scholarly journals Microbial-Derived Metabolites Induce Epithelial Recovery Via the Sting Pathway in Mice and Men and Protect from Graft-Versus-Host Disease

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 87-87
Author(s):  
Erik Thiele Orberg ◽  
Sascha Göttert ◽  
Andreas Hiergeist ◽  
Elisabeth Meedt ◽  
Karin Kleigrewe ◽  
...  

Abstract Background: Graft-versus-host disease (GVHD) is a dreaded complication after allogeneic stem-cell transplantation (ASCT). Previously, we and others showed that activation of type I interferon (IFN-I) inducing pathways such as RIG-I/MAVS or cGAS-STING can promote the integrity of the intestinal barrier and limit GVHD (Fischer et al., Sci Transl Med, 2017). However, the signals that drive these protective IFN-I responses are poorly understood. Commensal microbiota can (i) have distant effects on immune responses through modulation of IFN-I signaling (Steed et al, Science, 2017; Swimm et al, Blood, 2018) and (ii) predict mortality in ASCT patients (Peled et al., N Engl J Med, 2020). We hypothesized that microbiota-derived products such as microbial metabolites engage IFN-I signaling in immune and non-immune cells poising them for induction of protective responses. We established a prospective, multi-centric clinical study in patients newly diagnosed with acute leukemia and performed longitudinal stool sampling to track changes in microbial community composition and metabolites expression levels. Submitted as a separate abstract to ASH 2021 (Orberg & Meedt et al.), we show that patients with high metabolite expression going into ASCT are less likely to develop GVHD. In this study, we translate our clinical observations to mouse models of acute GVHD and human and mouse intestinal organoids to uncover the molecular mechanisms via which metabolites protect the intestinal barrier during ASCT. Methods: Stool samples from ASCT patients were obtained in Munich and at Regensburg in accordance to IRB-approved study protocols. Patients were sampled at initial diagnosis (Dx), prior to conditioning and weekly after ASCT up to day 28. We analyzed samples by 16S rRNA sequencing and mass spectrometry to obtain a complete picture of microbiome composition and function. Next, we tested metabolites which we detected in patients (desaminotyrosine [DAT], indole-3-carboxaldehyde [ICA]) as treatment in preclinical models in ASCT and GVHD mouse models. Outcomes were assessed by a novel organoid recovery assay in addition to established read-outs. To obtain a mechanistic understanding of the signaling pathways involved, we stimulated WT or IFN-I-signaling-impaired mouse (incl. STING -/-, MAVS -/-, IFNαR -/-) as well as human intestinal crypt-derived organoids with metabolites. Results: Here, we present a 64-year-old female patient diagnosed with AML who received a 9/10 HLA-matched ASCT. At day 7, i.v. antibiotics were started due to fever and Enterococcus bacteraemia. At day 15, the patient developed skin and GI GVHD (Glucksberg III). At the timepoint initial diagnosis (Dx), i.e. the timepoint when we diagnosed AML but before therapy was initiated, we detected rich alpha diversity (Panel 1a) in the patient's stool. Flavonifractor plautii, a producer of the metabolite DAT, was detectable (Panel 1b). We observed high-level expression of metabolites including short-chain fatty acids, DAT, ICA and secondary bile acids (Panel 1c). Following ASCT, and especially at the early time-points day 0 and day 7, alpha diversity and metabolite expression declined drastically. We confirmed this trend in our multi-centric cohort of ASCT patients by comparing levels of DAT and ICA sampled at admission to the transplantation ward (Conditioning) versus at clinical diagnosis of GVHD: in patients with GVHD, metabolite levels were drastically reduced (Panel 2). Next, we prophylactically administered metabolites in a major mismatch mouse ASCT model. Metabolite-treated mice showed significantly showed better outcomes in our organoid recovery assay, which measures the ability of intestinal stem cells to recover after allogeneic injury (Panel 3). This effect that was abrogated in STING -/- recipients. Metabolite stimulation of mouse small intestinal organoids promoted organoid numbers and size, and required intact STING signaling (Panel 4a). Human colon organoids also responded to DAT and ICA, however the metabolite effect was lost when co-administered with the STING-inhibitor H151. Conclusions: We identify that microbial-derived metabolites detected in patients can engage the STING pathway in humans and mice to confer resistance from immune damage. Thus, prophylactic administration of metabolite cocktails or bacterial consortia that can produce these metabolites may reduce occurrence of GVHD in ASCT patients. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.

2021 ◽  
Vol 30 ◽  
pp. 096368972110337
Author(s):  
Xi Sun ◽  
Qiaomei He ◽  
Jun Yang ◽  
Andi Wang ◽  
Fang Zhang ◽  
...  

Acute graft-versus-host disease (aGVHD) is one of the most common complications of allogeneic hematopoietic stem cell transplantation (allo-HSCT). Janus kinase (JAK) inhibitors are considered as reliable and promising agents for patients with aGVHD. The prophylactic and therapeutic effects of SHR0302, a novel JAK inhibitor, were evaluated in aGVHD mouse models. The overall survival (OS), progression-free survival (PFS), bodyweight of mice, GVHD scores were observed and recorded. The bone marrow and spleen samples of diseased model mice or peripheral blood of patients were analyzed. SHR0302 could prevent and reverse aGVHD in mouse models with preserving graft-versus-tumor effect. Functionally, SHR0302 improved the OS and PFS, restored bodyweight, reduced GVHD scores, and reduced immune cells infiltrated in target tissues. SHR0302 treatment also enhanced the hematopoietic reconstruction compared to the control group. Mechanistically, our results suggested that SHR0302 could inhibit the activation of T cells and modulate the differentiation of helper T (Th) cells by reducing Th1 and increasing regulatory T (Treg) cells. In addition, SHR0302 decreased the expression of chemokine receptor CXCR3 on donor T cells and the secretion of cytokines or chemokines including interleukin (IL)-6, interferon γ (IFN-γ), tumor necrosis factor α (TNF-α), CXCL10, etc. thereby destroying the IFN-γ/CXCR3/CXCL10 axis which promotes the progression of GVHD. Besides, SHR0302 decreased the phosphorylation of JAK and its downstream STATs, AKT and ERK1/2, which ultimately regulated the activation, proliferation, and differentiation of lymphocytes. Experiments on primary cells from aGVHD patients also confirmed the results. In summary, our results indicated that JAK inhibitor SHR0302 might be used as a novel agent for patients with aGVHD.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 5075-5075
Author(s):  
Jan-Erik Johansson ◽  
Tor Ekman

Abstract Background Graft-versus-host disease (GVHD) is the primary complication of allogeneic, haemopoietic, stem-cell transplantation (HSCT). From murine models, it has been revealed that gut toxicity increases the translocation of inflammatory mediators through the impaired intestinal barrier, which aggravates systemic GVHD. In patients, the intestinal barrier is disrupted after myeloablative conditioning but preserved after reduced intensity conditioning (RIC). The present study investigates, in a clinical situation, whether the severity of acute GVHD depends on intestinal-barrier function. Methods In 38 patients (21 myeloablative, 17 RIC), intestinal permeability was assessed before transplant and during the transplantation course (day −1 to +14). Blood levels of cytokines were assessed in parallel. Results Intestinal barrier integrity was preserved in RIC patients but disrupted in myeloablatively conditioned on day 4 (p = 0.0091) and on day 7 (p = 0.0014) compared with RIC patients. Patients with mild, acute GVHD (grades 0–1) had a significantly better preserved intestinal-barrier function (p = 0.042 on day 4) and lower TNF-a levels (p = 0.013 and 0.025 on day −1 and day +7 respectively), compared with patients with more pronounced GVHD (grades 2–4). Conclusion Intestinal barrier function predicts the severity of acute GVHD in patients and gives support to the assumption that gut protective strategies may diminish acute GVHD.


PLoS ONE ◽  
2017 ◽  
Vol 12 (8) ◽  
pp. e0183284 ◽  
Author(s):  
Gideon Steinbach ◽  
David M. Hockenbery ◽  
Gerwin Huls ◽  
Terry Furlong ◽  
David Myerson ◽  
...  

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3207-3207
Author(s):  
Erik Thiele Orberg ◽  
Sascha Göttert ◽  
Hendrik Poeck

Background: Graft-versus-host disease (GVHD) is a dreaded complication after stem-cell transplantation (SCT). Standard treatment relies on immunosuppressants but is associated with an increased risk of infection and relapse. Up 50% of patients develop steroid-refractory GVHD, with dismal impact on SCT outcomes. New studies in mice suggest that induction of type-I interferon (IFN-I) signalling or activation of IFN-I inducing pathways such as cGAS/STING or RIG-I/MAVS can promote gut barrier integrity and limit GVHD. However, the endogenous ligands that drive this "protective" IFN-I response are poorly defined. Recent data in mice and humans suggest that microbial-derived metabolites such as small-chain fatty acids or indoles can decrease GHVD mortality. Here, we describe IFN-I inducing metabolites that improve outcomes in mouse models of gut epithelial damage and acute GVHD via a mechanism dependent on STING signalling. Methods: To investigate which cell types mediate protection and how, we generated intestinal organoids and antigen presenting cells (APC) from bone marrow of WT or genetically deficient mice (MAVS-/-, STING-/-, IFNAR-/-) under steady state conditions versus chemotherapy, total body irradiation and after allogeneic SCT in the presence or absence of microbial metabolites. Analysis was performed by microscopy, immunoblotting, qPCR, ELISA and flow cytometry. Outcomes of gut-injured mice were assessed by clinical scoring, histopathology, flow cytometry and organoid recovery. Results: Metabolite treatment promoted regeneration of intestinal organoids as assayed by organoid numbers as well as proliferation. These effects were dependent on IFN-I and STING signalling. In addition, we found activated pro-inflammatory NFkB signalling and decreased apoptosis as evidenced by reduced caspase-3 cleavage. In APCs, IFN-I responses were enhanced in the presence of metabolites including increased IFN-β production and upregulation of IFN stimulated genes. Metabolite-treated mice showed improved recovery of intestinal stem cells following gut injury by chemotherapy and irradiation and increased survival in acute GVHD. Conclusions: Our findings uncover a mechanism by which microbial metabolites amplify IFN-I signals, promote tissue regeneration and thereby prevent allo-activation and GVHD. We show for the first time that these IFN-I inducing metabolites are protective in diverse models of gut damage -- from chemotherapy and radiation to immune-mediated damage -- via a STING-dependent mechanism. Perhaps the poor prognosis of GVHD patients exhibiting a loss of microbiota diversity can be explained in part by an absence of "protective" metabolites able to amplify critical IFN signals. We are currently studying whether metabolite levels correlate with severity and outcome of GVHD in humans. Figure Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Vol 10 (11) ◽  
pp. 2508
Author(s):  
Martyna Tyszka ◽  
Jarosław Biliński ◽  
Grzegorz Władysław Basak

The intestinal barrier consists of an epithelial lining covered with specialized mucus inhabited by intestinal microbiota. An intact gut barrier ensures a resistance to bacteria and toxins translocation. On the other hand, gut permeability allows the absorption of essential nutrients, fluids and ions. This balance is achieved only by the complex structure and functional characteristics of the intestinal barrier. Allogenic hematopoietic cell transplantation remains the only curative treatment for many hematological diseases, but its application is limited because of possible transplant-related mortality mainly due to graft-versus-host disease and infectious complications. The intestinal barrier has been extensively studied in recent years as the primary site of graft-versus-host disease initiation and propagation. In the present review, we focused on the physiological structure and function of the gut barrier and the evidence of how the disruption of the gut barrier and increased intestinal permeability affects transplant recipients. Finally, therapeutic strategies aiming at intestinal barrier protection with a special focus on microbiome preservation and restoration in the allogenic hematopoietic cell transplantation setting are discussed.


2014 ◽  
Vol 6 (243) ◽  
pp. 243ra87-243ra87 ◽  
Author(s):  
S. C. Nalle ◽  
H. Aimee Kwak ◽  
K. L. Edelblum ◽  
N. E. Joseph ◽  
G. Singh ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document