Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.351
Filter
1.
Nat Immunol ; 19(8): 859-870, 2018 08.
Article in English | MEDLINE | ID: mdl-30013146

ABSTRACT

IgE is an ancient and conserved immunoglobulin isotype with potent immunological function. Nevertheless, the regulation of IgE responses remains an enigma, and evidence of a role for IgE in host defense is limited. Here we report that topical exposure to a common environmental DNA-damaging xenobiotic initiated stress surveillance by γδTCR+ intraepithelial lymphocytes that resulted in class switching to IgE in B cells and the accumulation of autoreactive IgE. High-throughput antibody sequencing revealed that γδ T cells shaped the IgE repertoire by supporting specific variable-diversity-joining (VDJ) rearrangements with unique characteristics of the complementarity-determining region CDRH3. This endogenous IgE response, via the IgE receptor FcεRI, provided protection against epithelial carcinogenesis, and expression of the gene encoding FcεRI in human squamous-cell carcinoma correlated with good disease prognosis. These data indicate a joint role for immunosurveillance by T cells and by B cells in epithelial tissues and suggest that IgE is part of the host defense against epithelial damage and tumor development.


Subject(s)
B-Lymphocytes/physiology , Carcinoma, Squamous Cell/immunology , Epithelial Cells/physiology , Immunoglobulin E/metabolism , Intraepithelial Lymphocytes/physiology , Neoplasms, Experimental/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, IgE/metabolism , Animals , Anthracenes/toxicity , Carcinoma, Squamous Cell/diagnosis , Cell Death , Cells, Cultured , Complementarity Determining Regions/genetics , DNA Damage , Female , High-Throughput Nucleotide Sequencing , Immunoglobulin Class Switching , Immunoglobulin E/genetics , Immunologic Surveillance , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/chemically induced , Piperidines/toxicity , Prognosis , Receptors, Antigen, T-Cell, gamma-delta/genetics
2.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Article in English | MEDLINE | ID: mdl-33762306

ABSTRACT

High levels of the intermediate filament protein keratin 17 (K17) are associated with poor prognoses for several human carcinomas. Studies in mouse models have shown that K17 expression is positively associated with growth, survival, and inflammation in skin and that lack of K17 delays onset of tumorigenesis. K17 occurs in the nucleus of human and mouse tumor keratinocytes where it impacts chromatin architecture, gene expression, and cell proliferation. We report here that K17 is induced following DNA damage and promotes keratinocyte survival. The presence of nuclear K17 is required at an early stage of the double-stranded break (DSB) arm of the DNA damage and repair (DDR) cascade, consistent with its ability to associate with key DDR effectors, including γ-H2A.X, 53BP1, and DNA-PKcs. Mice lacking K17 or with attenuated K17 nuclear import showed curtailed initiation in a two-step skin carcinogenesis paradigm. The impact of nuclear-localized K17 on DDR and cell survival provides a basis for the link between K17 induction and poor clinical outcomes for several human carcinomas.


Subject(s)
Carcinoma/genetics , DNA Repair , Keratin-17/metabolism , Keratins/metabolism , Neoplasms, Experimental/genetics , 9,10-Dimethyl-1,2-benzanthracene/administration & dosage , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Active Transport, Cell Nucleus , Animals , Carcinogenesis/chemically induced , Carcinogenesis/genetics , Carcinogenesis/pathology , Carcinoma/chemically induced , Carcinoma/pathology , Cell Nucleus/metabolism , Cell Survival/genetics , DNA Breaks, Double-Stranded/drug effects , Female , Gene Knockout Techniques , HeLa Cells , Humans , Intravital Microscopy , Keratin-17/genetics , Keratinocytes , Keratins/genetics , Male , Mice, Knockout , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/pathology , Time-Lapse Imaging
3.
Proc Natl Acad Sci U S A ; 117(50): 32029-32037, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33257580

ABSTRACT

Disease tolerance, the capacity of tissues to withstand damage caused by a stimulus without a decline in host fitness, varies across tissues, environmental conditions, and physiologic states. While disease tolerance is a known strategy of host defense, its role in noninfectious diseases has been understudied. Here, we provide evidence that a thermogenic fat-epithelial cell axis regulates intestinal disease tolerance during experimental colitis. We find that intestinal disease tolerance is a metabolically expensive trait, whose expression is restricted to thermoneutral mice and is not transferable by the microbiota. Instead, disease tolerance is dependent on the adrenergic state of thermogenic adipocytes, which indirectly regulate tolerogenic responses in intestinal epithelial cells. Our work has identified an unexpected mechanism that controls intestinal disease tolerance with implications for colitogenic diseases.


Subject(s)
Adipose Tissue, Brown/metabolism , Colitis/immunology , Colonic Neoplasms/immunology , Disease Resistance , Enterobacteriaceae Infections/immunology , Adipocytes/metabolism , Adipose Tissue, Brown/cytology , Animals , Azoxymethane/administration & dosage , Cell Communication , Citrobacter rodentium/pathogenicity , Colitis/chemically induced , Colitis/microbiology , Colitis/pathology , Colonic Neoplasms/chemically induced , Colonic Neoplasms/pathology , Dextran Sulfate/toxicity , Enterobacteriaceae Infections/chemically induced , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae Infections/pathology , Epithelial Cells/metabolism , Female , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Male , Mice , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Thermogenesis/immunology
4.
Proc Natl Acad Sci U S A ; 117(11): 6075-6085, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32123074

ABSTRACT

MicroRNA-31 (miR-31) is overexpressed in esophageal squamous cell carcinoma (ESCC), a deadly disease associated with dietary Zn deficiency and inflammation. In a Zn deficiency-promoted rat ESCC model with miR-31 up-regulation, cancer-associated inflammation, and a high ESCC burden following N-nitrosomethylbenzylamine (NMBA) exposure, systemic antimiR-31 delivery reduced ESCC incidence from 85 to 45% (P = 0.038) and miR-31 gene knockout abrogated development of ESCC (P = 1 × 10-6). Transcriptomics, genome sequencing, and metabolomics analyses in these Zn-deficient rats revealed the molecular basis of ESCC abrogation by miR-31 knockout. Our identification of EGLN3, a known negative regulator of nuclear factor κB (NF-κB), as a direct target of miR-31 establishes a functional link between oncomiR-31, tumor suppressor target EGLN3, and up-regulated NF-κB-controlled inflammation signaling. Interaction among oncogenic miR-31, EGLN3 down-regulation, and inflammation was also documented in human ESCCs. miR-31 deletion resulted in suppression of miR-31-associated EGLN3/NF-κB-controlled inflammatory pathways. ESCC-free, Zn-deficient miR-31-/- rat esophagus displayed no genome instability and limited metabolic activity changes vs. the pronounced mutational burden and ESCC-associated metabolic changes of Zn-deficient wild-type rats. These results provide conclusive evidence that miR-31 expression is necessary for ESCC development.


Subject(s)
Esophageal Neoplasms/genetics , Esophageal Squamous Cell Carcinoma/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , MicroRNAs/metabolism , Neoplasms, Experimental/genetics , Animals , Carcinogens/toxicity , Cell Line, Tumor , Dietary Supplements , Esophageal Neoplasms/chemically induced , Esophageal Neoplasms/pathology , Esophageal Neoplasms/prevention & control , Esophageal Squamous Cell Carcinoma/chemically induced , Esophageal Squamous Cell Carcinoma/pathology , Esophageal Squamous Cell Carcinoma/prevention & control , Esophagus/pathology , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Humans , Male , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , NF-kappa B/metabolism , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/pathology , Neoplasms, Experimental/prevention & control , Nitrosamines/toxicity , Rats , Rats, Transgenic , Signal Transduction/genetics , Zinc/administration & dosage , Zinc/deficiency
5.
Proc Natl Acad Sci U S A ; 117(50): 31993-32004, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33262282

ABSTRACT

Effective cancer prevention requires the discovery and intervention of a factor critical to cancer development. Here we show that ovarian progesterone is a crucial endogenous factor inducing the development of primary tumors progressing to metastatic ovarian cancer in a mouse model of high-grade serous carcinoma (HGSC), the most common and deadliest ovarian cancer type. Blocking progesterone signaling by the pharmacologic inhibitor mifepristone or by genetic deletion of the progesterone receptor (PR) effectively suppressed HGSC development and its peritoneal metastases. Strikingly, mifepristone treatment profoundly improved mouse survival (∼18 human years). Hence, targeting progesterone/PR signaling could offer an effective chemopreventive strategy, particularly in high-risk populations of women carrying a deleterious mutation in the BRCA gene.


Subject(s)
BRCA1 Protein/genetics , Cystadenocarcinoma, Serous/prevention & control , Mifepristone/pharmacology , Ovarian Neoplasms/prevention & control , Progesterone/antagonists & inhibitors , Adult , Animals , Breast/pathology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/prevention & control , Cystadenocarcinoma, Serous/chemistry , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , Disease Models, Animal , Estradiol/administration & dosage , Female , Humans , Mice , Middle Aged , Mifepristone/therapeutic use , Mutation , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Neoplasms, Experimental/prevention & control , Ovarian Neoplasms/chemically induced , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovary/pathology , Ovary/surgery , Progesterone/administration & dosage , Progesterone/metabolism , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Salpingo-oophorectomy , Signal Transduction/drug effects , Signal Transduction/genetics
6.
Gastroenterology ; 160(4): 1179-1193.e14, 2021 03.
Article in English | MEDLINE | ID: mdl-32920015

ABSTRACT

BACKGROUND & AIMS: Streptococcus thermophilus was identified to be depleted in patients with colorectal cancer (CRC) by shotgun metagenomic sequencing of 526 multicohort fecal samples. Here, we aim to investigate whether this bacterium could act as a prophylactic for CRC prevention. METHODS: The antitumor effects of S thermophilus were assessed in cultured colonic epithelial cells and in 2 murine models of intestinal tumorigenesis. The tumor-suppressive protein produced by S thermophilus was identified by mass spectrometry and followed by ß-galactosidase activity assay. The mutant strain of S thermophilus was constructed by homologous recombination. The effect of S thermophilus on the gut microbiota composition was assessed by shotgun metagenomic sequencing. RESULTS: Oral gavage of S thermophilus significantly reduced tumor formation in both Apcmin/+ and azoxymethane-injected mice. Coincubation with S thermophilus or its conditioned medium decreased the proliferation of cultured CRC cells. ß-Galactosidase was identified as the critical protein produced by S thermophilus by mass spectrometry screening and ß-galactosidase activity assay. ß-Galactosidase secreted by S thermophilus inhibited cell proliferation, lowered colony formation, induced cell cycle arrest, and promoted apoptosis of cultured CRC cells and retarded the growth of CRC xenograft. The mutant S thermophilus without functional ß-galactosidase lost its tumor-suppressive effect. Also, S thermophilus increased the gut abundance of known probiotics, including Bifidobacterium and Lactobacillus via ß-galactosidase. ß-Galactosidase-dependent production of galactose interfered with energy homeostasis to activate oxidative phosphorylation and downregulate the Hippo pathway kinases, which partially mediated the anticancer effects of S thermophilus. CONCLUSION: S thermophilus is a novel prophylactic for CRC prevention in mice. The tumor-suppressive effect of S thermophilus is mediated at least by the secretion of ß-galactosidase.


Subject(s)
Bacterial Proteins/metabolism , Colorectal Neoplasms/prevention & control , Probiotics/administration & dosage , Streptococcus thermophilus/enzymology , beta-Galactosidase/metabolism , Adenomatous Polyposis Coli Protein/genetics , Animals , Azoxymethane/administration & dosage , Azoxymethane/toxicity , Bacterial Proteins/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/chemically induced , Colon/microbiology , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/genetics , Colorectal Neoplasms/microbiology , Humans , Intestinal Mucosa/microbiology , Male , Mice , Mice, Transgenic , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/genetics , Neoplasms, Experimental/microbiology , Neoplasms, Experimental/prevention & control , Probiotics/metabolism , Streptococcus thermophilus/genetics , beta-Galactosidase/genetics
7.
Gastroenterology ; 160(4): 1284-1300.e16, 2021 03.
Article in English | MEDLINE | ID: mdl-33217448

ABSTRACT

BACKGROUND & AIMS: RNA N6-methyladenosine (m6A) modification has recently emerged as a new regulatory mechanism in cancer progression. We aimed to explore the role of the m6A regulatory enzyme METTL3 in colorectal cancer (CRC) pathogenesis and its potential as a therapeutic target. METHODS: The expression and clinical implication of METTL3 were investigated in multiple human CRC cohorts. The underlying mechanisms of METTL3 in CRC were investigated by integrative m6A sequencing, RNA sequencing, and ribosome profiling analyses. The efficacy of targeting METTL3 in CRC treatment was elucidated in CRC cell lines, patient-derived CRC organoids, and Mettl3-knockout mouse models. RESULTS: Using targeted clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 dropout screening, we identified METTL3 as the top essential m6A regulatory enzyme in CRC. METTL3 was overexpressed in 62.2% (79/127) and 88.0% (44/50) of primary CRCs from 2 independent cohorts. High METTL3 expression predicted poor survival in patients with CRC (n = 374, P < .01). Functionally, silencing METTL3 suppressed tumorigenesis in CRC cells, human-derived primary CRC organoids, and Mettl3-knockout mouse models. We discovered the novel functional m6A methyltransferase domain of METTL3 in CRC cells by domain-focused CRISPR screening and mutagenesis assays. Mechanistically, METTL3 directly induced the m6A-GLUT1-mTORC1 axis as identified by integrated m6A sequencing, RNA sequencing, ribosome sequencing, and functional validation. METTL3 induced GLUT1 translation in an m6A-dependent manner, which subsequently promoted glucose uptake and lactate production, leading to the activation of mTORC1 signaling and CRC development. Furthermore, inhibition of mTORC1 potentiated the anticancer effect of METTL3 silencing in CRC patient-derived organoids and METTL3 transgenic mouse models. CONCLUSIONS: METTL3 promotes CRC by activating the m6A-GLUT1-mTORC1 axis. METTL3 is a promising therapeutic target for the treatment of CRC.


Subject(s)
Colorectal Neoplasms/genetics , Glucose Transporter Type 1/genetics , Methyltransferases/metabolism , Neoplasms, Experimental/genetics , Adenosine/analogs & derivatives , Adenosine/metabolism , Aged , Animals , Azoxymethane/administration & dosage , Azoxymethane/toxicity , Carcinogenesis , Cell Line, Tumor , Cohort Studies , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , DNA Methylation , Dextran Sulfate/administration & dosage , Dextran Sulfate/toxicity , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Methyltransferases/genetics , Mice, Knockout , Middle Aged , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/pathology , Signal Transduction/genetics , Up-Regulation
8.
Gastroenterology ; 160(3): 781-796, 2021 02.
Article in English | MEDLINE | ID: mdl-33129844

ABSTRACT

BACKGROUND & AIMS: Immune checkpoint inhibitors have limited efficacy in many tumors. We investigated mechanisms of tumor resistance to inhibitors of programmed cell death-1 (PDCD1, also called PD-1) in mice with gastric cancer, and the role of its ligand, PD-L1. METHODS: Gastrin-deficient mice were given N-methyl-N-nitrosourea (MNU) in drinking water along with Helicobacter felis to induce gastric tumor formation; we also performed studies with H/K-ATPase-hIL1B mice, which develop spontaneous gastric tumors at the antral-corpus junction and have parietal cells that constitutively secrete interleukin 1B. Mice were given injections of an antibody against PD-1 or an isotype control before tumors developed, or anti-PD-1 and 5-fluorouracil and oxaliplatin, or an antibody against lymphocyte antigen 6 complex locus G (also called Gr-1), which depletes myeloid-derived suppressor cells [MDSCs]), after tumors developed. We generated knock-in mice that express PD-L1 specifically in the gastric epithelium or myeloid lineage. RESULTS: When given to gastrin-deficient mice before tumors grew, anti-PD-1 significantly reduced tumor size and increased tumor infiltration by T cells. However, anti-PD-1 alone did not have significant effects on established tumors in these mice. Neither early nor late anti-PD-1 administration reduced tumor growth in the presence of MDSCs in H/K-ATPase-hIL-1ß mice. The combination of 5-fluorouracil and oxaliplatin reduced MDSCs, increased numbers of intra-tumor CD8+ T cells, and increased the response of tumors to anti-PD-1; however, this resulted in increased tumor expression of PD-L1. Expression of PD-L1 by tumor or immune cells increased gastric tumorigenesis in mice given MNU. Mice with gastric epithelial cells that expressed PD-L1 did not develop spontaneous tumors, but they developed more and larger tumors after administration of MNU and H felis, with accumulation of MDSCs. CONCLUSIONS: In mouse models of gastric cancer, 5-fluorouracil and oxaliplatin reduce numbers of MDSCs to increase the effects of anti-PD-1, which promotes tumor infiltration by CD8+ T cells. However, these chemotherapeutic agents also induce expression of PD-L1 by tumor cells. Expression of PD-L1 by gastric epithelial cells increases tumorigenesis in response to MNU and H felis, and accumulation of MDSCs, which promote tumor progression. The timing and site of PD-L1 expression is therefore important in gastric tumorigenesis and should be considered in design of therapeutic regimens.


Subject(s)
Helicobacter Infections/immunology , Myeloid-Derived Suppressor Cells/immunology , Neoplasms, Experimental/immunology , Programmed Cell Death 1 Receptor/metabolism , Stomach Neoplasms/immunology , Administration, Oral , Animals , Carcinogenesis/chemically induced , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinogenesis/immunology , Gastric Mucosa/immunology , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Gastrins/genetics , Helicobacter Infections/chemically induced , Helicobacter Infections/genetics , Helicobacter Infections/microbiology , Helicobacter felis/immunology , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Methylnitrosourea/administration & dosage , Mice , Mice, Knockout , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/microbiology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/immunology , Stomach Neoplasms/chemically induced , Stomach Neoplasms/drug therapy , Stomach Neoplasms/microbiology , Tumor Microenvironment/immunology
9.
Hepatology ; 74(2): 797-815, 2021 08.
Article in English | MEDLINE | ID: mdl-33650193

ABSTRACT

BACKGROUND AND AIMS: Intrahepatic cholangiocarcinoma (iCCA) is closely correlated with hepatic progenitor cell (HPC) expansion and liver fibrosis. Brahma-related gene 1 (Brg1), an enzymatic subunit of the switch/sucrose nonfermentable complex that is critical in stem cell maintenance and tumor promotion, is prominently up-regulated in both HPCs and iCCA; however, its role in this correlation remains undefined. APPROACH AND RESULTS: A retrospective cohort study indicated that high Brg1 expression suggests poor prognosis in patients with iCCA. In chronically injured livers induced by a 0.1% 3,5-diethoxycarbonyl-1,4-dihydrocollidine diet or bile duct ligation surgery, HPCs were dramatically activated, as indicated by their enhanced expression of Brg1 and a subset of stem cell markers; however, Brg1 ablation in HPCs strongly suppressed HPC expansion and liver fibrosis. Furthermore, in a chemically induced iCCA model, inhibition of Brg1 by a specific inhibitor or inducible gene ablation markedly improved histology and suppressed iCCA growth. Mechanistically, in addition to transcriptionally promoting both Wnt receptor genes and target genes, Brg1 was found to bind to the ß-catenin/transcription factor 4 transcription complex, suggesting a possible approach for regulation of Wnt/ß-catenin signaling. CONCLUSIONS: We have demonstrated the function of Brg1 in promoting HPC expansion, liver cirrhosis, and, ultimately, iCCA development in chronically injured livers, which is largely dependent on Wnt/ß-catenin signaling. Our data suggest that therapies targeting Brg1-expressing HPCs are promising for the treatment of liver cirrhosis and iCCA.


Subject(s)
Bile Duct Neoplasms/genetics , Cholangiocarcinoma/genetics , DNA Helicases/genetics , Liver Cirrhosis/genetics , Neoplasm Recurrence, Local/epidemiology , Nuclear Proteins/genetics , Transcription Factors/genetics , Adult , Aged , Animals , Azabicyclo Compounds/pharmacology , Azabicyclo Compounds/therapeutic use , Bile Duct Neoplasms/chemically induced , Bile Duct Neoplasms/mortality , Bile Duct Neoplasms/therapy , Bile Ducts, Intrahepatic/pathology , Bile Ducts, Intrahepatic/surgery , Cholangiocarcinoma/chemically induced , Cholangiocarcinoma/mortality , Cholangiocarcinoma/therapy , DNA Helicases/antagonists & inhibitors , DNA Helicases/metabolism , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Liver/pathology , Liver/surgery , Liver Cirrhosis/diagnosis , Liver Cirrhosis/pathology , Liver Cirrhosis/prevention & control , Male , Mice , Mice, Transgenic , Middle Aged , Neoplasm Recurrence, Local/genetics , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/genetics , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Prognosis , Pyridines/pharmacology , Pyridines/therapeutic use , Retrospective Studies , Stem Cells/metabolism , Stem Cells/pathology , Thioacetamide/administration & dosage , Thioacetamide/toxicity , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Up-Regulation , Wnt Signaling Pathway/genetics
10.
Carcinogenesis ; 42(2): 232-242, 2021 02 25.
Article in English | MEDLINE | ID: mdl-32816038

ABSTRACT

More than a million cases of cutaneous squamous cell carcinoma are diagnosed in the USA each year, and its incidence is increasing. Most of these malignancies arise from premalignant lesions, providing an opportunity for intervention before malignant progression. We previously documented how cytoplasmic mislocalization of CDC25A in premalignant and malignant skin cancers confers resistance to apoptotic cell death via a mechanism that depends on its interaction with 14-3-3ε. From these data, we hypothesized that 14-3-3ε overexpression drives skin tumor development and progression, such that targeting 14-3-3ε may be a useful strategy for skin cancer treatment. Like CDC25A, 14-3-3ε was overexpressed and mislocalized to the cytoplasm of both benign and malignant human skin cancer. Skin-targeted deletion of the 14-3-3ε gene reduced skin tumor development by 75% and blocked malignant progression. 14-3-3ε suppressed apoptosis through activation of Akt, leading to inhibition of BCL2 associated agonist of cell death and upregulation of Survivin. Using virtual tetrapeptide libraries, we developed a novel peptide that specifically blocked 14-3-3ε heterodimerization and thereby prevented its interaction with CDC25A. The peptide reduced prosurvival signaling, killed skin cancer cells and reduced skin tumor growth in xenograft. Normal skin keratinocytes were unaffected by inhibition or deletion of 14-3-3ε. Thus, targeting of 14-3-3ε dimerization is a promising strategy for the treatment of premalignant skin lesions.


Subject(s)
14-3-3 Proteins/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , Skin Neoplasms/drug therapy , cdc25 Phosphatases/metabolism , 14-3-3 Proteins/genetics , 14-3-3 Proteins/metabolism , 9,10-Dimethyl-1,2-benzanthracene/administration & dosage , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Carcinogens/administration & dosage , Carcinogens/toxicity , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cytoplasm/drug effects , Cytoplasm/metabolism , Female , Humans , Keratinocytes , Male , Mice , Mice, Knockout , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Protein Multimerization/drug effects , Skin Neoplasms/pathology , Tetradecanoylphorbol Acetate/administration & dosage , Tetradecanoylphorbol Acetate/analogs & derivatives , Tetradecanoylphorbol Acetate/toxicity , Xenograft Model Antitumor Assays
11.
J Am Chem Soc ; 143(37): 15233-15242, 2021 09 22.
Article in English | MEDLINE | ID: mdl-34514797

ABSTRACT

Taking advantage of cancer cells' endogenous characters, the responsive activation of DNA nanomachines has achieved great success in tumor therapy. Combining with extra stimuli such as external light irradiation provided spatiotemporal control of DNA nanomachine activation. However, specific activation at the cellular level is still challenging considering the macroscopic-scale exposure area of usual light sources. DNA logic gates located at the cell membrane contributed to cellular specificity, but the free diffusion of input DNA strands during the operation process would impair efficiency and result in side effects to circumjacent normal cells in solid tumors. Here we design a transmembrane DNA logical computation strategy to activate a DNA nanomachine only in cancer cells from a complex solid tumor microenvironment. The DNA nanomachine multishell UCNPs-DNA is prepared by modifying DNA strands on upconversion nanoparticles. LA-apt, a DNA strand anchoring to a cancer cell membrane overexpressed receptor, and intracellular miRNA-21 served as inputs 1 and 2, respectively. Hybridization with input 1 at the cell membrane not only exposes the miRNA-21 recognition region at the DNA nanomachine, but also delivers it into cancer cells. The cascade hybridization with intracellular input 2 completes the "AND" gate operation and releases a DNA strand L2 as output. L2 acts as the trigger to operate the DNA nanomachine and correspondingly activates the photosensitizer Rose Bengal for reactive oxygen species generation. Through the "AND" gate operation of the DNA nanomachine across the cancer cell membrane, highly precise therapy only to cancer cells is achieved in a complex solid tumor microenvironment, which could become a promising modality for precise therapy of solid tumors.


Subject(s)
Breast Neoplasms/therapy , Chemical Engineering/methods , DNA/chemistry , Nanostructures/therapeutic use , Neoplasms/therapy , Animals , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Nanostructures/chemistry , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/diagnostic imaging
12.
Int J Cancer ; 148(1): 226-237, 2021 01 01.
Article in English | MEDLINE | ID: mdl-32700769

ABSTRACT

Hepatocellular carcinoma (HCC) is highly resistant to anticancer therapy and novel therapeutic strategies are needed. Chronotherapy may become a promising approach because it may improve the efficacy of antimitotic radiation and chemotherapy by considering timing of treatment. To date little is known about time-of-day dependent changes of proliferation and DNA damage in HCC. Using transgenic c-myc/transforming growth factor (TGFα) mice as HCC animal model, we immunohistochemically demonstrated Ki67 as marker for proliferation and γ-H2AX as marker for DNA damage in HCC and surrounding healthy liver (HL). Core clock genes (Per1, Per2, Cry1, Cry2, Bmal 1, Rev-erbα and Clock) were examined by qPCR. Data were obtained from samples collected ex vivo at four different time points and from organotypic slice cultures (OSC). Significant differences were found between HCC and HL. In HCC, the number of Ki67 immunoreactive cells showed two peaks (ex vivo: ZT06 middle of day and ZT18 middle of night; OSC: CT04 and CT16). In ex vivo samples, the number of γ-H2AX positive cells in HCC peaked at ZT18 (middle of the night), while in OSC their number remained high during subjective day and night. In both HCC and HL, clock gene expression showed a time-of-day dependent expression ex vivo but no changes in OSC. The expression of Per2 and Cry1 was significantly lower in HCC than in HL. Our data support the concept of chronotherapy of HCC. OSC may become useful to test novel cancer therapies.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Neoplasms, Experimental/genetics , Period Circadian Proteins/genetics , Animals , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Cell Proliferation/genetics , Chlorides/administration & dosage , Chlorides/toxicity , Chronotherapy , DNA Damage , Gene Expression Regulation, Neoplastic , Humans , Liver/pathology , Liver Neoplasms/chemically induced , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Mice , Mice, Transgenic , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/therapy , Photoperiod , Proto-Oncogene Proteins c-myc/genetics , Transforming Growth Factor alpha/genetics , Tumor Cells, Cultured , Zinc Compounds/administration & dosage , Zinc Compounds/toxicity
13.
Gastroenterology ; 159(6): 2146-2162.e33, 2020 12.
Article in English | MEDLINE | ID: mdl-32805281

ABSTRACT

BACKGROUND & AIMS: Chromosomal instability (CIN) is a carcinogenesis event that promotes metastasis and resistance to therapy by unclear mechanisms. Expression of the colon cancer-associated transcript 2 gene (CCAT2), which encodes a long noncoding RNA (lncRNA), associates with CIN, but little is known about how CCAT2 lncRNA regulates this cancer enabling characteristic. METHODS: We performed cytogenetic analysis of colorectal cancer (CRC) cell lines (HCT116, KM12C/SM, and HT29) overexpressing CCAT2 and colon organoids from C57BL/6N mice with the CCAT2 transgene and without (controls). CRC cells were also analyzed by immunofluorescence microscopy, γ-H2AX, and senescence assays. CCAT2 transgene and control mice were given azoxymethane and dextran sulfate sodium to induce colon tumors. We performed gene expression array and mass spectrometry to detect downstream targets of CCAT2 lncRNA. We characterized interactions between CCAT2 with downstream proteins using MS2 pull-down, RNA immunoprecipitation, and selective 2'-hydroxyl acylation analyzed by primer extension analyses. Downstream proteins were overexpressed in CRC cells and analyzed for CIN. Gene expression levels were measured in CRC and non-tumor tissues from 5 cohorts, comprising more than 900 patients. RESULTS: High expression of CCAT2 induced CIN in CRC cell lines and increased resistance to 5-fluorouracil and oxaliplatin. Mice that expressed the CCAT2 transgene developed chromosome abnormalities, and colon organoids derived from crypt cells of these mice had a higher percentage of chromosome abnormalities compared with organoids from control mice. The transgenic mice given azoxymethane and dextran sulfate sodium developed more and larger colon polyps than control mice given these agents. Microarray analysis and mass spectrometry indicated that expression of CCAT2 increased expression of genes involved in ribosome biogenesis and protein synthesis. CCAT2 lncRNA interacted directly with and stabilized BOP1 ribosomal biogenesis factor (BOP1). CCAT2 also increased expression of MYC, which activated expression of BOP1. Overexpression of BOP1 in CRC cell lines resulted in chromosomal missegregation errors, and increased colony formation, and invasiveness, whereas BOP1 knockdown reduced viability. BOP1 promoted CIN by increasing the active form of aurora kinase B, which regulates chromosomal segregation. BOP1 was overexpressed in polyp tissues from CCAT2 transgenic mice compared with healthy tissue. CCAT2 lncRNA and BOP1 mRNA or protein were all increased in microsatellite stable tumors (characterized by CIN), but not in tumors with microsatellite instability compared with nontumor tissues. Increased levels of CCAT2 lncRNA and BOP1 mRNA correlated with each other and with shorter survival times of patients. CONCLUSIONS: We found that overexpression of CCAT2 in colon cells promotes CIN and carcinogenesis by stabilizing and inducing expression of BOP1 an activator of aurora kinase B. Strategies to target this pathway might be developed for treatment of patients with microsatellite stable colorectal tumors.


Subject(s)
Chromosomal Instability , Colorectal Neoplasms/genetics , Neoplasms, Experimental/genetics , RNA, Long Noncoding/metabolism , RNA-Binding Proteins/genetics , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Aurora Kinase B/metabolism , Azoxymethane/toxicity , Carcinogenesis/genetics , Cell Line, Tumor , Colon/cytology , Colon/pathology , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/pathology , Cytogenetic Analysis , Dextrans/toxicity , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/pathology , Male , Mice , Mice, Transgenic , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/pathology , Organoids , Primary Cell Culture , Proto-Oncogene Proteins c-myc/metabolism , RNA, Long Noncoding/genetics , RNA-Binding Proteins/metabolism , Signal Transduction/genetics
14.
Mol Cell Biochem ; 476(4): 1765-1781, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33433833

ABSTRACT

NF-κB is the principle transcription factor and plays the central role in orchestrating chronic inflammation by regulating levels of cytokines, chemokines and growth factors. Piperlongumine (PL), a major alkaloid in the fruit of Piper longum Linn. has gained worldwide attention for its anticancer properties, however, its mechanism of action in the chemoprevention of colon cancer has not been investigated yet. Therefore, the present study was designed to elucidate the underlying molecular mechanism of PL in preventing DMH/DSS induced experimental colon cancer in mice. In the current study well established DMH/DSS induced experimental colon cancer mouse model was used to demonstrate the chemopreventive potential of PL. The expression of NF-κB and its downstream target proteins was evaluated mainly through western blotting. In addition, CAM assay, immunohistochemical staining and gelatin zymography was used to show anti-angiogenic and anti-invasive potential of PL. Additionally, important tumor biomarkers such as TSA, LASA, LDH and IL-6 levels were also estimated. The results of current study showed that PL was capable to inhibit NF-κB activation as well as its nuclear translocation. PL administration to DMH/DSS treated mice also inhibited the NF-κB downstream signaling cascades such as including COX-2 pathway, JAK/STAT pathway, ß-catenin, Notch signaling pathway, angiogenesis and epithelial to mesenchymal transition pathway. The findings of the present study have claimed PL as promising chemopreventive agent for colon cancer with pleiotropic action. The current study emphasizes that regular consumption of PL can be an effective approach in the prevention of colon cancer in humans.


Subject(s)
Colonic Neoplasms/metabolism , Dioxolanes/pharmacology , NF-kappa B/metabolism , Neoplasm Proteins/metabolism , Neoplasms, Experimental/metabolism , Signal Transduction/drug effects , Animals , Colonic Neoplasms/chemically induced , Colonic Neoplasms/pathology , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/pathology
15.
J Biochem Mol Toxicol ; 35(5): e22723, 2021 May.
Article in English | MEDLINE | ID: mdl-33511709

ABSTRACT

Gastric carcinoma is one of the most aggressive types of cancer that ranks fifth among all cancer incidences and third in cancer mortality. As it exhibits a prolonged asymptomatic condition and high recurrence rate, it is a great challenge to treat gastric cancer. Traditional medicine that utilizes herbal phytochemicals to treat various diseases is a potent alternative for current allopathic treatment. Hence, we evaluated the potency of a phytochemical bilobalide for treating gastric cancer in in vitro and in vivo models. Bilobalide, a sesquiterpenoid, is present in the Ginkgo biloba plant that belongs to the family of Ginkgoaceae. The cytotoxicity effect of bilobalide was evaluated in both gastric cancer (AGS) cells and normal gastric epithelial cells. Apoptosis-inducing property of bilobalide against the AGS cell line was analyzed with different fluorescent staining techniques and terminal deoxynucleotidyl transferase dUTP nick-end labeling assay, and cell cycle analysis was carried out by flow cytometry. The in vivo studies were assessed with N-methyl-N-nitrosourea (MNU)-induced gastric cancer in rats. Serum-specific gastric markers were quantified and histopathological analysis of stomach tissue was performed. The expression of target-signaling molecules was analyzed by a reverse-transcription polymerase chain reaction. The in vitro results proved that bilobalide effectively suppressed the AGS cell growth and induced cell death by nuclear damage and apoptosis induction. The bilobalide treatment effectively arrested the cell cycle of AGS cells via inhibiting the PI3K-signaling pathway. Our in vivo results also confirmed that the bilobalide persuasively inhibited the MNU-induced gastric carcinoma via inhibiting the thioredoxin-fold family proteins and inflammatory markers' expression. Overall, our results authentically prove that bilobalide possesses therapeutic potency to cure gastric carcinoma.


Subject(s)
Apoptosis/drug effects , Bilobalides/pharmacology , Cell Proliferation/drug effects , Neoplasms, Experimental/drug therapy , Plant Extracts/chemistry , Stomach Neoplasms/drug therapy , Animals , Bilobalides/chemistry , Cell Line, Tumor , Ginkgo biloba , Humans , Male , Methylnitrosourea/toxicity , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Rats , Rats, Wistar , Stomach Neoplasms/chemically induced , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
16.
J Biochem Mol Toxicol ; 35(9): e22838, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34273909

ABSTRACT

Colorectal cancer (CRC) is the third most common type of cancer. Here, we studied the inhibitory effect of IRAK1 and IRAK4 as a preventive strategy using a colitis-induced tumorigenesis mouse model. CRC clinical data were obtained from the Gene Expression Omnibus (GEO). An experimental inflammation-dependent CRC model was induced by treatment with azoxymethane (AOM) and then dextran sodium sulfate (DSS) in C57BL/6 mice. Mice were administered an IRAK1/4 inhibitor by intraperitoneal injection at 3 mg/kg twice each week for 9 weeks. The IRAK1/4 inhibitor attenuated histological changes and prevented tumor growth. Tumor-associated proteins, including p65 and Ki-67, were downregulated by the IRAK1/4 inhibitor in AOM/DSS-treated mice. Additionally, IRAK1/4 inhibitor administration effectively decreased the expression of inflammatory cytokines. Furthermore, we observed that IRAK1/4 inhibitor treatment attenuated colitis-induced tumorigenesis by inhibiting epithelial-mesenchymal transition. These observations indicate that inhibition of IRAK1 and IRAK4 may suppress experimental colitis-induced tumorigenesis by inhibiting inflammatory responses and epithelial-mesenchymal transition.


Subject(s)
Carcinogenesis/drug effects , Colitis-Associated Neoplasms/drug therapy , Colitis/drug therapy , Epithelial-Mesenchymal Transition/drug effects , Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Neoplasms, Experimental/drug therapy , Protein Kinase Inhibitors/pharmacology , Animals , Carcinogenesis/chemically induced , Carcinogenesis/metabolism , Colitis/chemically induced , Colitis/enzymology , Colitis-Associated Neoplasms/chemically induced , Colitis-Associated Neoplasms/enzymology , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/enzymology , Interleukin-1 Receptor-Associated Kinases/metabolism , Male , Mice , Neoplasm Proteins/metabolism , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/enzymology
17.
Regul Toxicol Pharmacol ; 124: 104977, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34174380

ABSTRACT

The National Toxicology Program (NTP) reported that chronic dietary exposure to 4-methylimidazole (4-MeI) increased the incidence of lung adenomas/carcinomas beyond the normally high spontaneous rate in B6C3F1 mice. To examine plausible modes of action (MoAs) for mouse lung tumors (MLTs) upon exposure to high levels of 4-MeI, and their relevance in assessing human risk, a systematic approach was used to identify and evaluate mechanistic data (in vitro and in vivo) in the primary and secondary literature, along with high-throughput screening assay data. Study quality, relevance, and activity of mechanistic data identified across the evidence-base were organized according to key characteristics of carcinogens (KCCs) to identify potential key events in known or novel MLT MoAs. Integration of these evidence streams provided confirmation that 4-MeI lacks genotoxic and cytotoxic activity with some evidence to support a lack of mitogenic activity. Further evaluation of contextual and chemical-specific characteristics of 4-MeI was consequently undertaken. Due to lack of genotoxicity, along with transcriptomic and histopathological lung changes up to 28 and 90 days of exposure, the collective evidence suggests MLTs observed following exposure to high levels of 4-MeI develop at a late stage in the mouse chronic bioassay, albeit the exact MoA remains unclear.


Subject(s)
Carcinogens/toxicity , Imidazoles/toxicity , Lung Neoplasms/epidemiology , Neoplasms, Experimental/epidemiology , Toxicity Tests, Chronic/statistics & numerical data , Animals , Carcinogens/administration & dosage , Data Interpretation, Statistical , Disease Progression , Dose-Response Relationship, Drug , Imidazoles/administration & dosage , Incidence , Lung/drug effects , Lung/pathology , Lung Neoplasms/chemically induced , Lung Neoplasms/pathology , Mice , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/pathology , Risk Assessment/methods , Risk Assessment/statistics & numerical data , Toxicity Tests, Chronic/methods
18.
Regul Toxicol Pharmacol ; 124: 104961, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34015422

ABSTRACT

Ethyl acrylate (EA) was classified by IARC as a Group-2B Carcinogen based, in part, on data suggesting increased incidence of thyroid neoplasia in rats and mice exposed chronically to EA vapors. We examined chronic exposure of rats and mice to EA vapors, evaluated the data on the incidence of thyroid follicular neoplasia, and determined the relevance of thyroid tumors to human health risk. The data revealed a small statistically significant increase in thyroid tumors in EA-exposed male rats and mice. The tumor incidences were within the range of historical controls and were not consistently dose-dependent. Most thyroid tumors in exposed animals were benign. Chronic exposure of EA to rats and mice (drinking water or gavage) and dogs (capsules) had no evidence of thyroid neoplasia. Results from chronic studies, in vivo and in vitro data, and ToxCastTM/Tox 21 HTPS did not support genotoxic/mutagenic potential for EA. This suggests that the associations between EA exposure and thyroid neoplasia represent chance or random observations rather than a compound-mediated effect. Due to species-specific physiological differences, the hypothalamic-pituitary-thyroid axis of rodents is more sensitive to endocrine disruptive chemicals than that of humans which further suggests that findings in rodents have questionable relevance to human health.


Subject(s)
Acrylates/toxicity , Carcinogens/toxicity , Neoplasms, Experimental/chemically induced , Stomach Neoplasms/chemically induced , Thyroid Neoplasms/chemically induced , Animals , Dogs , Female , Humans , Male , Mice , Neoplasms, Experimental/blood , Neoplasms, Experimental/pathology , Rats , Species Specificity , Stomach/drug effects , Stomach/pathology , Stomach Neoplasms/pathology , Thyroid Gland/drug effects , Thyroid Gland/metabolism , Thyroid Gland/pathology , Thyroid Hormones/blood , Thyroid Hormones/metabolism , Thyroid Neoplasms/blood , Thyroid Neoplasms/pathology , Toxicity Tests, Chronic/statistics & numerical data
19.
Int J Mol Sci ; 22(19)2021 Sep 29.
Article in English | MEDLINE | ID: mdl-34638879

ABSTRACT

Colorectal cancer (CRC) is one of the most frequently diagnosed cancers in humans. At early stages CRC is treated by surgery and at advanced stages combined with chemotherapy. We examined here the potential effect of glucosylceramide synthase (GCS)-inhibition on CRC biology. GCS is the rate-limiting enzyme in the glycosphingolipid (GSL)-biosynthesis pathway and overexpressed in many human tumors. We suppressed GSL-biosynthesis using the GCS inhibitor Genz-123346 (Genz), NB-DNJ (Miglustat) or by genetic targeting of the GCS-encoding gene UDP-glucose-ceramide-glucosyltransferase- (UGCG). GCS-inhibition or GSL-depletion led to a marked arrest of the cell cycle in Lovo cells. UGCG silencing strongly also inhibited tumor spheroid growth in Lovo cells and moderately in HCT116 cells. MS/MS analysis demonstrated markedly elevated levels of sphingomyelin (SM) and phosphatidylcholine (PC) that occurred in a Genz-concentration dependent manner. Ultrastructural analysis of Genz-treated cells indicated multi-lamellar lipid storage in vesicular compartments. In mice, Genz lowered the incidence of experimentally induced colorectal tumors and in particular the growth of colorectal adenomas. These results highlight the potential for GCS-based inhibition in the treatment of CRC.


Subject(s)
Cell Cycle/drug effects , Colonic Neoplasms , Dioxanes/pharmacology , Glycosphingolipids , Pyrrolidines/pharmacology , Spheroids, Cellular , Animals , Colonic Neoplasms/chemically induced , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Glucosyltransferases/antagonists & inhibitors , Glucosyltransferases/metabolism , Glycosphingolipids/biosynthesis , Glycosphingolipids/genetics , HCT116 Cells , Humans , Mice , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology
20.
Carcinogenesis ; 41(8): 1134-1144, 2020 08 12.
Article in English | MEDLINE | ID: mdl-31740923

ABSTRACT

Increased expression of osteopontin (secreted phosphoprotein 1, SPP1) is associated with aggressive human lung adenocarcinoma (LADC), but its function remains unknown. Our aim was to determine the role of SPP1 in smoking-induced LADC. We combined mouse models of tobacco carcinogen-induced LADC, of deficiency of endogenous Spp1 alleles, and of adoptive pulmonary macrophage reconstitution to map the expression of SPP1 and its receptors and determine its impact during carcinogenesis. Co-expression of Spp1 and mutant KrasG12C in benign cells was employed to investigate SPP1/KRAS interactions in oncogenesis. Finally, intratracheal adenovirus encoding Cre recombinase was delivered to LSL.KRASG12D mice lacking endogenous or overexpressing transgenic Spp1 alleles. SPP1 was overexpressed in experimental and human LADC and portended poor survival. In response to two different smoke carcinogens, Spp1-deficient mice developed fewer and smaller LADC with decreased cellular survival and angiogenesis. Both lung epithelial- and macrophage-secreted SPP1 drove tumor-associated inflammation, while epithelial SPP1 promoted early tumorigenesis by fostering the survival of KRAS-mutated cells. Finally, loss and overexpression of Spp1 was, respectively, protective and deleterious for mice harboring KRASG12D-driven LADC. Our data support that SPP1 is functionally involved in early stages of airway epithelial carcinogenesis driven by smoking and mutant KRAS and may present an important therapeutic target.


Subject(s)
Adenocarcinoma of Lung/pathology , Carcinogenesis/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Osteopontin/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Smoking/adverse effects , Adenocarcinoma of Lung/chemically induced , Adenocarcinoma of Lung/genetics , Animals , HEK293 Cells , Humans , Lung Neoplasms/chemically induced , Mice , Mice, Inbred C57BL , Mutation , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Osteopontin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL