ArticlePDF Available

Immune protection is dependent on the gut microbiome in a lethal mouse gammaherpesviral infection

Authors:

Abstract and Figures

Immunopathogenesis in systemic viral infections can induce a septic state with leaky capillary syndrome, disseminated coagulopathy, and high mortality with limited treatment options. Murine gammaherpesvirus-68 (MHV-68) intraperitoneal infection is a gammaherpesvirus model for producing severe vasculitis, colitis and lethal hemorrhagic pneumonia in interferon gamma receptor-deficient (IFNγR−/−) mice. In prior work, treatment with myxomavirus-derived Serp-1 or a derivative peptide S-7 (G305TTASSDTAITLIPR319) induced immune protection, reduced disease severity and improved survival after MHV-68 infection. Here, we investigate the gut bacterial microbiome in MHV-68 infection. Antibiotic suppression markedly accelerated MHV-68 pathology causing pulmonary consolidation and hemorrhage, increased mortality and specific modification of gut microbiota. Serp-1 and S-7 reduced pulmonary pathology and detectable MHV-68 with increased CD3 and CD8 cells. Treatment efficacy was lost after antibiotic treatments with associated specific changes in the gut bacterial microbiota. In summary, transkingdom host-virus-microbiome interactions in gammaherpesvirus infection influences gammaherpesviral infection severity and reduces immune modulating therapeutic efficacy.
This content is subject to copyright. Terms and conditions apply.
1
SCIENTIFIC REPORTS | (2020) 10:2371 | https://doi.org/10.1038/s41598-020-59269-9
www.nature.com/scientificreports
Immune protection is dependent
on the gut microbiome in a lethal
mouse gammaherpesviral infection
Jordan R. Yaron
1,2,13, Sriram Ambadapadi 1,2,13, Liqiang Zhang 1,2, Ramani N. Chavan3,
Scott A. Tibbetts4, Shahar Keinan 5, Arvind Varsani
3,6,7,8, Juan Maldonado
3,9,
Simona Kraberger2,3, Amanda M. Tafoya1,2, Whitney L. Bullard4, Jacquelyn Kilbourne1,2,3,
Alison Stern-Harbutte4, Rosa Krajmalnik-Brown3,10,11, Barbara H. Munk1, Erling O. Koppang12,
Efrem S. Lim
3* & Alexandra R. Lucas1,2,4*
Immunopathogenesis in systemic viral infections can induce a septic state with leaky capillary
syndrome, disseminated coagulopathy, and high mortality with limited treatment options. Murine
gammaherpesvirus-68 (MHV-68) intraperitoneal infection is a gammaherpesvirus model for producing
severe vasculitis, colitis and lethal hemorrhagic pneumonia in interferon gamma receptor-decient
(IFNγR/) mice. In prior work, treatment with myxomavirus-derived Serp-1 or a derivative peptide
S-7 (G305TTASSDTAITLIPR319) induced immune protection, reduced disease severity and improved
survival after MHV-68 infection. Here, we investigate the gut bacterial microbiome in MHV-68 infection.
Antibiotic suppression markedly accelerated MHV-68 pathology causing pulmonary consolidation and
hemorrhage, increased mortality and specic modication of gut microbiota. Serp-1 and S-7 reduced
pulmonary pathology and detectable MHV-68 with increased CD3 and CD8 cells. Treatment ecacy
was lost after antibiotic treatments with associated specic changes in the gut bacterial microbiota. In
summary, transkingdom host-virus-microbiome interactions in gammaherpesvirus infection inuences
gammaherpesviral infection severity and reduces immune modulating therapeutic ecacy.
Viral infections induce potent immune responses, an immunopathogenesis that can lead to severe complications
with sepsis or leaky capillary syndromes and very high mortality and limited eective treatments, a true unmet
clinical need. Sepsis has an associated risk of disseminated intravascular coagulation (DIC) with thrombosis,
hemorrhage and shock13. One such group of viruses with known severe complications are the gammaherpesvi-
ruses (GHV). e murine gammaherpesvirus-68 (MHV-68) is a widely used, well-controlled laboratory model
of GHV host-pathogen interaction with genetic similarity to the human viruses Epstein-Barr virus (EBV) and
Kaposi’s sarcoma-associated herpesvirus (KSHV)4.
Inammatory vasculitic syndromes (IVS) are a group of rare, heterogeneous and devastating inammatory
conditions of the body’s extensive system of blood vessels with increased morbidity, including sudden loss of
vision, aneurysm, aortic arch syndrome, stroke, and associated increases in mortality57. e etiology of many
systemic vasculitides is currently unknown, with proposed mechanisms ranging from induction by fungal spores
1Center for Personalized Diagnostics, The Biodesign Institute, Arizona State University, Tempe, Arizona, USA.
2Center for Immunotherapy, Vaccines and Virotherapy, The Biodesign Institute, Arizona State University, Tempe,
Arizona, USA. 3Center for Fundamental and Applied Microbiomics, The Biodesign Institute, Arizona State University,
Tempe, Arizona, USA. 4Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida,
Gainesville, Florida, USA. 5Cloud Pharmaceuticals, Research Triangle Park (RTP), North Carolina, USA. 6School of
Life Sciences, Arizona State University, Tempe, Arizona, USA. 7Center of Evolution and Medicine Arizona State
University, Tempe, Arizona, USA. 8Structural Biology Research Unit, Department of Integrative Biomedical Sciences,
University of Cape Town, Rondebosch, Cape Town, South Africa. 9KED Genomics Core, Arizona State University,
Tempe, Arizona, USA. 10Swette Center for Environmental Biotechnology, The Biodesign Institute, Arizona State
University, Tempe, Arizona, USA. 11School of Sustainable Engineering and the Built Environment, Arizona State
University, Tempe, Arizona, USA. 12Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary
Medicine, Norwegian University of Life Sciences, Oslo, Norway. 13These authors contributed equally: Jordan R. Yaron
and Sriram Ambadapadi. *email: efrem.lim@asu.edu; arlucas5@asu.edu
OPEN
Content courtesy of Springer Nature, terms of use apply. Rights reserved
2
SCIENTIFIC REPORTS | (2020) 10:2371 | https://doi.org/10.1038/s41598-020-59269-9
www.nature.com/scientificreports
www.nature.com/scientificreports/
to herpesviruses infections (e.g., zoster)7,8. A lethal IVS large vessel arteritis model which closely mimics human
Giant Cell Arteritis, Kawasaki’s disease and Takayasu’s arteritis can be induced by high dose intraperitoneal
MHV-68 infection of interferon gamma receptor knockout (IFNγR/) mice9. Many infected mice display exten-
sive pulmonary hemorrhage and consolidation, mimicking DIC in viral sepsis, which has a very high attendant
mortality10. A signicant number of mice also have marked colon dilatation reminiscent of toxic megacolon11, in
addition to aggressive inammatory cell invasion with hemorrhagic lung consolidation. Herpesvirus infections
have been associated with inammatory bowel diseases1214 and gastrointestinal involvement in systemic vascu-
litides has been reported by other groups15,16. As other gastrointestinal inammatory conditions are associated
with, or driven by, microbiome changes, the gut microbiome may have a role in the pathogenesis of GHV and
specically MHV-68 infections.
We have previously reported that immune modulating proteins from myxomavirus (MYXV) provide a new
class of anti-inammatory treatments when delivered as recombinant protein or by ectopic expression aer
Adeno-associated virus (AAV) delivery1720. MYXV is the causative agent of lethal myxomatosis in the European
rabbit (Oryctolagus cuniculus), expressing highly potent immune evasion proteins that act as virulence factors in
MYXV infections. MYXV is not a pathogen for mice or humans. When puried as isolated recombinant proteins,
these immune modulating biologics can modify disease progression in a wide range of inammatory diseases
in preclinical animals and man21. In prior work, the MYXV-derived serine protease inhibitor (serpin) Serp-1
and a Serp-1 reactive center loop (RCL) peptide S-7 (G305TTASSDTAITLIPR319) signicantly improved survival,
reducing pulmonary and aortic inammation as well as hemorrhagic lung consolidation aer MHV-68 infec-
tion10,22. Serp-1 targets the coagulation pathways, both thrombolytic and thrombotic, and has proven, highly
eective anti-inammatory functions23,24. Antibiotic treatment of IFNγR/ mice prior to MHV-68 infection
abrogated the ecacy of S-7 treatment for lethal MHV-68 infections, leading to signicantly reduced survival.
erapeutically modied S-7 peptides (MPS7–8 and MPS7–9), designed based upon the serpin crystal structure,
maintained ecacy in this model with improved survival aer MHV-68 infection25. e MPS7 peptides have
predicted increased hydrogen bonds when compared to the native S-7 peptide25.
e pathophysiologic role of the gut bacterial microbiome in gammaherpesviral infections and on immune
modulating treatments has not previously been investigated. We report here a systematic examination of the role
of the gut bacterial microbiome in MHV-68 infection and on Serp-1 and Serp-1-derived S-7 peptide treatment
in MHV-68 infections.
Results
Serp-1 protein and S-7 peptide treatments improve survival after MHV-68 infection in
IFNγR/ mice. MHV-68 infections establish latency with chronic infection in wildtype mice26, causing
only u-like symptoms. Conversely, intraperitoneal infection of mice with underlying immune deciency, as
for IFNγR/ mice, results in a lethal large vessel vasculitis, colitis and hemorrhagic pneumonia with early mor-
tality9,27. Intranasal infection, an alternative model for MHV-68, is not a model for severe vasculitic syndromes.
Our group and others have used this intraperitoneal infection model of vasculitis to study the pathogenesis of
gammaherpesvirus host-pathogen interactions, as well as to investigate the potential for anti-inammatory treat-
ment of associated inammatory vasculitic syndromes10,11,22,25,28,29. Intraperitoneal infection of IFNγR/ mice
with 12.5 × 106 pfu was performed with follow-up for 150 days to determine survival rates. MHV-68 infection
with administration of control saline treatment alone for 30 days was lethal, with a median survival of 41 days.
Serp-1 (100 ng/g) or S-7 (100 ng/g) intraperitoneal (IP) injections for 30 days improved survival to 60% at 150
days (p = 0.0022 and p = 0.0218, respectively) (Fig.1B). us, Serp-1 and S-7 protect IFNγR/ mice aer lethal
MHV-68 infection10,22.
Loss of gut bacteria accelerates MHV-68 induced disease. We hypothesized that the gut bacterial
microbiome may play a role in the host-pathogen interactions of MHV-68 induced disease through transking-
dom interactions. We completed a systematic analysis of the eect of microbiome depletion on MHV-68 induced
disease progression (Fig.2A). Mice were maintained on medicated water containing a broad-spectrum cocktail
Figure 1. Serp-1 and Serp-1 RCL-derived peptide S-7 improve survival in lethal MHV-68 infection. (A)
Structure of Serp-1 with A-beta sheet (blue) and reactive center loop (RCL; red) highlighted. RCL-derived
peptide S-7 illustrated in green. (B) Kaplan-Meier curve depicting survival of IFNγR/ mice infected with
MHV-68 and treated with saline (le; N = 12 and 5), Serp-1 (middle; N = 5 and 5), or S-7 (right; N = 5 and 5)
without or with antibiotics.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
3
SCIENTIFIC REPORTS | (2020) 10:2371 | https://doi.org/10.1038/s41598-020-59269-9
www.nature.com/scientificreports
www.nature.com/scientificreports/
of four antibiotics: bacitracin, gentamicin, streptomycin and ciprooxacin (Table1) in order to suppress gut bac-
terial populations. During this time the density of the gut bacteria (as measured by colony forming units [CFU]
per gram fecal pellet) was markedly reduced, validating a decrease in bacterial load (Fig.2B). Aer ten days, mice
were returned to normal drinking water for 24 hours, during which time the antibiotic treatment maintained sup-
pression of the microbiome (Fig.2B). Mice were subsequently infected by IP injection of MHV-68 and injected
with control saline daily for 30 days and survival assessed for up to 150 days(Fig. 1B).
Suppression of gut bacteria by antibiotic treatment markedly accelerated the course of MHV-68 infection.
Saline-treated mice exhibited a median survival of 41 days, while antibiotic-treated mice had a median survival of
19 days (p = 0.0411; Fig.1B). 16 S rRNA gene amplicon sequencing was subsequently performed on DNA extracts
from the large intestines of individual mice at 3 days post- MHV-68 infection in order to assess early changes in
the gut microbiota. We detected a distinct dysbiosis in mice pre-treated with antibiotics prior to MHV-68 infec-
tion versus mice with a stable bacterial microbiome prior to infection (Fig.2C). is dysbiosis was associated
with a non-statistically signicant trend towards increased diversity in amplicon sequence variant (ASV) richness
(Fig.2D; p = 0.329). Sub-population analysis revealed signicant changes in candidate ASVs in MHV-68 infected
mice that were associated with antibiotic treatment (Fig.2E, top panels; increased abundance aer antibiotics) or
with no antibiotic treatment (Fig.2E, bottom panels; decreased abundance aer antibiotics). ese results suggest
that the gut bacterial microbiome is a signicant determinant of MHV-68 induced disease progression.
Serp-1 and S-7 peptide treatment ecacy is dependent on the gut bacterial microbiota. We
next investigated the potential for dependence of Serp-1 and S-7 therapeutic ecacy on the gut bacterial microbi-
ome in MHV-68 infected IFNγR/ mice. Mice were treated with medicated drinking water (Table1) for 10 days,
Figure 2. Antibiotic treatment accelerates MHV-68 lethality and alters the gut bacterial microbiome. (A)
Overview of experimental design. Mice are treated with a broad-spectrum antibiotic cocktail in their drinking
water for 10 days, placed on normal water for 1 day, infected with MHV-68 with or without treatments with
follow-up for survival. (B) Antibiotic-medicated water completely ablates bacterial microbiome contents during
the course of the 10-day pre-treatment, which recovers by three days aer removal of antibiotics. (C) 16 S
microbiome relative abundance (genus level) and (D) Bacterial alpha diversity (Shannon Index) is shown for
saline-treated mice with and without antibiotics. (E) Heatmap showing ASV abundance in mice treated with
saline in the presence or absence of antibiotic pre-treatment.
Antibiotic Dose Target class Mechanism of action
Bacitracin 1 g/L Gram-positive Cell wall synthesis inhibition88
Gentamicin 0.5 g/L Gram-negative and Gram-positive Ribosome inhibition89
Streptomycin 2 g/L Gram-negative and Gram-positive Ribosome inhibition90
Ciprooxacin 0.125 g/L Gram-negative and Gram-positive DNA replication inhibition91
Table 1. Antibiotic cocktail components.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
4
SCIENTIFIC REPORTS | (2020) 10:2371 | https://doi.org/10.1038/s41598-020-59269-9
www.nature.com/scientificreports
www.nature.com/scientificreports/
placed on normal water for 24 hours, then infected with MHV-68 and concurrently treated for 30 days with either
Serp-1 or S-7 by IP injection. Mice were followed for survival or 3-day follow-up (Fig.2A).
As previously reported, Serp-1 and S-7 peptide improved survival with 60% of mice surviving to 150 days
aer MHV-68 infection. In contrast, antibiotic-treated mice had accelerated disease, with a median survival of 19
(Serp-1; p = 0.0025; Fig.1B) and 18 days (S-7; p = 0.0339; Fig.1B). Serp-1 or S-7 ecacy aer antibiotic treatment
is not signicantly dierent from saline treatment aer antibiotics (Serp-1 + Abx vs Saline + Abx, p = 0.2012;
S-7 + Abx vs Saline + Abx, p = 0.9164). us, the ecacy of Serp-1 or S-7 treatment and protection in lethal
MHV-68 infections was dependent on the gut bacterial microbiome.
Microbiome analysis identifies candidate taxa associated with Serp-1 and S-7 treat-
ments. Based on the ndings that (1) antibiotic-treated mice accelerated and exacerbated MHV-68 disease,
reducing survival time and increasing mortality, and that (2) immune protection conferred by Serp-1 and S-7 was
highly sensitive to antibiotic treatment, we proled the gut bacterial microbiota of mice treated with Serp-1 or
S-7. 16S rRNA gene amplicon sequencing was performed on DNA extracts derived from the large intestines of
individual mice 3 days post-infection with MHV-68 and aer treatment with Serp-1 or S-7 (Fig.3). Serp-1 and
S-7 treated mice, in the absence of antibiotics, had a higher diversity of microbiota (median = 1.270, p = 0.0079;
median = 0.862, p = 0.1508respectively) than saline treated mice (median = 0.385) (Fig.3B). Principal coordi-
nate analyses indicate that although the gut bacterial microbiomes of Serp-1 and S-7 treated mice diered from
saline treated mice, there was variability within Serp-1 and S-7 treated mice (Fig.3C, Supplementary Fig.1B).
is nding was consistent with the phenotypic variability observed in vivo. Specically, there was partial protec-
tion by Serp-1 and S-7 treatment with 60% survival (Fig.1B).
We hypothesized that responsiveness to Serp-1 and S-7 treatment may be driven by the presence of “pro-
tective” ASVs (dierentially associated with Serp-1 and/or S-7 treatment) or the absence of “potentiator” ASVs
(dierentially associated with saline treatment). erefore, to identify potential microbe(s) responsible for the
microbiome-mediated protection by Serp-1 and S-7, we analyzed the gut bacterial microbiota for discriminant
ASVs that were dierentially represented in Serp-1 and/or S-7 treated mice compared to saline treated mice, in
the absence of antibiotics (Supplementary Fig.1C–E). Of the 15 ASVs identied as dierentially associated with
Serp-1/peptide treatments, ASV4, was consistently associated with both Serp-1 and S-7 treatment (Fig.3D). ASV4
was most closely-related to the sequence of Bacillus massiliogorillae30. Conversely, ASV1 (most closely-related to
Frondihabitans peucedani31) and ASV123 (most closely-related to Enterococcus saigonensis32) were identied from
8 ASV candidates as being associated with saline treatment when compared to both Serp-1 or S-7 treatments.
Based upon the fact that Serp-1 and S-7 protected mice in an antibiotic-dependent manner, we reasoned that
the relative abundance of the discriminant ASVs would be altered in mice that were pre-treated with antibiotics.
As predicted, the relative abundance of Serp-1- or S-7-associated ASVs was decreased in antibiotic pre-treated
mice, albeit to a more moderate extent in S-7 (Fig.3E,F, compare “No Abx” to “ + Abx”). ese results suggest
that the interplay between with bacterial microbiota like ASV4, that are associated with a protective phenotype,
and microbiota such as ASV1 and ASV123 that are associated with a patho-exacerbative phenotype, can inuence
the outcome of immune modulating treatments. In summary, responsiveness to immune modulating therapy in
MHV-68 induced disease is associated with specic alterations in the gut bacterial microbiota.
Antibiotic-dependent exacerbation of early lung pathology of MHV-68 induced disease. MHV-
68 persistent infection33 leads to severe hemorrhagic and consolidating pulmonary pathology3436. Hence, we
investigated whether the pulmonary pathology induced by MHV-68 infection reected the antibiotic-dependent
exacerbation we observed in survival analysis. We performed quantitative morphometric analyses of lungs at an
early, 3-day follow-up aer infection. In the absence of antibiotics, considerable early pulmonary consolidation
and inammation was observed in MHV-68-infected lung tissue in IFNγR/ mice treated with control saline
alone. is severe pulmonary pathology was considerably reduced by Serp-1 or S-7 peptide treatment (Fig.4A)10.
Early lung pathology was markedly worse aer antibiotic treatment in all conditions (saline, Serp-1 or S-7), with
most animals in the saline group also showing hemorrhage in aected regions.
Previous reports have demonstrated that thickening of the alveolar wall/septa and reduction of alveolar lumen
area are reliable indicators of pulmonary inammation in acute laboratory models3739. In this study, quantitative
morphometry of the lungs revealed a signicant reduction in alveolar wall thickness in MHV-68-infected mice
treated with Serp-1 or S-7 versus saline-treated controls that was lost aer treatment with antibiotics (Fig.4B).
Without antibiotic pre-treatment and when compared to saline-treated controls, alveolar lumen area was signi-
cantly increased in mice treated with S-7 (p = 0.0067) with a trend toward an increase in mice treated with Serp-1.
Antibiotic treatment nullied any level of protection by treatment with S-7 (Fig.4C). No diagnostic pathology
nor signicant changes in inammatory cell inltrates on histological analysis was noted in the gut or aorta at
this early, 3-day follow-up time (not shown). ese results indicate that immune protection against pulmonary
inammation and early stage hemorrhage promoted by Serp-1 and S-7 treatment are microbiome dependent.
Serp-1 and S-7 promote an increased gut microbiome-dependent CD3+ occupancy in the lungs.
T-cell responses play a crucial role in limiting active infection and have been reported as central mediators for
managing chronic MHV-68 infection4042. Hence, we examined early, acute phase CD3+ T-cell recruitment to the
lungs in MHV-68 infections. Antibiotic treatment signicantly reduced CD3+ cells in the lungs of saline-treated
mice (Fig.4E, p = 0.0155). Further, S-7 signicantly increased, and Serp-1 trended towards increasing, the detect-
able CD3+ cells in the lungs of infected mice (Fig.4D,E), indicating that the serpin-mediated protection against
MHV-68 induced disease is closely associated with increased pulmonary T-cell activity. Further staining indi-
cates no appreciable eect of Serp-1 and S-7 on pulmonary CD4+ cells (Fig.4F), while CD8+ staining showed
signicant increases with S-7 (p = 0.0312) and a strong trend towards increase with Serp-1 (p = 0.1734) treatment
Content courtesy of Springer Nature, terms of use apply. Rights reserved
5
SCIENTIFIC REPORTS | (2020) 10:2371 | https://doi.org/10.1038/s41598-020-59269-9
www.nature.com/scientificreports
www.nature.com/scientificreports/
(Fig.4G). Remarkably, antibiotic treatment that depleted the gut microbiome led to a loss of this observed CD3
inltration-promoting eects and a loss of CD8 bias of both Serp-1 and S-7 (Fig.4E-G). Taken together, these
results suggest protection from MHV-68 induced disease via CD3 recruitment and surveillance in the lungs, with
a CD8 bias, enhanced by Serp-1- or S-7, proceeds via a microbiome-dependent mechanism.
Figure 3. 16 S microbiome analysis. (A) 16S microbiome relative abundance (genus level) is shown. (B)
Bacterial alpha diversity (Shannon Index) is shown. Statistical signicance was assessed by Mann-Whitney
test. (C) Principal coordinate analysis (PCoA) plot of weighted UniFrac distances. (D) Heatmap showing ASV
abundance in mice treated with saline, Serp-1 or S7, in the absence of antibiotics. ASV shown were identied by
dierential analyses to be associated with Serp-1 or S7 (top section), or saline (bottom section). (E) Heatmaps of
dierential ASV from (D) are compared in the absence or presence of antibiotics for Serp-1 treated mice (le)
or S-7 treated mice (right). (F) Relative abundance of ASV4 (identied as dierentially associated with Serp-1
and S-7 treatment), and ASV1 and 123 (identied as dierentially associated with saline) in individual mouse
microbiomes. ASV taxonomic classication of the most closely related bacterial taxa are shown. Saline + Abx,
N = 6; Saline No Abx, N = 6; Serp-1 + Abx, N = 6; Serp-1 No Abx, N = 5; S7 + Abx, N = 6; S7 No Abx, N = 5.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
6
SCIENTIFIC REPORTS | (2020) 10:2371 | https://doi.org/10.1038/s41598-020-59269-9
www.nature.com/scientificreports
www.nature.com/scientificreports/
Serp-1 and S-7 reduce pulmonary MHV-68 levels in a gut bacterial microbiome-dependent
manner. Because T-cells are crucial for control of MHV-68 infection, we hypothesized that the promotion
of CD3 T-cell inltration would lead to reduced viral levels in the lungs. Antibiotic treatment did not change the
amount of MHV-68 antigen staining in the lungs of mice treated with saline control alone (Fig.5A,B). However,
Serp-1 and S-7 signicantly reduced MHV-68 antigen levels in the lungs in mice without antibiotic treatment
(Fig.5B; Serp-1, p = 0.0009; S-7, p = 0.0003). ese decreases in detectable MHV68 were partially reversed aer
antibiotic pre-treatment in MHV-68-infected mice (Figs.5 and S3). us, Serp-1 and S-7 initiate a gut bacterial
microbiome-dependent reduction in MHV-68 levels in the lungs.
Discussion
Compelling evidence has established an important role for the gut microbiome in immune responsiveness4345 as
well as the pathogenesis of various diseases, including those with primary involvement outside of the gut, such
as respiratory46, hepatic47, renal48, neurologic49, autoimmune50, rheumatoid51 and cardiovascular5254 conditions.
Some research groups have demonstrated that targeted manipulation of the gut microbiome to correct dysbiosis,
as for fecal transplants, may aid in treatment of certain diseases. For example, a small, randomized clinical trial
(NCT02636647) found improved outcomes in recurrent hepatic encephalopathy patients treated by fecal micro-
biota transplant from a single, rationally selected donor enriched with Lachnospiraceae and Ruminococcaceae
(taxa determined to be depleted in hepatic encephalopathy patients)55. A second trial (NTR1776) found that
fecal microbiota transfer from lean donors to obese donors partially reversed symptoms of metabolic syn-
drome, including improvements in insulin resistance56. Aside from correcting dysbiosis, recent evidence sup-
ports the hypothesis that microbiome composition also plays a signicant role in determining the ecacy of
some treatments. It was recently found that low gut microbiome levels of Akkermansia muciniphila caused
non-responsiveness in patients receiving PD-1 blockade immunotherapy for epithelial tumors5759, reinforcing
the importance of the microbiome in treatment ecacy, as highlighted by a critical earlier study that identied a
positive correlation between Bacteroides fragilis and anti-CTLA4 therapy ecacy for melanoma60. Alternatively,
some treatments may inadvertently alter the gut microbiome and predispose to increased pathogenic infec-
tions. For example, it was recently demonstrated that proton pump inhibitors signicantly reduced microbial
diversity with changes in more than 20% of bacterial taxa, leading to increases in Enterococcus, Streptococcus,
Staphylococcus and the potentially pathogenic Escherichia coli species61. us, an understanding of how the gut
microbiome inuences, and is inuenced by, new therapeutic treatments is crucial.
Trans-kingdom interactions may also play a crucial role in dictating disease pathogenesis in mammalian hosts.
For viruses which require enteric bacteria for replication, antibiotics may reduce viral load. Numerous studies
have demonstrated this viral-bacterial interaction in laboratory systems, such as murine and human norovirus
Figure 4. Microbiome-dependent Serp-1 and S-7 ecacy is associated with pulmonary inammation and
increased occupancy of CD3+ and CD8+ cells (A) Representative H&E sections of mouse lungs at 3 days
follow-up aer MHV-68 infection without antibiotic pre-treatment (top row) or aer antibiotic pre-treatment
(bottom row) and with saline, Serp-1 or S-7 treatment. Scale bar is 100 µm. (B) Alveolar wall thickness and (C)
Alveolar lumen area of mouse lungs at 3 days follow-up as in (A). (D) Representative CD3 IHC micrographs
of mouse lungs at 3 days follow-up aer MHV-68 infection without antibiotic pre-treatment (top row) or
aer antibiotic pre-treatment (bottom row) and with saline, Serp-1 or S-7 treatment. Scale bar is 20 µm. (E)
Quantication of CD3+, CD4+ and CD8+ cells per 40× eld from 3–6 elds per mouse. Statistics in panels
B, C and E performed by Two-Way ANOVA with Fisher’s LSD post-hoc analysis. *p < 0.05, **p < 0.01,
***p < 0.001; n.s. is not signicant. N = 3 for all conditions.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
7
SCIENTIFIC REPORTS | (2020) 10:2371 | https://doi.org/10.1038/s41598-020-59269-9
www.nature.com/scientificreports
www.nature.com/scientificreports/
infection62,63. For other viruses, however, loss of the bacterial microbiome may lead to worsening clinical disease.
A recent study by ackray et al. reports that oral antibiotic (ampicillin) treatment exacerbates disease severity in
avivirus infections by modulating avivirus-specic CD8 responses64, suggesting a microbiome, immune system
and virus interaction. ese disparate observations, which depend substantially on (a) the virus being studied, (b)
the host system and (c) the antibiotic regimen, highlight how the virome is still a poorly understood, and conse-
quently under-appreciated, component of health and disease65.
We recently reported a severe gastrointestinal pathology in a signicant fraction of IFNγR/ mice intraperi-
toneally infected with MHV-6811, a model for systemic arteritis and hemorrhagic pneumonia9,10. In this same
model, we also previously reported therapeutic ecacy with the MYXV-derived serine protease inhibitor Serp-
110 as well as peptides derived from the Serp-1 reactive center loop22,25. On this basis, we undertook a systematic
analysis of the eect of Serp-1 and the RCL-derived Serp-1 peptide S-7 on MHV-68 induced disease.
A broad-spectrum antibiotic cocktail was used to suppress the gut microbiome prior to MHV-68 infection.
We found that suppression of the gut microbiota prior to MHV-68 infection accelerated disease, with increased
lethality and earlier death occurring at approximately twice the rate of untreated mice (Fig.2C). is acceleration
of MHV-68 disease was accompanied by a dramatic post-antibiotic dysbiosis and a suite of candidate ASVs with
increased or decreased relative abundance associated with worsened disease outcomes.
Furthermore, we demonstrate here that bacterial microbiome interactions were required for a protective
immune response induced by Serp-1 or S-710. Microbiome ablation completely abolished treatment ecacy and
improved survival (Fig.1B). Probing further, we found that Serp-1 and S-7 facilitated protection against lung
inammation and consolidation associated with an increase in CD3+ cell invasion (with a CD8+ bias) and a
reduction of MHV-68 staining in the lungs (Figs.4 and 5). is nding is in agreement with previous reports that
T-cell surveillance (eected by CD8+ cells) is critical for limiting acute GHV infection and that a progressive
loss of T-cells is associated with worsened GHV disease4042. e loss of Serp-1-based treatment ecacy was
not expected to be due to direct interactions between the antibiotics and immune modulating treatments for a
number of reasons: (1) the antibiotics used in this study have a relatively short half-life (1–7 hours) and previ-
ous studies report negligible concentrations 24 hours aer dosing6669 – a timeline we incorporated in this study
design; (2) we observed a rapid recovery of microbial burden within three days (Fig.2B); (3) the antibiotics in
our cocktail are not known to suppress immune responses on their own, and in some reports are observed to
enhance T-cell activity70; (4) it is not expected that the loss of activity is due to direct interactions with the anti-
biotics because in vitro inhibition of uPA activity by Serp-1 is unaected by the presence of the same antibiotic
cocktail (Fig.S2). A recent report by Yang et al. indicated that in the short-term, antibiotics can induce metabolite
changes which have systemic, suppressant eects on immune function in the peritoneal space by altering respira-
tory activity44. Yang et al., however, utilized an E. coli infection and the antibiotic, ciprooxacin, was given within
4 hours of infection and maintained during the course of the infection. Here, we utilize a gammaherpesvirus
infection with longer pathogenic kinetics and remove antibiotics 24 hours prior to infection, wherein any trace of
ciprooxacin would be undetectable66, and we are not aware of any other reports suggesting immune suppressant
eects of the other antibiotics in our cocktail. Taken together, these data provide the rst evidence that microbi-
ome interactions are essential for a protective immune response, induced here by Serp-1 and S-7 treatment. We
have previously found Serp-1 to function through the urokinase-type plasminogen activator receptor (uPAR) in
systems involving immune cell inltration (e.g., wound healing71, chemokine-induced ascites72). Levels of uPAR
have been found to increase substantially in incidences of infection and sepsis73. us, future work will investigate
the possibility that the microbiome is modulating levels of binding partners essential for Serp-1 and S-7 function
in MHV-68 infection.
We propose here that transkingdom interactions with the gut bacterial microbiome are required for host
innate immunity to mount a protective response against lethal gammaherpesviral disease (Fig.6). When
Figure 5. Serp-1 and S-7 reduce MHV-68 presence in a gut bacterial microbiome-dependent manner (A)
Representative IHC images of 100 × elds depicting MHV-68 antigen staining in the lungs of mice at 3 days
follow-up aer MHV-68 infection without antibiotic pre-treatment (top row) or aer antibiotic pre-treatment
(bottom row) and with saline, Serp-1 or S-7 treatment. (B) Quantication of percent of MHV-68 positively
stained cells per 100 × eld from 3 elds per mouse. N = 3 for all conditions.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
8
SCIENTIFIC REPORTS | (2020) 10:2371 | https://doi.org/10.1038/s41598-020-59269-9
www.nature.com/scientificreports
www.nature.com/scientificreports/
MHV-68 infection causes viral pulmonary inammation and sepsis, as induced in the MHV-68 infection model,
host innate immune responses mediated by gut bacterial microbiota interactions leads to the recruitment of
immune cells (e.g. T-cells) to the lungs. However, antibiotic ablation of the gut bacterial microbiota alters patho-
logic outcome by preventing eective immune response responses leading to acceleration of MHV-68 inamma-
tory pathogenesis. Immunomodulatory treatments (like Serp-1 and S-7) can induce a stronger innate immune
response (such as T-cell recruitment) through (other) bacterial microbiota interactions. us, outcomes of GHV
infection are determined by the interplay of host innate immunity and microbiota interactions.
Interestingly, the gut microbiota of all Serp-1 and S-7 treated mice did not clearly cluster separately from
saline treated mice. us, the dynamics of selected increased or decreased microbiome ASVs is associated with
worsened pathology or alternatively protective immune modulation. Given that not all mice were fully protected
by Serpin treatment, we speculate that the gut microbiota heterogeneity of Serp-1- or peptide-treated mice may
reect the true variability in immunological outcomes (i.e. 60% protection). We found specic ASVs that were
either increased in response to Serp-1/peptide treatment (e.g. ASV4), or decreased when compared to saline
treatment (e.g. ASV1 and ASV123).
Although the current study design could not assess the predictive value of these discriminant ASVs on indi-
vidual mice survival outcomes, future studies will need to be performed in order to stratify the role of dierent
microbiome sub-populations by specic (i.e., single) antibiotic treatment as well as dened microbial recon-
stitution (e.g., with the Altered Shaedler Flora community or similar)74. Taken together, these ndings suggest
that the responsiveness to immune modulations in MHV-68 disease involves a combination of interactions with
protective microbiota and patho-exacerbative microbiota. Further work will be needed to establish a direct role
for bacteria related to these select ASVs to prove cause-and-eect relationships and to dene a precise mechanism
of bacterial microbiome inuence on immune responses in GHV disease.
With this study, we report that the pathogenicity of MHV-68 infection and eective treatment with the
immune modulating serpin, Serp-1, and serpin peptide, S-7, is substantially accelerated by suppression of the
gut bacterial microbiome with antibiotics. MHV-68 is a model virus for studying GHV pathogenesis and severe
viral sepsis4,75, therefore this nding may have implications for the pathogenesis and treatments of other GHV,
such as KSHV and EBV, where adverse responses to antibiotics are clinically recognized76. We further report here
the modulation of gut microbiome composition by an immune modulating protein and peptide. In conclusion,
(1) microbiome changes may have signicant impact on therapeutic immune modulating proteins77,78 and pep-
tides79,80, and (2), antibiotic treatment may increase severity of GHV infections.
Methods
Ethics statement. All animal studies conform to local and national guidelines for animal care and exper-
imentation. Protocols were approved by the local Institutional Animal Care and Use Committee (IACUC) of
University of Florida (#201604234_01).
Figure 6. Proposed model (Le) In uninterrupted lethal MHV-68 infection of IFNγR/ mice, the gut
microbiome and immune response interact to mount an ultimately insucient response of cells such as T-cells
to aected tissues (e.g., the lungs), leading to severe disease and death. Antibiotics suppresses the immune
stimulatory eects of the bacterial microbiome, reducing further the immune response and worsening
the disease. (Right) In immune modulator-mediated protection by treatments such as Serp-1 or S-7, the
interactions leading to a suciently mounted immune response are enhanced, promoting an increased T-cell
inltration to aected tissues, reducing disease pathology and leading to survival.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
9
SCIENTIFIC REPORTS | (2020) 10:2371 | https://doi.org/10.1038/s41598-020-59269-9
www.nature.com/scientificreports
www.nature.com/scientificreports/
Animals. IFNγR/ mice (B6.129S7-Ifngr1tm1Agt/J) were purchased from the Jackson Laboratory (Bar Harbor,
ME, USA). Animals were housed in barrier conditions at the University of Florida Animal Care Services vivar-
ium and bred under specic pathogen-free conditions. Mice were weaned at 3 weeks, maintained on a 12-hour
light-dark cycle and were fed water and standard rodent chow ad libitum.
Antibiotic treatment. At 4 weeks of age, IFNγR/ mouse cohorts were transferred from the ABSL1 colony
to a separate, ABSL2 colony. Gut microbiome suppression was achieved by replacing standard drinking water
with autoclaved reverse osmosis water (obtained from the same animal care facility) containing an antibiotic
cocktail (Table1) composed of Streptomycin (2 g/L), Gentamicin (0.5 g/L), Bacitracin (1 g/L) and Ciprooxacin
(0.125 g/L) for 10 days. One day (24 hours) prior to infection, medicated water was replaced with standard animal
care facility water, which was maintained for the remainder of the experiment.
MHV-68 infection. On day 11 (aer 10 days of antibiotics) mice (5 weeks old) were infected with MHV-68 at
a dose of 12.5 × 106 PFU in 0.1 mL DMEM by intraperitoneal (IP) injection as previously described22,25,27,29. Mice
were returned to the colony and monitored for signs of distress for the duration of the experiment. Mice were
either followed for 150 days to determine survival or euthanized at 3 days post-infection and organs harvested
into formalin for histology or RNAlater (ermo Scientic, USA) for microbiome analyses. Details on the num-
bers of MHV-68-infected mice in this study are detailed in Table2.
Gut bacterial microbiome sample processing. Large intestine samples preserved in RNAlater were
processed for total genomic DNA isolation using the ZymoBiomics® miniprep kit (Zymo Research) according
to manufacturer’s recommended procedure. Isolated DNA was quantied using a NanoDrop 2000C (ermo
Scientic) and stored at 80 °C. Samples were analyzed in the Arizona State University KED Genomics Core
for whole-sample 16 S rRNA gene amplicon sequencing. DNA library preparation for Illumina® MiSeq plat-
form was prepared according to the protocol from Earth Microbiome Project (http://www.earthmicrobiome.
org/emp-standard-protocols/16s). e 16 S primer set 515f-806r81 was used for 2 × 150 pair-ended sequencing.
16S rRNA gene amplicon sequencing and analysis. Illumina MiSeq sequencing reads (2 × 150 bp)
of the 16 S rRNA gene V4 region82 were analyzed with QIIME 2 (ver 2017.12)83 for 34 samples: Saline + Abx
(n = 6), Saline No Abx (n = 6), Serp-1 + Abx (n = 6), Serp-1 No Abx (n = 5), S7 + Abx (n = 6), S7 No Abx (n = 5).
Sequencing reads were processed with DADA2 to infer Amplicon Sequence Variants (ASVs) at a 97% identity
threshold using Greengenes database (version 13.8)84. To account for inter-sample depth variability, all samples
were rareed to 55,000 reads per sample (10 iterations) (Fig.S1A). One sample (12,107 reads; Saline No Abx
group) was omitted due to insucient reads and the remaining 33 samples were preserved for further analy-
sis. ASV richness, alpha diversity (Shannon’s Diversity Index) and beta diversity (UniFrac distance) were cal-
culated using QIIME 2. Statistical analyses (read depth, ASV richness and Shannon Index comparisons) were
performed with Mann-Whitney test. Bars are median ± standard error. P-value less than 0.05 was considered
signicant. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001; n.s. is not signicant. PCoA was performed with
weighted unifrac distance in QIIME 2. To identify discriminating ASVs associated with the respective treatments,
LEfSe (Linear discriminant analysis Eect Size)85, DESeq. 2 (version 1.20.0)86 and likelihood ratio tests were
performed in R studio (Version 1.1.456) (Fig.S1C–E). Discriminating analyses were rst performed compared
Serp-1 (no Abx) to saline (no Abx) mice, S-7 (no Abx) to saline (no Abx) mice, and nally as a combined group of
Serpin-treated mice [Serp-1 (no Abx) and S-7 (no Abx)] to saline (no Abx mice). Discriminant ASVs identied in
these analyses were pooled and validated in heatmap and abundance analyses (Fig.4D–F).
Histopathology. Mice were euthanized at 3 days follow-up aer infection by carbon dioxide asphyxiation
followed by cervical dislocation. Organs were harvested and xed in 10% neutral buered formalin. Samples
were dehydrated through graded alcohol, paraffin-embedded, sectioned into 4–6 µm ribbons, captured on
Treatment Antibiotics (ABX) Follow-up # of mice
Saline
No 3 days 6
Yes 3 days 6
No 150 days 12
Yes 150 days 5
Serp-1
No 3 days 6
Yes 3 days 6
No 150 days 5
Yes 150 days 5
S-7
No 3 days 6
Yes 3 days 6
No 150 days 5
Yes 150 days 5
Total # Infected Mice 73
Table 2. MHV-68-infected mice in this study.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
10
SCIENTIFIC REPORTS | (2020) 10:2371 | https://doi.org/10.1038/s41598-020-59269-9
www.nature.com/scientificreports
www.nature.com/scientificreports/
charged glass and dried overnight at 37 °C prior to processing for histopathology. Slides were rehydrated, stained
with Gill’s hematoxylin No.3 and Eosin Y (H&E) according to standard procedure, dehydrated and mounted in
Cytoseal XYL (ermo Fisher Scientic, USA).
Quantitative morphometry. H&E-stained aorta, lung and colon sections were imaged with a 20 × /0.5NA
objective on an Olympus BX51 microscope equipped with an Olympus DP74 camera operated by cellSens
Dimensions v1.16 soware. Objective-calibrated measurements of alveolar septal thickness and lumen area were
collected using cellSens Dimensions. At least 50 measurements were collected and averaged for each mouse, and
at least three mice were examined for each group. Images were processed for visualization in gures using ImageJ/
FIJI v1.52i87.
Immunohistochemistry. FFPE blocks containing lung tissue were sectioned into 4–6 µm ribbons, cap-
tured on charged glass and dried overnight at 37 °C prior to processing. Slides were rehydrated and epitopes
retrieved by boiling in sodium citrated buer. Endogenous peroxidases were quenched with 3% hydrogen per-
oxide, slides were blocked in 5% bovine serum albumin in TBS/0.1% Tween 20 and sections were probed with
rabbit polyclonal antibody against CD3 (Abcam ab5690; 1:200), CD4 (Abcam ab183685; 1:1000) or CD8 (Abcam
ab209775; 1:2000). For MHV-68 detection, 1:500 rabbit anti-serum or pre-immune serum (as control) were used
for immunostaining (kind gi of Dr. H.W. Virgin III9). 1:500 goat anti-rabbit HRP was used for secondary stain-
ing (Jackson Immuno Research 111-035-144). Antigens were revealed with ImmPACT DAB (Vector Labs, USA)
and mounted with Cytoseal XYL. Sections were imaged with a 40 × /0.75NA objective.
Viral load determination by qPCR. Total DNA was isolated from each FFPE sample (4 × 5 µm thick
sections for each sample was used) using the QIAamp DNA FFPE Tissue Kit according to the manufacturer’s
instructions. Samples were quantied using a DS-11 series spectrophotometer/uorometer (Denovix, USA).
Quantitative PCR (qPCR) was undertaken to investigate relative viral load using the following reaction per sam-
ple: 10 µL SsoAdvanced Universal SYBR Green Supermix (Bio-rad Laboratories Inc., USA), 0.4 µl each of the
primers 65 F (5-GTCAGGGCCCAGTCCGTA-3) and 65 R (5-TGGCCCTCTACCTTCTGTTGA-3), 200 ng of
DNA and water up to 20 µL total volume. Reactions were run in triplicate with controls, and a standard curve
using pCR2.1 Topo plasmid (ermo Fisher Scientic, USA) containing the cloned target MHV region35. e
following cycling conditions used on a CFX96 (Bio-rad Laboratories, Inc., USA) instrument: 95 °C for 20 sec, 40
cycles (95 °C for 15 sec, 60 °C for 20 sec), followed by a melt curve analysis.
Statistical analysis. Survival and pathology statistics were analyzed with GraphPad Prism v8.0.1.
Kaplan-Meier survival statistics were calculated using Log-rank (Mantel-Cox) testing. For visualization, individ-
ual comparison curves are presented. Lung pathology statistics were compared using a Two-Way ANOVA with
a Fisher’s LSD or Tukey’s post-hoc test. Bars are mean ± standard error. P-value less than 0.05 was considered
signicant. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001; n.s. is not signicant.
Data availability
Sequence data has been deposited to the NCBI Sequence Read Archive under BioProject accession number
PRJNA517927.
Received: 1 August 2019; Accepted: 22 January 2020;
Published: xx xx xxxx
References
1. Xie, Z. et al. Vascular endothelial hyperpermeability induces the clinical symptoms of Clarson disease (the systemic capillary lea
syndrome). Blood 119, 4321–4332 (2012).
2. Albe, J. . et al. Vascular permeability in the brain is a late pathogenic event during i Valley fever virus encephalitis in rats.
Virology 526, 173–179 (2018).
3. Winler, C. W., ace, B., Phillips, . & Peterson, . E. Capillaries in the olfactory bulb but not the cortex are highly susceptible to
virus-induced vascular lea and promote viral neuroinvasion. Acta Neuropathol. 130, 233–245 (2015).
4. Pedro Simas, J. & Efstathiou, S. Murine gammaherpesvirus 68: A model for the study of gammaherpesvirus pathogenesis. Trends
Microbiol. 6, 276–282 (1998).
5. Elefante, E., Tripoli, A., Ferro, F. & Baldini, C. One year in review: systemic vasculitis. Clin. Exp. heumatol. 34, S1–6 (2016).
6. Elefante, E. et al. One year in review 2017: systemic vasculitis. Clin. Exp. heumatol. 35(Suppl 1), 5–26 (2017).
7. Elefante, E. et al. One year in review 2018: systemic vasculitis. Clin. Exp. heumatol. 36(Suppl 1), 12–32 (2018).
8. odo, X. et al. Tropospheric winds from northeastern China carry the etiologic agent of awasai disease from its source to Japan.
Proc. Natl. Acad. Sci. 111, 7952–7957 (2014).
9. Wec, . E. et al. Murine gamma-herpesvirus 68 causes severe large-vessel arteritis in mice lacing interferon-gamma
responsiveness: a new model for virus-induced vascular disease. Nat. Med. 3, 1346–53 (1997).
10. Chen, H. et al. Myxomavirus-derived serpin prolongs survival and reduces inammation and hemorrhage in an unrelated lethal
mouse viral infection. Antimicrob. Agents Chemother. 57, 4114–4127 (2013).
11. Chen, H. et al. Mouse gamma herpesvirus MHV-68 induces severe gastrointestinal (GI) dilatation in interferon gamma receptor-
decient mice (IFNγ/) that is bloced by interleuin-10. Viruses 10, (2018).
12. Waeeld, A. J. et al. Detection of herpesvirus DNA in the large intestine of patients with ulcerative colitis and Crohn’s disease using
the nested polymerase chain reaction. J. Med. Virol. 38, 183–190 (1992).
13. Nagasai, S., Oita, ., Mitani, N., Shimizu, N. & Yanai, H. Epstein-Barr virus infection of the colon with inammatory bowel
disease. Am. J. Gastroenterol. 94, 1582–1586 (2004).
14. yan, J. L. et al. Epstein-barr virus infection is common in inamed gastrointestinal mucosa. Dig. Dis. Sci. 57, 1887–1898 (2012).
15 . Soowamber, M., Weizman, A. V. & Pagnoux, C. Gastrointestinal aspects of vasculitides. Nat. ev. Gastroenterol. Hepatol. 14, 185–194
(2017).
16. Hatemi, I., Hatemi, G. & Çeli, A. F. Systemic vasculitis and the gut. Curr. Opin. heumatol. 29, 33–38 (2017).
Content courtesy of Springer Nature, terms of use apply. Rights reserved
11
SCIENTIFIC REPORTS | (2020) 10:2371 | https://doi.org/10.1038/s41598-020-59269-9
www.nature.com/scientificreports
www.nature.com/scientificreports/
17. Lucas, A., Yaron, J. ., Zhang, L., Macaulay, C. & McFadden, G. Serpins: Development for erapeutic Applications. Methods Mol.
Biol. 1826, 255–265 (2018).
18. Ildefonso, C. J. et al. Gene Delivery of a Viral Anti-Inammatory Protein to Combat Ocular Inammation. Hum. Gene er. 26,
59–68 (2015).
19. Miller, L. W. et al. Inhibition of transplant vasculopathy in a rat aortic allogra model aer infusion of anti-inammatory viral
serpin. Circulation 101, 1598–605 (2000).
20. wiecien, J. M. et al. Myxoma virus derived immune modulating proteins, M-T7 and Serp-1, reduce early inammation aer spinal
cord injury in the rat model. Folia Neuropathol. 57, 41–50 (2019).
21. Tardif, J.-C. et al. A randomized controlled, phase 2 trial of the viral serpin Serp-1 in patients with acute coronary syndromes
undergoing percutaneous coronary intervention. Circ. Cardiovasc. Interv. 3, 543–8 (2010).
22. Ambadapadi, S. et al. eactive Center Loop (CL) peptides derived from serpins display independent coagulation and immune
modulating activities. J. Biol. Chem. 291, 2874–2887 (2016).
23. Nash, P., Whitty, A., Handwerer, J., Macen, J. & McFadden, G. Inhibitory specicity of the anti-inammatory myxoma virus serpin,
SEP-1. J. Biol. Chem. 273, 20982–20991 (1998).
24. Viswanathan, . et al. Myxoma viral serpin, Serp-1, a unique interceptor of coagulation and innate immune pathways. romb.
Haemost. 95, 499–510 (2006).
25. Mahon, B. P. et al. Crystal Structure of Cleaved Serp-1, a Myxomavirus-Derived Immune Modulating Serpin: Structural Design of
Serpin eactive Center Loop Peptides with Improved erapeutic Function. Biochemistry 57, 1096–1107 (2018).
26. Flano, E., Husain, S. M., Sample, J. T., Woodland, D. L. & Blacman, M. A. Latent Murine -Herpesvirus Infection Is Established in
Activated B Cells, Dendritic Cells, and Macrophages. J. Immunol. 165, 1074–1081 (2000).
27. Dal Canto, A. J., Swanson, P. E., O’Guin, A. ., Spec, S. H. & Virgin, H. W. IFN-gamma action in the media of the great elastic
arteries, a novel immunoprivileged site. J. Clin. Invest. 107, 15–22 (2001).
28. Spieeroetter, E. et al. eactivation of gammaHV68 induces neointimal lesions in pulmonary arteries of S100A4/Mts1-
overexpressing mice in association with degradation of elastin. Am. J. Physiol Lung Cell. Mol. Physiol. 294, L276–L289 (2008).
29. Chen, H. et al. Analysis of In Vivo Serpin Functions in Models of Inammatory Vascular Disease. Methods Mol. Biol. 1826, 157–182
(2018).
30. eita, M. B. et al. Non-contiguous nished genome sequence and description of Bacillus massiliogorillae sp. nov. Stand. Genomic
Sci. 9, 93–105 (2013).
31. Lee, S. D. Frondihabitans peucedani sp. nov., an actinobacterium isolated from rhizosphere soil, and emended description of the
genus Frondihabitans Greene et al. 2009. Int. J. Syst. Evol. Microbiol. 60, 1740–1744 (2010).
32. Harada, T. et al. Enterococcus saigonensis sp. nov., isolated from retail chicen meat and liver. Int. J. Syst. Evol. Microbiol. 66,
3779–3785 (2016).
33. Stewart, J. P., Usherwood, E. J., oss, A., Dyson, H. & Nash, T. Lung Epithelial Cells Are a Major Site of Murine Gammaherpesvirus
Persistence. J. Exp. Med. 187, 1941–1951 (1998).
34. Coen, N. et al. Activity and Mechanism of Action of HDVD, a Novel Pyrimidine Nucleoside Derivative with High Levels of
Selectivity and Potency against Gammaherpesviruses. J. Virol. 87, 3839–3851 (2013).
35. anai, . et al. Murine γ-Herpesvirus 68 Induces Severe Lung Inammation in IL-27–Decient Mice with Liver Dysfunction
Preventable by Oral Neomycin. J. Immunol. 200, 2703–2713 (2018).
36. Mora, A. L. et al. Lung infection with γ-herpesvirus induces progressive pulmonary brosis in 2-biased mice. Am. J. Physiol. Cell.
Mol. Physiol. 289, L711–L721 (2005).
37. Hanaoa, M. et al. Immunomodulatory strategies prevent the development of autoimmune emphysema. espir. es. 11, 179 (2010).
38. Organ, L. et al. Structural and functional correlations in a large animal model of bleomycin-induced pulmonary brosis. BMC Pulm.
Med. 15, (2015).
39. Costola-de-Souza, C. et al. Monoacylglycerol Lipase (MAGL) Inhibition Attenuates Acute Lung Injury in Mice. PLoS One 8, 1–15
(2013).
40. Cardin, . D., Broos, J. W., Sarawar, S. . & Doherty, P. C. Progressive loss of CD8+ T cell-mediated control of a gamma-
herpesvirus in the absence of CD4+ T cells. J. Exp. Med. 184, 863–71 (1996).
41. Spars-issen, . L., Braaten, D. C., reher, S., Spec, S. H. & Virgin, H. W. An optimized CD4 T-cell response can control
productive and latent gammaherpesvirus infection. J. Virol. 78, 6827–35 (2004).
42. Spars-issen, . L. et al. CD4 T cell control of acute and latent murine gammaherpesvirus infection requires IFNγ. Virology 338,
201–208 (2005).
43. Schirmer, M. et al. Lining the Human Gut Microbiome to Inammatory Cytoine Production Capacity. Cell 167, 1125–1136.e8
(2016).
44 . Yang, J. H. et al. Antibiotic-Induced Changes to the Host Metabolic Environment Inhibit Drug Ecacy and Alter Immune Function.
Cell Host Microbe 22, 1–9 (2017).
45. Blander, J. M., Longman, . S., Iliev, I. D., Sonnenberg, G. F. & Artis, D. egulation of inammation by microbiota interactions with
the host. Nat. Immunol. 18, 851–860 (2017).
46. Budden, . F. et al. Emerging pathogenic lins between microbiota and the gut-lung axis. Nat. ev. Microbiol. 15, 55–63 (2017).
47. Tripathi, A. et al. e gut-liver axis and the intersection with the microbiome. Nat. ev. Gastroenterol. Hepatol. 15, 397–411 (2018).
48. amezani, A. & aj, D. S. e Gut Microbiome, idney Disease, and Targeted Interventions. J. Am. Soc. Nephrol. 25, 657–670
(2014).
49. Scheperjans, F. et al. Gut microbiota are related to Parinsons disease and clinical phenotype. Mov. Disord. 30, 350–358 (2015).
50. Hevia, A. et al. Intestinal dysbiosis associated with systemic lupus erythematosus. Mbio. 5, 1–10 (2014).
51. Scher, J. U. et al. Expansion of intestinal Prevotella copri correlates with enhanced susceptibility to arthritis. Elife. 2013, 1–20 (2013).
52. Chen, X. et al. e cardiovascular macrophage: a missing lin between gut microbiota and cardiovascular diseases? Eur. ev. Med.
Pharmacol. Sci. 22, 1860–1872 (2018).
53. asselman, L. J., Vernice, N. A., DeLeon, J. & eiss, A. B. e gut microbiome and elevated cardiovascular ris in obesity and
autoimmunity. Atherosclerosis 271, 203–213 (2018).
54. Singh, V., Yeoh, B. S. & Vijay-umar, M. Gut microbiome as a novel cardiovascular therapeutic target. Curr. Opin. Pharmacol. 27,
8–12 (2016).
55. Bajaj, J. S. et al. Fecal microbiota transplant from a rational stool donor improves hepatic encephalopathy: A randomized clinical
trial. Hepatology 66, 1727–1738 (2017).
56. Vrieze, A. et al. Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic
syndrome. Gastroenterology 143, 913–916.e7 (2012).
57. outy, B. et al. Gut microbiome inuences ecacy of PD-1–based immunotherapy against epithelial tumors. Science (80-.). 359,
91–97 (2018).
58. Matson, V. et al. e commensal microbiome is associated with anti–PD-1 ecacy in metastatic melanoma patients. Science (80-.).
359, 104–108 (2018).
59. Gopalarishnan, V. et al. Gut microbiome modulates response to anti–PD-1 immunotherapy in melanoma patients. Science (80-.).
359, 97–103 (2018).
Content courtesy of Springer Nature, terms of use apply. Rights reserved
12
SCIENTIFIC REPORTS | (2020) 10:2371 | https://doi.org/10.1038/s41598-020-59269-9
www.nature.com/scientificreports
www.nature.com/scientificreports/
60. Vetizou, M. et al. Anticancer immunotherapy by CTLA-4 blocade relies on the gut microbiota. Science (80-.). 350, 1079–1084
(2015).
61. Imhann, F. et al. Proton pump inhibitors aect the gut microbiome. Gut 65, 740–748 (2016).
62. Jones, M. . et al. Enteric bacteria promote human and mouse norovirus infection of B cells. Science (80-.). 346, 755–759 (2014).
63. Baldridge, M. T. et al. Commensal microbes and interferon-λ determine persistence of enteric murine norovirus infection. Science
(80-.). 347, 266–269 (2015).
64. acray, L. B. et al. Oral Antibiotic Treatment of Mice Exacerbates the Disease Severity of Multiple Flavivirus Infections. Cell ep.
22, 3440–3453.e6 (2018).
65. Handley, S. A. e virome: A missing component of biological interaction networs in health and disease. Genome Med. 8, 32–34
(2016).
66. Bédos, J.-P. et al. Pharmacodynamic activities of ciprooxacin and sparoxacin in a murine pneumococcal pneumonia model:
elevance for drug ecacy. J. Pharmacol. Exp. er. 286, 29–35 (1998).
67. Swenson, C. E., Stewart, . A., Hammett, J. L., Fitzsimmons, W. E. & Ginsberg, . S. Pharmacoinetics and in vivo activity of
liposome-encapsulated gentamicin. Antimicrob. Agents Chemother. 34, 235–240 (1990).
68. A., B. Inuence of the destabilisation of the maternal digestive microora on that of the newborn rat. Biol. Neonate 63, 236–245
(1993).
69. itschel, W. A. Biological Half-Lives of Drugs. Drug Intell. Clin. Pharm. 4, 332–347 (1970).
70. Stünel, . G. E., Hewlett, G. & Zeiler, H. J. Ciprooxacin enhances T cell function by modulating interleuin activities. Clin. Exp.
Immunol. 86, 525–531 (1991).
71. Zhang, L. et al. A Virus-Derived Immune Modulating Serpin Accelerates Wound Closure with Improved Collagen emodeling. J.
Clin. Med. 8, 1626 (2019).
72. Viswanathan, . et al. Myxoma viral serpin, Serp-1, inhibits human monocyte adhesion through regulation of actin-binding protein
lamin B. J. Leuoc. Biol. 85, 418–26 (2009).
73. Florquin, S. et al. elease of uroinase plasminogen activator receptor during urosepsis and endotoxemia. idney Int. 59, 2054–2061
(2001).
74. Wymore Brand, M. et al. e Altered Schaedler Flora: Continued Applications of a Dened Murine Microbial Community. ILA J.
56, 169–178 (2015).
75. Olivadoti, M., Toth, L. A., Weinberg, J. & Opp, M. . Murine gammaherpesvirus 68: A model for the study of Epstein-Barr virus
infections and related diseases. Comp. Med. 57, 44–50 (2007).
76. Carlson, J. A., Perlmutter, A., Tobin, E., ichardson, D. & ohwedder, A. Adverse antibiotic-induced eruptions associated with
epstein barr virus infection and showing iuchi-Fujimoto disease-lie histology. Am. J. Dermatopathol. 28, 48–55 (2006).
77. Fei, . et al. Anti-inammatory activity of a thermophilic serine protease inhibitor from extremophile Pyrobaculum neutrophilum.
Eur. J. Inamm. 15, 143–151 (2017).
78. Ehlers, M. . Immune-modulating eects of alpha-1 antitrypsin. Biol. Chem. 395, 1187–1193 (2014).
79. Badawi, A. H. & Siahaan, T. J. Immune modulating peptides for the treatment and suppression of multiple sclerosis. Clin. Immunol.
144, 127–138 (2012).
80. I.,  . et al. β-cell function in new-onset type 1 diabetes and immunomodulation with a heat-shoc protein peptide (DiaPep277): A
randomised, double-blind, phase II trial. Lancet 358, 1749–1753 (2001).
81. Caporaso, J. G. et al. Global patterns of 16S rNA diversity at a depth of millions of sequences per sample. Proc. Natl. Acad. Sci. 108,
4516–4522 (2011).
82. Maldonado, J., Yaron, J. ., Zhang, L. & Lucas, A. Next-Generation Sequencing Library Preparation for 16S rNA Microbiome
Analysis Aer Serpin Treatment. Methods Mol. Biol. 1826, 213–221 (2018).
83. Bolyen, E. et al. QIIME 2: eproducible, interactive, scalable, and extensible microbiome data science. PeerJ Prepr. 6, e27295v1
(2018).
84. DeSantis, T. Z. et al. Greengenes, a chimera-checed 16S rNA gene database and worbench compatible with AB. Appl. Environ.
Microbiol. 72, 5069–5072 (2006).
85. Segata, N. et al. Metagenomic biomarer discovery and explanation. Genome Biol. 12, (2011).
86. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for NA-seq data with DESeq. 2. Genome
Biol. 15, 1–21 (2014).
87. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).
88. Stone, . J. & Strominger, J. L. Mechanism of Action of Bacitracin: Complexation with Metal Ion and C55-Isoprenyl Pyrophosphate.
Proc. Natl. Acad. Sci. 68, 3223–3227 (1971).
89. otra, L. P., Haddad, J. & Mobashery, S. Aminoglycosides: Perspectives on mechanisms of action and resistance and strategies to
counter resistance. Antimicrob. Agents Chemother. 44, 3249–3256 (2000).
90. Luzzatto, L., Apirion, D. & Schlessinger, D. Mechanism of action of streptomycin in E. coli: interruption of the ribosome cycle at the
initiation of protein synthesis. Proc. Natl. Acad. Sci. USA 60, 873–80 (1968).
91. Sanders, C. C. Ciprooxacin: In Vit ro Activity, Mechanism of Action, and esistance. Clin. Infect. Dis. 10, 516–527 (1988).
Acknowledgements
e authors gratefully acknowledge Dr. Grant McFadden and Dr. Christian Jobin for many helpful discussions.
The authors acknowledge resources and support from the KED Genomics Core, part of the Biosciences
Core Facilities at Arizona State University. This study was financially supported by grants from the NIH
(1R01AI100987-01A1 and 1RC1HL100202), American Heart Association (17GRNT33460327), University of
Florida Gatorade Fund (00115070) and start-up funds from the Biodesign Institute at Arizona State University
all to ARL.
Author contributions
J.R.Y., S.A., L.Z., and A.R.L. designed research; J.R.Y., L.Z., R.C., E.L., A.V., S.Kraberger., J.M., R.K-B., B.H.M.
E.O.K. and A.R.L. analyzed data. J.R.Y. S.A. L.Z. S.Kraberger., A.S-H., A.M.T. and J.K. performed research; S.A.T.,
S.Keinan. and W.B. contributed new reagents or analytic tools; J.R.Y. and A.R.L. wrote the paper. All authors
reviewed the paper.
Competing interests
ARL is a listed inventor on issued patents US20060122115A1 and US7419670B2 on the therapeutic uses of
Serp-1, and on patent application US20180319868A1 on the therapeutic uses of Serp-1-derived RCL peptides.
ARL and JRY are listed inventors on provisional patent application 62/792,201and PCT/US20/13398 on
microbiome eects in inammatory vasculitis. ere are no other competing interests.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
13
SCIENTIFIC REPORTS | (2020) 10:2371 | https://doi.org/10.1038/s41598-020-59269-9
www.nature.com/scientificreports
www.nature.com/scientificreports/
Additional information
Supplementary information is available for this paper at https://doi.org/10.1038/s41598-020-59269-9.
Correspondence and requests for materials should be addressed to E.S.L. or A.R.L.
Reprints and permissions information is available at www.nature.com/reprints.
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and
institutional aliations.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International
License, which permits use, sharing, adaptation, distribution and reproduction in any medium or
format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Cre-
ative Commons license, and indicate if changes were made. e images or other third party material in this
article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the
material. If material is not included in the article’s Creative Commons license and your intended use is not per-
mitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the
copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.
© e Author(s) 2020
Content courtesy of Springer Nature, terms of use apply. Rights reserved
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... Mikrobiyota, hematopoez, inflamasyon ve immünite başta olmak üzere birçok fizyolojik fonksiyon üzerinde etkisi bulunmaktadır. Mikrobiyotanın insan sağlığı ve kanser dahil pek çok hastalıkla ilişkisinin gösterilmesi ile bu alanda çalışmalar hız kazanmış ve mikrobiyotanın artık, yeni bir organ olarak değerlendirilmesi gerektiği görüşü ortaya çıkmıştır (8)(9)(10)(11)(12)(13)(14)(15)(16)(17). Yaş, coğrafi farklılıklar, diyet alışkanlıkları, antibiyotikler, probiyotikler gibi faktörler intestinal motilite, pH, redoks durumu, mikronutrientler, sekresyonlar (asit, safra,mukus, enzimler) gibi tanımlanmış birçok etiyolojik faktör, mikrobiyotanın kompozisyonunun farklılık göstermesine yol açabilmekte, ve bu değişiklikler diabetes mellitus, kardiyovasküler hastalıklar ve kanser dahil birçok kronik hastalık ile ilişkili bulunmuştur (9)(10). ...
... Mikrobiyotanın insan sağlığı ve kanser dahil pek çok hastalıkla ilişkisinin gösterilmesi ile bu alanda çalışmalar hız kazanmış ve mikrobiyotanın artık, yeni bir organ olarak değerlendirilmesi gerektiği görüşü ortaya çıkmıştır (8)(9)(10)(11)(12)(13)(14)(15)(16)(17). Yaş, coğrafi farklılıklar, diyet alışkanlıkları, antibiyotikler, probiyotikler gibi faktörler intestinal motilite, pH, redoks durumu, mikronutrientler, sekresyonlar (asit, safra,mukus, enzimler) gibi tanımlanmış birçok etiyolojik faktör, mikrobiyotanın kompozisyonunun farklılık göstermesine yol açabilmekte, ve bu değişiklikler diabetes mellitus, kardiyovasküler hastalıklar ve kanser dahil birçok kronik hastalık ile ilişkili bulunmuştur (9)(10). Tüm kanserlerin yaklaşık %15 'inde patojenik mikroorganizmalar etken olarak tanımlanmıştı. ...
... Bu ajanlar, tümör supresyonunun bozulması, konakçı DNA'sında virusun onkogenik etkileşimi, lokal immün yanıtı bozması gibi farklı biyolojik ONCOLOGY NURSING CONGRESS [18][19] December 2020, Online Congress yöntemlerle tümör oluşumuna neden olmaktadırlar. Son zamanlarda yapılan çalışmalarda, Mikrobiyotanın, özelliklede barsak Mikrobiyotasının kanserin inisiasyonu, progresyonu ve vücuttaki yaygınlığı ile yakın bir ilişki içinde bulunduğu, yani karsinogenezin her aşamasında anahtar bir rol oynayabileceği, ek olarak da, anti kanser tedavilere yanıt ve toksik yan etki eğilimlerini de değiştirebileceği ile ilgili yeni kanıtlar ortaya çıkmaktadır (8)(9)(10)(11) . ...
Conference Paper
Full-text available
Özet: Kanserden ölüm oranı kalp damar hastalıklarından sonra 2. sırada yeralmaktadır. Kanser kelime anlamıyla kontrolsüz hücre çoğalmasıdır. İnsan vücudunda kanserin çoğalmasının engelleyen en önemli faktör bağışıklık sistemidir. İmmun sistemin 'mikrobiyota gibi ' gibi çok önemli bir destek sistemi bulunmaktadır. Son zamanlarda oldukça önem kazanmaktadır. Amaç: kanser gelişiminde mikrobiyotanın ilişkinisin önemini açıklamak amacıyla çalışılmıştır. Kanser tedavi güç ve uzun zamana yayılan, mortalite ve morbidite oranı yüksek olan zor bir hastalıktır. Tedavi seçenekleri olmasına karşın başarı oranı ve hayatta kalım oranı hala daha istendik seviyelerde olamayabilmektedir. Kullanılan tedavi yöntemleri yan etkileri oldukça fazla olup bireyin yaşamını olumsuz etkileyebilmektedir. Tüm bu olumsuzlar nedeniyle kanser için risk faktörlerinin belirlenip korunmaya çalışılmak gerekmektedir. İnsan vücudunda kanser gelişmesini engelleyebilecek çok önemli mekanizmalara sahiptir. Fakat gelişen teknoloji ve radyoaktif maruziyeti gibi birçok dış etkenlerden dolayı bu mekanizmalar etkilenerek kanser gelişmek için uygun ortamı bulmaktadır. Mikrobiyota immun sisteminin şüphesiz olarak önemli bir parçası haline gelmiştir. İçerdiği birçok bakteri ve mikroogranizma sayesinde sayısız fonksiyona sahip olup, birçok hastalık ile de yakından ilişkilidir. Son yıllarda özellikle bağırsak mikrobiyotasına yoğunlaşan araştırmacılar, obesite merkezi sinir sistemi hastalıkları, depresyon ve anksiyete, kolon kanseri, alzheimer gibi hastalıklarının gelişmesinde önemli ilişkisi olduğu savunulmaktadır. Mikrobiyota zenginliği sadece bağarsaklarda olduğu savunulmaktayken son çalışmalarda akciğer, deri, vajina bölgelerinin kendi flora sahip oldukları kanıtlanmıştır. Mikrobiyota zenginliği, beslenme alışkanlıkları, genetik yatkınlıklar, geçirilen hastalıklar, maruz kalınan patojenler, tüketilen ilaçlar gibi birçok değişkene bağlı olarak sürekli bir değişmektedir. Kullanılan antibiyotiklerin yararlı bakterileri öldürmesi ve kalan bakterilerin baskınlığının artması mikrobiyota dengesini değiştirmekte ve yine hastalıkların ortaya çıkmasına neden olmaktadır. İnce bağarsak florasındaki zenginlik kalın bağarsağa göre daha fazla olup kalın bağarsağın daha mikrop açısından daha temiz olmaktadır. Benzer olarak kalın bağarsakta ince bağarsağa oranla daha fazla kanser geliştiği çok yangındır. Bu iki bilginin ilişkisi olduğu savunulmaktadır. Ancak bazı patojenlere maruz kalarak da bu sistemin bozulmasına neden olunabilir. mide kanserinin sebeplerinden biri mide mikrobiyotasının bozulması olarak gösterilmektedir. Mide florasının bozulmasını sağlayabilecek olan ve dışarıdan gelen "Helicobakter pylori" ise en büyük tehditlerden birini oluşturmaktadır. Hemşireler olarak bu bilgiler ışığında hastalarımıza bütüncül yaklaşarak beslenme alışkanlıkları ve yaşam stilini sorgulamak ve öykü almak oldukça önemli bir görevi olabilmektedir. Sonuç: Kanser insan hayatı için önemli bir tehdit olup gelişmesi bağışıklık sistemini güçlendirerek mümkün olabilmektedir. Bağışıklığın yanı sıra mikrobiyota zenginliğinide çok önemli olduğu savunulmaktadır. Bu doğrultuda bakıldığında hemşirelerin hastaları beslenme alışkanlıklarına göre değerlendirip risk gruplarını belirliyebilmektedir. Kanserde korunmada bu durumun değerlendirmesi önerilmektedir.
... The treatment of MHV-68 infections in interferon gamma receptor knockout mice was further tested as a model for lethal vasculitis, DIC, colitis, and lung hemorrhage in a study investigating the gut microbiome. The suppression of gut bacteria by the oral administration of an antibiotic cocktail worsened MHV-68 infection and blocked Serp-1 treatment efficacy, indicating that the Serp-1 therapeutic efficacy in this MHV68 model was dependent upon an intact gut microbiota [27]. While acute transplant rejection is well-controlled with current immunosuppressants, chronic transplant vasculopathy and rejection remain a barrier to long-term transplant function. ...
Article
Full-text available
Serine protease inhibitors, SERPINS, are a highly conserved family of proteins that regulate serine proteases in the central coagulation and immune pathways, representing 2–10% of circulating proteins in the blood. Serine proteases form cascades of sequentially activated enzymes that direct thrombosis (clot formation) and thrombolysis (clot dissolution), complement activation in immune responses and also programmed cell death (apoptosis). Virus-derived serpins have co-evolved with mammalian proteases and serpins, developing into highly effective inhibitors of mammalian proteolytic pathways. Through interacting with extracellular and intracellular serine and cysteine proteases, viral serpins provide a new class of highly active virus-derived coagulation-, immune-, and apoptosis-modulating drug candidates. Viral serpins have unique characteristics: (1) function at micrograms per kilogram doses; (2) selectivity in targeting sites of protease activation; (3) minimal side effects at active concentrations; and (4) the demonstrated capacity to be modified, or fine-tuned, for altered protease targeting. To date, the virus-derived serpin class of biologics has proven effective in a wide range of animal models and in one clinical trial in patients with unstable coronary disease. Here, we outline the known viral serpins and review prior studies with viral serpins, considering their potential for application as new sources for immune-, coagulation-, and apoptosis-modulating therapeutics.
... Activation of latent Epstein-Barr virus (EBV) infection is linked to the development of MS (144,145). EBV is a common virus that is considered part of the commensal microbiome (146). EBV infects B cells and epithelial cells, and because it shares molecular mimicry to some host protein, the viral genome integrates within the host DNA. ...
Article
Full-text available
Trillions of microbes survive and thrive inside the human body. These tiny creatures are crucial to the development and maturation of our immune system and to maintain gut immune homeostasis. Microbial dysbiosis is the main driver of local inflammatory and autoimmune diseases such as colitis and inflammatory bowel diseases. Dysbiosis in the gut can also drive systemic autoimmune diseases such as type 1 diabetes, rheumatic arthritis, and multiple sclerosis. Gut microbes directly interact with the immune system by multiple mechanisms including modulation of the host microRNAs affecting gene expression at the post-transcriptional level or production of microbial metabolites that interact with cellular receptors such as TLRs and GPCRs. This interaction modulates crucial immune functions such as differentiation of lymphocytes, production of interleukins, or controlling the leakage of inflammatory molecules from the gut to the systemic circulation. In this review, we compile and analyze data to gain insights into the underpinning mechanisms mediating systemic autoimmune diseases. Understanding how gut microbes can trigger or protect from systemic autoimmune diseases is crucial to (1) tackle these diseases through diet or lifestyle modification, (2) develop new microbiome-based therapeutics such as prebiotics or probiotics, (3) identify diagnostic biomarkers to predict disease risk, and (4) observe and intervene with microbial population change with the flare-up of autoimmune responses. Considering the microbiome signature as a crucial player in systemic autoimmune diseases might hold a promise to turn these untreatable diseases into manageable or preventable ones.
... This virus, with its surface protein (S) can bind to the host receptor angiotensin converting enzyme 2 (ACE2) to enter the host cell. ACE2 is a peptidase and is attached to the cell membrane in several tissues throughout the body, including the heart, gastrointestinal tract, kidneys, and lungs [5][6][7]. The most common symptoms of COVID-19 infection are respiratory symptoms, fever, cough, dyspnea, malaise, and fatigue. ...
Article
Full-text available
Background: Healthcare providers are high-risk groups for the novel coronavirus disease (COVID-19) infection. Nursing students are an essential part of high-risk healthcare providers. The aim of this study was to evaluate the knowledge, attitude, and practice of nursing and midwifery students at Golestan University of Medical Sciences (GOUMS) toward the prevention of COVID-19. Methods: This cross-sectional study was conducted on 174 nursing and midwifery students of GOUMS (northeast of IRAN) in 2020. Data were gathered using an online questionnaire comprising three parts – Knowledge (8 questions), Attitude (10 questions), and Practice (8 questions) toward the prevention of COVID-19. The range of scores in each subscale (knowledge, attitude, and preventive practices) were 8–24, 10–50, and 8–24, respectively. Data were analyzed using the SPSS v.16 software. The Mann–Whitney and Kruskal–Wallis tests were used for the analysis. Results: Overall, in this study, nursing and midwifery students had a good knowledge (mean score: 23.19 ± 2.56), a positive attitude (mean score: 45.48 ± 4.21), and appropriate practice (mean score: 23.30 ± 3.51) regarding COVID-19 prevention. Also, the results revealed a positive correlation between knowledge and attitude (r = 0.1, P = 0.18) and attitude and practice (r = 0.2, P = 0.01) among the students. Conclusion: The findings demonstrated a good preventive knowledge, attitude, and practice toward COVID-19 among nursing and midwifery students.
... In contrast to the above, the gut microbiome can affect the infection processes of various eukaryotic enteric viruses in the gastrointestinal tract [194] and can induce an immune response that controls the infection process of eukaryotic viruses [195]. The enteric microbiota enhances the production of mucus and the synthesis of potential antiviral compounds, such as lactic acid, acetic acid, reactive oxygen species, and defensins, which inhibit local viral replication [196]. ...
Article
Full-text available
The core microbiome, which refers to a set of consistent microbial features across populations, is of major interest in microbiome research and has been addressed by numerous studies. Understanding the core microbiome can help identify elements that lead to dysbiosis, and lead to treatments for microbiome-related health states. However, defining the core microbiome is a complex task at several levels. In this review, we consider the current state of core human microbiome research. We consider the knowledge that has been gained, the factors limiting our ability to achieve a reliable description of the core human microbiome, and the fields most likely to improve that ability. DNA sequencing technologies and the methods for analyzing metagenomics and amplicon data will most likely facilitate higher accuracy and resolution in describing the microbiome. However, more effort should be invested in characterizing the microbiome’s interactions with its human host, including the immune system and nutrition. Other components of this holobiontic system should also be emphasized, such as fungi, protists, lower eukaryotes, viruses, and phages. Most importantly, a collaborative effort of experts in microbiology, nutrition, immunology, medicine, systems biology, bioinformatics, and machine learning is probably required to identify the traits of the core human microbiome.
... and depleted in Clostridia spp. In an experiment with microbiome-depleted hosts, Yaron et al. (53) inoculated axenic mice with murine gammaherpesvirus 68 (MHV-68). These mice had a lower survival rate than the control group. ...
Article
Full-text available
The microorganisms associated with an organism, the microbiome, have a strong and wide impact in their host biology. In particular, the microbiome modulates both the host defense responses and immunity, thus influencing the fate of infections by pathogens. Indeed, this immune modulation and/or interaction with pathogenic viruses can be essential to define the outcome of viral infections. Understanding the interplay between the microbiome and pathogenic viruses opens future venues to fight viral infections and enhance the efficacy of antiviral therapies. An increasing number of researchers are focusing on microbiome-virus interactions, studying diverse combinations of microbial communities, hosts, and pathogenic viruses. Here, we aim to review these studies, providing an integrative overview of the microbiome impact on viral infection across different pathosystems.
... Furthermore, growing evidence suggests that alterations in the human microbiome may also occur in response to viral infections (12)(13)(14)(15)(16)(17). Bacteria can also play important roles during viral infection processes, ranging from offering protection against viral agents (18)(19)(20)(21)(22) to facilitating viral infections or participating in bacterial-viral coinfections (23)(24)(25)(26)(27). The continuous emergence of novel pathogenic viruses at a global scale, such as the H1N1 influenza A virus (28,29), or the coronaviruses responsible for the severe acute respiratory syndrome (SARS-CoV) (30,31), the Middle East respiratory syndrome (MERS-CoV) (32,33), or the more recent SARS-CoV-2 causative of the current COVID-19 pandemic (34,35), emphasizes the need to understand how microbial communities might be related to these pathogens and whether they modulate infection risk. ...
Article
Full-text available
The global emergence of novel pathogenic viruses presents an important challenge for research, as high biosafety levels are required to process samples. While inactivation of infectious agents facilitates the use of less stringent safety conditions, its effect on other biological entities of interest present in the sample is generally unknown. Here, we analyzed the effect of five inactivation methods (heat, ethanol, formaldehyde, psoralen, and TRIzol) on microbiome composition and diversity in samples collected from four different body sites (gut, nasal, oral, and skin) and compared them against untreated samples from the same tissues. We performed 16S rRNA gene sequencing and estimated abundance and diversity of bacterial taxa present in all samples. Nasal and skin samples were the most affected by inactivation, with ethanol and TRIzol inducing the largest changes in composition, and heat, formaldehyde, TRIzol, and psoralen inducing the largest changes in diversity. Oral and stool microbiomes were more robust to inactivation, with no significant changes in diversity and only moderate changes in composition. Firmicutes was the taxonomic group least affected by inactivation, while Bacteroidetes had a notable enrichment in nasal samples and moderate enrichment in fecal and oral samples. Actinobacteria were more notably depleted in fecal and skin samples, and Proteobacteria exhibited a more variable behavior depending on sample type and inactivation method. Overall, our results demonstrate that inactivation methods can alter the microbiome in a tissue-specific manner and that careful consideration should be given to the choice of method based on the sample type under study.
Article
Full-text available
SARS-CoV-2 mainly affects the respiratory system, but the gastrointestinal tract is also a target. Prolonged gut disorders, in COVID-19 patients, were correlated with decreased richness and diversity of the gut microbiota, immune deregulation and delayed viral clearance. Although there are no definitive conclusions, ample evidence would suggest that the gut microbiome composition and function play a role in COVID-19 progression. Microbiome modulation strategies for population stratification and management of COVID-19 infection are under investigation, representing an area of interest in the ongoing pandemic. In this review, we present the existing data related to the interaction between gut microbes and the host’s immune response to SARS-CoV-2 and discuss the implications for current disease management and readiness to face future pandemics.
Article
Peptides, short stretches of amino acids or small proteins that occupy a strategic position between proteins and amino acids, are readily accessible by chemical and biological methods. With ideal properties for forming high-affinity and specific interactions with host target proteins, they have established an important niche in the drug development spectrum complementing small molecule and biological therapeutics. Among the most successful biomedicines in use today, peptide-based drugs show great promise. This, coupled with recent advances in synthetic and nanochemical biology, has led to the creation of tailor-made peptide therapeutics for improved biocompatibility. This review presents an overview of the latest research on pathogen-derived, host-cell-interacting peptides. It also highlights strategies for using peptide-based therapeutics that address cellular transport challenges through the introduction of nanoparticles that serve as platforms to facilitate the delivery of peptide biologics and therapeutics for treating various inflammatory diseases. Finally, this paper describes future perspectives, specific pathogen-based peptides that can enhance specificity, efficiency, and capacity in functional peptide-based therapeutics, which are in the spotlight as new treatment alternatives for various diseases, and also presents verified sequences and targets that can increase chemical and pharmacological value.
Article
Full-text available
Yeni koronavius hastalığı "COVID-19" dünyada birçok insanın hayatını tehdit eden ciddi bir halk sağlığı haline gelmiştir. Bu yeni virüsten korunmada ve hastalığın seyrini iyileştirmede, SARS-CoV-2 için halen spesifik bir tedavinin olmadığı da göz önüne alındığında, bağışıklık sistemin aktif ve güçlü tutulması önemlidir. Bağışıklık sisteminde bağırsakların ve içerisindeki mikroorganizmaların çeşitliliği ile oluşturduğu mikrobiyotanın oldukça önemli yer edinmektedir. Aynı bu yararlı patojenlerin denge halinde sürdürülmesi bağışıklığı artırmaktadır. Epidemiyolojik ve deneysel çalışmalar beslenme, bağışıklık sistemi ve enfeksiyon üçgeninde yiyecek çeşitliliğinin önemine işaret etmektedir. COVID-19 salgını, sağlıklı yaşam, sağlığın korunması, güçlendirme ve bağışıklık sistemi bileşenlerinin gibi profilaktik yaklaşımların önemini bir kez daha göstermiştir. Mikrobiyota kaynaklarının nasıl elde edilip kullanılacağı, mikrobiyota düzenleyici-destekleyici ürünlerin uygulamasının standardizasyonu ve beslenmenin düzenlenmesinin tedavideki rolü gibi pek çok konuda ileri araştırmalara ihtiyaç vardır.
Article
Full-text available
Numerous treatments have been developed to promote wound healing based on current understandings of the healing process. Hemorrhaging, clotting, and associated inflammation regulate early wound healing. We investigated treatment with a virus-derived immune modulating serine protease inhibitor (SERPIN), Serp-1, which inhibits thrombolytic proteases and inflammation, in a mouse excisional wound model. Saline or recombinant Serp-1 were applied directly to wounds as single doses of 1 μg or 2 µg or as two 1 µg boluses. A chitosan-collagen hydrogel was also tested for Serp-1 delivery. Wound size was measured daily for 15 days and scarring assessed by Masson’s trichrome, Herovici’s staining, and immune cell dynamics and angiogenesis by immunohistochemistry. Serp-1 treatment significantly accelerated wound healing, but was blocked by urokinase-type plasminogen activator (uPAR) antibody. Repeated dosing at a lower concentration was more effective than single high-dose serpin. A single application of Serp-1-loaded chitosan-collagen hydrogel was as effective as repeated aqueous Serp-1 dosing. Serp-1 treatment of wounds increased arginase-1-expressing M2-polarized macrophage counts and periwound angiogenesis in the wound bed. Collagen staining also demonstrated that Serp-1 improves collagen maturation and organization at the wound site. Serp-1 has potential as a safe and effective immune modulating treatment that targets thrombolytic proteases, accelerating healing and reducing scar in deep cutaneous wounds.
Article
Full-text available
Spinal cord injury (SCI)-initiated inflammation was treated with anti-inflammatory reagents. We compared local spinal cord or intraperitoneal infusion of two Myxoma virus derived immune modulating proteins, Serp-1 and M-T7, with dexamethasone (DEX). Hemorrhage and necrosis after SCI initiate a complex pathogenesis dominated by early, severe and highly destructive inflammatory macrophage infiltration. We examined sustained, 7-day, subdural infusion of either M-T7, a chemokine modulator or Serp-1, a plasminogen activator and factor inhibitor. Mature male rats had epidural balloon crush SCI and sustained subdural infusion of Serp-1, M-T7, DEX or saline for 7 days via the osmotic pump. A separate group of rats with SCI had intra-peritoneal infusion. Clinical evaluation included endpoint monitoring with body weight, hemorrhagic cystitis and bilateral toe pinch response. Sections of the spinal cord were analyzed histologically and macrophage numbers counted by standardized protocol in the cavity of injury (COI). While the rats administered DEX demonstrated substantial body weight loss, dehydration and dermal atrophy consistent with steroid toxicity, rats infused with Serp-1 and M-T7 had no toxicity. Serp-1 improved withdrawal responses. Subdural infusion of Serp-1, M-T7 and DEX significantly reduced numbers of phagocytic, CD68-positive macrophages. With intraperitoneal infusion only M-T7 reduced macrophage counts, Serp-1 showed only a trend. Local infusion of highly active immune modulating proteins; Serp-1 and M-T7, targeting serine protease and chemokine pathways demonstrated excellent potential for neuroprotection after severe SCI in a rat model, without adverse side effects. Sustained subdural infusion offers an alternative route of administration for treatment of SCI.
Article
Full-text available
Inflammatory bowel disease (IBD) and Clostridium difficile infection cause gastrointestinal (GI) distension and, in severe cases, toxic megacolon with risk of perforation and death. Herpesviruses have been linked to severe GI dilatation. MHV-68 is a model for human gamma herpesvirus infection inducing GI dilatation in interleukin-10 (IL-10)-deficient mice but is benign in wildtype mice. MHV-68 also causes lethal vasculitis and pulmonary hemorrhage in interferon gamma receptor-deficient (IFNγR −/−) mice, but GI dilatation has not been reported. In prior work the Myxomavirus-derived anti-inflammatory serpin, Serp-1, improved survival, reducing vasculitis and pulmonary hemorrhage in MHV-68-infected IFNγR −/− mice with significantly increased IL-10. IL-10 has been investigated as treatment for GI dilatation with variable efficacy. We report here that MHV-68 infection produces severe GI dilatation with inflammation and gut wall degradation in 28% of INFγR-/-mice. Macrophage invasion and smooth muscle degradation were accompanied by decreased concentrations of T helper (Th2), B, monocyte, and dendritic cells. Plasma and spleen IL-10 were significantly reduced in mice with GI dilatation, while interleukin-1 beta (IL-1β), IL-6, tumor necrosis factor alpha (TNFα) and INFγ increased. Treatment of gamma herpesvirus-infected mice with exogenous IL-10 prevents severe GI inflammation and dilatation, suggesting benefit for herpesvirus-induced dilatation.
Article
Full-text available
In the past decade, an exciting realization has been that diverse liver diseases - ranging from nonalcoholic steatohepatitis, alcoholic steatohepatitis and cirrhosis to hepatocellular carcinoma - fall along a spectrum. Work on the biology of the gut-liver axis has assisted in understanding the basic biology of both alcoholic fatty liver disease and nonalcoholic fatty liver disease (NAFLD). Of immense importance is the advancement in understanding the role of the microbiome, driven by high-throughput DNA sequencing and improved computational techniques that enable the complexity of the microbiome to be interrogated, together with improved experimental designs. Here, we review gut-liver communications in liver disease, exploring the molecular, genetic and microbiome relationships and discussing prospects for exploiting the microbiome to determine liver disease stage and to predict the effects of pharmaceutical, dietary and other interventions at a population and individual level. Although much work remains to be done in understanding the relationship between the microbiome and liver disease, rapid progress towards clinical applications is being made, especially in study designs that complement human intervention studies with mechanistic work in mice that have been humanized in multiple respects, including the genetic, immunological and microbiome characteristics of individual patients. These 'avatar mice' could be especially useful for guiding new microbiome-based or microbiome-informed therapies.
Article
Rift Valley fever virus (RVFV) is a zoonotic disease of livestock that causes several clinical outcomes in people including febrile disease, hemorrhagic fever, and/or encephalitis. After aerosol infection with RVFV, Lewis rats develop lethal encephalitic disease, and we use this as a model for studying disease mechanisms of RVFV infection in the brain. Permeability of the brain vasculature in relation to virus invasion and replication is not known. Here, we found that vascular permeability in the brain occurred late in the course of infection and corresponded temporally to expression of matrix metalloproteinase-9 (MMP-9). Virus replication was ongoing within the central nervous system for several days prior to detectable vascular leakage. Based on this study, vascular permeability was not required for entry of RVFV into the brain of rats. Prevention of vascular leakage late in infection may be an important component for prevention of lethal neurological disease in the rat model.
Chapter
Serpins have a wide range of functions in regulation of serine proteases in the thrombotic cascade and in immune responses, representing up to 2–10% of circulating proteins in the blood. Selected serpins also have cross-class inhibitory actions for cysteine proteases in inflammasome and apoptosis pathways. The arterial and venous systems transport blood throughout the mammalian body representing a central site for interactions between coagulation proteases and circulating blood cells (immune cells) and target tissues, a very extensive and complex interaction. While analysis of serpin functions in vitro in kinetics or gel shift assays or in tissue culture provides very necessary information on molecular mechanisms, the penultimate assessment of biological or physiological functions and efficacy for serpins as therapeutics requires study in vivo in whole animal models (some also consider cell culture to be an in vivo approach).
Chapter
Serine protease inhibitors, serpins, can have profound effects on many systems in the body including immune defense systems. The microbiome, specifically the gut and lung bacterial microbiota, is now known, under some conditions, to alter immune defenses. DNA library preparation for microbiome studies is a procedure that prepares microbial genomic DNA to be sequenced in next-generation sequencing platforms. The construction involves a PCR reaction that will amplify the 16S rRNA gene and will incorporate a specific index and adaptors to the fragments. After confirmation of the product amplification by gel electrophoresis, samples will be later normalized to the same DNA amount of 240 ng. Final concentration of the library is validated by quantitative PCR (qPCR).
Chapter
Serine protease inhibitors, or serpins, function as central regulators for many vital processes in the mammalian body, maintaining homeostasis for clot formation and breakdown, immune responses, lung function, and hormone or central nervous system activity, among many others. When serine protease activity or serpin-mediated regulation becomes unbalanced or dysfunctional, then severe disease states and pathogenesis can ensue. With serpinopathies, genetic mutations lead to inactive serpins or protein aggregation with loss of function. With other disorders, such as sepsis, atherosclerosis, cancer, obesity, and the metabolic syndrome, the thrombotic and thrombolytic cascades and/or inflammatory responses become unbalanced, with excess bleeding and clotting and upregulation of adverse immune responses. Returning overall balance can be engineered through introduction of a beneficial serpin replacement as a therapeutic or through blockade of serpins that are detrimental. Several drugs have been developed and are currently in use and/or in development both to replace dysfunctional serpins and to block adverse effects induced by aberrant protease or serpin actions.
Article
Systemic vasculitis are heterogeneous, complex and disabling disorders. Following the previous annual reviews of this series, this paper gives a brief overview on current knowledge about recent literature on small- and large-vessel systemic vasculitis, with a specific focus on pathogenetic and clinical aspects, novel possible disease-related biomarkers and current and future therapies that are in the pipeline.