Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Emerg Infect Dis ; 30(4): 817-821, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38526320

ABSTRACT

Orthohantaviruses cause hantavirus cardiopulmonary syndrome; most cases occur in the southwest region of the United States. We discuss a clinical case of orthohantavirus infection in a 65-year-old woman in Michigan and the phylogeographic link of partial viral fragments from the patient and rodents captured near the presumed site of infection.


Subject(s)
Hantavirus Infections , Orthohantavirus , Female , Humans , Aged , Michigan/epidemiology , Phylogeography , Syndrome
2.
PLoS Negl Trop Dis ; 18(1): e0011672, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38215158

ABSTRACT

BACKGROUND: Hantaviruses are negative-stranded RNA viruses that can sometimes cause severe disease in humans; however, they are maintained in mammalian host populations without causing harm. In Panama, sigmodontine rodents serve as hosts to transmissible hantaviruses. Due to natural and anthropogenic forces, these rodent populations are having increased contact with humans. METHODS: We extracted RNA and performed Illumina deep metatranscriptomic sequencing on Orthohantavirus seropositive museum tissues from rodents. We acquired sequence reads mapping to Choclo virus (CHOV, Orthohantavirus chocloense) from heart and kidney tissue of a two-decade old frozen museum sample from a Costa Rican pygmy rice rat (Oligoryzomys costaricensis) collected in Panama. Reads mapped to the CHOV reference were assembled and then validated by visualization of the mapped reads against the assembly. RESULTS: We recovered a 91% complete consensus sequence from a reference-guided assembly to CHOV with an average of 16X coverage. The S and M segments used in our phylogenetic analyses were nearly complete (98% and 99%, respectively). There were 1,199 ambiguous base calls of which 93% were present in the L segment. Our assembled genome varied 1.1% from the CHOV reference sequence resulting in eight nonsynonymous mutations. Further analysis of all publicly available partial S segment sequences support a clear relationship between CHOV clinical cases and O. costaricensis acquired strains. CONCLUSIONS: Viruses occurring at extremely low abundances can be recovered from deep metatranscriptomics of archival tissues housed in research natural history museum biorepositories. Our efforts resulted in the second CHOV genome publicly available. This genomic data is important for future surveillance and diagnostic tools as well as understanding the evolution and pathogenicity of CHOV.


Subject(s)
Orthohantavirus , Sigmodontinae , Animals , Rats , Humans , Phylogeny , Rodentia , Biological Specimen Banks
3.
PLoS One ; 19(1): e0296718, 2024.
Article in English | MEDLINE | ID: mdl-38236803

ABSTRACT

Orthohantaviruses are diverse zoonotic RNA viruses. Small mammals, such as mice and rats are common chronic, asymptomatic hosts that transmit the virus through their feces and urine. In North America, hantavirus infection primarily causes hantavirus cardiopulmonary syndrome (HCPS), which has a mortality rate of nearly 36%. In the United States of America, New Mexico (NM) is leading the nation in the number of HCPS-reported cases (N = 129). However, no reported cases of HCPS have occurred within eastern NM. In this study, we assessed the prevalence of Sin Nombre virus (SNV) in rodent assemblages across eastern NM, using RT-qPCR. We screened for potential rodent hosts in the region, as well as identified areas that may pose significant infection risk to humans. We captured and collected blood and lung tissues from 738 rodents belonging to 23 species. 167 individuals from 16 different species were positive for SNV RNA by RT-qPCR, including 6 species unreported in the literature: Onychomys leucogaster (Northern grasshopper mouse), Dipodomys merriami (Merriam's kangaroo rat), Dipodomys ordii (Ord's kangaroo rat), Dipodomys spectabilis (Banner-tailed kangaroo rat), Perognathus flavus (Silky pocket mouse), and Chaetodipus hispidus (Hispid pocket mouse). The infection rates did not differ between sexes or rodent families (i.e., Cricetidae vs. Heteromyidae). Generalized linear model showed that disturbed habitat types positively influenced the prevalence of SNV at sites of survey. Overall, the results of this study indicate that many rodent species in east New Mexico have the potential to maintain SNV in the environment, but further research is needed to assess species specific infectivity mechanisms and potential risk to humans.


Subject(s)
Hantavirus Infections , Hantavirus Pulmonary Syndrome , Orthohantavirus , Sin Nombre virus , Humans , Animals , Mice , Rodentia , Dipodomys , Sin Nombre virus/genetics , New Mexico/epidemiology , Prevalence , Hantavirus Infections/epidemiology , Hantavirus Infections/veterinary , Orthohantavirus/genetics , Arvicolinae , Hantavirus Pulmonary Syndrome/epidemiology , Hantavirus Pulmonary Syndrome/veterinary
4.
Viruses ; 14(4)2022 03 25.
Article in English | MEDLINE | ID: mdl-35458412

ABSTRACT

Orthohantaviruses are negative-stranded RNA viruses with trisegmented genomes that can cause severe disease in humans and are carried by several host reservoirs throughout the world. Old World orthohantaviruses are primarily located throughout Europe and Asia, causing hemorrhagic fever with renal syndrome, and New World orthohantaviruses are found in North, Central, and South America, causing hantavirus cardiopulmonary syndrome (HCPS). In the United States, Sin Nombre orthohantavirus (SNV) is the primary cause of HCPS with a fatality rate of ~36%. The primary SNV host reservoir is thought to be the North American deer mouse, Peromyscus maniculatus. However, it has been shown that other species of Peromyscus can carry different orthohantaviruses. Few studies have systemically surveyed which orthohantaviruses may exist in wild-caught rodents or monitored spillover events into additional rodent reservoirs. A method for the rapid detection of orthohantaviruses is needed to screen large collections of rodent samples. Here, we report a pan-orthohantavirus, two-step reverse-transcription quantitative real-time PCR (RT-qPCR) tool designed to detect both Old and New World pathogenic orthohantavirus sequences of the S segment of the genome and validated them using plasmids and authentic viruses. We then performed a screening of wild-caught rodents and identified orthohantaviruses in lung tissue, and we confirmed the findings by Sanger sequencing. Furthermore, we identified new rodent reservoirs that have not been previously reported as orthohantavirus carriers. This novel tool can be used for the efficient and rapid detection of various orthohantaviruses, while uncovering potential new orthohantaviruses and host reservoirs that may otherwise go undetected.


Subject(s)
Hantavirus Infections , Hantavirus Pulmonary Syndrome , Orthohantavirus , Rodent Diseases , Sin Nombre virus , Animals , Disease Reservoirs , Orthohantavirus/genetics , Hantavirus Infections/diagnosis , Hantavirus Infections/epidemiology , Hantavirus Infections/veterinary , Peromyscus , Rodent Diseases/epidemiology , Rodentia
5.
Cell Biol Toxicol ; 38(1): 31-41, 2022 02.
Article in English | MEDLINE | ID: mdl-34021430

ABSTRACT

Anti-inflammatory and proinflammatory responses in macrophages are influenced by cellular metabolism. Macrophages are the primary phagocyte in mucosal environments (i.e., intestinal tract and lungs) acting as first-line defense against microorganisms and environmental pollutants. Given the extensive contamination of our food and water sources with microplastics, we aimed to examine the metabolic response in macrophages to microplastic particles (MPs). Utilizing murine macrophages, we assessed the metabolic response of macrophages after polystyrene MP phagocytosis. The phagocytosis of MP by macrophages induced a metabolic shift toward glycolysis and a reduction in mitochondrial respiration that was associated with an increase of cell surface markers CD80 and CD86 and cytokine gene expression associated with glycolysis. The gastrointestinal consequences of this metabolic switch in the context of an immune response remain uncertain, but the global rise of plastic pollution and MP ingestion potentially poses an unappreciated health risk. Macrophage phagocytosis of microplastics alters cellular metabolism. - Macrophages cannot degrade PS MP. - MP phagocytosis increases glycolysis in murine macrophages. - MP phagocytosis reduces mitochondrial respiration in murine macrophages.


Subject(s)
Microplastics , Water Pollutants, Chemical , Animals , Gastrointestinal Tract , Macrophages/chemistry , Mice , Microplastics/toxicity , Plastics , Polystyrenes/toxicity , Water Pollutants, Chemical/analysis
6.
J Crohns Colitis ; 16(2): 259-274, 2022 Feb 23.
Article in English | MEDLINE | ID: mdl-34374750

ABSTRACT

Intestinal myeloid cells play a critical role in balancing intestinal homeostasis and inflammation. Here, we report that expression of the autophagy-related 5 [Atg5] protein in myeloid cells prevents dysbiosis and excessive intestinal inflammation by limiting IL-12 production. Mice with a selective genetic deletion of Atg5 in myeloid cells [Atg5ΔMye] showed signs of dysbiosis preceding colitis, and exhibited severe intestinal inflammation upon colitis induction that was characterised by increased IFNγ production. The exacerbated colitis was linked to excess IL-12 secretion from Atg5-deficient myeloid cells and gut dysbiosis. Restoration of the intestinal microbiota or genetic deletion of IL-12 in Atg5ΔMye mice attenuated the intestinal inflammation in Atg5ΔMye mice. Additionally, Atg5 functions to limit IL-12 secretion through modulation of late endosome [LE] acidity. Last, the autophagy cargo receptor NBR1, which accumulates in Atg5-deficient cells, played a role by delivering IL-12 to LE. In summary, Atg5 expression in intestinal myeloid cells acts as an anti-inflammatory brake to regulate IL-12, thus preventing dysbiosis and uncontrolled IFNγ-driven intestinal inflammation.


Subject(s)
Colitis , Dysbiosis , Animals , Autophagy/genetics , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Colitis/chemically induced , Colitis/prevention & control , Inflammation/metabolism , Interleukin-12 , Intracellular Signaling Peptides and Proteins , Mice , Mice, Inbred C57BL
7.
J Virol ; 95(23): e0153421, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34549977

ABSTRACT

Sin Nombre orthohantavirus (SNV), a negative-sense, single-stranded RNA virus that is carried and transmitted by the North American deer mouse Peromyscus maniculatus, can cause infection in humans through inhalation of aerosolized excreta from infected rodents. This infection can lead to hantavirus cardiopulmonary syndrome (HCPS), which has an ∼36% case-fatality rate. We used reverse transcriptase quantitative PCR (RT-qPCR) to confirm SNV infection in a patient and identified SNV in lung tissues in wild-caught rodents from potential sites of exposure. Using viral whole-genome sequencing (WGS), we identified the likely site of transmission and discovered SNV in multiple rodent species not previously known to carry the virus. Here, we report, for the first time, the use of SNV WGS to pinpoint a likely site of human infection and identify SNV simultaneously in multiple rodent species in an area of known host-to-human transmission. These results will impact epidemiology and infection control for hantaviruses by tracing zoonotic transmission and investigating possible novel host reservoirs. IMPORTANCE Orthohantaviruses cause severe disease in humans and can be lethal in up to 40% of cases. Sin Nombre orthohantavirus (SNV) is the main cause of hantavirus disease in North America. In this study, we sequenced SNV from an infected patient and wild-caught rodents to trace the location of infection. We also discovered SNV in rodent species not previously known to carry SNV. These studies demonstrate for the first time the use of virus sequencing to trace the transmission of SNV and describe infection in novel rodent species.


Subject(s)
Disease Reservoirs/virology , Hantavirus Pulmonary Syndrome/transmission , Hantavirus Pulmonary Syndrome/veterinary , Hantavirus Pulmonary Syndrome/virology , Rodent Diseases/transmission , Rodent Diseases/virology , Rodentia/virology , Sin Nombre virus , Animals , Antibodies, Viral , Base Sequence , Female , Orthohantavirus/genetics , Hantavirus Infections/genetics , Hantavirus Infections/transmission , Hantavirus Infections/veterinary , Hantavirus Pulmonary Syndrome/epidemiology , Humans , Lung , Male , Mice , North America , Peromyscus/virology , Prevalence , RNA, Viral/genetics , Rodent Diseases/epidemiology , Sin Nombre virus/genetics , White People , Whole Genome Sequencing
8.
Viruses ; 13(8)2021 08 12.
Article in English | MEDLINE | ID: mdl-34452463

ABSTRACT

Pathogenic New World orthohantaviruses cause hantavirus cardiopulmonary syndrome (HCPS), a severe immunopathogenic disease in humans manifested by pulmonary edema and respiratory distress, with case fatality rates approaching 40%. High levels of inflammatory mediators are present in the lungs and systemic circulation of HCPS patients. Previous studies have provided insights into the pathophysiology of HCPS. However, the longitudinal correlations of innate and adaptive immune responses and disease outcomes remain unresolved. This study analyzed serial immune responses in 13 HCPS cases due to Sin Nombre orthohantavirus (SNV), with 11 severe cases requiring extracorporeal membrane oxygenation (ECMO) treatment and two mild cases. We measured viral load, levels of various cytokines, urokinase plasminogen activator (uPA), and plasminogen activator inhibitor-1 (PAI-1). We found significantly elevated levels of proinflammatory cytokines and PAI-1 in five end-stage cases. There was no difference between the expression of active uPA in survivors' and decedents' cases. However, total uPA in decedents' cases was significantly higher compared to survivors'. In some end-stage cases, uPA was refractory to PAI-1 inhibition as measured by zymography, where uPA and PAI-1 were strongly correlated to lymphocyte counts and IFN-γ. We also found bacterial co-infection influencing the etiology and outcome of immune response in two cases. Unsupervised Principal Component Analysis and hierarchical cluster analyses resolved separate waves of correlated immune mediators expressed in one case patient due to a sequential co-infection of bacteria and SNV. Overall, a robust proinflammatory immune response, characterized by an imbalance in T helper 17 (Th17) and regulatory T-cells (Treg) subsets, was correlated with dysregulated inflammation and mortality. Our sample size is small; however, the core differences correlated to survivors and end-stage HCPS are instructive.


Subject(s)
Cytokines/genetics , Cytokines/immunology , Hantavirus Infections/complications , Hantavirus Infections/immunology , Hantavirus Pulmonary Syndrome/immunology , Plasminogen/genetics , Sin Nombre virus/pathogenicity , Adolescent , Adult , Coinfection/complications , Coinfection/microbiology , Coinfection/virology , Cytokines/classification , Female , Hantavirus Infections/physiopathology , Hantavirus Pulmonary Syndrome/physiopathology , Humans , Inflammation/immunology , Inflammation/virology , Longitudinal Studies , Lung/immunology , Lung/pathology , Lung/virology , Male , Middle Aged , Patient Acuity , Plasminogen/analysis , Plasminogen/immunology , Retrospective Studies , Sin Nombre virus/immunology , Young Adult
9.
Elife ; 102021 07 06.
Article in English | MEDLINE | ID: mdl-34232859

ABSTRACT

Hantaviruses are RNA viruses with known epidemic threat and potential for emergence. Several rodent-borne hantaviruses cause zoonoses accompanied by severe illness and death. However, assessments of zoonotic risk and the development of countermeasures are challenged by our limited knowledge of the molecular mechanisms of hantavirus infection, including the identities of cell entry receptors and their roles in influencing viral host range and virulence. Despite the long-standing presumption that ß3/ß1-containing integrins are the major hantavirus entry receptors, rigorous genetic loss-of-function evidence supporting their requirement, and that of decay-accelerating factor (DAF), is lacking. Here, we used CRISPR/Cas9 engineering to knockout candidate hantavirus receptors, singly and in combination, in a human endothelial cell line that recapitulates the properties of primary microvascular endothelial cells, the major targets of viral infection in humans. The loss of ß3 integrin, ß1 integrin, and/or DAF had little or no effect on entry by a large panel of hantaviruses. By contrast, loss of protocadherin-1, a recently identified entry receptor for some hantaviruses, substantially reduced hantavirus entry and infection. We conclude that major host molecules necessary for endothelial cell entry by PCDH1-independent hantaviruses remain to be discovered.


Subject(s)
Endothelial Cells/virology , Orthohantavirus/physiology , Receptors, Cell Surface/metabolism , Viral Proteins/metabolism , Cell Line , Humans
10.
Evol Ecol ; 34(3): 339-359, 2020.
Article in English | MEDLINE | ID: mdl-32508375

ABSTRACT

Mutations can occur throughout the virus genome and may be beneficial, neutral or deleterious. We are interested in mutations that yield a C next to a G, producing CpG sites. CpG sites are rare in eukaryotic and viral genomes. For the eukaryotes, it is thought that CpG sites are rare because they are prone to mutation when methylated. In viruses, we know less about why CpG sites are rare. A previous study in HIV suggested that CpG-creating transition mutations are more costly than similar non-CpG-creating mutations. To determine if this is the case in other viruses, we analyzed the allele frequencies of CpG-creating and non-CpG-creating mutations across various strains, subtypes, and genes of viruses using existing data obtained from Genbank, HIV Databases, and Virus Pathogen Resource. Our results suggest that CpG sites are indeed costly for most viruses. By understanding the cost of CpG sites, we can obtain further insights into the evolution and adaptation of viruses.

SELECTION OF CITATIONS
SEARCH DETAIL
...