Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 1121, 2023 02 27.
Article in English | MEDLINE | ID: mdl-36849569

ABSTRACT

Liver tumour-initiating cells (TICs) contribute to tumour initiation, metastasis, progression and drug resistance. Metabolic reprogramming is a cancer hallmark and plays vital roles in liver tumorigenesis. However, the role of metabolic reprogramming in TICs remains poorly explored. Here, we identify a mitochondria-encoded circular RNA, termed mcPGK1 (mitochondrial circRNA for translocating phosphoglycerate kinase 1), which is highly expressed in liver TICs. mcPGK1 knockdown impairs liver TIC self-renewal, whereas its overexpression drives liver TIC self-renewal. Mechanistically, mcPGK1 regulates metabolic reprogramming by inhibiting mitochondrial oxidative phosphorylation (OXPHOS) and promoting glycolysis. This alters the intracellular levels of α-ketoglutarate and lactate, which are modulators in Wnt/ß-catenin activation and liver TIC self-renewal. In addition, mcPGK1 promotes PGK1 mitochondrial import via TOM40 interactions, reprogramming metabolism from oxidative phosphorylation to glycolysis through PGK1-PDK1-PDH axis. Our work suggests that mitochondria-encoded circRNAs represent an additional regulatory layer controlling mitochondrial function, metabolic reprogramming and liver TIC self-renewal.


Subject(s)
Liver , Oxidative Phosphorylation , Humans , Carcinogenesis , Lactic Acid , Mitochondria , RNA, Circular , RNA, Mitochondrial , Phosphoglycerate Kinase/genetics
2.
Int J Oncol ; 62(2)2023 02.
Article in English | MEDLINE | ID: mdl-36633145

ABSTRACT

MicroRNA (miRNA), a non­coding single­stranded RNA molecule with a length of 21­25 nucleotides transcripts, has been identified to play important roles in tumorigenesis and shows great potential applications in cancer diagnosis, prognosis and therapy. Brain derived neurotrophic factor (BDNF) is a member of the nerve growth factor family and usually serves as a biomarker in neurological and neuropsychiatric diseases for diagnosis and treatment by regulating its high­affinity receptor TrkB (Tyrosine Kinase Receptor B). Abnormal expression of BDNF is also closely related to the development of cancer, cancer­related pain and depression. However, little significant progress has been made in the application of BDNF in cancers. Recent studies have shown that the expression of BDNF is directly regulated by a cluster of miRNAs. This review concluded and discussed the role and mechanism of miRNAs targeting BDNF in cancers, and provided novel insights into the diagnosis and therapy of cancer in the future.


Subject(s)
Brain-Derived Neurotrophic Factor , MicroRNAs , Neoplasms , Humans , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasms/diagnosis , Neoplasms/genetics , Neoplasms/therapy , Prognosis , Receptor, trkB/genetics , Receptor, trkB/metabolism
3.
Front Oncol ; 12: 816270, 2022.
Article in English | MEDLINE | ID: mdl-35756642

ABSTRACT

Background: Perineural invasion (PNI) is a malignant metastatic mode of tumors and has been reported in many tumors including esophageal cancer (EC). However, the role of PNI in EC has been reported differently. This systematic review and meta-analysis aims to focus on the role of PNI in EC. Methods: Eight databases of CNKI, VIP, Wanfang, Scopus, Wiley, ISI, PubMed, and EBSCO are used for literature search. The association of PNI with gender, pathological stages of T and N (pT and pN), lymphovascular invasion (LVI), lymph node metastasis, 5-year overall survival (OS), and 5-year disease-free survival (DFS) was examined in the meta-analysis by Revman5.0 Software. The pooled OR/HR and 95% CI were used to assess the risk and prognostic value. Results: Sixty-nine published studies were screened for analysis of PNI in EC. The incidence of PNI in esophageal squamous carcinoma (ESCC) and esophageal adenocarcinoma (EAC) was different, but not statistically significant (p > 0.05). The PNI-positive patients had a significantly higher risk of pT stage (OR = 3.85, 95% CI = 2.45-6.05, p < 0.00001), pN stage (OR = 1.86, 95% CI = 1.52-2.28, p < 0.00001), LVI (OR = 2.44, 95% CI = 1.55-3.85, p = 0.0001), and lymph node metastasis (OR = 2.87, 95% CI = 1.56-5.29, p = 0.0007). Furthermore, the cumulative analysis revealed a significant correlation between PNI and poor OS (HR = 1.37, 95% CI = 1.24-1.51, p < 0.0001), as well as poor DFS (HR = 1.55, 95% CI = 1.38-1.74, p < 0.0001). Conclusion: PNI occurrence is significantly related to tumor stage, LVI, lymph node metastasis, OS, and DFS. These results indicate that PNI can serve as an indicator of high malignant degree and poor prognosis in EC.

4.
Neuron ; 110(14): 2268-2282.e4, 2022 07 20.
Article in English | MEDLINE | ID: mdl-35550066

ABSTRACT

Colorectal cancer stem cells (CSCs) contribute to colorectal tumorigenesis and metastasis. Colorectal CSCs reside within specialized niches and harbor self-renewal and differentiation capacities. However, the niche regulations of CSCs remain unclear. Here, we show that intestinal nerve cells are required for CSC self-renewal and colorectal tumorigenesis. Enteric serotonergic neurons produce 5-hydroxytryptamine (5-HT) to function as a modulator of CSC self-renewal. 5-HT receptors HTR1B/1D/1F are highly expressed in colorectal CSCs and engage with 5-HT to initiate Wnt/ß-catenin signaling. Mechanistically, colorectal cancer (CRC)-enriched microbiota metabolite isovalerate suppresses the enrichment of the NuRD complex onto Tph2 promoter to initiate Tph2 expression, leading to 5-HT production. 5-HT signaling is correlated with CRC severity. Blocking 5-HT signaling in mice not only inhibits the self-renewal of colorectal CSCs but also displays therapeutic efficacy against CRC tumors. Our findings reveal a cross talk between intestinal neurons and tumor cells that serves as an additional layer for CSC regulation.


Subject(s)
Cell Self Renewal , Colorectal Neoplasms , Animals , Carcinogenesis , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Mice , Serotonergic Neurons/metabolism , Serotonin , Wnt Signaling Pathway
5.
Adv Sci (Weinh) ; 9(13): e2105160, 2022 05.
Article in English | MEDLINE | ID: mdl-35233964

ABSTRACT

Colorectal cancer (CRC) is one of the most common tumors around the world. Circular RNA is widely involved in tumor progression via unclear mechanisms. Here, circREEP3 is found to be upregulated in CRC tissues. circREEP3 upregulation predicts poor patient survival. circREEP3 knockout suppresses CRC tumorigenesis and metastasis, and impairs stem cell-like phenotype. Mechanistically, circREEP3 recruits the chromatin remodeling protein CHD7 to FKBP10 promoter and activates its transcription. Moreover, circREEP3 restricts RIG-1-dependent antitumor immunity. FKBP10 is highly expressed in CRC tissues and associated with poor prognosis. FKBP10 ectopic expression partially rescues the potential of proliferation and metastasis in circREEP3-deficient CRC cells. Thus, the findings support circREEP3-FKBP10 axis drives CRC progression and may be a critical prognostic marker.


Subject(s)
Colorectal Neoplasms , Gene Expression Regulation, Neoplastic , Carcinogenesis/genetics , Cell Proliferation/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic/genetics , Humans , RNA, Circular/genetics , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism
6.
Br J Cancer ; 126(7): 1037-1046, 2022 04.
Article in English | MEDLINE | ID: mdl-34912075

ABSTRACT

BACKGROUND: Due to the high recurrence and low 5-year survival rates of esophageal squamous cell carcinoma (ESCC) after treatment, the discovery of novel drugs for recurrence chemoprevention is of particular importance. METHODS: We screened the FDA-approved drug library and found that Nuplazid, an atypical antipsychotic that acts as an effective 5-HT 2 A receptor inverse agonist, could potentially exert anticancer effects in vitro and in vivo on ESCC. RESULTS: Pull-down results indicated that Nuplazid binds with p21-activated kinase 4 (PAK4), and a kinase assay showed that Nuplazid strongly suppressed PAK4 kinase activity. Moreover, Nuplazid exhibited inhibitory effects on ESCC in vivo. CONCLUSIONS: Our findings indicate that Nuplazid can suppress ESCC progression through targeting PAK4.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Cell Line, Tumor , Cell Proliferation , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/metabolism , Gene Expression Regulation, Neoplastic , Humans , Piperidines , Urea/analogs & derivatives , p21-Activated Kinases/metabolism
7.
Mol Cancer ; 20(1): 132, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34649567

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is one of the most intractable tumors in the world due to its high rate of recurrence and heterogeneity. Liver cancer initiating cells also called cancer stem cells (CSCs) play a critical role in resistance against typical therapy and high tumor-initiating potential. However, the role of the novel circular RNA (circRNA) circIPO11 in the maintenance of liver cancer initiating cells remains elusive. METHODS: CircRNAs highly conserved in humans and mice were identified from 3 primary HCC samples by circRNA array. The expression and function of circIPO11 were further evaluated by Northern blot, limiting dilution xenograft analysis, chromatin isolation by RNA purification-PCR assay (ChIRP) and HCC patient-derived tumor cells (PDC) models. CircIpo11 knockout (KO) mice were generated by a CRISPR/Cas9 technology. RESULTS: CircIPO11 is highly expressed in HCC tumor tissues and liver CSCs. CircIPO11 is required for the self-renewal maintenance of liver CSCs to initiate HCC development. Mechanistically, circIPO11 recruits TOP1 to GLI1 promoter to trigger its transcription, leading to the activation of Hedgehog signaling. Moreover, GLI1 is also highly expressed in HCC tumor tissues and liver CSCs, and TOP1 expression levels positively correlate with the metastasis, recurrence and survival of HCC patients. Additionally, circIPO11 knockout in mice suppresses the progression of chemically induced liver cancer development. CONCLUSION: Our findings reveal that circIPO11 drives the self-renewal of liver CSCs and promotes the propagation of HCC via activating Hedgehog signaling pathway. Antisense oligonucleotides (ASOs) against circIPO11 combined with TOP1 inhibitor camptothecin (CPT) exert synergistic antitumor effect. Therefore, circIPO11 and the Hedgehog signaling pathway may provide new potential targets for the treatment of HCC patients.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Self Renewal/genetics , Hedgehog Proteins/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , RNA, Circular , beta Karyopherins/genetics , Animals , Biomarkers, Tumor , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Disease Models, Animal , Disease Susceptibility , Gene Dosage , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Liver Neoplasms/pathology , Mice , Mice, Knockout , Neoplastic Stem Cells/pathology , Promoter Regions, Genetic , Signal Transduction
9.
Nat Commun ; 11(1): 4076, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32796851

ABSTRACT

Group 3 innate lymphoid cells (ILC3) are an important regulator for immunity, inflammation and tissue homeostasis in the intestine, but how ILC3 activation is regulated remains elusive. Here we identify a new circular RNA (circRNA) circKcnt2 that is induced in ILC3s during intestinal inflammation. Deletion of circKcnt2 causes gut ILC3 activation and severe colitis in mice. Mechanistically, circKcnt2, as a nuclear circRNA, recruits the nucleosome remodeling deacetylase (NuRD) complex onto Batf promoter to inhibit Batf expression; this in turn suppresses Il17 expression and thereby ILC3 inactivation to promote innate colitis resolution. Furthermore, Mbd3-/-Rag1-/- and circKcnt2-/-Rag1-/- mice develop severe innate colitis following dextran sodium sulfate (DSS) treatments, while simultaneous deletion of Batf promotes colitis resolution. In summary, our data support a function of the circRNA circKcnt2 in regulating ILC3 inactivation and resolution of innate colitis.


Subject(s)
Colitis/immunology , Colitis/metabolism , Lymphocytes/metabolism , Potassium Channels, Sodium-Activated/metabolism , RNA, Circular/metabolism , Animals , Colitis/pathology , DNA-Binding Proteins/genetics , Disease Models, Animal , Female , Homeodomain Proteins/genetics , Homeostasis , Humans , Immunity, Innate , Inflammation/immunology , Inflammation/pathology , Intestines/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Potassium Channels, Sodium-Activated/genetics , RNA, Circular/genetics , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Transcription Factors/genetics
10.
J Exp Med ; 217(2)2020 02 03.
Article in English | MEDLINE | ID: mdl-31699823

ABSTRACT

All hematopoietic lineages are derived from a limited pool of hematopoietic stem cells (HSCs). Although the mechanisms underlying HSC self-renewal have been extensively studied, little is known about the role of protein glutamylation and deglutamylation in hematopoiesis. Here, we show that carboxypeptidase CCP3 is most highly expressed in BM cells among CCP members. CCP3 deficiency impairs HSC self-renewal and hematopoiesis. Deubiquitinase BAP1 is a substrate for CCP3 in HSCs. BAP1 is glutamylated at Glu651 by TTLL5 and TTLL7, and BAP1-E651A mutation abrogates BAP1 glutamylation. BAP1 glutamylation accelerates its ubiquitination to trigger its degradation. CCP3 can remove glutamylation of BAP1 to promote its stability, which enhances Hoxa1 expression, leading to HSC self-renewal. Bap1E651A mice produce higher numbers of LT-HSCs and peripheral blood cells. Moreover, TTLL5 and TTLL7 deficiencies sustain BAP1 stability to promote HSC self-renewal and hematopoiesis. Therefore, glutamylation and deglutamylation of BAP1 modulate HSC self-renewal and hematopoiesis.


Subject(s)
Cell Self Renewal/genetics , Glutamic Acid/metabolism , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , Ubiquitination/genetics , Animals , Bone Marrow Transplantation , CRISPR-Cas Systems , Carrier Proteins/metabolism , Cells, Cultured , Female , Granzymes/metabolism , HEK293 Cells , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mice , Mice, Knockout , Peptide Synthases/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/genetics
11.
J Exp Med ; 216(11): 2653-2668, 2019 11 04.
Article in English | MEDLINE | ID: mdl-31434684

ABSTRACT

Innate lymphoid cells (ILCs) play critical roles in defending infections and maintaining mucosal homeostasis. All ILCs arise from common lymphoid progenitors (CLPs) in bone marrow. However, how CLPs stratify and differentiate into ILC lineages remains elusive. Here, we showed that Yeats4 is highly expressed in ILCs and their progenitors. Yeats4 conditional KO in the hematopoietic system causes decreased numbers of ILCs and impairs their effector functions. Moreover, Yeats4 regulates α4ß7 + CLP differentiation toward common helper ILC progenitors (CHILPs). Mechanistically, Yeats4 recruits the Dot1l-RNA Pol II complex onto Lmo4 promoter through recognizing H3K27ac modification to initiate Lmo4 transcription in α4ß7 + CLPs. Additionally, Lmo4 deficiency also impairs ILC lineage differentiation and their effector functions. Collectively, the Yeats4-Lmo4 axis is required for ILC lineage commitment.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cell Lineage/genetics , LIM Domain Proteins/genetics , Lymphocytes/metabolism , Transcription Factors/genetics , Transcription, Genetic , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Differentiation/genetics , Female , LIM Domain Proteins/deficiency , LIM Domain Proteins/metabolism , Lymphocytes/cytology , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/metabolism , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Mice, Transgenic , Transcription Factors/metabolism
12.
Nat Immunol ; 20(2): 183-194, 2019 02.
Article in English | MEDLINE | ID: mdl-30643264

ABSTRACT

Intestinal stem cells (ISCs) are maintained by stemness signaling for precise modulation of self-renewal and differentiation under homeostasis. However, the way in which intestinal immune cells regulate the self-renewal of ISCs remains elusive. Here we found that mouse and human Lgr5+ ISCs showed high expression of the immune cell-associated circular RNA circPan3 (originating from the Pan3 gene transcript). Deletion of circPan3 in Lgr5+ ISCs impaired their self-renewal capacity and the regeneration of gut epithelium in a manner dependent on immune cells. circPan3 bound mRNA encoding the cytokine IL-13 receptor subunit IL-13Rα1 (Il13ra1) in ISCs to increase its stability, which led to the expression of IL-13Rα1 in ISCs. IL-13 produced by group 2 innate lymphoid cells in the crypt niche engaged IL-13Rα1 on crypt ISCs and activated signaling mediated by IL-13‒IL-13R, which in turn initiated expression of the transcription factor Foxp1. Foxp1 is associated with ß-catenin in rendering its nuclear translocation, which caused activation of the ß-catenin pathway and the maintenance of Lgr5+ ISCs.


Subject(s)
Cell Self Renewal/immunology , Interleukin-13/metabolism , Intestinal Mucosa/immunology , RNA/metabolism , Stem Cells/physiology , Animals , Carrier Proteins/genetics , Cell Differentiation/immunology , Cell Self Renewal/genetics , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/immunology , Dextran Sulfate/toxicity , Disease Models, Animal , Female , Humans , Interleukin-13/immunology , Interleukin-13 Receptor alpha1 Subunit/genetics , Interleukin-13 Receptor alpha1 Subunit/immunology , Interleukin-13 Receptor alpha1 Subunit/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , RNA/genetics , RNA/immunology , RNA, Circular , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/metabolism , Regeneration/genetics , Regeneration/immunology , Signal Transduction/genetics , Signal Transduction/immunology , beta Catenin/immunology , beta Catenin/metabolism
13.
J Hepatol ; 70(5): 918-929, 2019 05.
Article in English | MEDLINE | ID: mdl-30582981

ABSTRACT

BACKGROUND & AIMS: Liver cancer is the second leading cause of cancer death worldwide. Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer in adults. The aim of this study was to define the role of the long non-coding RNA lncHDAC2 in the tumorigenesis of HCC. METHODS: CD13+CD133+ cells (hereafter called liver cancer stem cells [CSCs]) and CD13-CD133- cells (referred to as non-CSCs) were sorted from 3 primary HCC tumor tissues and followed by transcriptome microarray. The expression and function of lncHDAC2 were further assessed by northern blot, sphere formation and xenograft tumor models. RESULTS: LncHDAC2 is highly expressed in HCC tumors and liver CSCs. LncHDAC2 promotes the self-renewal of liver CSCs and tumor propagation. In liver CSCs, lncHDAC2 recruits the NuRD complex onto the promoter of PTCH1 to inhibit its expression, leading to activation of Hedgehog signaling. Moreover, HDAC2 expression levels are positively related to HCC severity and PTCH1 levels are negatively related to HCC severity. Additionally, the Smo inhibitor cyclopamine was shown to impair the self-renewal of liver CSCs and suppress tumor propagation. CONCLUSION: Our findings reveal that lncHDAC2 promotes the self-renewal of liver CSCs and tumor propagation by activating the Hedgehog signaling pathway. Downregulating lncHDAC2 is a promising antitumor strategy in HCC. LAY SUMMARY: Liver cancer stem cells harbor high tumor-initiating potential and confer resistance to typical therapies, but the mechanism underlying their self-renewal remains elusive. LncHDAC2 augments the self-renewal of these cells, promoting tumor propagation. In liver cancer stem cells, lncHDAC2 activates Hedgehog signaling to initiate liver tumorigenesis. Therefore, lncHDAC2 and the Hedgehog signaling pathway may serve as biomarkers and potential drug targets for hepatocellular carcinoma.


Subject(s)
Cell Self Renewal , Hedgehog Proteins/physiology , Histone Deacetylase 2/genetics , Liver Neoplasms/pathology , Neoplastic Stem Cells/physiology , RNA, Long Noncoding/physiology , Signal Transduction , Cell Line, Tumor , Humans , Patched-1 Receptor/genetics , Promoter Regions, Genetic , Signal Transduction/physiology
14.
Biomed Pharmacother ; 108: 845-851, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30372896

ABSTRACT

Glioma is one of the most aggressive and lethal human cancers with a low cure rate. LASP1 plays an oncogenic role in multiple human cancers; however, its role in glioma remains largely unknown. Here, we found that LASP1 was highly expressed in glioma tissue samples. Functionally, knockdown of LASP1 significantly suppressed glioma cell proliferation and migration in vitro and tumorigenicity in vivo. These effects were found to be mechanistically associated with suppression of AKT activity. Furthermore, we identified LASP1 as a direct target of miR-377-3p. Overexpression of miR-377-3p reduced the expression of LASP1 and suppressed the proliferation and migration of glioma cells. Restoration of LASP1 expression in miR-377-3p-overexpressing cells attenuated the inhibition of glioma cell malignancy and reversed the dephosphorylation of AKT. Taken together, our results suggest that LASP1 activates the PI3K/AKT signaling pathway and is downregulated by miR-377-3p during glioma progression. These data provide a new possible therapeutic target in glioma.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cell Movement/genetics , Cell Proliferation/genetics , Cytoskeletal Proteins/genetics , Glioma/genetics , Glioma/pathology , LIM Domain Proteins/genetics , MicroRNAs/genetics , Animals , Carcinogenesis/genetics , Cell Line, Tumor , Down-Regulation/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics
15.
Nat Cell Biol ; 20(10): 1134-1144, 2018 10.
Article in English | MEDLINE | ID: mdl-30224759

ABSTRACT

The intestinal epithelium harbours remarkable self-renewal capacity that is driven by Lgr5+ intestinal stem cells (ISCs) at the crypt base. However, the molecular mechanism controlling Lgr5+ ISC stemness is incompletely understood. We show that a Gata6 long noncoding RNA (lncGata6) is highly expressed in ISCs. LncGata6 knockout or conditional knockout in ISCs impairs the stemness of ISCs and epithelial regeneration. Mechanistically, lncGata6 recruits the NURF complex onto the Ehf promoter to induce its transcription, which promotes the expression of Lgr4/5 to enhance Wnt signalling activation. Moreover, the human orthologue lncGATA6 is highly expressed in the cancer stem cells of colorectal cancer and promotes tumour initiation and progression. Antisense oligonucleotides against lncGATA6 exhibit strong therapeutic efficacy on colorectal cancer. Thus, targeting lncGATA6 will have potential clinical applications in colorectal cancer treatment as an ideal therapeutic target.


Subject(s)
Cell Transformation, Neoplastic/genetics , Intestinal Mucosa/metabolism , RNA, Long Noncoding/genetics , Stem Cells/metabolism , Animals , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Gene Expression Profiling/methods , Humans , Intestinal Mucosa/cytology , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mice, Transgenic , RNA, Long Noncoding/metabolism , Transplantation, Heterologous , Tumor Cells, Cultured
16.
J Hepatol ; 69(4): 861-872, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29653123

ABSTRACT

BACKGROUND & AIMS: Long noncoding RNAs (lncRNAs) play important roles in various biological processes, regulating gene expression by diverse mechanisms. However, how lncRNAs regulate liver repopulation is unknown. Herein, we aimed to identify lncRNAs that regulate liver repopulation and elucidate the signaling pathways involved. METHODS: Herein, we performed 70% partial hepatectomy in wild-type and gene knockout mice. We then performed transcriptomic analyses to identify a divergent lncRNA termed lncHand2 that is highly expressed during liver regeneration. RESULTS: LncHand2 is constitutively expressed in the nuclei of pericentral hepatocytes in mouse and human livers. LncHand2 knockout abrogates liver regeneration and repopulation capacity. Mechanistically, lncHand2 recruits the Ino80 remodeling complex to initiate expression of Nkx1-2 in trans, which triggers c-Met (Met) expression in hepatocytes. Finally, knockout of both Nkx1-2 and c-Met causes more severe liver injury and poorer repopulation ability. Thus, lncHand2 promotes liver repopulation via initiating Nkx1-2-induced c-Met signaling. CONCLUSIONS: Our findings reveal that lncHand2 acts as a critical mediator regulating liver repopulation. It does this by inducing Nkx1-2 expression, which in turn triggers c-Met signaling. LAY SUMMARY: Long noncoding RNAs play important roles in various biological processes. While long noncoding RNAs do not directly code proteins, they can regulate gene expression by diverse mechanisms. We identified the long noncoding RNA, termed lncHand2 because of its proximity to the gene Hand2, to be an important determinant of liver regeneration through c-Met signaling.


Subject(s)
Liver Regeneration , Proto-Oncogene Proteins c-met/physiology , RNA, Long Noncoding/physiology , Signal Transduction/physiology , ATPases Associated with Diverse Cellular Activities , Adenosine Triphosphatases/physiology , Animals , DNA-Binding Proteins , Female , Gene Expression Profiling , Hepatocytes/metabolism , Homeodomain Proteins/genetics , Humans , Male , Mice , Mice, Knockout , Nuclear Proteins/genetics , RNA, Long Noncoding/analysis , Transcription Factors/genetics
17.
Nat Commun ; 9(1): 1261, 2018 03 28.
Article in English | MEDLINE | ID: mdl-29593216

ABSTRACT

Temporal and spatial-specific regulation of pluripotency networks is largely dependent on the precise modifications of core transcription factors. Misregulation of glutamylation is implicated in severe physiological abnormalities. However, how glutamylation regulates cell reprogramming and pluripotency networks remains elusive. Here we show that cytosolic carboxypeptidases 1 (CCP1) or CCP6 deficiency substantially promotes induced pluripotent cell (iPSC) induction and pluripotency of embryonic stem cells (ESCs). Klf4 polyglutamylation at Glu381 by tubulin tyrosine ligase-like 4 (TTLL4) and TTLL1 during cell reprogramming impedes its lysine 48-linked ubiquitination and sustains Klf4 stability. Klf4-E381A knockin mice display impaired blastocyst development and embryonic lethality. Deletion of TTLL4 or TTLL1 abrogates cell reprogramming and early embryogenesis. Thus, Klf4 polyglutamylation plays a critical role in the regulation of cell reprogramming and pluripotency maintenance.


Subject(s)
Cellular Reprogramming , Gene Expression Regulation, Developmental , Glutamine/chemistry , Induced Pluripotent Stem Cells/cytology , Kruppel-Like Transcription Factors/metabolism , Peptide Synthases/metabolism , Animals , Carboxypeptidases/deficiency , Cell Differentiation , Embryonic Stem Cells/cytology , Female , Fibroblasts/cytology , Gene Deletion , Gene Knock-In Techniques , HEK293 Cells , Humans , Kruppel-Like Factor 4 , Mice , Nerve Tissue Proteins/deficiency , Peptides/chemistry
18.
Clin Cancer Res ; 23(21): 6673-6685, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28765327

ABSTRACT

Purpose: Bladder cancer is one of the most common urinary malignancies worldwide characterized by a high rate of recurrence and no targeted therapy method. Bladder cancer stem cells (BCSCs) play a crucial role in tumor initiation, metastasis, and drug resistance. However, the regulatory signaling and self-renewal mechanisms of BCSCs remain largely unknown. Here, we identified a novel signal, the KMT1A-GATA3-STAT3 circuit, which promoted the self-renewal and tumorigenicity of human BCSCs.Experimental Design: In a discovery step, human BCSCs and bladder cancer non-stem cells (BCNSCs) isolated from primary bladder cancer samples #1 and #2, and the bladder cancer cell line EJ were analyzed by transcriptome microarray. In a validation step, 10 paired bladder cancer and normal tissues, different tumor cell lines, the public microarray datasets of human bladder cancer, and The Cancer Genome Atlas database were applied for the verification of gene expression.Results: KMT1A was highly expressed and responsible for the increase of tri-methylating lysine 9 of histone H3 (H3K9me3) modification in BCSCs compared with either BCNSCs or normal bladder tissue. GATA3 bound to the -1710∼-1530 region of STAT3 promoter and repressed its transcription. H3K9me3 modification on the -1351∼-1172bp region of the GATA3 promoter mediated by KMT1A repressed the transcription of GATA3 and upregulated the expression of STAT3. In addition, the activated STAT3 triggered self-renewal of BCSCs. Furthermore, depletion of KMT1A or STAT3 abrogated the formation of BCSC tumorspheres and xenograft tumors.Conclusions: KMT1A positively regulated the self-renewal and tumorigenicity of human BCSCs via KMT1A-GATA3-STAT3 circuit, in which KMT1A could be a promising target for bladder cancer therapy. Clin Cancer Res; 23(21); 6673-85. ©2017 AACR.


Subject(s)
GATA3 Transcription Factor/genetics , Methyltransferases/genetics , Repressor Proteins/genetics , STAT3 Transcription Factor/genetics , Urinary Bladder Neoplasms/genetics , Adult , Aged , Cell Line, Tumor , Cell Self Renewal/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/pathology , Signal Transduction/genetics , Urinary Bladder Neoplasms/pathology
19.
Eur Urol ; 71(1): 8-12, 2017 01.
Article in English | MEDLINE | ID: mdl-27387124

ABSTRACT

Cancer stem cells are considered responsible for many important aspects of tumors such as their self-renewal, tumor-initiating, drug-resistance and metastasis. However, the genetic basis and origination of human bladder cancer stem cells (BCSCs) remains unknown. Here, we conducted single-cell sequencing on 59 cells including BCSCs, bladder cancer non-stem cells (BCNSCs), bladder epithelial stem cells (BESCs) and bladder epithelial non-stem cells (BENSCs) from three bladder cancer (BC) specimens. Specifically, BCSCs demonstrate clonal homogeneity and suggest their origin from BESCs or BCNSCs through phylogenetic analysis. Moreover, 21 key altered genes were identified in BCSCs including six genes not previously described in BC (ETS1, GPRC5A, MKL1, PAWR, PITX2 and RGS9BP). Co-mutations of ARID1A, GPRC5A and MLL2 introduced by CRISPR/Cas9 significantly enhance the capabilities of self-renewal and tumor-initiating of BCNSCs. To our knowledge, our study first provides an overview of the genetic basis of human BCSCs with single-cell sequencing and demonstrates the biclonal origin of human BCSCs via evolution analysis. PATIENT SUMMARY: Human bladder cancer stem cells show the high level of consistency and may derived from bladder epithelial stem cells or bladder cancer non-stem cells. Mutations of ARID1A, GPRC5A and MLL2 grant bladder cancer non-stem cells the capability of self-renewal.


Subject(s)
DNA-Binding Proteins/genetics , Neoplasm Proteins/genetics , Neoplastic Stem Cells/metabolism , Nuclear Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Transcription Factors/genetics , Urinary Bladder Neoplasms/genetics , Epithelial Cells/metabolism , Epithelial Cells/physiology , Humans , Mutation , Neoplastic Stem Cells/physiology , Polymorphism, Single Nucleotide , Urinary Bladder Neoplasms/physiopathology , Exome Sequencing
20.
Oncotarget ; 7(3): 2629-45, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26625313

ABSTRACT

Bladder cancer (BC) is distinguished by high rate of recurrence after surgery, but the underlying mechanisms remain poorly understood. Here we performed the whole-exome sequencing of 37 BC individuals including 20 primary and 17 recurrent samples in which the primary and recurrent samples were not from the same patient. We uncovered that MLL, EP400, PRDM2, ANK3 and CHD5 exclusively altered in recurrent BCs. Specifically, the recurrent BCs and bladder cancer cells with MLL mutation displayed increased histone H3 tri-methyl K4 (H3K4me3) modification in tissue and cell levels and showed enhanced expression of GATA4 and ETS1 downstream. What's more, MLL mutated bladder cancer cells obtained with CRISPR/Cas9 showed increased ability of drug-resistance to epirubicin (a chemotherapy drug for bladder cancer) than wild type cells. Additionally, the BC patients with high expression of GATA4 and ETS1 significantly displayed shorter lifespan than patients with low expression. Our study provided an overview of the genetic basis of recrudescent bladder cancer and discovered that genetic alterations of MLL were involved in BC relapse. The increased modification of H3K4me3 and expression of GATA4 and ETS1 would be the promising targets for the diagnosis and therapy of relapsed bladder cancer.


Subject(s)
Biomarkers, Tumor/genetics , Exome/genetics , High-Throughput Nucleotide Sequencing/methods , Histone-Lysine N-Methyltransferase/genetics , Mutation/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Neoplasm Recurrence, Local/genetics , Urinary Bladder Neoplasms/genetics , Animals , Apoptosis , Blotting, Western , Case-Control Studies , Cell Proliferation , Chromatin Immunoprecipitation , Female , GATA4 Transcription Factor/genetics , Humans , Immunoenzyme Techniques , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Recurrence, Local/pathology , Prognosis , Proto-Oncogene Protein c-ets-1/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Tumor Cells, Cultured , Urinary Bladder Neoplasms/pathology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...