Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Psychother Res ; 32(4): 428-439, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34338165

RESUMO

OBJECTIVE: To provide evidence of the effectiveness of a brief relapse prevention intervention using implementation intentions (Self-Management after Therapy, SMArT), following remission from depression and to identify effective relapse prevention strategies. METHOD: The SMArT intervention was provided to 107 patients who were recovered after psychological therapy for depression. Relapse events were calculated as reliable and clinically significant increases in PHQ-scores. Sixteen patients receiving the intervention and eight practitioners providing it were interviewed. Framework Analysis identified seven themes which highlighted effective relapse prevention strategies and effective implementation of the SMArT intervention. RESULTS: Relapse rates at the final SMArT session (four months after the end of acute stage therapy) were 11%. Seven themes were identified that supported effective self-management: (1) Relationship with the practitioner-feeling supported; (2) Support networks; (3) Setting goals, implementing plans and routine; (4) Changing views of recovery; (5) The SMArT sessions-mode, content, timing, duration; (6) Suitability for the person; and (7) Suitability for the service. CONCLUSION: The study provides some support for the effectiveness of the SMArT intervention, although a randomized controlled trial is required; and identifies important relapse prevention strategies.


Assuntos
Depressão , Intenção , Doença Crônica , Depressão/terapia , Humanos , Recidiva , Prevenção Secundária
2.
J Psychiatr Ment Health Nurs ; 27(2): 162-171, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31495046

RESUMO

WHAT IS KNOWN ON THE SUBJECT?: Addressing spiritual issues to maintain a sense of hope, meaning and purpose can be an important aspect of mental health care which goes beyond simply providing facilities for religious observance. Expressions of spiritual need from service users can potentially be confused with symptoms of mental ill health. Little is known about how mental health nurses understand or provide this aspect of care for service users. WHAT THE PAPER ADDS TO EXISTING KNOWLEDGE?: An understanding from the mental health nurse perspective of how mental health nurses understand and care for service users' spiritual needs, and what influences their practice in this area. Ideas about how education and opportunities for good practice in this area might be advanced. WHAT ARE THE IMPLICATIONS FOR PRACTICE?: Nurses need more education and guidance as well as supportive team and management cultures so that they feel comfortable and able to deliver this important aspect of care. Abstract Introduction Mental health nurses have a professional obligation to attend to service users' spiritual needs, but little is known about specific issues related to provision of care for spiritual need faced by mental health nurses or how nurses understand this aspect of care and deliver it in practice. Aim/Question To explore mental health nurses' ́understandings of spiritual need and their experiences of delivering this care for service users. Method A qualitative study was conducted in one NHS mental health service. Interviews were undertaken with seventeen mental health nurses practising in a variety of areas. Results Four themes were generated from thematic analysis of data in the template style: Expressing personal perspectives on spirituality; Expressing perspectives on spirituality as a nursing professional; Nursing spiritually; and Permeating anxiety (integrative). Discussion Participants had complex understandings of spiritual need and evident anxieties in relation to this area of care. Two different approaches to nursing spiritually are characterised as (a) pragmatic (concerned with procedural aspects of care) and (b) spiritually empathetic. Mental health nurses were uncertain about the acceptability of attention to spiritual issues as part of care and anxious about distinguishing between symptoms of mental ill health and spiritual needs. Implications for practice Educational experiences need to emphasise both pragmatic and empathetic approaches, and work needs to be organised to support good practice.


Assuntos
Atitude do Pessoal de Saúde , Transtornos Mentais/enfermagem , Serviços de Saúde Mental , Papel do Profissional de Enfermagem , Recursos Humanos de Enfermagem , Enfermagem Psiquiátrica , Espiritualidade , Adulto , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Pesquisa Qualitativa
3.
PLoS Pathog ; 14(4): e1006989, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29652922

RESUMO

The OAS/RNase L pathway is one of the best-characterized effector pathways of the IFN antiviral response. It inhibits the replication of many viruses and ultimately promotes apoptosis of infected cells, contributing to the control of virus spread. However, viruses have evolved a range of escape strategies that act against different steps in the pathway. Here we unraveled a novel escape strategy involving Theiler's murine encephalomyelitis virus (TMEV) L* protein. Previously we found that L* was the first viral protein binding directly RNase L. Our current data show that L* binds the ankyrin repeats R1 and R2 of RNase L and inhibits 2'-5' oligoadenylates (2-5A) binding to RNase L. Thereby, L* prevents dimerization and oligomerization of RNase L in response to 2-5A. Using chimeric mouse hepatitis virus (MHV) expressing TMEV L*, we showed that L* efficiently inhibits RNase L in vivo. Interestingly, those data show that L* can functionally substitute for the MHV-encoded phosphodiesterase ns2, which acts upstream of L* in the OAS/RNase L pathway, by degrading 2-5A.


Assuntos
2',5'-Oligoadenilato Sintetase/metabolismo , Nucleotídeos de Adenina/metabolismo , Endorribonucleases/antagonistas & inibidores , Vírus da Hepatite Murina/fisiologia , Oligorribonucleotídeos/metabolismo , Theilovirus/metabolismo , Proteínas Virais/metabolismo , Animais , Antivirais/metabolismo , Endorribonucleases/fisiologia , Células HeLa , Hepatite Viral Animal/metabolismo , Hepatite Viral Animal/virologia , Interações Hospedeiro-Patógeno , Humanos , Camundongos
4.
J Virol ; 92(1)2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29046453

RESUMO

Coronaviruses (CoVs) are positive-sense RNA viruses that infect numerous mammalian and avian species and are capable of causing severe and lethal disease in humans. CoVs encode several innate immune antagonists that counteract the host innate immune response to facilitate efficient viral replication. CoV nonstructural protein 14 (nsp14) encodes 3'-to-5' exoribonuclease activity (ExoN), which performs a proofreading function and is required for high-fidelity replication. Outside of the order Nidovirales, arenaviruses are the only RNA viruses that encode an ExoN, which functions to degrade double-stranded RNA (dsRNA) replication intermediates. In this study, we tested the hypothesis that CoV ExoN also functions to antagonize the innate immune response. We demonstrate that viruses lacking ExoN activity [ExoN(-)] are sensitive to cellular pretreatment with interferon beta (IFN-ß) in a dose-dependent manner. In addition, ExoN(-) virus replication was attenuated in wild-type bone marrow-derived macrophages (BMMs) and partially restored in interferon alpha/beta receptor-deficient (IFNAR-/-) BMMs. ExoN(-) virus replication did not result in IFN-ß gene expression, and in the presence of an IFN-ß-mediated antiviral state, ExoN(-) viral RNA levels were not substantially reduced relative to those of untreated samples. However, ExoN(-) virus generated from IFN-ß-pretreated cells had reduced specific infectivity and decreased relative fitness, suggesting that ExoN(-) virus generated during an antiviral state is less viable to establish a subsequent infection. Overall, our data suggest murine hepatitis virus (MHV) ExoN activity is required for resistance to the innate immune response, and antiviral mechanisms affecting the viral RNA sequence and/or an RNA modification act on viruses lacking ExoN activity.IMPORTANCE CoVs encode multiple antagonists that prevent or disrupt an efficient innate immune response. Additionally, no specific antiviral therapies or vaccines currently exist for human CoV infections. Therefore, the study of CoV innate immune antagonists is essential for understanding how CoVs overcome host defenses and to maximize potential therapeutic interventions. Here, we sought to determine the contributions of nsp14 ExoN activity in the induction of and resistance to the innate immune response. We show that viruses lacking nsp14 ExoN activity are more sensitive than wild-type MHV to restriction by exogenous IFN-ß and that viruses produced in the presence of an antiviral state are less capable of establishing a subsequent viral infection. Our results support the hypothesis that murine hepatitis virus ExoN activity is required for resistance to the innate immune response.


Assuntos
Exorribonucleases/genética , Exorribonucleases/metabolismo , Imunidade Inata , Vírus da Hepatite Murina/enzimologia , Vírus da Hepatite Murina/imunologia , Proteínas não Estruturais Virais/metabolismo , Animais , Antivirais/farmacologia , Genoma Viral , Interferon beta/farmacologia , Camundongos , Vírus da Hepatite Murina/efeitos dos fármacos , Vírus da Hepatite Murina/genética , Mutagênese , Mutação , RNA Viral/metabolismo , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/imunologia , Replicação Viral/efeitos dos fármacos
5.
J Neurovirol ; 23(6): 845-854, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28895072

RESUMO

The inflammasome, a cytosolic protein complex that mediates the processing and secretion of pro-inflammatory cytokines, is one of the first responders during viral infection. The cytokines secreted following inflammasome activation, which include IL-1 and IL-18, regulate cells of both the innate and adaptive immune system, guiding the subsequent immune responses. In this study, we used murine coronavirus, mouse hepatitis virus (MHV), infection of the central nervous system and liver to assess of the role of the inflammasome and its related cytokines on pathogenesis and host defense during viral infection. Mice lacking all inflammasome signaling due to the absence of caspase-1 and -11 were more vulnerable to infection, with poor survival and elevated viral replication compared to wild-type mice. Mice lacking IL-1 signaling experienced elevated viral replication but similar survival compared to wild-type controls. In the absence of IL-18, mice had elevated viral replication and poor survival, and this protective effect of IL-18 was found to be due to promotion of interferon gamma production in αß T cells. These data suggest that inflammasome signaling is largely protective during murine coronavirus infection, in large part due to the pro-inflammatory effects of IL-18.


Assuntos
Infecções por Coronavirus/imunologia , Interleucina-18/imunologia , Interleucina-1/imunologia , Vírus da Hepatite Murina/imunologia , Transdução de Sinais/imunologia , Imunidade Adaptativa , Animais , Caspase 1/deficiência , Caspase 1/genética , Caspase 1/imunologia , Caspases/deficiência , Caspases/genética , Caspases/imunologia , Caspases Iniciadoras , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/patologia , Sistema Nervoso Central/virologia , Infecções por Coronavirus/mortalidade , Infecções por Coronavirus/patologia , Infecções por Coronavirus/virologia , Regulação da Expressão Gênica , Imunidade Inata , Inflamassomos/imunologia , Inflamassomos/metabolismo , Interferon gama/genética , Interferon gama/imunologia , Interleucina-1/deficiência , Interleucina-1/genética , Interleucina-18/deficiência , Interleucina-18/genética , Fígado/imunologia , Fígado/patologia , Fígado/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Vírus da Hepatite Murina/patogenicidade , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Análise de Sobrevida , Linfócitos T/imunologia , Linfócitos T/virologia , Carga Viral , Replicação Viral
6.
J Virol ; 91(5)2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28003490

RESUMO

Viruses in the family Coronaviridae, within the order Nidovirales, are etiologic agents of a range of human and animal diseases, including both mild and severe respiratory diseases in humans. These viruses encode conserved replicase and structural proteins as well as more diverse accessory proteins, encoded in the 3' ends of their genomes, that often act as host cell antagonists. We previously showed that 2',5'-phosphodiesterases (2',5'-PDEs) encoded by the prototypical Betacoronavirus, mouse hepatitis virus (MHV), and by Middle East respiratory syndrome-associated coronavirus antagonize the oligoadenylate-RNase L (OAS-RNase L) pathway. Here we report that additional coronavirus superfamily members, including lineage A betacoronaviruses and toroviruses infecting both humans and animals, encode 2',5'-PDEs capable of antagonizing RNase L. We used a chimeric MHV system (MHVMut) in which exogenous PDEs were expressed from an MHV backbone lacking the gene for a functional NS2 protein, the endogenous RNase L antagonist. With this system, we found that 2',5'-PDEs encoded by the human coronavirus HCoV-OC43 (OC43; an agent of the common cold), human enteric coronavirus (HECoV), equine coronavirus (ECoV), and equine torovirus Berne (BEV) are enzymatically active, rescue replication of MHVMut in bone marrow-derived macrophages, and inhibit RNase L-mediated rRNA degradation in these cells. Additionally, PDEs encoded by OC43 and BEV rescue MHVMut replication and restore pathogenesis in wild-type (WT) B6 mice. This finding expands the range of viruses known to encode antagonists of the potent OAS-RNase L antiviral pathway, highlighting its importance in a range of species as well as the selective pressures exerted on viruses to antagonize it.IMPORTANCE Viruses in the family Coronaviridae include important human and animal pathogens, including the recently emerged viruses severe acute respiratory syndrome-associated coronavirus (SARS-CoV) and Middle East respiratory syndrome-associated coronavirus (MERS-CoV). We showed previously that two viruses within the genus Betacoronavirus, mouse hepatitis virus (MHV) and MERS-CoV, encode 2',5'-phosphodiesterases (2',5'-PDEs) that antagonize the OAS-RNase L pathway, and we report here that these proteins are furthermore conserved among additional coronavirus superfamily members, including lineage A betacoronaviruses and toroviruses, suggesting that they may play critical roles in pathogenesis. As there are no licensed vaccines or effective antivirals against human coronaviruses and few against those infecting animals, identifying viral proteins contributing to virulence can inform therapeutic development. Thus, this work demonstrates that a potent antagonist of host antiviral defenses is encoded by multiple and diverse viruses within the family Coronaviridae, presenting a possible broad-spectrum therapeutic target.


Assuntos
Endorribonucleases/metabolismo , Coronavírus da Síndrome Respiratória do Oriente Médio/enzimologia , Vírus da Hepatite Murina/enzimologia , Diester Fosfórico Hidrolases/fisiologia , Torovirus/enzimologia , Proteínas não Estruturais Virais/fisiologia , Nucleotídeos de Adenina/química , Sequência de Aminoácidos , Animais , Domínio Catalítico , Linhagem Celular , Sequência Conservada , Cricetinae , Ativação Enzimática , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oligorribonucleotídeos/química , Diester Fosfórico Hidrolases/química , Proteínas não Estruturais Virais/química , Replicação Viral
7.
Int Urogynecol J ; 27(9): 1393-400, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26942594

RESUMO

INTRODUCTION AND HYPOTHESIS: This study was designed to evaluate the effects of CP55,940 on normal bladder function in vivo and examine whether it suppresses urinary frequency induced by nociceptive stimuli in the bladder. Cannabinoid receptor (CBR) activity may be involved in the regulation of bladder function. However, the role of CBR subtypes in micturition has yet to be established. CP55,940 is a synthetic analogue of tetrahydrocannabidiol, which is a psychoactive ingredient of the Cannabis plant. METHODS: Cystometry under urethane anaesthesia was performed to evaluate the effect of intravesical delivery of CP55,940 with or without administration of CB1 antagonist AM251 or CB2 antagonist AM630 on bladder function in female rats. The effects of CP55,940 were also examined in rats with urinary irritation induced by intravesical infusion of acetic acid. RESULTS: Infusion of CP55,940 significantly (p < 0.05) increased micturition interval (MI) and bladder capacity (BC) by 52 % and decreased maximal voiding pressure (MP) by 25 %. Pretreatment with AM251 or AM630 before CP55,940 administration prevented CP55,940-induced increases in MI, BC and reduced MP. Acetic acid induced urinary frequency as evidenced by a reduction in MI and was suppressed by CP55,940. CONCLUSIONS: CP55,940 decreases bladder activity and urinary frequency induced by nociceptive stimuli, probably by suppression of bladder afferent activity. Effects of CP55,940 were abolished by both CBR antagonists. This data implicates a role for the endocannabinoid system in bladder mechanoafferent function in rats. In addition, our results show that CP55,940 reverses urinary frequency exemplified in an overactive bladder model, suggesting it could be an effective treatment for patients with lower urinary tract symptoms.


Assuntos
Agonistas de Receptores de Canabinoides/farmacologia , Cicloexanóis/farmacologia , Bexiga Urinária Hiperativa/tratamento farmacológico , Bexiga Urinária/efeitos dos fármacos , Ácido Acético , Administração Intravesical , Animais , Modelos Animais de Doenças , Feminino , Indóis/administração & dosagem , Piperidinas/administração & dosagem , Pirazóis/administração & dosagem , Ratos , Ratos Sprague-Dawley , Resultado do Tratamento , Bexiga Urinária/fisiopatologia , Bexiga Urinária Hiperativa/induzido quimicamente , Bexiga Urinária Hiperativa/fisiopatologia , Micção/efeitos dos fármacos , Urodinâmica/efeitos dos fármacos
8.
mBio ; 7(2): e00258, 2016 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-27025250

RESUMO

UNLABELLED: Middle East respiratory syndrome coronavirus (MERS-CoV) is the first highly pathogenic human coronavirus to emerge since severe acute respiratory syndrome coronavirus (SARS-CoV) in 2002. Like many coronaviruses, MERS-CoV carries genes that encode multiple accessory proteins that are not required for replication of the genome but are likely involved in pathogenesis. Evasion of host innate immunity through interferon (IFN) antagonism is a critical component of viral pathogenesis. The IFN-inducible oligoadenylate synthetase (OAS)-RNase L pathway activates upon sensing of viral double-stranded RNA (dsRNA). Activated RNase L cleaves viral and host single-stranded RNA (ssRNA), which leads to translational arrest and subsequent cell death, preventing viral replication and spread. Here we report that MERS-CoV, a lineage CBetacoronavirus, and related bat CoV NS4b accessory proteins have phosphodiesterase (PDE) activity and antagonize OAS-RNase L by enzymatically degrading 2',5'-oligoadenylate (2-5A), activators of RNase L. This is a novel function for NS4b, which has previously been reported to antagonize IFN signaling. NS4b proteins are distinct from lineage ABetacoronavirusPDEs and rotavirus gene-encoded PDEs, in having an amino-terminal nuclear localization signal (NLS) and are localized mostly to the nucleus. However, the expression level of cytoplasmic MERS-CoV NS4b protein is sufficient to prevent activation of RNase L. Finally, this is the first report of an RNase L antagonist expressed by a human or bat coronavirus and provides a specific mechanism by which this occurs. Our findings provide a potential mechanism for evasion of innate immunity by MERS-CoV while also identifying a potential target for therapeutic intervention. IMPORTANCE: Middle East respiratory syndrome coronavirus (MERS-CoV) is the first highly pathogenic human coronavirus to emerge since severe acute respiratory syndrome coronavirus (SARS-CoV). MERS-CoV, like other coronaviruses, carries genes that encode accessory proteins that antagonize the host antiviral response, often the type I interferon response, and contribute to virulence. We found that MERS-CoV NS4b and homologs from related lineage C bat betacoronaviruses BtCoV-SC2013 (SC2013) and BtCoV-HKU5 (HKU5) are members of the 2H-phosphoesterase (2H-PE) enzyme family with phosphodiesterase (PDE) activity. Like murine coronavirus NS2, a previously characterized PDE, MERS NS4b, can antagonize activation of the OAS-RNase L pathway, an interferon-induced potent antiviral activity. Furthermore, MERS-CoV mutants with deletion of genes encoding accessory proteins NS3 to NS5 or NS4b alone or inactivation of the PDE can activate RNase L during infection of Calu-3 cells. Our report may offer a potential target for therapeutic intervention if NS4b proves to be critical to pathogenesis inin vivomodels of MERS-CoV infection.


Assuntos
Nucleotídeos de Adenina/metabolismo , Endorribonucleases/antagonistas & inibidores , Interações Hospedeiro-Patógeno , Coronavírus da Síndrome Respiratória do Oriente Médio/fisiologia , Oligorribonucleotídeos/metabolismo , Proteínas não Estruturais Virais/metabolismo , Animais , Cricetinae , Humanos , Evasão da Resposta Imune , Camundongos , Sinais de Localização Nuclear , Proteínas não Estruturais Virais/genética
9.
J Virol ; 90(6): 3160-72, 2016 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-26739051

RESUMO

UNLABELLED: The oligoadenylate synthetase (OAS)-RNase L pathway is a potent interferon (IFN)-induced antiviral activity. Upon sensing double-stranded RNA, OAS produces 2',5'-oligoadenylates (2-5A), which activate RNase L. Murine coronavirus (mouse hepatitis virus [MHV]) nonstructural protein 2 (ns2) is a 2',5'-phosphodiesterase (PDE) that cleaves 2-5A, thereby antagonizing RNase L activation. PDE activity is required for robust replication in myeloid cells, as a mutant of MHV (ns2(H126R)) encoding an inactive PDE fails to antagonize RNase L activation and replicates poorly in bone marrow-derived macrophages (BMM), while ns2(H126R) replicates to high titer in several types of nonmyeloid cells, as well as in IFN receptor-deficient (Ifnar1(-/-)) BMM. We reported previously that myeloid cells express significantly higher basal levels of OAS transcripts than nonmyeloid cells. Here, we investigated the contributions of Oas gene expression, basal IFN signaling, and virus-induced IFN to RNase L activation. Infection with ns2(H126R) activated RNase L in Ifih1(-/-) BMM to a similar extent as in wild-type (WT) BMM, despite the lack of IFN induction in the absence of MDA5 expression. However, ns2(H126R) failed to induce RNase L activation in BMM treated with IFNAR1-blocking antibody, as well as in Ifnar1(-/-) BMM, both expressing low basal levels of Oas genes. Thus, activation of RNase L does not require virus-induced IFN but rather correlates with adequate levels of basal Oas gene expression, maintained by basal IFN signaling. Finally, overexpression of RNase L is not sufficient to compensate for inadequate basal OAS levels. IMPORTANCE: The oligoadenylate synthetase (OAS)-RNase L pathway is a potent antiviral activity. Activation of RNase L during murine coronavirus (mouse hepatitis virus [MHV]) infection of myeloid cells correlates with high basal Oas gene expression and is independent of virus-induced interferon secretion. Thus, our data suggest that cells with high basal Oas gene expression levels can activate RNase L and thereby inhibit virus replication early in infection upon exposure to viral double-stranded RNA (dsRNA) before the induction of interferon and prior to transcription of interferon-stimulated antiviral genes. These findings challenge the notion that activation of the OAS-RNase L pathway requires virus to induce type I IFN, which in turn upregulates OAS gene expression, as well as to provide dsRNA to activate OAS. Our data further suggest that myeloid cells may serve as sentinels to restrict viral replication, thus protecting other cell types from infection.


Assuntos
2',5'-Oligoadenilato Sintetase/metabolismo , Endorribonucleases/biossíntese , Expressão Gênica , Interações Hospedeiro-Patógeno , Vírus da Hepatite Murina/fisiologia , Células Mieloides/enzimologia , Células Mieloides/virologia , Animais , Células Cultivadas , Camundongos , Camundongos Knockout
10.
Int Urogynecol J ; 27(1): 129-39, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26224382

RESUMO

INTRODUCTION: Immunohistochemical (IHC) evidence shows that cannabinoid receptors (CB) are expressed in human bladders and cannabinoid agonists are known to inhibit detrusor contractility. However, the mechanism for this inhibition remains unknown. In addition, the role of CB in detrusor overactivity (DO) is under-investigated. The aim of this study was to compare CB expression in normal and DO human bladders and to further characterise these receptors. METHODS: Polymer chain reaction (PCR) was used to detect differences in CB transcripts in bladder samples. Differences in CB protein expression was assessed by IHC. Immunofluorescence (IF) was used to evaluate co-localisation of CB with nerve fibres. Receptor density and binding affinity were measured using the cannabinoid radioligand [(3)H]-CP-55,940. RESULTS: There were higher levels of CB1 transcripts in the urothelium of patients with DO and lower levels in the detrusor, compared with normal bladders. Radioligand binding revealed CB density of 421 ± 104 fmol/mg protein in normal human bladders. IHC confirmed these findings at the protein level. IF staining demonstrated co-localisation of CB1 with choline acetyltransferase-(ChAT)-positive nerves in the detrusor and co-localisation with PGP9.5 in both urothelium and detrusor. CB2 was co-localised with both ChAT and PGP9.5 in the urothelium and the detrusor. CONCLUSIONS: Cannabinoid receptor expression is reduced in the detrusor of patients with DO, which may play a role in the pathophysiology of the disease. Co-localisation of CB receptors with cholinergic nerves may suggest that CB1, being localised on pre- and postsynaptic terminals, could influence neurotransmitter release. Our findings suggest the potential role of cannabinoid agonists in overactive bladder pharmacotherapy.


Assuntos
Receptores de Canabinoides/biossíntese , Bexiga Urinária Hiperativa/metabolismo , Bexiga Urinária/metabolismo , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Bexiga Urinária Hiperativa/fisiopatologia
11.
J Virol ; 89(24): 12330-40, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26423942

RESUMO

UNLABELLED: Infection with the murine coronavirus mouse hepatitis virus (MHV) activates the pattern recognition receptors melanoma differentiation-associated gene 5 (MDA5) and Toll-like receptor 7 (TLR7) to induce transcription of type I interferon. Type I interferon is crucial for control of viral replication and spread in the natural host, but the specific contributions of MDA5 signaling to this pathway as well as to pathogenesis and subsequent immune responses are largely unknown. In this study, we use MHV infection of the liver as a model to demonstrate that MDA5 signaling is critically important for controlling MHV-induced pathology and regulation of the immune response. Mice deficient in MDA5 expression (MDA5(-/-) mice) experienced more severe disease following MHV infection, with reduced survival, increased spread of virus to additional sites of infection, and more extensive liver damage than did wild-type mice. Although type I interferon transcription decreased in MDA5(-/-) mice, the interferon-stimulated gene response remained intact. Cytokine production by innate and adaptive immune cells was largely intact in MDA5(-/-) mice, but perforin induction by natural killer cells and levels of interferon gamma, interleukin-6 (IL-6), and tumor necrosis factor alpha (TNF-α) in serum were elevated. These data suggest that MDA5 signaling reduces the severity of MHV-induced disease, at least in part by reducing the intensity of the proinflammatory cytokine response. IMPORTANCE: Multicellular organisms employ a wide range of sensors to detect viruses and other pathogens. One such sensor, MDA5, detects viral RNA and triggers induction of type I interferons, chemical messengers that induce inflammation and help regulate the immune responses. In this study, we sought to determine the role of MDA5 during infection with mouse hepatitis virus, a murine coronavirus used to model viral hepatitis as well as other human diseases. We found that mice lacking the MDA5 sensor were more susceptible to infection than were mice with MDA5 and experienced decreased survival. Viral replication in the liver was similar in mice with and without MDA5, but liver damage was increased in MDA5(-/-) mice, suggesting that the immune response is causing the damage. Production of several proinflammatory cytokines was elevated in MDA5(-/-) mice, suggesting that MDA5 may be responsible for keeping pathological inflammatory responses in check.


Assuntos
Infecções por Coronavirus/imunologia , RNA Helicases DEAD-box/imunologia , Vírus da Hepatite Murina/imunologia , Transdução de Sinais/imunologia , Animais , Linhagem Celular , Infecções por Coronavirus/genética , RNA Helicases DEAD-box/genética , Humanos , Interferon Tipo I/genética , Interferon Tipo I/imunologia , Helicase IFIH1 Induzida por Interferon , Interleucina-6/genética , Interleucina-6/imunologia , Camundongos , Camundongos Knockout , Vírus da Hepatite Murina/genética , Transdução de Sinais/genética , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
12.
mBio ; 5(4): e01312-14, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24987090

RESUMO

Viral 2',5'-phosphodiesterases (2',5'-PDEs) help disparate RNA viruses evade the antiviral activity of interferon (IFN) by degrading 2',5'-oligoadenylate (2-5A) activators of RNase L. A kinase anchoring proteins (AKAPs) bind the regulatory subunits of protein kinase A (PKA) to localize and organize cyclic AMP (cAMP) signaling during diverse physiological processes. Among more than 43 AKAP isoforms, AKAP7 appears to be unique in its homology to viral 2',5'-PDEs. Here we show that mouse AKAP7 rapidly degrades 2-5A with kinetics similar to that of murine coronavirus (mouse hepatitis virus [MHV]) strain A59 ns2 and human rotavirus strain WA VP3 proteins. To determine whether AKAP7 could substitute for a viral 2',5'-PDE, we inserted AKAP7 cDNA into an MHV genome with an inactivated ns2 gene. The AKAP7 PDE domain or N-terminally truncated AKAP7 (both lacking a nuclear localization motif), but not full-length AKAP7 or a mutant, AKAP7(H185R), PDE domain restored the infectivity of ns2 mutant MHV in bone marrow macrophages and in livers of infected mice. Interestingly, the AKAP7 PDE domain and N-terminally deleted AKAP7 were present in the cytoplasm (the site of MHV replication), whereas full-length AKAP7 was observed only in nuclei. We suggest the possibility that viral acquisition of the host AKAP7 PDE domain might have occurred during evolution, allowing diverse RNA viruses to antagonize the RNase L pathway. Importance: Early virus-host interactions determine whether an infection is established, highlighting the need to understand fundamental mechanisms regulating viral pathogenesis. Recently, our laboratories reported a novel mode of regulation of the IFN antiviral response. We showed that the coronavirus MHV accessory protein ns2 antagonizes the type I IFN response, promoting viral replication and hepatitis. ns2 confers virulence by cleaving 2',5'-oligoadenylate (2-5A) activators of RNase L in macrophages. We also reported that the rotavirus VP3 C-terminal domain (VP3-CTD) cleaves 2-5A and that it may rescue ns2 mutant MHV. Here we report that a cellular protein, AKAP7, has an analogous 2',5'-phosphodiesterase (2',5'-PDE) domain that is able to restore the growth of chimeric MHV expressing inactive ns2. The proviral effect requires cytoplasmic localization of the AKAP7 PDE domain. We speculate that AKAP7 is the ancestral precursor of viral proteins, such as ns2 and VP3, that degrade 2-5A to evade the antiviral activity of RNase L.


Assuntos
Proteínas de Ancoragem à Quinase A/química , Proteínas de Ancoragem à Quinase A/metabolismo , Coronavirus/metabolismo , Proteínas não Estruturais Virais/metabolismo , Proteínas de Ancoragem à Quinase A/genética , Animais , Western Blotting , Linhagem Celular , Coronavirus/genética , Proteínas Quinases Dependentes de AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Endorribonucleases/genética , Endorribonucleases/metabolismo , Humanos , Camundongos , Camundongos Knockout , Proteínas não Estruturais Virais/genética
13.
Antimicrob Agents Chemother ; 58(8): 4894-8, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24841268

RESUMO

We have previously shown that SSYA10-001 blocks severe acute respiratory syndrome coronavirus (SARS-CoV) replication by inhibiting SARS-CoV helicase (nsp13). Here, we show that SSYA10-001 also inhibits replication of two other coronaviruses, mouse hepatitis virus (MHV) and Middle Eastern respiratory syndrome coronavirus (MERS-CoV). A putative binding pocket for SSYA10-001 was identified and shown to be similar in SARS-CoV, MERS-CoV, and MHV helicases. These studies show that it is possible to target multiple coronaviruses through broad-spectrum inhibitors.


Assuntos
Antivirais/farmacologia , DNA Helicases/antagonistas & inibidores , Coronavírus da Síndrome Respiratória do Oriente Médio/efeitos dos fármacos , Vírus da Hepatite Murina/efeitos dos fármacos , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/efeitos dos fármacos , Triazóis/farmacologia , Proteínas Virais/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Antivirais/química , Sítios de Ligação , Linhagem Celular , Chlorocebus aethiops , DNA Helicases/química , Relação Dose-Resposta a Droga , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Fibroblastos/virologia , Humanos , Concentração Inibidora 50 , Camundongos , Coronavírus da Síndrome Respiratória do Oriente Médio/fisiologia , Simulação de Acoplamento Molecular , Dados de Sequência Molecular , Vírus da Hepatite Murina/fisiologia , Ligação Proteica , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/fisiologia , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Triazóis/química , Células Vero , Proteínas Virais/química , Replicação Viral/efeitos dos fármacos
14.
Naunyn Schmiedebergs Arch Pharmacol ; 387(6): 581-9, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24652077

RESUMO

To characterize human urothelial cell lines' cannabinoid receptor expression and evaluate their possible use for studying signalling interactions with purinergic and muscarinic receptor activation. PCR was used to detect cannabinoid (CB), muscarinic and purinergic receptor transcripts in HCV29 and UROtsa cells, whilst immunofluorescence evaluated protein expression and localization of cannabinoid receptors. The effect of CB1 agonist (ACEA) on carbachol- and ATP-induced changes in intracellular calcium ([Ca(2+)]i) levels was measured using fluorimetry. The ability of ACEA to reduce intracellular cAMP was investigated in HCV29 cells. CB1 and GPR55 receptor transcripts were detected in HCV29 and UROtsa cells, respectively. Immunofluorescence showed positive staining for CB1 in the HCV29 cells. Both cell lines expressed transcript levels for muscarinic receptors, but carbachol did not raise [Ca(2+)]i levels indicating a lack or low expression of G(q)-coupled muscarinic receptors. Transcripts for purinergic receptors were detected; ATP significantly increased [Ca(2+)]i in HCV29 and UROtsa cells by 395 ± 61 and 705 ± 100 nM (mean ± SEM, n = 6), respectively. ACEA did not alter ATP-induced [Ca(2+)]i or cAMP levels in HCV29 cells. Whilst HCV29 cells expressed CB1 and UROtsa cells expressed GPR55 receptors, these were not functionally coupled to the existing purinergic-driven increase in Ca2+ as such they do not represent a good model to study signalling interactions.


Assuntos
Canabinoides/metabolismo , Receptor CB1 de Canabinoide/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Bexiga Urinária/metabolismo , Urotélio/metabolismo , Linhagem Celular , Humanos , Ligação Proteica/fisiologia , Receptores de Canabinoides , Bexiga Urinária/citologia , Urotélio/citologia
15.
PLoS One ; 8(9): e75346, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24058676

RESUMO

Persistent infection of the mouse central nervous system (CNS) with mouse hepatitis virus (MHV) induces a demyelinating disease pathologically similar to multiple sclerosis and is therefore used as a model system. There is little information regarding the host factors that correlate with and contribute to MHV-induced demyelination. Here, we detail the genes and pathways associated with MHV-induced demyelinating disease in the spinal cord. High-throughput sequencing of the host transcriptome revealed that demyelination is accompanied by numerous transcriptional changes indicative of immune infiltration as well as changes in the cytokine milieu and lipid metabolism. We found evidence that a Th1-biased cytokine/chemokine response and eicosanoid-derived inflammation accompany persistent MHV infection and that antigen presentation is ongoing. Interestingly, increased expression of genes involved in lipid transport, processing, and catabolism, including some with known roles in neurodegenerative diseases, coincided with demyelination. Lastly, expression of several genes involved in osteoclast or bone-resident macrophage function, most notably TREM2 and DAP12, was upregulated in persistently infected mouse spinal cord. This study highlights the complexity of the host antiviral response, which accompany MHV-induced demyelination, and further supports previous findings that MHV-induced demyelination is immune-mediated. Interestingly, these data suggest a parallel between bone reabsorption by osteoclasts and myelin debris clearance by microglia in the bone and the CNS, respectively. To our knowledge, this is the first report of using an RNA-seq approach to study the host CNS response to persistent viral infection.


Assuntos
Infecções por Coronavirus/metabolismo , Doenças Desmielinizantes/metabolismo , Vírus da Hepatite Murina/metabolismo , Medula Espinal/metabolismo , Transcriptoma , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Animais , Infecções por Coronavirus/patologia , Citocinas/biossíntese , Citocinas/metabolismo , Doenças Desmielinizantes/patologia , Doenças Desmielinizantes/virologia , Feminino , Masculino , Glicoproteínas de Membrana/biossíntese , Camundongos , Proteínas do Tecido Nervoso/biossíntese , Receptores Imunológicos/biossíntese , Medula Espinal/patologia , Células Th1/metabolismo , Células Th1/patologia
16.
Proc Natl Acad Sci U S A ; 110(32): 13114-9, 2013 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-23878220

RESUMO

Efficient and productive virus infection often requires viral countermeasures that block innate immunity. The IFN-inducible 2',5'-oligoadenylate (2-5A) synthetases (OASs) and ribonuclease (RNase) L are components of a potent host antiviral pathway. We previously showed that murine coronavirus (MHV) accessory protein ns2, a 2H phosphoesterase superfamily member, is a phosphodiesterase (PDE) that cleaves 2-5A, thereby preventing activation of RNase L. The PDE activity of ns2 is required for MHV replication in macrophages and for hepatitis. Here, we show that group A rotavirus (RVA), an important cause of acute gastroenteritis in children worldwide, encodes a similar PDE. The RVA PDE forms the carboxy-terminal domain of the minor core protein VP3 (VP3-CTD) and shares sequence and predicted structural homology with ns2, including two catalytic HxT/S motifs. Bacterially expressed VP3-CTD exhibited 2',5'-PDE activity, which cleaved 2-5A in vitro. In addition, VP3-CTD expressed transiently in mammalian cells depleted 2-5A levels induced by OAS activation with poly(rI):poly(rC), preventing RNase L activation. In the context of recombinant chimeric MHV expressing inactive ns2, VP3-CTD restored the ability of the virus to replicate efficiently in macrophages or in the livers of infected mice, whereas mutant viruses expressing inactive VP3-CTD (H718A or H798R) were attenuated. In addition, chimeric viruses expressing either active ns2 or VP3-CTD, but not nonfunctional equivalents, were able to protect ribosomal RNA from RNase L-mediated degradation. Thus, VP3-CTD is a 2',5'-PDE able to functionally substitute for ns2 in MHV infection. Remarkably, therefore, two disparate RNA viruses encode proteins with homologous 2',5'-PDEs that antagonize activation of innate immunity.


Assuntos
Imunidade Inata/imunologia , Diester Fosfórico Hidrolases/imunologia , Vírus de RNA/imunologia , Proteínas não Estruturais Virais/imunologia , 2',5'-Oligoadenilato Sintetase/imunologia , 2',5'-Oligoadenilato Sintetase/metabolismo , Nucleotídeos de Adenina/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação/genética , Sítios de Ligação/imunologia , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/imunologia , Proteínas do Capsídeo/metabolismo , Linhagem Celular , Células Cultivadas , Endorribonucleases/genética , Endorribonucleases/imunologia , Endorribonucleases/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Humanos , Immunoblotting , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/virologia , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Vírus da Hepatite Murina/imunologia , Vírus da Hepatite Murina/metabolismo , Vírus da Hepatite Murina/fisiologia , Mutação , Oligorribonucleotídeos/metabolismo , Diester Fosfórico Hidrolases/genética , Diester Fosfórico Hidrolases/metabolismo , Infecções por Vírus de RNA/imunologia , Infecções por Vírus de RNA/virologia , Vírus de RNA/metabolismo , Vírus de RNA/fisiologia , Rotavirus/imunologia , Rotavirus/metabolismo , Rotavirus/fisiologia , Homologia de Sequência de Aminoácidos , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo
17.
J Virol ; 87(15): 8408-18, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23698313

RESUMO

Previous studies have demonstrated that the murine coronavirus mouse hepatitis virus (MHV) nonstructural protein 2 (ns2) is a 2',5'-phosphodiesterase that inhibits activation of the interferon-induced oligoadenylate synthetase (OAS)-RNase L pathway. Enzymatically active ns2 is required for efficient MHV replication in macrophages, as well as for the induction of hepatitis in C57BL/6 mice. In contrast, following intranasal or intracranial inoculation, efficient replication of MHV in the brain is not dependent on an enzymatically active ns2. The replication of wild-type MHV strain A59 (A59) and a mutant with an inactive phosphodiesterase (ns2-H126R) was assessed in primary hepatocytes and primary central nervous system (CNS) cell types-neurons, astrocytes, and oligodendrocytes. A59 and ns2-H126R replicated with similar kinetics in all cell types tested, except macrophages and microglia. RNase L activity, as assessed by rRNA cleavage, was induced by ns2-H126R, but not by A59, and only in macrophages and microglia. Activation of RNase L correlated with the induction of type I interferon and the consequent high levels of OAS mRNA induced in these cell types. Pretreatment of nonmyeloid cells with interferon restricted A59 and ns2-H126R to the same extent and failed to activate RNase L following infection, despite induction of OAS expression. However, rRNA degradation was induced by treatment of astrocytes or oligodendrocytes with poly(I·C). Thus, RNase L activation during MHV infection is cell type specific and correlates with relatively high levels of expression of OAS genes, which are necessary but not sufficient for induction of an effective RNase L antiviral response.


Assuntos
2',5'-Oligoadenilato Sintetase/metabolismo , Endorribonucleases/metabolismo , Interações Hospedeiro-Patógeno , Vírus da Hepatite Murina/enzimologia , Vírus da Hepatite Murina/fisiologia , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Animais , Células Cultivadas , Deleção de Genes , Camundongos , Camundongos Endogâmicos C57BL , Vírus da Hepatite Murina/genética , Vírus da Hepatite Murina/imunologia , Proteínas não Estruturais Virais/genética
18.
Int Urogynecol J ; 24(5): 855-63, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23081739

RESUMO

INTRODUCTION AND HYPOTHESIS: Our aim was to compare expression and distribution of cannabinoid receptors CB1 and CB2, transient receptor potential vanilloid receptor 1 (TRPV1), and modulating enzymes in human and rat bladder. We also evaluated effects of cannabinoid agonists (ACEA, agonist of CB1; GP1A, agonist of CB2) on contractile responses of rat bladder strips. METHODS: Distribution and expression of CB1, CB2 and TRPV1 receptors and enzymes fatty acid amide hydrolase (FAAH) and N-acyl phosphatidylethanolamine-hydrolyzing phospholipase D (NAPE-PLD) was studied using immunohistochemistry and immunoblotting on human and Wistar rat bladders. The effects of cannabinoid agonists on contractile responses of isolated rat bladder strips to electrical-field stimulation (EFS) or carbachol-evoked responses were determined. RESULTS: Immunoreactivity for CB1 and TRPV1 receptors and FAAH and NAPE-PLD was present in the bladder of both species. CB1 proteins were of different sizes in rat (57 kDa) and human (40 kDa) bladder. CB2 (45 kDa in both species) immunolocalised to both urothelium and detrusor muscle in human bladder but only to detrusor muscle in rat. FAAH proteins were found at 55 kDa for both species. Rat NAPE-PLD protein (44 kDa) was similar in size to that in human bladder (45 kDa). TRPV1 proteins were found at 104 kDa in both species. ACEA (10(-4) M) attenuated bladder contractions by 35 ± 5.4 % (p < 0.001); GP1a had no effect despite the EC50 values for the carbachol dose-response curves for both agonists being significantly shifted to the right. CONCLUSIONS: The endocannabinoid system is functionally expressed in both species, with CB1 receptors showing both pre- and postsynaptic inhibitory effects on rat bladder contraction, whereas CB2 acts only postsynaptically.


Assuntos
Endocanabinoides/metabolismo , Receptor CB1 de Canabinoide/metabolismo , Receptor CB2 de Canabinoide/metabolismo , Bexiga Urinária/metabolismo , Idoso , Animais , Células CHO , Cricetinae , Cricetulus , Estimulação Elétrica , Endocanabinoides/fisiologia , Feminino , Humanos , Pessoa de Meia-Idade , Ratos , Ratos Wistar , Receptor CB1 de Canabinoide/agonistas , Receptor CB2 de Canabinoide/agonistas , Canais de Cátion TRPV/metabolismo , Bexiga Urinária/fisiologia
19.
Cell Host Microbe ; 11(6): 607-16, 2012 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-22704621

RESUMO

Many viruses induce hepatitis in humans, highlighting the need to understand the underlying mechanisms of virus-induced liver pathology. The murine coronavirus, mouse hepatitis virus (MHV), causes acute hepatitis in its natural host and provides a useful model for understanding virus interaction with liver cells. The MHV accessory protein, ns2, antagonizes the type I interferon response and promotes hepatitis. We show that ns2 has 2',5'-phosphodiesterase activity, which blocks the interferon inducible 2',5'-oligoadenylate synthetase (OAS)-RNase L pathway to facilitate hepatitis development. Ns2 cleaves 2',5'-oligoadenylate, the product of OAS, to prevent activation of the cellular endoribonuclease RNase L and consequently block viral RNA degradation. An ns2 mutant virus was unable to replicate in the liver or induce hepatitis in wild-type mice, but was highly pathogenic in RNase L deficient mice. Thus, RNase L is a critical cellular factor for protection against viral infection of the liver and the resulting hepatitis.


Assuntos
2',5'-Oligoadenilato Sintetase/antagonistas & inibidores , Endorribonucleases/antagonistas & inibidores , Fígado/virologia , Vírus da Hepatite Murina/patogenicidade , Proteínas não Estruturais Virais/metabolismo , Fatores de Virulência/metabolismo , Replicação Viral , Nucleotídeos de Adenina/metabolismo , Animais , Interferons/imunologia , Fígado/patologia , Camundongos , Vírus da Hepatite Murina/fisiologia , Oligorribonucleotídeos/metabolismo , Estabilidade de RNA , RNA Viral/metabolismo
20.
J Urol ; 186(6): 2455-62, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22019168

RESUMO

PURPOSE: Artificial sweeteners augment bladder contraction. We hypothesized that artificial sweeteners activate sweet taste receptors in the bladder. Thus, we investigated the expression of sweet taste receptors in human and rat bladders. MATERIALS AND METHODS: Sections of human and rat bladders were cut from paraffin blocks and stained by immunohistochemistry for the expression of T1R2/3 sweet taste receptors. Bladder homogenates were subjected to sodium dodecyl sulfate-polyacrylamide electrophoresis, followed by immunoblotting for T1R2/3 receptor expression. Rat bladder strips with and without urothelium were suspended in organ baths. The contractile response to 10 Hz electrical field stimulation was determined in the absence and presence of saccharin (Sigma-Aldrich®) (10(-8) to 10(-3) M). Responses to KCl in the absence and presence of saccharin, and saccharin plus zinc were also determined. RESULTS: T1R2/3 sweet taste receptors were expressed in human and rat bladder urothelium. Immunostaining was evident in the plasma membrane of the 3 urothelial cell types, particularly umbrella cells. Immunoblotting revealed bands at expected molecular weights in human and rat bladder homogenates. Saccharin augmented rat bladder smooth muscle contraction due to electrical field stimulation only when urothelium was present in the bladder strip. Zinc blocked the enhancing effect of saccharin on responses to KCl. CONCLUSIONS: T1R2/3 sweet taste receptors are expressed in human and rat bladder urothelium. Activation of these receptors by artificial sweeteners may result in augmented bladder contraction.


Assuntos
Receptores Acoplados a Proteínas G/análise , Receptores Acoplados a Proteínas G/biossíntese , Bexiga Urinária/química , Bexiga Urinária/metabolismo , Animais , Feminino , Humanos , Técnicas In Vitro , Isoformas de Proteínas , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...