Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 190
Filter
1.
Cells ; 12(23)2023 12 04.
Article in English | MEDLINE | ID: mdl-38067194

ABSTRACT

The bone marrow (BM) hematopoietic system (HS) gives rise to blood cells originating from hematopoietic stem cells (HSCs), including megakaryocytes (MKs) and red blood cells (erythrocytes; RBCs). Many steps of the cell-fate decision remain to be elucidated, being important for cancer treatment. To explore the role of Wnt/ß-catenin for MK and RBC differentiation, we activated ß-catenin signaling in platelet-derived growth factor b (Pdgfb)-expressing cells of the HS using a Cre-lox approach (Ctnnb1BM-GOF). FACS analysis revealed that Pdgfb is mainly expressed by megakaryocytic progenitors (MKPs), MKs and platelets. Recombination resulted in a lethal phenotype in mutants (Ctnnb1BM-GOFwt/fl, Ctnnb1BM-GOFfl/fl) 3 weeks after tamoxifen injection, showing an increase in MKs in the BM and spleen, but no pronounced anemia despite reduced erythrocyte counts. BM transplantation (BMT) of Ctnnb1BM-GOF BM into lethally irradiated wildtype recipients (BMT-Ctnnb1BM-GOF) confirmed the megakaryocytic, but not the lethal phenotype. CFU-MK assays in vitro with BM cells of Ctnnb1BM-GOF mice supported MK skewing at the expense of erythroid colonies. Molecularly, the runt-related transcription factor 1 (RUNX1) mRNA, known to suppress erythropoiesis, was upregulated in Ctnnb1BM-GOF BM cells. In conclusion, ß-catenin activation plays a key role in cell-fate decision favoring MK development at the expense of erythroid production.


Subject(s)
Megakaryocytes , Thrombopoiesis , beta Catenin , Animals , Mice , beta Catenin/metabolism , Megakaryocyte-Erythroid Progenitor Cells , Megakaryocytes/metabolism , Proto-Oncogene Proteins c-sis/metabolism , Thrombopoiesis/physiology
2.
Dev Cell ; 58(23): 2732-2745.e5, 2023 Dec 04.
Article in English | MEDLINE | ID: mdl-37909044

ABSTRACT

The extrahepatic branches of the biliary tree have glands that connect to the surface epithelium through narrow pits. The duct epithelia undergo homeostatic renewal, yet the identity and multiplicity of cells that maintain this tissue is unknown. Using marker-free and targeted clonal fate mapping in mice, we provide evidence that the extrahepatic bile duct is compartmentalized. Pit cholangiocytes of extramural glands renewed the surface epithelium, whereas basally oriented cholangiocytes maintained the gland itself. In contrast, basally positioned cholangiocytes replenished the surface epithelium in mural glands. Single-cell sequencing identified genes enriched in the base and surface epithelial populations, with trajectory analysis showing graded gene expression between these compartments. Epithelia were plastic, changing cellular identity upon fasting and refeeding. Gain of canonical Wnt signaling caused basal cell expansion, gastric chief cell marker expression, and a decrease in surface epithelial markers. Our results identify the cellular hierarchy governing extrahepatic biliary epithelial renewal.


Subject(s)
Bile Ducts, Extrahepatic , Biliary Tract , Animals , Mice , Epithelium , Epithelial Cells , Cell Proliferation
3.
JCI Insight ; 8(8)2023 04 24.
Article in English | MEDLINE | ID: mdl-37092550

ABSTRACT

The main cause of malignancy-related mortality is metastasis. Although metastatic progression is driven by diverse tumor-intrinsic mechanisms, there is a growing appreciation for the contribution of tumor-extrinsic elements of the tumor microenvironment, especially macrophages, which correlate with poor clinical outcomes. Macrophages consist of bone marrow-derived and tissue-resident populations. In contrast to bone marrow-derived macrophages, the transcriptional pathways that govern the pro-metastatic activities of tissue-resident macrophages (TRMs) remain less clear. Alveolar macrophages (AMs) are a TRM population with critical roles in tissue homeostasis and metastasis. Wnt/ß-catenin signaling is a hallmark of cancer and has been identified as a pathologic regulator of AMs in infection. We tested the hypothesis that ß-catenin expression in AMs enhances metastasis in solid tumor models. Using a genetic ß-catenin gain-of-function approach, we demonstrated that (a) enhanced ß-catenin in AMs heightened lung metastasis; (b) ß-catenin activity in AMs drove a dysregulated inflammatory program strongly associated with Tnf expression; and (c) localized TNF-α blockade abrogated this metastatic outcome. Last, ß-catenin gene CTNNB1 and TNF expression levels were positively correlated in AMs of patients with lung cancer. Overall, our findings revealed a Wnt/ß-catenin/TNF-α pro-metastatic axis in AMs with potential therapeutic implications against tumors refractory to the antineoplastic actions of TNF-α.


Subject(s)
Lung Neoplasms , Macrophages, Alveolar , Humans , Macrophages, Alveolar/metabolism , Tumor Necrosis Factor-alpha/metabolism , beta Catenin/genetics , beta Catenin/metabolism , Lung Neoplasms/pathology , Wnt Signaling Pathway , Tumor Microenvironment
4.
Bioact Mater ; 21: 110-128, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36093329

ABSTRACT

Cell source is the key to decellularized matrix (DM) strategy. This study compared 3 cell types, osteocytes with/without dominant active Wnt/ß-catenin signaling (daCO and WTO) and bone marrow stromal cells (BMSCs) for their DMs in bone repair. Decellularization removes all organelles and >95% DNA, and retained >74% collagen and >71% GAG, maintains the integrity of cell basement membrane with dense boundaries showing oval and honeycomb structure in osteocytic DM and smooth but irregular shape in the BMSC-DM. DM produced higher cell survival rate (90%) and higher proliferative activity. In vitro, daCO-DM induces more and longer stress fibers in BMSCs, conducive to cell adhesion, spreading, and osteogenic differentiation. 8-wk after implantation of the critical-sized parietal bone defect model, daCO-DM formed tight structures, composed of a large number of densely-arranged type-I collagen under polarized light microscope, which is similar to and integrated with host bone. BV/TV (>54%) was 1.5, 2.9, and 3.5 times of WTO-DM, BMSC-DM, and none-DM groups, and N.Ob/T.Ar (3.2 × 102/mm2) was 1.7, 2.9, and 3.3 times. At 4-wk, daCO-DM induced osteoclastogenesis, 2.3 times higher than WTO-DM; but BMSC-DM or none-DM didn't. daCO-DM increased the expression of RANKL and MCSF, Vegfa and Angpt1, and Ngf in BMSCs, which contributes to osteoclastogenesis, angiogenesis, and neurogenesis, respectively. daCO-DM promoted H-type vessel formation and nerve markers ß3-tubulin and NeuN expression. Conclusion: daCO-DM produces metabolic and neurovascularized organoid bone to accelerate the repair of bone defects. These features are expected to achieve the effect of autologous bone transplantation, suitable for transformation application.

5.
Genes Dev ; 36(11-12): 737-751, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35798383

ABSTRACT

The primary cilium, a signaling organelle projecting from the surface of a cell, controls cellular physiology and behavior. The presence or absence of primary cilia is a distinctive feature of a given tumor type; however, whether and how the primary cilium contributes to tumorigenesis are unknown for most tumors. Medulloblastoma (MB) is a common pediatric brain cancer comprising four groups: SHH, WNT, group 3 (G3), and group 4 (G4). From 111 cases of MB, we show that primary cilia are abundant in SHH and WNT MBs but rare in G3 and G4 MBs. Using WNT and G3 MB mouse models, we show that primary cilia promote WNT MB by facilitating translation of mRNA encoding ß-catenin, a major oncoprotein driving WNT MB, whereas cilium loss promotes G3 MB by disrupting cell cycle control and destabilizing the genome. Our findings reveal tumor type-specific ciliary functions and underlying molecular mechanisms. Moreover, we expand the function of primary cilia to translation control and reveal a molecular mechanism by which cilia regulate cell cycle progression, thereby providing new frameworks for studying cilium function in normal and pathologic conditions.


Subject(s)
Brain Neoplasms , Cerebellar Neoplasms , Medulloblastoma , Animals , Brain Neoplasms/pathology , Cell Cycle/genetics , Cerebellar Neoplasms/genetics , Cilia/genetics , Humans , Medulloblastoma/genetics , Mice
6.
Nat Commun ; 13(1): 1500, 2022 03 21.
Article in English | MEDLINE | ID: mdl-35314700

ABSTRACT

Although the mammalian intestinal epithelium manifests robust regenerative capacity after various cytotoxic injuries, the underlying mechanism has remained unclear. Here we identify the cyclin-dependent kinase inhibitor p57 as a specific marker for a quiescent cell population located around the +4 position of intestinal crypts. Lineage tracing reveals that the p57+ cells serve as enteroendocrine/tuft cell precursors under normal conditions but dedifferentiate and act as facultative stem cells to support regeneration after injury. Single-cell transcriptomics analysis shows that the p57+ cells undergo a dynamic reprogramming process after injury that is characterized by fetal-like conversion and metaplasia-like transformation. Population-level analysis also detects such spatiotemporal reprogramming widely in other differentiated cell types. In intestinal adenoma, p57+ cells manifest homeostatic stem cell activity, in the context of constitutively activated spatiotemporal reprogramming. Our results highlight a pronounced plasticity of the intestinal epithelium that supports maintenance of tissue integrity in normal and neoplastic contexts.


Subject(s)
Intestinal Mucosa , Neoplasms , Animals , Cell Differentiation , Intestinal Mucosa/metabolism , Intestines , Mammals , Neoplasms/metabolism , Stem Cells/metabolism
7.
Elife ; 112022 01 19.
Article in English | MEDLINE | ID: mdl-35042581

ABSTRACT

In the thymus, the thymic epithelium provides a microenvironment essential for the development of functionally competent and self-tolerant T cells. Previous findings showed that modulation of Wnt/ß-catenin signaling in mouse thymic epithelial cells (TECs) disrupts embryonic thymus organogenesis. However, the role of ß-catenin in TECs for postnatal T-cell development remains to be elucidated. Here, we analyzed gain-of-function (GOF) and loss-of-function (LOF) of ß-catenin highly specific in mouse TECs. We found that GOF of ß-catenin in TECs results in severe thymic dysplasia and T-cell deficiency beginning from the embryonic period. By contrast, LOF of ß-catenin in TECs reduces the number of cortical TECs and thymocytes modestly and only postnatally. These results indicate that fine-tuning of ß-catenin expression within a permissive range is required for TECs to generate an optimal microenvironment to support postnatal T-cell development.


Subject(s)
Epithelial Cells/metabolism , T-Lymphocytes/metabolism , Thymus Gland/metabolism , beta Catenin/metabolism , Animals , Female , Mice
8.
FASEB J ; 35(11): e21957, 2021 11.
Article in English | MEDLINE | ID: mdl-34606641

ABSTRACT

The Wnt signaling antagonist, sclerostin, is a potent suppressor of bone acquisition that also mediates endocrine communication between bone and adipose. As a result, Sost-/- mice exhibit dramatic increases in bone formation but marked decreases in visceral and subcutaneous adipose that are secondary to alterations in lipid synthesis and utilization. While interrogating the mechanism by which sclerostin influences adipocyte metabolism, we observed paradoxical increases in the adipogenic potential and numbers of CD45- :Sca1+ :PDGFRα+ adipoprogenitors in the stromal vascular compartment of fat pads isolated from male Sost-/- mice. Lineage tracing studies indicated that sclerostin deficiency blocks the differentiation of PDGFRα+ adipoprogenitors to mature adipocytes in association with increased Wnt/ß-catenin signaling. Importantly, osteoblast/osteocyte-specific Sost gene deletion mirrors the accumulation of PDGFRα+ adipoprogenitors, reduction in fat mass, and improved glucose metabolism evident in Sost-/- mice. These data indicate that bone-derived sclerostin regulates multiple facets of adipocyte physiology ranging from progenitor cell commitment to anabolic metabolism.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adipocytes/metabolism , Adipogenesis/genetics , Bone and Bones/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Stem Cells/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway/genetics , beta Catenin/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adipose Tissue/metabolism , Animals , Blood Glucose/metabolism , Cells, Cultured , Gene Knockout Techniques/methods , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoblasts/metabolism , Osteocytes/metabolism , Osteogenesis/genetics
9.
Nat Commun ; 11(1): 1680, 2020 04 03.
Article in English | MEDLINE | ID: mdl-32245949

ABSTRACT

Rosettes are widely used in epithelial morphogenesis during embryonic development and organogenesis. However, their role in postnatal development and adult tissue maintenance remains largely unknown. Here, we show zona glomerulosa cells in the adult adrenal cortex organize into rosettes through adherens junction-mediated constriction, and that rosette formation underlies the maturation of adrenal glomerular structure postnatally. Using genetic mouse models, we show loss of ß-catenin results in disrupted adherens junctions, reduced rosette number, and dysmorphic glomeruli, whereas ß-catenin stabilization leads to increased adherens junction abundance, more rosettes, and glomerular expansion. Furthermore, we uncover numerous known regulators of epithelial morphogenesis enriched in ß-catenin-stabilized adrenals. Among these genes, we show Fgfr2 is required for adrenal rosette formation by regulating adherens junction abundance and aggregation. Together, our data provide an example of rosette-mediated postnatal tissue morphogenesis and a framework for studying the role of rosettes in adult zona glomerulosa tissue maintenance and function.


Subject(s)
Adherens Junctions/metabolism , Morphogenesis , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Zona Glomerulosa/growth & development , beta Catenin/metabolism , Adherens Junctions/genetics , Adherens Junctions/ultrastructure , Adrenal Gland Neoplasms/surgery , Animals , Animals, Newborn , Female , Humans , Male , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Receptor, Fibroblast Growth Factor, Type 2/genetics , Zona Glomerulosa/cytology , Zona Glomerulosa/metabolism , Zona Glomerulosa/ultrastructure , beta Catenin/genetics
10.
Cell Stem Cell ; 24(5): 753-768.e6, 2019 05 02.
Article in English | MEDLINE | ID: mdl-30982770

ABSTRACT

Cell-autonomous Wnt signaling has well-characterized functions in controlling stem cell activity, including in the prostate. While niche cells secrete Wnt ligands, the effects of Wnt signaling in niche cells per se are less understood. Here, we show that stromal cells in the proximal prostatic duct near the urethra, a mouse prostate stem cell niche, not only produce multiple Wnt ligands but also exhibit strong Wnt/ß-catenin activity. The non-canonical Wnt ligand Wnt5a, secreted by proximal stromal cells, directly inhibits proliefration of prostate epithelial stem or progenitor cells whereas stromal cell-autonomous canonical Wnt/ß-catenin signaling indirectly suppresses prostate stem or progenitor activity via the transforming growth factor ß (TGFß) pathway. Collectively, these pathways restrain the proliferative potential of epithelial cells in the proximal prostatic ducts. Human prostate likewise exhibits spatially restricted distribution of stromal Wnt/ß-catenin activity, suggesting a conserved mechanism for tissue patterning. Thus, this study shows how distinct stromal signaling mechanisms within the prostate cooperate to regulate tissue homeostasis.


Subject(s)
Epithelial Cells/physiology , Prostate/cytology , Stem Cells/physiology , Stromal Cells/physiology , Wnt-5a Protein/metabolism , Animals , Cell Proliferation , Cells, Cultured , Humans , Male , Mice , Mice, Inbred C57BL , Mice, SCID , Receptor Cross-Talk , Stem Cell Niche , Transforming Growth Factor beta/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism
11.
Mol Neurobiol ; 56(1): 553-566, 2019 Jan.
Article in English | MEDLINE | ID: mdl-29737454

ABSTRACT

ß-catenin (key mediator in the Wnt signaling pathway) contributes to the pathophysiology of mood disorders, associated to neurogenesis and neuroplasticity. Decreased ß-catenin protein levels have been observed in the hippocampus and prefrontal cortex of depressed subjects. Additionally, the antidepressants exert, at least in part, their neurogenic effects by increasing ß-catenin levels in the subgranular zone of the hippocampus. To further understand the role of ß-catenin in depression and anxiety, we generated two conditional transgenic mice in which ß-catenin was either inactivated or stabilized in cells expressing CreERT under the control of the astrocyte-specific glutamate transporter (GLAST) promoter inducible by tamoxifen, which presents high expression levels on the subgranular zone of the hippocampus. Here, we show that ß-catenin inactivation in GLAST-expressing cells enhanced anxious/depressive-like responses. These behavioral changes were associated with impaired hippocampal proliferation and markers of immature neurons as doublecortin. On the other hand, ß-catenin stabilization induced an anxiolytic-like effect in the novelty suppressed feeding test and tended to ameliorate depressive-related behaviors. In these mice, the control over the Wnt/ß-catenin pathway seems to be tighter as evidenced by the lack of changes in some proliferation markers. Moreover, animals with stabilized ß-catenin showed resilience to some anxious/depressive manifestations when subjected to the corticosterone model of depression. Our findings demonstrate that ß-catenin present in GLAST-expressing cells plays a critical role in the development of anxious/depressive-like behaviors and resilience, which parallels its regulatory function on hippocampal proliferation. Further studies need to be done to clarify the importance of these changes in other brain areas also implicated in the neurobiology of anxiety and depressive disorders.


Subject(s)
Anxiety/metabolism , Behavior, Animal , Depression/metabolism , Excitatory Amino Acid Transporter 1/metabolism , Hippocampus/pathology , beta Catenin/metabolism , Animals , Anxiety/complications , Behavior, Animal/drug effects , Cell Cycle/drug effects , Cell Proliferation/drug effects , Corticosterone/administration & dosage , Corticosterone/pharmacology , Dentate Gyrus/pathology , Depression/complications , Glial Fibrillary Acidic Protein/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Protein Stability/drug effects , SOXB1 Transcription Factors/metabolism
12.
Kidney Int ; 93(6): 1367-1383, 2018 06.
Article in English | MEDLINE | ID: mdl-29605095

ABSTRACT

Imbalance of Wnt/ß-catenin signaling in renal cells is associated with renal dysfunction, yet the precise mechanism is poorly understood. Previously we observed activated Wnt/ß-catenin signaling in renal tubules during proteinuric nephropathy with an unknown net effect. Therefore, to identify the definitive role of tubular Wnt/ß-catenin, we generated a novel transgenic "Tubcat" mouse conditionally expressing stabilized ß-catenin specifically in renal tubules following tamoxifen administration. Four weeks after tamoxifen injection, uninephrectomized Tubcat mice displayed proteinuria and elevated blood urea nitrogen levels compared to non-transgenic mice, implying a detrimental effect of the activated signaling. This was associated with infiltration of the tubulointerstitium predominantly by M1 macrophages and overexpression of the inflammatory chemocytokines CCL-2 and RANTES. Induction of overload proteinuria by intraperitoneal injection of low-endotoxin bovine serum albumin following uninephrectomy for four weeks aggravated proteinuria and increased blood urea nitrogen levels to a significantly greater extent in Tubcat mice. Renal dysfunction correlated with the degree of M1 macrophage infiltration in the tubulointerstitium and renal cortical up-regulation of CCL-2, IL-17A, IL-1ß, CXCL1, and ICAM-1. There was overexpression of cortical TLR-4 and NLRP-3 in Tubcat mice, independent of bovine serum albumin injection. Finally, there was no fibrosis, activation of epithelial-mesenchymal transition or non-canonical Wnt pathways observed in the kidneys of Tubcat mice. Thus, conditional activation of renal tubular Wnt/ß-catenin signaling in a novel transgenic mouse model demonstrates that this pathway enhances intrarenal inflammation via the TLR-4/NLRP-3 inflammasome axis in overload proteinuria.


Subject(s)
Inflammation Mediators/metabolism , Kidney Tubules/metabolism , Macrophages/metabolism , Nephritis/metabolism , Proteinuria/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Chemokine CCL2/metabolism , Chemokine CCL5/metabolism , Disease Models, Animal , Inflammasomes/metabolism , Kidney Tubules/pathology , Kidney Tubules/physiopathology , Mice, Transgenic , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nephrectomy , Nephritis/genetics , Nephritis/pathology , Nephritis/physiopathology , Proteinuria/genetics , Proteinuria/pathology , Proteinuria/physiopathology , Serum Albumin, Bovine , Toll-Like Receptor 4/metabolism , Up-Regulation , Wnt Signaling Pathway/genetics , beta Catenin/genetics
13.
Oncotarget ; 9(13): 11243-11257, 2018 Feb 16.
Article in English | MEDLINE | ID: mdl-29541410

ABSTRACT

BACKGROUND: Wnt/ß-catenin signaling plays a crucial role in embryogenesis, tissue homeostasis, metabolism and malignant transformation of different organs including the liver. Continuous ß-catenin signaling due to somatic mutations in exon 3 of the Ctnnb1 gene is associated with different liver diseases including cancer and cholestasis. RESULTS: Expression of a degradation resistant form of ß-catenin in hepatocytes resulted in 100% mortality within 31 days after birth. Ctnnb1CAhep mice were characterized by reduced body weight, significantly enlarged livers with hepatocellular fat accumulation around central veins and increased hepatic triglyceride content. Proteomics analysis using whole liver tissue revealed significant deregulation of proteins involved in fat, glucose and mitochondrial energy metabolism, which was also reflected in morphological anomalies of hepatocellular mitochondria. Key enzymes involved in transport and synthesis of fatty acids and cholesterol were significantly deregulated in livers of Ctnnb1CAhep mice. Furthermore, carbohydrate metabolism was substantially disturbed in mutant mice. CONCLUSION: Continuous ß-catenin signaling in hepatocytes results in premature death due to severe disturbances of liver associated metabolic pathways and mitochondrial dysfunction. METHODS: To investigate the influence of permanent ß-catenin signaling on liver biology we analyzed mice with hepatocyte specific expression of a dominant stable form of ß-catenin (Ctnnb1CAhep ) and their WT littermates by serum biochemistry, histology, electron microscopy, mRNA profiling and proteomic analysis of the liver.

14.
Development ; 145(1)2018 01 08.
Article in English | MEDLINE | ID: mdl-29217753

ABSTRACT

Several studies have demonstrated a multiphasic role for Wnt signaling during embryonic cardiogenesis and developed protocols that enrich for cardiac derivatives during in vitro differentiation of human pluripotent stem cells (hPSCs). However, few studies have investigated the role of Wnt signaling in the specification of cardiac progenitor cells (CPCs) toward downstream fates. Using transgenic mice and hPSCs, we tracked endothelial cells (ECs) that originated from CPCs expressing NKX2.5. Analysis of EC-fated CPCs at discrete phenotypic milestones during hPSC differentiation identified reduced Wnt activity as a hallmark of EC specification, and the enforced activation or inhibition of Wnt reduced or increased, respectively, the degree of vascular commitment within the CPC population during both hPSC differentiation and mouse embryogenesis. Wnt5a, which has been shown to exert an inhibitory influence on Wnt signaling during cardiac development, was dynamically expressed during vascular commitment of hPSC-derived CPCs, and ectopic Wnt5a promoted vascular specification of hPSC-derived and mouse embryonic CPCs.


Subject(s)
Embryo, Mammalian/metabolism , Endothelial Cells/metabolism , Heart/embryology , Pluripotent Stem Cells/metabolism , Wnt Signaling Pathway/physiology , Animals , Embryo, Mammalian/cytology , Endothelial Cells/cytology , Humans , Mice , Mice, Transgenic , Pluripotent Stem Cells/cytology , Wnt-5a Protein/genetics , Wnt-5a Protein/metabolism
15.
Dev Cell ; 43(6): 673-688.e5, 2017 12 18.
Article in English | MEDLINE | ID: mdl-29103956

ABSTRACT

Mechanistic target of rapamycin (MTOR) cooperates with Hedgehog (HH) signaling, but the underlying mechanisms are incompletely understood. Here we provide genetic, biochemical, and pharmacologic evidence that MTOR complex 1 (mTORC1)-dependent translation is a prerequisite for HH signaling. The genetic loss of mTORC1 function inhibited HH signaling-driven growth of the cerebellum and medulloblastoma. Inhibiting translation or mTORC1 blocked HH signaling. Depleting 4EBP1, an mTORC1 target that inhibits translation, alleviated the dependence of HH signaling on mTORC1. Consistent with this, phosphorylated 4EBP1 levels were elevated in HH signaling-driven medulloblastomas in mice and humans. In mice, an mTORC1 inhibitor suppressed medulloblastoma driven by a mutant SMO that is inherently resistant to existing SMO inhibitors, prolonging the survival of the mice. Our study reveals that mTORC1-mediated translation is a key component of HH signaling and an important target for treating medulloblastoma and other cancers driven by HH signaling.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Carrier Proteins/antagonists & inhibitors , Cerebellar Neoplasms/metabolism , Hedgehog Proteins/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Medulloblastoma/metabolism , Phosphoproteins/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/metabolism , Animals , Carrier Proteins/metabolism , Cell Cycle Proteins , Cell Proliferation/physiology , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/pathology , Eukaryotic Initiation Factors , Hedgehog Proteins/genetics , Humans , Kruppel-Like Transcription Factors/metabolism , Medulloblastoma/genetics , Medulloblastoma/pathology , Mice , Phosphoproteins/metabolism , Phosphorylation , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction/genetics , Smoothened Receptor/genetics , Smoothened Receptor/metabolism , Zinc Finger Protein Gli2/genetics , Zinc Finger Protein Gli2/metabolism
16.
Cancer Discov ; 7(5): 522-538, 2017 05.
Article in English | MEDLINE | ID: mdl-28202625

ABSTRACT

Obesity increases the risk of cancers, including hepatocellular carcinomas (HCC). However, the precise molecular mechanisms through which obesity promotes HCC development are still unclear. Recent studies have shown that gut microbiota may influence liver diseases by transferring its metabolites and components. Here, we show that the hepatic translocation of obesity-induced lipoteichoic acid (LTA), a Gram-positive gut microbial component, promotes HCC development by creating a tumor-promoting microenvironment. LTA enhances the senescence-associated secretory phenotype (SASP) of hepatic stellate cells (HSC) collaboratively with an obesity-induced gut microbial metabolite, deoxycholic acid, to upregulate the expression of SASP factors and COX2 through Toll-like receptor 2. Interestingly, COX2-mediated prostaglandin E2 (PGE2) production suppresses the antitumor immunity through a PTGER4 receptor, thereby contributing to HCC progression. Moreover, COX2 overexpression and excess PGE2 production were detected in HSCs in human HCCs with noncirrhotic, nonalcoholic steatohepatitis (NASH), indicating that a similar mechanism could function in humans.Significance: We showed the importance of the gut-liver axis in obesity-associated HCC. The gut microbiota-driven COX2 pathway produced the lipid mediator PGE2 in senescent HSCs in the tumor microenvironment, which plays a pivotal role in suppressing antitumor immunity, suggesting that PGE2 and its receptor may be novel therapeutic targets for noncirrhotic NASH-associated HCC. Cancer Discov; 7(5); 522-38. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 443.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Dinoprostone/metabolism , Gastrointestinal Microbiome/physiology , Liver Neoplasms/metabolism , Obesity/complications , Animals , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/microbiology , Female , Humans , Lipopolysaccharides/metabolism , Liver Neoplasms/immunology , Liver Neoplasms/microbiology , Male , Mice, Inbred C57BL , Obesity/microbiology , Teichoic Acids/metabolism , Tumor Escape/immunology , Tumor Microenvironment/immunology
17.
Clin Cancer Res ; 23(3): 833-844, 2017 Feb 01.
Article in English | MEDLINE | ID: mdl-27492974

ABSTRACT

PURPOSE: We have reported loss of SMAD4 promotes expression of CCL15 from colorectal cancer to recruit CCR1+ myeloid cells through the CCL15-CCR1 axis, which contributes to invasion and liver metastasis. However, the molecular mechanism of lung metastasis is yet to be elucidated. Our purpose is to determine whether similar mechanism is involved in the lung metastasis of colorectal cancer. EXPERIMENTAL DESIGN: In a mouse model, we examined whether SMAD4 could affect the metastatic activity of colorectal cancer cells to the lung through the CCL15-CCR1 axis. We immunohistochemically analyzed expression of SMAD4, CCL15, and CCR1 with 107 clinical specimens of colorectal cancer lung metastases. We also characterized the CCR1+ myeloid cells using several cell-type-specific markers. RESULTS: In a mouse model, CCL15 secreted from SMAD4-deficient colorectal cancer cells recruited CCR1+ cells, promoting their metastatic activities to the lung. Immunohistochemical analysis of lung metastases from colorectal cancer patients revealed that CCL15 expression was significantly correlated with loss of SMAD4, and that CCL15-positive metastases recruited approximately 1.9 times more numbers of CCR1+ cells than CCL15-negative metastases. Importantly, patients with CCL15-positive metastases showed a significantly shorter relapse-free survival (RFS) than those with CCL15-negative metastases, and multivariate analysis indicated that CCL15 expression was an independent predictor of shorter RFS. Immunofluorescent staining showed that most CCR1+ cells around lung metastases were tumor-associated neutrophil, although a minor fraction was granulocytic myeloid-derived suppressor cell. CONCLUSIONS: CCL15-CCR1 axis may be a therapeutic target to prevent colorectal cancer lung metastasis. CCL15 can be a biomarker indicating poor prognosis of colorectal cancer patients with lung metastases. Clin Cancer Res; 23(3); 833-44. ©2016 AACR.


Subject(s)
Chemokines, CC/physiology , Colorectal Neoplasms/pathology , Lung Neoplasms/secondary , Macrophage Inflammatory Proteins/physiology , Neoplasm Proteins/deficiency , Neutrophil Infiltration , Receptors, CCR1/physiology , Smad4 Protein/deficiency , Animals , Cell Line, Tumor , Cell Movement , Chemokines, CC/biosynthesis , Chemokines, CC/genetics , Colorectal Neoplasms/metabolism , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Genes, Reporter , Heterografts , Humans , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Macrophage Inflammatory Proteins/biosynthesis , Macrophage Inflammatory Proteins/genetics , Mice , Mice, Nude , Mice, SCID , Myeloid Cells/metabolism , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Prognosis , Proportional Hazards Models , Smad4 Protein/physiology
18.
Oncotarget ; 7(52): 86985-86998, 2016 Dec 27.
Article in English | MEDLINE | ID: mdl-27895309

ABSTRACT

BACKGROUND: The Wnt/ß-catenin signaling pathway plays a crucial role in embryonic development, tissue homeostasis, wound healing and malignant transformation in different organs including the liver. The consequences of continuous ß-catenin signaling in hepatocytes remain elusive. RESULTS: Livers of Ctnnb1CA hep mice were characterized by disturbed liver architecture, proliferating cholangiocytes and biliary type of fibrosis. Serum ALT and bile acid levels were significantly increased in Ctnnb1CA hep mice. The primary bile acid synthesis enzyme Cyp7a1 was increased whereas Cyp27 and Cyp8b1 were reduced in Ctnnb1CA hep mice. Expression of compensatory bile acid transporters including Abcb1, Abcb4, Abcc2 and Abcc4 were significantly increased in Ctnnb1CA hep mice while Ntcp was reduced. Accompanying changes of bile acid transporters favoring excretion of bile acids were observed in intestine and kidneys of Ctnnb1CA hep mice. Additionally, disturbed bile acid regulation through the FXR-FGF15-FGFR4 pathway was observed in mice with activated ß-catenin. MATERIALS AND METHODS: Mice with a loxP-flanked exon 3 of the Ctnnb1 gene were crossed to Albumin-Cre mice to obtain mice with hepatocyte-specific expression of a dominant stable form of ß-catenin (Ctnnb1CA hep mice). Ctnnb1CA hep mice were analyzed by histology, serum biochemistry and mRNA profiling. CONCLUSIONS: Expression of a dominant stable form of ß-catenin in hepatocytes results in severe cholestasis and biliary type fibrosis.


Subject(s)
Cholestasis/etiology , Hepatocytes/metabolism , beta Catenin/physiology , Animals , Bile Acids and Salts/metabolism , Cholestanetriol 26-Monooxygenase/genetics , Cholesterol 7-alpha-Hydroxylase/genetics , Liver Cirrhosis, Biliary/etiology , Mice , Mice, Inbred C57BL , Signal Transduction/physiology
19.
Dev Biol ; 412(2): 234-49, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26934381

ABSTRACT

Organ shape and size are important determinants of their physiological functions. Epithelial tubes are anlagen of many complex organs. How these tubes acquire their complex shape and size is a fundamental question in biology. In male mice, the Wolffian duct (WD; postnatally known as epididymis) undergoes an astonishing transformation, where a straight tube only a few millimetres long elongates to over 1000 times its original length and fits into a very small space, due to extensive coiling of epithelium, to perform the highly specialized function of sperm maturation. Defective coiling disrupts sperm maturation and leads to male infertility. Recent work has shown that epithelial cell proliferation is a major driver of WD coiling. Still, very little is known about the molecular signals involved in this process. Testicular androgens are known regulators of WD development. However, epithelial androgen receptor signalling is dispensable for WD coiling. In this study, we have shown that Wnt signalling is highly active in the entire WD epithelium during its coiling, and is limited to only a few segments of the epididymis in later life. Pharmacological and genetic suppression of Wnt signalling inhibited WD coiling by decreasing cell proliferation and promoting apoptosis. Comparative gene expression analysis identified Fibroblast growth factor 7 (Fgf7) as a prime Wnt target gene involved in WD coiling and in vitro treatment with Fgf7 protein increased coiling of WDs. In summary, our work has established that epithelial canonical Wnt signalling is a critical regulator of WD coiling and its precise regulation is essential for WD/epididymal differentiation.


Subject(s)
Epididymis/metabolism , Epithelium/metabolism , Wnt Signaling Pathway/genetics , Wolffian Ducts/metabolism , Animals , Epididymis/embryology , Epithelium/embryology , Fibroblast Growth Factor 7/genetics , Fibroblast Growth Factor 7/metabolism , Gene Expression Regulation, Developmental , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Reverse Transcriptase Polymerase Chain Reaction , Wolffian Ducts/embryology , beta Catenin/genetics , beta Catenin/metabolism
20.
Genes Dev ; 29(14): 1493-506, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26193883

ABSTRACT

Mutations in Adenomatous polyposis coli (APC) underlie familial adenomatous polyposis (FAP), an inherited cancer syndrome characterized by the widespread development of colorectal polyps. APC is best known as a scaffold protein in the ß-catenin destruction complex, whose activity is antagonized by canonical Wnt signaling. Whether other effector pathways mediate APC's tumor suppressor function is less clear. Here we report that activation of YAP, the downstream effector of the Hippo signaling pathway, is a general hallmark of tubular adenomas from FAP patients. We show that APC functions as a scaffold protein that facilitates the Hippo kinase cascade by interacting with Sav1 and Lats1. Consistent with the molecular link between APC and the Hippo signaling pathway, genetic analysis reveals that YAP is absolutely required for the development of APC-deficient adenomas. These findings establish Hippo-YAP signaling as a critical effector pathway downstream from APC, independent from its involvement in the ß-catenin destruction complex.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli/physiopathology , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Adenoma/enzymology , Adenoma/physiopathology , Adenomatous Polyposis Coli/enzymology , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli Protein/metabolism , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Cycle Proteins , Cells, Cultured , Hippo Signaling Pathway , Humans , Intestines/physiopathology , Mice , Transcription Factors , YAP-Signaling Proteins , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...