Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.610
Filter
1.
Zhen Ci Yan Jiu ; 49(8): 829-835, 2024 Aug 25.
Article in English, Chinese | MEDLINE | ID: mdl-39318312

ABSTRACT

OBJECTIVES: To observe the effect of electroacupuncture (EA) on pain, anxiety like behavior, and substance P(SP) /neurokinin-1 receptor (NK1R) /ß -arrestin 1(ARRB1) pathway related protein expression in hippocampus of chronic constriction injury (CCI) rats, so as to explore its mechanisms underlying improvement of neuropathic pain. METHODS: Twenty-seven male SD rats were randomly divided into sham operation, model and EA groups, with 9 rats in each group. The CCI model was established by ligature of the left sciatic nerve. On the 8th day following modeling, EA (2 Hz, 0.5-1.5 mA) was applied to the left "Huantiao" (GB34) and "Yanglingquan" (GB34) for 30 min, once every other day for 13 times. Mechanical paw withdrawal threshold (MWT), thermal paw withdrawal threshold (TWL) and difference of the weight distribution of the hind limbs were detected before operation and at the 5th, 9th, 17th, 25th and 33rd days after operation. Open field test was used to evaluate the anxiety-like behavior of rats. The content of SP in hippocampus was determined by ELISA. The protein expression of NK1R and ARRB1 in hippocampus was detected by Western blot. RESULTS: Compared with the sham operation group, the MWT and TWL of the left hind limb at the 5th, 9th, 17th, 25th and 33rd days after operation, the time of entering the central area and the total distance of movement, and the content of SP in the hippocampus were significantly decreased (P<0.001, P<0.01), while the difference of the weight distribution of the hind limbs at the 5th, 9th, 17th, 25th and 33rd days after operation and the protein expression of NK1R and ARRB1 were significantly increased (P<0.001, P<0.05) in the model group. After EA intervention, the MWT and TWL of the left hind limb, the time of entering the central area and the total moving distance, and the expression of SP in the hippocampus were significantly increased (P<0.01, P<0.001, P<0.05), while the difference in the weight distribution of the hind limbs was significantly reduced, and the expression of NK1R and ARRB1 protein in the hippocampus were significantly decreased (P<0.001, P<0.05) in the EA group. CONCLUSIONS: EA can effectively improve the pain and anxiety behaviors in CCI rats, and reverse the abnormal expression of SP, NK1R and ARRB1 proteins in the hippocampus, which may be related to its effects in regulating the SP/NK1R/ARRB1 pathway in the hippocampus.


Subject(s)
Electroacupuncture , Hippocampus , Neuralgia , Rats, Sprague-Dawley , Receptors, Neurokinin-1 , Substance P , Animals , Male , Rats , Hippocampus/metabolism , Neuralgia/therapy , Neuralgia/metabolism , Neuralgia/genetics , Receptors, Neurokinin-1/metabolism , Receptors, Neurokinin-1/genetics , Humans , Substance P/metabolism , Substance P/genetics , beta-Arrestin 1/metabolism , beta-Arrestin 1/genetics , Acupuncture Points , Signal Transduction
2.
Nature ; 633(8028): 207-215, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39112700

ABSTRACT

Tumour innervation is associated with worse patient outcomes in multiple cancers1,2, which suggests that it may regulate metastasis. Here we observed that highly metastatic mouse mammary tumours acquired more innervation than did less-metastatic tumours. This enhanced innervation was driven by expression of the axon-guidance molecule SLIT2 in tumour vasculature. Breast cancer cells induced spontaneous calcium activity in sensory neurons and elicited release of the neuropeptide substance P (SP). Using three-dimensional co-cultures and in vivo models, we found that neuronal SP promoted breast tumour growth, invasion and metastasis. Moreover, patient tumours with elevated SP exhibited enhanced lymph node metastatic spread. SP acted on tumoral tachykinin receptors (TACR1) to drive death of a small population of TACR1high cancer cells. Single-stranded RNAs (ssRNAs) released from dying cells acted on neighbouring tumoural Toll-like receptor 7 (TLR7) to non-canonically activate a prometastatic gene expression program. This SP- and ssRNA-induced Tlr7 gene expression signature was associated with reduced breast cancer survival outcomes. Therapeutic targeting of this neuro-cancer axis with the TACR1 antagonist aprepitant, an approved anti-nausea drug, suppressed breast cancer growth and metastasis in multiple models. Our findings reveal that tumour-induced hyperactivation of sensory neurons regulates multiple aspects of metastatic progression in breast cancer through a therapeutically targetable neuropeptide/extracellular ssRNA sensing axis.


Subject(s)
Breast Neoplasms , Neoplasm Metastasis , RNA , Sensory Receptor Cells , Substance P , Toll-Like Receptor 7 , Animals , Female , Humans , Mice , Breast Neoplasms/blood supply , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Death/drug effects , Cell Line, Tumor , Disease Progression , Gene Expression Regulation, Neoplastic , Intercellular Signaling Peptides and Proteins/metabolism , Lymphatic Metastasis , Neoplasm Invasiveness , Nerve Tissue Proteins/metabolism , RNA/metabolism , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/drug effects , Substance P/metabolism , Survival Analysis , Toll-Like Receptor 7/metabolism , Receptors, Neurokinin-1/metabolism
3.
Brain Res ; 1842: 149107, 2024 Nov 01.
Article in English | MEDLINE | ID: mdl-38977236

ABSTRACT

Fever elicited by bacterial lypopolyssacharide (LPS) is mediated by pro-inflammatory cytokines, which activate central mediators and regulate the hypothalamic temperature setpoint. This response is often accompanied by morphological changes involving the extracellular matrix, neurons and glial cells, with significant health impacts. The NK1 receptor is involved in the febrile response induced by LPS but its effects over the extracellular matrix in the context of neuroinflammation remain unknown. The present work aims to clarify the extracellular changes associated with NK1 signaling in LPS-induced fever. Male Wistar rats were exposed to LPS intraperitoneally. Experimental groups were pre-treated intracerebroventricularly with the NK1 selective inhibitor SR140333B or saline. Histological changes involving the brain extracellular matrix were evaluated using hematoxylin and eosin, Mason's trichrome, picrosirius, alcian blue, periodic acid Schiff's stains. The expression of matrix metalloproteinase 9 (MMP9) was studied using confocal microscopy. Fever was accompanied by edema, perivascular lymphoplamacytic and neutrophylic infiltration, spongiosis and MMP9 overexpression. SR140333B significantly reduced LPS-induced fever (p < 0.0001), MMP9 overexpression (p < 0.01) and associated histological changes. These results contribute to characterize cerebral extracellular matrix changes associated with LPS-induced fever. Overall, the present work supports a role for NK1 receptor in these neuroinflammatory changes, involving MMP9 overexpression, edema and leukocytic infiltration.


Subject(s)
Fever , Lipopolysaccharides , Rats, Wistar , Receptors, Neurokinin-1 , Animals , Male , Fever/chemically induced , Fever/metabolism , Lipopolysaccharides/pharmacology , Rats , Receptors, Neurokinin-1/metabolism , Matrix Metalloproteinase 9/metabolism , Brain/metabolism , Brain/pathology , Brain/drug effects , Extracellular Matrix/metabolism , Extracellular Matrix/drug effects , Neurons/metabolism , Neurons/pathology , Neurons/drug effects , Neurokinin-1 Receptor Antagonists/pharmacology
4.
Physiol Rep ; 12(12): e16125, 2024 Jun.
Article in English | MEDLINE | ID: mdl-39031618

ABSTRACT

Stimulation of the calcium-sensing receptor (CaSR) regulates vascular contractility, but cellular mechanisms involved remain unclear. This study investigated the role of perivascular sensory nerves in CaSR-induced relaxations of male rat mesenteric arteries. In fluorescence studies, colocalisation between synaptophysin, a synaptic vesicle marker, and the CaSR was present in the adventitial layer of arterial segments. Using wire myography, increasing external Ca2+ concentration ([Ca2+]o) from 1 to 10 mM induced vasorelaxations, previously shown to involve the CaSR, which were inhibited by pretreatment with capsaicin. [Ca2+]o-induced vasorelaxations were partially reduced by the calcitonin gene-related peptide (CGRP) receptor blockers, CGRP 8-37 and BIBN 4096, and the neurokinin 1 (NK1) receptor blocker L733,060. The inhibitory effect of CGRP 8-37 required a functional endothelium whereas the inhibitory action of L733,060 did not. Complete inhibition of [Ca2+]o-induced vasorelaxations occurred when CGRP 8-37 and L733,060 were applied together. [Ca2+]o-induced vasorelaxations in the presence of capsaicin were abolished by the ATP-dependent K+ channel (KATP) blocker PNU 37883, but unaffected by the endothelium nitric oxide synthase (eNOS) inhibitor L-NAME. We suggest that the CaSR on perivascular sensory nerves mediate relaxations in rat mesenteric arteries via endothelium-dependent and -independent mechanisms involving CGRP and NK1 receptor-activated NO production and KATP channels, respectively.


Subject(s)
Calcitonin Gene-Related Peptide , Mesenteric Arteries , Receptors, Calcium-Sensing , Receptors, Neurokinin-1 , Vasodilation , Animals , Male , Receptors, Calcium-Sensing/metabolism , Calcitonin Gene-Related Peptide/metabolism , Calcitonin Gene-Related Peptide/pharmacology , Receptors, Neurokinin-1/metabolism , Mesenteric Arteries/drug effects , Mesenteric Arteries/physiology , Mesenteric Arteries/metabolism , Rats , Vasodilation/drug effects , Vasodilation/physiology , Rats, Wistar , Neurokinin-1 Receptor Antagonists/pharmacology , Calcium/metabolism , Capsaicin/pharmacology , Calcitonin Gene-Related Peptide Receptor Antagonists/pharmacology , Signal Transduction/physiology
5.
J Ocul Pharmacol Ther ; 40(7): 445-451, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38829162

ABSTRACT

Purpose: To evaluate the therapeutic efficacy of topical application of a neurokinin-1 receptor (NK1R) antagonist in a rabbit model of nonallergic ocular redness. Methods: Nonallergic ocular redness was induced in rabbits by a single, topical application of dapiparzole hydrochloride eye drops (0.5%, 1%, 2%, or 5%). The NK1R antagonist L-703,606 was topically applied to the eye at the same time of induction or 20 min after induction, and phosphate buffered saline (PBS) treatment served as the control. Superior bulbar conjunctival images were taken every 30 s for the first 2 min, followed by every 4 min for 8 min, and then every 10 min until 1 h. The severity of ocular redness was evaluated on the images using ImageJ-based ocular redness index (ORI) calculations. Results: The ORI scores were significantly increased after the application of 0.5%, 1%, 2%, or 5% dapiparzole at each time point evaluated, with the most severe redness induced by the 5% dapiprazole that led to a maximal mean increase in ORI score of 14 at 20 min post-induction and thus used for subsequent evaluation of therapeutic efficacy of NK1R antagonism. Topical L-703,606, when applied at the same time as dapiprazole induction, significantly suppressed the increase of ORI scores at all time points (∼40% decrease). Furthermore, when applied at 20 min after dapiprazole induction, L-703,606 rapidly and effectively suppressed the increase of ORI scores at 30, 40, 50, and 60 min (∼30% decrease). Conclusions: Topical blockade of NK1R effectively prevents and alleviates nonallergic ocular redness in a novel animal model.


Subject(s)
Disease Models, Animal , Neurokinin-1 Receptor Antagonists , Animals , Rabbits , Neurokinin-1 Receptor Antagonists/pharmacology , Neurokinin-1 Receptor Antagonists/administration & dosage , Ophthalmic Solutions/administration & dosage , Male , Receptors, Neurokinin-1/metabolism , Dose-Response Relationship, Drug , Conjunctiva/drug effects , Conjunctiva/metabolism
6.
Int J Mol Sci ; 25(12)2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38928206

ABSTRACT

Substance P (SP), encoded by the Tac1 gene, has been shown to promote leukocyte infiltration and organ impairment in mice with sepsis. Neurokinin-1 receptor (NK1R) is the major receptor that mediates the detrimental impact of SP on sepsis. This investigation studied whether SP affects the expression of adhesion molecules, including intercellular cell adhesion molecule-1 (ICAM1) and vascular cell adhesion molecule-1 (VCAM1) on vascular endothelial cells in the liver and lungs, contributing to leukocyte infiltration in these tissues of mice with sepsis. Sepsis was induced by caecal ligation and puncture (CLP) surgery in mice. The actions of SP were inhibited by deleting the Tac1 gene, blocking NK1R, or combining these two methods. The activity of myeloperoxidase and the concentrations of ICAM1 and VCAM1 in the liver and lungs, as well as the expression of ICAM1 and VCAM1 on vascular endothelial cells in these tissues, were measured. The activity of myeloperoxidase and the concentration of ICAM1 and VCAM1 in the liver and lungs, as well as the expression of ICAM1 and VCAM1 on vascular endothelial cells in these tissues, increased in mice with CLP surgery-induced sepsis. Suppressing the biosynthesis of SP and its interactions with NK1R attenuated CLP surgery-induced alterations in the liver and lungs of mice. Our findings indicate that SP upregulates the expression of ICAM1 and VCAM1 on vascular endothelial cells in the liver and lungs, thereby increasing leukocyte infiltration in these tissues of mice with CLP surgery-induced sepsis by activating NK1R.


Subject(s)
Endothelial Cells , Intercellular Adhesion Molecule-1 , Liver , Lung , Receptors, Neurokinin-1 , Sepsis , Substance P , Vascular Cell Adhesion Molecule-1 , Animals , Sepsis/metabolism , Sepsis/pathology , Mice , Substance P/metabolism , Lung/metabolism , Lung/pathology , Liver/metabolism , Liver/pathology , Intercellular Adhesion Molecule-1/metabolism , Intercellular Adhesion Molecule-1/genetics , Endothelial Cells/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Receptors, Neurokinin-1/metabolism , Receptors, Neurokinin-1/genetics , Male , Leukocytes/metabolism , Mice, Inbred C57BL , Peroxidase/metabolism , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules/genetics , Disease Models, Animal
7.
Am J Physiol Cell Physiol ; 327(1): C151-C167, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38798270

ABSTRACT

Substance P (SP) is released from sensory nerves in the arteries and heart. It activates neurokinin-1 receptors (NK1Rs) causing vasodilation, immune modulation, and adverse cardiac remodeling. The hypothesis was tested: SP and SP metabolites activate different second messenger signaling pathways. Macrophages, endothelial cells, and fibroblasts metabolized SP to N- and C-terminal metabolites to varying extents. SP 5-11 was the most abundant metabolite followed by SP 1-4, SP 7-11, SP 6-11, SP 3-11, and SP 8-11. In NK1R-expressing human embryonic kidney 293 (HEK293) cells, SP and some C-terminal SP metabolites stimulate the NK1R, promoting the dissociation of several Gα proteins, including Gαs and Gαq from their ßγ subunits. SP increases intracellular calcium concentrations ([Ca]i) and cyclic 3',5'-adenosine monophosphate (cAMP) accumulation with similar -log EC50 values of 8.5 ± 0.3 and 7.8 ± 0.1 M, respectively. N-terminal metabolism of SP by up to five amino acids and C-terminal deamidation of SP produce peptides that retain activity to increase [Ca]i but not to increase cAMP. C-terminal metabolism results in the loss of both activities. Thus, [Ca]i and cAMP signaling are differentially affected by SP metabolism. To assess the role of N-terminal metabolism, SP and SP 6-11 were compared with cAMP-mediated activities in NK1R-expressing 3T3 fibroblasts. SP inhibits nuclear factor κB (NF-κB) activity, cell proliferation, and wound healing and stimulates collagen production. SP 6-11 had little or no activity. Cyclooxygenase-2 (COX-2) expression is increased by SP but not by SP 6-11. Thus, metabolism may select the cellular response to SP by inhibiting or redirecting the second messenger signaling pathway activated by the NK1R.NEW & NOTEWORTHY Endothelial cells, macrophages, and fibroblasts metabolize substance P (SP) to N- and C-terminal metabolites with SP 5-11 as the most abundant metabolite. SP activates neurokinin-1 receptors to increase intracellular calcium and cyclic AMP. In contrast, SP metabolites of N-terminal metabolism and C-terminal deamidation retain the ability to increase calcium but lose the ability to increase cyclic AMP. These new insights indicate that the metabolism of SP directs cellular functions by regulating specific signaling pathways.


Subject(s)
Cyclic AMP , Receptors, Neurokinin-1 , Signal Transduction , Substance P , Substance P/metabolism , Receptors, Neurokinin-1/metabolism , Receptors, Neurokinin-1/agonists , Humans , Cyclic AMP/metabolism , Animals , HEK293 Cells , Mice , Fibroblasts/metabolism , Fibroblasts/drug effects , Calcium/metabolism
8.
Int J Biol Sci ; 20(7): 2507-2531, 2024.
Article in English | MEDLINE | ID: mdl-38725846

ABSTRACT

Neuropeptide substance P (SP) belongs to a family of bioactive peptides and regulates many human diseases. This study aims to investigate the role and underlying mechanisms of SP in colitis. Here, activated SP-positive neurons and increased SP expression were observed in dextran sodium sulfate (DSS)-induced colitis lesions in mice. Administration of exogenous SP efficiently ameliorated the clinical symptoms, impaired intestinal barrier function, and inflammatory response. Mechanistically, SP protected mitochondria from damage caused by DSS or TNF-α exposure, preventing mitochondrial DNA (mtDNA) leakage into the cytoplasm, thereby inhibiting the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway. SP can also directly prevent STING phosphorylation through the neurokinin-1 receptor (NK1R), thereby inhibiting the activation of the TBK1-IRF3 signaling pathway. Further studies revealed that SP alleviated the DSS or TNF-α-induced ferroptosis process, which was associated with repressing the cGAS-STING signaling pathway. Notably, we identified that the NK1R inhibition reversed the effects of SP on inflammation and ferroptosis via the cGAS-STING pathway. Collectively, we unveil that SP attenuates inflammation and ferroptosis via suppressing the mtDNA-cGAS-STING or directly acting on the STING pathway, contributing to improving colitis in an NK1R-dependent manner. These findings provide a novel mechanism of SP regulating ulcerative colitis (UC) disease.


Subject(s)
Colitis , Ferroptosis , Inflammation , Signal Transduction , Substance P , Animals , Male , Mice , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Dextran Sulfate , DNA, Mitochondrial/metabolism , Ferroptosis/drug effects , Inflammation/metabolism , Membrane Proteins/metabolism , Mice, Inbred C57BL , Nucleotidyltransferases/metabolism , Receptors, Neurokinin-1/metabolism , Signal Transduction/drug effects , Substance P/metabolism , Substance P/pharmacology , Tumor Necrosis Factor-alpha/metabolism
9.
J Neurophysiol ; 132(1): 108-129, 2024 07 01.
Article in English | MEDLINE | ID: mdl-38748514

ABSTRACT

µ-Opioid receptors (MORs) are responsible for mediating both the analgesic and respiratory effects of opioid drugs. By binding to MORs in brainstem regions involved in controlling breathing, opioids produce respiratory depressive effects characterized by slow and shallow breathing, with potential cardiorespiratory arrest and death during overdose. To better understand the mechanisms underlying opioid-induced respiratory depression, thorough knowledge of the regions and cellular subpopulations that may be vulnerable to modulation by opioid drugs is needed. Using in situ hybridization, we determined the distribution and coexpression of Oprm1 (gene encoding MORs) mRNA with glutamatergic (Vglut2) and neurokinin-1 receptor (Tacr1) mRNA in medullary and pontine regions involved in breathing control and modulation. We found that >50% of cells expressed Oprm1 mRNA in the preBötzinger complex (preBötC), nucleus tractus solitarius (NTS), nucleus ambiguus (NA), postinspiratory complex (PiCo), locus coeruleus (LC), Kölliker-Fuse nucleus (KF), and the lateral and medial parabrachial nuclei (LBPN and MPBN, respectively). Among Tacr1 mRNA-expressing cells, >50% coexpressed Oprm1 mRNA in the preBötC, NTS, NA, Bötzinger complex (BötC), PiCo, LC, raphe magnus nucleus, KF, LPBN, and MPBN, whereas among Vglut2 mRNA-expressing cells, >50% coexpressed Oprm1 mRNA in the preBötC, NTS, NA, BötC, PiCo, LC, KF, LPBN, and MPBN. Taken together, our study provides a comprehensive map of the distribution and coexpression of Oprm1, Tacr1, and Vglut2 mRNA in brainstem regions that control and modulate breathing and identifies Tacr1 and Vglut2 mRNA-expressing cells as subpopulations with potential vulnerability to modulation by opioid drugs.NEW & NOTEWORTHY Opioid drugs can cause serious respiratory side-effects by binding to µ-opioid receptors (MORs) in brainstem regions that control breathing. To better understand the regions and their cellular subpopulations that may be vulnerable to modulation by opioids, we provide a comprehensive map of Oprm1 (gene encoding MORs) mRNA expression throughout brainstem regions that control and modulate breathing. Notably, we identify glutamatergic and neurokinin-1 receptor-expressing cells as potentially vulnerable to modulation by opioid drugs and worthy of further investigation using targeted approaches.


Subject(s)
Receptors, Neurokinin-1 , Receptors, Opioid, mu , Vesicular Glutamate Transport Protein 2 , Animals , Receptors, Opioid, mu/metabolism , Receptors, Opioid, mu/genetics , Receptors, Neurokinin-1/metabolism , Receptors, Neurokinin-1/genetics , Mice , Vesicular Glutamate Transport Protein 2/metabolism , Vesicular Glutamate Transport Protein 2/genetics , Male , Brain Stem/metabolism , Brain Stem/drug effects , Mice, Inbred C57BL , RNA, Messenger/metabolism , RNA, Messenger/genetics , Respiratory Center/metabolism , Respiratory Center/drug effects
10.
Neuropsychopharmacology ; 49(11): 1689-1699, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38649427

ABSTRACT

Behavioral and clinical studies have revealed a critical role of substance P (SP) in aggression; however, the neural circuit mechanisms underlying SP and aggression remain elusive. Here, we show that tachykinin-expressing neurons in the medial amygdala (MeATac1 neurons) are activated during aggressive behaviors in male mice. We identified MeATac1 neurons as a key mediator of aggression and found that MeATac1→ventrolateral part of the ventromedial hypothalamic nucleus (VMHvl) projections are critical to the regulation of aggression. Moreover, SP/neurokinin-1 receptor (NK-1R) signaling in the VMHvl modulates aggressive behaviors in male mice. SP/NK-1R signaling regulates aggression by influencing glutamate transmission in neurons in the VMHvl. In summary, these findings place SP as a key node in aggression circuits.


Subject(s)
Aggression , Corticomedial Nuclear Complex , Substance P , Animals , Male , Mice , Aggression/physiology , Corticomedial Nuclear Complex/physiology , Corticomedial Nuclear Complex/metabolism , Corticomedial Nuclear Complex/drug effects , Glutamic Acid/metabolism , Mice, Inbred C57BL , Neural Pathways/physiology , Neurons/physiology , Neurons/metabolism , Receptors, Neurokinin-1/metabolism , Substance P/metabolism , Tachykinins/metabolism , Ventromedial Hypothalamic Nucleus/physiology , Ventromedial Hypothalamic Nucleus/metabolism , Ventromedial Hypothalamic Nucleus/drug effects
11.
Cancer Chemother Pharmacol ; 94(2): 237-250, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38678150

ABSTRACT

PURPOSE: The current candidate gene association study aims to investigate tag SNPs from the TACR1 gene as pharmacogenetic predictors of response to the antiemetic guidelines-recommended, NK-1 receptor antagonist-based, triple antiemetic regimens. METHODS: A set of eighteen tag SNPs of TACR1 were genotyped in breast cancer patients receiving anthracycline and cyclophosphamide (with/without docetaxel) applying real-time PCR-HRMA. Data analysis for 121 ultimately enrolled patients was initiated by defining haplotype blocks using PHASE v.2.1. The association of each tag SNP and haplotype alleles with failure to achieve the defined antiemetic regimen efficacy endpoints was tested using PLINK (v.1.9 and v.1.07, respectively) based on the logistic regression, adjusting for the previously known chemotherapy-induced nausea and vomiting (CINV) prognostic factors. All reported p-values were corrected using the permutation test (n = 100,000). RESULTS: Four variants of rs881, rs17010730, rs727156, and rs3755462, as well as haplotypes containing the mentioned variants, were significantly associated with failure to achieve at least one of the defined efficacy endpoints. Variant annotation via in-silico studies revealed that the non-seed sequence variant, rs881, is located in the miRNA (hsa-miR-613) binding site. The other three variants or a variant in complete linkage disequilibrium with them overlap a region of high H3K9ac-promoter-like signature or regions of high enhancer-like signature in the brain or gastrointestinal tissue. CONCLUSION: Playing an essential role in regulating TACR1 expression, gene polymorphisms of TACR1 serve as the potential pharmacogenetic predictors of response to the NK-1 receptor antagonist-based, triple antiemetic regimens. If clinically approved, modifying the NK-1 receptor antagonist dose leads to better management of CINV in risk-allele carriers.


Subject(s)
Antiemetics , Antineoplastic Combined Chemotherapy Protocols , Breast Neoplasms , Cyclophosphamide , Nausea , Polymorphism, Single Nucleotide , Receptors, Neurokinin-1 , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Middle Aged , Antiemetics/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Receptors, Neurokinin-1/genetics , Nausea/chemically induced , Nausea/genetics , Cyclophosphamide/adverse effects , Cyclophosphamide/therapeutic use , Cyclophosphamide/administration & dosage , Vomiting/chemically induced , Vomiting/genetics , Neurokinin-1 Receptor Antagonists/therapeutic use , Adult , Genetic Association Studies , Haplotypes , Aged , Docetaxel/therapeutic use , Docetaxel/adverse effects , Pharmacogenetics , Anthracyclines/adverse effects , Anthracyclines/therapeutic use , Genotype
12.
Eur J Pharmacol ; 973: 176587, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38642667

ABSTRACT

Agonist-induced phosphorylation is a crucial step in the activation/deactivation cycle of G protein-coupled receptors (GPCRs), but direct determination of individual phosphorylation events has remained a major challenge. We have recently developed a bead-based immunoassay for the quantitative assessment of agonist-induced GPCR phosphorylation that can be performed entirely in 96-well plates, thus eliminating the need for western blot analysis. In the present study, we adapted this assay to three novel phosphosite-specific antibodies directed against the neurokinin 1 (NK1) receptor, namely pS338/pT339-NK1, pT344/pS347-NK1, and pT356/pT357-NK1. We found that substance P (SP) stimulated concentration-dependent phosphorylation of all three sites, which could be completely blocked in the presence of the NK1 receptor antagonist aprepitant. The other two endogenous ligands of the tachykinin family, neurokinin A (NKA) and neurokinin B (NKB), were also able to induce NK1 receptor phosphorylation, but to a much lesser extent than substance P. Interestingly, substance P promoted phosphorylation of the two distal sites more efficiently than that of the proximal site. The proximal site was identified as a substrate for phosphorylation by protein kinase C. Analysis of GPCR kinase (GRK)-knockout cells revealed that phosphorylation was mediated by all four GRK isoforms to similar extents at the T344/S347 and the T356/T357 cluster. Knockout of all GRKs resulted in abolition of all phosphorylation signals highlighting the importance of these kinases in agonist-mediated receptor phosphorylation. Thus, the 7TM phosphorylation assay technology allows for rapid and detailed analyses of GPCR phosphorylation.


Subject(s)
Receptors, Neurokinin-1 , Substance P , Receptors, Neurokinin-1/metabolism , Receptors, Neurokinin-1/agonists , Phosphorylation/drug effects , Humans , Substance P/pharmacology , Animals , Immunoassay/methods , Cricetulus , CHO Cells , Mice , Neurokinin-1 Receptor Antagonists/pharmacology , Neurokinin A/pharmacology , Neurokinin A/metabolism
13.
Discov Med ; 36(183): 714-720, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38665020

ABSTRACT

BACKGROUND: Spinal cord injury (SCI) is usually caused by external direct or indirect factors, and with a high morbidity and mortality rate. The aim of this study was to observe the effects of Dexmedetomidine (DEX) combined with Esketamine (ESK) on pain behavior and potential analgesic mechanisms in rats with SCI. The goal was to provide a reliable multimodal analgesic medication regimen for SCI. METHODS: Thirty rats were divided into five groups with six rats in each group: Sham group, SCI group, DEX group, ESK group, and DEX+ESK group. The SCI model in rats was constructed, and the motor function of hind limbs of rats was measured using Basso Beattie Bresnahan (BBB) locomotor rating scale and inclined plate test. The levels of interleukin 18 (IL-18), interleukin 1ß (IL-1ß), and tumor necrosis factor-α (TNF-α) in the spinal cord were determined by enzyme-linked immunosorbent assay (ELISA). The expressions of substance P (SP), neurokinin-1 receptor (NK-1R), B cell lymphoma-2 (Bcl-2), and Bcl2-associated X protein (Bax) in the rats' spinal cord were measured by Western blot assay. The viability of spinal astrocytes was evaluated by 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. RESULTS: After 7 days, the BBB scores were significantly higher in the DEX, ESK, and DEX+ESK groups compared to the SCI group (p < 0.01). Additionally, the DEX+ESK group had significantly higher scores than both the DEX and ESK groups (p < 0.01). The maximum angle of the DEX (p < 0.05), ESK (p < 0.05), and DEX+ESK groups (p < 0.01) were higher than the SCI group, and the maximum angle of DEX+ESK group was higher than DEX and ESK groups (p < 0.05). The levels of IL-18, IL-1ß, and TNF-α in the DEX, ESK, and DEX+ESK groups were lower than the SCI group (p < 0.01), while the DEX+ESK group had significantly lower IL-18, IL-1ß, and TNF-α levels than the DEX and ESK groups (p < 0.01). The levels of SP (p < 0.01) and NK-1R (p < 0.05) were lower in the DEX, ESK, and DEX+ESK groups compared to the SCI group, and the levels of SP and NK-1R were lower in the DEX+ESK group compared to the DEX and ESK groups (p < 0.01). The DEX and ESK groups suppressed the activity of spinal astrocytes (p < 0.01), however, the DEX+ESK group had larger effects on spinal astrocytes than the ESK group (p < 0.05). CONCLUSIONS: Treatment using DEX combined with ESK improves the motor function, inhibits inflammation and astrocyte activity, and exerts analgesic effects on rats with SCI. These findings can serve as a reference for the selection of multi-modal analgesics.


Subject(s)
Dexmedetomidine , Ketamine , Rats, Sprague-Dawley , Spinal Cord Injuries , Animals , Dexmedetomidine/pharmacology , Dexmedetomidine/therapeutic use , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/pathology , Spinal Cord Injuries/metabolism , Rats , Ketamine/pharmacology , Ketamine/therapeutic use , Male , Analgesics/pharmacology , Analgesics/therapeutic use , Spinal Cord/drug effects , Spinal Cord/pathology , Spinal Cord/metabolism , Substance P/metabolism , Disease Models, Animal , Tumor Necrosis Factor-alpha/metabolism , Receptors, Neurokinin-1/metabolism , Interleukin-1beta/metabolism
14.
Toxins (Basel) ; 16(4)2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38668612

ABSTRACT

Accidents caused by Bothrops jararaca (Bj) snakes result in several local and systemic manifestations, with pain being a fundamental characteristic. The inflammatory process responsible for hyperalgesia induced by Bj venom (Bjv) has been studied; however, the specific roles played by the peripheral and central nervous systems in this phenomenon remain unclear. To clarify this, we induced hyperalgesia in rats using Bjv and collected tissues from dorsal root ganglia (DRGs) and spinal cord (SC) at 2 and 4 h post-induction. Samples were labeled for Iba-1 (macrophage and microglia), GFAP (satellite cells and astrocytes), EGR1 (neurons), and NK1 receptors. Additionally, we investigated the impact of minocycline, an inhibitor of microglia, and GR82334 antagonist on Bjv-induced hyperalgesia. Our findings reveal an increase in Iba1 in DRG at 2 h and EGR1 at 4 h. In the SC, markers for microglia, astrocytes, neurons, and NK1 receptors exhibited increased expression after 2 h, with EGR1 continuing to rise at 4 h. Minocycline and GR82334 inhibited venom-induced hyperalgesia, highlighting the crucial roles of microglia and NK1 receptors in this phenomenon. Our results suggest that the hyperalgesic effects of Bjv involve the participation of microglial and astrocytic cells, in addition to the activation of NK1 receptors.


Subject(s)
Bothrops , Crotalid Venoms , Ganglia, Spinal , Hyperalgesia , Receptors, Neurokinin-1 , Animals , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Crotalid Venoms/toxicity , Male , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Receptors, Neurokinin-1/metabolism , Minocycline/pharmacology , Spinal Cord/drug effects , Spinal Cord/metabolism , Early Growth Response Protein 1/metabolism , Early Growth Response Protein 1/genetics , Microglia/drug effects , Microglia/metabolism , Neuroglia/drug effects , Neuroglia/metabolism , Rats , Glial Fibrillary Acidic Protein/metabolism , Calcium-Binding Proteins/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Microfilament Proteins/metabolism , Neurokinin-1 Receptor Antagonists/pharmacology , Rats, Sprague-Dawley
15.
Liver Int ; 44(7): 1651-1667, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38554043

ABSTRACT

BACKGROUND: Intrahepatic cholangiocarcinoma (iCCA) has a poor prognosis and limited treatment options. Aprepitant, a selective NK-1R antagonist, can inhibit the growth of various tumours in vitro and in vivo. However, it remains unclear whether aprepitant has cytotoxic effects on iCCA. METHODS: We measured the expression of SP/NK-1R in clinical samples of iCCA by immunohistochemistry. Then, we detected the cytotoxic effects of aprepitant on iCCA cells via MTT, EdU and colony formation assay. We constructed a subcutaneous xenograft model of BALB/c nude mice by using HCCC-9810 and RBE cell lines to explore the effects of aprepitant in vivo. To elucidate the potential mechanisms, we explored the pro-apoptotic effect of aprepitant by flow cytometric, western blotting, ROS detection and JC-1 staining. Furthermore, we detected the autophagic level of HCCC-9810 and RBE by western blotting, mRFP-eGFP-LC3 adenovirus transfection and electron microscope. RESULTS: SP/NK-1R is significantly expressed in iCCA. Aprepitant inhibited human iCCA xenograft growth and dose-dependently decreased the viability of RBE and HCCC-9810 cells. Aprepitant-induced mitochondria-dependent apoptosis through ROS/JNK pathway. Additionally, pretreatment with z-VAD-fmk partly reversed the effect of aprepitant on cell viability, while NAC completely attenuated the cytotoxic effects of aprepitant in vitro. Furthermore, we observed the dynamic changes of autophagosome in RBE and HCCC-9810 cells treated with aprepitant. CONCLUSION: SP/NK-1R signalling is significantly activated in iCCA and promotes the proliferation of iCCA cells. By contrast, aprepitant can induce autophagy and apoptosis in iCCA cells via ROS accumulation and subsequent activation of JNK.


Subject(s)
Apoptosis , Aprepitant , Autophagy , Bile Duct Neoplasms , Cholangiocarcinoma , Mice, Inbred BALB C , Mice, Nude , Neurokinin-1 Receptor Antagonists , Reactive Oxygen Species , Xenograft Model Antitumor Assays , Aprepitant/pharmacology , Aprepitant/therapeutic use , Animals , Humans , Autophagy/drug effects , Apoptosis/drug effects , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/pathology , Cholangiocarcinoma/metabolism , Reactive Oxygen Species/metabolism , Cell Line, Tumor , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/pathology , Bile Duct Neoplasms/metabolism , Neurokinin-1 Receptor Antagonists/pharmacology , Mice , Male , Female , Receptors, Neurokinin-1/metabolism , Middle Aged , Cell Proliferation/drug effects
16.
Int J Mol Sci ; 25(6)2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38542509

ABSTRACT

Traumatic brain injuries represent a leading cause of death and disability in the paediatric and adult populations. Moderate-to-severe injuries are associated with blood-brain barrier dysfunction, the development of cerebral oedema, and neuroinflammation. Antagonists of the tachykinin NK1 receptor have been proposed as potential agents for the post-injury treatment of TBI. We report on the identification of EUC-001 as a potential clinical candidate for development as a novel TBI therapy. EUC-001 is a selective NK1 antagonist with a high affinity for the human NK1 receptor (Ki 5.75 × 10-10 M). It has sufficient aqueous solubility to enable intravenous administration, whilst still retaining good CNS penetration as evidenced by its ability to inhibit the gerbil foot-tapping response. Using an animal model of TBI, the post-injury administration of EUC-001 was shown to restore BBB function in a dose-dependent manner. EUC-001 was also able to ameliorate cerebral oedema. These effects were associated with a significant reduction in post-TBI mortality. In addition, EUC-001 was able to significantly reduce functional deficits, both motor and cognitive, that normally follow a severe injury. EUC-001 is proposed as an ideal candidate for clinical development for TBI.


Subject(s)
Brain Edema , Brain Injuries, Traumatic , Animals , Humans , Child , Receptors, Neurokinin-1 , Substance P , Neurokinin-1 Receptor Antagonists/pharmacology , Neurokinin-1 Receptor Antagonists/therapeutic use , Brain Injuries, Traumatic/drug therapy , Infusions, Intravenous
17.
Am J Physiol Cell Physiol ; 326(5): C1482-C1493, 2024 05 01.
Article in English | MEDLINE | ID: mdl-38525537

ABSTRACT

Corneal fibroblasts maintain homeostasis of the corneal stroma by mediating the synthesis and degradation of extracellular collagen, and these actions are promoted by transforming growth factor-ß (TGF-ß) and interleukin-1ß (IL-1ß), respectively. The cornea is densely innervated with sensory nerve fibers that are not only responsible for sensation but also required for physiological processes such as tear secretion and wound healing. Loss or dysfunction of corneal nerves thus impairs corneal epithelial wound healing and can lead to neurotrophic keratopathy. The sensory neurotransmitter substance P (SP) promotes corneal epithelial wound healing by enhancing the stimulatory effects of growth factors and fibronectin. We have now investigated the role of SP in collagen metabolism mediated by human corneal fibroblasts in culture. Although SP alone had no effect on collagen synthesis or degradation by these cells, it promoted the stimulatory effect of TGF-ß on collagen type I synthesis without affecting that of IL-1ß on the expression of matrix metalloproteinase-1. This effect of SP on TGF-ß-induced collagen synthesis was accompanied by activation of p38 mitogen-activated protein kinase (MAPK) signaling and was attenuated by pharmacological inhibition of p38 or of the neurokinin-1 receptor. Our results thus implicate SP as a modulator of TGF-ß-induced collagen type I synthesis by human corneal fibroblasts, and they suggest that loss of this function may contribute to the development of neurotrophic keratopathy.NEW & NOTEWORTHY This study investigates the role of substance P (SP) in collagen metabolism mediated by human corneal fibroblasts in culture. We found that, although SP alone had no effect on collagen synthesis or degradation by corneal fibroblasts, it promoted the stimulatory effect of transforming growth factor-ß on collagen type I synthesis without affecting that of interleukin-1ß on the expression of matrix metalloproteinase-1.


Subject(s)
Fibroblasts , Interleukin-1beta , Substance P , Transforming Growth Factor beta , p38 Mitogen-Activated Protein Kinases , Humans , Substance P/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Transforming Growth Factor beta/metabolism , Fibroblasts/metabolism , Fibroblasts/drug effects , Cells, Cultured , Interleukin-1beta/metabolism , Collagen Type I/metabolism , Collagen Type I/biosynthesis , Receptors, Neurokinin-1/metabolism , Cornea/metabolism , Cornea/drug effects , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 1/genetics , Collagen/metabolism , Collagen/biosynthesis , Signal Transduction/drug effects , Corneal Stroma/metabolism , Corneal Stroma/drug effects , Corneal Keratocytes/metabolism , Corneal Keratocytes/drug effects
18.
Naunyn Schmiedebergs Arch Pharmacol ; 397(8): 5875-5882, 2024 08.
Article in English | MEDLINE | ID: mdl-38334824

ABSTRACT

Substance P (SP), an important neuropeptide, has a crucial role in the progression of several cancers, including prostate cancer, through interacting with the neurokinin-1 receptor (NK1R). Oxidative stress is also involved in the onset and progression of prostate cancer. However, no studies have been performed on the cross-talk between the SP/NK1R system and cellular redox balance in prostate cancer, and how it is involved in tumorogenesis. We aimed to investigate the effect of the SP/NK1R system and the blockage of NK1R with its specific antagonist (aprepitant) on the cellular redox status of the prostate cancer cell line (PC3 and LNCaP). We performed the resazurin assay to evaluate the toxicity of the aprepitant on the PC3 and LNCaP cell lines. The intracellular reactive oxygen species (ROS) level was measured after SP and aprepitant treatment. The alterations of expression and activity of two crucial cellular oxidoreductases, glutaredoxin, and thioredoxin were evaluated by qRT-PCR and commercial kits (ZellBio GmbH), respectively. Our results revealed that SP increased ROS production and decreased the expression and activity of glutaredoxin and thioredoxin. On the other hand, treatment of cells with aprepitant showed reverse results. In conclusion, we found that the SP/NK1R system could promote prostate cancer progression by inducing oxidative stress. In addition, the inhibition of NK1R by aprepitant modulated the effect of the SP/NK1R system on the cellular redox system. Aprepitant might therefore be introduced as a candidate for the treatment of prostate cancer; however, more studies are required to confirm the validation of this hypothesis.


Subject(s)
Aprepitant , Glutaredoxins , Prostatic Neoplasms , Reactive Oxygen Species , Receptors, Neurokinin-1 , Substance P , Thioredoxins , Humans , Male , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/drug therapy , Thioredoxins/metabolism , Reactive Oxygen Species/metabolism , Aprepitant/pharmacology , Receptors, Neurokinin-1/metabolism , Cell Line, Tumor , Glutaredoxins/metabolism , Glutaredoxins/genetics , Substance P/metabolism , Substance P/pharmacology , Neurokinin-1 Receptor Antagonists/pharmacology , Oxidative Stress/drug effects , PC-3 Cells
19.
J Zhejiang Univ Sci B ; 25(2): 91-105, 2024 Feb 15.
Article in English, Chinese | MEDLINE | ID: mdl-38303494

ABSTRACT

Recently, the substance P (SP)/neurokinin-1 receptor (NK-1R) system has been found to be involved in various human pathophysiological disorders including the symptoms of coronavirus disease 2019 (COVID-19). Besides, studies in the oncological field have demonstrated an intricate correlation between the upregulation of NK-1R and the activation of SP/NK-1R system with the progression of multiple carcinoma types and poor clinical prognosis. These findings indicate that the modulation of SP/NK-1R system with NK-1R antagonists can be a potential broad-spectrum antitumor strategy. This review updates the latest potential and applications of NK-1R antagonists in the treatment of human diseases and cancers, as well as the underlying mechanisms. Furthermore, the strategies to improve the bioavailability and efficacy of NK-1R antagonist drugs are summarized, such as solid dispersion systems, nanonization, and nanoencapsulation. As a radiopharmaceutical therapeutic, the NK-1R antagonist aprepitant was originally developed as radioligand receptor to target NK-1R-overexpressing tumors. However, combining NK-1R antagonists with other drugs can produce a synergistic effect, thereby enhancing the therapeutic effect, alleviating the symptoms, and improving patients quality of life in several diseases and cancers.


Subject(s)
Neoplasms , Neurokinin-1 Receptor Antagonists , Humans , Neurokinin-1 Receptor Antagonists/pharmacology , Neurokinin-1 Receptor Antagonists/therapeutic use , Quality of Life , Substance P , Receptors, Neurokinin-1 , Neoplasms/drug therapy
20.
Addict Biol ; 29(1): e13357, 2024 01.
Article in English | MEDLINE | ID: mdl-38221805

ABSTRACT

There is a high frequency of comorbidity of alcohol use disorder (AUD) and depression in human populations. We have studied this relationship in our lab using the social defeat stress (SDS) model, which results in both depression-like behaviours and increased alcohol consumption in male mice. However, standard SDS procedures are difficult to use in female mice due to a lack of territorial aggression. In the experiments presented here, we used vicarious defeat stress (VDS) to assess social withdrawal and alcohol consumption in female C57BL6/J mice. We also assessed the expression of interleukin-6 (IL6), which is a proinflammatory cytokine that is associated with depression in humans and sensitivity to SDS in mice. In these experiments, C57BL/6 female mice underwent 10 days of VDS where they witnessed the physical defeat of a male conspecific by an aggressive CD1 mouse. After the end of VDS, mice were either given access to alcohol or sacrificed for the measurement of IL6 expression. We found that VDS increased alcohol consumption and IL6 expression in the frontal cortex and hippocampus. Given that the neurokinin-1 receptor (NK1R) can mediate both stress-induced alcohol consumption and IL6 expression, we tested the ability of NK1R antagonism to reduce VDS-induced alcohol consumption and found that this treatment reduced alcohol intake in both VDS-exposed mice and in unstressed controls. The observed increase in alcohol consumption suggests that VDS is a model that can be utilized to study stress-induced alcohol consumption in female mice, and that this is sensitive to NK1R antagonism.


Subject(s)
Alcohol Drinking , Interleukin-6 , Receptors, Neurokinin-1 , Stress, Psychological , Animals , Female , Male , Mice , Alcohol Drinking/metabolism , Interleukin-6/metabolism , Mice, Inbred C57BL , Receptors, Neurokinin-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL