41 results on '"Vannella KM"'
Search Results
2. TLR9 Signaling in Alveolar Epithelial Cells Protects from Gammaherpesvirus-Induced Apoptosis.
- Author
-
McMillan, TR, primary, Vannella, KM, additional, Stoolman, JS, additional, Standiford, TJ, additional, Toews, GB, additional, and Moore, BB, additional
- Published
- 2009
- Full Text
- View/download PDF
3. Latent herpesvirus infection augments experimental pulmonary fibrosis.
- Author
-
Vannella KM, Luckhardt TR, Wilke CA, van Dyk LF, Toews GB, Moore BB, Vannella, Kevin M, Luckhardt, Tracy R, Wilke, Carol A, van Dyk, Linda F, Toews, Galen B, and Moore, Bethany B
- Abstract
Rationale: No effective treatment exists for idiopathic pulmonary fibrosis, and its pathogenesis remains unclear. Accumulating evidence implicates herpesviruses as cofactors (either initiating or exacerbating agents) of fibrotic lung disease, but a role for latent herpesvirus infection has not been studied.Objectives: To develop a murine model to determine whether latent herpesvirus infection can augment fibrotic responses and to gain insight into potential mechanisms of enhanced fibrogenesis.Methods: Mice were infected with murine gammaherpesvirus 14 to 70 days before a fibrotic challenge with fluorescein isothiocyanate or bleomycin so that the virus was latent at the time of fibrotic challenge. Measurements were made after viral infection alone or after the establishment of fibrosis.Measurements and Main Results: gammaHerpesvirus is latent by 14 days post infection, and infection 14 to 70 days before fibrotic challenge augmented fibrosis. Fibrotic augmentation was not dependent on reactivation of the latent virus to a lytic state. Total cell numbers and fibrocyte numbers were increased in the lungs of latently infected mice administered fibrotic challenge compared with mock-infected mice that received fibrotic challenge. Latent infection up-regulates expression of proinflammatory chemokines, transforming growth factor-beta1, and cysteinyl leukotrienes in alveolar epithelial cells.Conclusions: Latent gammaherpesvirus infection augments subsequent fibrotic responses in mice. Enhanced fibrosis is associated with the induction of profibrotic factors and the recruitment of fibrocytes. Our data complement existing human and animal data supporting the hypothesis that gammaherpesviruses can serve as initiating cofactors in the pathogenesis of pulmonary fibrosis. [ABSTRACT FROM AUTHOR]- Published
- 2010
- Full Text
- View/download PDF
4. Exacerbation of established pulmonary fibrosis in a murine model by gammaherpesvirus.
- Author
-
McMillan TR, Moore BB, Weinberg JB, Vannella KM, Fields WB, Christensen PJ, van Dyk LF, Toews GB, McMillan, Tracy R, Moore, Bethany B, Weinberg, Jason B, Vannella, Kevin M, Fields, W Brad, Christensen, Paul J, van Dyk, Linda F, and Toews, Galen B
- Abstract
Rationale: Idiopathic pulmonary fibrosis is a progressive disease with high mortality. Although most patients have a slow, progressive course, some patients will have an acute deterioration in function or acute exacerbation, which carries a poor prognosis. In some cases, acute deterioration is associated with infection. Herpesviruses have been associated with this disease. Fibrocytes have also been shown to be important in the pathogenesis of pulmonary fibrosis.Objectives: To develop a murine model for infectious exacerbation of preexisting fibrosis, and provide mechanistic insight into the role of herpesviruses in fibrotic disease.Methods: We used a model of fluorescein isothiocyanate-induced pulmonary fibrosis in mice. Infection with a murine gammaherpesvirus was given at time of established lung fibrosis. Measurements were made at the time of peak lytic viral replication.Measurements and Main Results: We demonstrate that infection with gammaherpesvirus can exacerbate established fluorescein isothiocyanate-induced fibrosis evidenced by increased total lung collagen, histologic changes of acute lung injury, and diminished lung function. Gammaherpesvirus can exacerbate preexisting fibrosis in a Th1 cytokine environment and in the absence of Th2 cytokines. Gammaherpesvirus increases fibrocyte recruitment to the lung in wild-type, but not CCR2(-/-) mice, in part because viral infection up-regulates production of CCL2 and CCL12, chemokines important for fibrocyte recruitment. In contrast, mouse adenovirus infection did not exacerbate collagen deposition.Conclusions: These data provide a new model for gammaherpesvirus exacerbation of established pulmonary fibrosis. The up-regulation of chemokines during viral infection and subsequent recruitment of fibrocytes to the lung likely contribute to augmentation of pulmonary fibrosis. [ABSTRACT FROM AUTHOR]- Published
- 2008
- Full Text
- View/download PDF
5. Evidence of SARS-CoV-2-specific T-cell-mediated myocarditis in a MIS-A case
- Author
-
Vannella, KM, primary, Oguz, C, additional, Stein, SR, additional, Pittaluga, S, additional, Dikoglu, E, additional, Kanwal, A, additional, Ramelli, SC, additional, Briese, T, additional, Su, L, additional, Wu, X, additional, Ramos-Benitez, MJ, additional, Perez-Valencia, LJ, additional, Babyak, A, additional, Cha, NR, additional, Chung, JY, additional, Ylaya, K, additional, Madathil, RJ, additional, Saharia, KK, additional, Scalea, TM, additional, Tran, QK, additional, Herr, DL, additional, Kleiner, DE, additional, Hewitt, SM, additional, Notarangelo, LD, additional, Grazioli, A, additional, and Chertow, DS, additional
- Full Text
- View/download PDF
6. SARS-CoV-2 infection perturbs the gastrointestinal tract and induces modest microbial translocation across the intestinal barrier.
- Author
-
Brooks K, Nelson CE, Aguilar C, Hoang TN, Ortiz AM, Langner CA, Yee DS, Flynn JK, Vrba S, Laidlaw E, Vannella KM, Grazioli A, Saharia KK, Purcell M, Singireddy S, Wu J, Stankiewicz J, Chertow DS, Sereti I, Paiardini M, Hickman HD, Via LE, Barber DL, and Brenchley JM
- Subjects
- Animals, Humans, Male, Gastrointestinal Tract microbiology, Gastrointestinal Tract virology, Gastrointestinal Tract immunology, Intestinal Mucosa microbiology, Intestinal Mucosa immunology, Intestinal Mucosa virology, Intestinal Mucosa metabolism, Female, Haptoglobins metabolism, B-Lymphocytes immunology, Middle Aged, Protein Precursors, Macaca mulatta, COVID-19 immunology, COVID-19 virology, COVID-19 microbiology, SARS-CoV-2 immunology, Bacterial Translocation, Lipopolysaccharide Receptors metabolism, Interleukin-6 metabolism
- Abstract
SARS-CoV-2 infects via the respiratory tract, but COVID-19 includes an array of non-respiratory symptoms, among them gastrointestinal (GI) manifestations such as vomiting and diarrhea. Here we investigated the GI pathology of SARS-CoV-2 infections in rhesus macaques and humans. Macaques experienced mild infection with USA-WA1/2020 and shed viral RNA in the respiratory tract and stool, including subgenomic RNA indicative of replication in the GI tract. Intestinal immune cell populations were disturbed, with significantly fewer proliferating (Ki67+) jejunal B cells in SARS-CoV-2-infected macaques than uninfected ones. Modest translocation of bacteria/bacterial antigen was observed across the colonic epithelium, with a corresponding significant increase in plasma soluble CD14 (sCD14) that may be induced by LPS. Human plasma demonstrated significant decreases in interleukin (IL)-6 and sCD14 upon recovery from COVID-19, suggesting resolution of inflammation and response to translocated bacteria. sCD14 significantly positively correlated with zonulin, an indicator of gut barrier integrity, and IL-6. These results demonstrate that GI perturbations such as microbial translocation can occur in even mild SARS-CoV-2 infections and may contribute to the COVID-19 inflammatory state.IMPORTANCEThis study investigates gastrointestinal (GI) barrier disruption in SARS-CoV-2 infections and how it may contribute to disease. We observed bacteria or bacterial products crossing from the colon interior (the lumen) to the lamina propria during SARS-CoV-2 infection in macaques. Bacteria/bacterial products are tolerated in the lumen but may induce immune responses if they translocate to the lamina propria. We also observed a significant increase in soluble CD14, which is associated with an immune response to bacterial products. In addition, we observed that humans recovering from COVID-19 experienced a significant decrease in soluble CD14, as well as the inflammatory marker interleukin (IL)-6. IL-6 and sCD14 correlated significantly across macaque and human samples. These findings suggest that SARS-CoV-2 infection results in GI barrier disruption that permits microbial translocation and a corresponding immune response. These findings could aid in developing interventions to improve COVID-19 patient outcomes., Competing Interests: The authors declare no conflict of interest.
- Published
- 2024
- Full Text
- View/download PDF
7. Initiator cell death event induced by SARS-CoV-2 in the human airway epithelium.
- Author
-
Liang K, Barnett KC, Hsu M, Chou WC, Bais SS, Riebe K, Xie Y, Nguyen TT, Oguin TH 3rd, Vannella KM, Hewitt SM, Chertow DS, Blasi M, Sempowski GD, Karlsson A, Koller BH, Lenschow DJ, Randell SH, and Ting JP
- Subjects
- Humans, Animals, Mice, Cell Death immunology, Angiotensin-Converting Enzyme 2 metabolism, Angiotensin-Converting Enzyme 2 genetics, Apoptosis immunology, SARS-CoV-2 immunology, COVID-19 immunology, COVID-19 pathology, Necroptosis immunology, Pyroptosis, Respiratory Mucosa virology, Respiratory Mucosa immunology, Respiratory Mucosa pathology, RNA-Binding Proteins metabolism, RNA-Binding Proteins genetics
- Abstract
Virus-induced cell death is a key contributor to COVID-19 pathology. Cell death induced by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is well studied in myeloid cells but less in its primary host cell type, angiotensin-converting enzyme 2 (ACE2)-expressing human airway epithelia (HAE). SARS-CoV-2 induces apoptosis, necroptosis, and pyroptosis in HAE organotypic cultures. Single-cell and limiting-dilution analysis revealed that necroptosis is the primary cell death event in infected cells, whereas uninfected bystanders undergo apoptosis, and pyroptosis occurs later during infection. Mechanistically, necroptosis is induced by viral Z-RNA binding to Z-DNA-binding protein 1 (ZBP1) in HAE and lung tissues from patients with COVID-19. The Delta (B.1.617.2) variant, which causes more severe disease than Omicron (B1.1.529) in humans, is associated with orders of magnitude-greater Z-RNA/ZBP1 interactions, necroptosis, and disease severity in animal models. Thus, Delta induces robust ZBP1-mediated necroptosis and more disease severity.
- Published
- 2024
- Full Text
- View/download PDF
8. Clinical and Immunologic Correlates of Vasodilatory Shock Among Ebola Virus-Infected Nonhuman Primates in a Critical Care Model.
- Author
-
Stein SR, Platt AP, Teague HL, Anthony SM, Reeder RJ, Cooper K, Byrum R, Drawbaugh DJ 2nd, Liu DX, Burdette TL, Hadley K, Barr B, Warner S, Rodriguez-Hernandez F, Johnson C, Stanek P, Hischak J, Kendall H, Huzella LM, Strich JR, Herbert R, St Claire M, Vannella KM, Holbrook MR, and Chertow DS
- Subjects
- Animals, Humans, Macaca mulatta, Leukocytes, Mononuclear, Viremia, Critical Care, Ebolavirus, Hemorrhagic Fever, Ebola
- Abstract
Background: Existing models of Ebola virus infection have not fully characterized the pathophysiology of shock in connection with daily virologic, clinical, and immunologic parameters. We implemented a nonhuman primate critical care model to investigate these associations., Methods: Two rhesus macaques received a target dose of 1000 plaque-forming units of Ebola virus intramuscularly with supportive care initiated on day 3. High-dimensional spectral cytometry was used to phenotype neutrophils and peripheral blood mononuclear cells daily., Results: We observed progressive vasodilatory shock with preserved cardiac function following viremia onset on day 5. Multiorgan dysfunction began on day 6 coincident with the nadir of circulating neutrophils. Consumptive coagulopathy and anemia occurred on days 7 to 8 along with irreversible shock, followed by death. The monocyte repertoire began shifting on day 4 with a decline in classical and expansion of double-negative monocytes. A selective loss of CXCR3-positive B and T cells, expansion of naive B cells, and activation of natural killer cells followed viremia onset., Conclusions: Our model allows for high-fidelity characterization of the pathophysiology of acute Ebola virus infection with host innate and adaptive immune responses, which may advance host-targeted therapy design and evaluation for use after the onset of multiorgan failure., Competing Interests: Potential conflicts of interest. All authors: No reported conflicts. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed., (Published by Oxford University Press on behalf of Infectious Diseases Society of America 2023.)
- Published
- 2023
- Full Text
- View/download PDF
9. Histopathology and SARS-CoV-2 Cellular Localization in Eye Tissues of COVID-19 Autopsies.
- Author
-
Sen HN, Vannella KM, Wang Y, Chung JY, Kodati S, Ramelli SC, Lee JW, Perez P, Stein SR, Grazioli A, Dickey JM, Ylaya K, Singh M, Yinda KC, Platt A, Ramos-Benitez MJ, Zerbe C, Munster VJ, de Wit E, Warner BM, Herr DL, Rabin J, Saharia KK, Kleiner DE, Hewitt SM, Chan CC, and Chertow DS
- Subjects
- Humans, SARS-CoV-2, Autopsy, RNA, Viral analysis, Inflammation, COVID-19
- Abstract
Ophthalmic manifestations and tissue tropism of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have been reported in association with coronavirus disease 2019 (COVID-19), but the pathology and cellular localization of SARS-CoV-2 are not well characterized. The objective of this study was to evaluate macroscopic and microscopic changes and investigate cellular localization of SARS-CoV-2 across ocular tissues at autopsy. Ocular tissues were obtained from 25 patients with COVID-19 at autopsy. SARS-CoV-2 nucleocapsid gene RNA was previously quantified by droplet digital PCR from one eye. Herein, contralateral eyes from 21 patients were fixed in formalin and subject to histopathologic examination. Sections of the droplet digital PCR-positive eyes from four other patients were evaluated by in situ hybridization to determine the cellular localization of SARS-CoV-2 spike gene RNA. Histopathologic abnormalities, including cytoid bodies, vascular changes, and retinal edema, with minimal or no inflammation in ocular tissues were observed in all 21 cases evaluated. In situ hybridization localized SARS-CoV-2 RNA to neuronal cells of the retinal inner and outer layers, ganglion cells, corneal epithelia, scleral fibroblasts, and oligodendrocytes of the optic nerve. In conclusion, a range of common histopathologic alterations were identified within ocular tissue, and SARS-CoV-2 RNA was localized to multiple cell types. Further studies will be required to determine whether the alterations observed were caused by SARS-CoV-2 infection, the host immune response, and/or preexisting comorbidities., (Copyright © 2023 American Society for Investigative Pathology. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
10. Pulmonary Co-Infections Detected Premortem Underestimate Postmortem Findings in a COVID-19 Autopsy Case Series.
- Author
-
Platt AP, Bradley BT, Nasir N, Stein SR, Ramelli SC, Ramos-Benitez MJ, Dickey JM, Purcell M, Singireddy S, Hays N, Wu J, Raja K, Curto R, Salipante SJ, Chisholm C, Carnes S, Marshall DA, Cookson BT, Vannella KM, Madathil RJ, Soherwardi S, McCurdy MT, Saharia KK, Rabin J, Nih Covid-Autopsy Consortium, Grazioli A, Kleiner DE, Hewitt SM, Lieberman JA, and Chertow DS
- Abstract
Bacterial and fungal co-infections are reported complications of coronavirus disease 2019 (COVID-19) in critically ill patients but may go unrecognized premortem due to diagnostic limitations. We compared the premortem with the postmortem detection of pulmonary co-infections in 55 fatal COVID-19 cases from March 2020 to March 2021. The concordance in the premortem versus the postmortem diagnoses and the pathogen identification were evaluated. Premortem pulmonary co-infections were extracted from medical charts while applying standard diagnostic definitions. Postmortem co-infection was defined by compatible lung histopathology with or without the detection of an organism in tissue by bacterial or fungal staining, or polymerase chain reaction (PCR) with broad-range bacterial and fungal primers. Pulmonary co-infection was detected premortem in significantly fewer cases (15/55, 27%) than were detected postmortem (36/55, 65%; p < 0.0001). Among cases in which co-infection was detected postmortem by histopathology, an organism was identified in 27/36 (75%) of cases. Pseudomonas , Enterobacterales , and Staphylococcus aureus were the most frequently identified bacteria both premortem and postmortem. Invasive pulmonary fungal infection was detected in five cases postmortem, but in no cases premortem. According to the univariate analyses, the patients with undiagnosed pulmonary co-infection had significantly shorter hospital ( p = 0.0012) and intensive care unit ( p = 0.0006) stays and significantly fewer extra-pulmonary infections ( p = 0.0021). Bacterial and fungal pulmonary co-infection are under-recognized complications in critically ill patients with COVID-19.
- Published
- 2023
- Full Text
- View/download PDF
11. Ebola virus shed glycoprotein is toxic to human T, B, and natural killer lymphocytes.
- Author
-
Perez-Valencia LJ, Vannella KM, Ramos-Benitez MJ, Sun J, Abu-Asab M, Dorward DW, Awad KS, Platt A, Jacobson E, Kindrachuk J, and Chertow DS
- Abstract
Lymphocyte depletion is a distinctive feature of Ebola virus (EBOV) disease. The ectodomain of EBOV glycoprotein (GP) is cleaved off the surface of infected cells into circulation as shed GP. To test the hypothesis that shed GP induces lymphocyte death, we cultured primary human B, NK, or T cells with shed GP in vitro . We found that shed GP dependably decreased B, NK, and T cell viability across donors. B and NK cells exhibited higher susceptibility than T cells. Continuous monitoring revealed shed GP began to kill B and NK cells by 4 h and T cells by 5 h. We also demonstrated that shed GP-induced lymphocyte death can be both caspase dependent and caspase independent. Our data are evidence that the cytotoxic effect of shed GP on lymphocytes may contribute to EBOV disease and highlight the need for further research to clarify mechanisms of shed GP-induced death., Competing Interests: The authors declare no competing interests., (© 2023 The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
12. Clinical and biological heterogeneity of multisystem inflammatory syndrome in adults following SARS-CoV-2 infection: a case series.
- Author
-
Barth KE, Spottiswoode N, Hurabielle C, Subbaraj L, Calfee CS, Matthay MA, French S, Connolly A, Hewitt SM, Vannella KM, Barnett C, Langelier CR, and Patterson S
- Abstract
Importance: Multisystem inflammatory syndrome in adults (MIS-A) is a poorly understood complication of SARS-CoV-2 infection with significant morbidity and mortality., Objective: Identify clinical, immunological, and histopathologic features of MIS-A to improve understanding of the pathophysiology and approach to treatment., Design: Three cases of MIS-A following SARS-CoV-2 infection were clinically identified between October 2021 - March 2022 using the U.S. Centers for Disease Control and Prevention diagnostic criteria. Clinical, laboratory, imaging, and tissue data were assessed., Findings: All three patients developed acute onset cardiogenic shock and demonstrated elevated inflammatory biomarkers at the time of hospital admission that resolved over time. One case co-occurred with new onset Type 1 diabetes and sepsis. Retrospective analysis of myocardial tissue from one case identified SARS-CoV-2 RNA. All three patients fully recovered with standard of care interventions plus immunomodulatory therapy that included intravenous immunoglobulin, corticosteroids, and in two cases, anakinra., Conclusion: MIS-A is a severe post-acute sequela of COVID-19 characterized by systemic elevation of inflammatory biomarkers. In this series of three cases, we find that although clinical courses and co-existent diseases vary, even severe presentations have potential for full recovery with prompt recognition and treatment. In addition to cardiogenic shock, glucose intolerance, unmasking of autoimmune disease, and sepsis can be features of MIS-A, and SARS-CoV-2 myocarditis can lead to a similar clinical syndrome., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Barth, Spottiswoode, Hurabielle, Subbaraj, COMET Consortium, Calfee, Matthay, French, Connolly, Hewitt, Vannella, Barnett, Langelier and Patterson.)
- Published
- 2023
- Full Text
- View/download PDF
13. Successful lung transplantation using an allograft from a COVID-19-recovered donor: a potential role for subgenomic RNA to guide organ utilization.
- Author
-
Saharia KK, Ramelli SC, Stein SR, Roder AE, Kreitman A, Banakis S, Chung JY, Burbelo PD, Singh M, Reed RM, Patel V, Rabin J, Krupnick AS, Cohen JI, de Wit E, Ghedin E, Hewitt SM, Vannella KM, Chertow DS, and Grazioli A
- Subjects
- Humans, SARS-CoV-2 genetics, Subgenomic RNA, RNA, Viral genetics, Retrospective Studies, Allografts, COVID-19, Lung Transplantation
- Abstract
Although the risk of SARS-CoV-2 transmission through lung transplantation from acutely infected donors is high, the risks of virus transmission and long-term lung allograft outcomes are not as well described when using pulmonary organs from COVID-19-recovered donors. We describe successful lung transplantation for a COVID-19-related lung injury using lungs from a COVID-19-recovered donor who was retrospectively found to have detectable genomic SARS-CoV-2 RNA in the lung tissue by multiple highly sensitive assays. However, SARS-CoV-2 subgenomic RNA (sgRNA), a marker of viral replication, was not detectable in the donor respiratory tissues. One year after lung transplantation, the recipient has a good functional status, walking 1 mile several times per week without the need for supplemental oxygen and without any evidence of donor-derived SARS-CoV-2 transmission. Our findings highlight the limitations of current clinical laboratory diagnostic assays in detecting the persistence of SARS-CoV-2 RNA in the lung tissue. The persistence of SARS-CoV-2 RNA in the donor tissue did not appear to represent active viral replication via sgRNA testing and, most importantly, did not negatively impact the allograft outcome in the first year after lung transplantation. sgRNA is easily performed and may be a useful assay for assessing viral infectivity in organs from donors with a recent infection., (Copyright © 2022 American Society of Transplantation & American Society of Transplant Surgeons. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
14. SARS-CoV-2 infection and persistence in the human body and brain at autopsy.
- Author
-
Stein SR, Ramelli SC, Grazioli A, Chung JY, Singh M, Yinda CK, Winkler CW, Sun J, Dickey JM, Ylaya K, Ko SH, Platt AP, Burbelo PD, Quezado M, Pittaluga S, Purcell M, Munster VJ, Belinky F, Ramos-Benitez MJ, Boritz EA, Lach IA, Herr DL, Rabin J, Saharia KK, Madathil RJ, Tabatabai A, Soherwardi S, McCurdy MT, Peterson KE, Cohen JI, de Wit E, Vannella KM, Hewitt SM, Kleiner DE, and Chertow DS
- Subjects
- Humans, RNA, Viral analysis, Virus Replication, Time Factors, Respiratory System pathology, Respiratory System virology, Autopsy, Brain virology, COVID-19 virology, SARS-CoV-2 genetics, SARS-CoV-2 isolation & purification, SARS-CoV-2 pathogenicity, SARS-CoV-2 physiology, Organ Specificity
- Abstract
Coronavirus disease 2019 (COVID-19) is known to cause multi-organ dysfunction
1-3 during acute infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), with some patients experiencing prolonged symptoms, termed post-acute sequelae of SARS-CoV-2 (refs.4,5 ). However, the burden of infection outside the respiratory tract and time to viral clearance are not well characterized, particularly in the brain3,6-14 . Here we carried out complete autopsies on 44 patients who died with COVID-19, with extensive sampling of the central nervous system in 11 of these patients, to map and quantify the distribution, replication and cell-type specificity of SARS-CoV-2 across the human body, including the brain, from acute infection to more than seven months following symptom onset. We show that SARS-CoV-2 is widely distributed, predominantly among patients who died with severe COVID-19, and that virus replication is present in multiple respiratory and non-respiratory tissues, including the brain, early in infection. Further, we detected persistent SARS-CoV-2 RNA in multiple anatomic sites, including throughout the brain, as late as 230 days following symptom onset in one case. Despite extensive distribution of SARS-CoV-2 RNA throughout the body, we observed little evidence of inflammation or direct viral cytopathology outside the respiratory tract. Our data indicate that in some patients SARS-CoV-2 can cause systemic infection and persist in the body for months., (© 2022. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)- Published
- 2022
- Full Text
- View/download PDF
15. Antiviral innate immunity is diminished in the upper respiratory tract of severe COVID-19 patients.
- Author
-
Ramos-Benitez MJ, Strich JR, Alehashemi S, Stein S, Rastegar A, de Jesus AA, Bhuyan F, Ramelli S, Babyak A, Perez-Valencia L, Vannella KM, Grubbs G, Khurana S, Gross R, Hadley K, Liang J, Mazur S, Postnikova E, Warner S, Holbrook MR, Busch LM, Warner B, Applefeld W, Warner S, Kadri SS, Davey RT, Goldbach-Mansky R, and Chertow DS
- Abstract
Understanding early innate immune responses to coronavirus disease 2019 (COVID-19) is crucial to developing targeted therapies to mitigate disease severity. Severe acute respiratory syndrome coronavirus (SARS-CoV)-2 infection elicits interferon expression leading to transcription of IFN-stimulated genes (ISGs) to control viral replication and spread. SARS-CoV-2 infection also elicits NF-κB signaling which regulates inflammatory cytokine expression contributing to viral control and likely disease severity. Few studies have simultaneously characterized these two components of innate immunity to COVID-19. We designed a study to characterize the expression of interferon alpha-2 ( IFNA2 ) and interferon beta-1 ( IFNB1 ), both type-1 interferons (IFN-1), interferon-gamma ( IFNG ), a type-2 interferon (IFN-2), ISGs, and NF-κB response genes in the upper respiratory tract (URT) of patients with mild (outpatient) versus severe (hospitalized) COVID-19. Further, we characterized the weekly dynamics of these responses in the upper and lower respiratory tracts (LRTs) and blood of severe patients to evaluate for compartmental differences. We observed significantly increased ISG and NF-κB responses in the URT of mild compared with severe patients early during illness. This pattern was associated with increased IFNA2 and IFNG expression in the URT of mild patients, a trend toward increased IFNB1 -expression and significantly increased STING/IRF3 /cGAS expression in the URT of severe patients. Our by-week across-compartment analysis in severe patients revealed significantly higher ISG responses in the blood compared with the URT and LRT of these patients during the first week of illness, despite significantly lower expression of IFNA2 , IFNB1 , and IFNG in blood. NF-κB responses, however, were significantly elevated in the LRT compared with the URT and blood of severe patients during peak illness (week 2). Our data support that severe COVID-19 is associated with impaired interferon signaling in the URT during early illness and robust pro-inflammatory responses in the LRT during peak illness., Competing Interests: Conflict of Interest The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
- Published
- 2022
- Full Text
- View/download PDF
16. SARS-CoV-2 mRNA vaccine induced higher antibody affinity and IgG titers against variants of concern in post-partum vs non-post-partum women.
- Author
-
Lee Y, Grubbs G, Ramelli SC, Levine AR, Bathula A, Saharia K, Purcell M, Singireddy S, Dugan CL, Kirchoff L, Lankford A, Cipriano S, Curto RA, Wu J, Raja K, Kelley E, Herr D, Vannella KM, Ravichandran S, Tang J, Harris A, Sajadi M, Chertow DS, Grazioli A, and Khurana S
- Subjects
- Antibodies, Viral, Antibody Affinity, Female, Humans, Immunoglobulin G, Postpartum Period, SARS-CoV-2 genetics, Vaccines, Synthetic, mRNA Vaccines, COVID-19 prevention & control, COVID-19 Vaccines
- Abstract
Background: Limited knowledge exists in post-partum women regarding durability of SARS-CoV-2 vaccine-induced antibody responses and their neutralising ability against SARS-CoV-2 variants of concern (VOC)., Methods: We elucidated longitudinal mRNA vaccination-induced antibody profiles of 13 post-partum and 13 non-post-partum women (control)., Findings: The antibody neutralisation titres against SARS-CoV-2 WA-1 strain were comparable between post-partum and non-post-partum women and these levels were sustained up to four months post-second vaccination in both groups. However, neutralisation titers declined against several VOCs, including Beta and Delta. Higher antibody binding was observed against SARS-CoV-2 receptor-binding domain (RBD) mutants with key VOC amino acids when tested with post-second vaccination plasma from post-partum women compared with controls. Importantly, post-vaccination plasma antibody affinity against VOCs RBDs was significantly higher in post-partum women compared with controls., Interpretation: This study demonstrates that there is a differential vaccination-induced immune responses in post-partum women compared with non-post-partum women, which could help inform future vaccination strategies for these groups., Funding: The antibody characterisation work described in this manuscript was supported by FDA's Medical Countermeasures Initiative (MCMi) grant #OCET 2021-1565 to S.K and intramural FDA-CBER COVID-19 supplemental funds., Competing Interests: Declaration of interests The authors have nothing to declare., (Published by Elsevier B.V.)
- Published
- 2022
- Full Text
- View/download PDF
17. Autocrine vitamin D signaling switches off pro-inflammatory programs of T H 1 cells.
- Author
-
Chauss D, Freiwald T, McGregor R, Yan B, Wang L, Nova-Lamperti E, Kumar D, Zhang Z, Teague H, West EE, Vannella KM, Ramos-Benitez MJ, Bibby J, Kelly A, Malik A, Freeman AF, Schwartz DM, Portilla D, Chertow DS, John S, Lavender P, Kemper C, Lombardi G, Mehta NN, Cooper N, Lionakis MS, Laurence A, Kazemian M, and Afzali B
- Subjects
- 25-Hydroxyvitamin D3 1-alpha-Hydroxylase metabolism, Basic-Leucine Zipper Transcription Factors metabolism, Bronchoalveolar Lavage Fluid cytology, COVID-19 immunology, COVID-19 pathology, Complement C3a immunology, Complement C3b immunology, Humans, JNK Mitogen-Activated Protein Kinases metabolism, Lymphocyte Activation immunology, Receptors, Calcitriol metabolism, Respiratory Distress Syndrome immunology, Respiratory Distress Syndrome pathology, Respiratory Distress Syndrome virology, STAT3 Transcription Factor metabolism, Signal Transduction immunology, Transcription, Genetic genetics, Interferon-gamma immunology, Interleukin-10 immunology, SARS-CoV-2 immunology, Th1 Cells immunology, Vitamin D metabolism
- Abstract
The molecular mechanisms governing orderly shutdown and retraction of CD4
+ type 1 helper T (TH 1) cell responses remain poorly understood. Here we show that complement triggers contraction of TH 1 responses by inducing intrinsic expression of the vitamin D (VitD) receptor and the VitD-activating enzyme CYP27B1, permitting T cells to both activate and respond to VitD. VitD then initiated the transition from pro-inflammatory interferon-γ+ TH 1 cells to suppressive interleukin-10+ cells. This process was primed by dynamic changes in the epigenetic landscape of CD4+ T cells, generating super-enhancers and recruiting several transcription factors, notably c-JUN, STAT3 and BACH2, which together with VitD receptor shaped the transcriptional response to VitD. Accordingly, VitD did not induce interleukin-10 expression in cells with dysfunctional BACH2 or STAT3. Bronchoalveolar lavage fluid CD4+ T cells of patients with COVID-19 were TH 1-skewed and showed de-repression of genes downregulated by VitD, from either lack of substrate (VitD deficiency) and/or abnormal regulation of this system., (© 2021. This is a U.S. government work and not under copyright protection in the U.S.; foreign copyright protection may apply.)- Published
- 2022
- Full Text
- View/download PDF
18. Evidence of SARS-CoV-2-Specific T-Cell-Mediated Myocarditis in a MIS-A Case.
- Author
-
Vannella KM, Oguz C, Stein SR, Pittaluga S, Dikoglu E, Kanwal A, Ramelli SC, Briese T, Su L, Wu X, Ramos-Benitez MJ, Perez-Valencia LJ, Babyak A, Cha NR, Chung JY, Ylaya K, Madathil RJ, Saharia KK, Scalea TM, Tran QK, Herr DL, Kleiner DE, Hewitt SM, Notarangelo LD, Grazioli A, and Chertow DS
- Subjects
- Adult, COVID-19 immunology, COVID-19 pathology, Humans, Male, Myocarditis immunology, RNA, Viral analysis, SARS-CoV-2, Systemic Inflammatory Response Syndrome immunology, COVID-19 complications, Myocarditis pathology, Myocarditis virology, Systemic Inflammatory Response Syndrome pathology, T-Lymphocytes immunology
- Abstract
A 26-year-old otherwise healthy man died of fulminant myocarditis. Nasopharyngeal specimens collected premortem tested negative for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Histopathological evaluation of the heart showed myocardial necrosis surrounded by cytotoxic T-cells and tissue-repair macrophages. Myocardial T-cell receptor (TCR) sequencing revealed hyper-dominant clones with highly similar sequences to TCRs that are specific for SARS-CoV-2 epitopes. SARS-CoV-2 RNA was detected in the gut, supporting a diagnosis of multisystem inflammatory syndrome in adults (MIS-A). Molecular targets of MIS-associated inflammation are not known. Our data indicate that SARS-CoV-2 antigens selected high-frequency T-cell clones that mediated fatal myocarditis., Competing Interests: Authors CO, LS and XW were employed by company Leidos Biomedical Research, Inc. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Vannella, Oguz, Stein, Pittaluga, Dikoglu, Kanwal, Ramelli, Briese, Su, Wu, Ramos-Benitez, Perez-Valencia, Babyak, Cha, Chung, Ylaya, Madathil, Saharia, Scalea, Tran, Herr, Kleiner, Hewitt, Notarangelo, Grazioli and Chertow.)
- Published
- 2021
- Full Text
- View/download PDF
19. In Utero Severe Acute Respiratory Syndrome Coronavirus 2 Infection.
- Author
-
Von Kohorn I, Stein SR, Shikani BT, Ramos-Benitez MJ, Vannella KM, Hewitt SM, Kleiner DE, Alejo JC, Burbelo P, Cohen JI, Wiedermann BL, and Chertow DS
- Subjects
- COVID-19 complications, COVID-19 Testing, Female, Fetal Blood virology, Humans, Infant, Newborn, Infant, Newborn, Diseases diagnosis, Infant, Newborn, Diseases etiology, Male, Placenta virology, Pregnancy, RNA, Viral blood, RNA, Viral urine, SARS-CoV-2 genetics, COVID-19 transmission, Infant, Newborn, Diseases virology, Infectious Disease Transmission, Vertical, Pregnancy Complications, Infectious virology
- Abstract
Evidence for in utero transmission of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is growing but not definitive. We present a case of neonatal infection that supports in utero transmission of SARS-CoV-2 and provides insight into the hematogenous spread from mother to fetus., (© The Author(s) 2020. Published by Oxford University Press on behalf of The Journal of the Pediatric Infectious Diseases Society. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2020
- Full Text
- View/download PDF
20. Nonhuman primates exposed to Zika virus in utero are not protected against reinfection at 1 year postpartum.
- Author
-
Vannella KM, Stein S, Connelly M, Swerczek J, Amaro-Carambot E, Coyle EM, Babyak A, Winkler CW, Saturday G, Gai ND, Hammoud DA, Dowd KA, Valencia LP, Ramos-Benitez MJ, Kindrachuk J, Pierson TC, Peterson KE, Brenchley JM, Whitehead SS, Khurana S, Herbert R, and Chertow DS
- Subjects
- Animals, Female, Humans, Macaca mulatta, Postpartum Period, Pregnancy, Reinfection, Pregnancy Complications, Infectious, Zika Virus, Zika Virus Infection
- Abstract
There is limited information about the impact of Zika virus (ZIKV) exposure in utero on the anti-ZIKV immune responses of offspring. We infected six rhesus macaque dams with ZIKV early or late in pregnancy and studied four of their offspring over the course of a year postpartum. Despite evidence of ZIKV exposure in utero, we observed no structural brain abnormalities in the offspring. We detected infant-derived ZIKV-specific immunoglobulin A antibody responses and T cell memory responses during the first year postpartum in the two offspring born to dams infected with ZIKV early in pregnancy. Critically, although the infants had acquired some immunological memory of ZIKV, it was not sufficient to protect them against reinfection with ZIKV at 1 year postpartum. The four offspring reexposed to ZIKV at 1 year postpartum all survived but exhibited acute viremia and viral tropism to lymphoid tissues; three of four reexposed offspring exhibited spinal cord pathology. These data suggest that macaque infants born to dams infected with ZIKV during pregnancy remain susceptible to postnatal infection and consequent neuropathology., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2020
- Full Text
- View/download PDF
21. Anti-IL-13Rα2 therapy promotes recovery in a murine model of inflammatory bowel disease.
- Author
-
Karmele EP, Pasricha TS, Ramalingam TR, Thompson RW, Gieseck RL 3rd, Knilans KJ, Hegen M, Farmer M, Jin F, Kleinman A, Hinds DA, Almeida Pereira T, de Queiroz Prado R, Bing N, Tchistiakova L, Kasaian MT, Wynn TA, and Vannella KM
- Subjects
- Animals, Crohn Disease etiology, Crohn Disease metabolism, Crohn Disease pathology, Dextran Sulfate adverse effects, Disease Models, Animal, Disease Susceptibility, Eosinophils immunology, Eosinophils metabolism, Gain of Function Mutation, Genetic Variation, Humans, Immunity, Inflammatory Bowel Diseases drug therapy, Inflammatory Bowel Diseases etiology, Inflammatory Bowel Diseases pathology, Interleukin-13 Receptor alpha2 Subunit genetics, Mice, Odds Ratio, Anti-Inflammatory Agents pharmacology, Antibodies, Monoclonal pharmacology, Inflammatory Bowel Diseases metabolism, Interleukin-13 Receptor alpha2 Subunit antagonists & inhibitors, Interleukin-13 Receptor alpha2 Subunit metabolism
- Abstract
There continues to be a major need for more effective inflammatory bowel disease (IBD) therapies. IL-13Rα2 is a decoy receptor that binds the cytokine IL-13 with high affinity and diminishes its STAT6-mediated effector functions. Previously, we found that IL-13Rα2 was necessary for IBD in mice deficient in the anti-inflammatory cytokine IL-10. Here, we tested for the first time a therapeutic antibody specifically targeting IL-13Rα2. We also used the antibody and Il13ra2
-/- mice to dissect the role of IL-13Rα2 in IBD pathogenesis and recovery. Il13ra2- /- mice were modestly protected from induction of dextran sodium sulfate (DSS)-induced colitis. Following a 7-day recovery period, Il13ra2-/- mice or wild-type mice administered the IL-13Rα2-neutralizing antibody had significantly improved colon health compared to control mice. Neutralizing IL-13Rα2 to increase IL-13 bioavailability promoted resolution of IBD even if neutralization occurred only during recovery. To link our observations in mice to a large human cohort, we conducted a phenome-wide association study of a more active variant of IL-13 (R130Q) that has reduced affinity for IL-13Rα2. Human subjects carrying R130Q reported a lower risk for Crohn's disease. Our findings endorse moving anti-IL-13Rα2 into preclinical drug development with the goal of accelerating recovery and maintaining remission in Crohn's disease patients.- Published
- 2019
- Full Text
- View/download PDF
22. Fibroblast-specific integrin-alpha V differentially regulates type 17 and type 2 driven inflammation and fibrosis.
- Author
-
Sciurba JC, Gieseck RL, Jiwrajka N, White SD, Karmele EP, Redes J, Vannella KM, Henderson NC, Wynn TA, and Hart KM
- Subjects
- Animals, Asthma metabolism, Asthma prevention & control, Disease Models, Animal, Female, Fibrosis, Gene Deletion, Inflammation pathology, Integrin alpha5 genetics, Interleukin-13 antagonists & inhibitors, Interleukin-13 immunology, Liver Cirrhosis immunology, Liver Cirrhosis pathology, Liver Cirrhosis prevention & control, Male, Mice, Knockout, Pulmonary Fibrosis immunology, Pulmonary Fibrosis pathology, Pulmonary Fibrosis prevention & control, Fibroblasts metabolism, Inflammation metabolism, Integrin alpha5 physiology
- Abstract
Fibroproliferative diseases affect a significant proportion of the world's population. Despite this, core mechanisms driving organ fibrosis of diverse etiologies remain ill defined. Recent studies suggest that integrin-alpha V serves as a master driver of fibrosis in multiple organs. Although diverse mechanisms contribute to the progression of fibrosis, TGF-β and IL-13 have emerged as central mediators of fibrosis during type 1/type 17, and type 2 polarized inflammatory responses, respectively. To investigate if integrin-alpha V interactions or signaling is critical to the development of type 2 fibrosis, we analyzed fibroblast-specific integrin-alpha V knockout mice in three type 2-driven inflammatory disease models. While we confirmed a role for integrin-alpha V in type 17-associated fibrosis, integrin-alpha V was not critical to the development of type 2-driven fibrosis. Additionally, our studies support a novel mechanism through which fibroblasts, via integrin-alpha V expression, are capable of regulating immune polarization. We show that when integrin-alpha V is deleted on fibroblasts, initiation of type 17 inflammation is inhibited leading to a deregulation of type 2 inflammation. This mechanism is most evident in a model of severe asthma, which is characterized by a mixed type 2/type 17 inflammatory response. Together, these findings suggest dual targeting of integrin-alpha V and type 2 pathways may be needed to ameliorate fibrosis and prevent rebound of opposing pro-fibrotic and inflammatory mechanisms. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd., (Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
23. Type 2 immunity is protective in metabolic disease but exacerbates NAFLD collaboratively with TGF-β.
- Author
-
Hart KM, Fabre T, Sciurba JC, Gieseck RL 3rd, Borthwick LA, Vannella KM, Acciani TH, de Queiroz Prado R, Thompson RW, White S, Soucy G, Bilodeau M, Ramalingam TR, Arron JR, Shoukry NH, and Wynn TA
- Subjects
- Adipose Tissue pathology, Animals, Diet, High-Fat, Eosinophils pathology, Humans, Inflammation pathology, Interferon-gamma deficiency, Interferon-gamma metabolism, Liver Cirrhosis pathology, Male, Mice, Inbred C57BL, Non-alcoholic Fatty Liver Disease pathology, Obesity pathology, Disease Progression, Immunity, Metabolic Diseases immunology, Metabolic Diseases prevention & control, Non-alcoholic Fatty Liver Disease immunology, Non-alcoholic Fatty Liver Disease prevention & control, Transforming Growth Factor beta metabolism
- Abstract
Nonalcoholic fatty liver disease (NAFLD) is now the most common progressive liver disease in developed countries and is the second leading indication for liver transplantation due to the extensive fibrosis it causes. NAFLD progression is thought to be tied to chronic low-level type 1 inflammation originating in the adipose tissue during obesity; however, the specific immunological mechanisms regulating the progression of NAFLD-associated fibrosis in the liver are unclear. To investigate the immunopathogenesis of NAFLD more completely, we investigated adipose dysfunction, nonalcoholic steatohepatitis (NASH), and fibrosis in mice that develop polarized type 1 or type 2 immune responses. Unexpectedly, obese interleukin-10 (IL-10)/IL-4-deficient mice (type 1-polarized) were highly resistant to NASH. This protection was associated with an increased hepatic interferon-γ (IFN-γ) signature. Conversely, IFN-γ-deficient mice progressed rapidly to NASH with evidence of fibrosis dependent on transforming growth factor-β (TGF-β) and IL-13 signaling. Unlike increasing type 1 inflammation and the marked loss of eosinophils seen in expanding adipose tissue, progression of NASH was associated with increasing eosinophilic type 2 liver inflammation in mice and human patient biopsies. Finally, simultaneous inhibition of TGF-β and IL-13 signaling attenuated the fibrotic machinery more completely than TGF-β alone in NAFLD-associated fibrosis. Thus, although type 2 immunity maintains healthy metabolic signaling in adipose tissues, it exacerbates the progression of NAFLD collaboratively with TGF-β in the liver., (Copyright © 2017 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2017
- Full Text
- View/download PDF
24. Mechanisms of Organ Injury and Repair by Macrophages.
- Author
-
Vannella KM and Wynn TA
- Subjects
- Animals, Cell Differentiation physiology, Homeostasis physiology, Humans, Macrophages physiology, Regeneration physiology, Wound Healing physiology
- Abstract
Macrophages regulate tissue regeneration following injury. They can worsen tissue injury by producing reactive oxygen species and other toxic mediators that disrupt cell metabolism, induce apoptosis, and exacerbate ischemic injury. However, they also produce a variety of growth factors, such as IGF-1, VEGF-α, TGF-β, and Wnt proteins that regulate epithelial and endothelial cell proliferation, myofibroblast activation, stem and tissue progenitor cell differentiation, and angiogenesis. Proresolving macrophages in turn restore tissue homeostasis by functioning as anti-inflammatory cells, and macrophage-derived matrix metalloproteinases regulate fibrin and collagen turnover. However, dysregulated macrophage function impairs wound healing and contributes to the development of fibrosis. Consequently, the mechanisms that regulate these different macrophage activation states have become active areas of research. In this review, we discuss the common and unique mechanisms by which macrophages instruct tissue repair in the liver, nervous system, heart, lung, skeletal muscle, and intestine and illustrate how macrophages might be exploited therapeutically.
- Published
- 2017
- Full Text
- View/download PDF
25. Interleukin-13 Activates Distinct Cellular Pathways Leading to Ductular Reaction, Steatosis, and Fibrosis.
- Author
-
Gieseck RL 3rd, Ramalingam TR, Hart KM, Vannella KM, Cantu DA, Lu WY, Ferreira-González S, Forbes SJ, Vallier L, and Wynn TA
- Subjects
- Animals, Bile Acids and Salts biosynthesis, Cell Proliferation, Cells, Cultured, Fibrosis, Humans, Interleukin-13 genetics, Interleukin-13 immunology, Mice, Mice, Inbred C57BL, Mice, Knockout, Signal Transduction, Th2 Cells immunology, Fatty Liver immunology, Fibroblasts immunology, Interleukin-13 metabolism, Liver pathology, Liver Cirrhosis, Biliary immunology, Schistosoma mansoni immunology, Schistosomiasis mansoni immunology
- Abstract
Fibroproliferative diseases are driven by dysregulated tissue repair responses and are a major cause of morbidity and mortality because they affect nearly every organ system. Type 2 cytokine responses are critically involved in tissue repair; however, the mechanisms that regulate beneficial regeneration versus pathological fibrosis are not well understood. Here, we have shown that the type 2 effector cytokine interleukin-13 simultaneously, yet independently, directed hepatic fibrosis and the compensatory proliferation of hepatocytes and biliary cells in progressive models of liver disease induced by interleukin-13 overexpression or after infection with Schistosoma mansoni. Using transgenic mice with interleukin-13 signaling genetically disrupted in hepatocytes, cholangiocytes, or resident tissue fibroblasts, we have revealed direct and distinct roles for interleukin-13 in fibrosis, steatosis, cholestasis, and ductular reaction. Together, these studies show that these mechanisms are simultaneously controlled but distinctly regulated by interleukin-13 signaling. Thus, it may be possible to promote interleukin-13-dependent hepatobiliary expansion without generating pathological fibrosis. VIDEO ABSTRACT., (Published by Elsevier Inc.)
- Published
- 2016
- Full Text
- View/download PDF
26. Enhanced protection from fibrosis and inflammation in the combined absence of IL-13 and IFN-γ.
- Author
-
Ramalingam TR, Gieseck RL, Acciani TH, M Hart K, Cheever AW, Mentink-Kane MM, Vannella KM, and Wynn TA
- Subjects
- Animals, Antibodies, Neutralizing, Female, Granuloma, Humans, Inflammation, Interferon-gamma metabolism, Interleukin-13 metabolism, Liver metabolism, Liver pathology, Liver Cirrhosis immunology, Liver Cirrhosis pathology, Lung metabolism, Lung pathology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Pulmonary Fibrosis immunology, Pulmonary Fibrosis pathology, Schistosomiasis mansoni parasitology, Schistosomiasis mansoni pathology, Signal Transduction, Tumor Necrosis Factor-alpha genetics, Tumor Necrosis Factor-alpha metabolism, Interferon-gamma genetics, Interleukin-13 genetics, Liver Cirrhosis prevention & control, Pulmonary Fibrosis prevention & control, Schistosoma mansoni immunology, Schistosomiasis mansoni prevention & control
- Abstract
Persistent or dysregulated IL-13 responses are key drivers of fibrosis in multiple organ systems, and this identifies this cytokine as an important therapeutic target. Nevertheless, the mechanisms by which IL-13 blockade leads to the amelioration of fibrosis remain unclear. Because IFN-γ exhibits potent anti-fibrotic activity, and IL-4Rα signalling antagonizes IFN-γ effector function, compensatory increases in IFN-γ activity following IL-13/IL-4Rα blockade might contribute to the reduction in fibrosis. To investigate the role of IFN-γ, we developed novel IL-13(-/-) /IFN-γ(-/-) double cytokine-deficient mice and examined disease progression in models of type 2-driven fibrosis. As predicted, we showed that fibrosis in the lung and liver are both highly dependent on IL-13. We also observed increased IFN-γ production and inflammatory activity in the tissues of IL-13-deficient mice. Surprisingly, however, an even greater reduction in fibrosis was observed in IL-13/IFN-γ double deficient mice, most notably in the livers of mice chronically infected with Schistosoma mansoni. The increased protection was associated with marked decreases in Tgfb1, Mmp12, and Timp1 mRNA expression in the tissues; reduced inflammation; and decreased expression of important pro-inflammatory mediators such as TNF-α. Experiments conducted with neutralizing monoclonal antibodies to IL-13 and IFN-γ validated the findings with the genetically deficient mice. Together, these studies demonstrate that the reduction in fibrosis observed when IL-13 signalling is suppressed is not dependent on increased IFN-γ activity. Instead, by reducing compensatory increases in type 1-associated inflammation, therapeutic strategies that block IFN-γ and IL-13 activity simultaneously can confer greater protection from progressive fibrosis than IL-13 blockade alone. Published 2016. This article is a U.S. Government work and is in the public domain in the USA., (Published 2016. This article is a U.S. Government work and is in the public domain in the USA.)
- Published
- 2016
- Full Text
- View/download PDF
27. Combinatorial targeting of TSLP, IL-25, and IL-33 in type 2 cytokine-driven inflammation and fibrosis.
- Author
-
Vannella KM, Ramalingam TR, Borthwick LA, Barron L, Hart KM, Thompson RW, Kindrachuk KN, Cheever AW, White S, Budelsky AL, Comeau MR, Smith DE, and Wynn TA
- Subjects
- Animals, Antibodies, Monoclonal therapeutic use, Cytokines antagonists & inhibitors, Cytokines genetics, Female, Fibrosis drug therapy, Fibrosis therapy, Granuloma drug therapy, Granuloma immunology, Granuloma parasitology, Granuloma therapy, Inflammation drug therapy, Interleukin-13 antagonists & inhibitors, Interleukin-13 genetics, Interleukin-13 metabolism, Interleukin-17 antagonists & inhibitors, Interleukin-17 genetics, Interleukin-33 antagonists & inhibitors, Interleukin-33 genetics, Interleukin-4 antagonists & inhibitors, Interleukin-4 genetics, Interleukin-4 metabolism, Lung Neoplasms drug therapy, Lung Neoplasms parasitology, Lung Neoplasms therapy, Male, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Knockout, Schistosoma mansoni immunology, Schistosoma mansoni pathogenicity, Thymic Stromal Lymphopoietin, Cytokines metabolism, Fibrosis immunology, Inflammation immunology, Inflammation therapy, Interleukin-17 metabolism, Interleukin-33 metabolism, Lung Neoplasms immunology
- Abstract
Thymic stromal lymphopoietin (TSLP), interleukin-25 (IL-25), and IL-33 are important initiators of type 2-associated mucosal inflammation and immunity. However, their role in the maintenance of progressive type 2 inflammation and fibrosis is much less clear. Using chronic models of helminth infection and allergic lung inflammation, we show that collective disruption of TSLP, IL-25, and IL-33 signaling suppresses chronic and progressive type 2 cytokine-driven inflammation and fibrosis. In a schistosome lung granuloma model or during chronic Schistosoma mansoni infection in the liver, individual ablation of TSLP, IL-25, or IL-33/ST2 had no impact on the development of IL-4/IL-13-dependent inflammation or fibrosis. However, significant reductions in granuloma-associated eosinophils, hepatic fibrosis, and IL-13-producing type 2 innate lymphoid cells (ILC2s) were observed when signaling of all three mediators was simultaneously disrupted. Combined blockade through monoclonal antibody (mAb) treatment also reduced IL-5 and IL-13 expression during primary and secondary granuloma formation in the lungs. In a model of chronic house dust mite-induced allergic lung inflammation, combined mAb treatment did not decrease established inflammation or fibrosis. TSLP/IL-33 double-knockout mice treated with anti-IL-25 mAb during priming, however, displayed decreased inflammation, mucus production, and lung remodeling in the chronic phase. Together, these studies reveal partially redundant roles for TSLP, IL-25, and IL-33 in the maintenance of type 2 pathology and suggest that in some settings, early combined targeting of these mediators is necessary to ameliorate progressive type 2-driven disease., (Copyright © 2016, American Association for the Advancement of Science.)
- Published
- 2016
- Full Text
- View/download PDF
28. Acidic chitinase primes the protective immune response to gastrointestinal nematodes.
- Author
-
Vannella KM, Ramalingam TR, Hart KM, de Queiroz Prado R, Sciurba J, Barron L, Borthwick LA, Smith AD, Mentink-Kane M, White S, Thompson RW, Cheever AW, Bock K, Moore I, Fitz LJ, Urban JF Jr, and Wynn TA
- Subjects
- Animals, Chitinases genetics, Chitinases metabolism, Chloride Channels genetics, Chloride Channels immunology, Chloride Channels metabolism, Flow Cytometry, Gastrointestinal Tract metabolism, Gastrointestinal Tract parasitology, Gene Expression immunology, Hormones, Ectopic genetics, Hormones, Ectopic immunology, Hormones, Ectopic metabolism, Host-Parasite Interactions immunology, Hypersensitivity genetics, Hypersensitivity immunology, Hypersensitivity metabolism, Immunity genetics, Intercellular Signaling Peptides and Proteins, Interleukin-13 genetics, Interleukin-13 immunology, Interleukin-13 metabolism, Lectins genetics, Lectins immunology, Lectins metabolism, Lung immunology, Lung metabolism, Lung pathology, Mice, Inbred C57BL, Mice, Knockout, Microscopy, Fluorescence, Nematospiroides dubius immunology, Nematospiroides dubius physiology, Nippostrongylus immunology, Nippostrongylus physiology, Reverse Transcriptase Polymerase Chain Reaction, Strongylida Infections metabolism, Strongylida Infections parasitology, beta-N-Acetylhexosaminidases genetics, beta-N-Acetylhexosaminidases immunology, beta-N-Acetylhexosaminidases metabolism, Chitinases immunology, Gastrointestinal Tract immunology, Immunity immunology, Strongylida Infections immunology
- Abstract
Acidic mammalian chitinase (AMCase) is known to be induced by allergens and helminths, yet its role in immunity is unclear. Using AMCase-deficient mice, we show that AMCase deficiency reduced the number of group 2 innate lymphoid cells during allergen challenge but was not required for establishment of type 2 inflammation in the lung in response to allergens or helminths. In contrast, AMCase-deficient mice showed a profound defect in type 2 immunity following infection with the chitin-containing gastrointestinal nematodes Nippostrongylus brasiliensis and Heligmosomoides polygyrus bakeri. The impaired immunity was associated with reduced mucus production and decreased intestinal expression of the signature type 2 response genes Il13, Chil3, Retnlb, and Clca1. CD103(+) dendritic cells, which regulate T cell homing, were also reduced in mesenteric lymph nodes of infected AMCase-deficient mice. Thus, AMCase functions as a critical initiator of protective type 2 responses to intestinal nematodes but is largely dispensable for allergic responses in the lung.
- Published
- 2016
- Full Text
- View/download PDF
29. Macrophages in Tissue Repair, Regeneration, and Fibrosis.
- Author
-
Wynn TA and Vannella KM
- Subjects
- Animals, Cell Communication, Cell Differentiation, Fibrosis, Humans, Macrophage Activation, Macrophages pathology, Phenotype, Macrophages physiology, Regeneration, Wound Healing
- Abstract
Inflammatory monocytes and tissue-resident macrophages are key regulators of tissue repair, regeneration, and fibrosis. After tissue injury, monocytes and macrophages undergo marked phenotypic and functional changes to play critical roles during the initiation, maintenance, and resolution phases of tissue repair. Disturbances in macrophage function can lead to aberrant repair, such that uncontrolled production of inflammatory mediators and growth factors, deficient generation of anti-inflammatory macrophages, or failed communication between macrophages and epithelial cells, endothelial cells, fibroblasts, and stem or tissue progenitor cells all contribute to a state of persistent injury, and this could lead to the development of pathological fibrosis. In this review, we discuss the mechanisms that instruct macrophages to adopt pro-inflammatory, pro-wound-healing, pro-fibrotic, anti-inflammatory, anti-fibrotic, pro-resolving, and tissue-regenerating phenotypes after injury, and we highlight how some of these mechanisms and macrophage activation states could be exploited therapeutically., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
30. Macrophages are critical to the maintenance of IL-13-dependent lung inflammation and fibrosis.
- Author
-
Borthwick LA, Barron L, Hart KM, Vannella KM, Thompson RW, Oland S, Cheever A, Sciurba J, Ramalingam TR, Fisher AJ, and Wynn TA
- Subjects
- Animals, Antigens, Differentiation genetics, Antigens, Differentiation immunology, Antigens, Ly genetics, Antigens, Ly immunology, CD11b Antigen genetics, CD11b Antigen immunology, Fibrosis, Gene Expression Regulation, Heparin-binding EGF-like Growth Factor genetics, Heparin-binding EGF-like Growth Factor immunology, Interleukin-13 genetics, Lung immunology, Lung parasitology, Lung pathology, Macrophages, Alveolar parasitology, Macrophages, Alveolar pathology, Mice, Mice, Transgenic, Nippostrongylus immunology, Nippostrongylus pathogenicity, Pneumonia parasitology, Pneumonia pathology, Pyroglyphidae immunology, Schistosoma mansoni immunology, Schistosoma mansoni pathogenicity, Schistosomiasis mansoni parasitology, Schistosomiasis mansoni pathology, Signal Transduction, Strongylida Infections parasitology, Strongylida Infections pathology, Th2 Cells parasitology, Th2 Cells pathology, Interleukin-13 immunology, Macrophages, Alveolar immunology, Pneumonia immunology, Schistosomiasis mansoni immunology, Strongylida Infections immunology, Th2 Cells immunology
- Abstract
The roles of macrophages in type 2-driven inflammation and fibrosis remain unclear. Here, using CD11b-diphtheria toxin receptor (DTR) transgenic mice and three models of interleukin 13 (IL-13)-dependent inflammation, fibrosis, and immunity, we show that CD11b(+) F4/80(+) Ly6C(+) macrophages are required for the maintenance of type 2 immunity within affected tissues but not secondary lymphoid organs. Direct depletion of macrophages during the maintenance or resolution phases of secondary Schistosoma mansoni egg-induced granuloma formation caused a profound decrease in inflammation, fibrosis, and type 2 gene expression. Additional studies with CD11c-DTR and CD11b/CD11c-DTR double-transgenic mice suggested that macrophages but not dendritic cells were critical. Mechanistically, macrophage depletion impaired effector CD4(+) T helper type 2 (Th2) cell homing and activation within the inflamed lung. Depletion of CD11b(+) F4/80(+) Ly6C(+) macrophages similarly reduced house dust mite-induced allergic lung inflammation and suppressed IL-13-dependent immunity to the nematode parasite Nippostrongylus brasiliensis. Consequently, therapeutic strategies targeting macrophages offer a novel approach to ameliorate established type 2 inflammatory diseases.
- Published
- 2016
- Full Text
- View/download PDF
31. TH2 and TH17 inflammatory pathways are reciprocally regulated in asthma.
- Author
-
Choy DF, Hart KM, Borthwick LA, Shikotra A, Nagarkar DR, Siddiqui S, Jia G, Ohri CM, Doran E, Vannella KM, Butler CA, Hargadon B, Sciurba JC, Gieseck RL, Thompson RW, White S, Abbas AR, Jackman J, Wu LC, Egen JG, Heaney LG, Ramalingam TR, Arron JR, Wynn TA, and Bradding P
- Subjects
- Animals, Cells, Cultured, Female, Humans, Interleukin-13 metabolism, Interleukin-17 metabolism, Mice, Mice, Inbred BALB C, Signal Transduction, Asthma immunology, Asthma metabolism, Th17 Cells metabolism, Th2 Cells metabolism
- Abstract
Increasing evidence suggests that asthma is a heterogeneous disorder regulated by distinct molecular mechanisms. In a cross-sectional study of asthmatics of varying severity (n = 51), endobronchial tissue gene expression analysis revealed three major patient clusters: TH2-high, TH17-high, and TH2/17-low. TH2-high and TH17-high patterns were mutually exclusive in individual patient samples, and their gene signatures were inversely correlated and differentially regulated by interleukin-13 (IL-13) and IL-17A. To understand this dichotomous pattern of T helper 2 (TH2) and TH17 signatures, we investigated the potential of type 2 cytokine suppression in promoting TH17 responses in a preclinical model of allergen-induced asthma. Neutralization of IL-4 and/or IL-13 resulted in increased TH17 cells and neutrophilic inflammation in the lung. However, neutralization of IL-13 and IL-17 protected mice from eosinophilia, mucus hyperplasia, and airway hyperreactivity and abolished the neutrophilic inflammation, suggesting that combination therapies targeting both pathways may maximize therapeutic efficacy across a patient population comprising both TH2 and TH17 endotypes., (Copyright © 2015, American Association for the Advancement of Science.)
- Published
- 2015
- Full Text
- View/download PDF
32. Incomplete deletion of IL-4Rα by LysM(Cre) reveals distinct subsets of M2 macrophages controlling inflammation and fibrosis in chronic schistosomiasis.
- Author
-
Vannella KM, Barron L, Borthwick LA, Kindrachuk KN, Narasimhan PB, Hart KM, Thompson RW, White S, Cheever AW, Ramalingam TR, and Wynn TA
- Subjects
- Animals, Cells, Cultured, Chronic Disease, Cytokines genetics, Cytokines metabolism, Disease Models, Animal, Female, Fibrosis parasitology, Fibrosis pathology, Inflammation parasitology, Inflammation pathology, Integrases metabolism, Macrophages, Peritoneal parasitology, Macrophages, Peritoneal pathology, Male, Mice, Mice, Inbred BALB C, Neutrophils immunology, Neutrophils parasitology, Neutrophils pathology, RNA, Messenger genetics, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, Schistosomiasis parasitology, Schistosomiasis pathology, Fibrosis immunology, Inflammation immunology, Macrophages, Peritoneal immunology, Receptors, Cell Surface physiology, Schistosoma mansoni pathogenicity, Schistosomiasis immunology
- Abstract
Mice expressing a Cre recombinase from the lysozyme M-encoding locus (Lyz2) have been widely used to dissect gene function in macrophages and neutrophils. Here, we show that while naïve resident tissue macrophages from IL-4Rαf(lox/delta)LysM(Cre) mice almost completely lose IL-4Rα function, a large fraction of macrophages elicited by sterile inflammatory stimuli, Schistosoma mansoni eggs, or S. mansoni infection, fail to excise Il4rα. These F4/80(hi)CD11b(hi) macrophages, in contrast to resident tissue macrophages, express lower levels of Lyz2 explaining why this population resists LysM(Cre)-mediated deletion. We show that in response to IL-4 and IL-13, Lyz2(lo)IL-4Rα(+) macrophages differentiate into an arginase 1-expressing alternatively-activated macrophage (AAM) population, which slows the development of lethal fibrosis in schistosomiasis. In contrast, we identified Lyz2(hi)IL-4Rα(+) macrophages as the key subset of AAMs mediating the downmodulation of granulomatous inflammation in chronic schistosomiasis. Our observations reveal a limitation on using a LysMCre mouse model to study gene function in inflammatory settings, but we utilize this limitation as a means to demonstrate that distinct populations of alternatively activated macrophages control inflammation and fibrosis in chronic schistosomiasis.
- Published
- 2014
- Full Text
- View/download PDF
33. Chitinase dependent control of protozoan cyst burden in the brain.
- Author
-
Nance JP, Vannella KM, Worth D, David C, Carter D, Noor S, Hubeau C, Fitz L, Lane TE, Wynn TA, and Wilson EH
- Subjects
- Animals, Brain microbiology, Brain pathology, Brain Diseases microbiology, Brain Diseases pathology, Chitinases immunology, Cysts immunology, Cysts pathology, Macrophages pathology, Mice, Th1 Cells pathology, Toxoplasmosis pathology, Brain immunology, Brain Diseases immunology, Macrophages immunology, Th1 Cells immunology, Toxoplasma immunology, Toxoplasmosis immunology
- Abstract
Chronic infections represent a continuous battle between the host's immune system and pathogen replication. Many protozoan parasites have evolved a cyst lifecycle stage that provides it with increased protection from environmental degradation as well as endogenous host mechanisms of attack. In the case of Toxoplasma gondii, these cysts are predominantly found in the immune protected brain making clearance of the parasite more difficult and resulting in a lifelong infection. Currently, little is known about the nature of the immune response stimulated by the presence of these cysts or how they are able to propagate. Here we establish a novel chitinase-dependent mechanism of cyst control in the infected brain. Despite a dominant Th1 immune response during Toxoplasma infection there exists a population of alternatively activated macrophages (AAMØ) in the infected CNS. These cells are capable of cyst lysis via the production of AMCase as revealed by live imaging, and this chitinase is necessary for protective immunity within the CNS. These data demonstrate chitinase activity in the brain in response to a protozoan pathogen and provide a novel mechanism to facilitate cyst clearance during chronic infections.
- Published
- 2012
- Full Text
- View/download PDF
34. TLR9-induced interferon β is associated with protection from gammaherpesvirus-induced exacerbation of lung fibrosis.
- Author
-
Luckhardt TR, Coomes SM, Trujillo G, Stoolman JS, Vannella KM, Bhan U, Wilke CA, Moore TA, Toews GB, Hogaboam C, and Moore BB
- Abstract
Background: We have shown previously that murine gammaherpesvirus 68 (γHV68) infection exacerbates established pulmonary fibrosis. Because Toll-like receptor (TLR)-9 may be important in controlling the immune response to γHV68 infection, we examined how TLR-9 signaling effects exacerbation of fibrosis in response to viral infection, using models of bleomycin- and fluorescein isothiocyanate-induced pulmonary fibrosis in wild-type (Balb/c) and TLR-9-/- mice., Results: We found that in the absence of TLR-9 signaling, there was a significant increase in collagen deposition following viral exacerbation of fibrosis. This was not associated with increased viral load in TLR-9-/- mice or with major alterations in T helper (Th)1 and Th2 cytokines. We examined alveolar epithelial-cell apoptosis in both strains, but this could not explain the altered fibrotic outcomes. As expected, TLR-9-/- mice had a defect in the production of interferon (IFN)-β after viral infection. Balb/c fibroblasts infected with γHV68 in vitro produced more IFN-β than did infected TLR-9-/- fibroblasts. Accordingly, in vitro infection of Balb/c fibroblasts resulted in reduced proliferation rates whereas infection of TLR-9-/- fibroblasts did not. Finally, therapeutic administration of CpG oligodeoxynucleotides ameliorated bleomycin-induced fibrosis in wild-type mice., Conclusions: These results show a protective role for TLR-9 signaling in murine models of lung fibrosis, and highlight differences in the biology of TLR-9 between mice and humans.
- Published
- 2011
- Full Text
- View/download PDF
35. New concepts of IL-10-induced lung fibrosis: fibrocyte recruitment and M2 activation in a CCL2/CCR2 axis.
- Author
-
Sun L, Louie MC, Vannella KM, Wilke CA, LeVine AM, Moore BB, and Shanley TP
- Subjects
- Animals, Antibodies, Blocking pharmacology, Bronchoalveolar Lavage Fluid cytology, Cells, Cultured, Collagen biosynthesis, Collagen genetics, Fibroblasts drug effects, Fibroblasts metabolism, Gene Expression Regulation drug effects, Humans, Immunohistochemistry, Lung drug effects, Lung metabolism, Lung pathology, Lymphocytes cytology, Lymphocytes drug effects, Mice, Pulmonary Fibrosis metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Cell Movement drug effects, Chemokine CCL2 metabolism, Fibroblasts pathology, Interleukin-10 metabolism, Macrophage Activation drug effects, Pulmonary Fibrosis pathology, Receptors, CCR2 metabolism
- Abstract
IL-10 is most commonly recognized as an anti-inflammatory cytokine possessing immunosuppressive effects necessary for regulated resolution of proinflammation. However, its role in the development of fibrosis during inflammatory resolution has not been clear. Few prior studies have linked IL-10 with the inhibition of fibrosis principally on the basis of regulating inflammation thought to be driving fibroproliferation. In contrast, in a model of long-term overexpression of IL-10, we observed marked induction of lung fibrosis in mice. The total cell number retrieved by bronchoalveolar lavage (BAL) increased 10-fold in the IL-10 overexpression (IL-10 OE) mice, with significant infiltration of T and B lymphocytes and collagen-producing cells. The presence of increased fibrocytes, isolated from collagenase-digested lungs, was identified by flow cytometry using dual staining of CD45 and collagen 1. Quantitative PCR analysis on an array of chemokine/chemokine receptor genes showed that receptor CCR2 and its ligand, CCL2, were highly upregulated in IL-10 OE mice, suggesting that IL-10-induced fibrocyte recruitment was CCL2/CCR2 specific. Given the prior association of alternatively activated (M(2)) macrophages with development of fibrosis in other disease states, we also examined the effect of IL-10 OE on the M(2) macrophage axis. We observed significantly increased numbers of M(2) macrophages in both BAL and whole lung tissue from the IL-10 OE mice. Administration of rabbit anti-CCL2 antiserum to IL-10 OE mice for three consecutive weeks significantly decreased fibrosis as evidenced by lung hydroxyproline content, compared with mice that received preimmune rabbit serum. These results indicate that overexpression of IL-10 induces fibrosis, in part, by fibrocyte recruitment and M(2) macrophage activation, and likely in a CCL2/CCR2 axis.
- Published
- 2011
- Full Text
- View/download PDF
36. Latent infection by γherpesvirus stimulates profibrotic mediator release from multiple cell types.
- Author
-
Stoolman JS, Vannella KM, Coomes SM, Wilke CA, Sisson TH, Toews GB, and Moore BB
- Subjects
- Animals, Base Sequence, Chemokine CCL2 biosynthesis, DNA Primers genetics, DNA, Viral genetics, Disease Models, Animal, Herpesviridae Infections genetics, Herpesviridae Infections virology, Interferon-gamma biosynthesis, Interleukin-10 biosynthesis, Lung immunology, Lung virology, Macrophage Activation, Macrophages, Alveolar immunology, Macrophages, Alveolar virology, Male, Mesoderm immunology, Mesoderm virology, Mice, Mice, Inbred C57BL, Monocyte Chemoattractant Proteins biosynthesis, Pulmonary Fibrosis genetics, Pulmonary Fibrosis immunology, Pulmonary Fibrosis virology, Spleen immunology, Spleen virology, T-Lymphocytes immunology, T-Lymphocytes virology, Transforming Growth Factor beta1 biosynthesis, Tumor Necrosis Factor-alpha biosynthesis, Tumor Virus Infections genetics, Tumor Virus Infections virology, Viral Load, Cytokines biosynthesis, Herpesviridae Infections immunology, Pulmonary Fibrosis etiology, Rhadinovirus pathogenicity, Tumor Virus Infections immunology
- Abstract
Although γherpesvirus infections are associated with enhanced lung fibrosis in both clinical and animal studies, there is limited understanding about fibrotic effects of γherpesviruses on cell types present in the lung, particularly during latent infection. Wild-type mice were intranasally infected with a murine γherpesvirus (γHV-68) or mock-infected with saline. Twenty-eight days postinfection (dpi), ∼14 days following clearance of the lytic infection, alveolar macrophages (AMs), mesenchymal cells, and CD19-enriched cell populations from the lung and spleen express M(3) and/or glycoprotein B (gB) viral mRNA and harbor viral genome. AMs from infected mice express more transforming growth factor (TGF)-β(1), CCL2, CCL12, TNF-α, and IFN-γ than AMs from mock-infected mice. Mesenchymal cells express more total TGF-β(1), CCL12, and TNF-α than mesenchymal cells from mock-infected mice. Lung and spleen CD19-enriched cells express more total TGF-β(1) 28 dpi compared with controls. The CD19-negative fraction of the spleen overexpresses TGF-β(1) and harbors viral genome, but this likely represents infection of monocytes. Purified T cells from the lung harbor almost no viral genome. Purified T cells overexpress IL-10 but not TGF-β(1). Intracellular cytokine staining demonstrated that lung T cells at 28 dpi produce IFN-γ but not IL-4. Thus infection with a murine γherpesvirus is sufficient to upregulate profibrotic and proinflammatory factors in a variety of lung resident and circulating cell types 28 dpi. Our results provide new information about possible contributions of these cells to fibrogenesis in the lungs of individuals harboring a γherpesvirus infection and may help explain why γHV-68 infection can augment or exacerbate fibrotic responses in mice.
- Published
- 2011
- Full Text
- View/download PDF
37. The antifibrotic effects of plasminogen activation occur via prostaglandin E2 synthesis in humans and mice.
- Author
-
Bauman KA, Wettlaufer SH, Okunishi K, Vannella KM, Stoolman JS, Huang SK, Courey AJ, White ES, Hogaboam CM, Simon RH, Toews GB, Sisson TH, Moore BB, and Peters-Golden M
- Subjects
- Adult, Animals, Bleomycin adverse effects, Bleomycin metabolism, Bleomycin pharmacology, Collagen adverse effects, Collagen metabolism, Collagen pharmacology, Dinoprostone metabolism, Dinoprostone pharmacology, Extracellular Matrix metabolism, Fibrinolysin, Fibroblasts drug effects, Fibroblasts metabolism, Fibroblasts pathology, Fibrosis metabolism, Fibrosis pathology, Humans, Lung drug effects, Lung metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Plasminogen adverse effects, Plasminogen pharmacology, Plasminogen Activator Inhibitor 1 adverse effects, Plasminogen Activator Inhibitor 1 pharmacology, Pulmonary Fibrosis chemically induced, Pulmonary Fibrosis pathology, Receptor, PAR-1 metabolism, Cyclooxygenase 2 biosynthesis, Dinoprostone biosynthesis, Plasminogen metabolism, Plasminogen Activator Inhibitor 1 metabolism, Pulmonary Fibrosis metabolism
- Abstract
Plasminogen activation to plasmin protects from lung fibrosis, but the mechanism underlying this antifibrotic effect remains unclear. We found that mice lacking plasminogen activation inhibitor-1 (PAI-1), which are protected from bleomycin-induced pulmonary fibrosis, exhibit lung overproduction of the antifibrotic lipid mediator prostaglandin E2 (PGE2). Plasminogen activation upregulated PGE2 synthesis in alveolar epithelial cells, lung fibroblasts, and lung fibrocytes from saline- and bleomycin-treated mice, as well as in normal fetal and adult primary human lung fibroblasts. This response was exaggerated in cells from Pai1-/- mice. Although enhanced PGE2 formation required the generation of plasmin, it was independent of proteinase-activated receptor 1 (PAR-1) and instead reflected proteolytic activation and release of HGF with subsequent induction of COX-2. That the HGF/COX-2/PGE2 axis mediates in vivo protection from fibrosis in Pai1-/- mice was demonstrated by experiments showing that a selective inhibitor of the HGF receptor c-Met increased lung collagen to WT levels while reducing COX-2 protein and PGE2 levels. Of clinical interest, fibroblasts from patients with idiopathic pulmonary fibrosis were found to be defective in their ability to induce COX-2 and, therefore, unable to upregulate PGE2 synthesis in response to plasmin or HGF. These studies demonstrate crosstalk between plasminogen activation and PGE2 generation in the lung and provide a mechanism for the well-known antifibrotic actions of the fibrinolytic pathway.
- Published
- 2010
- Full Text
- View/download PDF
38. Secondary lymphoid organs contribute to, but are not required for the induction of graft-versus-host responses following allogeneic bone marrow transplantation: a shifting paradigm for T cell allo-activation.
- Author
-
Silva IA, Olkiewicz K, Askew D, Fisher JM, Chaudhary MN, Vannella KM, Deurloo DT, Choi SW, Pierce EM, Clouthier SG, Liu C, and Cooke KR
- Subjects
- Animals, Bone Marrow Transplantation adverse effects, Cell Proliferation, Cytotoxicity, Immunologic, Mice, Mice, Mutant Strains, Transplantation, Homologous, Bone Marrow Transplantation immunology, Graft vs Host Disease immunology, Lymphocyte Activation immunology, Lymphoid Tissue immunology, T-Lymphocytes immunology
- Abstract
Graft-versus-host disease (GVHD) remains the major complication of allogeneic bone marrow transplantation (allo-BMT). GVHD fundamentally depends upon the activation of donor T cells by host antigen-presenting cells (APCs), but the precise location of these interactions remains uncertain. We examined the role of secondary lymphoid organs (SLO) in the induction of GVHD by using homozygous aly/aly mice that are deficient in lymph nodes (LNs) and Peyer's patches (PPs). Lethally irradiated, splenectomized, aly/aly (LN/PP/Sp-/-) mice and sham-splenectomized, aly/+ (LN/PP/Sp+/+) mice received BMT from either syngeneic (aly/aly) or allogeneic, major histocompatibility complex (MHC) disparate donors. Surprisingly, although LN/PP/Sp-/- allo-BMT recipients experience a survival advantage, they developed significant systemic and target organ GVHD that is comparable to LN/PP/Sp+/+ controls. Early after allo-BMT, the activation and proliferation of donor T cells was significantly greater in the BM cavity of LN/PP/Sp-/- mice compared to LN/PP/Sp+/+ controls. Donor T cells in LN/PP/Sp-/- mice demonstrated cytolytic activity in vitro, but Graft vs Leukemia (GVL) activity could be overcome by increasing the tumor burden. These data suggest that SLO contribute to, but are not required for, allogeneic T cell responses, and suggest that the BM may represent an alternative, albeit less efficient site for T cell activation following allo-BMT., (Copyright 2010 American Society for Blood and Marrow Transplantation. Published by Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
39. Viruses as co-factors for the initiation or exacerbation of lung fibrosis.
- Author
-
Vannella KM and Moore BB
- Abstract
Idiopathic pulmonary fibrosis (IPF) remains exactly that. The disease originates from an unknown cause, and little is known about the mechanisms of pathogenesis. While the disease is likely multi-factorial, evidence is accumulating to implicate viruses as co-factors (either as initiating or exacerbating agents) of fibrotic lung disease. This review summarizes the available clinical and experimental observations that form the basis for the hypothesis that viral infections may augment fibrotic responses. We review the data suggesting a link between hepatitis C virus, adenovirus, human cytomegalovirus and, in particular, the Epstein-Barr gammaherpesvirus, in IPF. In addition, we highlight the recent associations made between gammaherpesvirus infection and lung fibrosis in horses and discuss the various murine models that have been used to investigate the contribution of gammaherpesviruses to fibrotic progression. We review the work demonstrating that gammaherpesvirus infection of Th2-biased mice leads to multi-organ fibrosis and highlight studies showing that gammaherpesviral infections of mice either pre- or post-fibrotic challenge can augment the development of fibrosis. Finally, we discuss potential mechanisms whereby viral infections may amplify the development of fibrosis. While none of these studies prove causality, we believe the evidence suggests that viral infections should be considered as potential initiators or exacerbating agents in at least some cases of IPF and thereby justify further study.
- Published
- 2008
- Full Text
- View/download PDF
40. Cysteinyl leukotrienes are autocrine and paracrine regulators of fibrocyte function.
- Author
-
Vannella KM, McMillan TR, Charbeneau RP, Wilke CA, Thomas PE, Toews GB, Peters-Golden M, and Moore BB
- Subjects
- Animals, Arachidonate 5-Lipoxygenase deficiency, Cell Proliferation drug effects, Chemotaxis drug effects, Chemotaxis immunology, Cysteine pharmacology, Disease Models, Animal, Dose-Response Relationship, Drug, Fluorescein-5-isothiocyanate pharmacology, Humans, Leukotrienes pharmacology, Membrane Proteins drug effects, Membrane Proteins genetics, Mesenchymal Stem Cells drug effects, Mice, Mice, Inbred C57BL, Mice, Knockout, Pulmonary Fibrosis prevention & control, RNA, Messenger drug effects, RNA, Messenger genetics, Receptors, Leukotriene drug effects, Receptors, Leukotriene genetics, Reverse Transcriptase Polymerase Chain Reaction, Structure-Activity Relationship, Autocrine Communication immunology, Cysteine physiology, Leukotrienes physiology, Membrane Proteins metabolism, Mesenchymal Stem Cells immunology, Paracrine Communication immunology, Pulmonary Fibrosis immunology, Receptors, Leukotriene metabolism
- Abstract
Pulmonary fibrosis is characterized by the accumulation of fibroblasts and myofibroblasts. These cells may accumulate from three potential sources: the expansion of resident lung fibroblasts, the process of epithelial-mesenchymal transition, or the recruitment and differentiation of circulating mesenchymal precursors known as fibrocytes. We have previously demonstrated that fibrocytes participate in lung fibrogenesis following administration of FITC to mice. We now demonstrate that leukotriene-deficient 5-LO(-/-) mice are protected from FITC-induced fibrosis. Both murine and human fibrocytes express both cysteinyl leukotriene receptor (CysLT) 1 and CysLT2. In addition, fibrocytes are capable of producing CysLTs and can be regulated via the autocrine or paracrine secretion of these lipid mediators. Exogenous administration of leukotriene (LT) D(4), but not LTC(4) induces proliferation of both murine and human fibrocytes in a dose-dependent manner. Consistent with this result, CysLT1 receptor antagonists are able to block the mitogenic effects of exogenous LTD(4) on fibrocytes. Endogenous production of CysLTs contributes to basal fibrocyte proliferation, but does not alter fibrocyte responses to basic fibroblast growth factor. Although CysLTs can induce the migration of fibrocytes in vitro, they do not appear to be essential for fibrocyte recruitment to the lung in vivo, possibly due to compensatory chemokine-mediated recruitment signals. However, CysLTs do appear to regulate the proliferation of fibrocytes once they are recruited to the lung. These data provide mechanistic insight into the therapeutic benefit of leukotriene synthesis inhibitors and CysLT1 receptor antagonists in animal models of fibrosis.
- Published
- 2007
- Full Text
- View/download PDF
41. Functional divergence of a unique C-terminal domain of leucyl-tRNA synthetase to accommodate its splicing and aminoacylation roles.
- Author
-
Hsu JL, Rho SB, Vannella KM, and Martinis SA
- Subjects
- Alternative Splicing, Amino Acid Sequence, Crystallography, X-Ray, DNA-Directed RNA Polymerases chemistry, Escherichia coli genetics, Escherichia coli metabolism, Mitochondria metabolism, Molecular Sequence Data, Protein Structure, Tertiary, RNA Splicing, Sequence Homology, Amino Acid, Two-Hybrid System Techniques, Viral Proteins chemistry, Yeasts metabolism, Leucine-tRNA Ligase chemistry
- Abstract
Leucyl-tRNA synthetase (LeuRS) performs dual essential roles in group I intron RNA splicing as well as protein synthesis within the yeast mitochondria. Deletions of the C terminus differentially impact the two functions of the enzyme in splicing and aminoacylation in vivo. Herein, we determined that a fiveamino acid C-terminal deletion of LeuRS, which does not complement a null strain, can form a ternary complex with the bI4 intron and its maturase splicing partner. However, the complex fails to stimulate splicing activity. The x-ray co-crystal structure of LeuRS showed that a C-terminal extension of about 60 amino acids forms a discrete domain, which is unique among the LeuRSs and interacts with the corner of the L-shaped tRNALeu. Interestingly, deletion of the entire yeast mitochondrial LeuRS C-terminal domain enhanced its aminoacylation and amino acid editing activities. In striking contrast, deletion of the corresponding C-terminal domain of Escherichia coli LeuRS abolished aminoacylation of tRNALeu and also amino acid editing of mischarged tRNA molecules. These results suggest that the role of the leucine-specific C-terminal domain in tRNA recognition for aminoacylation and amino acid editing has adapted differentially and with surprisingly opposite effects. We propose that the secondary role of yeast mitochondrial LeuRS in RNA splicing has impacted the functional evolution of this critical C-terminal domain.
- Published
- 2006
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.