Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(10)2024 May 08.
Article in English | MEDLINE | ID: mdl-38791161

ABSTRACT

Adult neurogenesis in the dentate gyrus (DG) is impaired during Alzheimer's disease (AD) progression. Curcumin has been reported to reduce cell apoptosis and stimulate neurogenesis. This study aimed to investigate the influence of curcumin on adult neurogenesis in AD mice and its potential mechanism. Two-month-old male C57BL/6J mice were injected with soluble ß-amyloid (Aß1-42) using lateral ventricle stereolocalization to establish AD models. An immunofluorescence assay, including bromodeoxyuridine (BrdU), doublecortin (DCX), and neuron-specific nuclear antigen (NeuN), was used to detect hippocampal neurogenesis. Western blot and an enzyme-linked immunosorbent assay (ELISA) were used to test the expression of related proteins and the secretion of brain-derived neurotrophic factor (BDNF). A Morris water maze was used to detect the cognitive function of the mice. Our results showed that curcumin administration (100 mg/kg) rescued the impaired neurogenesis of Aß1-42 mice, shown as enhanced BrdU+/DCX+ and BrdU+/NeuN+ cells in DG. In addition, curcumin regulated the phosphatidylinositol 3 kinase (PI3K)/protein kinase B (Akt) -mediated glycogen synthase kinase-3ß (GSK3ß) /Wingless/Integrated (Wnt)/ß-catenin pathway and cyclic adenosine monophosphate response element-binding protein (CREB)/BDNF in Aß1-42 mice. Inhibiting Wnt/ß-catenin and depriving BDNF could reverse both the upregulated neurogenesis and cognitive function of curcumin-treated Aß1-42 mice. In conclusion, our study indicates that curcumin, through targeting PI3K/Akt, regulates GSK3ß/Wnt/ß-catenin and CREB/BDNF pathways, improving the adult neurogenesis of AD mice.


Subject(s)
Alzheimer Disease , Curcumin , Neurogenesis , Wnt Signaling Pathway , Animals , Male , Mice , Alzheimer Disease/metabolism , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , beta Catenin/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Curcumin/pharmacology , Disease Models, Animal , Doublecortin Protein/metabolism , Hippocampus/metabolism , Hippocampus/drug effects , Mice, Inbred C57BL , Neurogenesis/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Up-Regulation/drug effects , Wnt Signaling Pathway/drug effects
2.
Cell ; 187(4): 914-930.e20, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38280375

ABSTRACT

The gut and liver are recognized to mutually communicate through the biliary tract, portal vein, and systemic circulation. However, it remains unclear how this gut-liver axis regulates intestinal physiology. Through hepatectomy and transcriptomic and proteomic profiling, we identified pigment epithelium-derived factor (PEDF), a liver-derived soluble Wnt inhibitor, which restrains intestinal stem cell (ISC) hyperproliferation to maintain gut homeostasis by suppressing the Wnt/ß-catenin signaling pathway. Furthermore, we found that microbial danger signals resulting from intestinal inflammation can be sensed by the liver, leading to the repression of PEDF production through peroxisome proliferator-activated receptor-α (PPARα). This repression liberates ISC proliferation to accelerate tissue repair in the gut. Additionally, treating mice with fenofibrate, a clinical PPARα agonist used for hypolipidemia, enhances colitis susceptibility due to PEDF activity. Therefore, we have identified a distinct role for PEDF in calibrating ISC expansion for intestinal homeostasis through reciprocal interactions between the gut and liver.


Subject(s)
Intestines , Liver , Animals , Mice , Cell Proliferation , Liver/metabolism , PPAR alpha/metabolism , Proteomics , Stem Cells/metabolism , Wnt Signaling Pathway , Intestines/cytology , Intestines/metabolism
3.
Biomed Pharmacother ; 168: 115800, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37935070

ABSTRACT

Gamma-aminobutyric acid (GABA), a non-protein-producing amino acid synthesized from the excitatory amino acid glutamate via the enzyme glutamic acid decarboxylase, is extensively found in microorganisms, plants and vertebrates, and is abundantly expressed in the spinal cord and brain. It is the major inhibitory neurotransmitter in the mammalian nervous system. GABA plays crucial roles in the regulation of synaptic transmission, the promotion of neuronal development and relaxation, and the prevention of insomnia and depression. As the major inhibitory neurotransmitter, GABA plays pivotal roles in the regulation of pain sensation, which is initiated by the activation of peripheral nociceptors and transmitted to the spinal cord and brain along nerves. GABA exerts these roles by directly acting on three types of receptors: ionotropic GABAA and GABAC receptors and G protein-coupled GABAB receptor. The chloride-permeable ion channel receptors GABAA and GABAC mediate fast neurotransmission, while the metabotropic GABAB receptor mediates slow effect. Different GABA receptors regulate pain sensation via different signaling pathways. Here we highlight recent updates on the involvement of specific GABA receptors and their subtypes in the process of pain sensation. Further understanding of different GABA receptors and signaling pathways in pain sensation will benefit the development of novel analgesics for pain management by targeting specific GABA receptor subtypes and signaling pathways.


Subject(s)
Analgesia , Receptors, GABA , Animals , Receptors, GABA/metabolism , Pain Management , Pain/drug therapy , gamma-Aminobutyric Acid/metabolism , Chloride Channels , Receptors, G-Protein-Coupled/metabolism , Glutamic Acid , Neurotransmitter Agents , Mammals
4.
Sci Adv ; 9(28): eadf3924, 2023 07 14.
Article in English | MEDLINE | ID: mdl-37436991

ABSTRACT

Peripheral regulatory T (pTreg) cells are a key T cell lineage for mucosal immune tolerance and anti-inflammatory responses, and interleukin-2 receptor (IL-2R) signaling is critical for Treg cell generation, expansion, and maintenance. The expression of IL-2R on pTreg cells is tightly regulated to ensure proper induction and function of pTreg cells without a clear molecular mechanism. We here demonstrate that Cathepsin W (CTSW), a cysteine proteinase highly induced in pTreg cells under transforming growth factor-ß stimulation is essential for the restraint of pTreg cell differentiation in an intrinsic manner. Loss of CTSW results in elevated pTreg cell generation, protecting the animals from intestinal inflammation. Mechanistically, CTSW inhibits IL-2R signaling in pTreg cells by cytosolic interaction with and process of CD25, repressing signal transducer and activator of transcription 5 activation to restrain pTreg cell generation and maintenance. Hence, our data indicate that CTSW acts as a gatekeeper to calibrate pTreg cell differentiation and function for mucosal immune quiescence.


Subject(s)
T-Lymphocytes, Regulatory , Animals , Cathepsin W , Cell Differentiation , Cell Division , Cell Lineage
5.
Immunity ; 56(7): 1533-1547.e7, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37354904

ABSTRACT

The crosstalk between the immune and neuroendocrine systems is critical for intestinal homeostasis and gut-brain communications. However, it remains unclear how immune cells participate in gut sensation of hormones and neurotransmitters release in response to environmental cues, such as self-lipids and microbial lipids. We show here that lipid-mediated engagement of invariant natural killer T (iNKT) cells with enterochromaffin (EC) cells, a subset of intestinal epithelial cells, promoted peripheral serotonin (5-HT) release via a CD1d-dependent manner, regulating gut motility and hemostasis. We also demonstrated that inhibitory sphingolipids from symbiotic microbe Bacteroides fragilis represses 5-HT release. Mechanistically, CD1d ligation on EC cells transduced a signal and restrained potassium conductance through activation of protein tyrosine kinase Pyk2, leading to calcium influx and 5-HT secretion. Together, our data reveal that by engaging with iNKT cells, gut chemosensory cells selectively perceive lipid antigens via CD1d to control 5-HT release, modulating intestinal and systemic homeostasis.


Subject(s)
Natural Killer T-Cells , Serotonin , Serotonin/metabolism , Lipids , Antigens, CD1d/metabolism
6.
Front Chem ; 10: 1036954, 2022.
Article in English | MEDLINE | ID: mdl-36438868

ABSTRACT

Resin glycosides, mainly distributed in plants of the family Convolvulaceae, are a class of novel and complex glycolipids. Their structural complexity and significant biological activities have received much attention from synthetic chemists, and a number of interesting resin glycosides have been synthesized. The synthesized resin glycosides and their analogues not only helped in structural verification, structural modification, and further biological activity exploration but also provided enlightenment for the synthesis of glycoside compounds. Herein, the present review summarizes the application of various efforts toward the synthesis of resin glycosides in the last decade.

7.
J Neurosci ; 42(31): 6090-6107, 2022 08 03.
Article in English | MEDLINE | ID: mdl-35760529

ABSTRACT

Alzheimer's disease (AD) is characterized pathologically by the structural and functional impairments of synapses in the hippocampus, inducing the learning and memory deficiencies. Ras GTPase is closely related to the synaptic function and memory. This study was to investigate the effects of farnesyl transferase inhibitor lonafarnib on the synaptic structure and function in AD male mice and explore the potential mechanism. Our results showed 50 mg/kg lonafarnib (intraperitoneal) rescued the impaired spatial memory and improved the damaged synaptic transmission and plasticity of Aß1-42 mice. In addition, lonafarnib ameliorated the morphology of synaptic dendrites and spines in Aß1-42 mice. Furthermore, lonafarnib enhanced α7nAChR cell surface expression and phosphorylation of downstream Akt and CaMKII in Aß1-42 mice, which were inhibited by α7nAChR antagonist methyl lycaconitine (MLA), and increased the phosphorylation of CREB in a CaMKII- but not ERK-dependent way. Lonafarnib enhanced hippocampal brain-derived neurotrophic factor (BDNF) concentration in Aß1-42 mice, which was sensitive to MLA and KN93 (an inhibitor of CaMKII), but not related to ERK and Akt pathways. H-Ras, but not Rhes, was related to the lonafarnib induced improvement of α7nAChR cell surface expression and BDNF content. Interestingly, lonafarnib induced improvement of synaptic transmission, plasticity and spatial cognition in Aß1-42 mice was abolished by BDNF deprivation with TrkB/Fc chimera protein. Our results indicate that lonafarnib can rescue the structural and functional impairments of synapses in the Aß1-42 mice, which may be related to the improvement of BDNF content through the H-Ras-α7nAChR-dependent CaMKII-CREB pathway, leading to the improvement of spatial cognition.SIGNIFICANCE STATEMENT Alzheimer's disease (AD) is characterized pathologically by the structural and functional impairments of synapses in the hippocampus, inducing the learning and memory deficiencies. However, no effective drugs have not been developed for the treatment of AD synaptic. This study for the first time reported the beneficial effects of Ras inhibitor lonafarnib on the synaptic structure and function in AD mice, providing an alternative way for the treatment of "synaptic disease" in AD patients.


Subject(s)
Alzheimer Disease , Brain-Derived Neurotrophic Factor , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Hippocampus/metabolism , Male , Memory Disorders , Mice , Peptide Fragments , Piperidines , Proto-Oncogene Proteins c-akt/metabolism , Pyridines , Spatial Memory , Synapses/physiology , Up-Regulation , alpha7 Nicotinic Acetylcholine Receptor/metabolism
8.
Front Cell Neurosci ; 16: 799717, 2022.
Article in English | MEDLINE | ID: mdl-35317196

ABSTRACT

How the enteric nervous system determines the pacing and propagation direction of neurogenic contractions along the colon remains largely unknown. We used a chemogenetic strategy to ablate enteric neurons expressing calretinin (CAL). Mice expressing human diphtheria toxin receptor (DTR) in CAL neurons were generated by crossing CAL-ires-Cre mice with Cre-dependent ROSA26-DTR mice. Immunohistochemical analysis revealed treatment with diphtheria toxin incurred a 42% reduction in counts of Hu-expressing colonic myenteric neurons (P = 0.036), and 57% loss of CAL neurons (comprising ∼25% of all Hu neurons; P = 0.004) compared to control. As proportions of Hu-expressing neurons, CAL neurons that contained nitric oxide synthase (NOS) were relatively spared (control: 15 ± 2%, CAL-DTR: 13 ± 1%; P = 0.145), while calretinin neurons lacking NOS were significantly reduced (control: 26 ± 2%, CAL-DTR: 18 ± 5%; P = 0.010). Colonic length and pellet sizes were significantly reduced without overt inflammation or changes in ganglionic density. Interestingly, colonic motor complexes (CMCs) persisted with increased frequency (mid-colon interval 111 ± 19 vs. 189 ± 24 s, CAL-DTR vs. control, respectively, P < 0.001), decreased contraction size (mid-colon AUC 26 ± 24 vs. 59 ± 13 gram/seconds, CAL-DTR vs. control, respectively, P < 0.001), and lacked preferential anterograde migration (P < 0.001). The functional effects of modest calretinin neuron ablation, particularly increased neurogenic motor activity frequencies, differ from models that incur general enteric neuron loss, and suggest calretinin neurons may contribute to pacing, force, and polarity of CMCs in the large bowel.

9.
Cell ; 185(7): 1172-1188.e28, 2022 03 31.
Article in English | MEDLINE | ID: mdl-35303419

ABSTRACT

Intestinal mucus forms the first line of defense against bacterial invasion while providing nutrition to support microbial symbiosis. How the host controls mucus barrier integrity and commensalism is unclear. We show that terminal sialylation of glycans on intestinal mucus by ST6GALNAC1 (ST6), the dominant sialyltransferase specifically expressed in goblet cells and induced by microbial pathogen-associated molecular patterns, is essential for mucus integrity and protecting against excessive bacterial proteolytic degradation. Glycoproteomic profiling and biochemical analysis of ST6 mutations identified in patients show that decreased sialylation causes defective mucus proteins and congenital inflammatory bowel disease (IBD). Mice harboring a patient ST6 mutation have compromised mucus barriers, dysbiosis, and susceptibility to intestinal inflammation. Based on our understanding of the ST6 regulatory network, we show that treatment with sialylated mucin or a Foxo3 inhibitor can ameliorate IBD.


Subject(s)
Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Sialyltransferases/genetics , Animals , Homeostasis , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Mice , Mucus/metabolism , Sialyltransferases/metabolism , Symbiosis
10.
Blood ; 139(12): 1878-1891, 2022 03 24.
Article in English | MEDLINE | ID: mdl-34871362

ABSTRACT

Peripheral serotonin (5-HT) is mainly generated from the gastrointestinal tract and taken up and stored by platelets in the circulation. Although the gut is recognized as a major immune organ, how intestinal local immune responses control whole-body physiology via 5-HT remains unclear. Here, we show that intestinal inflammation enhances systemic platelet activation and blood coagulation. Intestinal epithelium damage induces elevated levels of the alarm cytokine interleukin-33 (IL-33), leading to platelet activation via promotion of gut-derived 5-HT release. More importantly, we found that loss of intestinal epithelial-derived IL-33 lowers peripheral 5-HT levels, resulting in compromised platelet activation and hemostasis. Functionally, intestinal IL-33 contributes to the recruitment of neutrophils to sites of acute inflammation by enhancing platelet activities. Genetic deletion of intestinal IL-33 or neutralization of peripheral IL-33 protects animals from lipopolysaccharide endotoxic shock through attenuated neutrophil extravasation. Therefore, our data establish a distinct role of intestinal IL-33 in activating platelets by promoting 5-HT release for systemic physiology and inflammation.


Subject(s)
Interleukin-33 , Serotonin , Animals , Inflammation , Neutrophil Infiltration , Neutrophils
11.
Ann Rheum Dis ; 80(12): 1604-1614, 2021 12.
Article in English | MEDLINE | ID: mdl-34663597

ABSTRACT

Crystal structures activate innate immune cells, especially macrophages and initiate inflammatory responses. We aimed to understand the role of the mechanosensitive TRPV4 channel in crystal-induced inflammation. Real-time RT-PCR, RNAscope in situ hybridisation, and Trpv4eGFP mice were used to examine TRPV4 expression and whole-cell patch-clamp recording and live-cell Ca2+ imaging were used to study TRPV4 function in mouse synovial macrophages and human peripheral blood mononuclear cells (PBMCs). Both genetic deletion and pharmacological inhibition approaches were used to investigate the role of TRPV4 in NLRP3 inflammasome activation induced by diverse crystals in vitro and in mouse models of crystal-induced pain and inflammation in vivo. TRPV4 was functionally expressed by synovial macrophages and human PBMCs and TRPV4 expression was upregulated by stimulation with monosodium urate (MSU) crystals and in human PBMCs from patients with acute gout flares. MSU crystal-induced gouty arthritis were significantly reduced by either genetic ablation or pharmacological inhibition of TRPV4 function. Mechanistically, TRPV4 mediated the activation of NLRP3 inflammasome by diverse crystalline materials but not non-crystalline NLRP3 inflammasome activators, driving the production of inflammatory cytokine interleukin-1ß which elicited TRPV4-dependent inflammatory responses in vivo. Moreover, chemical ablation of the TRPV1-expressing nociceptors significantly attenuated the MSU crystal-induced gouty arthritis. In conclusion, TRPV4 is a common mediator of inflammatory responses induced by diverse crystals through NLRP3 inflammasome activation in macrophages. TRPV4-expressing resident macrophages are critically involved in MSU crystal-induced gouty arthritis. A neuroimmune interaction between the TRPV1-expressing nociceptors and the TRPV4-expressing synovial macrophages contributes to the generation of acute gout flares.


Subject(s)
Arthralgia/metabolism , Arthritis/metabolism , Crystal Arthropathies/metabolism , Leukocytes, Mononuclear/metabolism , Macrophages/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Nociceptors/metabolism , TRPV Cation Channels/genetics , Adult , Animals , Arthralgia/immunology , Arthritis/immunology , Arthritis, Gouty/immunology , Arthritis, Gouty/metabolism , Crystal Arthropathies/immunology , Gout/immunology , Gout/metabolism , Humans , Inflammasomes/immunology , Inflammation , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Leukocytes, Mononuclear/immunology , Macrophages/immunology , Male , Mice , Middle Aged , Optical Imaging , Patch-Clamp Techniques , Synovial Membrane/cytology , THP-1 Cells , TRPV Cation Channels/agonists , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/metabolism , Uric Acid
12.
J Exp Med ; 218(9)2021 09 06.
Article in English | MEDLINE | ID: mdl-34287641

ABSTRACT

Mucus produced by goblet cells in the gastrointestinal tract forms a biological barrier that protects the intestine from invasion by commensals and pathogens. However, the host-derived regulatory network that controls mucus secretion and thereby changes gut microbiota has not been well studied. Here, we identify that Forkhead box protein O1 (Foxo1) regulates mucus secretion by goblet cells and determines intestinal homeostasis. Loss of Foxo1 in intestinal epithelial cells (IECs) results in defects in goblet cell autophagy and mucus secretion, leading to an impaired gut microenvironment and dysbiosis. Subsequently, due to changes in microbiota and disruption in microbiome metabolites of short-chain fatty acids, Foxo1 deficiency results in altered organization of tight junction proteins and enhanced susceptibility to intestinal inflammation. Our study demonstrates that Foxo1 is crucial for IECs to establish commensalism and maintain intestinal barrier integrity by regulating goblet cell function.


Subject(s)
Forkhead Box Protein O1/metabolism , Gastrointestinal Microbiome/physiology , Gastrointestinal Tract/physiology , Mucus/metabolism , Animals , Autophagy/physiology , Colitis/chemically induced , Colitis/metabolism , Colitis/microbiology , Dysbiosis/genetics , Fatty Acids, Volatile/metabolism , Female , Forkhead Box Protein O1/genetics , Goblet Cells/pathology , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Male , Mice, Inbred C57BL , Mucin-2/metabolism , Symbiosis/physiology
13.
J Neurochem ; 158(6): 1381-1393, 2021 09.
Article in English | MEDLINE | ID: mdl-33930191

ABSTRACT

Neurodegenerative disorders, such as Alzheimer's disease and Parkinson's disease, are devastating diseases in the elderly world, which are closely associated with progressive neuronal loss induced by a variety of genetic and/or environmental factors. Unfortunately, currently available treatments for neurodegenerative disorders can only relieve the symptoms but not modify the pathological processes. Over the past decades, our group by collaborating with Profs. Yuan-Ping Pang and Paul R. Carlier has developed three series of homo/hetero dimeric acetylcholinesterase inhibitors derived from tacrine and/or huperzine A. The representative dimers bis(3)-Cognitin (B3C), bis(12)-hupyridone, and tacrine(10)-hupyridone might possess disease-modifying effects through the modulation of N-methyl-d-aspartic acid receptors, the activation of myocyte enhancer factor 2D gene transcription, and the promotion of neurotrophic factor secretion. In this review, we summarize that the representative dimers, such as B3C, provide neuroprotection against a variety of neurotoxins via multiple targets, including the inhibitions of N-methyl-d-aspartic acid receptor with pathological-activated potential, neuronal nitric oxide synthase, and ß-amyloid cascades synergistically. More importantly, B3C might offer disease-modifying potentials by activating myocyte enhancer factor 2D transcription, inducing neuritogenesis, and promoting the expressions of neurotrophic factors in vitro and in vivo. Taken together, the novel dimers might offer synergistic disease-modifying effects, proving that dimerization might serve as one of the strategies to develop new generation of therapeutics for neurodegenerative disorders.


Subject(s)
Acetylcholinesterase/metabolism , Alkaloids/administration & dosage , Cholinesterase Inhibitors/administration & dosage , Drug Delivery Systems/methods , Neurodegenerative Diseases/drug therapy , Sesquiterpenes/administration & dosage , Tacrine/administration & dosage , Alkaloids/chemistry , Animals , Cholinesterase Inhibitors/chemistry , Drug Combinations , Drug Delivery Systems/trends , Humans , Neurodegenerative Diseases/diagnosis , Neurodegenerative Diseases/enzymology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/chemistry , Sesquiterpenes/chemistry , Tacrine/chemistry
14.
Immunity ; 54(1): 151-163.e6, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33220232

ABSTRACT

The gastrointestinal tract is known as the largest endocrine organ that encounters and integrates various immune stimulations and neuronal responses due to constant environmental challenges. Enterochromaffin (EC) cells, which function as chemosensors on the gut epithelium, are known to translate environmental cues into serotonin (5-HT) production, contributing to intestinal physiology. However, how immune signals participate in gut sensation and neuroendocrine response remains unclear. Interleukin-33 (IL-33) acts as an alarmin cytokine by alerting the system of potential environmental stresses. We here demonstrate that IL-33 induced instantaneous peristaltic movement and facilitated Trichuris muris expulsion. We found that IL-33 could be sensed by EC cells, inducing release of 5-HT. IL-33-mediated 5-HT release activated enteric neurons, subsequently promoting gut motility. Mechanistically, IL-33 triggered calcium influx via a non-canonical signaling pathway specifically in EC cells to induce 5-HT secretion. Our data establish an immune-neuroendocrine axis in calibrating rapid 5-HT release for intestinal homeostasis.


Subject(s)
Enterochromaffin Cells/physiology , Interleukin-33/metabolism , Intestines/physiology , Neurons/physiology , Serotonin/metabolism , Trichuriasis/immunology , Trichuris/physiology , Animals , Calcium Signaling , Homeostasis , Interleukin-33/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuroimmunomodulation , Peristalsis
15.
Br J Pharmacol ; 177(18): 4181-4192, 2020 09.
Article in English | MEDLINE | ID: mdl-32542737

ABSTRACT

BACKGROUND AND PURPOSE: Hair follicle telogen to anagen transition results in a break in cellular quiescence of the hair follicle stem cells, which subsequently promotes hair follicle regeneration. Many critical molecules and signalling pathways are involved in hair follicle cycle progression. Transient receptor potential vanilloid 4 (TRPV4) is a polymodal sensory transducer that regulates various cutaneous functions under both normal and disease conditions. However, the role of TRPV4 in hair follicle regeneration in vivo remains incompletely understood. EXPERIMENTAL APPROACH: Using adult C57BL/6J mice, keratinocyte (K14Cre ; Trpv4f/f ) and macrophage (Cx3cr1Cre ; Trpv4f/f ) Trpv4 conditional knockout (cKO) mice, Trpv4-/- mice, we investigated the effect of a single intradermal injection of GSK1016790A, a potent and selective small molecule TRPV4 activator, on hair follicle regeneration. Chemical cues and signal molecules involved in hair follicle cycle progression were measured by immunofluorescence staining, quantitative RT-PCR and western blotting. KEY RESULTS: Here, we show that a single intradermal injection of GSK1016790A is sufficient to induce telogen to anagen transition and hair follicle regeneration in mice by increasing the expression of the anagen-promoting growth factors and down-regulating the expression of growth factors that inhibit anagen. The action of GSK1016790A relies largely on the function of TRPV4 in skin and involves activation of downstream ERK signalling. CONCLUSION AND IMPLICATIONS: Our results suggest that transient chemical activation of TRPV4 in the skin induces hair follicle regeneration in mice, which might provide an effective therapeutic strategy for the treatment of hair loss and alopecia.


Subject(s)
Hair Follicle , Regeneration , TRPV Cation Channels , Alopecia/drug therapy , Animals , Female , Keratinocytes , Male , Mice , Mice, Inbred C57BL , TRPV Cation Channels/genetics
16.
Immunity ; 49(1): 107-119.e4, 2018 07 17.
Article in English | MEDLINE | ID: mdl-29958798

ABSTRACT

Intestinal macrophages are critical for gastrointestinal (GI) homeostasis, but our understanding of their role in regulating intestinal motility is incomplete. Here, we report that CX3C chemokine receptor 1-expressing muscularis macrophages (MMs) were required to maintain normal GI motility. MMs expressed the transient receptor potential vanilloid 4 (TRPV4) channel, which senses thermal, mechanical, and chemical cues. Selective pharmacologic inhibition of TRPV4 or conditional deletion of TRPV4 from macrophages decreased intestinal motility and was sufficient to reverse the GI hypermotility that is associated with chemotherapy treatment. Mechanistically, stimulation of MMs via TRPV4 promoted the release of prostaglandin E2 and elicited colon contraction in a paracrine manner via prostaglandin E receptor signaling in intestinal smooth muscle cells without input from the enteric nervous system. Collectively, our data identify TRPV4-expressing MMs as an essential component required for maintaining normal GI motility and provide potential drug targets for GI motility disorders.


Subject(s)
Colon/physiology , Gastrointestinal Motility , Macrophages/metabolism , Myocytes, Smooth Muscle/metabolism , Signal Transduction , TRPV Cation Channels/metabolism , Animals , CX3C Chemokine Receptor 1/metabolism , Colon/physiopathology , Cyclooxygenase 1/deficiency , Cyclooxygenase 1/metabolism , Dinoprostone/analysis , Dinoprostone/metabolism , Female , Gastric Mucosa/cytology , Gene Expression , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Mice , Mice, Knockout , Muscle Contraction , Receptors, Prostaglandin E/antagonists & inhibitors , Receptors, Prostaglandin E/metabolism , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/deficiency , TRPV Cation Channels/genetics
17.
Science ; 360(6388): 530-533, 2018 05 04.
Article in English | MEDLINE | ID: mdl-29724954

ABSTRACT

The somatosensory system relays many signals ranging from light touch to pain and itch. Touch is critical to spatial awareness and communication. However, in disease states, innocuous mechanical stimuli can provoke pathologic sensations such as mechanical itch (alloknesis). The molecular and cellular mechanisms that govern this conversion remain unknown. We found that in mice, alloknesis in aging and dry skin is associated with a loss of Merkel cells, the touch receptors in the skin. Targeted genetic deletion of Merkel cells and associated mechanosensitive Piezo2 channels in the skin was sufficient to produce alloknesis. Chemogenetic activation of Merkel cells protected against alloknesis in dry skin. This study reveals a previously unknown function of the cutaneous touch receptors and may provide insight into the development of alloknesis.


Subject(s)
Ion Channels/physiology , Mechanotransduction, Cellular/physiology , Merkel Cells/physiology , Pruritus/physiopathology , Skin/innervation , Touch , Animals , Gene Deletion , Ion Channels/genetics , Mechanotransduction, Cellular/genetics , Merkel Cells/drug effects , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Pruritus/genetics
18.
Gastroenterology ; 155(2): 514-528.e6, 2018 08.
Article in English | MEDLINE | ID: mdl-29782847

ABSTRACT

BACKGROUND & AIMS: Strategies are needed to increase gastrointestinal transit without systemic pharmacologic agents. We investigated whether optogenetics, focal application of light to control enteric nervous system excitability, could be used to evoke propagating contractions and increase colonic transit in mice. METHODS: We generated transgenic mice with Cre-mediated expression of light-sensitive channelrhodopsin-2 (ChR2) in calretinin neurons (CAL-ChR2 Cre+ mice); Cre- littermates served as controls. Colonic myenteric neurons were analyzed by immunohistochemistry, patch-clamp, and calcium imaging studies. Motility was assessed by mechanical, electrophysiological, and video recording in vitro and by fecal output in vivo. RESULTS: In isolated colons, focal light stimulation of calretinin enteric neurons evoked classic polarized motor reflexes (50/58 stimulations), followed by premature anterograde propagating contractions (39/58 stimulations). Light stimulation could evoke motility from sites along the entire colon. These effects were prevented by neural blockade with tetrodotoxin (n = 2), and did not occur in control mice (n = 5). Light stimulation of proximal colon increased the proportion of natural fecal pellets expelled over 15 minutes in vitro (75% ± 17% vs 32% ± 8% for controls) (P < .05). In vivo, activation of wireless light-emitting diodes implanted onto the colon wall significantly increased hourly fecal pellet output in conscious, freely moving mice (4.2 ± 0.4 vs 1.3 ± 0.3 in controls) (P < .001). CONCLUSIONS: In studies of mice, we found that focal activation of a subset of enteric neurons can increase motility of the entire colon in vitro, and fecal output in vivo. Optogenetic control of enteric neurons might therefore be used to modify gut motility.


Subject(s)
Colon/physiology , Enteric Nervous System/physiology , Gastrointestinal Transit/radiation effects , Light , Optogenetics/methods , Animals , Calbindin 2/genetics , Calbindin 2/metabolism , Channelrhodopsins/genetics , Channelrhodopsins/metabolism , Channelrhodopsins/radiation effects , Colon/innervation , Colon/radiation effects , Enteric Nervous System/cytology , Gastrointestinal Transit/genetics , Mice , Mice, Transgenic , Models, Animal , Neurons/metabolism , Neurons/radiation effects
19.
J Allergy Clin Immunol ; 141(2): 608-619.e7, 2018 02.
Article in English | MEDLINE | ID: mdl-28807414

ABSTRACT

BACKGROUND: Chronic itch is a highly debilitating symptom that underlies many medical disorders with no universally effective treatments. Although unique neuronal signaling cascades in the sensory ganglia and spinal cord have been shown to critically promote the pathogenesis of chronic itch, the role of skin-associated cells remains poorly understood. OBJECTIVE: We sought to examine the cutaneous mechanisms underlying transient receptor potential vanilloid 4 (TRPV4)-mediated allergic and nonallergic chronic itch. METHODS: Expression of TRPV4 in chronic itch and healthy control skin preparations was examined by using real-time RT-PCR. Trpv4eGFP mice were used to study the expression and function of TRPV4 in the skin by means of immunofluorescence staining, flow cytometry, calcium imaging, and patch-clamp recordings. Genetic and pharmacologic approaches were used to examine the role and underlying mechanisms of TRPV4 in mouse models of dry skin-associated chronic itch and spontaneous scratching associated with squaric acid dibutylester-induced allergic contact dermatitis. RESULTS: TRPV4 is selectively expressed by dermal macrophages and epidermal keratinocytes in mice. Lineage-specific deletion of TRPV4 in macrophages and keratinocytes reduces allergic and nonallergic chronic itch in mice, respectively. Importantly, TRPV4 expression is significantly increased in skin biopsy specimens from patients with chronic idiopathic pruritus in comparison with skin from healthy control subjects. Moreover, TRPV4-dependent chronic itch requires 5-hydroxytryptamine (5-HT) signaling secondary to activation of distinct 5-HT receptors in mice with allergic and those with nonallergic chronic itch conditions. CONCLUSION: Our study reveals previously unrecognized mechanisms by which TRPV4-expressing epithelial and immune cells in the skin critically and dynamically mediate chronic itch and unravels novel targets for therapeutics in the setting of chronic itch.


Subject(s)
Dermatitis, Allergic Contact/immunology , Dermis/immunology , Gene Expression Regulation/immunology , Keratinocytes/immunology , Macrophages/immunology , Pruritus/immunology , TRPV Cation Channels/immunology , Animals , Chronic Disease , Dermatitis, Allergic Contact/genetics , Dermatitis, Allergic Contact/pathology , Dermis/pathology , Female , Gene Expression Regulation/genetics , Humans , Keratinocytes/pathology , Macrophages/pathology , Male , Mice , Mice, Knockout , Pruritus/genetics , Pruritus/pathology , TRPV Cation Channels/genetics
20.
Eur J Pharmacol ; 820: 39-44, 2018 Feb 05.
Article in English | MEDLINE | ID: mdl-29198958

ABSTRACT

Gastrointestinal tract motility may be demoted significantly after surgery operations at least in part due to anaesthetic agents, but there is no comprehensive explanation of the molecular mechanism(s) of such adverse effects. Anesthetics are known to interact with various receptors and ion channels including several subtypes of transient receptor potential (TRP) channels. Two members of the canonical subfamily of TRP channels (TRPC), TRPC4 and TRPC6 are Ca2+-permeable cation channels involved in visceral smooth muscle contractility induced by acetylcholine, the primary excitatory neurotransmitter in the gut. In the present study, we aimed to study the effect of anesthetics on muscarinic receptor-mediated excitation and contraction of intestinal smooth muscle. Here we show that muscarinic cation current (mICAT) mediated by TRPC4 and TRPC6 channels in mouse ileal myocytes was strongly inhibited by isoflurane (0.5mM), one of the most commonly used inhalation anesthetics. Carbachol-activated mICAT was reduced by 63 ± 11% (n = 5), while GTPγS-induced (to bypass muscarinic receptors) current was inhibited by 44 ± 9% (n = 6). Furthermore, carbachol-induced ileum and colon contractions were inhibited by isoflurane by about 30%. We discuss the main sites of isoflurane action, which appear to be G-proteins and muscarinic receptors, rather than TRPC4/6 channels. These results contribute to our better understanding of the signalling pathways affected by inhalation anesthetics, which may cause ileus, and thus may be important for the development of novel treatment strategies during postoperative recovery.


Subject(s)
Carbachol/antagonists & inhibitors , Intestines/drug effects , Isoflurane/pharmacology , Muscle Contraction/drug effects , Muscle, Smooth/physiology , TRPC Cation Channels/metabolism , TRPC6 Cation Channel/metabolism , Anesthetics, Inhalation/pharmacology , Animals , Calcium/metabolism , Carbachol/pharmacology , Electrophysiological Phenomena/drug effects , Intestines/physiology , Intracellular Space/drug effects , Intracellular Space/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle, Smooth/cytology , Muscle, Smooth/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL