Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 13 de 13
Filtrer
1.
J Neurovirol ; 30(1): 86-99, 2024 02.
Article de Anglais | MEDLINE | ID: mdl-38453879

RÉSUMÉ

Simian varicella virus (SVV) produces peripheral inflammatory responses during varicella (primary infection) and zoster (reactivation) in rhesus macaques (RM). However, it is unclear if peripheral measures are accurate proxies for central nervous system (CNS) responses. Thus, we analyzed cytokine and Aß42/Aß40 changes in paired serum and cerebrospinal fluid (CSF) during the course of infection. During varicella and zoster, every RM had variable changes in serum and CSF cytokine and Aß42/Aß40 levels compared to pre-inoculation levels. Overall, peripheral infection appears to affect CNS cytokine and Aß42/Aß40 levels independent of serum responses, suggesting that peripheral disease may contribute to CNS disease.


Sujet(s)
Peptides bêta-amyloïdes , Cytokines , Macaca mulatta , Animaux , Peptides bêta-amyloïdes/liquide cérébrospinal , Peptides bêta-amyloïdes/sang , Cytokines/liquide cérébrospinal , Cytokines/sang , Activation virale , Fragments peptidiques/liquide cérébrospinal , Fragments peptidiques/sang , Varicellovirus/génétique , Varicellovirus/immunologie , Herpèsvirus humain de type 3/pathogénicité , Herpèsvirus humain de type 3/immunologie , Infections à Herpesviridae/liquide cérébrospinal , Infections à Herpesviridae/virologie , Infections à Herpesviridae/sang , Infections à Herpesviridae/immunologie , Mâle , Zona/liquide cérébrospinal , Zona/virologie , Zona/sang , Zona/immunologie , Maladies des singes/virologie , Maladies des singes/liquide cérébrospinal , Maladies des singes/sang
2.
Res Sq ; 2023 Oct 16.
Article de Anglais | MEDLINE | ID: mdl-37886544

RÉSUMÉ

Simian varicella virus (SVV) produces peripheral inflammatory responses during varicella (primary infection) and zoster (reactivation) in rhesus macaques (RM). However, it is unclear if peripheral measures are accurate proxies for central nervous system (CNS) responses. Thus, we analyzed cytokine and Aß42/Aß40 changes in paired serum and cerebrospinal fluid (CSF) during the course of infection. During varicella and zoster, every RM had variable changes in serum and CSF cytokine and Aß42/Aß40 levels compared to pre-inoculation levels. Overall, peripheral infection appears to affect CNS cytokine and Aß42/Aß40 levels independent of serum responses, suggesting that peripheral disease may contribute to CNS disease.

3.
J Technol Behav Sci ; : 1-8, 2023 Jan 30.
Article de Anglais | MEDLINE | ID: mdl-36742417

RÉSUMÉ

Most suicides occur among adults of working age and workplace suicide prevention is a public health priority. Workplace suicide prevention efforts, however, remain limited. This paper describes and evaluates a workplace-based suicide prevention gatekeeper training delivered in-person and virtually. VitalCog is a 2-h suicide prevention program designed specifically for the workplace and based on best practices for gatekeeper training. It is designed to be practical and interactive, with four modules (why prevention matters, what to do if someone is suicidal, conversations about suicide, and postvention), each containing related video, group discussion, and role play exercise components. It was delivered live by experienced trainers either in-person or using synchronous technologies between 2018 and 2021. A mixed methods pre- and post-training design with no control group was used to evaluate in-person vs. virtual delivery to determine knowledge gain, confidence identifying warning signs, and comfort levels talking about suicide. One thousand two-hundred and forty-four (1244) pre- and post-training responses were analyzed, with no significant (p > .05) socio-economic differences between the pre-training and post-training respondent samples. Both in-person (n = 841) and virtual (n = 403) training groups demonstrated statistically significant increases in knowledge about suicide prevention and seeking help, confidence to identify suicide warning signs, and comfort levels talking to someone about getting help. Interestingly, the virtual group showed higher post-training outcome scores than the in-person group. While COVID-19 significantly reduced in-person training opportunities, these results suggest that offering VitalCog virtually is as effective as in-person, and potentially has advantages over in-person training.

4.
Viruses ; 14(4)2022 03 25.
Article de Anglais | MEDLINE | ID: mdl-35458404

RÉSUMÉ

Virus infection of adrenal glands can disrupt secretion of mineralocorticoids, glucocorticoids, and sex hormones from the cortex and catecholamines from the medulla, leading to a constellation of symptoms such as fatigue, dizziness, weight loss, nausea, and muscle and joint pain. Specifically, varicella zoster virus (VZV) can produce bilateral adrenal hemorrhage and adrenal insufficiency during primary infection or following reactivation. However, the mechanisms by which VZV affects the adrenal glands are not well-characterized. Herein, we determined if primary human adrenal cortical cells (HAdCCs) infected with VZV support viral replication and produce a proinflammatory environment. Quantitative PCR showed VZV DNA increasing over time in HAdCCs, yet no cell death was seen at 3 days post-infection by TUNEL staining or Western Blot analysis with PARP and caspase 9 antibodies. Compared to conditioned supernatant from mock-infected cells, supernatant from VZV-infected cells contained significantly elevated IL-6, IL-8, IL-12p70, IL-13, IL-4, and TNF-α. Overall, VZV can productively infect adrenal cortical cells in the absence of cell death, suggesting that these cells may be a potential reservoir for ongoing viral replication and proinflammatory cytokine production, leading to chronic adrenalitis and dysfunction.


Sujet(s)
Mort cellulaire , Zona , Maladies virales , Cortex surrénal , Mort cellulaire/immunologie , Mort cellulaire/physiologie , Zona/métabolisme , Zona/anatomopathologie , Herpèsvirus humain de type 3/physiologie , Humains , Inflammation/métabolisme , Interleukines/métabolisme , Culture de cellules primaires , Facteur de nécrose tumorale alpha/métabolisme , Réplication virale
5.
Article de Anglais | MEDLINE | ID: mdl-34759019

RÉSUMÉ

BACKGROUND AND OBJECTIVES: Compared with stroke controls, patients with varicella zoster virus (VZV) vasculopathy have increased amyloid in CSF, along with increased amylin (islet amyloid polypeptide [IAPP]) and anti-VZV antibodies. Thus, we examined the gene expression profiles of VZV-infected primary human brain vascular adventitial fibroblasts (HBVAFs), one of the initial arterial cells infected in VZV vasculopathy, to determine whether they are a potential source of amyloid that can disrupt vasculature and potentiate inflammation. METHODS: Mock- and VZV-infected quiescent HBVAFs were harvested at 3 days postinfection. Targeted RNA sequencing of the whole-human transcriptome (BioSpyder Technologies, TempO-Seq) was conducted followed by gene set enrichment and pathway analysis. Selected pathways unique to VZV-infected cells were confirmed by enzyme-linked immunoassays, migration assays, and immunofluorescence analysis (IFA) that included antibodies against amylin and amyloid-beta, as well as amyloid staining by Thioflavin-T. RESULTS: Compared with mock, VZV-infected HBVAFs had significantly enriched gene expression pathways involved in vascular remodeling and vascular diseases; confirmatory studies showed secretion of matrix metalloproteinase-3 and -10, as well increased migration of infected cells and uninfected cells when exposed to conditioned media from VZV-infected cells. In addition, significantly enriched pathways involved in amyloid-associated diseases (diabetes mellitus, amyloidosis, and Alzheimer disease), tauopathy, and progressive neurologic disorder were identified; predicted upstream regulators included amyloid precursor protein, apolipoprotein E, microtubule-associated protein tau, presenilin 1, and IAPP. Confirmatory IFA showed that VZV-infected HBVAFs contained amyloidogenic peptides (amyloid-beta and amylin) and intracellular amyloid. DISCUSSION: Gene expression profiles and pathway enrichment analysis of VZV-infected HBVAFs, as well as phenotypic studies, reveal features of pathologic vascular remodeling (e.g., increased cell migration and changes in the extracellular matrix) that can contribute to cerebrovascular disease. Furthermore, the discovery of amyloid-associated transcriptional pathways and intracellular amyloid deposition in HBVAFs raise the possibility that VZV vasculopathy is an amyloid disease. Amyloid deposition may contribute to cell death and loss of vascular wall integrity, as well as potentiate chronic inflammation in VZV vasculopathy, with disease severity and recurrence determined by the host's ability to clear virus infection and amyloid deposition and by the coexistence of other amyloid-associated diseases (i.e., Alzheimer disease and diabetes mellitus).


Sujet(s)
Adventice , Peptides bêta-amyloïdes/métabolisme , Angiopathies intracrâniennes , Fibroblastes , Infection à virus varicelle-zona , Remodelage vasculaire , Adventice/cytologie , Adventice/métabolisme , Adventice/anatomopathologie , Adventice/virologie , Cellules cultivées , Angiopathies intracrâniennes/métabolisme , Angiopathies intracrâniennes/anatomopathologie , Angiopathies intracrâniennes/virologie , Fibroblastes/cytologie , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Fibroblastes/virologie , Humains , Analyse de séquence d'ARN , Transcriptome/physiologie , Infection à virus varicelle-zona/métabolisme , Infection à virus varicelle-zona/anatomopathologie , Infection à virus varicelle-zona/virologie , Remodelage vasculaire/physiologie
6.
Article de Anglais | MEDLINE | ID: mdl-34493606

RÉSUMÉ

BACKGROUND AND OBJECTIVES: Varicella zoster virus (VZV) antigen has been detected in temporal arteries (TAs) of individuals with giant cell arteritis (GCA), the most common systemic vasculitis in older adults. Thus, we explored the contribution of VZV to GCA pathogenesis. METHODS: Formalin-fixed, paraffin-embedded TA sections from biopsy-positive GCA participants with VZV antigen (GCA/VZV-positive; n = 20) and without (GCA/VZV-negative, n = 20) and from normal participants with VZV antigen (control/VZV-positive, n = 11) and without (control/VZV-negative, n = 20) were analyzed by targeted RNA sequencing of the whole human transcriptome (BioSpyder TempO-Seq). Ingenuity pathway analysis and R-computational program were used to identify differentially expressed genes and pathways between groups. RESULTS: Compared with control/VZV-negative TAs, GCA/VZV-negative and GCA/VZV-positive TAs were significantly enriched for human transcripts specific for pathways involved in viral infections, including viral entry, nuclear factor kappa B activation by viruses, and other pathogen-related immune activation pathways. Similarly, human gene sets supporting viral infection were found in control/VZV-positive TAs that showed no morphological signs of inflammation, suggesting that the enriched pathways were not nonspecific signatures of infiltrating immune cells. All GCA TAs and control/VZV-positive TAs showed enrichment of transcripts involved in vascular remodeling, including smooth muscle cell migration. DISCUSSION: The detection of viral and immune activation pathways in GCA TAs supports a role for virus infection in GCA pathogenesis. In addition, the detection of viral pathways in control/VZV-positive TAs, along with vascular remodeling pathways, suggests that these samples may represent early infection with progression to clinical disease, depending on host and other environmental factors.


Sujet(s)
Antigènes viraux/isolement et purification , ADN viral/isolement et purification , Artérite à cellules géantes/virologie , Herpèsvirus humain de type 3 , Artères temporales/virologie , Sujet âgé , Femelle , Formaldéhyde , Analyse de profil d'expression de gènes , Artérite à cellules géantes/anatomopathologie , Humains , Mâle , Adulte d'âge moyen , Inclusion en paraffine , Analyse de séquence d'ARN , Artères temporales/anatomopathologie , Fixation tissulaire
7.
Viruses ; 13(7)2021 06 26.
Article de Anglais | MEDLINE | ID: mdl-34206909

RÉSUMÉ

Latent varicella zoster virus (VZV) has been detected in human adrenal glands, raising the possibility of virus-induced adrenal damage and dysfunction during primary infection or reactivation. Rare cases of bilateral adrenal hemorrhage and insufficiency associated with VZV reactivation have been reported. Since there is no animal model for VZV infection of adrenal glands, we obtained adrenal glands from two non-human primates (NHPs) that spontaneously developed varicella from primary simian varicella virus (SVV) infection, the NHP VZV homolog. Histological and immunohistochemical analysis revealed SVV antigen and DNA in the adrenal medulla and cortex of both animals. Adrenal glands were observed to have Cowdry A inclusion bodies, cellular necrosis, multiple areas of hemorrhage, and varying amounts of polymorphonuclear cells. No specific association of SVV antigen with ßIII-tubulin-positive nerve fibers was found. Overall, we found that SVV can productively infect NHP adrenal glands, and is associated with inflammation, hemorrhage, and cell death. These findings suggest that further studies are warranted to examine the contribution of VZV infection to human adrenal disease. This study also suggests that VZV infection may present itself as acute adrenal dysfunction with "long-hauler" symptoms of fatigue, weakness, myalgias/arthralgias, and hypotension.


Sujet(s)
Glandes surrénales/anatomopathologie , Glandes surrénales/virologie , Infections à Herpesviridae/anatomopathologie , Herpèsvirus humain de type 3/pathogénicité , Glandes surrénales/cytologie , Animaux , Femelle , Infections à Herpesviridae/virologie , Techniques histologiques , Macaca fascicularis/virologie , Mâle
8.
J Neurol Sci ; 422: 117315, 2021 Mar 15.
Article de Anglais | MEDLINE | ID: mdl-33503519

RÉSUMÉ

OBJECTIVE: Varicella zoster virus (VZV) vasculopathy and cerebral amyloid angiopathy (CAA) have similar clinical presentations: both affect cerebrovasculature in the elderly, produce hemorrhage, and can have a protracted course of cognitive decline and other neurological deficits. The cause of CAA is unknown, but amyloid-beta (Aß) is found within arterial walls. Recent studies show that VZV induces Aß and amylin expression and an amyloid-promoting environment. Thus, we determined if VZV was present in CAA-affected arteries. METHODS: Two subjects with pathologically-verified CAA were identified postmortem and frontal lobes analyzed by immunohistochemistry for arteries containing VZV, Aß, and amylin and H&E for pathological changes. VZV antigen detection was confirmed by PCR for VZV DNA in the same region. RESULTS: In both CAA cases, sections with cerebral arteries containing VZV antigen with corresponding VZV DNA were identified; VZV antigen co-localized with Aß in media of arteries with histological changes characteristic of CAA. Amylin was also seen in the intima of a VZV-positive artery in the diabetic subject. Not all Aß-containing arteries had VZV, but all VZV-positive arteries contained Aß. CONCLUSIONS: VZV antigen co-localized with Aß in some affected arteries from two CAA cases, suggesting a possible association between VZV infection and CAA.


Sujet(s)
Angiopathie amyloïde cérébrale , Herpèsvirus humain de type 3 , Sujet âgé , Peptides bêta-amyloïdes , Artères cérébrales , ADN , Humains
9.
J Infect Dis ; 223(7): 1284-1294, 2021 04 08.
Article de Anglais | MEDLINE | ID: mdl-32809013

RÉSUMÉ

BACKGROUND: Varicella zoster virus (VZV) vasculopathy is characterized by persistent arterial inflammation leading to stroke. Studies show that VZV induces amyloid formation that may aggravate vasculitis. Thus, we determined if VZV central nervous system infection produces amyloid. METHODS: Aß peptides, amylin, and amyloid were measured in cerebrospinal fluid (CSF) from 16 VZV vasculopathy subjects and 36 stroke controls. To determine if infection induced amyloid deposition, mock- and VZV-infected quiescent primary human perineurial cells (qHPNCs), present in vasculature, were analyzed for intracellular amyloidogenic transcripts/proteins and amyloid. Supernatants were assayed for amyloidogenic peptides and ability to induce amyloid formation. To determine amylin's function during infection, amylin was knocked down with small interfering RNA and viral complementary DNA (cDNA) was quantitated. RESULTS: Compared to controls, VZV vasculopathy CSF had increased amyloid that positively correlated with amylin and anti-VZV antibody levels; Aß40 was reduced and Aß42 unchanged. Intracellular amylin, Aß42, and amyloid were seen only in VZV-infected qHPNCs. VZV-infected supernatant formed amyloid fibrils following addition of amyloidogenic peptides. Amylin knockdown decreased viral cDNA. CONCLUSIONS: VZV infection increased levels of amyloidogenic peptides and amyloid in CSF and qHPNCs, indicating that VZV-induced amyloid deposition may contribute to persistent arterial inflammation in VZV vasculopathy. In addition, we identified a novel proviral function of amylin.


Sujet(s)
Peptides bêta-amyloïdes , Amyloïde , Artérite , Zona , Polypeptide amyloïde des ilots , Fragments peptidiques , Amyloïde/liquide cérébrospinal , Peptides bêta-amyloïdes/liquide cérébrospinal , Artérite/liquide cérébrospinal , Artérite/diagnostic , Artérite/virologie , ADN complémentaire , ADN viral , Zona/liquide cérébrospinal , Zona/diagnostic , Herpèsvirus humain de type 3 , Humains , Polypeptide amyloïde des ilots/liquide cérébrospinal , Fragments peptidiques/liquide cérébrospinal , Accident vasculaire cérébral
10.
J Neurovirol ; 26(3): 422-428, 2020 06.
Article de Anglais | MEDLINE | ID: mdl-32385803

RÉSUMÉ

Herpes zoster is associated with an increased dementia and neovascular macular degeneration risk and a decline in glycemic control in diabetes mellitus. Because amyloid is present and pathogenic in these diseases, we quantified amyloid, Aß40, Aß42, and amylin in 14 zoster and 10 control plasmas. Compared with controls, zoster plasma had significantly elevated amyloid that correlated with Aß42 and amylin levels and increased amyloid aggregation with addition of exogenous Aß42 or amylin. These results suggest that zoster plasma contains factor(s) that promotes aggregation of amyloidogenic peptides, potentially contributing to the toxic amyloid burden and explaining accelerated disease progression following zoster.


Sujet(s)
Peptides bêta-amyloïdes/génétique , Zona/sang , Herpèsvirus humain de type 3/pathogénicité , Polypeptide amyloïde des ilots/génétique , Fragments peptidiques/génétique , Agrégation pathologique de protéines/sang , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Peptides bêta-amyloïdes/sang , Études cas-témoins , Femelle , Expression des gènes , Zona/génétique , Zona/anatomopathologie , Herpèsvirus humain de type 3/croissance et développement , Interactions hôte-pathogène/génétique , Humains , Polypeptide amyloïde des ilots/sang , Mâle , Adulte d'âge moyen , Fragments peptidiques/sang , Agrégats de protéines , Agrégation pathologique de protéines/génétique , Agrégation pathologique de protéines/anatomopathologie
11.
J Infect Dis ; 221(7): 1088-1097, 2020 03 16.
Article de Anglais | MEDLINE | ID: mdl-31665341

RÉSUMÉ

BACKGROUND: Herpes zoster is linked to amyloid-associated diseases, including dementia, macular degeneration, and diabetes mellitus, in epidemiological studies. Thus, we examined whether varicella-zoster virus (VZV)-infected cells produce amyloid. METHODS: Production of intracellular amyloidogenic proteins (amylin, amyloid precursor protein [APP], and amyloid-ß [Aß]) and amyloid, as well as extracellular amylin, Aß, and amyloid, was compared between mock- and VZV-infected quiescent primary human spinal astrocytes (qHA-sps). The ability of supernatant from infected cells to induce amylin or Aß42 aggregation was quantitated. Finally, the amyloidogenic activity of viral peptides was examined. RESULTS: VZV-infected qHA-sps, but not mock-infected qHA-sps, contained intracellular amylin, APP, and/or Aß, and amyloid. No differences in extracellular amylin, Aß40, or Aß42 were detected, yet only supernatant from VZV-infected cells induced amylin aggregation and, to a lesser extent, Aß42 aggregation into amyloid fibrils. VZV glycoprotein B (gB) peptides assembled into fibrils and catalyzed amylin and Aß42 aggregation. CONCLUSIONS: VZV-infected qHA-sps produced intracellular amyloid and their extracellular environment promoted aggregation of cellular peptides into amyloid fibrils that may be due, in part, to VZV gB peptides. These findings suggest that together with host and other environmental factors, VZV infection may increase the toxic amyloid burden and contribute to amyloid-associated disease progression.


Sujet(s)
Peptides bêta-amyloïdes , Astrocytes , Polypeptide amyloïde des ilots , Infection à virus varicelle-zona/métabolisme , Aciclovir/pharmacologie , Peptides bêta-amyloïdes/composition chimique , Peptides bêta-amyloïdes/métabolisme , Antiviraux/pharmacologie , Astrocytes/effets des médicaments et des substances chimiques , Astrocytes/métabolisme , Astrocytes/virologie , Cellules cultivées , Espace extracellulaire/métabolisme , Humains , Espace intracellulaire/métabolisme , Polypeptide amyloïde des ilots/composition chimique , Polypeptide amyloïde des ilots/métabolisme , Protéines de l'enveloppe virale/composition chimique , Protéines de l'enveloppe virale/métabolisme
12.
J Infect Dis ; 220(9): 1453-1461, 2019 09 26.
Article de Anglais | MEDLINE | ID: mdl-30835269

RÉSUMÉ

BACKGROUND: In temporal arteries (TAs) from patients with giant cell arteritis, varicella zoster virus (VZV) is seen in perineurial cells that surround adventitial nerve bundles and form the peripheral nerve-extrafascicular tissue barrier (perineurium). We hypothesized that during VZV reactivation from ganglia, virus travels transaxonally and disrupts the perineurium to infect surrounding cells. METHODS: Mock- and VZV-infected primary human perineurial cells (HPNCs) were examined for alterations in claudin-1, E-cadherin, and N-cadherin. Conditioned supernatant was analyzed for a soluble factor(s) mediating these alterations and for the ability to increase cell migration. To corroborate in vitro findings, a VZV-infected TA was examined. RESULTS: In VZV-infected HPNCs, claudin-1 redistributed to the nucleus; E-cadherin was lost and N-cadherin gained, with similar changes seen in VZV-infected perineurial cells in a TA. VZV-conditioned supernatant contained increased interleukin 6 (IL-6) that induced E-cadherin loss and N-cadherin gain and increased cell migration when added to uninfected HPNCs; anti-IL-6 receptor antibody prevented these changes. CONCLUSIONS: IL-6 secreted from VZV-infected HPNCs facilitated changes in E- and N-cadherin expression and cell migration, reminiscent of an epithelial-to-mesenchymal cell transition, potentially contributing to loss of perineurial cell barrier integrity and viral spread. Importantly, an anti-IL-6 receptor antibody prevented virus-induced perineurial cell disruption.


Sujet(s)
Antigènes CD/biosynthèse , Cadhérines/biosynthèse , Herpèsvirus humain de type 3/croissance et développement , Interactions hôte-pathogène , Interleukine-6/métabolisme , Myofibroblastes/virologie , Mouvement cellulaire , Cellules cultivées , Claudine-1/biosynthèse , Milieux de culture conditionnés , Expression des gènes , Humains , Myofibroblastes/métabolisme
13.
J Infect Dis ; 218(8): 1324-1335, 2018 09 08.
Article de Anglais | MEDLINE | ID: mdl-29788447

RÉSUMÉ

Background: Varicella zoster virus (VZV) can present as a myelopathy with spinal astrocyte infection. Recent studies support a role for the neurokinin-1 receptor (NK-1R) in virus infections, as well as for cytoskeletal alterations that may promote viral spread. Thus, we examined the role of NK-1R in VZV-infected primary human spinal astrocytes (HA-sps) to shed light on the pathogenesis of VZV myelopathy. Methods: Mock- and VZV-infected HA-sps were examined for substance P (subP) production, NK-1R localization, morphological changes, and viral spread in the presence or absence of the NK-1R antagonists aprepitant and rolapitant. Results: VZV infection of HA-sps induced nuclear localization of full-length and truncated NK-1R in the absence of the endogenous ligand, subP, and was associated with extensive lamellipodia formation and viral spread that was inhibited by NK-1R antagonists. Conclusions: We have identified a novel, subP-independent, proviral function of nuclear NK-1R associated with lamellipodia formation and viral spread that is distinct from subP-induced NK-1R cell membrane/cytoplasmic localization without lamellipodia formation. These results suggest that binding of a putative viral ligand to NK-1R produces a dramatically different NK-1R downstream effect than binding of subP. Finally, the NK-1R antagonists aprepitant and rolapitant provide promising alternatives to nucleoside analogs in treating VZV infections, including myelopathy.


Sujet(s)
Transport nucléaire actif/physiologie , Astrocytes/physiologie , Astrocytes/virologie , Herpèsvirus humain de type 3/physiologie , Pseudopodes/physiologie , Récepteur de la neurokinine 1/métabolisme , Aprépitant/pharmacologie , Cellules cultivées , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Humains , Antagonistes du récepteur de la neurokinine-1/pharmacologie , Isoformes de protéines , Spiranes/pharmacologie , Substance P
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...