Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Mol Carcinog ; 62(2): 145-159, 2023 02.
Article in English | MEDLINE | ID: mdl-36218231

ABSTRACT

Doublecortin like kinase 1 (DCLK1) plays a crucial role in several cancers including colon and pancreatic adenocarcinomas. However, its role in squamous cell carcinoma (SCC) remains unknown. To this end, we examined DCLK1 expression in head and neck SCC (HNSCC) and anal SCC (ASCC). We found that DCLK1 is elevated in patient SCC tissue, which correlated with cancer progression and poorer overall survival. Furthermore, DCLK1 expression is significantly elevated in human papilloma virus negative HNSCC, which are typically aggressive with poor responses to therapy. To understand the role of DCLK1 in tumorigenesis, we used specific shRNA to suppress DCLK1 expression. This significantly reduced tumor growth, spheroid formation, and migration of HNSCC cancer cells. To further the translational relevance of our studies, we sought to identify a selective DCLK1 inhibitor. Current attempts to target DCLK1 using pharmacologic approaches have relied on nonspecific suppression of DCLK1 kinase activity. Here, we demonstrate that DiFiD (3,5-bis [2,4-difluorobenzylidene]-4-piperidone) binds to DCLK1 with high selectivity. Moreover, DiFiD mediated suppression of DCLK1 led to G2/M arrest and apoptosis and significantly suppressed tumor growth of HNSCC xenografts and ASCC patient derived xenografts, supporting that DCLK1 is critical for SCC growth.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Humans , Apoptosis , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Doublecortin-Like Kinases , G2 Phase Cell Cycle Checkpoints , Intracellular Signaling Peptides and Proteins/genetics , Protein Serine-Threonine Kinases/metabolism , Squamous Cell Carcinoma of Head and Neck/genetics , Animals
2.
Cell Mol Gastroenterol Hepatol ; 13(2): 425-440, 2022.
Article in English | MEDLINE | ID: mdl-34563711

ABSTRACT

BACKGROUND & AIMS: Single immunoglobulin interleukin-1-related receptor (SIGIRR) is a major inhibitor of Toll-like receptor signaling. Our laboratory identified a novel SIGIRR stop mutation (p.Y168X) in an infant who died of severe necrotizing enterocolitis (NEC). Herein, we investigated the mechanisms by which SIGIRR mutations induce Toll-like receptor hyper-responsiveness in the neonatal gut, disrupting postnatal intestinal adaptation. METHODS: Clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 was used to generate transgenic mice encoding the SIGIRR p.Y168X mutation. Ileal lysates, mouse intestinal epithelial cell (IEC) lysates, and intestinal sections were used to assess inflammation, signal transducer and activator of transcription 3 (STAT3) phosphorylation, microRNA (miRNA), and interleukin-1-related-associated kinase 1 (IRAK1) expression. Western blot, quantitative reverse-transcription polymerase chain reaction(qRT-PCR), and luciferase assays were performed to investigate SIGIRR-STAT3 signaling in human intestinal epithelial cells (HIEC) expressing wild-type or SIGIRR (p.Y168X) plasmids. RESULTS: SigirrTg mice showed increased intestinal inflammation and nuclear factor-κB activation concomitant with decreased IEC expression of miR-146a and miR-155. Mechanistic studies in HIECs showed that although SIGIRR induced STAT3-mediated expression of miR-146a and miR-155, the p.Y168X mutation disrupted SIGIRR-mediated STAT3-dependent miRNA expression. Chromatin immunoprecipitation and luciferase assays showed that SIGIRR activation of STAT3-induced miRNA expression is dependent on IRAK1. Both in HIECs and in the mouse intestine, decreased expression of miR-146a observed with the p.Y168X mutation increased expression of IRAK1, a protein whose down-regulation is important for postnatal gut adaptation. CONCLUSIONS: Our results uncover a novel pathway (SIGIRR-STAT3-miRNA-IRAK1 repression) by which SIGIRR regulates postnatal intestine adaptation, which is disrupted by a SIGIRR mutation identified in human NEC. These data provide new insights into how human genetic mutations in SIGIRR identified in NEC result in loss of postnatal intestinal immune tolerance.


Subject(s)
Enterocolitis, Necrotizing , MicroRNAs , Animals , Humans , Immunoglobulins/genetics , Immunoglobulins/metabolism , Mice , MicroRNAs/genetics , Mutation/genetics , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
4.
Cell Death Discov ; 7(1): 169, 2021 Jun 17.
Article in English | MEDLINE | ID: mdl-34226497

ABSTRACT

Alternative promoter usage generates long and short isoforms (DCLK1-L and DCLK1-S) of doublecortin-like kinase-1 (DCLK1). Tight control of Notch signaling is important to prevent and restitute inflammation in the intestine. Our aim was to investigate whether Notch1-DCLK1 axis regulates the mucosal immune responses to infection and whether this is phenocopied in human models of colitis. In the FFPE (formalin-fixed paraffin-embedded) sections prepared from the colons of ulcerative colitis (UC) and immune-mediated colitis (IRAEC) patients, expression of DCLK1 isoforms correlated positively with Notch1 and negatively with a transcriptional repressor, FoxD3 (Forkhead Box D3). DCLK1 protein staining in these sections was predominantly sub-epithelial (stromal) wherein DCLK1 co-localized with NICD, CD68, CD11c, and neutrophil elastase (NE). NE also co-stained with Citrullinated-H3 indicating the presence of neutrophil extracellular traps. In human neutrophils, elevated levels of DCLK1-S, CXCL-10, Ly6G, MPO, NE, and Notch1/2 in LPS-treated cells were inhibited when LPS was added in conjunction with Notch blocker dibenzazepine (DBZ; LPS + DBZ group). In CR-infected Rag1-/- mice, higher levels of DCLK1 in the colonic crypts were inhibited when mice received DBZ for 10 days coincident with significant dysbiosis, barrier disruption, and colitis. Concurrently, DCLK1 immunoreactivity shifted toward the stroma in CR + DBZ mice with predominance of DCLK1-S that coincided with higher Notch1 levels. Upon antibiotic treatment, partial restoration of crypt DCLK1, reduction in MPO activity, and increased survival followed. When intestinal epithelial cell-specific Dclk1-knockout (Dclk1ΔIEC) or Dclk1ΔIEC;Rag1-/- double knockout (DKO) mice were infected with CR and given a single dose of DBZ, they developed barrier defect and severe colitis with higher levels of stromal DCLK1-S, Ly6G, NE, and Notch1. We therefore propose that, by regulating the mucosal immune responses, the Notch-DCLK1 axis may be integral to the development of murine or human colitis.

5.
Int J Mol Sci ; 23(1)2021 Dec 29.
Article in English | MEDLINE | ID: mdl-35008767

ABSTRACT

Decreases in short-chain-fatty-acids (SCFAs) are linked to inflammatory bowel disease (IBD). Yet, the mechanisms through which SCFAs promote wound healing, orchestrated by intestinal stem cells, are poorly understood. We discovered that, in mice with Citrobacter rodentium (CR)-induced infectious colitis, treatment with Pectin and Tributyrin diets reduced the severity of colitis by restoring Firmicutes and Bacteroidetes and by increasing mucus production. RNA-seq in young adult mouse colon (YAMC) cells identified higher expression of Lgr4, Lgr6, DCLK1, Muc2, and SIGGIR after Butyrate treatment. Lineage tracing in CR-infected Lgr5-EGFP-IRES-CreERT2/ROSA26-LacZ (Lgr5-R) mice also revealed an expansion of LacZ-labeled Lgr5(+) stem cells in the colons of both Pectin and Tributyrin-treated mice compared to control. Interestingly, gut microbiota was required for Pectin but not Tributyrin-induced Lgr5(+) stem cell expansion. YAMC cells treated with sodium butyrate exhibited increased Lgr5 promoter reporter activity due to direct Butyrate binding with Lgr5 at -4.0 Kcal/mol, leading to thermal stabilization. Upon ChIP-seq, H3K4me3 increased near Lgr5 transcription start site that contained the consensus binding motif for a transcriptional activator of Lgr5 (SPIB). Thus, a multitude of effects on gut microbiome, differential gene expression, and/or expansion of Lgr5(+) stem cells seem to underlie amelioration of colitis following dietary intervention.


Subject(s)
Colitis/microbiology , Colitis/pathology , Diet , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae Infections/pathology , Microbiota , Stem Cells/pathology , Animals , Biodiversity , Butyrates/pharmacology , Cell Proliferation/drug effects , Cell Proliferation/genetics , Citrobacter rodentium/physiology , Epithelium/pathology , Fermentation , Gene Expression Profiling , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Mice, Inbred C57BL , Mucin-2/metabolism , Pectins/pharmacology , Promoter Regions, Genetic/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Regeneration/drug effects , Transcription, Genetic/drug effects , Triglycerides/pharmacology
6.
PLoS One ; 13(11): e0206701, 2018.
Article in English | MEDLINE | ID: mdl-30383855

ABSTRACT

Intestinal mucus layer disruption and gut microflora modification in conjunction with tight junction (TJ) changes can increase colonic permeability that allows bacterial dissemination and intestinal and systemic disease. We showed previously that Citrobacter rodentium (CR)-induced colonic crypt hyperplasia and/or colitis is regulated by a functional cross-talk between the Notch and Wnt/ß-catenin pathways. In the current study, mucus analysis in the colons of CR-infected (108 CFUs) and Notch blocker Dibenzazepine (DBZ, i.p.; 10µmol/Kg b.w.)-treated mice revealed significant alterations in the composition of trace O-glycans and complex type and hybrid N-glycans, compared to CR-infected mice alone that preceded/accompanied alterations in 16S rDNA microbial community structure and elevated EUB338 staining. While mucin-degrading bacterium, Akkermansia muciniphila (A. muciniphila) along with Enterobacteriaceae belonging to Proteobacteria phyla increased in the feces, antimicrobial peptides Angiogenin-4, Intelectin-1 and Intelectin-2, and ISC marker Dclk1, exhibited dramatic decreases in the colons of CR-infected/DBZ-treated mice. Also evident was a loss of TJ and adherens junction protein immuno-staining within the colonic crypts that negatively impacted paracellular barrier. These changes coincided with the loss of Notch signaling and exacerbation of mucosal injury. In response to a cocktail of antibiotics (Metronidazole/ciprofloxacin) for 10 days, there was increased survival that coincided with: i) decreased levels of Proteobacteria, ii) elevated Dclk1 levels in the crypt and, iii) reduced paracellular permeability. Thus, enteric infections that interfere with Notch activity may promote mucosal dysbiosis that is preceded by changes in mucus composition. Controlled use of antibiotics seems to alleviate gut dysbiosis but may be insufficient to promote colonic crypt regeneration.


Subject(s)
Citrobacter rodentium , Colon/immunology , Enterobacteriaceae Infections/immunology , Mucus/immunology , Receptors, Notch/antagonists & inhibitors , Animals , Animals, Outbred Strains , Colitis/drug therapy , Colitis/immunology , Colitis/pathology , Colon/drug effects , Colon/microbiology , Colon/pathology , Dibenzazepines/pharmacology , Disease Models, Animal , Dysbiosis/drug therapy , Dysbiosis/immunology , Dysbiosis/pathology , Enterobacteriaceae Infections/drug therapy , Enterobacteriaceae Infections/pathology , Enzyme Inhibitors/pharmacology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mucus/microbiology , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction/drug effects , Tight Junctions/drug effects , Tight Junctions/immunology , Tight Junctions/pathology
7.
Nat Commun ; 7: 13561, 2016 11 25.
Article in English | MEDLINE | ID: mdl-27886186

ABSTRACT

The ability of cancer cells to survive and grow in anchorage- and serum-independent conditions is well correlated with their aggressiveness. Here, using a human whole-genome shRNA library, we identify TMIGD3 isoform1 (i1) as a factor that suppresses this ability in osteosarcoma (OS) cells, mainly by inhibiting NF-κB activity. Knockdown of TMIGD3 increases proliferation, tumour formation and metastasis of OS cells. Overexpression of TMIGD3 isoform1 (i1), but not isoform3 (i3) which shares a common C-terminal region, suppresses these malignant properties. Adenosine A3 receptor (A3AR) having an identical N-terminal region shows similar biological profiles to TMIGD3 i1. Protein expression of TMIGD3 and A3AR is lower in human OS tissues than normal tissues. Mechanistically, TMIGD3 i1 and A3AR commonly inhibit the PKA-Akt-NF-κB axis. However, TMIGD3 i1 only partially rescues phenotypes induced by A3AR knockdown, suggesting the presence of distinct pathways. Our findings reveal an unappreciated role for TMIGD3 i1 as a suppressor of NF-κB activity and OS progression.


Subject(s)
Bone Neoplasms/pathology , Lung Neoplasms/pathology , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , NF-kappa B/metabolism , Osteosarcoma/pathology , Receptor, Adenosine A3/metabolism , Animals , Bone Neoplasms/genetics , Cell Line, Tumor , Cell Movement/genetics , Disease Progression , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Membrane Glycoproteins/genetics , Membrane Proteins/genetics , Mice , Mice, Nude , Osteosarcoma/genetics , Osteosarcoma/secondary , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptor, Adenosine A3/genetics , Signal Transduction/genetics , Xenograft Model Antitumor Assays
8.
Microorganisms ; 4(2)2016 Jun 15.
Article in English | MEDLINE | ID: mdl-27681914

ABSTRACT

Inflammatory Bowel Disease (IBD) is a multifactorial disorder that conceptually occurs as a result of altered immune responses to commensal and/or pathogenic gut microbes in individuals most susceptible to the disease. During Crohn's Disease (CD) or Ulcerative Colitis (UC), two components of the human IBD, distinct stages define the disease onset, severity, progression and remission. Epigenetic, environmental (microbiome, metabolome) and nutritional factors are important in IBD pathogenesis. While the dysbiotic microbiota has been proposed to play a role in disease pathogenesis, the data on IBD and diet are still less convincing. Nonetheless, studies are ongoing to examine the effect of pre/probiotics and/or FODMAP reduced diets on both the gut microbiome and its metabolome in an effort to define the healthy diet in patients with IBD. Knowledge of a unique metabolomic fingerprint in IBD could be useful for diagnosis, treatment and detection of disease pathogenesis.

9.
Carcinogenesis ; 37(4): 385-96, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26785732

ABSTRACT

Phytochemicals modulate key cellular signaling pathways and have proven anticancer effects. Alcea rosea(AR; Hollyhock) is an ornamental plant with known anti-inflammatory properties. This study explored its role as an anticancer agent. The AR seed extract (AR extract) inhibited proliferation and colony formation in a dose- and time-dependent manner and promoted apoptosis as was evidenced by cleavage of PARP and increased expression of Bax accompanying reduced levels of BCL-xl protein in HCT116 and SW480 cells, respectively. In addition, AR extract-arrested cells at Go/G1 phase of cell cycle and exhibited decreases in Cyclin D1. AR extract-treated cells exhibited reduced number and size of colonospheres in a dose-dependent manner concomitant with decreases in cancer stem cell (CSC) markers ALDH1A1 and Dclk1. Relative levels of ß-catenin, Notch-ICD, Hes1 and EZH2 were also attenuated by AR extract. TOP-flash reporter activity, a measure of Wnt signaling, decreased significantly in response to treatment while overexpression of wild type but not mutant EZH2, reversed the inhibitory effects. Moreover, WIF1 (a Wnt antagonist) promoter activity increased dramatically following treatment with AR extract which phenocopied increases in WIF1 reporter activity following EZH2 knockdown.In vivo, AR extract attenuated tumor growth due probably to reduced levels of EZH2, ß-catenin, CyclinD1 and Ki-67 along with reduced levels of CSC markers. Since partial purification via HPLC yielded a prominent peak, efforts are underway to identify the active ingredient(s). Taken together, the results clearly suggest that AR extract/active component(s) can be an effective preventative/therapeutic agent to target colon cancer.


Subject(s)
Colonic Neoplasms/pathology , Epigenesis, Genetic , Neoplastic Stem Cells/pathology , Plants , Signal Transduction , Humans
10.
PLoS Genet ; 11(11): e1005638, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26528816

ABSTRACT

Adenomatous polyposis coli (APC) inactivating mutations are present in most human colorectal cancers and some other cancers. The APC protein regulates the ß-catenin protein pool that functions as a co-activator of T cell factor (TCF)-regulated transcription in Wnt pathway signaling. We studied effects of reduced dosage of the Ctnnb1 gene encoding ß-catenin in Apc-mutation-induced colon and ovarian mouse tumorigenesis and cell culture models. Concurrent somatic inactivation of one Ctnnb1 allele, dramatically inhibited Apc mutation-induced colon polyposis and greatly extended Apc-mutant mouse survival. Ctnnb1 hemizygous dose markedly inhibited increases in ß-catenin levels in the cytoplasm and nucleus following Apc inactivation in colon epithelium, with attenuated expression of key ß-catenin/TCF-regulated target genes, including those encoding the EphB2/B3 receptors, the stem cell marker Lgr5, and Myc, leading to maintenance of crypt compartmentalization and restriction of stem and proliferating cells to the crypt base. A critical threshold for ß-catenin levels in TCF-regulated transcription was uncovered for Apc mutation-induced effects in colon epithelium, along with evidence of a feed-forward role for ß-catenin in Ctnnb1 gene expression and CTNNB1 transcription. The active ß-catenin protein pool was highly sensitive to CTNNB1 transcript levels in colon cancer cells. In mouse ovarian endometrioid adenocarcinomas (OEAs) arising from Apc- and Pten-inactivation, while Ctnnb1 hemizygous dose affected ß-catenin levels and some ß-catenin/TCF target genes, Myc induction was retained and OEAs arose in a fashion akin to that seen with intact Ctnnb1 gene dose. Our findings indicate Ctnnb1 gene dose exerts tissue-specific differences in Apc mutation-instigated tumorigenesis. Differential expression of selected ß-catenin/TCF-regulated genes, such as Myc, likely underlies context-dependent effects of Ctnnb1 gene dosage in tumorigenesis.


Subject(s)
Colonic Neoplasms/genetics , Genes, APC , Mutation , Ovarian Neoplasms/genetics , beta Catenin/metabolism , Animals , Female , Mice
11.
Lung Cancer ; 70(2): 136-45, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20207041

ABSTRACT

Epithelial-mesenchymal transition (EMT) is a critical phenotypic alteration of cancer cells that triggers invasion and metastasis. Lung cancer cells often show mesenchymal phenotypes; however, a causative genetic alteration for the induction of EMT in lung cancer cells remains unknown. Recent studies have shown that the LKB1 gene is mutated in up to one-third of lung adenocarcinomas. Therefore, to pursue the possible involvement of LKB1 inactivation in the induction of EMT in lung carcinogenesis, we generated immortalized lung epithelial cells and lung adenocarcinoma cells with stable or transient LKB1 knockdown. LKB1 knockdown increased cell motility and invasiveness, and induced the expression of several mesenchymal marker proteins accompanied by the expression of ZEB1, a transcriptional repressor for E-cadherin and an EMT inducer. In agreement with the recent findings, expression of miR-200a/c was inversely correlated with that of ZEB1 in LKB1 knockdown clones with mesenchymal phenotype. Furthermore, transient knockdown of LKB1 induced ZEB1 mRNA and increased cell motility, and this motility was suppressed by ZEB1 repression. These results strongly indicate that LKB1 inactivation triggers EMT in lung cancer cells through the induction of ZEB1.


Subject(s)
Adenocarcinoma/metabolism , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Lung Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinase Kinases , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Cadherins/metabolism , Cell Line, Transformed , Cell Line, Tumor , Cell Movement/genetics , Cell Transformation, Neoplastic , Cells, Cultured , Epithelial Cells/pathology , Gene Knockdown Techniques , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , MicroRNAs/genetics , Mutation/genetics , Neoplasm Invasiveness/genetics , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/genetics , Transcription Factors/biosynthesis , Transcription Factors/genetics , Zinc Finger E-box-Binding Homeobox 1
12.
Genes Cells ; 7(6): 607-17, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12059963

ABSTRACT

BACKGROUND: The PSD-95 family of proteins possesses multiple protein binding domains, including three PDZ domains, an SH3 domain, a HOOK domain and a guanylate kinase-like (GK) domain. The PSD-95 proteins function as scaffolding proteins that link ion channels such as the N-methyl-d-aspartate-receptors (NMDA-Rs) with cytoskeletal networks and signalling molecules, thereby controlling synaptic plasticity and learning. RESULTS: We found that the PSD-95 family proteins interact via their GK domains with SPA-1-like protein (SPAL), a GTPase-activating protein (GAP) that is specific for Rap1. SPAL was contained within the NMDA-R-PSD-95 complex, and co-localized with PSD-95 and NMDA-R at the synapses in cultured hippocampal neurones. Furthermore, NMDA stimulation induced the dephosphorylation of SPAL in cultured hippocampal neurones. CONCLUSION: Our findings suggest that SPAL may be involved in the NMDA-mediated organization of cytoskeletal networks and signal transduction.


Subject(s)
GTPase-Activating Proteins/metabolism , Hippocampus/metabolism , Nerve Tissue Proteins/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , rap1 GTP-Binding Proteins/metabolism , Animals , Cells, Cultured , Disks Large Homolog 4 Protein , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Membrane Proteins , Neurons , Precipitin Tests , Rats , Two-Hybrid System Techniques
13.
Med Electron Microsc ; 32(1): 20-24, 1999 May.
Article in English | MEDLINE | ID: mdl-11810421

ABSTRACT

Spa-1-like protein (SPAL) is a novel GTPase-activating protein (GAP) family protein. We generated anti-SPAL antibody, and examined localization of SPAL in the rat esophagus and heart by immunofluorescence microscopy. In the esophagus, SPAL was highly expressed in the spinous and granular layers and localized at the cell-cell border of the epithelial cells. SPAL in the spinous layer was almost colocalized with beta-catenin, an intracellular anchor protein of E-cadherin, while colocalization of SPAL with the tight junction protein ZO-1 was shown in the granular layer. In the heart, SPAL was localized at the intercalated disks, and there colocalized with both beta-catenin and ZO-1. In addition, much punctate localization of SPAL was seen along lateral borders of the cardiomyocytes, where beta-catenin and ZO-1 were absent. From these results, it is suggested that SPAL may regulate cell-cell adhesion in the rat esophagus and heart.

SELECTION OF CITATIONS
SEARCH DETAIL
...