6): 952-954, 2019 09 25.
Article in English | MEDLINE | ID: mdl-31557456

ABSTRACT

Mechanical itch is a desire to scratch due to light mechanical stimuli. In this issue of Neuron, Pan et al. (2019) identify a feedforward inhibition circuit in the spinal cord dorsal horn that processes mechanical itch as well as spontaneous itch.


Subject(s)
Pruritus , Stroke , Humans , Neurons
20.
Nat Rev Neurosci ; 20(11): 667-685, 2019 11.
Article in English | MEDLINE | ID: mdl-31537912

ABSTRACT

Astrocytes are critical for maintaining the homeostasis of the CNS. Increasing evidence suggests that a number of neurological and neuropsychiatric disorders, including chronic pain, may result from astrocyte 'gliopathy'. Indeed, in recent years there has been substantial progress in our understanding of how astrocytes can regulate nociceptive synaptic transmission via neuronal-glial and glial-glial cell interactions, as well as the involvement of spinal and supraspinal astrocytes in the modulation of pain signalling and the maintenance of neuropathic pain. A role of astrocytes in the pathogenesis of chronic itch is also emerging. These developments suggest that targeting the specific pathways that are responsible for astrogliopathy may represent a novel approach to develop therapies for chronic pain and chronic itch.


Subject(s)
Astrocytes/metabolism , Cell Communication/physiology , Chronic Pain/metabolism , Pruritus/metabolism , Animals , Astrocytes/pathology , Chronic Pain/pathology , Homeostasis/physiology , Humans , Neuroglia/metabolism , Neuroglia/pathology , Pruritus/pathology , Synapses/metabolism , Synapses/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...