Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Breast Cancer Res ; 22(1): 89, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32795346

ABSTRACT

BACKGROUND: Combined targeting of CDK4/6 and ER is now the standard of care for patients with advanced ER+/HER2- breast cancer. However, acquired resistance to these therapies frequently leads to disease progression. As such, it is critical to identify the mechanisms by which resistance to CDK4/6-based therapies is acquired and also identify therapeutic strategies to overcome resistance. METHODS: In this study, we developed and characterized multiple in vitro and in vivo models of acquired resistance to CDK4/6-based therapies. Resistant models were screened by reverse phase protein array (RPPA) for cell signaling changes that are activated in resistance. RESULTS: We show that either a direct loss of Rb or loss of dependence on Rb signaling confers cross-resistance to inhibitors of CDK4/6, while PI3K/mTOR signaling remains activated. Treatment with the p110α-selective PI3K inhibitor, alpelisib (BYL719), completely blocked the progression of acquired CDK4/6 inhibitor-resistant xenografts in the absence of continued CDK4/6 inhibitor treatment in models of both PIK3CA mutant and wild-type ER+/HER2- breast cancer. Triple combination therapy against PI3K:CDK4/6:ER prevented and/or delayed the onset of resistance in treatment-naive ER+/HER2- breast cancer models. CONCLUSIONS: These data support the clinical investigation of p110α-selective inhibitors of PI3K, such as alpelisib, in patients with ER+/HER2- breast cancer who have progressed on CDK4/6:ER-based therapies. Our data also support the investigation of PI3K:CDK4/6:ER triple combination therapy to prevent the onset of resistance to the combination of endocrine therapy plus CDK4/6 inhibition.


Subject(s)
Breast Neoplasms/drug therapy , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Drug Resistance, Neoplasm , Estrogen Receptor alpha/antagonists & inhibitors , Phosphatidylinositol 3-Kinases/chemistry , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Evaluation, Preclinical , Female , Humans , Mice, Nude , Molecular Targeted Therapy , Phosphatidylinositol 3-Kinases/metabolism , Pregnancy , Protein Kinase Inhibitors/pharmacology , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays
2.
J Nurs Educ ; 59(8): 461-464, 2020 Aug 01.
Article in English | MEDLINE | ID: mdl-32757011

ABSTRACT

BACKGROUND: The purpose of our web-based, semi-virtual reality simulation was to raise baccalaureate nursing students' awareness of civility and incivility. Educators know civility in nursing is crucial to safe and effective patient care. However, nursing students have reported physiological and psychological distress after experiencing incivility. If nurse educators are unable to better promote civility, the quality of nursing education, and ultimately nursing practice, is threatened. METHOD: We implemented an innovative civility-related, semi-virtual reality, simulation with senior-level nursing students. After attending a faculty-led discussion on the importance of civility and incivility, students participated in a web-based, semi-virtual reality simulation, followed by a synchronous debriefing session. RESULTS: Students reported that the intervention raised their awareness of civility and incivility. CONCLUSION: This article describes the intervention and shares the lessons learned in implementing it so nurse educators may replicate this innovative strategy to raise awareness of civility in nursing education. [J Nurs Educ. 2020;59(8):461-464.].


Subject(s)
Education, Nursing , Incivility , Simulation Training , Virtual Reality , Education, Nursing/methods , Faculty, Nursing , Humans , Incivility/prevention & control , Students, Nursing
3.
Nurse Educ ; 45(3): 165-168, 2020.
Article in English | MEDLINE | ID: mdl-31335610

ABSTRACT

BACKGROUND: Incivility is a low-intensity, discourteous behavior intended to disrupt or harm positive interaction. If allowed, student-to-student incivility can undermine the educational environment. PURPOSE: The purpose of the integrative review was to examine factors influencing incivility among nursing students and teaching strategies used to reduce incivility in nursing education. METHODS: Qualitative and quantitative studies were reviewed. The Johns Hopkins Research Evidence Appraisal tool was used to narrow down the selection of articles. Content analysis was used to evaluate the qualitative research. RESULTS: Five major points of interest were identified: workload and high expectations contributed to incivility, degrees of incivility, effects of incivility, coping mechanisms among individuals, and effective teaching strategies addressing incivility. CONCLUSION: Continued research on innovative teaching strategies that raise awareness of civility while reducing incivility is warranted.


Subject(s)
Curriculum , Education, Nursing/organization & administration , Faculty, Nursing/psychology , Incivility/prevention & control , Students, Nursing/psychology , Adult , Female , Humans , Male , Middle Aged , Nursing Education Research , Qualitative Research , United States , Young Adult
4.
J Virol ; 90(6): 3160-72, 2016 Jan 06.
Article in English | MEDLINE | ID: mdl-26739051

ABSTRACT

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.


Subject(s)
2',5'-Oligoadenylate Synthetase/metabolism , Endoribonucleases/biosynthesis , Gene Expression , Host-Pathogen Interactions , Murine hepatitis virus/physiology , Myeloid Cells/enzymology , Myeloid Cells/virology , Animals , Cells, Cultured , Mice , Mice, Knockout
5.
J Virol ; 89(24): 12330-40, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26423942

ABSTRACT

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.


Subject(s)
Coronavirus Infections/immunology , DEAD-box RNA Helicases/immunology , Murine hepatitis virus/immunology , Signal Transduction/immunology , Animals , Cell Line , Coronavirus Infections/genetics , DEAD-box RNA Helicases/genetics , Humans , Interferon Type I/genetics , Interferon Type I/immunology , Interferon-Induced Helicase, IFIH1 , Interleukin-6/genetics , Interleukin-6/immunology , Mice , Mice, Knockout , Murine hepatitis virus/genetics , Signal Transduction/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
6.
Clin Cancer Res ; 21(3): 585-93, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25473003

ABSTRACT

PURPOSE: Distinct molecular subgroups of medulloblastoma, including hedgehog (Hh) pathway-activated disease, have been reported. We identified and clinically validated a five-gene Hh signature assay that can be used to preselect patients with Hh pathway-activated medulloblastoma. EXPERIMENTAL DESIGN: Gene characteristics of the Hh medulloblastoma subgroup were identified through published bioinformatic analyses. Thirty-two genes shown to be differentially expressed in fresh-frozen and formalin-fixed paraffin-embedded tumor samples and reproducibly analyzed by RT-PCR were measured in matched samples. These data formed the basis for building a multi-gene logistic regression model derived through elastic net methods from which the five-gene Hh signature emerged after multiple iterations. On the basis of signature gene expression levels, the model computed a propensity score to determine Hh activation using a threshold set a priori. The association between Hh activation status and tumor response to the Hh pathway inhibitor sonidegib (LDE225) was analyzed. RESULTS: Five differentially expressed genes in medulloblastoma (GLI1, SPHK1, SHROOM2, PDLIM3, and OTX2) were found to associate with Hh pathway activation status. In an independent validation study, Hh activation status of 25 medulloblastoma samples showed 100% concordance between the five-gene signature and Affymetrix profiling. Further, in medulloblastoma samples from 50 patients treated with sonidegib, all 6 patients who responded were found to have Hh-activated tumors. Three patients with Hh-activated tumors had stable or progressive disease. No patients with Hh-nonactivated tumors responded. CONCLUSIONS: This five-gene Hh signature can robustly identify Hh-activated medulloblastoma and may be used to preselect patients who might benefit from sonidegib treatment.


Subject(s)
Cerebellar Neoplasms/genetics , Hedgehog Proteins/antagonists & inhibitors , Medulloblastoma/genetics , Patient Selection , Transcriptome , Adolescent , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biphenyl Compounds/pharmacology , Biphenyl Compounds/therapeutic use , Cerebellar Neoplasms/drug therapy , Cerebellar Neoplasms/metabolism , Child , Child, Preschool , Computational Biology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Hedgehog Proteins/metabolism , Humans , Infant , Medulloblastoma/drug therapy , Medulloblastoma/metabolism , Models, Biological , Prognosis , Pyridines/pharmacology , Pyridines/therapeutic use , Reproducibility of Results , Signal Transduction/drug effects , Treatment Outcome
7.
Clin Cancer Res ; 20(7): 1900-9, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24523439

ABSTRACT

PURPOSE: This phase I trial was undertaken to determine the maximum tolerated dose (MTD), dose-limiting toxicities (DLT), safety, tolerability, pharmacokinetics, pharmacodynamics, and preliminary antitumor activity of the novel smoothened inhibitor sonidegib (LDE225), a potent inhibitor of hedgehog signaling, in patients with advanced solid tumors. EXPERIMENTAL DESIGN: Oral sonidegib was administered to 103 patients with advanced solid tumors, including medulloblastoma and basal cell carcinoma (BCC), at doses ranging from 100 to 3,000 mg daily and 250 to 750 mg twice daily, continuously, with a single-dose pharmacokinetics run-in period. Dose escalations were guided by a Bayesian logistic regression model. Safety, tolerability, efficacy, pharmacokinetics, and biomarkers in skin and tumor biopsies were assessed. RESULTS: The MTDs of sonidegib were 800 mg daily and 250 mg twice daily. The main DLT of reversible grade 3/4 elevated serum creatine kinase (18% of patients) was observed at doses ≥ the MTD in an exposure-dependent manner. Common grade 1/2 adverse events included muscle spasm, myalgia, gastrointestinal toxicities, increased liver enzymes, fatigue, dysgeusia, and alopecia. Sonidegib exposure increased dose proportionally up to 400 mg daily, and displayed nonlinear pharmacokinetics at higher doses. Sonidegib exhibited exposure-dependent reduction in GLI1 mRNA expression. Tumor responses observed in patients with medulloblastoma and BCC were associated with evidence of hedgehog pathway activation. CONCLUSIONS: Sonidegib has an acceptable safety profile in patients with advanced solid tumors and exhibits antitumor activity in advanced BCC and relapsed medulloblastoma, both of which are strongly associated with activated hedgehog pathway, as determined by gene expression.


Subject(s)
Biphenyl Compounds/administration & dosage , Carcinoma, Basal Cell/drug therapy , Medulloblastoma/drug therapy , Pyridines/administration & dosage , Receptors, G-Protein-Coupled/antagonists & inhibitors , Administration, Oral , Adult , Aged , Aged, 80 and over , Biphenyl Compounds/adverse effects , Biphenyl Compounds/pharmacokinetics , Carcinoma, Basal Cell/genetics , Carcinoma, Basal Cell/pathology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Maximum Tolerated Dose , Medulloblastoma/genetics , Medulloblastoma/pathology , Middle Aged , Neoplasm Staging , Pyridines/adverse effects , Pyridines/pharmacokinetics , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Smoothened Receptor , Transcription Factors/biosynthesis , Zinc Finger Protein GLI1
8.
J Virol ; 87(15): 8408-18, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23698313

ABSTRACT

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.


Subject(s)
2',5'-Oligoadenylate Synthetase/metabolism , Endoribonucleases/metabolism , Host-Pathogen Interactions , Murine hepatitis virus/enzymology , Murine hepatitis virus/physiology , Viral Nonstructural Proteins/metabolism , Virus Replication , Animals , Cells, Cultured , Gene Deletion , Mice , Mice, Inbred C57BL , Murine hepatitis virus/genetics , Murine hepatitis virus/immunology , Viral Nonstructural Proteins/genetics
9.
J Virol ; 85(19): 10058-68, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21752905

ABSTRACT

Previous studies have demonstrated that mouse hepatitis virus (MHV) hepatotropism is determined largely by postentry events rather than by availability of the viral receptor. In addition, mutation of MHV nonstructural protein 2 (ns2) abrogates the ability of the virus to replicate in the liver and induce hepatitis but does not affect replication in the central nervous system (CNS). Here we show that replication of ns2 mutant viruses is attenuated in bone marrow-derived macrophages (BMM) generated from wild-type (wt) mice but not in L2 fibroblasts, primary astrocytes, or BMM generated from type I interferon receptor-deficient (IFNAR(-/-)) mice. In addition, ns2 mutants are more sensitive than wt virus to pretreatment of BMM, but not L2 fibroblasts or primary astrocytes, with alpha/beta interferon (IFN-α/ß). The ns2 mutants induced similar levels of IFN-α/ß in wt and IFNAR(-/-) BMM, indicating that ns2 expression has no effect on the induction of IFN but rather that it antagonizes a later step in IFN signaling. Consistent with these in vitro data, the virulence of ns2 mutants increased to near that of wt virus after depletion of macrophages in vivo. These data imply that the ability of MHV to replicate in macrophages is a prerequisite for replication in the liver and induction of hepatitis but not for replication or disease in the CNS, underscoring the importance of IFN signaling in macrophages in vivo for protection of the host from hepatitis. Our results further support the notion that viral tissue tropism is determined in part by postentry events, including the early type I interferon response.


Subject(s)
Interferon Type I/immunology , Murine hepatitis virus/immunology , Murine hepatitis virus/physiology , Viral Tropism , Animals , Cells, Cultured , Coronavirus Infections/pathology , Coronavirus Infections/virology , Fibroblasts/virology , Hepatitis, Viral, Animal/pathology , Hepatitis, Viral, Animal/virology , Leukocyte Reduction Procedures , Macrophages/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Interferon alpha-beta/deficiency , Rodent Diseases/pathology , Rodent Diseases/virology , Viral Nonstructural Proteins/deficiency
10.
J Invest Dermatol ; 131(8): 1735-44, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21430703

ABSTRACT

Basal cell carcinoma (BCC) is a distinctive manifestation in nevoid basal cell carcinoma syndrome (NBCCS) patients. Both inherited and acquired mutations of patched 1 (PTCH1), a tumor-suppressor gene controlling the activity of Smoothened (SMO), are the primary cause of the constitutive activation of the Hedgehog (HH) pathway, leading to the emergence of BCCs in NBCCS. LDE225, a distinct, selective antagonist of SMO, showed potent inhibition of basaloid tumor nest formation and mediated regression of preformed basaloid tumors in organ cultures of skin derived from Ptch1 heterozygous knockout mice. In a double-blind, randomized, vehicle-controlled, intraindividual study, a total of 8 NBCCS patients presenting 27 BCCs were treated twice daily with 0.75% LDE225 cream or vehicle for 4 weeks. Application of 0.75% LDE225 cream was well tolerated and showed no skin irritation. Of 13 LDE225-treated BCCs, 3 showed a complete, 9 a partial, and only 1 no clinical response. Except for one partial response, the vehicle produced no clinical response in any of the 14 treated BCCs. Treatment with 0.75% LDE225 cream in NBCCS patients was very well tolerated and caused BCC regression, thus potentially offering an attractive therapeutic alternative to currently available therapies for this indication.JID JOURNAL CLUB ARTICLE: For questions, answers, and open discussion about this article, please go to http://www.nature.com/jid/journalclub.


Subject(s)
Antineoplastic Agents/administration & dosage , Biphenyl Compounds/administration & dosage , Carcinoma, Basal Cell/drug therapy , Pyridines/administration & dosage , Receptors, G-Protein-Coupled/antagonists & inhibitors , Signal Transduction/drug effects , Skin Neoplasms/drug therapy , Administration, Topical , Animals , Antineoplastic Agents/adverse effects , Biphenyl Compounds/adverse effects , Carcinoma, Basal Cell/pathology , Female , Hair/drug effects , Hair/growth & development , Hedgehog Proteins/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques , Patched Receptors , Patched-1 Receptor , Pregnancy , Pyridines/adverse effects , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , Skin Neoplasms/pathology , Smoothened Receptor
11.
Sci Transl Med ; 2(52): 52ra72, 2010 Oct 06.
Article in English | MEDLINE | ID: mdl-20926833

ABSTRACT

Interleukin-17A (IL-17A) is elaborated by the T helper 17 (T(H)17) subset of T(H) cells and exhibits potent proinflammatory properties in animal models of autoimmunity, including collagen-induced arthritis, experimental autoimmune encephalomyelitis, and experimental autoimmune uveitis. To determine whether IL-17A mediates human inflammatory diseases, we investigated the efficacy and safety of AIN457, a human antibody to IL-17A, in patients with psoriasis, rheumatoid arthritis, and chronic noninfectious uveitis. Patients with chronic plaque-type psoriasis (n = 36), rheumatoid arthritis (n = 52), or chronic noninfectious uveitis (n = 16) were enrolled in clinical trials to evaluate the effects of neutralizing IL-17A by AIN457 at doses of 3 to 10 mg/kg, given intravenously. We evaluated efficacy by measuring the psoriasis area and severity index (PASI), the American College of Rheumatology 20% response (ACR20) for rheumatoid arthritis, or the number of responders for uveitis, as defined by either vision improvement or reduction in ocular inflammation or corticosteroid dose. AIN457 treatment induced clinically relevant responses of variable magnitude in patients suffering from each of these diverse immune-mediated diseases. Variable response rates may be due to heterogeneity in small patient populations, differential pathogenic roles of IL-17A in these diseases, and the different involvement or activation of IL-17A-producing cells. The rates of adverse events, including infections, were similar in the AIN457 and placebo groups. These results support a role for IL-17A in the pathophysiology of diverse inflammatory diseases including psoriasis, rheumatoid arthritis, and noninfectious uveitis.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies , Arthritis, Rheumatoid/drug therapy , Interleukin-17/immunology , Psoriasis/drug therapy , Uveitis/drug therapy , Adolescent , Adult , Aged , Animals , Antibodies/immunology , Antibodies/therapeutic use , Antibodies, Monoclonal, Humanized , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Double-Blind Method , Humans , Middle Aged , Placebos/therapeutic use , Psoriasis/immunology , Psoriasis/pathology , Treatment Outcome , Uveitis/immunology , Uveitis/pathology , Young Adult
12.
J Virol ; 84(11): 5656-69, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20357099

ABSTRACT

The importance of the type I interferon (IFN-I) system in limiting coronavirus replication and dissemination has been unequivocally demonstrated by rapid lethality following infection of mice lacking the alpha/beta IFN (IFN-alpha/beta) receptor with mouse hepatitis virus (MHV), a murine coronavirus. Interestingly, MHV has a cell-type-dependent ability to resist the antiviral effects of IFN-alpha/beta. In primary bone-marrow-derived macrophages and mouse embryonic fibroblasts, MHV replication was significantly reduced by the IFN-alpha/beta-induced antiviral state, whereas IFN treatment of cell lines (L2 and 293T) has only minor effects on replication (K. M. Rose and S. R. Weiss, Viruses 1:689-712, 2009). Replication of other RNA viruses, including Theiler's murine encephalitis virus (TMEV), vesicular stomatitis virus (VSV), Sindbis virus, Newcastle disease virus (NDV), and Sendai virus (SeV), was significantly inhibited in L2 cells treated with IFN-alpha/beta, and MHV had the ability to rescue only SeV replication. We present evidence that MHV infection can delay interferon-stimulated gene (ISG) induction mediated by both SeV and IFN-beta but only when MHV infection precedes SeV or IFN-beta exposure. Curiously, we observed no block in the well-defined IFN-beta signaling pathway that leads to STAT1-STAT2 phosphorylation and translocation to the nucleus in cultures infected with MHV. This observation suggests that MHV must inhibit an alternative IFN-induced pathway that is essential for early induction of ISGs. The ability of MHV to delay SeV-mediated ISG production may partially involve limiting the ability of IFN regulatory factor 3 (IRF-3) to function as a transcription factor. Transcription from an IRF-3-responsive promoter was partially inhibited by MHV; however, IRF-3 was transported to the nucleus and bound DNA in MHV-infected cells superinfected with SeV.


Subject(s)
Coronavirus Infections/immunology , Coronavirus/physiology , Gene Expression Regulation , Interferon Regulatory Factor-3/metabolism , Interferon Type I/pharmacology , Active Transport, Cell Nucleus , Animals , DNA/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Interferon-beta/pharmacology , Mice , Protein Transport , Sendai virus , Transcription, Genetic
13.
Am J Ther ; 17(6): 543-52, 2010.
Article in English | MEDLINE | ID: mdl-19797939

ABSTRACT

A meta-analysis of 29 clinical studies on tegaserod revealed an imbalance of cardiovascular ischemic events in patients treated with drug versus placebo. Because increased platelet activity is known to attribute to cardiovascular events, we examined the presence of serotonin receptor type 4 (5-HT4) receptors and the effects of tegaserod on in vitro aggregation of human platelets. Blood samples were obtained from 20 healthy volunteers and 20 subjects with irritable bowel syndrome with constipation. Samples of whole blood-citrate mixtures were incubated with different tegaserod concentrations mimicking human Cmax values (10 nM), 3.3 times, and 10 times Cmax for at least 1 hour. Conventional plasma platelet aggregation was induced by adenosine diphosphate, collagen, thrombin receptor activating peptide, epinephrine, and serotonin plus adenosine diphosphate. Gene expression analyses targeting 5-HT4 and serotonin receptor type 2 receptors were carried out using human platelet RNA. The presence of 5-HT4 receptor protein was investigated by Western blot analysis using membrane fractions from human platelets. Preincubation with tegaserod resulted in mild but statistically significant increases in platelet aggregation induced by adenosine diphosphate, collagen, epinephrine, or serotonin, mostly at supratherapeutic concentrations of tegaserod. The effects were comparable using thrombocytes obtained from healthy volunteers and patients with irritable bowel syndrome with constipation. Expression analysis revealed that mRNA encoding both 5-HT4 and serotonin receptor type 2 receptors was present in human platelets. The expression of 5-HT4 receptor mRNA was approximately eightfold lower than serotonin receptor type 2 receptor mRNA. Results from Western blot analyses examining the presence of 5-HT4 receptor protein in human platelets were in agreement with the findings of the mRNA expression analysis. In platelets harvested from normal persons and patients with irritable bowel syndrome with constipation and exposed in vitro to tegaserod, we detected small but statistically significant concentration-dependent increases in induced platelet aggregation. The relationship of these in vitro effects to clinical cardiovascular ischemic events is presently unclear. Western blot analysis findings suggest the presence of 5-HT4 receptor protein on human platelets. Further investigations on the potential role of 5-HT4 receptors in human platelets may be warranted.


Subject(s)
Blood Platelets/drug effects , Indoles/pharmacology , Platelet Aggregation/drug effects , Receptors, Serotonin, 5-HT4/biosynthesis , Serotonin 5-HT4 Receptor Agonists/pharmacology , Adenosine Diphosphate/pharmacology , Adult , Aged , Blood Platelets/metabolism , Dose-Response Relationship, Drug , Epinephrine/pharmacology , Female , Humans , In Vitro Techniques , Male , Middle Aged , RNA, Messenger/biosynthesis , Receptors, Serotonin, 5-HT2/biosynthesis , Receptors, Serotonin, 5-HT2/genetics , Receptors, Serotonin, 5-HT4/genetics , Serotonin/pharmacology
14.
Viruses ; 1(3): 689-712, 2009 12 01.
Article in English | MEDLINE | ID: mdl-20221421

ABSTRACT

Coronaviruses infect many species of animal including humans, causing acute and chronic diseases of many organ systems. Murine coronavirus, mouse hepatitis virus (MHV) infection of the mouse, provides animal models for the study of central nervous system disease, including encephalitis and demyelinating diseases such as Multiple Sclerosis and for hepatitis. While there are many studies of the adaptive immune response to MHV, there has until recently been scant information on the type I interferon (IFN) response to MHV. The relationship between MHV and the IFN-alpha/beta response is paradoxical. While the type I IFN response is a crucial aspect of host defense against MHV in its natural host, there is little if any induction of IFN following infection of mouse fibroblast cell lines in vitro. Furthermore, MHV is relatively resistant to the antiviral effects of IFN-alpha/beta in mouse fibroblast cell lines and in human 293T cells. MHV can, under some circumstances, compromise the antiviral effects of IFN signaling. The nucleocapsid protein as well as the nsp1 and nsp3 proteins of MHV has been reported to have IFN antagonist activity. However, in primary cell types such as plasmacytoid dendritic cells (pDC) and macrophages, IFN is induced by MHV infection and an antiviral state is established. Other primary cell types such as neurons, astrocytes and hepatocytes fail to produce IFN following infection and, in vivo, likely depend on IFN produced by pDCs and macrophages for protection from MHV. Thus MHV induction of IFN-alpha/beta and the ability to induce an antiviral state in response to interferon is extremely cell type dependent. IFN induced protection from MHV pathogenesis likely requires the orchestrated activities of several cell types, however, the cell types involved in limiting MHV replication may be different in the liver and in the immune privileged CNS.

15.
J Virol ; 82(2): 987-98, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17977970

ABSTRACT

Vif(IIIB), which has been a standard model for the viral infectivity factor of human immunodeficiency virus type 1 (HIV-1), binds the cytidine deaminase APOBEC3G (A3G) and induces its degradation, thereby precluding its lethal incorporation into assembling virions. Additionally, Vif(IIIB) less efficiently degrades A3F, another potent anti-HIV-1 cytidine deaminase. Although the APOBEC3 paralogs A3A, A3B, and A3C have weaker anti-HIV-1 activities and are only partially degraded by Vif(IIIB), we found that Vif(IIIB) induces their emigration from the nucleus to the cytosol and thereby causes net increases in the cytosolic concentrations and anti-HIV-1 activities of A3A and A3B. In contrast, some other Vifs, exemplified by Vif(HXB2) and Vif(ELI-1), much more efficiently degrade and thereby neutralize all APOBEC3s. Studies focused mainly on A3F imply that it occurs associated with mRNA-PABP1 in translationally active polysomes and to a lesser extent in mRNA processing bodies (P-bodies). A3F appears to stabilize the P-bodies with which it is associated. A correspondingly small proportion of Vif(IIIB) also localizes in P-bodies in an A3F-dependent manner. Stress causes A3A, A3B, A3C, and A3F to colocalize efficiently with Vif(IIIB) and mRNA-PABP1 complexes in stress granules in a manner that is prevented by cycloheximide, an inhibitor of translational elongation. Coimmunoprecipitation studies suggest that Vifs from different HIV-1 isolates associate with all tested APOBEC3s. Thus, Vifs interact closely with structurally diverse APOBEC3s, with effects on their subcellular localization, degradation rates, and antiviral activities. Cytosolic APOBEC3-Vif complexes are predominantly bound to mRNAs that dynamically move between translationally active and storage or processing pools.


Subject(s)
Cytidine Deaminase/metabolism , Cytosine Deaminase/metabolism , HIV-1/metabolism , Proteins/metabolism , vif Gene Products, Human Immunodeficiency Virus/metabolism , Cell Line , Humans , Minor Histocompatibility Antigens , Poly(A)-Binding Protein I/metabolism , Protein Binding , Protein Transport , RNA, Messenger/metabolism
16.
J Biol Chem ; 281(39): 29105-19, 2006 Sep 29.
Article in English | MEDLINE | ID: mdl-16887808

ABSTRACT

Deoxycytidine deaminases APOBEC3G (A3G) and APOBEC3F (A3F) (members of the apolipoprotein B mRNA-editing catalytic polypeptide 3 family) have RNA-binding motifs, invade assembling human immunodeficiency virus (HIV-1), and hypermutate reverse transcripts. Antagonistically, HIV-1 viral infectivity factor degrades these enzymes. A3G is enzymatically inhibited by binding RNA within an unidentified large cytosolic ribonucleoprotein, implying that RNA degradation during reverse transcription may activate intravirion A3G at the necessary moment. We purified a biologically active tandem affinity-tagged A3G from human HEK293T cells. Mass spectrometry and coimmunoprecipitation from HEK293T and T lymphocyte extracts identified many RNA-binding proteins specifically associated with A3G and A3F, including poly(A)-binding proteins (PABPs), YB-1, Ro-La, RNA helicases, ribosomal proteins, and Staufen1. Most strikingly, nearly all A3G-associated proteins were known to bind exclusively or intermittently to translating and/or dormant mRNAs. Accordingly, A3G in HEK293T and T lymphocyte extracts was almost completely in A3G-mRNA-PABP complexes that shifted reversibly between polysomes and dormant pools in response to translational inhibitors. For example arsenite, which inhibits 5'-cap-dependent translational initiation, shifted mRNA-A3G-PABP from polysomes into stress granules in a manner that was blocked and reversed by the elongation inhibitor cycloheximide. Immunofluorescence microscopy showed A3G-mRNA-PABP stress granules only partially overlapping with Staufen1. A3G coimmunoprecipitated HIV-1 RNA and many mRNAs. Ribonuclease released nearly all A3G-associated proteins, including A3G homo-oligomers and A3G-A3F hetero-oligomers, but the viral infectivity factor remained bound. Many proteins and RNAs associated with A3G are excluded from A3G-containing virions, implying that A3G competitively partitions into virions based on affinity for HIV-1 RNA.


Subject(s)
HIV-1/metabolism , Nucleoside Deaminases/physiology , Polyribosomes/metabolism , Repressor Proteins/physiology , APOBEC-3G Deaminase , Cell Line , Cytidine Deaminase , Gene Products, vif/metabolism , HeLa Cells , Humans , Nucleoside Deaminases/chemistry , Peptides/chemistry , Protein Binding , RNA, Messenger/metabolism , Repressor Proteins/chemistry , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Transfection , Virus Replication , vif Gene Products, Human Immunodeficiency Virus
17.
AIDS Res Hum Retroviruses ; 21(7): 611-9, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16060832

ABSTRACT

APOBEC3G and 3F (A3G and A3F) cytidine deaminases incorporate into retroviral cores where they lethally hypermutate nascent DNA reverse transcripts. As substantiated here, the viral infectivity factor (Vif) encoded by human immunodeficiency virus type-1 (HIV-1) binds A3G and A3F and induces their degradation, thereby precluding their incorporation into viral progeny. Previous evidence suggested that A3G is expressed in H9 and other nonpermissive cells that contain this antiviral defense but not in several permissive cells, and that overexpression of A3G or A3F makes permissive cells nonpermissive. Using a broader panel of cell lines, we confirmed a correlation between A3G and cellular abilities to inactivate HIV-1(Deltavif). However, there was a quantitative discrepancy because several cells with weak antiviral activities had similar amounts of wild-type A3G mRNA and protein compared to H9 cells. Antiviral activity of H9 cells was also attenuated in some conditions. These quantitative discrepancies could not be explained by the presence of A3F or other A3G paralogs in some of the cell lines. Thus, A3A, A3B, and A3C had weak but significant anti-HIV-1 activities and did not dominantly interfere with A3G or A3F antiviral functions. Control of A3G synthesis by the protein kinase C/mitogen-activated protein kinase kinase/extracellular signal-regulated kinase pathway was also similar in permissive and nonpermissive cells. A3G in highly permissive cells is degraded by Vif, suggesting that it is not in a sequestered site, and is specifically incorporated in low amounts into HIV-1(Deltavif). Although A3G and/or A3F inactivate HIV-1(Deltavif) and are neutralized by Vif, the antiviral properties of cell lines are also influenced by other cellular and viral factors.


Subject(s)
Antiviral Agents , Cytidine Deaminase/biosynthesis , Cytosine Deaminase/biosynthesis , Nucleoside Deaminases/metabolism , Repressor Proteins/metabolism , APOBEC-3G Deaminase , Base Sequence , Blotting, Northern , Cytidine Deaminase/physiology , Cytosine Deaminase/physiology , DNA Primers , Gene Products, vif/physiology , HeLa Cells , Humans , Minor Histocompatibility Antigens , Molecular Sequence Data , Nucleoside Deaminases/physiology , Repressor Proteins/physiology , Reverse Transcriptase Polymerase Chain Reaction
18.
Toxicol Pathol ; 32(5): 577-90, 2004.
Article in English | MEDLINE | ID: mdl-15603542

ABSTRACT

Kidney slices represent an in vitro model that has the cellular complexity of in vivo tissue to provide insights into mechanisms of organ injury, as shown in this study with the model nephrotoxicant cisplatin. Cell pathways altered by cisplatin exposure are assessed by gene expression analysis, cell function, and morphology in human and rat kidney slices in comparison to rat kidney from an in vivo study. The acute nephrosis of the tubular epithelium induced by cisplatin in vivo was reproduced in both human and rat kidney slices, while the glomerulus appeared resistant even at high concentrations. Kidney gene expression changes of in vivo and in vitro samples were indicative of transcription, DNA damage, cell cycle, proliferation, and apoptosis that are in agreement with the mechanism of cisplatin causing DNA damage, growth arrest, and apoptosis; while genes indicative of protein damage, the disruption of transport and calcium homeostasis, cellular metabolism, and oxidative stress are pathways linked with cisplatin binding to various cellular proteins and macromolecules. Both concentration and time-dependent gene expression changes evident in the in vitro model preceded a change in tissue morphology. Functional assays confirming cell dysfunction and increased apoptosis revealed the rat kidney to be more sensitive to the effects of cisplatin than human kidney as demonstrated by significant decreases in slice ATP and GSH levels, significant increases in caspase 9 and 3 activity, p53 protein levels, and increased DNA laddering. The regional markers of proximal and distal tubular injury, alpha- and pi-glutathione S-transferases, were shown for the human kidney slices to be significantly increased by cisplatin. In this study, cisplatin-induced nephrotoxicity was demonstrated morphologically in rat and human kidney slices, and the associated gene expression and functional changes characterized the cellular pathways involved.


Subject(s)
Antineoplastic Agents/toxicity , Cisplatin/toxicity , Kidney Cortex/drug effects , Kidney Tubular Necrosis, Acute/chemically induced , Kidney/drug effects , Adult , Animals , Antineoplastic Agents/administration & dosage , Cisplatin/administration & dosage , Culture Media/chemistry , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Gene Expression Profiling , Humans , Injections, Intravenous , Kidney/metabolism , Kidney/pathology , Kidney Cortex/metabolism , Kidney Cortex/pathology , Kidney Function Tests , Kidney Tubular Necrosis, Acute/genetics , Kidney Tubular Necrosis, Acute/pathology , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Organ Culture Techniques , Rats , Rats, Wistar
19.
Toxicol Sci ; 82(2): 534-44, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15456927

ABSTRACT

Liver slice viability is extended to 96 h for rat, expanding the use of this in vitro model for studying mechanisms of injury and repair, including pathways of fibrosis. The contributing factors to increased organ slice survival consist of the use of a preservation solution for liver perfusion and slice preparation, obtaining rats that are within the weight range of 250-325 g, placing a cellulose filter atop the titanium mesh roller-insert to support the slice, and maintaining the slices in an optimized culture medium which is replaced daily. The liver slices remain metabolically active, synthesizing adenosine triphosphate (ATP), glutathione, and glycogen, and exhibit preserved organelle integrity and slice morphology. Slice preparation results in 2-cut surfaces which likely triggers a repair and regenerative response. The fibrogenic pathways are evident by the activation of stellate cells, the proliferation of myofibroblast-like cells, and an increased collagen deposition by 48 h. Markers indicative of activated stellate cells, alpha-smooth muscle actin, collagen 1a1, desmin, and HSP47 are substantiated by real time-PCR. Increased staining of alpha-smooth muscle actin initially around the vessels and by 72-96 h in the tissue is accompanied by increased collagen staining. Microarray gene expression revealed extracellular matrix changes with the up-regulation of cytoskeleton, filaments, collagens, and actin genes; and the down-regulation of genes linked with lipid metabolism. The improvements in extending liver slice survival, in conjunction with its three-dimensional multi-cellular complexity, increases the application of this in vitro model for investigating pathways of injury and repair, and fibrosis.


Subject(s)
Liver Cirrhosis/pathology , Liver/pathology , Organ Culture Techniques , Signal Transduction/physiology , Adenosine Triphosphate/metabolism , Animals , Caspases/metabolism , Collagen/metabolism , Extracellular Matrix/physiology , Extracellular Matrix/ultrastructure , Gene Expression/drug effects , Genetic Markers , Glutathione/metabolism , Immunohistochemistry , Liver/metabolism , Liver Cirrhosis/metabolism , Liver Regeneration/physiology , Male , Microscopy, Electron , Oligonucleotide Array Sequence Analysis , RNA/biosynthesis , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
20.
J Biol Chem ; 279(40): 41744-9, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15297452

ABSTRACT

Apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like 3G (APOBEC3G) is an antiretroviral deoxycytidine deaminase that lethally hypermutates human immunodeficiency virus type 1 (HIV-1) but is itself neutralized by the HIV-1-encoded viral infectivity factor. Accordingly, APOBEC3G occurs specifically in human T lymphocytic cell lines that contain this antiviral defense, including H9. Since the substrate specificities of related cytidine deaminases are strongly influenced by their intracellular quantities, we analyzed the factors that control APOBEC3G expression. The levels of APOBEC3G mRNA and protein were unaffected by treatment of proliferating H9 cells with interferons or tumor necrosis factor-alpha but were enhanced up to 20-fold by phorbol myristate acetate. This induction was mediated at the transcriptional level by a pathway that required activation of the protein kinase Calpha/betaI isozyme (PKC), mitogen-activated protein kinase kinase (MEK) 1 and 2, and extracellular signal-regulated kinase (ERK). Correspondingly, induction of APOBEC3G was blocked by multiple inhibitors that act at diverse steps of this pathway. The PKCalpha/betaI/MEK/ERK pathway also controlled basal levels of APOBEC3G mRNA and protein, which consequently declined when cells were treated with these inhibitors or arrested in the G(0) state of the cell cycle by serum starvation. We conclude that expression of the antiviral APOBEC3G editing enzyme is dynamically controlled by the PKCalpha/betaI/MEK/ERK protein kinase cascade in human T lymphocytes.


Subject(s)
Cytidine Deaminase/biosynthesis , Protein Biosynthesis , T-Lymphocytes/metabolism , Tetradecanoylphorbol Acetate/analogs & derivatives , Transcription, Genetic , Up-Regulation , APOBEC-3G Deaminase , Cell Line , Cytidine Deaminase/genetics , Cytidine Deaminase/physiology , HIV-1/genetics , Humans , MAP Kinase Kinase 1 , MAP Kinase Kinase 2 , Mitogen-Activated Protein Kinase Kinases , Mitogen-Activated Protein Kinases , Nucleoside Deaminases , Protein Kinase C , Protein Kinase C-alpha , Protein-Tyrosine Kinases , Proteins/genetics , Proteins/physiology , RNA, Messenger/analysis , Repressor Proteins , Signal Transduction , Tetradecanoylphorbol Acetate/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...