Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
AIDS ; 2024 May 31.
Article in English | MEDLINE | ID: mdl-38819840

ABSTRACT

People living with HIV (PLWH) are at greater risk of developing lung diseases even when they are antiretroviral therapy (ART)-adherent and virally suppressed. The most common pulmonary function abnormality in PLWH is that of impaired d iffusing capacity of the l ungs for c arbon monoxide (DL CO ), which is an independent risk factor for increased mortality in PLWH. Earlier work has identified several plasma biomarkers of inflammation and immune activation to be associated with decreased DL CO . However, the underpinning molecular mechanisms of HIV-associated impaired DL CO are largely unknown. We conducted a pilot study of PLWH with normal DL CO (values greater than or equal to the l ower l imit of n ormal, DL CO ≥LLN) or abnormal DL CO (DL CO

2.
PLoS Pathog ; 19(11): e1011771, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37934757

ABSTRACT

Kaposi sarcoma-associated herpesvirus (KSHV) inflammatory cytokine syndrome (KICS) is a newly described chronic inflammatory disease condition caused by KSHV infection and is characterized by high KSHV viral load and sustained elevations of serum KSHV-encoded IL-6 (vIL-6) and human IL-6 (hIL-6). KICS has significant immortality and greater risks of other complications, including malignancies. Although prolonged inflammatory vIL-6 exposure by persistent KSHV infection is expected to have key roles in subsequent disease development, the biological effects of prolonged vIL-6 exposure remain elusive. Using thiol(SH)-linked alkylation for the metabolic (SLAM) sequencing and Cleavage Under Target & Release Using Nuclease analysis (CUT&RUN), we studied the effect of prolonged vIL-6 exposure in chromatin landscape and resulting cytokine production. The studies showed that prolonged vIL-6 exposure increased Bromodomain containing 4 (BRD4) and histone H3 lysine 27 acetylation co-occupancies on chromatin, and the recruitment sites were frequently co-localized with poised RNA polymerase II with associated enzymes. Increased BRD4 recruitment on promoters was associated with increased and prolonged NF-κB p65 binding after the lipopolysaccharide stimulation. The p65 binding resulted in quicker and sustained transcription bursts from the promoters; this mechanism increased total amounts of hIL-6 and IL-10 in tissue culture. Pretreatment with the BRD4 inhibitors, OTX015 and MZ1, eliminated the enhanced inflammatory cytokine production. These findings suggest that persistent vIL-6 exposure may establish a chromatin landscape favorable for the reactivation of inflammatory responses in monocytes. This epigenetic memory may explain the greater risk of chronic inflammatory disease development in KSHV-infected individuals.


Subject(s)
Herpesviridae Infections , Herpesvirus 8, Human , Sarcoma, Kaposi , Humans , Herpesvirus 8, Human/physiology , Interleukin-6/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Cytokines/metabolism , Herpesviridae Infections/metabolism , Chromatin/metabolism , Epigenesis, Genetic , Cell Cycle Proteins/metabolism
3.
PLoS Pathog ; 19(10): e1011703, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37883374

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) is an oncogenic double-stranded DNA virus and the etiologic agent of Kaposi's sarcoma and hyperinflammatory lymphoproliferative disorders. Understanding the mechanism by which KSHV increases the infected cell population is crucial for curing KSHV-associated diseases. Using scRNA-seq, we demonstrate that KSHV preferentially infects CD14+ monocytes, sustains viral lytic replication through the viral interleukin-6 (vIL-6), which activates STAT1 and 3, and induces an inflammatory gene expression program. To study the role of vIL-6 in monocytes upon KSHV infection, we generated recombinant KSHV with premature stop codon (vIL-6(-)) and its revertant viruses (vIL-6(+)). Infection of the recombinant viruses shows that both vIL-6(+) and vIL-6(-) KSHV infection induced indistinguishable host anti-viral response with STAT1 and 3 activations in monocytes; however, vIL-6(+), but not vIL-6(-), KSHV infection promoted the proliferation and differentiation of KSHV-infected monocytes into macrophages. The macrophages derived from vIL-6(+) KSHV infection showed a distinct transcriptional profile of elevated IFN-pathway activation with immune suppression and were compromised in T-cell stimulation function compared to those from vIL-6(-) KSHV infection or uninfected control. Notably, a viral nuclear long noncoding RNA (PAN RNA), which is required for sustaining KSHV gene expression, was substantially reduced in infected primary monocytes upon vIL-6(-) KSHV infection. These results highlight the critical role of vIL-6 in sustaining KSHV transcription in primary monocytes. Our findings also imply a clever strategy in which KSHV utilizes vIL-6 to secure its viral pool by expanding infected monocytes via differentiating into longer-lived dysfunctional macrophages. This mechanism may facilitate KSHV to escape from host immune surveillance and to support a lifelong infection.


Subject(s)
Herpesviridae Infections , Herpesvirus 8, Human , Sarcoma, Kaposi , Humans , Herpesvirus 8, Human/physiology , Interleukin-6/metabolism , Monocytes/metabolism , Herpesviridae Infections/metabolism , Macrophages/metabolism , Immunologic Factors/metabolism , Virus Replication
4.
Cells ; 12(15)2023 07 31.
Article in English | MEDLINE | ID: mdl-37566056

ABSTRACT

CD40L is expressed in activated T cells, and it plays a major role in immune response and is a major therapeutic target for inflammation. High IgM syndrome type 1 (HIGM1) is a congenital functional defect in CD40L/CD40 signaling due to defective CD40L. CD40L is also stored in platelet granules and transported to the surface upon platelet activation. Platelet integrin αIIbß3 is known to bind to fibrinogen and activation of αIIbß3 is a key event that triggers platelet aggregation. Also, the KGD motif is critical for αIIbß3 binding and the interaction stabilizes thrombus. Previous studies showed that CD40L binds to and activates integrins αvß3 and α5ß1 and that HIGM1 mutations are clustered in the integrin-binding sites. However, the specifics of CD40L binding to αIIbß3 were unclear. Here, we show that CD40L binds to αIIbß3 in a KGD-independent manner using CD40L that lacks the KGD motif. Two HIGM1 mutants, S128E/E129G and L155P, reduced the binding of CD40L to the classical ligand-binding site (site 1) of αIIbß3, indicating that αIIbß3 binds to the outer surface of CD40L trimer. Also, CD40L bound to the allosteric site (site 2) of αIIbß3 and allosterically activated αIIbß3 without inside-out signaling. Two HIMG1 mutants, K143T and G144E, on the surface of trimeric CD40L suppressed CD40L-induced αIIbß3 activation. These findings suggest that CD40L binds to αIIbß3 in a manner different from that of αvß3 and α5ß1 and induces αIIbß3 activation. HIGM1 mutations are clustered in αIIbß3 binding sites in CD40L and are predicted to suppress thrombus formation and immune responses through αIIbß3.


Subject(s)
Hyper-IgM Immunodeficiency Syndrome, Type 1 , Thrombosis , Humans , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , CD40 Ligand/genetics , CD40 Ligand/metabolism , Allosteric Site , Binding Sites , Mutation/genetics , Integrin alpha5beta1/metabolism
5.
bioRxiv ; 2023 Jul 23.
Article in English | MEDLINE | ID: mdl-37503036

ABSTRACT

Kaposi sarcoma-associated herpesvirus (KSHV) inflammatory cytokine syndrome (KICS) is a newly described chronic inflammatory disease condition caused by KSHV infection and is characterized by high KSHV viral load and sustained elevations of serum KSHV-encoded IL-6 (vIL-6) and human IL-6 (hIL-6). KICS has significant immortality and possesses greater risks of having other complications, which include malignancies. Although prolonged inflammatory vIL-6 exposure by persistent KSHV infection is expected to have key roles in subsequent disease development, the biological effects of prolonged vIL-6 exposure remain elusive. Using thiol-Linked Alkylation for the Metabolic Sequencing and Cleavage Under Target & Release Using Nuclease analysis, we studied the effect of prolonged vIL-6 exposure in chromatin landscape and resulting cytokine production. The studies showed that prolonged vIL-6 exposure increased Bromodomain containing 4 (BRD4) and histone H3 lysine 27 acetylation co-occupancies on chromatin, and the recruitment sites were frequently co-localized with poised RNAPII with associated enzymes. Increased BRD4 recruitment on promoters was associated with increased and prolonged NF-κB p65 binding after the lipopolysaccharide stimulation. The p65 binding resulted in quicker and sustained transcription bursts from the promoters; this mechanism increased total amounts of hIL-6 and IL-10 in tissue culture. Pretreatment with the BRD4 inhibitor, OTX015, eliminated the enhanced inflammatory cytokine production. These findings suggest that persistent vIL-6 exposure may establish a chromatin landscape favorable for the reactivation of inflammatory responses in monocytes. This epigenetic memory may explain the greater risk of chronic inflammatory disease development in KSHV-infected individuals.

6.
bioRxiv ; 2023 Mar 26.
Article in English | MEDLINE | ID: mdl-36945595

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) is an oncogenic double-stranded DNA virus and the etiologic agent of Kaposi's sarcoma and hyperinflammatory lymphoproliferative disorders. Understanding the mechanism by which KSHV increases the infected cell population is crucial for curing KSHV-associated diseases. Here we demonstrate that KSHV preferentially infects CD14 + monocytes and sustains viral replication through the viral interleukin-6 (vIL6)-mediated activation of STAT1 and 3. Using vIL6-sufficient and vIL6-deficient recombinant KSHV, we demonstrated that vIL6 plays a critical role in promoting the proliferation and differentiation of KSHV-infected monocytes into macrophages. The macrophages derived from vIL6-sufficient KSHV infection showed a distinct transcriptional profile of elevated IFN-pathway activation with immune suppression and were compromised in T-cell stimulation function compared to those from vIL6-deficient KSHV infection or uninfected control. These results highlight a clever strategy, in which KSHV utilizes vIL6 to secure its viral pool by expanding infected dysfunctional macrophages. This mechanism also facilitates KSHV to escape from host immune surveillance and to establish a lifelong infection. 160. Summary: KSHV causes multiple inflammatory diseases, however, the underlying mechanism is not clear. Shimoda et al. demonstrate that KSHV preferentially infects monocytes and utilizes virally encoded interleukin-6 to expand and deregulate infected monocytes. This helps the virus escape from host immune surveillance.

7.
Cell Rep ; 39(6): 110788, 2022 05 10.
Article in English | MEDLINE | ID: mdl-35545047

ABSTRACT

Kaposi sarcoma-associated herpesvirus (KSHV) establishes a latent infection in the cell nucleus, but where KSHV episomal genomes are tethered and the mechanisms underlying KSHV lytic reactivation are unclear. Here, we study the nuclear microenvironment of KSHV episomes and show that the KSHV latency-lytic replication switch is regulated via viral long non-coding (lnc)RNA-CHD4 (chromodomain helicase DNA binding protein 4) interaction. KSHV episomes localize with CHD4 and ADNP proteins, components of the cellular ChAHP complex. The CHD4 and ADNP proteins occupy the 5'-region of the highly inducible lncRNAs and terminal repeats of the KSHV genome together with latency-associated nuclear antigen (LANA). Viral lncRNA binding competes with CHD4 DNA binding, and KSHV reactivation sequesters CHD4 from the KSHV genome, which is also accompanied by detachment of KSHV episomes from host chromosome docking sites. We propose a model in which robust KSHV lncRNA expression determines the latency-lytic decision by regulating LANA/CHD4 binding to KSHV episomes.


Subject(s)
Herpesvirus 8, Human , RNA, Long Noncoding , Sarcoma, Kaposi , Antigens, Viral/genetics , Antigens, Viral/metabolism , Chromosomes/metabolism , Herpesvirus 8, Human/genetics , Humans , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Plasmids , RNA, Long Noncoding/genetics , Tumor Microenvironment , Virus Latency/genetics
8.
Commun Biol ; 4(1): 1330, 2021 12 02.
Article in English | MEDLINE | ID: mdl-34857874

ABSTRACT

In herpesvirus replicating cells, host cell gene transcription is frequently down-regulated because important transcriptional apparatuses are appropriated by viral transcription factors. Here, we show a small peptide derived from the Kaposi's sarcoma-associated herpesvirus transactivator (K-Rta) sequence, which attenuates cellular MYC expression, reduces cell proliferation, and selectively kills cancer cell lines in both tissue culture and a xenograft tumor mouse model. Mechanistically, the peptide functions as a decoy to block the recruitment of coactivator complexes consisting of Nuclear receptor coactivator 2 (NCOA2), p300, and SWI/SNF proteins to the MYC promoter in primary effusion lymphoma cells. Thiol(SH)-linked alkylation for the metabolic sequencing of RNA (SLAM seq) with target-transcriptional analyses further confirm that the viral peptide directly attenuates MYC and MYC-target gene expression. This study thus provides a unique tool to control MYC activation, which may be used as a therapeutic payload to treat MYC-dependent diseases such as cancers and autoimmune diseases.


Subject(s)
Herpesvirus 8, Human/physiology , Leukemia/physiopathology , Lymphoma/physiopathology , Proto-Oncogene Proteins c-myc/genetics , Trans-Activators/genetics , Cell Line, Tumor , Cell Proliferation , Herpesvirus 8, Human/chemistry , Humans , Proto-Oncogene Proteins c-myc/metabolism , Trans-Activators/metabolism , Tumor Cells, Cultured
9.
J Biol Chem ; 296: 100399, 2021.
Article in English | MEDLINE | ID: mdl-33571526

ABSTRACT

CD40L is a member of the TNF superfamily that participates in immune cell activation. It binds to and signals through several integrins, including αvß3 and α5ß1, which bind to the trimeric interface of CD40L. We previously showed that several integrin ligands can bind to the allosteric site (site 2), which is distinct from the classical ligand-binding site (site 1), raising the question of if CD40L activates integrins. In our explorations of this question, we determined that integrin α4ß1, which is prevalently expressed on the same CD4+ T cells as CD40L, is another receptor for CD40L. Soluble (s)CD40L activated soluble integrins αvß3, α5ß1, and α4ß1 in cell-free conditions, indicating that this activation does not require inside-out signaling. Moreover, sCD40L activated cell-surface integrins in CHO cells that do not express CD40. To learn more about the mechanism of binding, we determined that sCD40L bound to a cyclic peptide from site 2. Docking simulations predicted that the residues of CD40L that bind to site 2 are located outside of the CD40L trimer interface, at a site where four HIGM1 (hyper-IgM syndrome type 1) mutations are clustered. We tested the effect of these mutations, finding that the K143T and G144E mutants were the most defective in integrin activation, providing support that this region interacts with site 2. We propose that allosteric integrin activation by CD40L also plays a role in CD40L signaling, and defective site 2 binding may be related to the impaired CD40L signaling functions of these HIGM1 mutants.


Subject(s)
CD40 Ligand/metabolism , Integrin alpha4beta1/metabolism , Integrin alpha5beta1/metabolism , Integrin alphaVbeta3/metabolism , Receptors, Cell Surface/chemistry , T-Lymphocytes/metabolism , Allosteric Site , Animals , CD40 Ligand/immunology , Cell Line , Cricetinae , Humans , Integrin alpha4beta1/immunology , Integrin alpha5beta1/immunology , Integrin alphaVbeta3/immunology , Molecular Docking Simulation , Protein Binding , Receptors, Cell Surface/metabolism , Signal Transduction , T-Lymphocytes/immunology
10.
Sci Rep ; 10(1): 17505, 2020 10 15.
Article in English | MEDLINE | ID: mdl-33060657

ABSTRACT

Alterations in the human glycome have been associated with cancer and autoimmunity. Thus, constructing a site-specific map of the human glycome for biomarker research and discovery has been a highly sought-after objective. However, due to analytical barriers, comprehensive site-specific glycoprofiling is difficult to perform. To develop a platform to detect easily quantifiable, site-specific, disease-associated glycan alterations for clinical applications, we have adapted the multiple reaction monitoring mass spectrometry method for use in glycan biomarker research. The adaptations allow for highly precise site-specific glycan monitoring with minimum sample prep. Using this technique, we successfully mapped out the relative abundances of the most common 159 glycopeptides in the plasma of 97 healthy volunteers. This plasma glycome map revealed 796 significant (FDR < 0.05) site-specific inter-protein and intra-protein glycan associations, of which the vast majority were previously unknown. Since age and gender are relevant covariants in biomarker research, these variables were also characterized. 13 glycopeptides were found to be associated with gender and 41 to be associated with age. Using just five age-associated glycopeptides, a highly accurate age prediction model was constructed and validated (r2 = 0.62 ± 0.12). The human plasma site-specific glycan map described herein has utility in applications ranging from glycan biomarker research and discovery to the development of novel glycan-altering interventions.


Subject(s)
Age Factors , Biomarkers/blood , Polysaccharides/blood , Sex Factors , Adult , Aged , Aged, 80 and over , Blood Proteins , Female , Glycomics , Glycopeptides/blood , Glycosylation , Healthy Volunteers , Humans , Immunoglobulin G/blood , Likelihood Functions , Male , Middle Aged , Spectrometry, Mass, Electrospray Ionization , Young Adult
11.
Proc Natl Acad Sci U S A ; 117(14): 7633-7644, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32213588

ABSTRACT

Membrane-bound oligosaccharides form the interfacial boundary between the cell and its environment, mediating processes such as adhesion and signaling. These structures can undergo dynamic changes in composition and expression based on cell type, external stimuli, and genetic factors. Glycosylation, therefore, is a promising target of therapeutic interventions for presently incurable forms of advanced cancer. Here, we show that cholangiocarcinoma metastasis is characterized by down-regulation of the Golgi α-mannosidase I coding gene MAN1A1, leading to elevation of extended high-mannose glycans with terminating α-1,2-mannose residues. Subsequent reshaping of the glycome by inhibiting α-mannosidase I resulted in significantly higher migratory and invasive capabilities while masking cell surface mannosylation suppressed metastasis-related phenotypes. Exclusive elucidation of differentially expressed membrane glycoproteins and molecular modeling suggested that extended high-mannose glycosylation at the helical domain of transferrin receptor protein 1 promotes conformational changes that improve noncovalent interaction energies and lead to enhancement of cell migration in metastatic cholangiocarcinoma. The results provide support that α-1,2-mannosylated N-glycans present on cancer cell membrane proteins may serve as therapeutic targets for preventing metastasis.


Subject(s)
Cholangiocarcinoma/metabolism , Cholangiocarcinoma/pathology , Mannose/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Female , Glycosylation , Humans , Membrane Glycoproteins/metabolism , Mice , Models, Molecular , Neoplasm Metastasis , Phenotype , Protein Multimerization
12.
Stem Cells ; 38(2): 231-245, 2020 02.
Article in English | MEDLINE | ID: mdl-31648388

ABSTRACT

Therapeutic applications for mesenchymal stem/stromal cells (MSCs) are growing; however, the successful implementation of these therapies requires the development of appropriate MSC delivery systems. Hydrogels are ideally suited to cultivate MSCs but tuning hydrogel properties to match their specific in vivo applications remains a challenge. Thus, further characterization of how hydrogel-based delivery vehicles broadly influence MSC function and fate will help lead to the next generation of more intelligently designed delivery vehicles. To date, few attempts have been made to comprehensively characterize hydrogel impact on the MSC transcriptome. Herein, we have synthesized cell-degradable hydrogels based on bio-inert poly(ethylene glycol) tethered with specific integrin-binding small molecules and have characterized their resulting effect on the MSC transcriptome when compared with 2D cultured and untethered 3D hydrogel cultured MSCs. The 3D culture systems resulted in alterations in the MSC transcriptome, as is evident by the differential expression of genes related to extracellular matrix production, glycosylation, metabolism, signal transduction, gene epigenetic regulation, and development. For example, genes important for osteogenic differentiation were upregulated in 3D hydrogel cultures, and the expression of these genes could be partially suppressed by tethering an integrin-binding RGD peptide within the hydrogel. Highlighting the utility of tunable hydrogels, when applied to ex vivo human wounds the RGD-tethered hydrogel was able to support wound re-epithelialization, possibly due to its ability to increase PDGF expression and decrease IL-6 expression. These results will aid in future hydrogel design for a broad range of applications.


Subject(s)
Hydrogels/therapeutic use , Integrins/metabolism , Mesenchymal Stem Cells/drug effects , Transcriptome/drug effects , Wound Healing/drug effects , Cell Differentiation , Humans
13.
J Immunol ; 203(5): 1383-1391, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31331973

ABSTRACT

CD40L plays a major role in immune response and is a major therapeutic target for inflammation. Integrin α5ß1 and CD40 simultaneously bind to CD40L. It is unclear if α5ß1 and CD40 work together in CD40/CD40L signaling or how α5ß1 binds to CD40L. In this article, we describe that the integrin-binding site of human CD40L is predicted to be located in the trimeric interface by docking simulation. Mutations in the predicted integrin-binding site markedly reduced the binding of α5ß1 to CD40L. Several CD40L mutants defective in integrin binding were defective in NF-κB activation and B cell activation and suppressed CD40L signaling induced by wild-type CD40L; however, they still bound to CD40. These findings suggest that integrin α5ß1 binds to monomeric CD40L through the binding site in the trimeric interface of CD40L, and this plays a critical role in CD40/CD40L signaling. Integrin αvß3, a widely distributed vascular integrin, bound to CD40L in a KGD-independent manner, suggesting that αvß3 is a new CD40L receptor. Several missense mutations in CD40L that induce immunodeficiency with hyper-IgM syndrome type 1 (HIGM1) are clustered in the integrin-binding site of the trimeric interface. These HIGM1 CD40L mutants were defective in binding to α5ß1 and αvß3 (but not to CD40), suggesting that the defect in integrin binding may be a causal factor of HIGM1. These findings suggest that α5ß1 and αvß3 bind to the overlapping binding site in the trimeric interface of monomeric CD40L and generate integrin-CD40L-CD40 ternary complex. CD40L mutants defective in integrins have potential as antagonists of CD40/CD40L signaling.


Subject(s)
CD40 Antigens/metabolism , CD40 Ligand/metabolism , Integrin alpha5beta1/metabolism , Integrin alphaVbeta3/metabolism , Signal Transduction/physiology , Animals , Binding Sites/physiology , CHO Cells , Cell Line , Cell Line, Tumor , Cricetulus , HEK293 Cells , Humans , Hyper-IgM Immunodeficiency Syndrome, Type 1/metabolism , K562 Cells , Mutation/physiology , Protein Binding/physiology
14.
JCI Insight ; 4(7)2019 04 04.
Article in English | MEDLINE | ID: mdl-30944245

ABSTRACT

Actinic keratosis (AK) is a precancerous skin lesion that is common in HIV-positive patients. Without effective treatment, AKs can progress to squamous cell carcinoma. Ingenol mebutate, a PKC agonist, is a US Food and Drug Administration-approved (FDA-approved) topical treatment for AKs. It can induce reactivation of latent HIV transcription in CD4+ T cells both in vitro and ex vivo. Although PKC agonists are known to be potent inducers of HIV expression from latency, their effects in vivo are not known because of the concerns of toxicity. Therefore, we sought to determine the effects of topical ingenol mebutate gel on the HIV transcription profile in HIV-infected individuals with AKs, specifically in the setting of suppressive antiretroviral therapy (ART). We found that AKs cleared following topical application of ingenol mebutate and detected marginal changes in immune activation in the peripheral blood and in skin biopsies. An overall increase in the level of HIV transcription initiation, elongation, and complete transcription was detected only in skin biopsies after the treatment. Our data demonstrate that application of ingenol mebutate to AKs in ART-suppressed HIV-positive patients can effectively cure AKs as well as disrupt HIV latency in the skin tissue microenvironment in vivo without causing massive immune activation.


Subject(s)
Diterpenes/administration & dosage , HIV Infections/drug therapy , Keratosis, Actinic/drug therapy , Virus Latency/drug effects , Administration, Cutaneous , Anti-HIV Agents/therapeutic use , Biopsy , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , HIV Infections/complications , HIV Infections/immunology , HIV Infections/virology , HIV-1/genetics , HIV-1/immunology , Humans , Keratosis, Actinic/complications , Keratosis, Actinic/immunology , Keratosis, Actinic/pathology , Male , Middle Aged , Pilot Projects , Skin/drug effects , Skin/immunology , Skin/pathology , Transcriptional Activation/drug effects , Transcriptional Activation/immunology , Treatment Outcome , United States , Virus Activation/drug effects , Virus Activation/immunology
15.
Front Immunol ; 10: 589, 2019.
Article in English | MEDLINE | ID: mdl-31019502

ABSTRACT

The present paradigm of psoriasis pathogenesis revolves around the IL-23/IL-17A axis. Dual-secreting Th17 T cells presumably are the predominant sources of the psoriasis phenotype-driving cytokines, IL-17A and IL-22. We thus conducted a meta-analysis of independently acquired RNA-seq psoriasis datasets to explore the relationship between the expression of IL17A and IL22. This analysis failed to support the existence of dual secreting IL-17A/IL-22 Th17 cells as a major source of these cytokines. However, variable relationships amongst the expression of psoriasis susceptibility genes and of IL17A, IL22, and IL23A were identified. Additionally, to shed light on gene expression relationships in psoriasis, we applied a machine learning nonlinear dimensionality reduction strategy (t-SNE) to display the entire psoriasis transcriptome as a 2-dimensonal image. This analysis revealed a variety of gene clusters, relevant to psoriasis pathophysiology but failed to support a relationship between IL17A and IL22. These results support existing theories on alternative sources of IL-17A and IL-22 in psoriasis such as a Th22 cells and non-T cell populations.


Subject(s)
Interleukin-17/biosynthesis , Interleukins/biosynthesis , Psoriasis/etiology , Psoriasis/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Transcriptome , Biomarkers , Computational Biology/methods , Cytokines/biosynthesis , Disease Susceptibility , Gene Expression Profiling , Gene Expression Regulation , Humans , Inflammation Mediators/metabolism , Molecular Imaging , Psoriasis/pathology , Interleukin-22
16.
JCI Insight ; 3(13)2018 07 12.
Article in English | MEDLINE | ID: mdl-29997305

ABSTRACT

Numerous studies of relatively few patients have linked T cell receptor (TCR) genes to psoriasis but have yielded dramatically conflicting results. To resolve these discrepancies, we have chosen to mine RNA-Seq datasets for patterns of TCR gene segment usage in psoriasis. A meta-analysis of 3 existing and 1 unpublished datasets revealed a statistically significant link between the relative expression of TRAJ23 and psoriasis and the psoriasis-associated cytokine IL-17A. TRGV5, a TCR-γ segment, was also associated with psoriasis but correlated instead with IL-36A, other IL-36 family members, and IL-17C (not IL-17A). In contrast, TRAJ39 was strongly associated with healthy skin. T cell diversity measurements and analysis of CDR3 sequences were also conducted, revealing no psoriasis-associated public CDR3 sequences. Finally, in comparison with the expression of TCR-αß genes, the expression of TCR-γδ genes was relatively low but mildly elevated in psoriatic skin. These results have implications for the development of targeted therapies for psoriasis and other autoimmune diseases. Also, the techniques employed in this study have applications in other fields, such as cancer immunology and infectious disease.


Subject(s)
Genes, T-Cell Receptor beta/genetics , Interleukin-17/genetics , Psoriasis/genetics , Sequence Analysis, RNA/methods , Animals , Autoimmune Diseases/genetics , Cytokines/metabolism , Gene Expression Regulation , Genes, T-Cell Receptor beta/immunology , Humans , Interleukin-17/immunology , Mice , Psoriasis/immunology , Skin , Transcription Factors
17.
Immunity ; 48(1): 91-106.e6, 2018 01 16.
Article in English | MEDLINE | ID: mdl-29343444

ABSTRACT

CD103+ dendritic cells are critical for cross-presentation of tumor antigens. Here we have shown that during immunotherapy, large numbers of cells expressing CD103 arose in murine tumors via direct differentiation of Ly6c+ monocytic precursors. These Ly6c+CD103+ cells could derive from bone-marrow monocytic progenitors (cMoPs) or from peripheral cells present within the myeloid-derived suppressor cell (MDSC) population. Differentiation was controlled by inflammation-induced activation of the transcription factor p53, which drove upregulation of Batf3 and acquisition of the Ly6c+CD103+ phenotype. Mice with a targeted deletion of p53 in myeloid cells selectively lost the Ly6c+CD103+ population and became unable to respond to multiple forms of immunotherapy and immunogenic chemotherapy. Conversely, increasing p53 expression using a p53-agonist drug caused a sustained increase in Ly6c+CD103+ cells in tumors during immunotherapy, which markedly enhanced the efficacy and duration of response. Thus, p53-driven differentiation of Ly6c+CD103+ monocytic cells represents a potent and previously unrecognized target for immunotherapy.


Subject(s)
Antigen-Presenting Cells/physiology , Monocytes/physiology , Myeloid Cells/metabolism , Neoplasms/immunology , Tumor Suppressor Protein p53/metabolism , Animals , Antigen-Presenting Cells/immunology , Antigens, CD/metabolism , Antigens, Ly/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Flow Cytometry , Humans , Immunotherapy/methods , Integrin alpha Chains/metabolism , Mice , Monocytes/immunology , Myeloid Cells/physiology
18.
Mol Cancer Ther ; 16(11): 2627-2638, 2017 11.
Article in English | MEDLINE | ID: mdl-28847988

ABSTRACT

Primary effusion lymphoma (PEL) is an aggressive subtype of non-Hodgkin lymphoma caused by Kaposi's sarcoma-associated herpesvirus (KSHV) infection. Currently, treatment options for patients with PEL are limited. Oncolytic viruses have been engineered as anticancer agents and have recently shown increased therapeutic promise. Similarly, lytic activation of endogenous viruses from latently infected tumor cells can also be applied as a cancer therapy. In theory, such a therapeutic strategy would induce oncolysis by viral replication, while simultaneously stimulating an immune response to viral lytic cycle antigens. We examined the combination of the FDA-approved drug ingenol-3-angelate (PEP005) with epigenetic drugs as a rational therapeutic approach for KSHV-mediated malignancies. JQ1, a bromodomain and extra terminal (BET) protein inhibitor, in combination with PEP005, not only robustly induced KSHV lytic replication, but also inhibited IL6 production from PEL cells. Using the dosages of these agents that were found to be effective in reactivating HIV (as a means to clear latent virus with highly active antiretroviral therapy), we were able to inhibit PEL growth in vitro and delay tumor growth in a PEL xenograft tumor model. KSHV reactivation was mediated by activation of the NF-κB pathway by PEP005, which led to increased occupancy of RNA polymerase II onto the KSHV genome. RNA-sequencing analysis further revealed cellular targets of PEP005, JQ1, and the synergistic effects of both. Thus, combination of PEP005 with a BET inhibitor may be considered as a rational therapeutic approach for the treatment of PEL. Mol Cancer Ther; 16(11); 2627-38. ©2017 AACR.


Subject(s)
Azepines/administration & dosage , Diterpenes/administration & dosage , Lymphoma, Primary Effusion/drug therapy , Sarcoma, Kaposi/therapy , Triazoles/administration & dosage , Animals , Cell Line, Tumor , DNA Replication/drug effects , Herpesvirus 8, Human/drug effects , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/pathogenicity , Humans , Lymphoma, Primary Effusion/etiology , Lymphoma, Primary Effusion/genetics , Lymphoma, Primary Effusion/virology , Mice , NF-kappa B/genetics , Oncolytic Virotherapy/methods , Oncolytic Viruses/drug effects , Oncolytic Viruses/genetics , Oncolytic Viruses/pathogenicity , Sarcoma, Kaposi/complications , Sarcoma, Kaposi/genetics , Sarcoma, Kaposi/virology , Virus Replication/drug effects , Xenograft Model Antitumor Assays
19.
Sci Rep ; 7: 43728, 2017 03 13.
Article in English | MEDLINE | ID: mdl-28287093

ABSTRACT

The goal of this pilot study was to determine whether HDL glycoprotein composition affects HDL's immunomodulatory function. HDL were purified from healthy controls (n = 13), subjects with metabolic syndrome (MetS) (n = 13), and diabetic hemodialysis (HD) patients (n = 24). Concentrations of HDL-bound serum amyloid A (SAA), lipopolysaccharide binding protein (LBP), apolipoprotein A-I (ApoA-I), apolipoprotein C-III (ApoC-III), α-1-antitrypsin (A1AT), and α-2-HS-glycoprotein (A2HSG); and the site-specific glycovariations of ApoC-III, A1AT, and A2HSG were measured. Secretion of interleukin 6 (IL-6) in lipopolysaccharide-stimulated monocytes was used as a prototypical assay of HDL's immunomodulatory capacity. HDL from HD patients were enriched in SAA, LBP, ApoC-III, di-sialylated ApoC-III (ApoC-III2) and desialylated A2HSG. HDL that increased IL-6 secretion were enriched in ApoC-III, di-sialylated glycans at multiple A1AT glycosylation sites and desialylated A2HSG, and depleted in mono-sialylated ApoC-III (ApoC-III1). Subgroup analysis on HD patients who experienced an infectious hospitalization event within 60 days (HD+) (n = 12), vs. those with no event (HD-) (n = 12) showed that HDL from HD+ patients were enriched in SAA but had lower levels of sialylation across glycoproteins. Our results demonstrate that HDL glycoprotein composition, including the site-specific glycosylation, differentiate between clinical groups, correlate with HDL's immunomodulatory capacity, and may be predictive of HDL's ability to protect from infection.


Subject(s)
Interleukin-6/metabolism , Lipopolysaccharides/immunology , Lipoproteins, HDL/metabolism , Monocytes/immunology , Monocytes/metabolism , Biomarkers , Cytokines/metabolism , Diabetes Mellitus/immunology , Diabetes Mellitus/metabolism , Glycopeptides/metabolism , Glycoproteins/metabolism , Glycosylation , Humans , Infections/immunology , Infections/metabolism , Lipoproteins, HDL/isolation & purification , Renal Dialysis
20.
Front Immunol ; 8: 1980, 2017.
Article in English | MEDLINE | ID: mdl-29379508

ABSTRACT

BACKGROUND: Pyoderma gangrenosum (PG) is a debilitating ulcerative skin disease that is one of the most common associated diseases seen in patients with inflammatory bowel disease and rheumatoid arthritis. Although PG is classified as a neutrophilic dermatosis, its pathophysiology is poorly understood. OBJECTIVE: Use data obtained from patient-reported histories, immunohistochemistry, and gene expression analysis to formulate a hypothesis on PG pathophysiology. METHODS: Ten PG patients participated and answered questions about new ulcer formation. Skin biopsies of healed prior ulcers and adjacent normal skin were obtained from four patients for immunohistochemistry. Scars from healthy patients and patients with discoid lupus were used as additional controls. New onset PG papules were analyzed using immunohistochemistry and gene expression analysis via quantitative real-time PCR. RESULTS: All PG patients reported that healed sites of previous ulceration are refractory to re-ulceration. Simultaneous biopsies of healed and uninvolved skin triggered ulceration only in the latter. On immunohistochemistry, healed PG scars showed complete loss of pilosebaceous units, which were present in normal skin, and to a lesser extent in control scars, and discoid scars. Early PG papules showed perivascular and peripilosebaceous T cell infiltrates, rather than neutrophils. These early inflammatory events were dominated by increased gene expression of CXCL9, CXCL10, CXCL11, IL-8, IL-17, IFNG, and IL-36G and transcription factors consistent with Th1 phenotype. LIMITATIONS: Small sample size was the main limitation. CONCLUSION: We put forth the hypothesis that PG is a T cell response resulting in the destruction of pilosebaceous units.

SELECTION OF CITATIONS
SEARCH DETAIL
...