scholarly journals Notch-IGF1 signaling during liver regeneration drives biliary epithelial cell expansion and inhibits hepatocyte differentiation

2021 ◽  
Vol 14 (688) ◽  
pp. eaay9185
Author(s):  
Sarah E. Minnis-Lyons ◽  
Sofía Ferreira-González ◽  
Niya Aleksieva ◽  
Tak Yung Man ◽  
Victoria L. Gadd ◽  
...  

In the adult liver, a population of facultative progenitor cells called biliary epithelial cells (BECs) proliferate and differentiate into cholangiocytes and hepatocytes after injury, thereby restoring liver function. In mammalian models of chronic liver injury, Notch signaling is essential for bile duct formation from these cells. However, the continual proliferation of BECs and differentiation of hepatocytes in these models have limited their use for determining whether Notch signaling is required for BECs to replenish hepatocytes after injury in the mammalian liver. Here, we used a temporally restricted model of hepatic repair in which large-scale hepatocyte injury and regeneration are initiated through the acute loss of Mdm2 in hepatocytes, resulting in the rapid, coordinated proliferation of BECs. We found that transient, early activation of Notch1- and Notch3-mediated signaling and entrance into the cell cycle preceded the phenotypic expansion of BECs into hepatocytes. Notch inhibition reduced BEC proliferation, which resulted in failure of BECs to differentiate into hepatocytes, indicating that Notch-dependent expansion of BECs is essential for hepatocyte regeneration. Notch signaling increased the abundance of the insulin-like growth factor 1 receptor (IGF1R) in BECs, and activating IGFR signaling increased BEC numbers but suppressed BEC differentiation into hepatocytes. These results suggest that different signaling mechanisms control BEC expansion and hepatocyte differentiation.

2021 ◽  
Author(s):  
Jonathan R Deans ◽  
Poonamjot Deol ◽  
Nina Titova ◽  
Sarah H Radi ◽  
Linh M Vuong ◽  
...  

Hepatocyte Nuclear Factor 4α (HNF4α), a master regulator of hepatocyte differentiation, is regulated by two promoters (P1 and P2). P1-HNF4α is the major isoform in the adult liver while P2-HNF4α is thought to be expressed only in fetal liver and liver cancer. Here, we show that P2-HNF4α is expressed at ZT9 and ZT21 in the normal adult liver and orchestrates a distinct transcriptome and metabolome via unique chromatin and protein-protein interactions. We demonstrate that while P1-HNF4α drives gluconeogenesis, P2-HNF4α drives ketogenesis and is required for elevated levels of ketone bodies in females. Exon swap mice expressing only P2- HNF4α exhibit subtle differences in circadian gene regulation and disruption of the clock increases expression of P2-HNF4α. Taken together, we propose that the highly conserved two-promoter structure of the Hnfa gene is an evolutionarily conserved mechanism to maintain the balance between gluconeogenesis and ketogenesis in the liver in a circadian fashion.


2019 ◽  
Vol 2019 ◽  
pp. 1-11 ◽  
Author(s):  
Jacquelyn O. Russell ◽  
Sungjin Ko ◽  
Satdarshan P. Monga ◽  
Donghun Shin

Liver regeneration after most forms of injury is mediated through the proliferation of hepatocytes. However, when hepatocyte proliferation is impaired, such as during chronic liver disease, liver progenitor cells (LPCs) arising from the biliary epithelial cell (BEC) compartment can give rise to hepatocytes to mediate hepatic repair. Promotion of LPC-to-hepatocyte differentiation in patients with chronic liver disease could serve as a potentially new therapeutic option, but first requires the identification of the molecular mechanisms driving this process. Notch signaling has been identified as an important signaling pathway promoting the BEC fate during development and has also been implicated in regulating LPC differentiation during regeneration. SRY-related HMG box transcription factor 9 (Sox9) is a direct target of Notch signaling in the liver, and Sox9 has also been shown to promote the BEC fate during development. We have recently shown in a zebrafish model of LPC-driven liver regeneration that inhibition of Hdac1 activity through MS-275 treatment enhances sox9b expression in LPCs and impairs LPC-to-hepatocyte differentiation. Therefore, we hypothesized that inhibition of Notch signaling would promote LPC-to-hepatocyte differentiation by repressing sox9b expression in zebrafish. We ablated the hepatocytes of Tg(fabp10a:CFP-NTR) larvae and blocked Notch activation during liver regeneration through treatment with γ-secretase inhibitor LY411575 and demonstrated enhanced induction of Hnf4a in LPCs. Alternatively, enhancing Notch signaling via Notch3 intracellular domain (N3ICD) overexpression impaired Hnf4a induction. Hepatocyte ablation in sox9b heterozygous mutant embryos enhanced Hnf4a induction, while BEC-specific Sox9b overexpression impaired LPC-to-hepatocyte differentiation. Our results establish the Notch-Sox9b signaling axis as inhibitory to LPC-to-hepatocyte differentiation in a well-established in vivo LPC-driven liver regeneration model.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 819-819
Author(s):  
Ivy T Tran ◽  
Ashley R Sandy ◽  
Alexis Carulli ◽  
Gloria T Shan ◽  
Vedran Radojcic ◽  
...  

Abstract Abstract 819 Notch signaling is a cell-cell communication pathway with multiple functions in health and disease. Notch ligands of the Delta-like (Dll1, 3, 4) or Jagged (Jagged1, 2) family interact with one of four mammalian Notch receptors (Notch1-4), leading to proteolytic activation of the receptors by gamma-secretase. We have discovered a critical role for Notch signaling in the differentiation of pathogenic host-reactive T cells during graft-versus-host disease (GVHD) after allogeneic bone marrow transplantation (allo-BMT). Expression of the pan-Notch inhibitor DNMAML in donor T cells led to markedly reduced GVHD severity, without causing global immunosuppression (Blood 2011, 117(1): 299–308). These findings identify Notch signaling in alloreactive T cells as an attractive therapeutic target after allo-BMT. To explore preclinical strategies of Notch blockade in GVHD, we first assessed the effects of systemic pan-Notch inhibition with gamma-secretase inhibitors. In the B6 anti-BALB/c MHC-mismatched model of allo-BMT, administration of the gamma-secretase inhibitor dibenzazepine was as efficient as genetic strategies at blocking Notch target gene expression and production of inflammatory cytokines in donor T cells (IFN-γ, TNF-α, IL-2). However, dibenzazepine induced severe gastrointestinal toxicity after total body irradiation due to inhibition of both Notch1 and Notch2 in the gut epithelium. To avoid these side effects, we hypothesized that targeting individual Notch receptors or ligands could provide safe therapeutic Notch blockade after allo-BMT. Among the four mammalian Notch receptors (Notch1-4), donor alloreactive T cells expressed Notch1 and Notch2. Host dendritic cells expressed Notch ligands of the Jagged and Delta-like (Dll) families, with markedly increased Dll4 but not Jagged1/2 transcripts after total body irradiation. This suggested that blockade of Notch1 and/or Notch2 in T cells or Delta-like Notch ligands in dendritic cells could abrogate GVHD. To explore this possibility, we used specific monoclonal antibodies to neutralize Notch receptors and ligands in vivo after allo-BMT (Nature 2006, 444(7122):1083–7; Nature 2010, 464(7291): 1052–7). Combined blockade of Notch1 and Notch2 in vivo reduced the production of key inflammatory cytokines by alloreactive CD4+ and CD8+ T cells to a similar extent as DNMAML-mediated pan-Notch inhibition. Inhibition of Notch1 alone led to a large decrease in cytokine secretion, indicating that Notch1 is a dominant non-redundant Notch receptor in alloreactive T cells. Consistently, transplantation of Notch1-deficient but not Notch2-deficient B6 T cells allowed for decreased GVHD and improved survival in BALB/c recipients, similarly to global Notch inhibition by DNMAML. We then studied the consequences of inhibiting Dll1, Dll4 or both Dll1/Dll4 Notch ligands during acute GVHD. Combined Dll1/Dll4 blockade was as potent as DNMAML expression in decreasing cytokine production by alloreactive T cells, demonstrating that Delta-like and not Jagged ligands are the key Notch agonists at the alloimmune synapse. Dll4 inhibition was superior to Dll1 blockade in reducing cytokine production, abrogating GVHD, and prolonging recipient survival. Importantly, combined Dll1/Dll4 inhibition provided long-term protection against GVHD morbidity and mortality, while avoiding severe gastrointestinal side effects from Notch inhibition. Protection was observed even upon transient Dll1/Dll4 blockade during 1–2 weeks after transplantation. Altogether, our data suggest that Notch1 and Dll4 preferentially interact during alloreactive T cell priming and identify novel strategies to safely and efficiently target individual elements of the Notch pathway after allo-BMT. Humanized antibodies against Notch receptors and ligands were designed to block both mouse and human proteins, thus our preclinical work could lead to new strategies for GVHD control in human patients. Disclosures: Shelton: Genentech Inc.: Employment. Yan:Genentech Inc.: Employment. Siebel:Genentech Inc.: Employment.


ASN NEURO ◽  
2016 ◽  
Vol 8 (2) ◽  
pp. 175909141663702 ◽  
Author(s):  
Maria da Conceição Pereira ◽  
Sara Morais ◽  
Jorge Sequeiros ◽  
Isabel Alonso

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1326-1326
Author(s):  
Adrian Schwarzer ◽  
Martin May ◽  
Harald Genth ◽  
Zhixiong Li ◽  
Christopher Baum ◽  
...  

Abstract Molecular hallmarks of T-ALL are the aberrant activation of NOTCH signaling and high activity of the PI3K-AKT-mTOR pathway. Upregulated mTOR and NOTCH have been linked to the resistance of T-ALL to chemotherapy as well as to high frequencies of leukemia-initiating cells. Hence, the mTOR and the NOTCH pathways are promising therapeutic targets in T-ALL. However, clinical success of the mTOR inhibitor Rapamycin in acute leukemia has been disappointing. Similar results have been observed in mouse models of T-ALL treated with Notch inhibitors. To investigate the impact of mTOR and Notch inhibition in a genetically complex T-ALL, we developed an aggressive murine T-ALL model, driven by tyrosine kinase signaling, loss of Pten, Cux1-haploinsufficiency and constitutive Notch signaling. In vitro, T-ALL blasts were highly sensitive to inhibition of AKT, mTOR and Notch signaling. We transplanted the leukemias into secondary recipients and initiated treatment with Rapamycin after the onset of leukemia. Rapamycin significantly prolonged survival of the animals (placebo: 27 days, Rapamycin 49 days, p<0.001). Eventually, all Rapamycin treated animals succumbed to the T-ALL that extensively infiltrated the bone marrow and solid organs despite continuous drug administration. When Rapamycin-resistant blasts were explanted and cultured in petri dishes they again became susceptible to Rapamycin, demonstrating a context-dependent resistance rather than outgrowth of intrinsically resistant clones. Gene set enrichment analysis revealed that Rapamycin-resistant T-ALL in vivo upregulated genetic networks associated with cell-cell interactions. Stromal cell support from OP9-cells as well as from mesenchymal stem cells recapitulated the in vivo effect and induced resistance to mTOR and Notch-inhibition in T-ALL blasts. Coating the tissue culture wells with Collagen, Fibronectin, Retronectin or Matrigel, did not elicit resistance. By using trans-well assays we show that the stroma-induced resistance was dependent on direct cell-cell interactions. Immunoblots and PhosFlow probing the mTORC1/C2 and Notch pathway demonstrated an identical drug effect on their intracellular targets in resistant T-ALL blasts cultured on stroma cells and susceptible cells in suspension. Since the number of molecules potentially involved in cell-to-cell contacts is vast, we focused on central nodes that organize this process in order to find a potentially druggable target that is critically involved in stroma-induced resistance. Transcriptome profiling pointed towards upregulation of Rac-associated pathways. We determined the activation of Rac1 by PAK-pull down assays in T-ALL blasts grown in suspension or on stromal cells. We observed an increase (FC=1.96 ± 0.58, p=0.04) in activated Rac1 in the T-ALL blasts in contact with a stromal layer. To determine whether Rac activation plays a role in stroma-induced resistance, we devised a strategy to abrogate Rac signaling in T-ALL blasts, but not in the stromal cells, since inhibition of Rac in stromal cells by the Rac-inhibitor NSC23766 led to the their detachment. Furthermore, Rac1,2 and 3 can be functionally redundant, making knock down experiments using shRNAs challenging. The Clostridium difficile serotype F strain 1470 produces toxin B isoform (TcdBF), that selectively glucosylates and inactivates Rac(1,2,3). We pretreated T-ALL blasts with TcdBF and observed a dose-dependent functional inhibition of Rac GTPases monitored by dephosphorylation of the Rac effector kinase pS144/141-PAK-1/2. T-ALL blasts were then incubated for 5 hours with increasing toxin doses, washed 3 times and incubated in toxin-free medium. Eighteen hours after the end of the exposure to the toxins, Rac was still inhibited. Strikingly, in the TcdBF-pretreated T-ALL, the stroma-induced resistance effect was abrogated and clusters of apoptotic cells were clearly visible (>2 fold reduction of the input, p=0.002). In contrast, the carrier-treated T-ALL exhibited resistance to the inhibitors on stroma (>10 fold expansion of the input, p<0.0003). Altogether, we identify the Rac-GTPases as a nexus of stroma-induced drug resistance and show that inhibition of Rac and mTOR is synthetically lethal to T-ALL blasts T-ALL blasts that are in contact with stromal cells, paving the way to augment the effectiveness of small molecule inhibitors in acute leukemia. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Author(s):  
Aresh Sahu ◽  
Sulochana Devi ◽  
Jonathan Jui ◽  
Daniel Goldman

AbstractUnlike mammals, zebrafish can regenerate a damaged retina. Key to this regenerative response are Müller glia (MG) that divide and produce progenitors for retinal repair. Although factors regulating MG’s decision to divide remain mostly unknown, a certain threshold of neuron death must be exceeded in order for MG to engage in a regenerative response. A role for Notch signaling in this process is indicated since its inhibition expands the zone of injury-responsive MG following a focal injury. Our data show that injury-dependent changes in Dll4 and Dlb control Notch signaling in MG and that Hey1 and Id2b are downstream effectors that regulate proliferation of MG and MG-derived progenitors. Although we find Hey1 and Id2b can inhibit proliferation of MG-derived progenitors, only Hey1 is able to regulate MG’s injury response threshold. Remarkably, Hey1 suppression is sufficient to recapitulate the effects of Notch inhibition on MG’s injury response threshold.


2019 ◽  
Vol 10 (1) ◽  
Author(s):  
Martina Legris ◽  
Yetkin Çaka Ince ◽  
Christian Fankhauser

AbstractPhytochromes are bilin-binding photosensory receptors which control development over a broad range of environmental conditions and throughout the whole plant life cycle. Light-induced conformational changes enable phytochromes to interact with signaling partners, in particular transcription factors or proteins that regulate them, resulting in large-scale transcriptional reprograming. Phytochromes also regulate promoter usage, mRNA splicing and translation through less defined routes. In this review we summarize our current understanding of plant phytochrome signaling, emphasizing recent work performed in Arabidopsis. We compare and contrast phytochrome responses and signaling mechanisms among land plants and highlight open questions in phytochrome research.


2006 ◽  
Vol 1 (2) ◽  
pp. 195-205 ◽  
Author(s):  
Cecilia Sahlgren ◽  
Urban Lendahl

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 340-340
Author(s):  
Ashley R Sandy ◽  
Jooho Chung ◽  
Ivy T Tran ◽  
Gloria T Shan ◽  
Ann Friedman ◽  
...  

Abstract Abstract 340 Graft-versus-host disease (GVHD) is a significant cause of morbidity and mortality following allogeneic bone marrow transplantation (allo-BMT). We previously identified Notch signaling as an essential regulator of allogeneic CD4+ T cell responses mediating GVHD after allo-BMT. Alloreactive CD4+ T cells expressing the pan-Notch inhibitor DNMAML induced markedly less severe GVHD as compared to wild-type T cells, leading to improved survival of the recipients. Notch-deprived T cells had preserved in vivo expansion and cytotoxicity. However, alloreactive DNMAML CD4+ T cells produced markedly decreased amounts of multiple proinflammatory cytokines, including TNF-alpha, IFN-gamma, and IL-2. This was associated with increased expansion of Foxp3+ CD4+ T regulatory cells. Thus, Notch signaling is an attractive new therapeutic target to control GVHD without eliminating the anti-cancer activity of allo-BMT. To elucidate the mechanisms of Notch action in GVHD, we studied the effects of Notch inhibition in alloreactive CD4+ and CD8+ T cells using minor and major histocompatibility antigen-mismatched models of allo-BMT. In the B6 anti-BALB/b minor antigen-mismatched model, recipients of B6 T cells were protected from lethal acute GVHD upon DNMAML expression in the CD4+, CD8+ or both T cell compartments. In the B6 anti-BALB/c MHC-mismatched model, DNMAML CD4+ or CD8+ T cells transplanted alone or in combination induced significantly less GVHD and resulted in improved survival compared to wild-type T cells. Upon ex vivo restimulation with anti-CD3/CD28 antibodies, both CD4+ and CD8+ DNMAML alloreactive T cells had markedly decreased production of IFN-gamma. These findings suggest that Notch signaling has parallel functions in CD4+ and CD8+ T cells. We then studied expression of Tbx21 (encoding T-bet) and Eomes, the key transcription factors regulating Ifng transcription in CD4+ Th1 and CD8+ T cells, respectively. DNMAML alloreactive T cells had preserved amounts of Tbx21 mRNA and T-bet protein, and increased levels of Eomes transcripts and protein. These data differ from past reports indicating that Notch signaling controls T cell differentiation through direct regulation of Tbx21 and Eomes expression. Ex vivo restimulation of DNMAML CD4+ and CD8+ T cells with PMA (diacylglycerol analog) and ionomycin (calcium ionophore) rescued IFN-gamma production by both T cell compartments and partially restored IL-2 production by CD4+ T cells, suggesting abnormal signaling downstream of the T cell receptor. After anti-CD3/CD28 restimulation, DNMAML alloreactive T cells showed markedly decreased phosphorylation of Mek1 and Erk1/2, indicating defective Ras/MAPK activation. PMA was sufficient to rescue Erk1/2 activation. NFkB activity was also significantly impaired in alloreactive DNMAML T cells as assessed with a NFkB-luciferase reporter transgene. Abnormal responsiveness was acquired in vivo during alloreactive T cell priming, since naïve DNMAML T cells had preserved Ras/MAPK activation. Moreover, alloreactive Notch-deprived T cells had elevated levels of intracellular cAMP and increased expression of the anergy-associated genes, Dgka and Egr3. Thus, alloreactive DNMAML T cells had features reminiscent of T cell anergy. Given that in vivo proliferation in irradiated recipients and cytotoxicity of DNMAML alloreactive T cells were largely preserved, our data suggest a “split anergy” phenotype with differential effects on distinct T cell effector functions. Altogether, our results reveal a parallel role for Notch signaling in both the CD4+ and CD8+ T cell compartments that differ from all previous reports of Notch action in mature T cells. Understanding the role of Notch signaling in alloreactive T cells is essential for harnessing the therapeutic potential of Notch inhibition in GVHD. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document