FG-4592 alleviates Radiation-Induced Intestinal Injury by facilitating recovery of intestinal stem cell and reducing damage of intestinal epithelial

Author(s):  
Penglin Xia ◽  
Yajie Yang ◽  
Ruling Liu ◽  
Zhenlan Feng ◽  
Yuhan Lin ◽  
...  
2018 ◽  
Vol 9 (1) ◽  
Author(s):  
Payel Bhanja ◽  
Andrew Norris ◽  
Pooja Gupta-Saraf ◽  
Andrew Hoover ◽  
Subhrajit Saha

2019 ◽  
Vol 10 (10) ◽  
Author(s):  
Bo Li ◽  
Carol Lee ◽  
Marissa Cadete ◽  
Haitao Zhu ◽  
Yuhki Koike ◽  
...  

Abstract Necrotizing enterocolitis (NEC) is a devastating neonatal disease characterized by acute intestinal injury. Intestinal stem cell (ISC) renewal is required for gut regeneration in response to acute injury. The Wnt/β-catenin pathway is essential for intestinal renewal and ISC maintenance. We found that ISC expression, Wnt activity and intestinal regeneration were all decreased in both mice with experimental NEC and in infants with acute active NEC. Moreover, intestinal organoids derived from NEC-injured intestine of both mice and humans failed to maintain proliferation and presented more differentiation. Administration of Wnt7b reversed these changes and promoted growth of intestinal organoids. Additionally, administration of exogenous Wnt7b rescued intestinal injury, restored ISC, and reestablished intestinal epithelial homeostasis in mice with NEC. Our findings demonstrate that during NEC, Wnt/β-catenin signaling is decreased, ISC activity is impaired, and intestinal regeneration is defective. Administration of Wnt resulted in the maintenance of intestinal epithelial homeostasis and avoidance of NEC intestinal injury.


2012 ◽  
Vol 302 (10) ◽  
pp. G1111-G1132 ◽  
Author(s):  
Laurianne Van Landeghem ◽  
M. Agostina Santoro ◽  
Adrienne E. Krebs ◽  
Amanda T. Mah ◽  
Jeffrey J. Dehmer ◽  
...  

Recent identification of intestinal epithelial stem cell (ISC) markers and development of ISC reporter mice permit visualization and isolation of regenerating ISCs after radiation to define their functional and molecular phenotypes. Previous studies in uninjured intestine of Sox9-EGFP reporter mice demonstrate that ISCs express low levels of Sox9-EGFP (Sox9-EGFP Low), whereas enteroendocrine cells (EEC) express high levels of Sox9-EGFP (Sox9-EGFP High). We hypothesized that Sox9-EGFP Low ISCs would expand after radiation, exhibit enhanced proliferative capacities, and adopt a distinct gene expression profile associated with rapid proliferation. Sox9-EGFP mice were given 14 Gy abdominal radiation and studied between days 3 and 9 postradiation. Radiation-induced changes in number, growth, and transcriptome of the different Sox9-EGFP cell populations were determined by histology, flow cytometry, in vitro culture assays, and microarray. Microarray confirmed that nonirradiated Sox9-EGFP Low cells are enriched for Lgr5 mRNA and mRNAs enriched in Lgr5-ISCs and identified additional putative ISC markers. Sox9-EGFP High cells were enriched for EEC markers, as well as Bmi1 and Hopx, which are putative markers of quiescent ISCs. Irradiation caused complete crypt loss, followed by expansion and hyperproliferation of Sox9-EGFP Low cells. From nonirradiated intestine, only Sox9-EGFP Low cells exhibited ISC characteristics of forming organoids in culture, whereas during regeneration both Sox9-EGFP Low and High cells formed organoids. Microarray demonstrated that regenerating Sox9-EGFP High cells exhibited transcriptomic changes linked to p53-signaling and ISC-like functions including DNA repair and reduced oxidative metabolism. These findings support a model in which Sox9-EGFP Low cells represent active ISCs, Sox9-EGFP High cells contain radiation-activatable cells with ISC characteristics, and both participate in crypt regeneration.


2020 ◽  
Vol 11 (1) ◽  
Author(s):  
Moritz Middelhoff ◽  
Henrik Nienhüser ◽  
Giovanni Valenti ◽  
H. Carlo Maurer ◽  
Yoku Hayakawa ◽  
...  

AbstractThe enteric neurotransmitter acetylcholine governs important intestinal epithelial secretory and immune functions through its actions on epithelial muscarinic Gq-coupled receptors such as M3R. Its role in the regulation of intestinal stem cell function and differentiation, however, has not been clarified. Here, we find that nonselective muscarinic receptor antagonism in mice as well as epithelial-specific ablation of M3R induces a selective expansion of DCLK1-positive tuft cells, suggesting a model of feedback inhibition. Cholinergic blockade reduces Lgr5-positive intestinal stem cell tracing and cell number. In contrast, Prox1-positive endocrine cells appear as primary sensors of cholinergic blockade inducing the expansion of tuft cells, which adopt an enteroendocrine phenotype and contribute to increased mucosal levels of acetylcholine. This compensatory mechanism is lost with acute irradiation injury, resulting in a paucity of tuft cells and acetylcholine production. Thus, enteroendocrine tuft cells appear essential to maintain epithelial homeostasis following modifications of the cholinergic intestinal niche.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Mara Martín-Alonso ◽  
Sharif Iqbal ◽  
Pia M. Vornewald ◽  
Håvard T. Lindholm ◽  
Mirjam J. Damen ◽  
...  

AbstractSmooth muscle is an essential component of the intestine, both to maintain its structure and produce peristaltic and segmentation movements. However, very little is known about other putative roles that smooth muscle cells may have. Here, we show that smooth muscle cells may be the dominant suppliers of BMP antagonists, which are niche factors essential for intestinal stem cell maintenance. Furthermore, muscle-derived factors render epithelium reparative and fetal-like, which includes heightened YAP activity. Mechanistically, we find that the membrane-bound matrix metalloproteinase MMP17, which is exclusively expressed by smooth muscle cells, is required for intestinal epithelial repair after inflammation- or irradiation-induced injury. Furthermore, we propose that MMP17 affects intestinal epithelial reprogramming after damage indirectly by cleaving diffusible factor(s) such as the matricellular protein PERIOSTIN. Together, we identify an important signaling axis that establishes a role for smooth muscle cells as modulators of intestinal epithelial regeneration and the intestinal stem cell niche.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1865-1865
Author(s):  
Rena Feinman ◽  
Iriana Colorado ◽  
Keyi Wang ◽  
Eugenia Dziopa ◽  
Leah Dziopa ◽  
...  

Abstract The failure to control both innate and adaptive immune responses in the gut has recently been implicated as a major pathogenic mechanism in the development of graft-versus-host disease (GVHD). Reduced oxygen availability in the intestine has been causally linked to gastrointestinal disease. During intestinal inflammation, increased metabolic activity of resident and infiltrating immune cells, bacteria and reduced blood flow may lead to a sharp decrease of oxygen, resulting in "inflammatory" hypoxia. The transcription factor family, hypoxia-inducible factor (HIF) originally discovered as a master regulator of the adaptive response to hypoxia, has recently emerged as a key regulator of the innate and adaptive immune responses. The HIF heterodimer consists of an oxygen-labile α subunit (HIFα) and a constitutively expressed HIF-1β subunit. Both HIF-1α and HIF-2α expression are markedly elevated in intestinal epithelial cells of patients with inflammatory bowel disease (IBD) and intestinal epithelial HIF-1 attenuates colitis in preclinical mouse models. Although HIF-2 has not been studied extensively in intestinal inflammation, it has emerged as a key regulator in intestinal iron homeostasis. Given that IBD and GVHD share many pathogenic mechanisms, we hypothesized that a sustained HIF response will protect the host intestinal epithelium from conditioning- and alloreactive T cell-induced gut damage. To determine the functional significance of intestinal epithelial HIF-1 and HIF-2 in gut GVHD, we generated conditional intestinal epithelial HIF-1α (HIF-1αΔIE) and HIF-2αΔIE vil-cre knockout (KO) mice on a C57BL/6 (B6) background lacking HIF-1α or HIF-2α in the host intestinal epithelium. Using a fully MHC mismatched B10.BR (H2k)→B6 (H2b) bone marrow transplant (BMT) model, loss of intestinal epithelial HIF-2 reduced the median survival time (43d) compared to wild-type (WT) recipients (58d, log-rank test, P < 0.005). Although intestinal epithelial HIF-1 deficiency shortened the median survival time (48.5d), it did not reach statistical difference. Loss of intestinal epithelial HIF-1 or HIF-2 worsened GVHD-induced histopathologic crypt damage compared to WT mice transplanted with T cell depleted bone marrow (BM) and enriched T cells (BM+T), 8d post- BMT. Pronounced subepithelial lifting, mucosal edema and sloughing were more evident in the villus tips of HIF-2αΔIE mice than HIF-1αΔIE mice. Hyperplastic crypts that are characteristic of regenerating crypts after radiation-induced damage were observed in Ki67-stained ileal/jejunal sections of WT mice post-BMT whereas fewer regenerating Ki67-labeled crypts were found in both HIF-1αΔIE and HIF-2αΔIE mice. In control T cell depleted BM groups (WT, HIF-1αΔIE and HIF-2αΔIE), Ki67+ -proliferating cells resided at the crypt base. Using quantitative real-time PCR analysis, we determined whether intestinal epithelial HIF-1 and HIF-2 differentially regulated the expression of Paneth cells and intestinal stem cell markers in the jejunum, 8d post-BMT. A 5-fold and 2-fold decrease in lysozyme (Lyz) mRNA levels occurred in WT (p<0.001) and HIF-1αΔIE BM+T mice (p<0.001) compared to their respective BM groups, 8d post-BMT. However, due to a 2-fold decrease in endogenous Lyz expression in HIF-1αΔIE (p<0.01) and HIF-2αΔIE (p<0.001) BM mice compared to WT BM mice, Lyz levels were not differentially changed among BM+T groups. Loss of either intestinal epithelial HIF-1 or HIF-2 reduced Reg3γ (p<0.05) and Sox9 (p<0.01) levels whereas only epithelial HIF-1 deficiency reduced Hes-1 by 2-fold (p<0.001) and prevented the recovery of the Lgr5 levels (p<0.01) compared to WT BM+T mice. In summary, we found that both intestinal epithelial HIF-1 and HIF-2 may protect the intestinal stem cell niche from GVHD-induced injury. Importantly, our results suggest that intestinal epithelial HIF-2 may be necessary in preventing GVHD-induced mortality. Disclosures No relevant conflicts of interest to declare.


2020 ◽  
Author(s):  
Mara Martín-Alonso ◽  
Håvard T. Lindholm ◽  
Sharif Iqbal ◽  
Pia Vornewald ◽  
Sigrid Hoel ◽  
...  

SUMMARYSmooth muscle is an essential component of the intestine, both to maintain its structure and produce peristaltic and segmentation movements. However, very little is known about other putative roles that smooth muscle may have. Here, we show that smooth muscle is the dominant supplier of BMP antagonists, which are niche factors that are essential for intestinal stem cell maintenance. Furthermore, muscle-derived factors can render epithelium reparative and fetal-like, which includes heightened YAP activity. Mechanistically, we find that the matrix metalloproteinase MMP17, which is exclusively expressed by smooth muscle, is required for intestinal epithelial repair after inflammation- or irradiation-induced injury. Furthermore, we provide evidence that MMP17 affects intestinal epithelial reprogramming indirectly by cleaving the matricellular protein PERIOSTIN, which itself is able to activate YAP. Together, we identify an important signaling axis that firmly establishes a role for smooth muscle as a modulator of intestinal epithelial regeneration and the intestinal stem cell niche.


2021 ◽  
Author(s):  
Jennifer Raisch ◽  
Anthony Côté-Biron ◽  
Marie-Josée Langlois ◽  
Caroline Leblanc ◽  
Nathalie Rivard

Abstract Intestinal epithelial self-renewal is tightly regulated by signaling pathways controlling stem cell proliferation, determination and differentiation. In particular, Wnt/β-catenin signaling controls crypt cell division and survival and is required for maintenance of the intestinal stem cell niche. Most colorectal cancers are also initiated by mutations activating the Wnt/β-catenin pathway. Wnt signals are transduced through Frizzled receptors and LRP5/LRP6 coreceptors to downregulate GSK3β activity, resulting in increased nuclear β-catenin. Herein, we explored if LRP6 expression is required for maintenance of intestinal homeostasis, regeneration and oncogenesis. Mice with an intestinal epithelial cell-specific deletion of Lrp6 (Lrp6IEC-KO) were generated and their phenotype analyzed. No difference in intestinal architecture or in proliferative and stem cell numbers was found in Lrp6IEC-KO mice in comparison to controls. Nevertheless, using ex vivo intestinal organoid cultures, we found that LRP6 expression was critical for crypt cell proliferation and stem cell maintenance. When exposed to dextran sodium sulfate, Lrp6IEC-KO mice developed more severe colitis than control mice. However, loss of LRP6 did not affect tumorigenesis in Apc Min/+ mice nor growth of human colorectal cancer cells. By contrast, Lrp6 silencing diminished anchorage-independent growth of BRafV600E-transformed IEC. Thus, LRP6 controls intestinal stem cell functionality and is necessary for BRAF-induced IEC oncogenesis.


2021 ◽  
Author(s):  
Wesley Tung ◽  
Ullas Valiya Chembazhi ◽  
Jing Yang ◽  
Ka Lam Nguyen ◽  
Aryan Lalwani ◽  
...  

Properly controlled intestinal epithelial cell regeneration is not only vital for protection against insults from environmental hazards but also crucial for preventing intestinal cancer. Intestinal stem cells located in the crypt region provide the driving force for epithelial regeneration, and thus their survival and death must be precisely regulated. We show here that polypyrimidine tract binding protein 1 (PTBP1, also called heterogeneous nuclear ribonucleoprotein I, or HNRNP I), an RNA-binding protein that post-transcriptionally regulates gene expression, is critical for intestinal stem cell survival and stemness. Mechanistically, we show that PTBP1 inhibits the expression of PHLDA3, an AKT repressor, and thereby maintains AKT activity in the intestinal stem cell compartment to promote stem cell survival and proliferation. Furthermore, we show that PTBP1 inhibits the expression of PTBP2, a paralog of PTBP1 that is known to induce neuron differentiation, through repressing inclusion of alternative exon 10 to Ptbp2 transcript. Loss of PTBP1 results in a significant upregulation of PTBP2, which is accompanied by splicing changes in genes that are important for neuron cell development. This finding suggests that PTBP1 prevents aberrant differentiation of intestinal stem cells into neuronal cells through inhibiting PTBP2. Our results thus reveal a novel mechanism whereby PTBP1 maintains intestinal stem cell survival and stemness through the control of gene function post-transcriptionally.


2020 ◽  
Author(s):  
Penglin Xia ◽  
Yajie Yang ◽  
Ruling Liu ◽  
Haibo Tang ◽  
Jicong Du ◽  
...  

Abstract Background: Radiation induced-intestinal injury (RIII) is a common complication after radiation therapy in patients with pelvic, abdominal or retroperitoneal tumors. The mechanism of RIII includes radiation-induced death of intestinal epithelial cells (IECs) and damage of intestinal stem cells (ISCs), among which damage of ISCs is main cause. Most recently, hypoxia Inducible factor (HIF) has been found to have effect on maintaining stemness and promoting proliferation of ISCs, which suggests a protective role of HIF in the RIII. In this study, we investigated the effect of FG4592, a novel up-regulator of HIF, on the protection of RIII, and further researched its function on ISCs.Methods: With/without FG4592 treatment, the abdomen of mice was radiated at dose of 25Gy, and then the degree of intestinal injury was assessed by H&E staining and Brdu label. By intestinal organoid culture, the multiplication capacity and differentiation features of ISCs were detected. Besides, the effects of FG4592 on the radiated IECs were also evaluated by detecting cell viability, apoptosis, and proliferation potential.Results: FG4592 could effectively up-regulate the expression of HIF-1α and HIF-2α in vivo. An abdominal radiation of 25Gy established the RIII model of mice, by which FG4592 was verified to have protective effect on the intestine from radiation. Morphology and Brdu test of intestinal organoid showed that FG4592 could promote regeneration and differentiation in ISCs after radiation, which were mediated by up-regulating HIF-2 rather than HIF-1.Conclusion: FG4592, a novel up-regulator of HIF could remit RIII and promote regeneration and differentiation of ISCs after radiation, which were depend on HIF-2 rather than HIF-1.


Sign in / Sign up

Export Citation Format

Share Document