Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Bioanalysis ; 12(20): 1439-1447, 2020 Oct.
Article in English | MEDLINE | ID: mdl-33006478

ABSTRACT

Aim: There is little information in the literature regarding assays for measuring CDH17 in tissues. Numerous studies indicate overexpression of CDH17 in a variety of diseases including hepatocellular carcinoma, colorectal and gastric cancer. Here we present an immunoaffinity enrichment LC-MS/MS approach for analysis of CDH17 in human tissues, plasma and serum as well as preclinical models. Results: CDH17 levels were measured in colon and ileum tissues from healthy donors and inflamed tissues from patients with Ulcerative Colitus or Crohn's disease. Applicability of the immunocapture LC-MS/MS approach is demonstrated for colon tissues from non-diseased mouse and cynomolgus monkey. Conclusion: The analytical approaches discussed here are suitable for quantitation of CDH17 in various tissues to enable both preclinical and clinical assessment.


Subject(s)
Cadherins/metabolism , Chromatography, Affinity/methods , Chromatography, Liquid/methods , Tandem Mass Spectrometry/methods , Humans
2.
Sci Rep ; 10(1): 14408, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32873881

ABSTRACT

Quantitatively determining in vivo achievable drug concentrations in targeted organs of animal models and subsequent target engagement confirmation is a challenge to drug discovery and translation due to lack of bioassay technologies that can discriminate drug binding with different mechanisms. We have developed a multiplexed and high-throughput method to quantify drug distribution in tissues by integrating high content screening (HCS) with U-Net based deep learning (DL) image analysis models. This technology combination allowed direct visualization and quantification of biologics drug binding in targeted tissues with cellular resolution, thus enabling biologists to objectively determine drug binding kinetics.


Subject(s)
Cadherins/immunology , Carbocyanines , Deep Learning , Fluorescent Dyes , High-Throughput Screening Assays/methods , Image Processing, Computer-Assisted/methods , Immunoconjugates/metabolism , Animals , Cadherins/metabolism , Colon/metabolism , Drug Discovery/methods , Intestine, Small/metabolism , Mice , Tissue Distribution
3.
J Exp Med ; 212(7): 1081-93, 2015 Jun 29.
Article in English | MEDLINE | ID: mdl-26077719

ABSTRACT

The protein tyrosine phosphatase PTPN22(C1858T) allelic polymorphism is associated with increased susceptibility for development of systemic lupus erythematosus (SLE) and other autoimmune diseases. PTPN22 (also known as LYP) and its mouse orthologue PEP play important roles in antigen and Toll-like receptor signaling in immune cell functions. We demonstrate here that PEP also plays an important inhibitory role in interferon-α receptor (IFNAR) signaling in mice. PEP co-immunoprecipitates with components of the IFNAR signaling complex. Pep(-/-) hematopoietic progenitors demonstrate increased IFNAR signaling, increased IFN-inducible gene expression, and enhanced proliferation and activation compared to Pep(+/+) progenitors in response to IFN-α. In addition, Pep(-/-) mice treated with IFN-α display a profound defect in hematopoiesis, resulting in anemia, thrombocytopenia, and neutropenia when compared to IFN-α-treated Pep(+/+) mice. As SLE patients carrying the PTPN22(C1858T) risk variant have higher serum IFN-α activity, these data provide a molecular basis for how type I IFNs and PTPN22 may cooperate to contribute to lupus-associated cytopenias.


Subject(s)
Lupus Erythematosus, Systemic/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 22/metabolism , Receptor, Interferon alpha-beta/metabolism , Signal Transduction/physiology , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Hematopoietic Stem Cells/metabolism , Immunoprecipitation , Interferon-alpha/blood , Mice , Mice, Knockout , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics
4.
EMBO J ; 34(2): 218-35, 2015 Jan 13.
Article in English | MEDLINE | ID: mdl-25398911

ABSTRACT

Mitogen-activated protein kinase (MAPK) activation controls diverse cellular functions including cellular survival, proliferation, and apoptosis. Tuning of MAPK activation is counter-regulated by a family of dual-specificity phosphatases (DUSPs). IL-33 is a recently described cytokine that initiates Th2 immune responses through binding to a heterodimeric IL-33Rα (ST2L)/IL-1α accessory protein (IL-1RAcP) receptor that coordinates activation of ERK and NF-κB pathways. We demonstrate here that DUSP5 is expressed in eosinophils, is upregulated following IL-33 stimulation and regulates IL-33 signaling. Dusp5(-/-) mice have prolonged eosinophil survival and enhanced eosinophil effector functions following infection with the helminth Nippostrongylus brasiliensis. IL-33-activated Dusp5(-/-) eosinophils exhibit increased cellular ERK1/2 activation and BCL-XL expression that results in enhanced eosinophil survival. In addition, Dusp5(-/-) eosinophils demonstrate enhanced IL-33-mediated activation and effector functions. Together, these data support a role for DUSP5 as a novel negative regulator of IL-33-dependent eosinophil function and survival.


Subject(s)
Dual-Specificity Phosphatases/physiology , Eosinophils/immunology , Interleukins/pharmacology , Killer Cells, Natural/immunology , Strongylida Infections/immunology , Animals , Blotting, Western , Cells, Cultured , DNA-Binding Proteins/physiology , Enzyme-Linked Immunosorbent Assay , Eosinophils/cytology , Eosinophils/drug effects , Eosinophils/parasitology , Female , Humans , Interleukin-33 , Killer Cells, Natural/cytology , Killer Cells, Natural/drug effects , Killer Cells, Natural/parasitology , Mice , Mice, Knockout , Nippostrongylus/pathogenicity , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Strongylida Infections/drug therapy , Strongylida Infections/mortality , Strongylida Infections/parasitology
5.
Virology ; 421(1): 12-8, 2011 Dec 05.
Article in English | MEDLINE | ID: mdl-21974802

ABSTRACT

We have previously reported that HIV-1 preferentially infects Foxp3+ Treg cells in vitro and in vivo, and Foxp3 enhances the HIV-1 LTR expression through epigenetic mechanisms in T cells. We report here that histone deacetylase inhibitor (HDACi) failed to further enhance HIV gene expression in FoxP3+ T cells. We discovered that Foxp3 inhibited cellular HDAC activity in T cells, and mutations in the forkhead domain that ablate Foxp3 function also abolished its ability to inhibit HDAC. When co-expressed, Foxp3 specifically inhibited the deacetylase activity of HDAC1. We further showed that Foxp3 was associated with HDAC1, and mutations in the forkhead domain that ablate Foxp3 function in Treg cells also inhibited Foxp3 association with and inhibition of HDAC1. Finally, Foxp3 failed to enhance HIV-1 gene expression in human T cells expressing HDAC1-specific shRNA. We conclude that Foxp3 modulates gene expression in human T cells at least partly by inhibiting HDAC1 activity.


Subject(s)
Down-Regulation , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Viral , HIV Infections/metabolism , HIV-1/physiology , Histone Deacetylase 1/metabolism , T-Lymphocytes/metabolism , Cell Line , Forkhead Transcription Factors/chemistry , Forkhead Transcription Factors/genetics , HIV Infections/enzymology , HIV Infections/genetics , HIV Infections/virology , HIV-1/genetics , Histone Deacetylase 1/genetics , Humans , Protein Binding , Protein Structure, Tertiary , T-Lymphocytes/enzymology , T-Lymphocytes/virology
6.
Immunol Res ; 41(3): 248-66, 2008.
Article in English | MEDLINE | ID: mdl-18726715

ABSTRACT

FoxP3(+)CD4(+)CD25(+) regulatory T (Treg) cells are implicated in a number of pathologic processes including elevated levels in cancers and infectious diseases, and reduced levels in autoimmune diseases. Treg cells are activated to modulate immune responses to avoid over-reactive immunity. However, conflicting findings are reported regarding relative levels of Treg cells during HIV-1 infection and disease progression. The role of Treg cells in HIV-1 diseases (aberrant immune activation) is poorly understood due to lack of a robust model. We summarize here the regulation and function of Foxp3 in Treg cells and in modulating HIV-1 replication. Based on recent findings from SIV/monkey and HIV/humanized mouse models, a model of the dual role of Treg cells in HIV-1 infection and immuno-pathogenesis is discussed.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Forkhead Transcription Factors/immunology , HIV Infections/immunology , HIV-1/immunology , T-Lymphocytes, Regulatory/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Cell Differentiation/immunology , Disease Models, Animal , Forkhead Transcription Factors/metabolism , HIV Infections/virology , HIV-1/metabolism , Humans , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/virology
7.
J Leukoc Biol ; 82(2): 361-9, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17502338

ABSTRACT

We have reported previously that p115Rho guanine nucleotide exchange factor, its upstream activator Galpha13, and its effector RhoA are able to inhibit HIV-1 replication. Here, we show that RhoA is able to inhibit HIV-1 gene expression through the NFAT-binding site in the HIV long-terminal repeat. Constitutively active NFAT counteracts the inhibitory activity of RhoA, and inhibition of NFAT activation also inhibits HIV-1 gene expression. We have shown further that RhoA inhibits NFAT-dependent transcription and IL-2 production in human T cells. RhoA does not inhibit nuclear localization of NFAT but rather, inhibits its transcriptional activity. In addition, RhoA decreases the level of acetylated histone H3, but not NFAT occupancy, at the IL-2 promoter. These data suggest that activation of RhoA can modulate IL-2 gene expression by inhibiting the transcriptional activity of NFAT and chromatin structure at the IL-2 promoter during T cell activation.


Subject(s)
Gene Expression Regulation , NFATC Transcription Factors/metabolism , Signal Transduction , T-Lymphocytes/metabolism , rhoA GTP-Binding Protein/physiology , Genes, Reporter , HIV Long Terminal Repeat/physiology , Humans , Jurkat Cells , Luciferases/metabolism , NFATC Transcription Factors/genetics , Retroviridae/genetics , Transduction, Genetic
8.
J Biol Chem ; 282(22): 15973-80, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17416586

ABSTRACT

FoxP3 determines the development of CD4+CD25+ regulatory T (Treg) cells and represses interleukin-2 (IL-2) expression in Treg cells. However, human immunodeficiency virus type 1 (HIV-1) infects and replicates efficiently in FoxP3+ Treg cells. We report that, while inhibiting IL-2 gene expression, FoxP3 enhances gene expression from HIV-1 long terminal repeat (LTR). This FoxP3 activity requires both the N- and C-terminal domains and is inactivated by human IPEX (immunodysregulation, polyendocrinopathy, enteropathy, X-linked syndrome) mutations. FoxP3 enhances HIV-1 LTR via its specific NFkappaB binding sequences in an NFkappaB-dependent fashion in T cells but not in HEK293 cells. FoxP3 decreases level of histone acetylation at the interleukin-2 locus but not at the HIV-1 LTR. Although NFkappaB nuclear translocation is not altered, FoxP3 enhances NFkappaB-p65 binding to HIV-1 LTR. These data suggest that FoxP3 modulates gene expression in a promoter sequence-dependent fashion by modulating chromatin structure and NFkappaB activity. HIV-1 LTR has evolved to both highjack the T-cell activation pathway for expression and to resist FoxP3-mediated suppression of T-cell activation.


Subject(s)
Cell Nucleus/metabolism , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Viral , HIV Long Terminal Repeat , HIV-1/metabolism , T-Lymphocytes, Regulatory/metabolism , Transcription Factor RelA/metabolism , Acetylation , Active Transport, Cell Nucleus/genetics , Cell Line , Cell Nucleus/genetics , Cell Nucleus/virology , Chromatin/metabolism , Chromatin/virology , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Viral/genetics , Histones/metabolism , Humans , Interleukin-2/biosynthesis , Mutation , Promoter Regions, Genetic , Quantitative Trait Loci , T-Lymphocytes, Regulatory/virology , Virus Replication/genetics
9.
Traffic ; 7(12): 1643-53, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17118119

ABSTRACT

RhoGTPases play important roles in the regulation of protein transport and membrane recycling. Little is known, however, about how RhoGTPases affect HIV-1 virion production, which is dependent on the endosomal sorting pathway. We report that ectopic expression of citron kinase (citron-K), a RhoA effector, preferentially enhances HIV-1 virion production. Depletion of endogenous citron-K inhibits HIV-1 virion production. Citron-N, which lacks the kinase domain, also enhances HIV-1 virion production. The leucine zipper, Rho-binding and zinc finger domains of citron-N are necessary for the enhancement activity. Citron-K also enhances murine leukemia virion production and the HIV-1 late domain is not required for the citron-K-mediated enhancement. Ectopic expression of citron-K leads to the formation of cytoplasmic structures containing citron-K and HIV-1 Gag proteins. HIV-1 and citron-K cooperatively enhance acidic endosome and lysosome compartments. Finally, citron-K promotes exocytosis of microvesicles or exosomes that co-purify with HIV-1 virions. We conclude that citron-K enhances HIV-1 virion production by stimulating the endosomal compartments and exocytosis.


Subject(s)
Exocytosis , HIV-1/metabolism , Protein Serine-Threonine Kinases/metabolism , Virion/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Cell Line , Endosomes/metabolism , Gene Deletion , Gene Products, gag/deficiency , Gene Products, gag/genetics , Gene Products, gag/metabolism , HIV-1/genetics , Humans , Intracellular Signaling Peptides and Proteins , Leucine Zippers , Lysosomes/metabolism , Mice , Protein Binding , Protein Serine-Threonine Kinases/genetics , Virus Replication , Zinc Fingers
10.
J Clin Microbiol ; 43(7): 3227-36, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16000440

ABSTRACT

The use of immunoglobulin M (IgM) antibody-capture enzyme-linked immunosorbent assay (MAC-ELISA) serves as a valuable tool for the diagnosis of acute flaviviral infections, since IgM antibody titers are detectable early, peak at about 2 weeks postinfection, and subsequently decline to lower levels over the next few months. Traditionally, virus-infected tissue culture or suckling mouse brain (SMB) has been the source of viral antigens used in the assay. In an effort to provide a reliable source of standardized viral antigens for serodiagnosis of the medically important flaviviruses, we have developed a eukaryotic plasmid vector to express the premembrane/membrane and envelope proteins which self-assemble into noninfectious virus-like particles (VLPs). In addition to the plasmids for Japanese encephalitis virus, West Nile virus (WNV), St. Louis encephalitis virus (SLEV), and dengue virus type 2 (DENV-2) reported earlier, we recently constructed the DENV-1, -3, and -4 VLP expression plasmids. Three blind-coded human serum panels were assembled from patients having recent DENV, SLEV, and WNV infections to assess the sensitivity and specificity of the MAC-ELISA using VLPs or SMB antigens. In addition, serum specimens from patients infected with either Powassan virus or La Crosse encephalitis virus were used to evaluate the cross-reactivity of seven mosquito-borne viral antigens. The results of the present studies showed higher sensitivity when using SLEV and WNV VLPs and higher specificity when using SLEV, WNV, and the mixture of DENV-1 to -4 VLPs in the MAC-ELISA than when using corresponding SMB antigens. Receiver operating characteristic (ROC) curve analysis, a plot of the sensitivity versus false positive rate (100 - specificity), was applied to discriminate the accuracy of tests comparing the use of VLPs and SMB antigen. The measurement of assay performance by the ROC analysis indicated that there were statistically significant differences in assay performance between DENV and WNV VLPs and the respective SMB antigens. Additionally, VLPs had a lower cutoff positive/negative ratio than corresponding SMB antigens when employed for the confirmation of current infections. The VLPs also performed better than SMB antigens in the MAC-ELISA, as indicated by a higher positive prediction value and positive likelihood ratio test. Cell lines continuously secreting these VLPs are therefore a significantly improved source of serodiagnostic antigens compared to the traditional sources of virus-infected tissue culture or suckling mouse brain.


Subject(s)
Antibodies, Viral/blood , Antigens, Viral/immunology , Flavivirus Infections/diagnosis , Flavivirus/immunology , Immunoglobulin M/blood , Virion/immunology , Animals , Animals, Suckling , Brain/virology , CHO Cells , COS Cells , Chlorocebus aethiops , Cricetinae , Enzyme-Linked Immunosorbent Assay , Flavivirus/classification , Flavivirus Infections/epidemiology , Flavivirus Infections/immunology , Flavivirus Infections/virology , Humans , Mice , ROC Curve , Sensitivity and Specificity
11.
Virology ; 306(1): 170-80, 2003 Feb 01.
Article in English | MEDLINE | ID: mdl-12620809

ABSTRACT

We have constructed a series of plasmids encoding premembrane (prM) and envelope (E) protein genes of dengue virus type 2 (DEN-2). These plasmids included an authentic DEN-2 prM-E construct (pCBD2-14-6), and two chimeric constructs, 90% DEN-2 E-10% Japanese encephalitis (JE) virus E (pCB9D2-1J-4-3) and 80% DEN-2 E-20% JE E (pCB8D2-2J-2-9-1). Monoclonal antibody (MAb) reactivity indicated that all three plasmids expressed authentic DEN-2 virus E protein epitopes representative of flavivirus domains 1, 2, and 3. However, only the pCB8D2-2J-2-9-1 construct secreted high levels of prM, M (membrane), and E proteins into the culture fluid of plasmid-transformed COS-1 cells. The major portion of the prM and E proteins expressed by COS-1 cells transformed by pCBD2-14-6 or pCB9D2-4-3 plasmids remained membrane-bound. The results supported the notion that an unidentified membrane retention sequence is located between E-397 and E-436 of DEN-2 virus E protein. Replacing the carboxyl-terminal 20% of DEN-2 E (397-450) with the corresponding JE sequence had no effect on anti-DEN-2 MAb reactivity, indicating that this region is antigenically inert, although it is required for antigen secretion. Plasmid pCBD2-2J-2-9-1, which expressed secreted forms of prM/M and E that have the potential to form subviral particles, was superior to other constructs in stimulating an antibody response. Ninety percent neutralization titers ranging from 1:40 to >1:1000 were observed in seven of nine serum specimens from pCB8D2-2J-2-9-1-immunized mice. Eleven of twelve 2-day-old neonatal mice, derived from a pCB8D2-2J-2-9-1 immunized female mouse, survived intraperitoneal challenge of DEN-2 New Guinea C virus.


Subject(s)
Dengue Virus/genetics , Encephalitis Virus, Japanese/genetics , Plasmids/genetics , Recombinant Fusion Proteins/metabolism , Viral Envelope Proteins , Animals , Antibodies, Viral/blood , COS Cells , Cell Membrane/metabolism , Chlorocebus aethiops , Dengue/immunology , Dengue/prevention & control , Dengue/virology , Dengue Virus/immunology , Dengue Virus/metabolism , Encephalitis Virus, Japanese/metabolism , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Female , Male , Mice , Mice, Inbred ICR , Neutralization Tests , Plasmids/immunology , Recombinant Fusion Proteins/genetics , Vaccination , Vero Cells , Viral Envelope Proteins/biosynthesis , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Viral Vaccines/immunology
12.
Med Educ ; 36(10): 979-84, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12390467

ABSTRACT

BACKGROUND AND STUDY AIMS: This paper reports on a 1-year, full-time widening access to medicine course that prepares mature adults for entry to selected UK medical schools. The course was developed in 1992-93 in co-operation with the University of Leicester Medical School and is restricted to adults over 21 years of age. The main subjects of the curriculum are biology, chemistry and physics. These are supported by mathematics, statistics and medical geography. METHODS: Three successive year groups were used as trial groups and nine medical schools agreed to participate in the trial. The successful trial students were monitored as they progressed through medical school. A total of 19% of successful trial students graduated from medical school with honours degrees. Evaluation of the trial years has led to changes in both the curriculum and assessment methods. FINDINGS: The longitudinal study of the course indicates that progression to medical school has increased from an average of 64% during the trial years to an average of 85% over the last 3 years. Graduate entry to the course has increased from an initial 10% of the intake to 50% over the same period. The role of the programme as a vehicle for widening access to medical school was monitored in the 4-year period 1997-2000. In these years, 41% of the student intake progressing to medical school came from socioeconomic groups IV and V, whilst 36% came from socioeconomic groups I and II. The 2001/2002 cohort numbers 45.


Subject(s)
College Admission Test , Curriculum , Education, Medical , Adult , England , Female , Humans , Male , Socioeconomic Factors , Students, Medical , Workforce
13.
Emerg Infect Dis ; 8(4): 380-6, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11971771

ABSTRACT

A total of 12 horses of different breeds and ages were infected with West Nile virus (WNV) via the bites of infected Aedes albopictus mosquitoes. Half the horses were infected with a viral isolate from the brain of a horse (BC787), and half were infected with an isolate from crow brain (NY99-6625); both were NY99 isolates. Postinfection, uninfected female Ae. albopictus fed on eight of the infected horses. In the first trial, Nt antibody titers reached >1:320, 1:20, 1:160, and 1:80 for horses 1 to 4, respectively. In the second trial, the seven horses with subclinical infections developed Nt antibody titers >1:10 between days 7 and 11 post infection. The highest viremia level in horses fed upon by the recipient mosquitoes was approximately 460 Vero cell PFU/mL. All mosquitoes that fed upon viremic horses were negative for the virus. Horses infected with the NY99 strain of WNV develop low viremia levels of short duration; therefore, infected horses are unlikely to serve as important amplifying hosts for WNV in nature.


Subject(s)
Horses/virology , West Nile Fever/transmission , West Nile virus/physiology , Aedes/physiology , Aedes/virology , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Bird Diseases/virology , Brain/virology , Chlorocebus aethiops , Female , Horse Diseases/blood , Horse Diseases/immunology , Horse Diseases/transmission , Horse Diseases/virology , Horses/blood , Horses/immunology , Insect Vectors/physiology , Insect Vectors/virology , Male , Saliva/immunology , Saliva/virology , Songbirds/virology , Vero Cells , Viremia/blood , Viremia/immunology , Viremia/transmission , Viremia/virology , West Nile Fever/blood , West Nile Fever/immunology , West Nile Fever/virology , West Nile virus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...