Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 98
Filter
Add more filters

Publication year range
1.
Immunity ; 57(3): 574-586.e7, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38430907

ABSTRACT

Continuously evolving influenza viruses cause seasonal epidemics and pose global pandemic threats. Although viral neuraminidase (NA) is an effective drug and vaccine target, our understanding of the NA antigenic landscape still remains incomplete. Here, we describe NA-specific human antibodies that target the underside of the NA globular head domain, inhibit viral propagation of a wide range of human H3N2, swine-origin variant H3N2, and H2N2 viruses, and confer both pre- and post-exposure protection against lethal H3N2 infection in mice. Cryo-EM structures of two such antibodies in complex with NA reveal non-overlapping epitopes covering the underside of the NA head. These sites are highly conserved among N2 NAs yet inaccessible unless the NA head tilts or dissociates. Our findings help guide the development of effective countermeasures against ever-changing influenza viruses by identifying hidden conserved sites of vulnerability on the NA underside.


Subject(s)
Influenza Vaccines , Influenza, Human , Orthomyxoviridae Infections , Humans , Animals , Mice , Swine , Viral Proteins/genetics , Neuraminidase , Influenza A Virus, H3N2 Subtype , Antibodies, Monoclonal , Antibodies, Viral
3.
Nat Immunol ; 20(3): 362-372, 2019 03.
Article in English | MEDLINE | ID: mdl-30742080

ABSTRACT

The present vaccine against influenza virus has the inevitable risk of antigenic discordance between the vaccine and the circulating strains, which diminishes vaccine efficacy. This necessitates new approaches that provide broader protection against influenza. Here we designed a vaccine using the hypervariable receptor-binding domain (RBD) of viral hemagglutinin displayed on a nanoparticle (np) able to elicit antibody responses that neutralize H1N1 influenza viruses spanning over 90 years. Co-display of RBDs from multiple strains across time, so that the adjacent RBDs are heterotypic, provides an avidity advantage to cross-reactive B cells. Immunization with the mosaic RBD-np elicited broader antibody responses than those induced by an admixture of nanoparticles encompassing the same set of RBDs as separate homotypic arrays. Furthermore, we identified a broadly neutralizing monoclonal antibody in a mouse immunized with mosaic RBD-np. The mosaic antigen array signifies a unique approach that subverts monotypic immunodominance and allows otherwise subdominant cross-reactive B cell responses to emerge.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/immunology , Influenza, Human/immunology , Nanoparticles/chemistry , Orthomyxoviridae Infections/immunology , Animals , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/virology , Cross Reactions/drug effects , Cross Reactions/immunology , Female , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Humans , Immunization , Influenza A Virus, H1N1 Subtype/metabolism , Influenza A Virus, H1N1 Subtype/physiology , Influenza Vaccines/administration & dosage , Influenza Vaccines/chemistry , Influenza, Human/prevention & control , Influenza, Human/virology , Mice, Inbred BALB C , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology
4.
Immunity ; 55(11): 2135-2148.e6, 2022 11 08.
Article in English | MEDLINE | ID: mdl-36306784

ABSTRACT

Epstein-Barr virus (EBV) is nearly ubiquitous in adults. EBV causes infectious mononucleosis and is associated with B cell lymphomas, epithelial cell malignancies, and multiple sclerosis. The EBV gH/gL glycoprotein complex facilitates fusion of virus membrane with host cells and is a target of neutralizing antibodies. Here, we examined the sites of vulnerability for virus neutralization and fusion inhibition within EBV gH/gL. We developed a panel of human monoclonal antibodies (mAbs) that targeted five distinct antigenic sites on EBV gH/gL and prevented infection of epithelial and B cells. Structural analyses using X-ray crystallography and electron microscopy revealed multiple sites of vulnerability and defined the antigenic landscape of EBV gH/gL. One mAb provided near-complete protection against viremia and lymphoma in a humanized mouse EBV challenge model. Our findings provide structural and antigenic knowledge of the viral fusion machinery, yield a potential therapeutic antibody to prevent EBV disease, and emphasize gH/gL as a target for herpesvirus vaccines and therapeutics.


Subject(s)
Epstein-Barr Virus Infections , Herpesvirus 4, Human , Cricetinae , Mice , Animals , Humans , Viral Envelope Proteins , Cricetulus , Membrane Glycoproteins , CHO Cells
5.
Immunity ; 52(5): 726-728, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32433944

ABSTRACT

Memory B cells (MBCs) expressing the transcription factor T-bet have been described in normal and dysregulated immune responses. In this issue of Immunity, Johnson et al. report that T-bet+ MBCs, formed in response to a primary influenza infection, contribute to protective antibody titers and persist mainly in the spleen with restricted trafficking between tissues.


Subject(s)
B-Lymphocyte Subsets , Animals , Antibody Specificity , B-Lymphocyte Subsets/metabolism , B-Lymphocytes/metabolism , Humans , Immunologic Memory , Mice , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Tissue Distribution
6.
Nature ; 614(7949): 752-761, 2023 02.
Article in English | MEDLINE | ID: mdl-36599369

ABSTRACT

Acute viral infections can have durable functional impacts on the immune system long after recovery, but how they affect homeostatic immune states and responses to future perturbations remain poorly understood1-4. Here we use systems immunology approaches, including longitudinal multimodal single-cell analysis (surface proteins, transcriptome and V(D)J sequences) to comparatively assess baseline immune statuses and responses to influenza vaccination in 33 healthy individuals after recovery from mild, non-hospitalized COVID-19 (mean, 151 days after diagnosis) and 40 age- and sex-matched control individuals who had never had COVID-19. At the baseline and independent of time after COVID-19, recoverees had elevated T cell activation signatures and lower expression of innate immune genes including Toll-like receptors in monocytes. Male individuals who had recovered from COVID-19 had coordinately higher innate, influenza-specific plasmablast, and antibody responses after vaccination compared with healthy male individuals and female individuals who had recovered from COVID-19, in part because male recoverees had monocytes with higher IL-15 responses early after vaccination coupled with elevated prevaccination frequencies of 'virtual memory'-like CD8+ T cells poised to produce more IFNγ after IL-15 stimulation. Moreover, the expression of the repressed innate immune genes in monocytes increased by day 1 to day 28 after vaccination in recoverees, therefore moving towards the prevaccination baseline of the healthy control individuals. By contrast, these genes decreased on day 1 and returned to the baseline by day 28 in the control individuals. Our study reveals sex-dimorphic effects of previous mild COVID-19 and suggests that viral infections in humans can establish new immunological set-points that affect future immune responses in an antigen-agnostic manner.


Subject(s)
COVID-19 , Immunity, Innate , Immunologic Memory , Influenza Vaccines , Sex Characteristics , T-Lymphocytes , Vaccination , Female , Humans , Male , CD8-Positive T-Lymphocytes/immunology , COVID-19/immunology , Influenza Vaccines/immunology , Influenza, Human/immunology , Influenza, Human/prevention & control , Interleukin-15/immunology , Toll-Like Receptors/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Monocytes , Immunity, Innate/genetics , Immunity, Innate/immunology , Single-Cell Analysis , Healthy Volunteers
7.
Immunity ; 51(2): 398-410.e5, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31350180

ABSTRACT

Vaccine-induced memory B cell responses to evolving viruses like influenza A involve activation of pre-existing immunity and generation of new responses. To define the contribution of these two types of responses, we analyzed the response to H7N9 vaccination in H7N9-naive adults. We performed comprehensive comparisons at the single-cell level of the kinetics, Ig repertoire, and activation phenotype of established pre-existing memory B cells recognizing conserved epitopes and the newly generated memory B cells directed toward H7 strain-specific epitopes. The recall response to conserved epitopes on H7 HA involved a transient expansion of memory B cells with little observed adaptation. However, the B cell response to newly encountered epitopes was phenotypically distinct and generated a sustained memory population that evolved and affinity matured months after vaccination. These findings establish clear differences between newly generated and pre-existing memory B cells, highlighting the challenges in achieving long-lasting, broad protection against an ever-evolving virus.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocytes/immunology , Influenza A Virus, H7N9 Subtype/physiology , Influenza Vaccines/immunology , Influenza, Human/immunology , Adult , Antibodies, Viral/metabolism , Antibody Formation , Cells, Cultured , Epitopes/immunology , Female , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Immunologic Memory , Lymphocyte Activation , Male , Middle Aged , Phenotype , Receptors, Antigen, B-Cell/genetics , Single-Cell Analysis , Vaccination , Young Adult
8.
Immunity ; 50(5): 1305-1316.e6, 2019 05 21.
Article in English | MEDLINE | ID: mdl-30979688

ABSTRACT

Epstein-Barr virus (EBV) causes infectious mononucleosis and is associated with epithelial-cell cancers and B cell lymphomas. An effective EBV vaccine is not available. We found that antibodies to the EBV glycoprotein gH/gL complex were the principal components in human plasma that neutralized infection of epithelial cells and that antibodies to gH/gL and gp42 contributed to B cell neutralization. Immunization of mice and nonhuman primates with nanoparticle vaccines that displayed components of the viral-fusion machinery EBV gH/gL or gH/gL/gp42 elicited antibodies that potently neutralized both epithelial-cell and B cell infection. Immune serum from nonhuman primates inhibited EBV-glycoprotein-mediated fusion of epithelial cells and B cells and targeted an epitope critical for virus-cell fusion. Therefore, unlike the leading EBV gp350 vaccine candidate, which only protects B cells from infection, these EBV nanoparticle vaccines elicit antibodies that inhibit the virus-fusion apparatus and provide cell-type-independent protection from virus infection.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , B-Lymphocytes/immunology , Epithelial Cells/immunology , Epstein-Barr Virus Infections/prevention & control , Herpesvirus 4, Human/immunology , Membrane Glycoproteins/immunology , Viral Envelope Proteins/immunology , Animals , B-Lymphocytes/virology , CHO Cells , Cell Fusion , Cell Line, Tumor , Cricetulus , Epithelial Cells/virology , Epstein-Barr Virus Infections/immunology , Female , HEK293 Cells , HeLa Cells , Humans , Immune Sera/administration & dosage , Macaca fascicularis , Male , Mice , Mice, Inbred BALB C , Vaccines, Virus-Like Particle/immunology , Viral Vaccines/immunology , Virus Attachment
9.
Genes Dev ; 33(5-6): 310-332, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30804224

ABSTRACT

Whether cell types exposed to a high level of environmental insults possess cell type-specific prosurvival mechanisms or enhanced DNA damage repair capacity is not well understood. BRN2 is a tissue-restricted POU domain transcription factor implicated in neural development and several cancers. In melanoma, BRN2 plays a key role in promoting invasion and regulating proliferation. Here we found, surprisingly, that rather than interacting with transcription cofactors, BRN2 is instead associated with DNA damage response proteins and directly binds PARP1 and Ku70/Ku80. Rapid PARP1-dependent BRN2 association with sites of DNA damage facilitates recruitment of Ku80 and reprograms DNA damage repair by promoting Ku-dependent nonhomologous end-joining (NHEJ) at the expense of homologous recombination. BRN2 also suppresses an apoptosis-associated gene expression program to protect against UVB-, chemotherapy- and vemurafenib-induced apoptosis. Remarkably, BRN2 expression also correlates with a high single-nucleotide variation prevalence in human melanomas. By promoting error-prone DNA damage repair via NHEJ and suppressing apoptosis of damaged cells, our results suggest that BRN2 contributes to the generation of melanomas with a high mutation burden. Our findings highlight a novel role for a key transcription factor in reprogramming DNA damage repair and suggest that BRN2 may impact the response to DNA-damaging agents in BRN2-expressing cancers.


Subject(s)
Apoptosis , DNA End-Joining Repair/genetics , Homeodomain Proteins/metabolism , Melanoma/genetics , Melanoma/physiopathology , Mutation/genetics , POU Domain Factors/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/genetics , Homeodomain Proteins/genetics , Humans , Ku Autoantigen/metabolism , POU Domain Factors/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Protein Binding , Protein Domains , Protein Transport
10.
PLoS Pathog ; 19(8): e1011514, 2023 08.
Article in English | MEDLINE | ID: mdl-37639457

ABSTRACT

Despite the availability of seasonal vaccines and antiviral medications, influenza virus continues to be a major health concern and pandemic threat due to the continually changing antigenic regions of the major surface glycoprotein, hemagglutinin (HA). One emerging strategy for the development of more efficacious seasonal and universal influenza vaccines is structure-guided design of nanoparticles that display conserved regions of HA, such as the stem. Using the H1 HA subtype to establish proof of concept, we found that tandem copies of an alpha-helical fragment from the conserved stem region (helix-A) can be displayed on the protruding spikes structures of a capsid scaffold. The stem region of HA on these designed chimeric nanoparticles is immunogenic and the nanoparticles are biochemically robust in that heat exposure did not destroy the particles and immunogenicity was retained. Furthermore, mice vaccinated with H1-nanoparticles were protected from lethal challenge with H1N1 influenza virus. By using a nanoparticle library approach with this helix-A nanoparticle design, we show that this vaccine nanoparticle construct design could be applicable to different influenza HA subtypes. Importantly, antibodies elicited by H1, H5, and H7 nanoparticles demonstrated homosubtypic and heterosubtypic cross-reactivity binding to different HA subtypes. Also, helix-A nanoparticle immunizations were used to isolate mouse monoclonal antibodies that demonstrated heterosubtypic cross-reactivity and provided protection to mice from viral challenge via passive-transfer. This tandem helix-A nanoparticle construct represents a novel design to display several hundred copies of non-trimeric conserved HA stem epitopes on vaccine nanoparticles. This design concept provides a new approach to universal influenza vaccine development strategies and opens opportunities for the development of nanoparticles with broad coverage over many antigenically diverse influenza HA subtypes.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza Vaccines , Influenza, Human , Nanoparticles , Animals , Mice , Humans , Hemagglutinins , Epitopes , Antibody Formation
11.
Alcohol Alcohol ; 59(1)2024 Jan 11.
Article in English | MEDLINE | ID: mdl-37873970

ABSTRACT

Increased alcohol consumption during the coronavirus disease 2019 pandemic is projected to impact alcohol-related liver disease (ALD) morbidity and mortality. Inter-hospital escalation-of-care referral requests to our tertiary-care hepatology unit were analyzed from January 2020 through December 2022. Most requests to our center were for ALD with an increase in requests from intermediate care units, suggestive of higher acuity illness.


Subject(s)
COVID-19 , Liver Diseases, Alcoholic , Humans , Liver Diseases, Alcoholic/epidemiology , Liver Diseases, Alcoholic/therapy , Alcohol Drinking/epidemiology , Pandemics , COVID-19/epidemiology , Referral and Consultation , Hospitals
12.
Proc Natl Acad Sci U S A ; 118(2)2021 01 12.
Article in English | MEDLINE | ID: mdl-33384332

ABSTRACT

Thrombopoietin (TPO) and the TPO-receptor (TPO-R, or c-MPL) are essential for hematopoietic stem cell (HSC) maintenance and megakaryocyte differentiation. Agents that can modulate TPO-R signaling are highly desirable for both basic research and clinical utility. We developed a series of surrogate protein ligands for TPO-R, in the form of diabodies (DBs), that homodimerize TPO-R on the cell surface in geometries that are dictated by the DB receptor binding epitope, in effect "tuning" downstream signaling responses. These surrogate ligands exhibit diverse pharmacological properties, inducing graded signaling outputs, from full to partial TPO agonism, thus decoupling the dual functions of TPO/TPO-R. Using single-cell RNA sequencing and HSC self-renewal assays we find that partial agonistic diabodies preserved the stem-like properties of cultured HSCs, but also blocked oncogenic colony formation in essential thrombocythemia (ET) through inverse agonism. Our data suggest that dampening downstream TPO signaling is a powerful approach not only for HSC preservation in culture, but also for inhibiting oncogenic signaling through the TPO-R.


Subject(s)
Receptors, Thrombopoietin/metabolism , Thrombopoietin/metabolism , Cell Differentiation/physiology , Cell Membrane/metabolism , Epitopes/immunology , Hematopoiesis/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Ligands , Megakaryocytes/metabolism , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Cytokine/metabolism , Receptors, Thrombopoietin/immunology , Receptors, Thrombopoietin/physiology , Signal Transduction/physiology , Thrombocythemia, Essential/metabolism , Thrombopoietin/physiology
13.
Liver Transpl ; 29(7): 757-767, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37016758

ABSTRACT

BACKGROUND: Alcohol accounts for a large disease burden in hepatology and liver transplantation (LT) and across the globe. Clinical evaluations and decisions about LT candidacy are challenging because they rely on detailed psychosocial assessments and interpretations of psychiatric and substance use disorder data, which often must occur rapidly according to the acuity of end-stage liver disease. Such difficulties commonly occur during the process of candidate selection and liver allocation, particularly during early LT (eLT) in patients with acute alcohol-associated hepatitis (AAH). Patients with AAH commonly have very recent or active substance use, high short-term mortality, psychiatric comorbidities, and compressed evaluation and treatment timetables. LT clinicians report that patients' alcohol-associated insight (AAI) is among the most relevant psychosocial data in this population, yet no studies exist examining how LT teams define and use AAI in eLT or its effect on clinical outcomes. In April 2022, we searched Ovid MEDLINE, Elsevier Embase, EBSCOhost PsycInfo and CINAHL, and Wiley Cochrane Central Register of Controlled Trials for reports describing AAH populations who underwent eLT, which also described psychosocial evaluation parameters. The searches retrieved 1603 unique reports. After eligibility screening, 8 were included in the qualitative analysis. This systematic review reveals that AAI is a poorly defined construct that is not measured in a standardized way. Yet it is a commonly cited parameter in articles that describe the psychosocial evaluation and decision-making of patients undergoing eLT for AAH. This article also discusses the general challenges of assessing AAI during eLT for AAH, existing AAI definitions and rating scales, how AAI has been used to date in the broader hepatology and LT literature, and future areas for clinical and research progress.


Subject(s)
Hepatitis, Alcoholic , Liver Transplantation , Humans , Liver Transplantation/adverse effects , Hepatitis, Alcoholic/diagnosis , Hepatitis, Alcoholic/surgery , Comorbidity
14.
Proc Natl Acad Sci U S A ; 115(37): E8668-E8677, 2018 09 11.
Article in English | MEDLINE | ID: mdl-30150413

ABSTRACT

The close integration of the MAPK, PI3K, and WNT signaling pathways underpins much of development and is deregulated in cancer. In principle, combinatorial posttranslational modification of key lineage-specific transcription factors would be an effective means to integrate critical signaling events. Understanding how this might be achieved is central to deciphering the impact of microenvironmental cues in development and disease. The microphthalmia-associated transcription factor MITF plays a crucial role in the development of melanocytes, the retinal pigment epithelium, osteoclasts, and mast cells and acts as a lineage survival oncogene in melanoma. MITF coordinates survival, differentiation, cell-cycle progression, cell migration, metabolism, and lysosome biogenesis. However, how the activity of this key transcription factor is controlled remains poorly understood. Here, we show that GSK3, downstream from both the PI3K and Wnt pathways, and BRAF/MAPK signaling converges to control MITF nuclear export. Phosphorylation of the melanocyte MITF-M isoform in response to BRAF/MAPK signaling primes for phosphorylation by GSK3, a kinase inhibited by both PI3K and Wnt signaling. Dual phosphorylation, but not monophosphorylation, then promotes MITF nuclear export by activating a previously unrecognized hydrophobic export signal. Nonmelanocyte MITF isoforms exhibit poor regulation by MAPK signaling, but instead their export is controlled by mTOR. We uncover here an unanticipated mode of MITF regulation that integrates the output of key developmental and cancer-associated signaling pathways to gate MITF flux through the import-export cycle. The results have significant implications for our understanding of melanoma progression and stem cell renewal.


Subject(s)
Cell Nucleus/metabolism , Glycogen Synthase Kinase 3/metabolism , MAP Kinase Signaling System , Microphthalmia-Associated Transcription Factor/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Active Transport, Cell Nucleus , Animals , Cell Line, Tumor , Cells, Cultured , HeLa Cells , Humans , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Microphthalmia-Associated Transcription Factor/genetics , Mutation , Phosphorylation , Protein Binding
15.
Circulation ; 140(6): 500-513, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31163988

ABSTRACT

BACKGROUND: Genome-wide association studies have identified chromosome 14q32 as a locus for coronary artery disease. The disease-associated variants fall in a hitherto uncharacterized gene called HHIPL1 (hedgehog interacting protein-like 1), which encodes a sequence homolog of an antagonist of hedgehog signaling. The function of HHIPL1 and its role in atherosclerosis are unknown. METHODS: HHIPL1 cellular localization, interaction with sonic hedgehog (SHH), and influence on hedgehog signaling were tested. HHIPL1 expression was measured in coronary artery disease-relevant human cells, and protein localization was assessed in wild-type and Apoe-/- (apolipoprotein E deficient) mice. Human aortic smooth muscle cell phenotypes and hedgehog signaling were investigated after gene knockdown. Hhipl1-/- mice were generated and aortic smooth muscle cells collected for phenotypic analysis and assessment of hedgehog signaling activity. Hhipl1-/- mice were bred onto both the Apoe-/- and Ldlr-/- (low-density lipoprotein receptor deficient) knockout strains, and the extent of atherosclerosis was quantified after 12 weeks of high-fat diet. Cellular composition and collagen content of aortic plaques were assessed by immunohistochemistry. RESULTS: In vitro analyses revealed that HHIPL1 is a secreted protein that interacts with SHH and increases hedgehog signaling activity. HHIPL1 expression was detected in human smooth muscle cells and in smooth muscle within atherosclerotic plaques of Apoe-/- mice. The expression of Hhipl1 increased with disease progression in aortic roots of Apoe-/- mice. Proliferation and migration were reduced in Hhipl1 knockout mouse and HHIPL1 knockdown aortic smooth muscle cells, and hedgehog signaling was decreased in HHIPL1-deficient cells. Hhipl1 knockout caused a reduction of >50% in atherosclerosis burden on both Apoe-/- and Ldlr-/- knockout backgrounds, and lesions were characterized by reduced smooth muscle cell content. CONCLUSIONS: HHIPL1 is a secreted proatherogenic protein that enhances hedgehog signaling and regulates smooth muscle cell proliferation and migration. Inhibition of HHIPL1 protein function might offer a novel therapeutic strategy for coronary artery disease.


Subject(s)
Atherosclerosis/genetics , Chromosomes, Human, Pair 14/genetics , Coronary Disease/genetics , Hedgehog Proteins/physiology , Intercellular Signaling Peptides and Proteins/physiology , Animals , Atherosclerosis/pathology , Cell Division , Cell Movement , Cells, Cultured , Humans , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout, ApoE , Myocytes, Smooth Muscle/metabolism , Plaque, Atherosclerotic/pathology , Receptors, LDL/deficiency , Signal Transduction
16.
Clin Immunol ; 215: 108440, 2020 06.
Article in English | MEDLINE | ID: mdl-32330555

ABSTRACT

Perinatally HIV-infected children (PHIV), despite successful antiretroviral therapy, present suboptimal responses to vaccinations compared to healthy-controls (HC). Here we investigated phenotypic and transcriptional signatures of H1N1-specific B-cells (H1N1-Sp) in PHIV, differentially responding to trivalent-influenza-vaccine (TIV), and HC. Patients were categorized in responders (R) and non-responders (NR) according to hemagglutination-inhibition-assay at baseline and 21 days after TIV. No differences in H1N1-Sp frequencies were found between groups. H1N1-Sp transcriptional analysis revealed a distinct signature between PHIV and HC. NR presented higher PIK3C2B and NOD2 expression compared to R, confirmed by downregulation of PIK3C2B in resting-memory of R after H1N1 in-vitro stimulation. In conclusion this study confirms that qualitative rather than quantitative analyses are needed to characterize immune responses in PHIV. These results further suggest that higher PIK3C2B in H1N1-Sp of NR is associated with lower H1N1 immunogenicity and may be targeted by future modulating strategies to improve TIV responses in PHIV.


Subject(s)
B-Lymphocytes/immunology , Class II Phosphatidylinositol 3-Kinases/immunology , Gene Expression/immunology , HIV Infections/immunology , Immunogenicity, Vaccine/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/immunology , Influenza, Human/immunology , Adolescent , Antibodies, Viral/immunology , Class II Phosphatidylinositol 3-Kinases/genetics , Down-Regulation/genetics , Down-Regulation/immunology , Female , Gene Expression/genetics , Hemagglutination Inhibition Tests/methods , Humans , Male , Transcription, Genetic/genetics , Transcription, Genetic/immunology , Vaccination/methods
17.
Psychosomatics ; 61(5): 450-455, 2020.
Article in English | MEDLINE | ID: mdl-32665149

ABSTRACT

BACKGROUND: The scientific literature in consultation-liaison psychiatry continually expands, and remaining familiar with the most current literature is challenging for practicing clinicians. The Guidelines and Evidence-Based Medicine Subcommittee of the Academy of Consultation-Liaison Psychiatry writes quarterly annotations of articles of interest to help Academy members gain familiarity with the most current evidence-based practices. These annotations are available on the Academy Website. OBJECTIVE: We identify the 10 most important manuscripts for clinical practice in consultation-liaison psychiatry from 2019. METHODS: Sixty-four abstracts were authored in 2019. Manuscripts were rated on clinical relevance to practice and quality of scholarship. The 10 articles with the highest aggregate scores from 19 raters are described. RESULTS: The resulting articles provide practical guidance for consultation psychiatrists on several topic areas including the treatment of substance use disorders. CONCLUSION: We suggest that these clinical findings should be familiar to all consultation-liaison psychiatrists regardless of practice area. Regular article reviews and summaries help busy clinicians deliver cutting-edge care and maintain a high standard of care across the specialty.


Subject(s)
Mental Disorders/therapy , Mental Health Services/organization & administration , Psychiatry/organization & administration , Referral and Consultation , Humans
18.
J Wound Ostomy Continence Nurs ; 47(6): 619-621, 2020.
Article in English | MEDLINE | ID: mdl-33201148

ABSTRACT

BACKGROUND: COVID-19 symptoms mimic many other common conditions, making it difficult to identify patients infected with COVID-19. Adult patients may exhibit what is called "COVID toes" later in their course of illness. Noteworthy is that COVID toes may be the only symptom in otherwise asymptomatic young adults and children. CASE: We present experience with an adult female patient residing in the United States with presumed COVID-19 infection who was self-isolating with illness not severe enough to require hospitalization. COVID toes were one symptom experienced as a part of her illness. Because COVID toes are usually seen in the recovery phase of COVID-19, and often in patients with less severe cases, many providers are unfamiliar with this new symptom. CONCLUSION: Familiarity with COVID toes will help foot care providers to identify patients who may not have been diagnosed with COVID-19 but could still be infectious and need to be isolated. COVID toe signs may be subtle and often mimic other conditions such as chilblains/pernio, thus a thorough history and physical examination are required.


Subject(s)
Betacoronavirus , Chilblains/diagnosis , Chilblains/virology , Coronavirus Infections/complications , Pneumonia, Viral/complications , Toes , COVID-19 , Chilblains/therapy , Coronavirus Infections/diagnosis , Coronavirus Infections/therapy , Female , Humans , Middle Aged , Pandemics , Pneumonia, Viral/diagnosis , Pneumonia, Viral/therapy , SARS-CoV-2
19.
J Virol ; 92(9)2018 05 01.
Article in English | MEDLINE | ID: mdl-29444938

ABSTRACT

Pandemic live attenuated influenza vaccines (pLAIV) prime subjects for a robust neutralizing antibody response upon subsequent administration of a pandemic inactivated subunit vaccine (pISV). However, a difference was not detected in H5-specific memory B cells in the peripheral blood between pLAIV-primed and unprimed subjects prior to pISV boost. To investigate the mechanism underlying pLAIV priming, we vaccinated groups of 12 African green monkeys (AGMs) with H5N1 pISV or pLAIV alone or H5N1 pLAIV followed by pISV and examined immunity systemically and in local draining lymph nodes (LN). The AGM model recapitulated the serologic observations from clinical studies. Interestingly, H5N1 pLAIV induced robust germinal center B cell responses in the mediastinal LN (MLN). Subsequent boosting with H5N1 pISV drove increases in H5-specific B cells in the axillary LN, spleen, and circulation in H5N1 pLAIV-primed animals. Thus, H5N1 pLAIV primes localized B cell responses in the MLN that are recalled systemically following pISV boost. These data provide mechanistic insights for the generation of robust humoral responses via prime-boost vaccination.IMPORTANCE We have previously shown that pandemic live attenuated influenza vaccines (pLAIV) prime for a rapid and robust antibody response on subsequent administration of inactivated subunit vaccine (pISV). This is observed even in individuals who had undetectable antibody (Ab) responses following the initial vaccination. To define the mechanistic basis of pLAIV priming, we turned to a nonhuman primate model and performed a detailed analysis of B cell responses in systemic and local lymphoid tissues following prime-boost vaccination with pLAIV and pISV. We show that the nonhuman primate model recapitulates the serologic observations from clinical studies. Further, we found that pLAIVs induced robust germinal center B cell responses in the mediastinal lymph node. Subsequent boosting with pISV in pLAIV-primed animals resulted in detection of B cells in the axillary lymph nodes, spleen, and peripheral blood. We demonstrate that intranasally administered pLAIV elicits a highly localized germinal center B cell response in the mediastinal lymph node that is rapidly recalled following pISV boost into germinal center reactions at numerous distant immune sites.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , B-Lymphocytes/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/immunology , Vaccines, Attenuated/immunology , Vaccines, Subunit/immunology , Administration, Intranasal , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Chlorocebus aethiops , Female , Humans , Influenza, Human/prevention & control , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymphocyte Count , Male , Vaccination
20.
Arterioscler Thromb Vasc Biol ; 38(8): 1711-1722, 2018 08.
Article in English | MEDLINE | ID: mdl-29794114

ABSTRACT

Objective- A large number of genetic loci have been associated with risk of coronary artery disease (CAD) through genome-wide association studies, however, for most loci the underlying biological mechanism is unknown. Determining the molecular pathways and cellular processes affected by these loci will provide new insights into CAD pathophysiology and may lead to new therapies. The CAD-associated variants at 10p11.23 fall in JCAD, which encodes an endothelial junction protein, however, its molecular function in endothelial cells is not known. In this study, we characterize the molecular role of JCAD (junctional cadherin 5 associated) in endothelial cells. Approach and Results- We show that JCAD knockdown in endothelial cells affects key phenotypes related to atherosclerosis including proliferation, migration, apoptosis, tube formation, and monocyte binding. We demonstrate that JCAD interacts with LATS2 (large tumor suppressor kinase 2) and negatively regulates Hippo signaling leading to increased activity of YAP (yes-associated protein), the transcriptional effector of the pathway. We also show by double siRNA knockdown that the phenotypes caused by JCAD knockdown require LATS2 and that JCAD is involved in transmission of RhoA-mediated signals into the Hippo pathway. In human tissues, we find that the CAD-associated lead variant, rs2487928, is associated with expression of JCAD in arteries, including atherosclerotic arteries. Gene co-expression analyses across disease-relevant tissues corroborate our phenotypic findings and support the link between JCAD and Hippo signaling. Conclusions- Our results show that JCAD negatively regulates Hippo signaling in endothelial cells and we suggest that JCAD contributes to atherosclerosis by mediating YAP activity and contributing to endothelial dysfunction.


Subject(s)
Cell Adhesion Molecules/metabolism , Coronary Artery Disease/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Neovascularization, Physiologic , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis , Cell Adhesion , Cell Adhesion Molecules/genetics , Cell Movement , Cell Proliferation , Coculture Techniques , Coronary Artery Disease/genetics , Coronary Artery Disease/pathology , HEK293 Cells , Hippo Signaling Pathway , Human Umbilical Vein Endothelial Cells/pathology , Humans , Monocytes/metabolism , Phenotype , Phosphoproteins/metabolism , Polymorphism, Single Nucleotide , Protein Serine-Threonine Kinases/genetics , THP-1 Cells , Transcription Factors , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , YAP-Signaling Proteins , rhoA GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL