scholarly journals The Bacterial Microbiota Promotes Normal Hematopoiesis Via Interferon Alpha and NOD1 Signaling Pathways

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 1080-1080
Author(s):  
Hannah Yan ◽  
Forrest C Walker ◽  
Hyojeong Han ◽  
Megan T Baldridge ◽  
Katherine Y. King

Abstract Long-term antibiotic therapy is associated with hematological side effects such as neutropenia and anemia. Our lab and others have shown that long-term antibiotic treatment in mice leads to bone marrow suppression and agranulocytosis in mice through depletion of the commensal bacteria. Our work further showed that Stat1-deficient mice phenocopy the bone marrow suppression phenotype of antibiotic-treated mice, suggesting that commensal microbiota mediate hematopoiesis via Stat1 signaling. However, the upstream actors of this pathway and the bacterial mediators required for commensal microbiota regulation of normal hematopoiesis still remain poorly understood. Such knowledge will be essential for understanding how to treat antibiotic-associated cytopenias. We hypothesize that microbial products detected by host cells trigger STAT1 signaling to potentiate normal hematopoiesis. To identify the host cells that require Stat1 for microbiota-promoted hematopoiesis, we treated conditional Stat1 knock-out mice with two weeks of antibiotic therapy. Of the four conditional knock-out mice we evaluated (LepR-Cre, Villin-Cre, Vav-iCre, LysM-Cre), only the mice deficient in STAT1 in hematopoietic cells (Vav-iCre Stat1 fl/fl) phenocopied the bone marrow suppression of antibiotic-treated mice. Our data suggest that STAT1 signaling is necessary in non-myeloid hematopoietic cells, but not intestinal epithelial cells or mesenchymal stromal cells for microbiota-promoted hematopoiesis. Non-competitive transplantation of Stat1 -/- bone marrow into wild type mice validated these findings; mice lacking STAT1 only in hematopoietic tissues phenocopied the Vav-iCre mice, consistent with a specific role for microbiota-mediated STAT1 signaling in the hematopoietic compartment. To assess the upstream mediator of STAT1 signaling in this biological context, we treated interferon (IFN) receptor knock out mice with two weeks of antibiotics. Of the three types of IFN receptor knock-outs evaluated, only mice deficient in type I IFN signaling phenocopied the bone marrow suppression of antibiotic-treated mice. These findings suggest that type I IFN signaling, and not type II or III, was required for microbiota-dependent hematopoiesis. These results were validated by showing that the administration of pegylated-IFNα was sufficient to rescue the depletion of type I IFN-STAT1 signaling in antibiotic-treated mice. To determine the microbial signals that may potentiate hematopoiesis, we evaluated two microbial products that were previously shown to alter hematopoiesis and to activate type I IFN signaling (Iwamura et al. Blood 2017 & Steed et al. Science 2017). We discovered that oral administration of these commensal-derived products, the metabolite desaminotyrosine (DAT) or NOD1 ligand (NOD1L), a motif of peptidoglycan, were each sufficient to rescue the hematopoietic defects induced by antibiotics in mice. To test whether these products rescue hematopoiesis by activating STAT1 signaling, we attempted to rescue the hematopoietic defects in Stat1 -/- mice. These studies showed that NOD1L rescues granulocyte but not progenitor counts in Stat1 -/- mice, suggesting that NOD1 and type I IFN signaling work together at the progenitor level, but independently at the downstream myeloid progenitor level to promote granulopoiesis. Overall, our studies expand our understanding of the signaling pathways by which the microbiota promotes normal hematopoiesis and identify novel therapeutic agents that can be used to ameliorate antibiotic-induced BM suppression. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. SCI-29-SCI-29
Author(s):  
Joel S Greenberger

Abstract Four different combinations of double knock-out mouse strains have been created (SMAD3-/- Fancd2-/-) consisting of combinations of the individual knock-out strains on either the C57BL/6 or 129/Sv background. Sets of breeding experiments were carried out and the frequency of detection of double knock-out mice was significantly below the expected frequency of detecting DKO mice by breeding double heterozygotes (1 out of 16 or 6.25%). In fact, DKO mice were detected at a frequency ranging from 3.4% to 0.54%. Mice of all four DKO genotypes showed resistance of bone marrow hematopoietic progenitor cells to canonical TGF-β signaling, consistent with the SMAD3-/- parent. However, the predominant phenotype was that of the Fancd2-/- parent including reduced duration of hematopoiesis in long-term bone marrow cultures, reduced marrow stem cell competitive repopulation capacity, and retained mitomycin C and radiation sensitivity of bone marrow stromal cells. These mice should be a valuable resource for elucidating the bypass pathways involved in the reduced, but successful gestation of SMAD3-/- Fancd2-/- mice, and the interactions of the FA and TGF-β signaling pathways. Supported by the NIAID/NIH U19-A168021 and the Fanconi Anemia Research Fund. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 317-317
Author(s):  
Ingrid G Winkler ◽  
Valerie Barbier ◽  
Andrew C Perkins ◽  
John L. Magnani ◽  
Jean-Pierre Levesque

Abstract We have previously identified a novel function for the adhesion molecule E-selectin – awakening otherwise dormant hematopoietic stem cells (HSC) and inducing lineage commitment (Winkler et al., Nat Med 2012). Now we show that therapeutic blockade of E-selectin in vivo specifically augments the mobilisation of HSC with highest self-renewal potential following G-CSF administration, and markedly improves subsequent engraftment and reconstitution in mice. From these data we hypothesise that vascular E-selectin acts as a gatekeeper influencing activation of transmigrating HSC. Firstly we found that administration of mobilizing doses of G-CSF increased the level of E-selectin expressed on the surface of bone marrow (BM) endothelial cells. To determine whether E-selectin influenced HSC mobilisation, we then compared G-CSF-mediated mobilisation in wildtype and E-selectin knock-out mice. We found that although absence of E-selectin did not significantly alter the number of phenotypic HSC or colony-forming cells mobilized into the blood following G-CSF, the absence of E-selectin in mobilised mice did increase the subsequent engraftment and reconstitution potential of mobilised blood analysed by competitive repopulation transplant assays. Next we investigated whether this beneficial effect could similarly be achieved by transient E-selectin blockade using therapeutic doses of GMI-1271, a small synthetic mimetic that specifically blocks the binding of E-selectin to its receptors. Wild-type mice were administered human G-CSF alone (125µg/kg subcutaneously twice daily for 3 days) ± GMI-1271 injections (20 mg/kg BID). The number of mobilized HSC were quantified by rigorous limiting-dilution transplantation of 0.3, 1, 5 or 20 µL mobilised blood in competition with 200,000 congenic BM cells into lethally-irradiated congenic recipients, to enable quantification of long-term reconstituting cells per mL of mobilised blood by Poisson’s distribution. We found the mobilized blood of donor mice injected with both GMI-1271 and G-CSF, showed faster engraftment and 25-fold increased reconstitution potential over blood from G-CSF alone injected controls (p<0.0001). That is after 3 days of G-CSF injections, blood from G-CSF plus GMI-1271 injected mice contained 476 reconstitution units (RU) /mL compared to 20 RU/mL in blood from mice mobilised with G-CSF alone. (One reconstitution unit is the same reconstitution as 100,000 normal bone marrow cells; 95% CI was 200 - 500 RU/mL compared to 11 – 36 RU/mL in blood from G-CSF plus GMI-1271 treated mice, compared to G-CSF alone injected mice respectively). Surprisingly, this dramatic boost to reconstitution potential with GMI-1271 co-administration was not reflected in the numbers of phenotypic HSPC, or number of colony-forming cells mobilised per mL of blood. Together these findings suggest that transient interactions between HSC extravasating across the BM endothelium during G-CSF administration, with E-selectin expressed on the BM vasculature, may inadvertently compromise the reconstitution potential of up to 96% of harvested peripheral blood HSC, indicating an unexpected disadvantage with current HSC harvesting procedures. These data are consistent with the role of E-selectin in the bone marrow (to awaken otherwise dormant HSC) that we have previously reported, and also point the way forward to a simple remedy (administration of E-selectin antagonist together with G-CSF) during HSC mobilisation to improve short- and long-term engraftment thus accelerate recovery in transplant recipients. In conclusion we propose the novel concept of E-selectin as a vascular ‘gate-keeper’ dampening the potential of migratory HSC. Disclosures Winkler: FibroGen Inc.: Research Funding. Magnani:GlycoMimetics Inc.: Employment, Equity Ownership, Membership on an entity's Board of Directors or advisory committees.


2021 ◽  
Vol 23 (Supplement_6) ◽  
pp. vi181-vi181
Author(s):  
Yuanfan Yang ◽  
Michael Brown ◽  
Kevin Stevenson ◽  
Giselle lopez ◽  
Reb Kornahrens ◽  
...  

Abstract Immunotherapy with polio:rhinovirus recombinant (PVSRIPO) has shown evidence of efficacy in a phase I clinical trial for recurrent GBM, resulting in durable radiographic responses and 21% long-term survival at 36 months. Ongoing research aims to enhance the clinical response rate by resolving the mechanisms of action and therapy resistance in vivo, thereby devising more effective therapies. Mouse glioma (CT2A) cells were intracranially implanted (day 0) in transgenic mice carrying poliovirus receptor CD155, and treated with intratumor PVSRIPO (5×105 pfu; day 6) to dissect early and late events following therapy. A blinded pathological review of 45 post-treatment tumors was performed. On day 8, a histological response, featured by tumor dissociation and shrinkage, with inflammation and microglia enrichment in the treated hemisphere, was common in PVSRIPO group (6/7) compared to controls (0/4). However, the response rate fell over time (7/12 on day 12; 1/7 on day 15) and the therapy was overcome by aggressive tumor regrowth. RNAseq was performed and Gene Set Enrichment Analysis of the tumor microenvironment revealed an acute type-I interferon (IFN)-related inflammation, correlating with the histological findings of profound proinflammatory engagement of microglia (Iba1+) widespread in the treated hemisphere. Microglia proliferation (Ki67+) was observed in the treated hemisphere, likely resulting from PVSRIPO infection, in CT2A and B16 intracranial models. This suggests an association of adaptive antitumor immunity—elicited by immediate intratumor type-I IFN-dominant inflammation—with tumor regression. Thus, buttressing type-I IFN directed antitumor CD8+T cell immunity, e.g. with blockade of the PD1:PD-L1 immune checkpoint, might contribute to tumor remission. Indeed, combination therapy with αPD-L1 antibody in the CT2A model showed longer median survival and higher long-term remission rate compared to monotherapy alone; CD8 T cell depletion can completely abrogate this efficacy with this therapy combination, confirming the role of anti-tumor immunity in this approach.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 53-53
Author(s):  
Harini Nivarthi ◽  
Andrea Majoros ◽  
Eva Hug ◽  
Ruochen Jia ◽  
Sarada Achyutuni ◽  
...  

The curative potential of Type I interferons for patients suffering from Myeloproliferative Neoplasms (MPNs) has been reported and these are the only class of drugs that can lead to reduction of the mutant allelic burden in patients. However, modelling IFN treatment in mice has been challenging. Here, we report the use of murine pegylated IFNα (murine ropeginterferon-a, mRopeg) developed by PharmaEssentia (Taipei, Taiwan) to model IFN treatment in transgenic MPN mouse models. We started treating JAK2V617Ff/+;vavCre and control vavCre mice (n=6-8) with PBS or mRopeg (600 ng/mouse/week), by subcutaneous injections from the time they were 4 weeks old. The mice were bled every 2 weeks from the facial vein and the blood parameters were monitored. We observed significant normalization of platelet and WBC counts in Jak2-V617F fl/+ vavCre mice to wild type levels. No effect on hematocrit and hemoglobin level was observed in the Jak2-V617F fl/+ vavCre mice. VavCre control animals showed no sign of negative effect such as cytopenia during the entire treatment course. We observed a highly significant prolongation of the survival of mRopeg treated JAK2V617Ff/+;vavCre mice over a duration of 80 days of treatment. While all the PBS treated JAK2V617Ff/+;vavCre mice died within 60 days, all the mRopeg treated mice were still alive till the end of the treatment duration. We also generated a novel transgenic mouse model that conditionally expresses hybrid mutant CALR protein (murine exons 1-8 and human CALR del52 exon9) from the endogenous murine Calr locus. We bred them into vavCre background (in both heterozyhous and homozygous states) to induce expression of CALR-del52 in hematopoietic cells. Upon Cre recombinase expression, the endogenous murine exon 9 is replaced by the human del52 exon 9 and the expression of the humanized Calr-del52 oncoprotein is detectable by Western blot analysis using mutant CALR specific antibodies. Calr-del52 animals develop an essential thrombocythemia (ET) like phenotype when expressed in a heterozygous state with elevated number of hematopoietic stem cells and megakaryocytes in the bone marrow. In the homozygous state, the thrombocythemia is more severe with splenomegaly and older animals show anemia with increased WBC. Bone marrow histology shows megakaryocytic hyperplasia with no sign of fibrosis up to age of one year. We treated a cohort of animals with 600 ng mRopeg/PBS once a week for 4 weeks. Peripheral blood counts were determined at baseline and at regular intervals during treatment. At the end of treatment, mice were sacrificed, and splenic and bone marrow cells were immunophenotyped and quantified by FACS. We observed correction of thrombocythemia in the homozygous Calr-del52 mice but no unspecific decrease of platelet count in the vavCre mRopeg treated animals. We observed significant specific reduction of the long-term hematopoietic stem cells (LT-HSCs/fraction A) in homozygous CALR-del52 mice. In conclusion, Type I IFN treatment significantly reduces platelet counts to normal levels in both JAK2 and CALR mutant driven MPN mouse models. The prolongation of survival of JAK2V617F transgenic mice upon Type I IFN treatment is particularly remarkable; as no survival data is reported until now in any clinical trials or other animal models. Further experiments are required to understand the mechanism of action of this phenomenon. Disclosures No relevant conflicts of interest to declare.


2016 ◽  
Vol 12 (10) ◽  
pp. e1005975 ◽  
Author(s):  
Emily Spaulding ◽  
David Fooksman ◽  
Jamie M. Moore ◽  
Alex Saidi ◽  
Catherine M. Feintuch ◽  
...  

2012 ◽  
Vol 26 (S1) ◽  
Author(s):  
Stephen James Hickner ◽  
Mariana Angoa-Perez ◽  
Donald M Kuhn ◽  
Jason H Mateika

2020 ◽  
Vol 127 (Suppl_1) ◽  
Author(s):  
Claire Zhang ◽  
David M Calcagno ◽  
Avinash Toomu ◽  
Kenneth M Huang ◽  
Zhenxing Fu ◽  
...  

Myocardial infarction (MI) elicits a rapid and vigorous reaction from the bone marrow hematopoietic compartment, inducing a massive efflux of myeloid first responders into the bloodstream. These cells traffic to the infarct, where they mediate cardiac remodeling and repair through inflammatory signaling and recruitment of additional immune cells to the injured myocardium. A hyperinflammatory myeloid compartment, as is produced by mutations in epigenetic regulator TET2 associated with clonal hematopoiesis, can thus drive adverse cardiac remodeling after MI and accelerate progression to heart failure. Whether loss of TET2 alters the transcriptional landscape of MI-induced myelopoiesis remains to be investigated in an unbiased fashion. Here, we performed single-cell RNA sequencing of >16,000 bone marrow myeloid cells isolated from wild-type and Tet2 -/- mice after MI to characterize the emergency hematopoietic response in the presence and absence of TET2. Our data capture distinct transitional states of myeloid lineage commitment and maturation, originating from myeloid progenitors and progressing along divergent granulocytic and monocytic differentiation trajectories. Additionally, we delineate a subpopulation of interferon (IFN)-activated myeloid progenitors, monocytes, and neutrophils characterized by the concerted upregulation of various Type I IFN-stimulated genes, and find the fraction of IFN-activated cells, as well as the degree of activation, to be markedly higher in Tet2 -/- mice. We have previously described activation of this pathway after MI in mice, and demonstrated cardioprotective effects of its genetic or pharmacological inhibition. Our findings reveal heightened activation of the antiviral Type I interferon response among bone marrow myeloid cells of Tet2 -/- mice during MI-induced emergency hematopoiesis. This highlights IFN signaling as a potential candidate driver of cardiovascular pathologies (including atherosclerosis, myocardial infarction, and heart failure) associated with TET2-mediated clonal hematopoiesis. Further studies are necessary to investigate whether Tet2 -/- mice exhibit enhanced response to blockade of Type I IFN signaling after MI, and to determine whether myeloid cells of TET2 -mutant humans are similarly activated.


2020 ◽  
Vol 117 (35) ◽  
pp. 21519-21526 ◽  
Author(s):  
Domonica N. Powell ◽  
Alyson Swimm ◽  
Robert Sonowal ◽  
Alexis Bretin ◽  
Andrew T. Gewirtz ◽  
...  

The intestinal epithelium is a highly dynamic structure that rejuvenates in response to acute stressors and can undergo alterations in cellular composition as animals age. The microbiota, acting via secreted factors related to indole, appear to regulate the sensitivity of the epithelium to stressors and promote epithelial repair via IL-22 and type I IFN signaling. As animals age, the cellular composition of the intestinal epithelium changes, resulting in a decreased proportion of goblet cells in the colon. We show that colonization of young or geriatric mice with bacteria that secrete indoles and various derivatives or administration of the indole derivative indole-3 aldehyde increases proliferation of epithelial cells and promotes goblet cell differentiation, reversing an effect of aging. To induce goblet cell differentiation, indole acts via the xenobiotic aryl hydrocarbon receptor to increase expression of the cytokine IL-10. However, the effects of indoles on goblet cells do not depend on type I IFN or on IL-22 signaling, pathways responsible for protection against acute stressors. Thus, indoles derived from the commensal microbiota regulate intestinal homeostasis, especially during aging, via mechanisms distinct from those used during responses to acute stressors. Indoles may have utility as an intervention to limit the decline of barrier integrity and the resulting systemic inflammation that occurs with aging.


Proceedings ◽  
2020 ◽  
Vol 50 (1) ◽  
pp. 74
Author(s):  
Wai-Yan Lui ◽  
Sonia Jangra ◽  
Kit-San Yuen ◽  
Michael George Botelho ◽  
Dong-Yan Jin

The Epstein–Barr virus (EBV) successfully infects 95% of all adults but causes Burkitt’s lymphoma, Hodgkin’s lymphoma, gastric carcinoma, nasopharyngeal carcinoma or other malignancies in only a small subset of infected individuals. The virus must have developed effective viral countermeasures to evade host innate immunity. In this study, we performed functional screens to identify EBV-encoded interferon (IFN) antagonists. Several tegument proteins were found to be potent suppressors of IFN production and/or signaling. The large tegument protein and deubiquitinase BPLF1 antagonized type I IFN production induced by DNA sensors cGAS and STING or RNA sensors RIG-I and MAVS. BPLF1’s ability to suppress innate immune signaling required its deubiquitinase activity. BPLF1 functioned as a catalytically active deubiquitinase for both K63- and K48-linked ubiquitin chains on STING and TBK1, with no ubiquitin linkage specificity. Induced expression of BPLF1 in EBV-infected cells through CRISPRa led to effective suppression of innate DNA and RNA sensing. Another EBV tegument protein, BGLF2, was found to suppress JAK-STAT signaling. This suppression was ascribed to more pronounced K48-linked polyubiquitination and proteasomal degradation of BGLF2-associated STAT2. In addition, BGLF2 also recruited tyrosine phosphatase SHP1 to inhibit tyrosine phosphorylation of JAK1 and STAT1. A BGLF2-deficient EBV activated type I IFN signaling more robustly. Taken together, we characterized the IFN antagonism of EBV tegument proteins BPLF1 and BGLF2, which modulate ubiquitination of key transducer proteins to counteract type I IFN production and signaling in host cells. Supported by HMRF 17160822, HMRF 18170942, and RGC C7027-16G.


Sign in / Sign up

Export Citation Format

Share Document