4,879 results on '"Lymphocytic Choriomeningitis Virus"'
Search Results
2. MitoTempo treatment as an approach to cure persistent viral infections?
- Author
-
Mischke, Jasmin, Klein, Sebastian, Cornberg, Markus, and Kraft, Anke R.M.
- Published
- 2024
- Full Text
- View/download PDF
3. Host specific sphingomyelin is critical for replication of diverse RNA viruses.
- Author
-
Han, Shuo, Ye, Xiaolei, Yang, Jintong, Peng, Xuefang, Jiang, Xiaming, Li, Jin, Zheng, Xiaojie, Zhang, Xinchen, Zhang, Yumin, Zhang, Lingyu, Wang, Wei, Li, Jiaxin, Xin, Wenwen, Zhang, Xiaoai, Xiao, Gengfu, Peng, Ke, Zhang, Leike, Du, Xuguang, Zhou, Lu, and Liu, Wei
- Subjects
- *
RNA replicase , *RNA virus infections , *HELIX-loop-helix motifs , *LYMPHOCYTIC choriomeningitis virus , *MOLECULAR structure , *GOLGI apparatus - Abstract
Lipids and lipid metabolism play an important role in RNA virus replication, which typically occurs on host cell endomembrane structures in the cytoplasm through mechanisms that are not yet fully identified. We conducted genome-scale CRISPR screening and identified sphingomyelin synthase 1 (SMS1; encoded by SGMS1) as a critical host factor for infection by severe fever with thrombocytopenia syndrome virus (SFTSV). SGMS1 knockout reduced sphingomyelin (SM) (d18:1/16:1) levels, inhibiting SFTSV replication. A helix-turn-helix motif in SFTSV RNA-dependent RNA polymerase (RdRp) directly binds to SM(d18:1/16:1) in Golgi apparatus, which was also observed in SARS-CoV-2 and lymphocytic choriomeningitis virus (LCMV), both showing inhibited replication in SGMS1 -KO cells. SM metabolic disturbance is associated with disease severity of viral infections. We designed a novel SMS1 inhibitor that protects mice against lethal SFTSV infection and reduce SARS-CoV-2 replication and pathogenesis. These findings highlight the critical role of SMS1 and SM(d18:1/16:1) in RNA virus replication, suggesting a broad-spectrum antiviral strategy. [Display omitted] • SGMS1 is identified as a critical host factor for SFTSV infection by a CRISPR screen • Deletion of SGMS1 inhibits SM biosynthesis and suppresses RNA virus replication • SM(d18:1/16:1) in Golgi apparatus directly binds to viral RNA-dependent RNA polymerase • A designed SMS1 inhibitor protects mice against lethal SFTSV infection High pathogenic RNA viruses pose a great threat to public health, yet there is still an urgent need for antiviral drugs available for the treatment of infections caused by these viruses. Uncovering the common mechanisms underlying RNA virus replication and virus-host interactions is crucial for identifying broad-spectrum antiviral targets and developing host-targeted antiviral strategies. Notably, many RNA viruses replicate within the cytoplasm of infected cells, frequently in specialized structures assembled on the surface of membrane-bound compartments. However, the host factors involved in the relationship between virus replication and cell membrane structure and their molecular mechanisms remain elusive. Here, we identify SGMS1 as an essential host factor for infections caused by various RNA viruses. We reveal that inhibitors interfering with any component of the SM biosynthesis pathway suppress viral infection in cells. We further demonstrate that a helix-turn-helix motif in viral RdRp directly binds to SM(d18:1/16:1) in the Golgi apparatus, thereby facilitating viral replication. Depletion of SMS1 strongly increases the survival rates of mice infected with SFTSV in a lethal model. These findings highlight the important role of SMS1 and SM(d18:1/16:1) in RNA virus replication and provide a promising target for the development of broad-spectrum antiviral strategies. Building on these insights, we further develop an SMS1 inhibitor that protects mice against lethal SFTSV infection and remarkedly reduces SARS-CoV-2 replication and pathogenesis in vivo. The designed inhibitor holds potential as a broad-spectrum antiviral approach. Han et al. identify SGMS1 as a critical host factor for infection of various RNA viruses. SM(d18:1/16:1) synthesized by SMS1 in Golgi apparatus directly binds to a helix-turn-helix motif in viral RdRp, facilitating the formation of viral replication complex. Inhibition of SMS1 activity offers therapeutical protection against viral infections. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
4. Evaluating Antigen- and Vector-Specific Immune Responses of a Recombinant Pichinde Virus-Based Vaccine Expressing the Lymphocytic Choriomeningitis Virus Nucleoprotein.
- Author
-
Cain, Michaela, Huang, Qinfeng, Sanchez, Shania, Ly, Hinh, and Liang, Yuying
- Subjects
LYMPHOCYTIC choriomeningitis virus ,IMMUNOLOGIC memory ,GENETIC vectors ,VIRAL vaccines ,IMMUNE response ,BACTERIAL vaccines - Abstract
Background: Live viral vector-based vaccines are known to elicit strong immune responses, but their use can be limited by anti-vector immunity. Here, we analyzed the immunological responses of a live-attenuated recombinant Pichinde virus (PICV) vector platform (rP18tri). Methods: To evaluate anti-PICV immunity in the development of vaccine antigen-specific immune responses, we generated a rP18tri-based vaccine expressing the lymphocytic choriomeningitis virus (LCMV) nucleoprotein (NP) and administered four doses of this rP18tri-NPLCMV vaccine to mice. Using MHC-I tetramers to detect PICV NP38-45 and LCMV NP396-404 epitope-specific CD8+ T cells, we monitored vector- and vaccine-antigen-specific immune responses after each vaccination dose. Results: LCMV NP396-404-specific effector and memory CD8+ T cells were detected after the first dose and peaked after the second dose, whereas PICV NP38-45-specific memory CD8+ T cells increased with each dose. PICV-binding IgG antibodies peaked after the second dose, while anti-PICV neutralizing antibodies (NAbs) remained low even after the fourth dose. Immunization with the rP18tri-NPLCMV vaccine significantly reduced LCMV viral titers in a chronic LCMV Clone 13 infection model, demonstrating the protective role of LCMV NP-specific T cells. Conclusion: These findings provide important insights into the antigen- and vector-specific immunity of the rP18tri-NPLCMV vaccine and support the development of NP-based vaccines against arenavirus pathogens. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
5. Rare perinatal infections
- Author
-
А. А. Fadeeva, H. A. Sarkisyan, O. V. Molochkova, N. Yu. Egorova, А. А. Komarova, D. M. Mushcherova, A. P. Khokhlova, O. I. Savateeva, and P. V. Shumilov
- Subjects
intrauterine infection ,congenital viral infection ,lymphocytic choriomeningitis virus ,zika virus ,congenital zika syndrome ,microcephaly ,intracranial calcification ,Pediatrics ,RJ1-570 - Abstract
Congenital viral infections are one of the pressing issues in modern neonatology. Due to the fact that viremia is a common stage in the infection process, the fetus is at high risk of intrauterine hematogenic infection, which can harm various organs and systems. The most frequently observed manifestations of congential viral infections are: intrauterine growth restriction (IUGR), micro- and macrocephaly, fetal hydrocephalus, hepatosplenomegaly, pneumonia, bone lesions, rash and haematological abnormalities. Even though these clinical manifestations are not specific, identifying the etiological factor is often a necessary step to determine the future management of the newborn. The aim of this paper is to collect and summarize literary data on lymphocytic choriomeningitis virus (LCMV) and Zika virus. Results: congenital infections caused by LCMV and Zika virus share similar clinical features. Laboratory and instrumental diagnostic methods facilitate carrying out the differential diagnosis and establishing prognosis and the best clinical approach to the patient.
- Published
- 2024
- Full Text
- View/download PDF
6. TET2 regulates early and late transitions in exhausted CD8+ T cell differentiation and limits CAR T cell function.
- Author
-
Dimitri, Alexander J., Baxter, Amy E., Chen, Gregory M., Hopkins, Caitlin R., Rouin, Geoffrey T., Hua Huang, Weimin Kong, Holliday, Christopher H., Wiebking, Volker, Bartoszek, Robert, Drury, Sydney, Dalton, Katherine, Koucky, Owen M., Zeyu Chen, Giles, Josephine R., Dils, Alexander T., In-Young Jung, O'Connor, Roddy, Collins, Sierra, and Everett, John K.
- Subjects
- *
T-cell exhaustion , *T cell differentiation , *T cells , *CELL physiology , *LYMPHOCYTIC choriomeningitis virus , *CHIMERIC antigen receptors - Abstract
CD8+ T cell exhaustion hampers control of cancer and chronic infections and limits chimeric antigen receptor (CAR) T cell efficacy. Targeting TET2 in CAR T cells provides therapeutic benefit; however, TET2's role in exhausted T cell (TEX) development is unclear. In chronic lymphocytic choriomeningitis virus (LCMV) infection, TET2 drove conversion from stem cell-like TEX progenitors toward terminally differentiated and effector (TEFF)-like TEX. TET2 also enforced a terminally differentiated state in the early bifurcation between TEFF and TEX, indicating broad roles for TET2 in acquisition of effector biology. To exploit the therapeutic potential of TET2, we developed clinically actionable TET2-targeted CAR T cells by disrupting TET2 via knock-in of a safety switch alongside CAR knock-in at the TRAC locus. TET2-targeted CAR T cells exhibited restrained terminal exhaustion in vitro and enhanced antitumor responses in vivo. Thus, TET2 regulates fate transitions in TEX differentiation and can be targeted with a safety mechanism in CAR T cells for improved tumor control. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
7. Type I interferon induced during chronic viral infection favors B‐cell development in the thymus.
- Author
-
Valbon, Stefanie F, Lebel, Marie‐Eve, Feldman, H Alex, Condotta, Stephanie A, Dong, Mengqi, Giordano, Daniela, Waggoner, Stephen N, Melichar, Heather J, and Richer, Martin J
- Subjects
- *
TYPE I interferons , *LYMPHOCYTIC choriomeningitis virus , *VIRUS diseases , *CELL populations , *THYMUS , *B cells - Abstract
Chronic viral infections cause thymic involution yet the potential for broader, longer‐term impact on thymic composition remains unexplored. Here we show that chronic, but not acute, lymphocytic choriomeningitis virus infection promotes a unique population of immature B cells in the thymus. We show that chronic viral infection promotes signals within the thymus, including the expression of B‐cell activating factor (BAFF), that favor the maturation of this population as these cells acquire expression of CD19 and immunoglobulin M. Mechanistically, type I interferon (IFN‐I), predominantly IFNβ, signals to thymic hematopoietic cells, strongly delaying T‐cell development at the earliest precursor stage. Furthermore, IFN‐I signaling to the nonhematopoietic compartment provides a second signal essential to favor B‐cell differentiation and maturation within the thymus. Importantly, chronic infection yields changes in the B‐cell population for at least 50 days following infection, long after thymic atrophy has subsided. Thus, the inflammatory milieu induced by chronic viral infection has a profound, and long‐lasting, effect on thymic composition leading to the generation of a novel population of thymic B cells. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
8. Architectural organization and in situ fusion protein structure of lymphocytic choriomeningitis virus.
- Author
-
Kang, Joon S., Kang Zhou, Hui Wang, Sijia Tang, Lyles, Kristin Van Mouwerik, Ming Luo, and Zhou, Z. Hong
- Subjects
- *
LYMPHOCYTIC choriomeningitis virus , *TRANSMEMBRANE domains , *RNA replicase , *HEMORRHAGIC fever , *VIRAL envelopes - Abstract
Arenaviruses exist globally and can cause hemorrhagic fever and neurological diseases, exemplified by the zoonotic pathogen lymphocytic choriomeningitis virus (LCMV). The structures of individual LCMV proteins or their fragments have been reported, but the architectural organization and the nucleocapsid assembly mechanism remain elusive. Importantly, the in situ structure of the arenavirus fusion protein complex (glycoprotein complex, GPC) as present on the virion prior to fusion, particularly with its integral stable signal peptide (SSP), has not been shown, hindering efforts such as structure-based vaccine design. Here, we have determined the in situ structure of LCMV proteins and their architectural organization in the virion by cryogenic electron tomography. The tomograms reveal the global distribution of GPC, matrix protein Z, and the contact points between the viral envelope and nucleocapsid. Subtomogram averaging yielded the in situ structure of the mature GPC with its transmembrane domain intact, revealing the GP2-SSP interface and the endodomain of GP2. The number of RNA-dependent RNA polymerase L molecules packaged within each virion varies, adding new perspectives to the infection mechanism. Together, these results delineate the structural organization of LCMV and offer new insights into its mechanism of LCMV maturation, egress, and cell entry. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. Generation of antigen-specific memory CD4 T cells by heterologous immunization enhances the magnitude of the germinal center response upon influenza infection.
- Author
-
Sircy, Linda M., Ramstead, Andrew G., Gibbs, Lisa C., Joshi, Hemant, Baessler, Andrew, Mena, Ignacio, García-Sastre, Adolfo, Emerson, Lyska L., Fairfax, Keke C., Williams, Matthew A., and Hale, J. Scott
- Subjects
- *
GERMINAL centers , *T helper cells , *IMMUNOLOGIC memory , *LYMPHOCYTIC choriomeningitis virus , *SEASONAL influenza , *B cells - Abstract
Current influenza vaccine strategies have yet to overcome significant obstacles, including rapid antigenic drift of seasonal influenza viruses, in generating efficacious long-term humoral immunity. Due to the necessity of germinal center formation in generating long-lived high affinity antibodies, the germinal center has increasingly become a target for the development of novel or improvement of less-efficacious vaccines. However, there remains a major gap in current influenza research to effectively target T follicular helper cells during vaccination to alter the germinal center reaction. In this study, we used a heterologous infection or immunization priming strategy to seed an antigen-specific memory CD4+ T cell pool prior to influenza infection in mice to evaluate the effect of recalled memory T follicular helper cells in increased help to influenza-specific primary B cells and enhanced generation of neutralizing antibodies. We found that heterologous priming with intranasal infection with acute lymphocytic choriomeningitis virus (LCMV) or intramuscular immunization with adjuvanted recombinant LCMV glycoprotein induced increased antigen-specific effector CD4+ T and B cellular responses following infection with a recombinant influenza strain that expresses LCMV glycoprotein. Heterologously primed mice had increased expansion of secondary Th1 and Tfh cell subsets, including increased CD4+ TRM cells in the lung. However, the early enhancement of the germinal center cellular response following influenza infection did not impact influenza-specific antibody generation or B cell repertoires compared to primary influenza infection. Overall, our study suggests that while heterologous infection or immunization priming of CD4+ T cells is able to enhance the early germinal center reaction, further studies to understand how to target the germinal center and CD4+ T cells specifically to increase long-lived antiviral humoral immunity are needed. Author summary: T follicular helper (Tfh) cells are specialized CD4+ T cells that provide help to B cells and are required to form germinal centers within secondary lymphoid organs during an immune response. Germinal centers are necessary for generating high affinity virus-specific antibodies necessary to clear influenza infections, though current vaccines fail to generate long-lived antibodies that universally recognize different influenza strains. We used a "heterologous priming" strategy in mice using a non-influenza viral infection or viral protein subunit vaccination to form memory CD4+ Tfh cells (in previously naïve mice) that can be rapidly recalled into secondary Tfh cells following influenza infection and ideally enhance the germinal center reaction and formation of high affinity antibodies to influenza better than primary Tfh cells. Our study showed that heterologous priming induced an increase in both CD4+ T and B cells early following influenza infection, suggesting we could successfully target enhancement of the germinal center. Despite the enhancement of the early germinal center cellular response, we did not see an increase in influenza-specific antiviral antibodies. Thus, while Tfh cells are critical for the generation of high affinity antibodies, other strategies to target expansion of Tfh cells during influenza vaccination will need to be developed. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
10. Cellular stress increases DRIP production and MHC Class I antigen presentation.
- Author
-
Pach, Natalie and Basler, Michael
- Subjects
LYMPHOCYTIC choriomeningitis virus ,VIRAL proteins ,ANTIGEN presentation ,ANTIGEN processing ,MANUFACTURING defects - Abstract
Background: Defective ribosomal products (DRiPs) are non-functional proteins rapidly degraded during or after translation being an essential source for MHC class I ligands. DRiPs are characterized to derive from a substantial subset of nascent gene products that degrade more rapidly than their corresponding native retiree pool. So far, mass spectrometry analysis revealed that a large number of HLA class I peptides derive from DRiPs. However, a specific viral DRiP on protein level was not described. In this study, we aimed to characterize and identify DRiPs derived from a viral protein. Methods: Using the nucleoprotein (NP) of the lymphocytic choriomeningitis virus (LCMV) which is conjugated N-terminally to ubiquitin, or the ubiquitin-like modifiers FAT10 or ISG15 the occurrence of DRiPs was studied. The formation and degradation of DRiPs was monitored by western blot with the help of a FLAG tag. Flow cytometry and cytotoxic T cells were used to study antigen presentation. Results: We identified several short lived DRiPs derived from LCMV-NP. Of note, these DRiPs could only be observed when the LCMV-NP was modified with ubiquitin or ubiquitin-like modifiers, but not in the wild type form. Using proteasome inhibitors, we could show that degradation of LCMV-NP derived DRiPs were proteasome dependent. Interestingly, the synthesis of DRiPs could be enhanced when cells were stressed with the help of FCS starvation. An enhanced NP118-126 presentation was observed when the LCMV-NP was modified with ubiquitin or ubiquitin-like modifiers, or under FCS starvation. Conclusion: Taken together, we visualize for the first time DRiPs derived from a viral protein. Furthermore, DRiPs formation, and therefore MHC-I presentation, is enhanced under cellular stress conditions. Our investigations on DRiPs in MHC class I antigen presentation open up new approaches for the development of vaccination strategies. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
11. Cellular N-Myristoyl Transferases Are Required for Mammarenavirus Multiplication.
- Author
-
Witwit, Haydar, Betancourt, Carlos Alberto, Cubitt, Beatrice, Khafaji, Roaa, Kowalski, Heinrich, Jackson, Nathaniel, Ye, Chengjin, Martinez-Sobrido, Luis, and de la Torre, Juan C.
- Subjects
- *
LYMPHOCYTIC choriomeningitis virus , *EXTRACELLULAR matrix proteins , *PEPTIDES , *MYRISTOYLATION , *PROTEOLYSIS - Abstract
The mammarenavirus matrix Z protein plays critical roles in virus assembly and cell egress. Meanwhile, heterotrimer complexes of a stable signal peptide (SSP) together with glycoprotein subunits GP1 and GP2, generated via co-and post-translational processing of the surface glycoprotein precursor GPC, form the spikes that decorate the virion surface and mediate virus cell entry via receptor-mediated endocytosis. The Z protein and the SSP undergo N-terminal myristoylation by host cell N-myristoyltransferases (NMT1 and NMT2), and G2A mutations that prevent myristoylation of Z or SSP have been shown to affect the Z-mediated virus budding and GP2-mediated fusion activity that is required to complete the virus cell entry process. In the present work, we present evidence that the validated on-target specific pan-NMT inhibitor DDD85646 exerts a potent antiviral activity against the prototypic mammarenavirus lymphocytic choriomeningitis virus (LCMV) that correlates with reduced Z budding activity and GP2-mediated fusion activity as well as with proteasome-mediated degradation of the Z protein. The potent anti-mammarenaviral activity of DDD85646 was also observed with the hemorrhagic-fever-causing Junin (JUNV) and Lassa (LASV) mammarenaviruses. Our results support the exploration of NMT inhibition as a broad-spectrum antiviral against human pathogenic mammarenaviruses. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
12. Manuel del Cerro (1931–2023).
- Author
-
Triarhou, Lazaros C.
- Subjects
- *
MEDICAL sciences , *CYTOLOGY , *NEURONS , *LYMPHOCYTIC choriomeningitis virus , *PURKINJE cells - Abstract
The article in the Journal of Neurology commemorates the life and work of Manuel del Cerro, an Argentinian-American neurobiologist born in 1931. Del Cerro made significant contributions to neuroscience, particularly in the study of the cerebellum and retinal cell transplantation. He was a dedicated educator and mentor, inspiring numerous students and professionals in the field. Del Cerro passed away in 2023, leaving behind a legacy of scientific achievement and a passion for microscopy. [Extracted from the article]
- Published
- 2024
- Full Text
- View/download PDF
13. Cell-Intrinsic CD38 Expression Sustains Exhausted CD8+ T Cells by Regulating Their Survival and Metabolism during Chronic Viral Infection
- Author
-
DeRogatis, Julia M, Neubert, Emily N, Viramontes, Karla M, Henriquez, Monique L, Nicholas, Dequina A, and Tinoco, Roberto
- Subjects
Medical Microbiology ,Biomedical and Clinical Sciences ,Immunology ,Biodefense ,Emerging Infectious Diseases ,Infectious Diseases ,2.1 Biological and endogenous factors ,Infection ,CD8-Positive T-Lymphocytes ,Cell Differentiation ,Lymphocytic Choriomeningitis ,Lymphocytic choriomeningitis virus ,Persistent Infection ,Animals ,Mice ,Cell Survival ,Up-Regulation ,Cell Proliferation ,CD38 ,LCMV ,T cell exhaustion ,effector T cell ,metabolism ,CD8(+) T cell ,chronic viral infection ,T cells ,CD8+ T cell ,Biological Sciences ,Agricultural and Veterinary Sciences ,Medical and Health Sciences ,Virology ,Agricultural ,veterinary and food sciences ,Biological sciences ,Biomedical and clinical sciences - Abstract
Acute and chronic viral infections result in the differentiation of effector and exhausted T cells with functional and phenotypic differences that dictate whether the infection is cleared or progresses to chronicity. High CD38 expression has been observed on CD8+ T cells across various viral infections and tumors in patients, suggesting an important regulatory function for CD38 on responding T cells. Here, we show that CD38 expression was increased and sustained on exhausted CD8+ T cells following chronic lymphocytic choriomeningitis virus (LCMV) infection, with lower levels observed on T cells from acute LCMV infection. We uncovered a cell-intrinsic role for CD38 expression in regulating the survival of effector and exhausted CD8+ T cells. We observed increased proliferation and function of Cd38-/- CD8+ progenitor exhausted T cells compared to those of wild-type (WT) cells. Furthermore, decreased oxidative phosphorylation and glycolytic potential were observed in Cd38-/- CD8+ T cells during chronic but not acute LCMV infection. Our studies reveal that CD38 has a dual cell-intrinsic function in CD8+ T cells, where it decreases proliferation and function yet supports their survival and metabolism. These findings show that CD38 is not only a marker of T cell activation but also has regulatory functions on effector and exhausted CD8+ T cells. IMPORTANCE Our study shows how CD38 expression is regulated on CD8+ T cells responding during acute and chronic viral infection. We observed higher CD38 levels on CD8+ T cells during chronic viral infection compared to levels during acute viral infection. Deleting CD38 had an important cell-intrinsic function in ensuring the survival of virus-specific CD8+ T cells throughout the course of viral infection. We found defective metabolism in Cd38-/- CD8+ T cells arising during chronic infection and changes in their progenitor T cell phenotype. Our studies revealed a dual cell-intrinsic role for CD38 in limiting proliferation and granzyme B production in virus-specific exhausted T cells while also promoting their survival. These data highlight new avenues for research into the mechanisms through which CD38 regulates the survival and metabolism of CD8+ T cell responses to viral infections.
- Published
- 2023
14. Mouse guanylate-binding proteins of the chromosome 3 cluster do not mediate antiviral activity in vitro or in mouse models of infection.
- Author
-
Tessema, Melkamu B., Feng, Shouya, Enosi Tuipulotu, Daniel, Farrukee, Rubaiyea, Ngo, Chinh, Gago da Graça, Catarina, Yamomoto, Masahiro, Utzschneider, Daniel T., Brooks, Andrew G., Londrigan, Sarah L., Man, Si Ming, and Reading, Patrick C.
- Subjects
- *
HUMAN herpesvirus 1 , *LYMPHOCYTIC choriomeningitis virus , *CHROMOSOMES , *LABORATORY mice - Abstract
Dynamin-like GTPase proteins, including myxoma (Mx) and guanylate-binding proteins (GBPs), are among the many interferon stimulated genes induced following viral infections. While studies report that human (h)GBPs inhibit different viruses in vitro, few have convincingly demonstrated that mouse (m)GBPs mediate antiviral activity, although mGBP-deficient mice have been used extensively to define their importance in immunity to diverse intracellular bacteria and protozoa. Herein, we demonstrate that individual (overexpression) or collective (knockout (KO) mice) mGBPs of the chromosome 3 cluster (mGBPchr3) do not inhibit replication of five viruses from different virus families in vitro, nor do we observe differences in virus titres recovered from wild type versus mGBPchr3 KO mice after infection with three of these viruses (influenza A virus, herpes simplex virus type 1 or lymphocytic choriomeningitis virus). These data indicate that mGBPchr3 do not appear to be a major component of cell-intrinsic antiviral immunity against the diverse viruses tested in our studies. Mouse guanylate binding proteins of the chromosome 3 cluster (mGBPchr3) do not mediate potent antiviral activity against different human or mouse viruses in vitro and in vivo. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
15. An attenuated lymphocytic choriomeningitis virus vector enhances tumor control in mice partly via IFN-I.
- Author
-
Chung, Young Rock, Awakoaiye, Bakare, Dangi, Tanushree, Irani, Nahid, Fourati, Slim, and Penaloza-MacMaster, Pablo
- Subjects
- *
LYMPHOCYTIC choriomeningitis virus , *GENETIC vectors , *MICE , *IMMUNOCOMPROMISED patients - Abstract
Viral vectors are being used for the treatment of cancer. Yet, their efficacy varies among tumors and their use poses challenges in immunosuppressed patients, underscoring the need for alternatives. We report striking antitumoral effects by a nonlytic viral vector based on attenuated lymphocytic choriomeningitis virus (r3LCMV). We show in multiple tumor models that injection of tumor-bearing mice with this vector results in improved tumor control and survival. Importantly, r3LCMV improved tumor control in immunodeficient Rag mice and MyD88 mice, suggesting that multiple pathways contributed to the antitumoral effects. The antitumoral effects of r3LCMV were also observed when this vector was administered several weeks before tumor challenges, suggesting the induction of trained immunity. Single-cell RNA sequencing analyses, antibody blockade experiments, and knockout models revealed a critical role for host-intrinsic IFN-I in the antitumoral efficacy of r3LCMV vectors. Collectively, these data demonstrate potent antitumoral effects by r3LCMV vectors and unveil multiple mechanisms underlying their antitumoral efficacy. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
16. T(r)icky Environments: Higher Prevalence of Tick-Borne Zoonotic Pathogens in Rodents from Natural Areas Compared with Urban Areas.
- Author
-
de Cock, Marieke P., Baede, Valérie O., Esser, Helen J., Fonville, Manoj, de Vries, Ankje, de Boer, Willem F., Mehl, Calvin, Ulrich, Rainer G., Schares, Gereon, Hakze-van der Honing, Renate W., van der Poel, Wim H. M., Sprong, Hein, and Maas, Miriam
- Subjects
- *
URBAN land use , *LYMPHOCYTIC choriomeningitis virus , *MICE , *METHICILLIN-resistant staphylococcus aureus , *ARTHROPOD vectors - Abstract
Background: Urban areas are unique ecosystems with stark differences in species abundance and composition compared with natural ecosystems. These differences can affect pathogen transmission dynamics, thereby altering zoonotic pathogen prevalence and diversity. In this study, we screened small mammals from natural and urban areas in the Netherlands for up to 19 zoonotic pathogens, including viruses, bacteria, and protozoan parasites. Materials and Methods: In total, 578 small mammals were captured, including wood mice (Apodemus sylvaticus), bank voles (Myodes glareolus), yellow-necked mice (Apodemus flavicollis), house mice (Mus musculus), common voles (Microtus arvalis), and greater white-toothed shrews (Crocidura russula). We detected a wide variety of zoonotic pathogens in small mammals from both urban and natural areas. For a subset of these pathogens, in wood mice and bank voles, we then tested whether pathogen prevalence and diversity were associated with habitat type (i.e., natural versus urban), degree of greenness, and various host characteristics. Results: The prevalence of tick-borne zoonotic pathogens (Borrelia spp. and Neoehrlichia mikurensis) was significantly higher in wood mice from natural areas. In contrast, the prevalence of Bartonella spp. was higher in wood mice from urban areas, but this difference was not statistically significant. Pathogen diversity was higher in bank voles from natural habitats and increased with body weight for both rodent species, although this relationship depended on sex for bank voles. In addition, we detected methicillin-resistant Staphylococcus aureus, extended-spectrum beta-lactamase/AmpC-producing Escherichia coli, and lymphocytic choriomeningitis virus for the first time in rodents in the Netherlands. Discussion: The differences between natural and urban areas are likely related to differences in the abundance and diversity of arthropod vectors and vertebrate community composition. With increasing environmental encroachment and changes in urban land use (e.g., urban greening), it is important to better understand transmission dynamics of zoonotic pathogens in urban environments to reduce potential disease risks for public health. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
17. Identification of residues in Lassa virus glycoprotein 1 involved in receptor switch.
- Author
-
Jiao Guo, Yi Wan, Yang Liu, Xiaoying Jia, Siqi Dong, Gengfu Xiao, and Wei Wang
- Subjects
LYMPHOCYTIC choriomeningitis virus ,INTERFEROMETRY ,LASSA fever ,MEMBRANE fusion ,MEMBRANE proteins - Abstract
Lassa virus (LASV) is an enveloped, negative-sense RNA virus that causes Lassa hemorrhagic fever. Successful entry of LASV requires the viral glycoprotein 1 (GP1) to undergo a receptor switch from its primary receptor alpha-dystroglycan (α-DG) to its endosomal receptor lysosome-associated membrane protein 1 (LAMP1). A conserved histidine triad in LASV GP1 has been reported to be responsible for receptor switch. To test the hypothesis that other non-conserved residues also contribute to receptor switch, we constructed a series of mutant LASV GP1 proteins and tested them for binding to LAMP1. Four residues, L84, K88, L107, and H170, were identified as critical for receptor switch. Substituting any of the four residues with the corresponding lymphocytic choriomeningitis virus (LCMV) residue (L84 N, K88E, L10F, and H170S) reduced the binding affinity of LASV GP1 for LAMP1. Moreover, all mutations caused decreases in glycoprotein precursor (GPC)-mediated membrane fusion at both pH 4.5 and 5.2. The infectivity of pseudotyped viruses bearing either GPC
L84N or GPCK88E decreased sharply in multiple cell types, while L107F and H170S had only mild effects on infectivity. Using biolayer light interferometry assay, we found that all four mutants had decreased binding affinity to LAMP1, in the order of binding affinity being L84 N > L107F > K88E > H170S. The four amino acid loci identified for the first time in this study have important reference significance for the in-depth investigation of the mechanism of receptor switching and immune escape of LASV occurrence and the development of reserve anti-LASV infection drugs. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
18. Lymphocytic Choriomeningitis Virus Infections in Hungary between 2017–2023—Investigation of the First Congenital Infections.
- Author
-
Koroknai, Anita, Nagy, Anna, Nagy, Orsolya, Csonka, Nikolett, Mezei, Eszter, Szomor, Katalin, and Takács, Mária
- Subjects
- *
LYMPHOCYTIC choriomeningitis virus , *CONGENITAL disorders , *LYMPHOCYTIC choriomeningitis , *NEUROLOGICAL disorders , *VIRUS diseases - Abstract
Lymphocytic choriomeningitis virus (LCMV) is a neglected rodent-borne arenavirus, primarily spread by common house mouse species. Acquired human infections range from asymptomatic to mild flu-like symptoms and self-resolving neurological diseases. In contrast, intrauterine LCMV infection is associated with high mortality and morbidity. Infection of the fetus often leads to fetal death, and surviving fetuses may develop vision impairment and central nervous system developmental disorders. LCMV is mainly diagnosed by serological methods using in-house indirect immunofluorescence assays. LCMV nucleic acid is detected by the nested RT-PCR method and confirmed by Sanger sequencing. In Hungary, 23 acquired lymphocytic choriomeningitis cases were diagnosed between 2017 and 2023. Ten out of 23 confirmed patients proved to be positive by the PCR method. Two cases of intrauterine LCMV infections were detected in 2019 and 2021, respectively. The IgG antibody titers measured in the infant's serum samples were much higher than the IgG titers of the maternal serum samples. Both IgM and IgA antibodies were detectable in the infants' sera. As the microbiological diagnosis of LCMV is rather challenging and the symptoms are very similar to the clinical picture of other common teratogenic pathogens such as cytomegalovirus or Toxoplasma gondii, intrauterine LCMV infections might still be underdiagnosed. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
19. The Significant Role of PA28αβ in CD8 + T Cell-Mediated Graft Rejection Contrasts with Its Negligible Impact on the Generation of MHC-I Ligands.
- Author
-
Inholz, Katharina, Bader, Ulrika, Mundt, Sarah, and Basler, Michael
- Subjects
- *
GRAFT rejection , *T cells , *T cell receptors , *CYTOTOXIC T cells , *LYMPHOCYTIC choriomeningitis virus , *CD8 antigen , *MAJOR histocompatibility complex - Abstract
The proteasome generates the majority of peptides presented on MHC class I molecules. The cleavage pattern of the proteasome has been shown to be changed via the proteasome activator (PA)28 alpha beta (PA28αβ). In particular, several immunogenic peptides have been reported to be PA28αβ-dependent. In contrast, we did not observe a major impact of PA28αβ on the generation of different major histocompatibility complex (MHC) classI ligands. PA28αβ-knockout mice infected with the lymphocytic choriomeningitis virus (LCMV) or vaccinia virus showed a normal cluster of differentiation (CD) 8 response and viral clearance. However, we observed that the adoptive transfer of wild-type cells into PA28αβ-knockout mice led to graft rejection, but not vice versa. Depletion experiments showed that the observed rejection was mediated by CD8+ cytotoxic T cells. These data indicate that PA28αβ might be involved in the development of the CD8+ T cell repertoire in the thymus. Taken together, our data suggest that PA28αβ is a crucial factor determining T cell selection and, therefore, impacts graft acceptance. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
20. An mRNA-LNP-based Lassa virus vaccine induces protective immunity in mice.
- Author
-
Mei Hashizume, Ayako Takashima, and Masaharu Iwasaki
- Subjects
- *
VIRAL vaccines , *LYMPHOCYTIC choriomeningitis virus , *HEMORRHAGIC fever , *LASSA fever , *HEMORRHAGIC diseases - Abstract
The mammarenavirus Lassa virus (LASV) causes the life-threatening hemorrhagic fever disease, Lassa fever. The lack of licensed medical countermeasures against LASV underscores the urgent need for the development of novel LASV vaccines, which has been hampered by the requirement for a biosafety level 4 facility to handle live LASV. Here, we investigated the efficacy of mRNA-lipid nanoparticle (mRNA-LNP)- based vaccines expressing the LASV glycoprotein precursor (LASgpc) or nucleoprotein (LCMnp) of the prototypic mammarenavirus, lymphocytic choriomeningitis virus (LCMV), in mice. Two doses of LASgpc- or LCMnp-mRNA-LNP administered intravenously (i.v.) protected C57BL/6 mice from a lethal challenge with a recombinant (r) LCMV expressing a modified LASgpc (rLCMV/LASgpc2m) inoculated intracranially. Intramuscular (i.m.) immunization with two doses of LASgpc- or LCMnp-mRNA-LNP significantly reduced the viral load in C57BL/6 mice inoculated i.v. with rLCMV/LASgpc2m. High levels of viremia and lethality were observed in CBA mice inoculated i.v. with rLCMV/LASgpc2m, which were abrogated by i.m. immunization with two doses of LASgpc-mRNA-LNP. The protective efficacy of two i.m. doses of LCMnp-mRNA-LNP was confirmed in a lethal hemorrhagic disease model of FVB mice i.v. inoculated with wild-type rLCMV. In all conditions tested, negligible and high levels of LASgpc- and LCMnp-specific antibodies were detected in mRNA-LNP-immunized mice, respectively, but robust LASgpc- and LCMnp-specific CD8+ T cell responses were induced. Accordingly, plasma from LASgpcmRNA-LNP-immunized mice did not exhibit neutralizing activity. Our findings and surrogate mouse models of LASV infection, which can be studied at a reduced biocontainment level, provide a critical foundation for the rapid development of mRNA-LNPbased LASV vaccines. IMPORTANCE Lassa virus (LASV) is a highly pathogenic mammarenavirus responsible for several hundred thousand infections annually in West African countries, causing a high number of lethal Lassa fever (LF) cases. Despite its significant impact on human health, clinically approved, safe, and effective medical countermeasures against LF are not available. The requirement of a biosafety level 4 facility to handle live LASV has been one of the main obstacles to the research and development of LASV countermeasures. Here, we report that two doses of mRNA-lipid nanoparticle-based vaccines expressing the LASV glycoprotein precursor (LASgpc) or nucleoprotein (LCMnp) of lymphocytic choriomeningitis virus (LCMV), a mammarenavirus genetically closely related to LASV, conferred protection to recombinant LCMV-based surrogate mouse models of lethal LASV infection. Notably, robust LASgpc- and LCMnp-specific CD8+ T cell responses were detected in mRNA-LNP-immunized mice, whereas no virus-neutralizing activity was observed. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
21. Characterization of non-standard viral genomes during arenavirus infections identifies prominent S RNA intergenic region deletions
- Author
-
Matthew Hackbart and Carolina B. López
- Subjects
arenavirus ,non-standard viral genomes ,deletions ,lymphocytic choriomeningitis virus ,viral interference ,Microbiology ,QR1-502 - Abstract
ABSTRACT Arenaviruses, a family of negative-sense RNA viruses spread by rodents, are a leading cause of severe hemorrhagic fever in humans. Due to a paucity of antivirals and vaccines for arenaviruses, there is a need to identify new mechanisms for interfering with arenavirus replication. In several negative-sense RNA viruses, natural viral interference results from the production of non-standard viral genomes (nsVGs) that activate the innate immune system and/or compete for essential viral products. Although it is well established that arenaviruses produce strong interfering activities, it is unknown if they produce interfering nsVGs. Here, we show that arenaviruses produce deletions within the intergenic region of their small (S) RNA genome, and these deletions inhibit viral glycoprotein production during minigenome replication. S RNA deletions are more abundant when arenaviruses are grown in high-interfering conditions and are associated with reduced viral replication. Overall, we found that arenaviruses produce internal deletions within the S RNA intergenic region that are capable of decreasing glycoprotein production. These natural arenavirus interfering molecules provide a new target for the generation of therapeutics against arenaviruses.IMPORTANCEArenaviruses are hemorrhagic fever-causing pathogens that infect millions of people a year. There are currently no approved antivirals that target arenaviruses, and understanding natural mechanisms that inhibit arenavirus replication is crucial for the development of effective therapeutics. Here, we identified multiple deletions within arenavirus genomes that remove major replicative elements of the viral genomes. We show that deletions that remove the intergenic region of the viral genome can prevent viral protein production. These deletions were found in all arenaviruses tested in this study representing a mechanism that could be harnessed for the development of antivirals that broadly target the arenavirus family.
- Published
- 2024
- Full Text
- View/download PDF
22. Shared and distinct biological circuits in effector, memory and exhausted CD8+ T cells revealed by temporal single-cell transcriptomics and epigenetics.
- Author
-
Wang, Ping, Staupe, Ryan, Abdel-Hakeem, Mohamed, Huang, Hua, Mathew, Divij, Painter, Mark, Wu, Jennifer, Giles, Josephine, Ngiow, Shin, Manne, Sasikanth, Baxter, Amy, Huang, Yinghui, Goel, Rishi, Yan, Patrick, Karakousis, Giorgos, Xu, Xiaowei, Mitchell, Tara, Huang, Alexander, Wherry, E, and Khan, Omar
- Subjects
Humans ,CD8-Positive T-Lymphocytes ,Transcriptome ,Lymphocytic choriomeningitis virus ,Epigenesis ,Genetic ,Chromatin ,Lymphocytic Choriomeningitis - Abstract
Naïve CD8+ T cells can differentiate into effector (Teff), memory (Tmem) or exhausted (Tex) T cells. These developmental pathways are associated with distinct transcriptional and epigenetic changes that endow cells with different functional capacities and therefore therapeutic potential. The molecular circuitry underlying these developmental trajectories and the extent of heterogeneity within Teff, Tmem and Tex populations remain poorly understood. Here, we used the lymphocytic choriomeningitis virus model of acute-resolving and chronic infection to address these gaps by applying longitudinal single-cell RNA-sequencing (scRNA-seq) and single-cell assay for transposase-accessible chromatin sequencing (scATAC-seq) analyses. These analyses uncovered new subsets, including a subpopulation of Tex cells expressing natural killer cell-associated genes that is dependent on the transcription factor Zeb2, as well as multiple distinct TCF-1+ stem/progenitor-like subsets in acute and chronic infection. These data also revealed insights into the reshaping of Tex subsets following programmed death 1 (PD-1) pathway blockade and identified a key role for the cell stress regulator, Btg1, in establishing the Tex population. Finally, these results highlighted how the same biological circuits such as cytotoxicity or stem/progenitor pathways can be used by CD8+ T cell subsets with highly divergent underlying chromatin landscapes generated during different infections.
- Published
- 2022
23. Retinal microglia express more MHC class I and promote greater T-cell-driven inflammation than brain microglia.
- Author
-
Bloomfield, Christina L., Gong, Joyce, Droho, Steven, Makinde, Hadijat M., Gurra, Miranda G., Stumpf, Cecilia H., Kharel, Arjun, Gadhvi, Gaurav, Winter, Deborah R., Weiguo Cui, Cuda, Carla M., and Lavine, Jeremy A.
- Subjects
MICROGLIA ,ENCEPHALITIS ,LYMPHOCYTIC choriomeningitis virus ,IMMUNOREGULATION ,GENE expression - Abstract
Introduction: Macrophage function is determined by microenvironment and origin. Brain and retinal microglia are both derived from yolk sac progenitors, yet their microenvironments differ. Utilizing single-cell RNA sequencing (scRNA-seq) data from mice, we tested the hypothesis that retinal and brain microglia exhibit distinct transcriptional profiles due to their unique microenvironments. Methods: Eyes and brains from 2-4 month wildtype mice were combined (20 eyes; 3 brains) to yield one biologically diverse sample per organ. Each tissue was digested into single cell suspensions, enriched for immune cells, and sorted for scRNA-seq. Analysis was performed in Seurat v3 including clustering, integration, and differential expression. Multi-parameter flow cytometry was used for validation of scRNA-seq results. Lymphocytic choriomeningitis virus (LCMV) Clone 13, which produces a systemic, chronic, and neurotropic infection, was used to validate scRNA-seq and flow cytometry results in vivo. Results: Cluster analysis of integrated gene expression data from eye and brain identified 6 Tmem119
+ P2ry12+ microglial clusters. Differential expression analysis revealed that eye microglia were enriched for more pro-inflammatory processes including antigen processing via MHC class I (14.0-fold, H2-D1 and H2-K1) and positive regulation of T-cell immunity (8.4-fold) compared to brain microglia. Multi-parameter flow cytometry confirmed that retinal microglia expressed 3.2-fold greater H2-Db and 263.3-fold more H2-Kb than brain microglia. On Day 13 and 29 after LCMV infection, CD8+ T-cell density was greater in the retina than the brain. Discussion: Our data demonstrate that the microenvironment of retina and brain differs, resulting in microglia-specific gene expression changes. Specifically, retinal microglia express greater MHC class I by scRNA-seq and multi-parameter flow cytometry, resulting in a possibly enhanced capability to stimulate CD8+ T-cell inflammation during LCMV infection. These results may explain tissue-specific differences between retina and brain during systemic viral infections and CD8+ T-cell driven autoimmune disease. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
24. Development of rhesus macaque astrocyte cell lines supporting infection with a panel of viruses.
- Author
-
Reiter, Stefanie, Sun, Ting, Gärtner, Sabine, Pöhlmann, Stefan, and Winkler, Michael
- Subjects
- *
RHESUS monkeys , *GLIAL fibrillary acidic protein , *LYMPHOCYTIC choriomeningitis virus , *VIRUS diseases , *VESICULAR stomatitis - Abstract
Non-human primate (NHP)-based model systems are highly relevant for biomedical research. However, only few NHP cell lines are available and the generation of additional cell lines is an urgent need to help in the refinement and replacement of these models. Using lentiviral transduction of c-Fos, we established cell lines from the brain of rhesus macaques (Macaca mulatta). Transcriptome analysis revealed that these cell lines are closely related to astrocytes, which was confirmed by immunoblot and immunofluorescence microscopy detecting expression of the astrocyte marker glial fibrillary acidic protein (GFAP). Quantitative real-time PCR (qRT-PCR) demonstrated that major pathways of the interferon (IFN) system are intact. Using retroviral pseudotypes we found that the cell lines are susceptible to entry driven by the glycoproteins of vesicular stomatitis virus (VSV), lymphocytic choriomeningitis virus (LCMV) and to a lesser extent influenza A virus (IAV). Finally, these cells supported growth of Zika virus (ZIKV) and Papiine alphaherpesvirus 2 (PaHV2). In summary, we developed IFN-responsive cell lines from the rhesus macaque brain that allowed entry driven by several viral glycoproteins and were permissive to infection with ZIKV and a primate simplexvirus. These cell lines will be useful for efforts to analyze neurotropic viral infections in rhesus macaque models. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
25. Anti-PD-L1 Immunotherapy of Chronic Virus Infection Improves Virus Control without Augmenting Tissue Damage by Fibrosis.
- Author
-
Casella, Valentina, Cebollada Rica, Paula, Argilaguet, Jordi, Vidal, Enric, González-Cao, María, Güerri-Fernandez, Roberto, Bocharov, Gennady, and Meyerhans, Andreas
- Subjects
- *
VIRUS diseases , *T-cell exhaustion , *LYMPHOCYTIC choriomeningitis virus , *IMMUNOTHERAPY , *FIBROSIS - Abstract
Immunotherapy with checkpoint inhibitors, albeit commonly used against tumors, is still at its infancy against chronic virus infections. It relies on the reinvigoration of exhausted T lymphocytes to eliminate virus-infected cells. Since T cell exhaustion is a physiological process to reduce immunopathology, the reinvigoration of these cells might be associated with an augmentation of pathological changes. To test this possibility, we here analyzed in the model system of chronic lymphocytic choriomeningitis virus (LCMV)-infected mice whether treatment with the checkpoint inhibitor anti-PD-L1 antibody would increase CD8 T cell-dependent fibrosis. We show that pre-existing spleen fibrosis did not worsen under conditions that increase CD8 T cell functionality and reduce virus loads suggesting that the CD8 T cell functionality increase remained below its pathogenicity threshold. These promising findings should further encourage immunotherapeutic trials against chronic virus infections. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
26. Neuroretinal degeneration in a mouse model of systemic chronic immune activation observed by proteomics.
- Author
-
Khan, Asif Manzoor, Steffensen, Maria Abildgaard, Paskeviciute, Egle, Abduljabar, Ahmed Basim, Sørensen, Torben Lykke, Vorum, Henrik, Nissen, Mogens Holst, and Honoré, Bent
- Subjects
MACULAR degeneration ,LIQUID chromatography-mass spectrometry ,LYMPHOCYTIC choriomeningitis virus ,RETINAL injuries ,PROTEOMICS ,LABORATORY mice ,NEUROECTODERMAL tumors - Abstract
Blindness or vision loss due to neuroretinal and photoreceptor degeneration affects millions of individuals worldwide. In numerous neurodegenerative diseases, including age-related macular degeneration, dysregulated immune responsemediated retinal degeneration has been found to play a critical role in the disease pathogenesis. To better understand the pathogenic mechanisms underlying the retinal degeneration, we used amouse model of systemic immune activation where we infected mice with lymphocytic choriomeningitis virus (LCMV) clone 13. Here, we evaluated the effects of LCMV infection and present a comprehensive discoverybased proteomic investigation using tandem mass tag (TMT) labeling and highresolution liquid chromatography-tandem mass spectrometry (LC-MS/MS). Changes in protein regulation in the posterior part of the eye, neuroretina, and RPE/choroid were compared to those in the spleen as a secondary lymphoid organ and to the kidney as a non-lymphoid but encapsulated organ at 1, 8, and 28 weeks of infection. Using bioinformatic tools, we found several proteins responsible for maintaining normal tissue homeostasis to be differentially regulated in the neuroretina and the RPE/choroid during the degenerative process. Additionally, in the organs we observed, several important protein pathways contributing to cellular homeostasis and tissue development were perturbed and associated with LCMVmediated inflammation, promoting disease progression. Our findings suggest that the response to a systemic chronic infection differs between the neuroretina and the RPE/choroid, and the processes induced by chronic systemic infection in the RPE/choroid are not unlike those induced in non-immune-privileged organs such as the kidney and spleen. Overall, our data provide detailed insight into several molecular mechanisms of neuroretinal degeneration and highlight various novel protein pathways that further suggest that the posterior part of the eye is not an isolated immunological entity despite the existence of neuroretinal immune privilege. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
27. Alternating Arenavirus Vector Immunization Generates Robust Polyfunctional Genotype Cross-Reactive Hepatitis B Virus–Specific CD8 T-Cell Responses and High Anti–Hepatitis B Surface Antigen Titers.
- Author
-
Schmidt, Sarah, Mengistu, Meron, Daffis, Stephane, Ahmadi-Erber, Sarah, Deutschmann, Daniela, Grigoriev, Tetiana, Chu, Ruth, Leung, Cleo, Tomkinson, Adrian, Uddin, Mohammad Nizam, Moshkani, Safiehkhatoon, Robek, Michael D, Perry, Jason, Lauterbach, Henning, Orlinger, Klaus, Fletcher, Simon P, and Balsitis, Scott
- Subjects
- *
CELL surface antigens , *HEPATITIS B , *CD8 antigen , *LYMPHOCYTIC choriomeningitis virus , *TITERS - Abstract
Hepatitis B Virus (HBV) is a major driver of infectious disease mortality. Curative therapies are needed and ideally should induce CD8 T cell-mediated clearance of infected hepatocytes plus anti-hepatitis B surface antigen (HBsAg) antibodies (anti-HBs) to neutralize residual virus. We developed a novel therapeutic vaccine using non-replicating arenavirus vectors. Antigens were screened for genotype conservation and magnitude and genotype reactivity of T cell response, then cloned into Pichinde virus (PICV) vectors (recombinant PICV, GS-2829) and lymphocytic choriomeningitis virus (LCMV) vectors (replication-incompetent, GS-6779). Alternating immunizations with GS-2829 and GS-6779 induced high-magnitude HBV T cell responses, and high anti-HBs titers. Dose schedule optimization in macaques achieved strong polyfunctional CD8 T cell responses against core, HBsAg, and polymerase and high titer anti-HBs. In AAV-HBV mice, GS-2829 and GS-6779 were efficacious in animals with low pre-treatment serum HBsAg. Based on these results, GS-2829 and GS-6779 could become a central component of cure regimens. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
28. A review of emerging health threats from zoonotic New World mammarenaviruses.
- Author
-
Lendino, Arianna, Castellanos, Adrian A., Pigott, David M., and Han, Barbara A.
- Subjects
- *
HEMORRHAGIC fever , *LYMPHOCYTIC choriomeningitis virus , *ZOONOSES , *ENDEMIC diseases , *MORTALITY , *LAND use - Abstract
Despite repeated spillover transmission and their potential to cause significant morbidity and mortality in human hosts, the New World mammarenaviruses remain largely understudied. These viruses are endemic to South America, with animal reservoir hosts covering large geographic areas and whose transmission ecology and spillover potential are driven in part by land use change and agriculture that put humans in regular contact with zoonotic hosts. We compiled published studies about Guanarito virus, Junin virus, Machupo virus, Chapare virus, Sabia virus, and Lymphocytic Choriomeningitis virus to review the state of knowledge about the viral hemorrhagic fevers caused by New World mammarenaviruses. We summarize what is known about rodent reservoirs, the conditions of spillover transmission for each of these pathogens, and the characteristics of human populations at greatest risk for hemorrhagic fever diseases. We also review the implications of repeated outbreaks and biosecurity concerns where these diseases are endemic, and steps that countries can take to strengthen surveillance and increase capacity of local healthcare systems. While there are unique risks posed by each of these six viruses, their ecological and epidemiological similarities suggest common steps to mitigate spillover transmission and better contain future outbreaks. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
29. Pathogenic and Apathogenic Strains of Lymphocytic Choriomeningitis Virus Have Distinct Entry and Innate Immune Activation Pathways.
- Author
-
Johnson, Dylan M., Khakhum, Nittaya, Wang, Min, Warner, Nikole L., Jokinen, Jenny D., Comer, Jason E., and Lukashevich, Igor S.
- Subjects
- *
LYMPHOCYTIC choriomeningitis virus , *INTERLEUKIN-1 receptors , *TOLL-like receptors , *EPITHELIAL cells , *CONFOCAL microscopy - Abstract
Lymphocytic choriomeningitis virus (LCMV) and Lassa virus (LASV) share many genetic and biological features including subtle differences between pathogenic and apathogenic strains. Despite remarkable genetic similarity, the viscerotropic WE strain of LCMV causes a fatal LASV fever-like hepatitis in non-human primates (NHPs) while the mouse-adapted Armstrong (ARM) strain of LCMV is deeply attenuated in NHPs and can vaccinate against LCMV-WE challenge. Here, we demonstrate that internalization of WE is more sensitive to the depletion of membrane cholesterol than ARM infection while ARM infection is more reliant on endosomal acidification. LCMV-ARM induces robust NF-κB and interferon response factor (IRF) activation while LCMV-WE seems to avoid early innate sensing and failed to induce strong NF-κB and IRF responses in dual-reporter monocyte and epithelial cells. Toll-like receptor 2 (TLR-2) signaling appears to play a critical role in NF-κB activation and the silencing of TLR-2 shuts down IL-6 production in ARM but not in WE-infected cells. Pathogenic LCMV-WE infection is poorly recognized in early endosomes and failed to induce TLR-2/Mal-dependent pro-inflammatory cytokines. Following infection, Interleukin-1 receptor-associated kinase 1 (IRAK-1) expression is diminished in LCMV-ARM- but not LCMV-WE-infected cells, which indicates it is likely involved in the LCMV-ARM NF-κB activation. By confocal microscopy, ARM and WE strains have similar intracellular trafficking although LCMV-ARM infection appears to coincide with greater co-localization of early endosome marker EEA1 with TLR-2. Both strains co-localize with Rab-7, a late endosome marker, but the interaction with LCMV-WE seems to be more prolonged. These findings suggest that LCMV-ARM's intracellular trafficking pathway may facilitate interaction with innate immune sensors, which promotes the induction of effective innate and adaptive immune responses. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
30. Lymphocytic Choriomeningitis Virus (LCMV): Current Status and Future Directions for Clinical and Molecular Diagnostic Techniques.
- Author
-
Eslami, Narges, Jahanabadi, Somaye, Ziaei, Elham, Shenagari, Mohammad, Salmanzadeh, Shokrollah, Abbasi, Samaneh, and Zandi, Milad
- Subjects
- *
CENTRAL nervous system viral diseases , *LYMPHOCYTIC choriomeningitis virus , *SYMPTOMS , *VIRUS diseases , *NEONATAL mortality - Abstract
Lymphocytic choriomeningitis virus is an RNA virus that is often overlooked despite the potential to cause severe illness. It is a significant cause of viral meningitis, particularly in specific clinical situations. LCMV is transmitted to humans when they come into contact with the secretions of infected mice, and its strong neurotropism primarily results in neurological symptoms. The most vulnerable populations are fetuses and immunosuppressed individuals. LCMV infection acquired through various means can manifest with a wide range of clinical symptoms, varying from being asymptomatic to severe manifestations. Additionally, in cases where individuals are affected by this viral infection, it can result in fatal central nervous system disorders. Specifically, in pregnant women, intrauterine LCMV infection has been observed to lead to fetal or neonatal mortality. Furthermore, it can cause chorioretinitis and hydrocephalus in infants, which not only causes significant harm but also results in long-term impairments. Timely identification and immediate intervention are crucial in improving the prognosis, especially among high-risk groups and regions where the infection is prevalent. Failure to promptly diagnose the condition can lead to significant mortality rates and leave survivors with long-term neurological complications. Consequently, it is imperative to utilize the most appropriate laboratory diagnostic approach, considering the patient's clinical symptoms, exposure history to the virus, and the prevalence of the pathogen in the area, to facilitate accurate clinical detection. This comprehensive review encompasses various diagnostic methodologies employed in managing LCMV, encompassing clinical manifestations, diagnosis, treatment, and potential complications associated with viral infections affecting the central nervous system. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
31. Autoimmunity-associated allele of tyrosine phosphatase gene PTPN22 enhances anti-viral immunity.
- Author
-
Orozco, Robin C., Marquardt, Kristi, Pratumchai, Isaraphorn, Shaikh, Anam Fatima, Mowen, Kerri, Domissy, Alain, Teijaro, John R., and Sherman, Linda A.
- Subjects
- *
PROTEIN-tyrosine phosphatase , *LYMPHOCYTIC choriomeningitis virus , *IMMUNITY , *T cells , *LYMPHOCYTE transformation - Abstract
The 1858C>T allele of the tyrosine phosphatase PTPN22 is present in 5–10% of the North American population and is strongly associated with numerous autoimmune diseases. Although research has been done to define how this allele potentiates autoimmunity, the influence PTPN22 and its pro-autoimmune allele has in anti-viral immunity remains poorly defined. Here, we use single cell RNA-sequencing and functional studies to interrogate the impact of this pro-autoimmune allele on anti-viral immunity during Lymphocytic Choriomeningitis Virus clone 13 (LCMV-cl13) infection. Mice homozygous for this allele (PEP-619WW) clear the LCMV-cl13 virus whereas wildtype (PEP-WT) mice cannot. This is associated with enhanced anti-viral CD4 T cell responses and a more immunostimulatory CD8α- cDC phenotype. Adoptive transfer studies demonstrated that PEP-619WW enhanced anti-viral CD4 T cell function through virus-specific CD4 T cell intrinsic and extrinsic mechanisms. Taken together, our data show that the pro-autoimmune allele of Ptpn22 drives a beneficial anti-viral immune response thereby preventing what is normally a chronic virus infection. Author summary: PTPN22 and its alternative allele, 1858C>T, has largely been studied in the context of autoimmunity. Through these studies, researchers defined roles for PTPN22 in regulating T lymphocyte activation, myeloid cell cytokine production, and macrophage polarization. Despite these immune pathways being critical for anti-viral immunity, little work has studied how this allele impacts virus infection. In this study, we examine gene expression and function of immune cell subsets to demonstrate how a common allelic variant of PTPN22, which strongly increases the risk of autoimmune disease, promotes successful clearance of an otherwise chronic viral infection. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
32. Central tolerance shapes the neutralizing B cell repertoire against a persisting virus in its natural host.
- Author
-
Florava, Marianna, Abreu-Mota, Tiago, Paesen, Guido C., Beetschen, Anna Sophia, Cornille, Karen, Marx, Anna-Friederike, Narr, Kerstin, Sahin, Mehmet, Dimitrova, Mirela, Swarnalekha, Nivedya, Beil-Wagner, Jane, Savic, Natasa, Pelczar, Pawel, Buch, Thorsten, King, Carolyn G., Bowden, Thomas A., and Pinschewer, Daniel D.
- Subjects
- *
B cells , *B cell receptors , *IMMUNOGLOBULIN class switching , *LYMPHOCYTIC choriomeningitis virus , *GERMINAL centers - Abstract
Viral mimicry of host cell structures has been postulated to curtail the B cell receptor (BCR) repertoire against persisting viruses through tolerance mechanisms. This concept awaits, however, experimental testing in a setting of natural virus--host relationship. We engineered mouse models expressing a monoclonal BCR specific for the envelope glycoprotein of lymphocytic choriomeningitis virus (LCMV), a naturally persisting mouse pathogen. When the heavy chain of the LCMV-neutralizing antibody KL25 was paired with its unmutated ancestor light chain, most B cells underwent receptor editing, a behavior reminiscent of autoreactive clones. In contrast, monoclonal B cells expressing the same heavy chain in conjunction with the hypermutated KL25 light chain did not undergo receptor editing but exhibited low levels of surface IgM, suggesting that light chain hypermutation had lessened KL25 autoreactivity. Upon viral challenge, these IgMlow cells were not anergic but up-regulated IgM, participated in germinal center reactions, produced antiviral antibodies, and underwent immunoglobulin class switch as well as further affinity maturation. These studies on a persisting virus in its natural host species suggest that central tolerance mechanisms prune the protective antiviral B cell repertoire. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
33. CD8 + T Cells Mediate Lethal Lung Pathology in the Absence of PD-L1 and Type I Interferon Signalling following LCMV Infection.
- Author
-
Spiteri, Alanna G., Suprunenko, Tamara, Cutts, Erin, Suen, Andrew, Ashhurst, Thomas M., Viengkhou, Barney, King, Nicholas J. C., and Hofer, Markus J.
- Subjects
- *
TYPE I interferons , *T cells , *INTERFERON receptors , *T-cell exhaustion , *PROGRAMMED death-ligand 1 , *LYMPHOCYTIC choriomeningitis virus , *CD8 antigen - Abstract
CD8+ T cells are critical to the adaptive immune response against viral pathogens. However, overwhelming antigen exposure can result in their exhaustion, characterised by reduced effector function, failure to clear virus, and the upregulation of inhibitory receptors, including programmed cell death 1 (PD-1). However, exhausted T cell responses can be "re-invigorated" by inhibiting PD-1 or the primary ligand of PD-1: PD-L1. Further, the absence of the type I interferon receptor IFNAR1 also results in T cell exhaustion and virus persistence in lymphocytic choriomeningitis virus Armstrong (LCMV-Arm)-infected mice. In this study, utilizing single- and double-knockout mice, we aimed to determine whether ablation of PD-1 could restore T cell functionality in the absence of IFNAR1 signalling in LCMV-Arm-infected mice. Surprisingly, this did not re-invigorate the T cell response and instead, it converted chronic LCMV-Arm infection into a lethal disease characterized by severe lung inflammation with an infiltration of neutrophils and T cells. Depletion of CD8+ T cells, but not neutrophils, rescued mice from lethal disease, demonstrating that IFNAR1 is required to prevent T cell exhaustion and virus persistence in LCMV-Arm infection, and in the absence of IFNAR1, PD-L1 is required for survival. This reveals an important interplay between IFNAR1 and PD-L1 with implications for therapeutics targeting these pathways. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
34. Activation of protein kinase receptor (PKR) plays a pro-viral role in mammarenavirus-infected cells.
- Author
-
Witwit, Haydar, Khafaji, Roaa, Salaniwal, Arul, Kim, Arthur S., Cubitt, Beatrice, Jackson, Nathaniel, Chengjin Ye, Weiss, Susan R., Martinez-Sobrido, Luis, and Carlos de la Torre, Juan
- Subjects
- *
PROTEIN receptors , *PROTEIN kinases , *LYMPHOCYTIC choriomeningitis virus , *DOUBLE-stranded RNA , *CELL anatomy - Abstract
Many viruses, including mammarenaviruses, have evolved mechanisms to counteract different components of the host cell innate immunity, which is required to facilitate robust virus multiplication. The double-stranded RNA (dsRNA) sensor protein kinase receptor (PKR) pathway plays a critical role in the cell anti-viral response. Whether PKR can restrict the multiplication of the Old World mammarenavirus lymphocytic choriomeningitis virus (LCMV) and the mechanisms by which LCMV may counteract the anti-viral functions of PKR have not yet been investigated. Here we present evidence that LCMV infection results in very limited levels of PKR activation, but LCMV multiplication is enhanced in the absence of PKR. In contrast, infection with a recombinant LCMV with a mutation affecting the 3'-5' exonuclease (ExoN) activity of the viral nucleoprotein resulted in robust PKR activation in the absence of detectable levels of dsRNA, which was associated with severely restricted virus multiplication that was alleviated in the absence of PKR. However, pharmacological inhibition of PKR activation resulted in reduced levels of LCMV multiplication. These findings uncovered a complex role of the PKR pathway in LCMV-infected cells involving both pro- and anti-viral activities. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
35. Lymphocytic choriomeningitis arenavirus requires cellular COPI and AP-4 complexes for efficient virion production.
- Author
-
Byford, Owen, Shaw, Amelia B., Hiu Nam Tse, Todd, Eleanor J. A. A., Álvarez-Rodríguez, Beatriz, Hewson, Roger, Fontana, Juan, and Barr, John N.
- Subjects
- *
LYMPHOCYTIC choriomeningitis , *LYMPHOCYTIC choriomeningitis virus , *ARENAVIRUSES , *GREEN fluorescent protein , *VIRAL envelope proteins , *ADAPTOR proteins , *VIRION , *COAT proteins (Viruses) - Abstract
Lymphocytic choriomeningitis virus (LCMV) is a bisegmented negativesense RNA virus classified within the Arenaviridae family of the Bunyavirales order. LCMV is associated with fatal disease in immunocompromized populations, and as the prototypical arenavirus, acts as a model for the many serious human pathogens within this group. Here, we examined the dependence of LCMV multiplication on cellular trafficking components using a recombinant LCMV expressing enhanced green fluorescent protein in conjunction with a curated siRNA library. The screen revealed a requirement for subunits of both the coat protein 1 (COPI) coatomer and adapter protein 4 (AP-4) complexes. By rescuing a recombinant LCMV harboring a FLAG-tagged glycoprotein (GP-1) envelope spike (rLCMV-GP1-FLAG), we showed infection resulted in marked co-localization of individual COPI and AP-4 components with both LCMV nucleoprotein (NP) and GP-1, consistent with their involvement in viral processes. To further investigate the role of both COPI and AP-4 complexes during LCMV infection, we utilized the ARF-I inhibitor brefeldin A (BFA) that prevents complex formation. Within a single 12-h cycle of virus multiplication, BFA pre-treatment caused no significant change in LCMV-specific RNA synthesis, alongside no significant change in LCMV NP expression, as measured by BFA time-of-addition experiments. In contrast, BFA addition resulted in a significant drop in released virus titers, approaching 50-fold over the same 12-h period, rising to over 600-fold over 24 h. Taken together, these findings suggest COPI and AP-4 complexes are important host cell factors required for the formation and release of infectious LCMV. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
36. The New South Wales Mouse Plague 2020-2021: A One Health description
- Author
-
Jennifer White, Joanne Taylor, Peter R. Brown, Steve Henry, Lucy Carter, Aditi Mankad, Wei-Shan Chang, Priscilla Stanley, Kerry Collins, David N. Durrheim, and Kirrilly Thompson
- Subjects
Australia ,Disaster ,Encephalomyocarditis virus ,Leptospirosis ,Lymphocytic choriomeningitis virus ,Mental health ,Medicine (General) ,R5-920 - Abstract
A mouse plague occurred in Eastern Australia from spring 2020 to winter 2021, impacting an area of around 180,000 km2. It harmed human physical and psychological health, damaged the natural and built environment, and endangered farmed, domestic and native animals. However, the mouse plague was overshadowed by the COVID-19 pandemic, especially as the end of the plague coincided with the arrival and surge of the COVID-19 delta strain in rural New South Wales (NSW). In this article, we systematically overview the multiple impacts of the plague and highlight their complex interactions. Using a One Health framework, we comprehensively review the i) human, ii) animal and iii) environmental impacts including economic dimensions. Given the damage that the mouse plague caused to infrastructure, we consider the environment from two perspectives: the natural and the built environment. This One Health description of the 2020–2021 mouse plague identifies priorities for preparedness, response and recovery at local, regional land levels to inform response and management of future mouse plague events in Australia. It also highlights the need for ongoing collaboration between researchers and practitioners in the human, animal and environmental health sectors.
- Published
- 2024
- Full Text
- View/download PDF
37. Genome-Wide Knockout Screen Identifies Human Sialomucin CD164 as an Essential Entry Factor for Lymphocytic Choriomeningitis Virus
- Author
-
Liu, Jamin, Knopp, Kristeene A, Rackaityte, Elze, Wang, Chung Yu, Laurie, Matthew T, Sunshine, Sara, Puschnik, Andreas S, and DeRisi, Joseph L
- Subjects
Prevention ,Biotechnology ,Emerging Infectious Diseases ,Vaccine Related ,Biodefense ,Infectious Diseases ,2.1 Biological and endogenous factors ,Aetiology ,2.2 Factors relating to the physical environment ,Infection ,Cysteine ,Endolyn ,Female ,Humans ,Lymphocytic Choriomeningitis ,Lymphocytic choriomeningitis virus ,Placenta ,Pregnancy ,Sialomucins ,CD164 ,CRISPR screen ,lymphocytic choriomeningitis virus ,viral entry ,Microbiology - Abstract
Lymphocytic choriomeningitis virus (LCMV) is a well-studied mammarenavirus that can be fatal in congenital infections. However, our understanding of LCMV and its interactions with human host factors remains incomplete. Here, host determinants affecting LCMV infection were investigated through a genome-wide CRISPR knockout screen in A549 cells, a human lung adenocarcinoma line. We identified and validated a variety of novel host factors that play a functional role in LCMV infection. Among these, knockout of the sialomucin CD164, a heavily glycosylated transmembrane protein, was found to ablate infection with multiple LCMV strains but not other hemorrhagic mammarenaviruses in several cell types. Further characterization revealed a dependency of LCMV entry on the cysteine-rich domain of CD164, including an N-linked glycosylation site at residue 104 in that region. Given the documented role of LCMV with respect to transplacental human infections, CD164 expression was investigated in human placental tissue and placental cell lines. CD164 was found to be highly expressed in the cytotrophoblast cells, an initial contact site for pathogens within the placenta, and LCMV infection in placental cells was effectively blocked using a monoclonal antibody specific to the cysteine-rich domain of CD164. Together, this study identifies novel factors associated with LCMV infection of human tissues and highlights the importance of CD164, a sialomucin that previously had not been associated with viral infection. IMPORTANCE Lymphocytic choriomeningitis virus (LCMV) is a human-pathogenic mammarenavirus that can be fatal in congenital infections. Although frequently used in the study of persistent infections in the field of immunology, aspects of this virus's life cycle remain incomplete. For example, while viral entry has been shown to depend on a cell adhesion molecule, DAG1, genetic knockout of this gene allows for residual viral infection, implying that additional receptors can mediate cell entry. The significance of our study is the identification of host factors important for successful infection, including the sialomucin CD164, which had not been previously associated with viral infection. We demonstrated that CD164 is essential for LCMV entry into human cells and can serve as a possible therapeutic target for treatment of congenital infection.
- Published
- 2022
38. Lymphocytic Choriomeningitis Virus in Person Living with HIV, Connecticut, USA, 2021
- Author
-
Jonathan Dyal, Shiv Gandhi, Caitlin M. Cossaboom, Austin Leach, Ketan Patel, Marjorie Golden, Joseph Canterino, Marie-Louise Landry, Debi Cannon, Mary Choi, Inna Krapiunaya, John D. Klena, and Trevor Shoemaker
- Subjects
Lymphocytic choriomeningitis virus ,HIV ,Connecticut ,United States ,arenavirus ,rodent-borne ,Medicine ,Infectious and parasitic diseases ,RC109-216 - Abstract
Lymphocytic choriomeningitis virus is an underreported cause of miscarriage and neurologic disease. Surveillance remains challenging because of nonspecific symptomatology, inconsistent case reporting, and difficulties with diagnostic testing. We describe a case of acute lymphocytic choriomeningitis virus disease in a person living with HIV in Connecticut, USA, identified by using quantitative reverse transcription PCR.
- Published
- 2023
- Full Text
- View/download PDF
39. Lymphocytic Choriomeningitis Virus Lineage V in Wood Mice, Germany
- Author
-
Calvin Mehl, Olayide Abraham Adeyemi, Claudia Wylezich, Dirk Höper, Martin Beer, Cornelia Triebenbacher, Gerald Heckel, and Rainer G. Ulrich
- Subjects
lymphocytic choriomeningitis virus ,LCMV ,lineage V ,wood mice ,Apodemus sylvaticus ,meningitis/encephalitis ,Medicine ,Infectious and parasitic diseases ,RC109-216 - Abstract
We identified a novel lineage of lymphocytic choriomeningitis virus, tentatively named lineage V, in wood mice (Apodemus sylvaticus) from Germany. Wood mouse–derived lymphocytic choriomeningitis virus can be found across a substantially greater range than previously thought. Increased surveillance is needed to determine its geographic range and zoonotic potential.
- Published
- 2024
- Full Text
- View/download PDF
40. Understanding the Neurotrophic Virus Mechanisms and Their Potential Effect on Systemic Lupus Erythematosus Development.
- Author
-
Uribe, Felipe R., González, Valentina P. I., Kalergis, Alexis M., Soto, Jorge A., and Bohmwald, Karen
- Subjects
- *
SYSTEMIC lupus erythematosus , *HUMAN herpesvirus 1 , *VIRUS diseases , *LYMPHOCYTIC choriomeningitis virus , *HERPES simplex virus , *CENTRAL nervous system viral diseases , *ADENOVIRUS diseases - Abstract
Central nervous system (CNS) pathologies are a public health concern, with viral infections one of their principal causes. These viruses are known as neurotropic pathogens, characterized by their ability to infiltrate the CNS and thus interact with various cell populations, inducing several diseases. The immune response elicited by neurotropic viruses in the CNS is commanded mainly by microglia, which, together with other local cells, can secrete inflammatory cytokines to fight the infection. The most relevant neurotropic viruses are adenovirus (AdV), cytomegalovirus (CMV), enterovirus (EV), Epstein–Barr Virus (EBV), herpes simplex virus type 1 (HSV-1), and herpes simplex virus type 2 (HSV-2), lymphocytic choriomeningitis virus (LCMV), and the newly discovered SARS-CoV-2. Several studies have associated a viral infection with systemic lupus erythematosus (SLE) and neuropsychiatric lupus (NPSLE) manifestations. This article will review the knowledge about viral infections, CNS pathologies, and the immune response against them. Also, it allows us to understand the relevance of the different viral proteins in developing neuronal pathologies, SLE and NPSLE. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
41. Armstrong strain lymphocytic choriomeningitis virus infection after accidental laboratory exposure.
- Author
-
Caron, Laurence, Delisle, Jean-Sébastien, Strong, James E, Deschambault, Yvon, Lombard-Vadnais, Félix, Labbé, Annie-Claude, and Lesage, Sylvie
- Subjects
- *
LYMPHOCYTIC choriomeningitis virus , *VIRUS diseases , *SUMATRIPTAN , *RESEARCH personnel , *EYE pain , *LABORATORY personnel , *IMMUNOGLOBULINS - Abstract
Background: Lymphocytic choriomeningitis virus (LCMV) is a human pathogen naturally present in wild rodents. In addition, LCMV is routinely used in immunology research as a model of viral infection in mice. The Armstrong common laboratory strain and the Clone-13 variant induce acute and chronic infections in mice, respectively. The frequent use of this virus in laboratory settings is associated with a risk of human infection for laboratory personnel. In contrast to LCMV Clone-13, few human laboratory infections with LCMV Armstrong have been reported, leading to a poor understanding of symptoms related to infection with this specific LCMV strain. Case presentation: A researcher accidentally infected herself percutaneously with LCMV Armstrong. Symptoms including headaches, dizziness, eye pain and nausea appeared seven days post-exposure and lasted ten days. LCMV-IgM antibodies were detected at 28 days post-infection and IgG seroconversion was observed later. Complete recovery was confirmed three months post exposure. Conclusions: Research involving live viruses comes with the risk of infection for research personnel. This case is the first reported accidental human infection with LCMV Armstrong. The symptoms differed from reported infections with LCMV Clone-13, by the absence of fever and vomiting, and presence of leg numbness. This report will therefore help clinicians and public health authorities to recognize the symptoms associated with LCMV Armstrong infections and to offer appropriate counselling to individuals who accidentally expose themselves. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
42. PDIA4 Is a Host Factor Important for Lymphocytic Choriomeningitis Virus Infection.
- Author
-
Xu, Mengwei, Xu, Huan, Wan, Weiwei, Jian, Xiaoqin, Jin, Runming, Wang, Lin, Wang, Jingshi, Xiao, Gengfu, Zhang, Leike, Chen, Hongbo, and Wen, Yuxi
- Subjects
- *
LYMPHOCYTIC choriomeningitis virus , *VIRUS diseases , *PROTEIN disulfide isomerase , *HEMORRHAGIC diseases , *RNA synthesis - Abstract
Mammalian arenaviruses are rodent-borne zoonotic viruses, some of which can cause fatal hemorrhagic diseases in humans. The first discovered arenavirus, lymphocytic choriomeningitis virus (LCMV), has a worldwide distribution and can be fatal for transplant recipients. However, no FDA-approved drugs or vaccines are currently available. In this study, using a quantitative proteomic analysis, we identified a variety of host factors that could be needed for LCMV infection, among which we found that protein disulfide isomerase A4 (PDIA4), a downstream factor of endoplasmic reticulum stress (ERS), is important for LCMV infection. Biochemical analysis revealed that LCMV glycoprotein was the main viral component accounting for PDIA4 upregulation. The inhibition of ATF6-mediated ERS could prevent the upregulation of PDIA4 that was stimulated by LCMV infection. We further found that PDIA4 can affect the LCMV viral RNA synthesis processes and release. In summary, we conclude that PDIA4 could be a new target for antiviral drugs against LCMV. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
43. Miscellaneous CNS Viral Infections: Underappreciated Causes of Neurologic Disease
- Author
-
Sami, Hiba, Firoze, Safiya, Khan, Parvez A., Sami, Hiba, editor, Firoze, Safiya, editor, and Khan, Parvez A., editor
- Published
- 2023
- Full Text
- View/download PDF
44. Lymphocytic Choriomeningitis Virus (LCMV) Infection in Children and Hearing Loss
- Author
-
Gülmez, Emrah, Yasar, Mehmet, Karpischenko, Sergei, Cingi, Cemal, Series Editor, Arısoy, Ayşe Engin, editor, Arısoy, Emin Sami, editor, Bayar Muluk, Nuray, editor, and Correa, Armando G., editor
- Published
- 2023
- Full Text
- View/download PDF
45. Pseudotyped Viruses for Mammarenavirus
- Author
-
Li, Qianqian, Huang, Weijing, Wang, Youchun, Crusio, Wim E., Series Editor, Dong, Haidong, Series Editor, Radeke, Heinfried H., Series Editor, Rezaei, Nima, Series Editor, Steinlein, Ortrud, Series Editor, Xiao, Junjie, Series Editor, and Wang, Youchun, editor
- Published
- 2023
- Full Text
- View/download PDF
46. Early innate and adaptive immune perturbations determine long-term severity of chronic virus and Mycobacterium tuberculosis coinfection
- Author
-
Xu, Wenxi, Snell, Laura M, Guo, Mengdi, Boukhaled, Giselle, Macleod, Bethany L, Li, Ming, Tullius, Michael V, Guidos, Cynthia J, Tsao, Ming-Sound, Divangahi, Maziar, Horwitz, Marcus A, Liu, Jun, and Brooks, David G
- Subjects
Medical Microbiology ,Biomedical and Clinical Sciences ,Immunology ,Rare Diseases ,Tuberculosis ,Infectious Diseases ,HIV/AIDS ,Lung ,2.2 Factors relating to the physical environment ,Aetiology ,2.1 Biological and endogenous factors ,Infection ,Good Health and Well Being ,Adaptive Immunity ,Animals ,Chronic Disease ,Coinfection ,Immunity ,Innate ,Lymphocytic Choriomeningitis ,Lymphocytic choriomeningitis virus ,Mice ,Mice ,Inbred C57BL ,Mice ,Transgenic ,Mycobacterium tuberculosis ,Neutrophils ,Phagocytosis ,Severity of Illness Index ,Th1 Cells ,Th17 Cells ,Time Factors ,CD4 T cells ,CyTOF ,LCMV ,T cell differentiation ,Th1 cells ,Th17 cells ,chronic viral infection ,coinfection ,neutrophil - Abstract
Chronic viral infections increase severity of Mycobacterium tuberculosis (Mtb) coinfection. Here, we examined how chronic viral infections alter the pulmonary microenvironment to foster coinfection and worsen disease severity. We developed a coordinated system of chronic virus and Mtb infection that induced central clinical manifestations of coinfection, including increased Mtb burden, extra-pulmonary dissemination, and heightened mortality. These disease states were not due to chronic virus-induced immunosuppression or exhaustion; rather, increased amounts of the cytokine TNFα initially arrested pulmonary Mtb growth, impeding dendritic cell mediated antigen transportation to the lymph node and subverting immune-surveillance, allowing bacterial sanctuary. The cryptic Mtb replication delayed CD4 T cell priming, redirecting T helper (Th) 1 toward Th17 differentiation and increasing pulmonary neutrophilia, which diminished long-term survival. Temporally restoring CD4 T cell induction overcame these diverse disease sequelae to enhance Mtb control. Thus, Mtb co-opts TNFα from the chronic inflammatory environment to subvert immune-surveillance, avert early immune function, and foster long-term coinfection.
- Published
- 2021
47. LYMPHOCYTIC CHORIOMENINGITIS VIRUS INFECTION
- Author
-
Teodora Gladnishka, Iva Trifonova, Vladislava Ivanova, and Iva Christova
- Subjects
Lymphocytic choriomeningitis virus ,Diagnosis ,serology ,Infectious and parasitic diseases ,RC109-216 - Abstract
Background: Lymphocytic choriomeningitis virus (LHMV) infection is a neglected rodent-borne zoonotic infection but it is found all over the world because of the cosmopolitan distribution of its reservoirs. The diagnostic of this disease is not widely applied that is why it has been underreported. The aim of this study is to investigate infection with LCMV in hospitalized patients in 2015-2022 in Bulgaria and to analyse the data compared to the worldwide data available in this field of research. Materials/methods: A total of 66 serum samples and 25 cerebrospinal fluid (CSF) samples from 73 patients with suspected LCMV infection from different hospitals in Bulgaria were collected. All samples were tested with a commercial enzyme-linked immunosorbent assay (Human LCMV-Ab ELISA, SSBT, China), based on the principle of double-antibody sandwich technique to detect Human LCMV-Antibody. Results: A total of 11/91 (12.09%) positive samples were found in 5 males and 6 females throughout the study period. The positive samples were from patients from the cities: Sofia, Stara Zagora, Montana. A total of 3/25 (12%) positive samples were from CSF samples and 8/66 positive samples (12.12%) were from serum samples. Conclusions: It’s found that this infection occurs in our country and should not be underestimated, due to the possible severe neurological course and the danger of fetal damage in pregnant women. The diagnosis of LCMV infection is based on previous experience, placed in the light of the continuous introduction of new more sensitive and specific approaches.
- Published
- 2024
- Full Text
- View/download PDF
48. Neuropilin-1 identifies a subset of highly activated CD8+ T cells during parasitic and viral infections.
- Author
-
Abberger, Hanna, Hose, Matthias, Ninnemann, Anne, Menne, Christopher, Eilbrecht, Mareike, Lang, Karl S., Matuschewski, Kai, Geffers, Robert, Herz, Josephine, Buer, Jan, Westendorf, Astrid M., and Hansen, Wiebke
- Subjects
- *
T cells , *CYTOTOXIC T cells , *PARASITIC diseases , *VIRUS diseases , *LYMPHOCYTIC choriomeningitis virus , *VASCULAR endothelial growth factors , *CEREBRAL malaria - Abstract
Neuropilin-1 (Nrp-1) expression on CD8+ T cells has been identified in tumor-infiltrating lymphocytes and in persistent murine gamma-herpes virus infections, where it interferes with the development of long-lived memory T cell responses. In parasitic and acute viral infections, the role of Nrp-1 expression on CD8+ T cells remains unclear. Here, we demonstrate a strong induction of Nrp-1 expression on CD8+ T cells in Plasmodium berghei ANKA (PbA)-infected mice that correlated with neurological deficits of experimental cerebral malaria (ECM). Likewise, the frequency of Nrp-1+CD8+ T cells was significantly elevated and correlated with liver damage in the acute phase of lymphocytic choriomeningitis virus (LCMV) infection. Transcriptomic and flow cytometric analyses revealed a highly activated phenotype of Nrp-1+CD8+ T cells from infected mice. Correspondingly, in vitro experiments showed rapid induction of Nrp-1 expression on CD8+ T cells after stimulation in conjunction with increased expression of activation-associated molecules. Strikingly, T cell-specific Nrp-1 ablation resulted in reduced numbers of activated T cells in the brain of PbA-infected mice as well as in spleen and liver of LCMV-infected mice and alleviated the severity of ECM and LCMV-induced liver pathology. Mechanistically, we identified reduced blood-brain barrier leakage associated with reduced parasite sequestration in the brain of PbA-infected mice with T cell-specific Nrp-1 deficiency. In conclusion, Nrp-1 expression on CD8+ T cells represents a very early activation marker that exacerbates deleterious CD8+ T cell responses during both, parasitic PbA and acute LCMV infections. Author summary: Malaria is caused by the parasite Plasmodium and remains one of the most life threatening infectious diseases worldwide. In 2021, WHO reported 247 million malaria cases and more than 600,000 deaths. In this study, we infect mice with Plasmodium berghei ANKA (PbA), a scientific model of experimental cerebral malaria (ECM), which affects the brain and is often fatal. During infection, CD8+ T cells are known to invade the brain and contribute to brain damage and inflammation through their activated and cytotoxic potential. We investigate whether T cell-expressed Neuropilin-1 (Nrp-1), a co-receptor for semaphorin and vascular endothelial growth factor (VEGF) known to play a role in immune cell activation and migration, has an impact on ECM manifestation. The results of our study demonstrate that Nrp-1 is associated with increased T cell activation and significantly aggravates the severity of ECM. Importantly, our results are also applicable to other CD8+ T cell-induced immunopathologies as we show similar effects during lymphocytic choriomeningitis virus (LCMV) infection. Together, this implies that Nrp-1 may be an early prognostic marker and blockade of Nrp-1 could be a promising target to control exacerbated T cell responses in different diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
49. c-Myc uses Cul4b to preserve genome integrity and promote antiviral CD8+ T cell immunity.
- Author
-
Dar, Asif A., Kim, Dale D., Gordon, Scott M., Klinzing, Kathleen, Rosen, Siera, Guha, Ipsita, Porter, Nadia, Ortega, Yohaniz, Forsyth, Katherine S., Roof, Jennifer, Fazelinia, Hossein, Spruce, Lynn A., Eisenlohr, Laurence C., Behrens, Edward M., and Oliver, Paula M.
- Subjects
T cells ,UBIQUITINATION ,DNA repair ,LONGEVITY ,LYMPHOCYTIC choriomeningitis virus ,DNA damage ,GENOMES ,VIRAL load - Abstract
During infection, virus-specific CD8
+ T cells undergo rapid bursts of proliferation and differentiate into effector cells that kill virus-infected cells and reduce viral load. This rapid clonal expansion can put T cells at significant risk for replication-induced DNA damage. Here, we find that c-Myc links CD8+ T cell expansion to DNA damage response pathways though the E3 ubiquitin ligase, Cullin 4b (Cul4b). Following activation, c-Myc increases the levels of Cul4b and other members of the Cullin RING Ligase 4 (CRL4) complex. Despite expressing c-Myc at high levels, Cul4b-deficient CD8+ T cells do not expand and clear the Armstrong strain of lymphocytic choriomeningitis virus (LCMV) in vivo. Cul4b-deficient CD8+ T cells accrue DNA damage and succumb to proliferative catastrophe early after antigen encounter. Mechanistically, Cul4b knockout induces an accumulation of p21 and Cyclin E2, resulting in replication stress. Our data show that c-Myc supports cell proliferation by maintaining genome stability via Cul4b, thereby directly coupling these two interdependent pathways. These data clarify how CD8+ T cells use c-Myc and Cul4b to sustain their potential for extraordinary population expansion, longevity and antiviral responses. Although T cells proliferate at an exceptionally fast rate following activation, the mechanisms sustaining such fast proliferation remain unclear. Here, the authors demonstrate that c-Myc upregulates the E3 ubiquitin ligase Cul4b to reduce replication stress in CD8+ T cells, allowing massive clonal expansion and viral clearance. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
50. Mathematical Model Predicting the Kinetics of Intracellular LCMV Replication.
- Author
-
Sergeeva, Julia, Grebennikov, Dmitry, Casella, Valentina, Cebollada Rica, Paula, Meyerhans, Andreas, and Bocharov, Gennady
- Subjects
- *
MATHEMATICAL models , *LYMPHOCYTIC choriomeningitis virus , *MULTISCALE modeling , *LIFE cycles (Biology) , *VIRAL variation - Abstract
The lymphocytic choriomeningitis virus (LCMV) is a non-cytopathic virus broadly used in fundamental immunology as a mouse model for acute and chronic virus infections. LCMV remains a cause of meningitis in humans, in particular the fatal LCMV infection in organ transplant recipients, which highlights the pathogenic potential and clinical significance of this neglected human pathogen. Paradoxically, the kinetics of the LCMV intracellular life cycle has not been investigated in detail. In this study, we formulate and calibrate a mathematical model predicting the kinetics of biochemical processes, including the transcription, translation, and degradation of molecular components of LCMV underlying its replication in infected cells. The model is used to study the sensitivity of the virus growth, providing a clear ranking of intracellular virus replication processes with respect to their contribution to net viral production. The stochastic formulation of the model enables the quantification of the variability characteristics in viral production, probability of productive infection and secretion of protein-deficient viral particles. As it is recognized that antiviral therapeutic options in human LCMV infection are currently limited, our results suggest potential targets for antiviral therapies. The model provides a currently missing building module for developing multi-scale mathematical models of LCMV infection in mice. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.