Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Cell ; 185(4): 614-629.e21, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35148840

ABSTRACT

Activation of the innate immune system via pattern recognition receptors (PRRs) is key to generate lasting adaptive immunity. PRRs detect unique chemical patterns associated with invading microorganisms, but whether and how the physical properties of PRR ligands influence the development of the immune response remains unknown. Through the study of fungal mannans, we show that the physical form of PRR ligands dictates the immune response. Soluble mannans are immunosilent in the periphery but elicit a potent pro-inflammatory response in the draining lymph node (dLN). By modulating the physical form of mannans, we developed a formulation that targets both the periphery and the dLN. When combined with viral glycoprotein antigens, this mannan formulation broadens epitope recognition, elicits potent antigen-specific neutralizing antibodies, and confers protection against viral infections of the lung. Thus, the physical properties of microbial ligands determine the outcome of the immune response and can be harnessed for vaccine development.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antigens, Viral/immunology , Candida albicans/chemistry , Mannans/immunology , Aluminum Hydroxide/chemistry , Animals , Antibodies, Neutralizing/immunology , Antibody Specificity/immunology , B-Lymphocytes/immunology , COVID-19/immunology , COVID-19/prevention & control , COVID-19/virology , Chlorocebus aethiops , Epitopes/immunology , Immunity, Innate , Immunization , Inflammation/pathology , Interferons/metabolism , Lectins, C-Type/metabolism , Ligands , Lung/immunology , Lung/pathology , Lung/virology , Lymph Nodes/immunology , Lymph Nodes/metabolism , Macrophages/metabolism , Mice, Inbred C57BL , Paranasal Sinuses/metabolism , Protein Subunits/metabolism , Sialic Acid Binding Ig-like Lectin 1/metabolism , Solubility , Spike Glycoprotein, Coronavirus/metabolism , T-Lymphocytes/immunology , Transcription Factor RelB/metabolism , Vero Cells , beta-Glucans/metabolism
2.
Nature ; 604(7904): 134-140, 2022 04.
Article in English | MEDLINE | ID: mdl-35130559

ABSTRACT

The SARS-CoV-2 virus has infected more than 261 million people and has led to more than 5 million deaths in the past year and a half1 ( https://www.who.org/ ). Individuals with SARS-CoV-2 infection typically develop mild-to-severe flu-like symptoms, whereas infection of a subset of individuals leads to severe-to-fatal clinical outcomes2. Although vaccines have been rapidly developed to combat SARS-CoV-2, there has been a dearth of antiviral therapeutics. There is an urgent need for therapeutics, which has been amplified by the emerging threats of variants that may evade vaccines. Large-scale efforts are underway to identify antiviral drugs. Here we screened approximately 18,000 drugs for antiviral activity using live virus infection in human respiratory cells and validated 122 drugs with antiviral activity and selectivity against SARS-CoV-2. Among these candidates are 16 nucleoside analogues, the largest category of clinically used antivirals. This included the antivirals remdesivir and molnupiravir, which have been approved for use in COVID-19. RNA viruses rely on a high supply of nucleoside triphosphates from the host to efficiently replicate, and we identified a panel of host nucleoside biosynthesis inhibitors as antiviral. Moreover, we found that combining pyrimidine biosynthesis inhibitors with antiviral nucleoside analogues synergistically inhibits SARS-CoV-2 infection in vitro and in vivo against emerging strains of SARS-CoV-2, suggesting a clinical path forward.


Subject(s)
Antiviral Agents , Drug Evaluation, Preclinical , Nucleosides , Pyrimidines , SARS-CoV-2 , Adenosine Monophosphate/analogs & derivatives , Alanine/analogs & derivatives , Antiviral Agents/pharmacology , COVID-19/virology , Cell Line , Cytidine/analogs & derivatives , Humans , Hydroxylamines , Nucleosides/analogs & derivatives , Nucleosides/pharmacology , Pyrimidines/pharmacology , SARS-CoV-2/drug effects , COVID-19 Drug Treatment
3.
PLoS Pathog ; 20(5): e1011669, 2024 May.
Article in English | MEDLINE | ID: mdl-38781259

ABSTRACT

The virus severe acute respiratory syndrome coronavirus 2, SARS-CoV-2, is the causative agent of the current COVID-19 pandemic. It possesses a large 30 kilobase (kb) genome that encodes structural, non-structural, and accessory proteins. Although not necessary to cause disease, these accessory proteins are known to influence viral replication and pathogenesis. Through the synthesis of novel infectious clones of SARS-CoV-2 that lack one or more of the accessory proteins of the virus, we have found that one of these accessory proteins, ORF8, is critical for the modulation of the host inflammatory response. Mice infected with a SARS-CoV-2 virus lacking ORF8 exhibit increased weight loss and exacerbated macrophage infiltration into the lungs. Additionally, infection of mice with recombinant SARS-CoV-2 viruses encoding ORF8 mutations found in variants of concern reveal that naturally occurring mutations in this protein influence disease severity. Our studies with a virus lacking this ORF8 protein and viruses possessing naturally occurring point mutations in this protein demonstrate that this protein impacts pathogenesis.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , SARS-CoV-2/genetics , COVID-19/virology , COVID-19/immunology , COVID-19/pathology , COVID-19/genetics , Mice , Humans , Disease Progression , Viral Proteins/genetics , Viral Proteins/metabolism , Lung/virology , Lung/pathology , Virus Replication , Pneumonia/virology , Pneumonia/pathology , Chlorocebus aethiops , Mutation , Vero Cells , Female
4.
Proc Natl Acad Sci U S A ; 119(37): e2204717119, 2022 09 13.
Article in English | MEDLINE | ID: mdl-36040867

ABSTRACT

The ongoing COVID-19 pandemic is a major public health crisis. Despite the development and deployment of vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the pandemic persists. The continued spread of the virus is largely driven by the emergence of viral variants, which can evade the current vaccines through mutations in the spike protein. Although these differences in spike are important in terms of transmission and vaccine responses, these variants possess mutations in the other parts of their genome that may also affect pathogenesis. Of particular interest to us are the mutations present in the accessory genes, which have been shown to contribute to pathogenesis in the host through interference with innate immune signaling, among other effects on host machinery. To examine the effects of accessory protein mutations and other nonspike mutations on SARS-CoV-2 pathogenesis, we synthesized both viruses possessing deletions in the accessory genes as well as viruses where the WA-1 spike is replaced by each variant spike gene in a SARS-CoV-2/WA-1 infectious clone. We then characterized the in vitro and in vivo replication of these viruses and compared them to both WA-1 and the full variant viruses. Our work has revealed that the accessory proteins contribute to SARS-CoV-2 pathogenesis and the nonspike mutations in variants can contribute to replication of SARS-CoV-2 and pathogenesis in the host. This work suggests that while spike mutations may enhance receptor binding and entry into cells, mutations in accessory proteins may alter clinical disease presentation.


Subject(s)
COVID-19 , Mutation , SARS-CoV-2 , Viral Regulatory and Accessory Proteins , Virulence , COVID-19/virology , Humans , SARS-CoV-2/classification , SARS-CoV-2/genetics , SARS-CoV-2/pathogenicity , Spike Glycoprotein, Coronavirus/genetics , Viral Regulatory and Accessory Proteins/genetics , Virulence/genetics , Virus Replication/genetics
5.
J Virol ; 97(11): e0133623, 2023 Nov 30.
Article in English | MEDLINE | ID: mdl-37846985

ABSTRACT

IMPORTANCE: Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has caused a wide spectrum of diseases in the human population, from asymptomatic infections to death. It is important to study the host differences that may alter the pathogenesis of this virus. One clinical finding in coronavirus disease 2019 (COVID-19) patients is that people with obesity or diabetes are at increased risk of severe illness from SARS-CoV-2 infection. We used a high-fat diet model in mice to study the effects of obesity and type 2 diabetes on SARS-CoV-2 infection as well as how these comorbidities alter the response to vaccination. We find that diabetic/obese mice have increased disease after SARS-CoV-2 infection and they have slower clearance of the virus. We find that the lungs of these mice have increased neutrophils and that removing these neutrophils protects diabetic/obese mice from disease. This demonstrates why these diseases have increased risk of severe disease and suggests specific interventions upon infection.


Subject(s)
COVID-19 Vaccines , Diabetes Mellitus, Type 2 , Obesity , Vaccine Efficacy , Animals , Humans , Mice , COVID-19/prevention & control , Diabetes Mellitus, Type 2/complications , Diet , Mice, Obese , Obesity/complications , SARS-CoV-2 , COVID-19 Vaccines/administration & dosage
6.
J Allergy Clin Immunol ; 152(5): 1107-1120.e6, 2023 11.
Article in English | MEDLINE | ID: mdl-37595760

ABSTRACT

BACKGROUND: Obesity and type 2 diabetes mellitus (T2DM) are associated with an increased risk of severe outcomes from infectious diseases, including coronavirus disease 2019. These conditions are also associated with distinct responses to immunization, including an impaired response to widely used severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) mRNA vaccines. OBJECTIVE: We sought to establish a connection between reduced immunization efficacy via modeling the effects of metabolic diseases on vaccine immunogenicity that is essential for the development of more effective vaccines for this distinct vulnerable population. METHODS: A murine model of diet-induced obesity and insulin resistance was used to model the effects of comorbid T2DM and obesity on vaccine immunogenicity and protection. RESULTS: Mice fed a high-fat diet (HFD) developed obesity, hyperinsulinemia, and glucose intolerance. Relative to mice fed a normal diet, HFD mice vaccinated with a SARS-CoV-2 mRNA vaccine exhibited significantly lower anti-spike IgG titers, predominantly in the IgG2c subclass, associated with a lower type 1 response, along with a 3.83-fold decrease in neutralizing titers. Furthermore, enhanced vaccine-induced spike-specific CD8+ T-cell activation and protection from lung infection against SARS-CoV-2 challenge were seen only in mice fed a normal diet but not in HFD mice. CONCLUSIONS: The study demonstrated impaired immunity following SARS-CoV-2 mRNA immunization in a murine model of comorbid T2DM and obesity, supporting the need for further research into the basis for impaired anti-SARS-CoV-2 immunity in T2DM and investigation of novel approaches to enhance vaccine immunogenicity among those with metabolic diseases.


Subject(s)
COVID-19 , Diabetes Mellitus, Type 2 , Insulin Resistance , Viral Vaccines , Animals , Humans , Mice , COVID-19 Vaccines , SARS-CoV-2 , COVID-19/prevention & control , Disease Models, Animal , Immunogenicity, Vaccine , Diet , Obesity , RNA, Messenger , Antibodies, Viral , Antibodies, Neutralizing
7.
Exp Dermatol ; 32(5): 641-647, 2023 05.
Article in English | MEDLINE | ID: mdl-36727558

ABSTRACT

Wrinkling is the hallmark of skin ageing. We previously reported that perioral wrinkling is more severe in females; however, the molecular basis is unknown. This study assessed sex differences in the molecular expression of key ageing regulators in perioral skin. Twelve subjects (n = 6 male/female) were enrolled in this cross-sectional study and biopsies were taken from the perioral and periocular regions. RNA expression of collagen I, collagen III, cysteine-rich angiogenic inducer 61 (CYR61) and insulin-like growth factor 1 (IGF-1) was assessed by qPCR. There was no difference between females' and males' Griffith's grades (6 and 5.67, respectively, p = 0.092) or periocular wrinkling grades (3.2 and 2.6, p = 0.421), but females had more severe perioral wrinkling grades than males (6.2 and 2.8, p = 0.020). Females not only expressed significantly more CYR61 (p = 0.018) in the perioral region than malesm but also expressed more collagen III (p = 0.016). There was no difference in collagen I (p = 0.115) or IGF-1 (p = 0.124) expression in the perioral region between sexes. In the periocular region, there were no significant differences between sexes in the expression of all four markers. The significant molecular differences in the perioral region between the sexes may contribute to the greater perioral skin wrinkling seen clinically in females.


Subject(s)
Skin Aging , Humans , Female , Male , Insulin-Like Growth Factor I/metabolism , Sex Characteristics , Cross-Sectional Studies , Collagen/metabolism , Collagen Type I/metabolism , Oxidative Stress
8.
Proc Natl Acad Sci U S A ; 116(22): 10917-10926, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31088972

ABSTRACT

T cell cytokines contribute to immunity against Staphylococcus aureus, but the predominant T cell subsets involved are unclear. In an S. aureus skin infection mouse model, we found that the IL-17 response was mediated by γδ T cells, which trafficked from lymph nodes to the infected skin to induce neutrophil recruitment, proinflammatory cytokines IL-1α, IL-1ß, and TNF, and host defense peptides. RNA-seq for TRG and TRD sequences in lymph nodes and skin revealed a single clonotypic expansion of the encoded complementarity-determining region 3 amino acid sequence, which could be generated by canonical nucleotide sequences of TRGV5 or TRGV6 and TRDV4 However, only TRGV6 and TRDV4 but not TRGV5 sequences expanded. Finally, Vγ6+ T cells were a predominant γδ T cell subset that produced IL-17A as well as IL-22, TNF, and IFNγ, indicating a broad and substantial role for clonal Vγ6+Vδ4+ T cells in immunity against S. aureus skin infections.


Subject(s)
Interleukin-17/physiology , Staphylococcal Infections/immunology , Staphylococcus aureus/pathogenicity , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , Animals , Disease Models, Animal , Humans , Lymph Nodes/immunology , Mice , Staphylococcal Infections/microbiology
9.
EMBO Rep ; 19(12)2018 12.
Article in English | MEDLINE | ID: mdl-30337494

ABSTRACT

Chitin is the second most abundant polysaccharide in nature and linked to fungal infection and asthma. However, bona fide immune receptors directly binding chitin and signaling immune activation and inflammation have not been clearly identified because polymeric crude chitin with unknown purity and molecular composition has been used. By using defined chitin (N-acetyl-glucosamine) oligomers, we here identify six-subunit-long chitin chains as the smallest immunologically active motif and the innate immune receptor Toll-like receptor (TLR2) as a primary fungal chitin sensor on human and murine immune cells. Chitin oligomers directly bind TLR2 with nanomolar affinity, and this fungal TLR2 ligand shows overlapping and distinct signaling outcomes compared to known mycobacterial TLR2 ligands. Unexpectedly, chitin oligomers composed of five or less subunits are inactive, hinting to a size-dependent system of immuno-modulation that appears conserved in plants and humans. Since blocking of the chitin-TLR2 interaction effectively prevents chitin-mediated inflammation in vitro and in vivo, our study highlights the chitin-TLR2 interaction as a potential target for developing novel therapies in chitin-related pathologies and fungal disease.


Subject(s)
Chitin/chemistry , Chitin/metabolism , Fungi/metabolism , Inflammation/metabolism , Inflammation/pathology , Toll-Like Receptor 2/metabolism , Animals , Cell Wall/drug effects , Cell Wall/metabolism , Chitinases/metabolism , Female , Humans , Hydrophobic and Hydrophilic Interactions , Immunologic Factors/pharmacology , Ligands , Lymphocytes/drug effects , Lymphocytes/metabolism , Mice, Inbred C57BL , Mice, Knockout , THP-1 Cells , Toll-Like Receptor 1/agonists , Toll-Like Receptor 1/metabolism , Toll-Like Receptor 2/chemistry , Zymosan/metabolism
10.
J Immunol ; 201(6): 1651-1661, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30045969

ABSTRACT

IL-17 is a potent proinflammatory cytokine that drives pathogenesis of multiple autoimmune diseases, including psoriasis. A major source of pathogenic IL-17 is a subset of γδ T cells (Tγδ17) that acquires the ability to produce IL-17 while developing in the thymus. The mechanisms that regulate homeostasis of Tγδ17 cells and their roles in psoriasis, however, are not fully understood. In this paper, we show that the heparan sulfate proteoglycan syndecan-1 (sdc1) plays a critical role in regulating homeostasis of Tγδ17 cells and modulating psoriasis-like skin inflammation in mice. sdc1 was predominantly expressed by Tγδ17 cells (but not IL-17- Tγδ cells) in the thymus, lymph nodes, and dermis. sdc1 deficiency significantly and selectively increased the frequency and absolute numbers of Tγδ17 cells by mechanisms that included increased proliferation and decreased apoptosis. Adoptive transfer experiments ruled out a significant role of sdc1 expressed on nonhematopoietic cells in halting expansion and proliferation of sdc1-deficient Tγδ17 cells. When subjected to imiquimod-induced psoriasiform dermatitis, Tγδ17 cells in sdc1KO mice displayed heightened responses accompanied by significantly increased skin inflammation than their wild-type counterparts. Furthermore, transferred sdc1-deficient γδ T cells caused more severe psoriasiform dermatitis than their sdc1-sufficient counterparts in TCR-ßδ KO hosts. The results uncover a novel role for sdc1 in controlling homeostasis of Tγδ17 cells and moderating host responses to psoriasis-like inflammation.


Subject(s)
Dermatitis/immunology , Interleukin-17/immunology , Psoriasis/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Syndecan-1/immunology , T-Lymphocytes/immunology , Animals , Dermatitis/genetics , Dermatitis/pathology , Disease Models, Animal , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-17/genetics , Mice , Mice, Knockout , Psoriasis/genetics , Psoriasis/pathology , Receptors, Antigen, T-Cell, gamma-delta/genetics , Syndecan-1/genetics , T-Lymphocytes/pathology
11.
Proc Natl Acad Sci U S A ; 114(26): E5094-E5102, 2017 06 27.
Article in English | MEDLINE | ID: mdl-28607050

ABSTRACT

Infection is a major complication of implantable medical devices, which provide a scaffold for biofilm formation, thereby reducing susceptibility to antibiotics and complicating treatment. Hematogenous implant-related infections following bacteremia are particularly problematic because they can occur at any time in a previously stable implant. Herein, we developed a model of hematogenous infection in which an orthopedic titanium implant was surgically placed in the legs of mice followed 3 wk later by an i.v. exposure to Staphylococcus aureus This procedure resulted in a marked propensity for a hematogenous implant-related infection comprised of septic arthritis, osteomyelitis, and biofilm formation on the implants in the surgical legs compared with sham-operated surgical legs without implant placement and with contralateral nonoperated normal legs. Neutralizing human monoclonal antibodies against α-toxin (AT) and clumping factor A (ClfA), especially in combination, inhibited biofilm formation in vitro and the hematogenous implant-related infection in vivo. Our findings suggest that AT and ClfA are pathogenic factors that could be therapeutically targeted against Saureus hematogenous implant-related infections.


Subject(s)
Antibodies, Bacterial/pharmacology , Antibodies, Neutralizing/pharmacology , Arthritis, Infectious , Biofilms/drug effects , Implants, Experimental/microbiology , Osteomyelitis , Staphylococcal Infections , Staphylococcus aureus/physiology , Animals , Arthritis, Infectious/drug therapy , Arthritis, Infectious/etiology , Arthritis, Infectious/microbiology , Disease Models, Animal , Humans , Male , Mice , Osteomyelitis/drug therapy , Osteomyelitis/etiology , Osteomyelitis/microbiology , Staphylococcal Infections/drug therapy , Staphylococcal Infections/etiology , Staphylococcal Infections/microbiology , Titanium
12.
J Allergy Clin Immunol ; 143(4): 1426-1443.e6, 2019 04.
Article in English | MEDLINE | ID: mdl-30240702

ABSTRACT

BACKGROUND: Atopic dermatitis (AD) is associated with epidermal barrier defects, dysbiosis, and skin injury caused by scratching. In particular, the barrier-defective epidermis in patients with AD with loss-of-function filaggrin mutations has increased IL-1α and IL-1ß levels, but the mechanisms by which IL-1α, IL-1ß, or both are induced and whether they contribute to the aberrant skin inflammation in patients with AD is unknown. OBJECTIVE: We sought to determine the mechanisms through which skin injury, dysbiosis, and increased epidermal IL-1α and IL-1ß levels contribute to development of skin inflammation in a mouse model of injury-induced skin inflammation in filaggrin-deficient mice without the matted mutation (ft/ft mice). METHODS: Skin injury of wild-type, ft/ft, and myeloid differentiation primary response gene-88-deficient ft/ft mice was performed, and ensuing skin inflammation was evaluated by using digital photography, histologic analysis, and flow cytometry. IL-1α and IL-1ß protein expression was measured by means of ELISA and visualized by using immunofluorescence and immunoelectron microscopy. Composition of the skin microbiome was determined by using 16S rDNA sequencing. RESULTS: Skin injury of ft/ft mice induced chronic skin inflammation involving dysbiosis-driven intracellular IL-1α release from keratinocytes. IL-1α was necessary and sufficient for skin inflammation in vivo and secreted from keratinocytes by various stimuli in vitro. Topical antibiotics or cohousing of ft/ft mice with unaffected wild-type mice to alter or intermix skin microbiota, respectively, resolved the skin inflammation and restored keratinocyte intracellular IL-1α localization. CONCLUSIONS: Taken together, skin injury, dysbiosis, and filaggrin deficiency triggered keratinocyte intracellular IL-1α release that was sufficient to drive chronic skin inflammation, which has implications for AD pathogenesis and potential therapeutic targets.


Subject(s)
Dermatitis, Atopic/metabolism , Inflammation/metabolism , Interleukin-1alpha/metabolism , Intermediate Filament Proteins/deficiency , Keratinocytes/metabolism , Animals , Dermatitis, Atopic/immunology , Dermatitis, Atopic/microbiology , Dysbiosis/immunology , Dysbiosis/metabolism , Filaggrin Proteins , Inflammation/immunology , Inflammation/microbiology , Interleukin-1alpha/immunology , Keratinocytes/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout
13.
J Immunol ; 199(5): 1772-1782, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28733486

ABSTRACT

The immune response to Staphylococcus aureus infection in skin involves the recruitment of polymorphonuclear neutrophils (PMNs) from the bone marrow via the circulation and local granulopoiesis from hematopoietic stem and progenitor cells (HSPCs) that also traffic to infected skin wounds. We focus on regulation of PMN number and function and the role of pore-forming α-toxin (AT), a virulence factor that causes host cell lysis and elicits inflammasome-mediated IL-1ß secretion in wounds. Infection with wild-type S. aureus enriched in AT reduced PMN recruitment and resulted in sustained bacterial burden and delayed wound healing. In contrast, PMN recruitment to wounds infected with an isogenic AT-deficient S. aureus strain was unimpeded, exhibiting efficient bacterial clearance and hastened wound resolution. HSPCs recruited to infected wounds were unaffected by AT production and were activated to expand PMN numbers in proportion to S. aureus abundance in a manner regulated by TLR2 and IL-1R signaling. Immunodeficient MyD88-knockout mice infected with S. aureus experienced lethal sepsis that was reversed by PMN expansion mediated by injection of wild-type HSPCs directly into wounds. We conclude that AT-induced IL-1ß promotes local granulopoiesis and effective resolution of S. aureus-infected wounds, revealing a potential antibiotic-free strategy for tuning the innate immune response to treat methicillin-resistant S. aureus infection in immunodeficient patients.


Subject(s)
Bacterial Toxins/immunology , Granulocytes/immunology , Hematopoietic Stem Cells/physiology , Hemolysin Proteins/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/physiology , Virulence Factors/immunology , Wound Infection/immunology , Animals , Bacterial Load , Bacterial Toxins/genetics , Cell Differentiation , Cell Proliferation , Granulocytes/microbiology , Hemolysin Proteins/genetics , Immunomodulation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Myeloid Differentiation Factor 88/genetics , Receptors, Interleukin-1/metabolism , Signal Transduction , Toll-Like Receptor 2/metabolism , Virulence Factors/genetics
14.
Proc Natl Acad Sci U S A ; 113(45): E6919-E6928, 2016 Nov 08.
Article in English | MEDLINE | ID: mdl-27791154

ABSTRACT

Bacterial biofilm formation is a major complication of implantable medical devices that results in therapeutically challenging chronic infections, especially in cases involving antibiotic-resistant bacteria. As an approach to prevent these infections, an electrospun composite coating comprised of poly(lactic-coglycolic acid) (PLGA) nanofibers embedded in a poly(ε-caprolactone) (PCL) film was developed to locally codeliver combinatorial antibiotics from the implant surface. The release of each antibiotic could be adjusted by loading each drug into the different polymers or by varying PLGA:PCL polymer ratios. In a mouse model of biofilm-associated orthopedic-implant infection, three different combinations of antibiotic-loaded coatings were highly effective in preventing infection of the bone/joint tissue and implant biofilm formation and were biocompatible with enhanced osseointegration. This nanofiber composite-coating technology could be used to tailor the delivery of combinatorial antimicrobial agents from various metallic implantable devices or prostheses to effectively decrease biofilm-associated infections in patients.

15.
Article in English | MEDLINE | ID: mdl-29311091

ABSTRACT

Staphylococcus aureus wound infections delay healing and result in invasive complications such as osteomyelitis, especially in the setting of diabetic foot ulcers. In preclinical animal models of S. aureus skin infection, antibody neutralization of alpha-toxin (AT), an S. aureus-secreted pore-forming cytolytic toxin, reduces disease severity by inhibiting skin necrosis and restoring effective host immune responses. However, whether therapeutic neutralization of alpha-toxin is effective against S. aureus-infected wounds is unclear. Herein, the efficacy of prophylactic treatment with a human neutralizing anti-AT monoclonal antibody (MAb) was evaluated in an S. aureus skin wound infection model in nondiabetic and diabetic mice. In both nondiabetic and diabetic mice, anti-AT MAb treatment decreased wound size and bacterial burden and enhanced reepithelialization and wound resolution compared to control MAb treatment. Anti-AT MAb had distinctive effects on the host immune response, including decreased neutrophil and increased monocyte and macrophage infiltrates in nondiabetic mice and decreased neutrophil extracellular traps (NETs) in diabetic mice. Similar therapeutic efficacy was achieved with an active vaccine targeting AT. Taken together, neutralization of AT had a therapeutic effect against S. aureus-infected wounds in both nondiabetic and diabetic mice that was associated with differential effects on the host immune response.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Bacterial Toxins/antagonists & inhibitors , Diabetes Mellitus, Experimental/immunology , Hemolysin Proteins/antagonists & inhibitors , Staphylococcal Skin Infections/drug therapy , Wound Healing/drug effects , Wounds, Nonpenetrating/drug therapy , Animals , Bacterial Load/drug effects , Bacterial Toxins/immunology , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/microbiology , Extracellular Traps/drug effects , Extracellular Traps/microbiology , Hemolysin Proteins/immunology , Humans , Immunity, Innate/drug effects , Macrophages/drug effects , Macrophages/microbiology , Male , Mice , Mice, Inbred C57BL , Monocytes/drug effects , Monocytes/microbiology , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/microbiology , Skin/drug effects , Skin/immunology , Skin/microbiology , Staphylococcal Skin Infections/complications , Staphylococcal Skin Infections/immunology , Staphylococcal Skin Infections/microbiology , Staphylococcal Vaccines/pharmacology , Wound Healing/immunology , Wounds, Nonpenetrating/complications , Wounds, Nonpenetrating/immunology , Wounds, Nonpenetrating/microbiology
16.
J Allergy Clin Immunol ; 140(4): 1054-1067.e10, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28216434

ABSTRACT

BACKGROUND: The Nod-like receptor NACHT, LRR, and PYD domain-containing protein 3 (NLRP3) and Bruton tyrosine kinase (BTK) are protagonists in innate and adaptive immunity, respectively. NLRP3 senses exogenous and endogenous insults, leading to inflammasome activation, which occurs spontaneously in patients with Muckle-Wells syndrome; BTK mutations cause the genetic immunodeficiency X-linked agammaglobulinemia (XLA). However, to date, few proteins that regulate NLRP3 inflammasome activity in human primary immune cells have been identified, and clinically promising pharmacologic targeting strategies remain elusive. OBJECTIVE: We sought to identify novel regulators of the NLRP3 inflammasome in human cells with a view to exploring interference with inflammasome activity at the level of such regulators. METHODS: After proteome-wide phosphoproteomics, the identified novel regulator BTK was studied in human and murine cells by using pharmacologic and genetic BTK ablation. RESULTS: Here we show that BTK is a critical regulator of NLRP3 inflammasome activation: pharmacologic (using the US Food and Drug Administration-approved inhibitor ibrutinib) and genetic (in patients with XLA and Btk knockout mice) BTK ablation in primary immune cells led to reduced IL-1ß processing and secretion in response to nigericin and the Staphylococcus aureus toxin leukocidin AB (LukAB). BTK affected apoptosis-associated speck-like protein containing a CARD (ASC) speck formation and caspase-1 cleavage and interacted with NLRP3 and ASC. S aureus infection control in vivo and IL-1ß release from cells of patients with Muckle-Wells syndrome were impaired by ibrutinib. Notably, IL-1ß processing and release from immune cells isolated from patients with cancer receiving ibrutinib therapy were reduced. CONCLUSION: Our data suggest that XLA might result in part from genetic inflammasome deficiency and that NLRP3 inflammasome-linked inflammation could potentially be targeted pharmacologically through BTK.


Subject(s)
Agammaglobulinemia/genetics , Cryopyrin-Associated Periodic Syndromes/genetics , Genetic Diseases, X-Linked/genetics , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protein-Tyrosine Kinases/metabolism , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Adaptive Immunity , Adaptor Proteins, Signal Transducing , Agammaglobulinaemia Tyrosine Kinase , Animals , Apoptosis Regulatory Proteins , Bacterial Proteins/immunology , Cells, Cultured , Humans , Immunity, Innate , Leukocidins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Targeted Therapy , NLR Proteins , Nigericin/immunology , Protein-Tyrosine Kinases/genetics , Proteomics , Pyrin Domain/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Lamin B Receptor
17.
Open Forum Infect Dis ; 11(2): ofad625, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38352152

ABSTRACT

Nirmatrelvir/ritonavir (NMV/r) is used for the treatment of coronavirus disease 2019 (COVID-19) infection. However, rebound COVID-19 infections can occur after taking NMV/r. We examined neutralizing antibodies to the severe acute respiratory syndrome coronavirus 2 spike protein before and after infection in people who did and did not take NMV/r to determine if NMV/r impedes the humoral immune response.

18.
JAMA Dermatol ; 160(7): 717-724, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38837144

ABSTRACT

Importance: Prurigo nodularis (PN) and chronic pruritus of unknown origin (CPUO) are chronic pruritic diseases that dramatically impair quality of life, but therapeutic options are limited. Abrocitinib, a Janus kinase 1 inhibitor, represents a promising therapy for both conditions. Objective: To assess the efficacy and safety of 200-mg oral abrocitinib administered once daily in adults with moderate to severe PN or CPUO. Design, Setting, and Participants: This phase 2, open-label, nonrandomized controlled trial conducted between September 2021 and July 2022 took place at a single center in the US. A total of 25 adult patients with moderate to severe PN or CPUO were screened. Ten patients with PN and 10 patients with CPUO were enrolled. All 20 patients completed the 12-week treatment period, 18 of whom completed the 4-week follow-up period. Intervention: Abrocitinib, 200 mg, by mouth once daily for 12 weeks. Main Outcomes and Measures: The primary efficacy end point was the percent change in weekly Peak Pruritus Numerical Rating Scale (PP-NRS) scores from baseline to week 12. Key secondary end points included the percentage of patients achieving at least a 4-point reduction in weekly PP-NRS score from baseline to week 12 and the percent change in Dermatology Life Quality Index (DLQI) scores. Results: A total of 10 patients with PN (mean [SD] age, 58.6 [13.1] years; all were female) and 10 patients with CPUO (mean [SD] age, 70.7 [5.6] years; 2 were female) enrolled in the study. The mean (SD) baseline PP-NRS score was 9.2 (1.0) for PN and 8.2 (1.2) for CPUO. PP-NRS scores decreased by 78.3% in PN (95% CI, -118.5 to -38.1; P < .001) and 53.7% in CPUO (95% CI, -98.8 to -8.6; P = .01) by week 12. From baseline to week 12, 8 of 10 patients with PN and 6 of 10 patients with CPUO achieved at least a 4-point improvement on the PP-NRS. Both groups experienced significant improvement in quality of life as demonstrated by percent change in DLQI scores (PN: -53.2% [95% CI, -75.3% to -31.1%]; P = .002; CPUO: -49.0% [95% CI, -89.6% to -8.0%]; P = .02). The most common adverse event among patients was acneiform eruption in 2 of 20 patients (10%). No serious adverse events occurred. Conclusions and Relevance: The results of this nonrandomized controlled trial suggest that abrocitinib monotherapy may be effective and tolerated well in adults with PN or CPUO. Randomized, double-blind, placebo-controlled trials are warranted to validate these findings. Trial Registration: ClinicalTrials.gov Identifier: NCT05038982.


Subject(s)
Prurigo , Pruritus , Pyrimidines , Quality of Life , Severity of Illness Index , Humans , Female , Male , Middle Aged , Prurigo/drug therapy , Prurigo/complications , Prurigo/diagnosis , Pruritus/drug therapy , Pruritus/etiology , Aged , Treatment Outcome , Chronic Disease , Adult , Pyrimidines/administration & dosage , Pyrimidines/adverse effects , Administration, Oral , Sulfonamides/administration & dosage , Sulfonamides/adverse effects
19.
mBio ; 14(5): e0158723, 2023 Oct 31.
Article in English | MEDLINE | ID: mdl-37581442

ABSTRACT

IMPORTANCE: Pyronaridine tetraphosphate is on the WHO Essential Medicine List for its importance as a widely available and safe treatment for malaria. We find that pyronaridine is a highly effective antiviral therapeutic across mouse models using multiple variants of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), and the highly pathogenic viruses SARS-CoV-1 and Middle East respiratory syndrome coronavirus responsible for previous coronavirus outbreaks. Additionally, we find that pyronaridine additively combines with current COVID-19 treatments such as nirmatrelvir (protease inhibitor in Paxlovid) and molnupiravir to further inhibit SARS-CoV-2 infections. There are many antiviral compounds that demonstrate efficacy in cellular models, but few that show this level of impact in multiple mouse models and represent a promising therapeutic for the current coronavirus pandemic as well as future outbreaks as well.


Subject(s)
COVID-19 , Middle East Respiratory Syndrome Coronavirus , Mice , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Naphthyridines/pharmacology , SARS-CoV-2
20.
bioRxiv ; 2023 May 05.
Article in English | MEDLINE | ID: mdl-36299426

ABSTRACT

Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2), the causative agent of Coronavirus disease 2019 (COVID-19), emerged in Wuhan, China, in December 2019. As of October 2022, there have been over 625 million confirmed cases of COVID-19, including over 6.5 million deaths. Epidemiological studies have indicated that comorbidities of obesity and diabetes mellitus are associated with increased morbidity and mortality following SARS-CoV-2 infection. We determined how the comorbidities of obesity and diabetes affect morbidity and mortality following SARS-CoV-2 infection in unvaccinated and adjuvanted spike nanoparticle (NVX-CoV2373) vaccinated mice. We find that obese/diabetic mice infected with SARS-CoV-2 have increased morbidity and mortality compared to age matched normal mice. Mice fed a high-fat diet (HFD) then vaccinated with NVX-CoV2373 produce equivalent neutralizing antibody titers to those fed a normal diet (ND). However, the HFD mice have reduced viral clearance early in infection. Analysis of the inflammatory immune response in HFD mice demonstrates a recruitment of neutrophils that was correlated with increased mortality and reduced clearance of the virus. Depletion of neutrophils in diabetic/obese vaccinated mice reduced disease severity and protected mice from lethality. This model recapitulates the increased disease severity associated with obesity and diabetes in humans with COVID-19 and is an important comorbidity to study with increasing obesity and diabetes across the world.

SELECTION OF CITATIONS
SEARCH DETAIL