Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 174
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Immunity ; 55(2): 237-253.e8, 2022 02 08.
Article in English | MEDLINE | ID: mdl-35081371

ABSTRACT

The Th17 cell-lineage-defining cytokine IL-17A contributes to host defense and inflammatory disease by coordinating multicellular immune responses. The IL-17 receptor (IL-17RA) is expressed by diverse intestinal cell types, and therapies targeting IL-17A induce adverse intestinal events, suggesting additional tissue-specific functions. Here, we used multiple conditional deletion models to identify a role for IL-17A in secretory epithelial cell differentiation in the gut. Paneth, tuft, goblet, and enteroendocrine cell numbers were dependent on IL-17A-mediated induction of the transcription factor ATOH1 in Lgr5+ intestinal epithelial stem cells. Although dispensable at steady state, IL-17RA signaling in ATOH1+ cells was required to regenerate secretory cells following injury. Finally, IL-17A stimulation of human-derived intestinal organoids that were locked into a cystic immature state induced ATOH1 expression and rescued secretory cell differentiation. Our data suggest that the cross talk between immune cells and stem cells regulates secretory cell lineage commitment and the integrity of the mucosa.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Intestinal Mucosa/cytology , Receptors, G-Protein-Coupled/metabolism , Receptors, Interleukin-17/metabolism , Stem Cells/metabolism , Animals , Cell Communication , Cell Differentiation/drug effects , Cell Lineage/drug effects , Colitis/chemically induced , Colitis/metabolism , Colitis/pathology , Dextran Sulfate/adverse effects , Humans , Interleukin-17/metabolism , Interleukin-17/pharmacology , Intestinal Mucosa/metabolism , Intestines/drug effects , Intestines/metabolism , Intestines/pathology , Mice , Mice, Knockout , NF-kappa B/metabolism , Receptors, Interleukin-17/deficiency , SOX9 Transcription Factor/metabolism , Signal Transduction , Stem Cells/cytology
3.
Oncologist ; 28(11): e1065-e1074, 2023 Nov 02.
Article in English | MEDLINE | ID: mdl-37156009

ABSTRACT

INTRODUCTION: Immune checkpoint inhibitor (ICI) pneumonitis causes substantial morbidity and mortality. Estimates of real-world incidence and reported risk factors vary substantially. METHODS: We conducted a retrospective review of 419 patients with advanced non-small cell lung cancer (NSCLC) who were treated with anti-PD-(L)1 with or without anti-CTLA-4 therapy. Clinical, imaging, and microbiological data were evaluated by multidisciplinary adjudication teams. The primary outcome of interest was grade ≥2 (CTCAEv5) pneumonitis. Clinicopathologic variables, tobacco use, cancer therapies, and preexisting lung disease were assessed for univariate effects using Cox proportional hazards models. We created multivariate Cox proportional hazards models to assess risk factors for pneumonitis and mortality. Pneumonitis, pneumonia, and progression were modeled as time-dependent variables in mortality models. RESULTS: We evaluated 419 patients between 2013 and 2021. The cumulative incidence of pneumonitis was 9.5% (40/419). In a multivariate model, pneumonitis increased the risk for mortality (HR 1.6, 95% CI, 1.0-2.5), after adjustment for disease progression (HR 1.6, 95% CI, 1.4-1.8) and baseline shortness of breath (HR 1.5, 95% CI, 1.2-2.0). Incomplete resolution was more common with more severe pneumonitis. Interstitial lung disease was associated with higher risk for pneumonitis (HR 5.4, 95% CI, 1.1-26.6), particularly in never smokers (HR 26.9, 95% CI, 2.8-259.0). CONCLUSION: Pneumonitis occurred at a high rate and significantly increased mortality. Interstitial lung disease, particularly in never smokers, increased the risk for pneumonitis.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Diseases, Interstitial , Lung Neoplasms , Pneumonia , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Incidence , Lung Neoplasms/drug therapy , Pneumonia/epidemiology , Risk Factors , Lung Diseases, Interstitial/complications , Retrospective Studies
4.
PLoS Pathog ; 17(12): e1010103, 2021 12.
Article in English | MEDLINE | ID: mdl-34871329

ABSTRACT

Yersinia pseudotuberculosis is a foodborne pathogen that subverts immune function by translocation of Yersinia outer protein (Yop) effectors into host cells. As adaptive γδ T cells protect the intestinal mucosa from pathogen invasion, we assessed whether Y. pseudotuberculosis subverts these cells in mice and humans. Tracking Yop translocation revealed that the preferential delivery of Yop effectors directly into murine Vγ4 and human Vδ2+ T cells inhibited anti-microbial IFNγ production. Subversion was mediated by the adhesin YadA, injectisome component YopB, and translocated YopJ effector. A broad anti-pathogen gene signature and STAT4 phosphorylation levels were inhibited by translocated YopJ. Thus, Y. pseudotuberculosis attachment and translocation of YopJ directly into adaptive γδ T cells is a major mechanism of immune subversion in mice and humans. This study uncovered a conserved Y. pseudotuberculosis pathway that subverts adaptive γδ T cell function to promote pathogenicity.


Subject(s)
Bacterial Proteins/immunology , Immune Evasion/immunology , Interferon-gamma/biosynthesis , Intraepithelial Lymphocytes/immunology , Yersinia pseudotuberculosis Infections/immunology , Animals , Humans , Mice , Yersinia pseudotuberculosis/immunology
5.
Circ Res ; 128(3): 335-357, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33539225

ABSTRACT

RATIONALE: Diabetic cardiomyopathy (DbCM) is a major complication in type-1 diabetes, accompanied by altered cardiac energetics, impaired mitochondrial function, and oxidative stress. Previous studies indicate that type-1 diabetes is associated with increased cardiac expression of KLF5 (Krüppel-like factor-5) and PPARα (peroxisome proliferator-activated receptor) that regulate cardiac lipid metabolism. OBJECTIVE: In this study, we investigated the involvement of KLF5 in DbCM and its transcriptional regulation. METHODS AND RESULTS: KLF5 mRNA levels were assessed in isolated cardiomyocytes from cardiovascular patients with diabetes and were higher compared with nondiabetic individuals. Analyses in human cells and diabetic mice with cardiomyocyte-specific FOXO1 (Forkhead box protein O1) deletion showed that FOXO1 bound directly on the KLF5 promoter and increased KLF5 expression. Diabetic mice with cardiomyocyte-specific FOXO1 deletion had lower cardiac KLF5 expression and were protected from DbCM. Genetic, pharmacological gain and loss of KLF5 function approaches and AAV (adeno-associated virus)-mediated Klf5 delivery in mice showed that KLF5 induces DbCM. Accordingly, the protective effect of cardiomyocyte FOXO1 ablation in DbCM was abolished when KLF5 expression was rescued. Similarly, constitutive cardiomyocyte-specific KLF5 overexpression caused cardiac dysfunction. KLF5 caused oxidative stress via direct binding on NADPH oxidase (NOX)4 promoter and induction of NOX4 (NADPH oxidase 4) expression. This was accompanied by accumulation of cardiac ceramides. Pharmacological or genetic KLF5 inhibition alleviated superoxide formation, prevented ceramide accumulation, and improved cardiac function in diabetic mice. CONCLUSIONS: Diabetes-mediated activation of cardiomyocyte FOXO1 increases KLF5 expression, which stimulates NOX4 expression, ceramide accumulation, and causes DbCM.


Subject(s)
Diabetic Cardiomyopathies/metabolism , Forkhead Box Protein O1/metabolism , Kruppel-Like Transcription Factors/metabolism , Myocytes, Cardiac/metabolism , Oxidative Stress , PPAR alpha/metabolism , Aged , Animals , Cell Line , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/pathology , Disease Models, Animal , Female , Forkhead Box Protein O1/genetics , Gene Expression Regulation , Humans , Kruppel-Like Transcription Factors/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Myocytes, Cardiac/pathology , PPAR alpha/genetics , Transcription, Genetic
6.
Circulation ; 143(11): 1139-1156, 2021 03 16.
Article in English | MEDLINE | ID: mdl-33430631

ABSTRACT

BACKGROUND: We previously showed that cardiomyocyte Krϋppel-like factor (KLF) 5 regulates cardiac fatty acid oxidation. As heart failure has been associated with altered fatty acid oxidation, we investigated the role of cardiomyocyte KLF5 in lipid metabolism and pathophysiology of ischemic heart failure. METHODS: Using real-time polymerase chain reaction and Western blot, we investigated the KLF5 expression changes in a myocardial infarction (MI) mouse model and heart tissue from patients with ischemic heart failure. Using 2D echocardiography, we evaluated the effect of KLF5 inhibition after MI using pharmacological KLF5 inhibitor ML264 and mice with cardiomyocyte-specific KLF5 deletion (αMHC [α-myosin heavy chain]-KLF5-/-). We identified the involvement of KLF5 in regulating lipid metabolism and ceramide accumulation after MI using liquid chromatography-tandem mass spectrometry, and Western blot and real-time polymerase chain reaction analysis of ceramide metabolism-related genes. We lastly evaluated the effect of cardiomyocyte-specific KLF5 overexpression (αMHC-rtTA [reverse tetracycline-controlled transactivator]-KLF5) on cardiac function and ceramide metabolism, and rescued the phenotype using myriocin to inhibit ceramide biosynthesis. RESULTS: KLF5 mRNA and protein levels were higher in human ischemic heart failure samples and in rodent models at 24 hours, 2 weeks, and 4 weeks post-permanent left coronary artery ligation. αMHC-KLF5-/- mice and mice treated with ML264 had higher ejection fraction and lower ventricular volume and heart weight after MI. Lipidomic analysis showed that αMHC-KLF5-/- mice with MI had lower myocardial ceramide levels compared with littermate control mice with MI, although basal ceramide content of αMHC-KLF5-/- mice was not different in control mice. KLF5 ablation suppressed the expression of SPTLC1 and SPTLC2 (serine palmitoyltransferase [SPT] long-chain base subunit ()1 2, respectively), which regulate de novo ceramide biosynthesis. We confirmed our previous findings that myocardial SPTLC1 and SPTLC2 levels are increased in heart failure patients. Consistently, αMHC-rtTA-KLF5 mice showed increased SPTLC1 and SPTLC2 expression, higher myocardial ceramide levels, and systolic dysfunction beginning 2 weeks after KLF5 induction. Treatment of αMHC-rtTA-KLF5 mice with myriocin that inhibits SPT, suppressed myocardial ceramide levels and alleviated systolic dysfunction. CONCLUSIONS: KLF5 is induced during the development of ischemic heart failure in humans and mice and stimulates ceramide biosynthesis. Genetic or pharmacological inhibition of KLF5 in mice with MI prevents ceramide accumulation, alleviates eccentric remodeling, and increases ejection fraction. Thus, KLF5 emerges as a novel therapeutic target for the treatment of ischemic heart failure.


Subject(s)
Cardiomyopathies/physiopathology , Ceramides/metabolism , Kruppel-Like Transcription Factors/metabolism , Myocytes, Cardiac/metabolism , Ventricular Remodeling/physiology , Animals , Disease Models, Animal , Humans , Male , Mice
7.
Lasers Surg Med ; 54(5): 790-803, 2022 07.
Article in English | MEDLINE | ID: mdl-35212407

ABSTRACT

OBJECTIVES: The purpose of this study was to show that optical coherence tomography (OCT) and thermal imaging can be used to monitor changes in the structure and activity of caries lesions over time after treatment with silver diamine fluoride (SDF). METHODS: Artificial caries lesions were formed on enamel and dentin bovine blocks. Each block was partitioned into five windows with the central three windows exposed to a demineralization solution to create lesions: one sound window served as a sound control (SC), one sound window was exposed to SDF to serve as a test control (SCT), one lesion window served as a lesion control (LC), one lesion window received one application of SDF (L1), while the other lesion window received two applications of SDF (L2). Each window was scanned using OCT before SDF application, and every week subsequently, for 12 weeks after initial SDF treatment. Changes in the mean intensity and the width of the peak of increased reflectivity due to the lesion and SDF along with the intensity at a depth of 180 µm from the surface representing optical penetration through the lesion were monitored. Changes in the heat lost, ΔQ (temperature integrated over time) of each window during drying with air were also monitored using a thermal imaging camera. Transverse microradiography (TMR), and high-resolution microscopy were also used for the analysis of selected samples. RESULTS: The reflectivity and optical penetration of sound and lesion areas of enamel and dentin manifested significant changes in OCT images after SDF application. Thermal imaging showed significant differences in ΔQ indicative of permeability changes in the sound and lesion areas of enamel and dentin after SDF application.


Subject(s)
Dental Caries , Tomography, Optical Coherence , Animals , Cattle , Dental Caries/pathology , Dentin/pathology , Fluorides, Topical , Proof of Concept Study , Quaternary Ammonium Compounds , Silver Compounds , Tomography, Optical Coherence/methods
8.
Gastroenterology ; 157(5): 1413-1428.e11, 2019 11.
Article in English | MEDLINE | ID: mdl-31352001

ABSTRACT

BACKGROUND & AIMS: Obesity is a risk factor for pancreatic cancer. In mice, a high-fat diet (HFD) and expression of oncogenic KRAS lead to development of invasive pancreatic ductal adenocarcinoma (PDAC) by unknown mechanisms. We investigated how oncogenic KRAS regulates the expression of fibroblast growth factor 21, FGF21, a metabolic regulator that prevents obesity, and the effects of recombinant human FGF21 (rhFGF21) on pancreatic tumorigenesis. METHODS: We performed immunohistochemical analyses of FGF21 levels in human pancreatic tissue arrays, comprising 59 PDAC specimens and 45 nontumor tissues. We also studied mice with tamoxifen-inducible expression of oncogenic KRAS in acinar cells (KrasG12D/+ mice) and fElasCreERT mice (controls). KrasG12D/+ mice were placed on an HFD or regular chow diet (control) and given injections of rhFGF21 or vehicle; pancreata were collected and analyzed by histology, immunoblots, quantitative polymerase chain reaction, and immunohistochemistry. We measured markers of inflammation in the pancreas, liver, and adipose tissue. Activity of RAS was measured based on the amount of bound guanosine triphosphate. RESULTS: Pancreatic tissues of mice expressed high levels of FGF21 compared with liver tissues. FGF21 and its receptor proteins were expressed by acinar cells. Acinar cells that expressed KrasG12D/+ had significantly lower expression of Fgf21 messenger RNA compared with acinar cells from control mice, partly due to down-regulation of PPARG expression-a transcription factor that activates Fgf21 transcription. Pancreata from KrasG12D/+ mice on a control diet and given injections of rhFGF21 had reduced pancreatic inflammation, infiltration by immune cells, and acinar-to-ductal metaplasia compared with mice given injections of vehicle. HFD-fed KrasG12D/+ mice given injections of vehicle accumulated abdominal fat, developed extensive inflammation, pancreatic cysts, and high-grade pancreatic intraepithelial neoplasias (PanINs); half the mice developed PDAC with liver metastases. HFD-fed KrasG12D/+ mice given injections of rhFGF21 had reduced accumulation of abdominal fat and pancreatic triglycerides, fewer pancreatic cysts, reduced systemic and pancreatic markers of inflammation, fewer PanINs, and longer survival-only approximately 12% of the mice developed PDACs, and none of the mice had metastases. Pancreata from HFD-fed KrasG12D/+ mice given injections of rhFGF21 had lower levels of active RAS than from mice given vehicle. CONCLUSIONS: Normal acinar cells from mice and humans express high levels of FGF21. In mice, acinar expression of oncogenic KRAS significantly reduces FGF21 expression. When these mice are placed on an HFD, they develop extensive inflammation, pancreatic cysts, PanINs, and PDACs, which are reduced by injection of FGF21. FGF21 also reduces the guanosine triphosphate binding capacity of RAS. FGF21 might be used in the prevention or treatment of pancreatic cancer.


Subject(s)
Acinar Cells/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Cell Transformation, Neoplastic/metabolism , Diet, High-Fat , Fibroblast Growth Factors/metabolism , Pancreatic Intraductal Neoplasms/metabolism , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Acinar Cells/pathology , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/prevention & control , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Down-Regulation , Fibroblast Growth Factors/genetics , Gene Expression Regulation, Neoplastic , Humans , Klotho Proteins , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Transgenic , Mutation , PPAR gamma/genetics , PPAR gamma/metabolism , Pancreatic Cyst/genetics , Pancreatic Cyst/metabolism , Pancreatic Cyst/pathology , Pancreatic Intraductal Neoplasms/genetics , Pancreatic Intraductal Neoplasms/pathology , Pancreatic Intraductal Neoplasms/prevention & control , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/prevention & control , Pancreatitis/genetics , Pancreatitis/metabolism , Pancreatitis/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
9.
Development ; 144(5): 737-754, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28246209

ABSTRACT

Krüppel-like factors (KLFs) are a family of zinc-finger transcription factors that are found in many species. Recent studies have shown that KLFs play a fundamental role in regulating diverse biological processes such as cell proliferation, differentiation, development and regeneration. Of note, several KLFs are also crucial for maintaining pluripotency and, hence, have been linked to reprogramming and regenerative medicine approaches. Here, we review the crucial functions of KLFs in mammalian embryogenesis, stem cell biology and regeneration, as revealed by studies of animal models. We also highlight how KLFs have been implicated in human diseases and outline potential avenues for future research.


Subject(s)
Embryonic Development , Kruppel-Like Transcription Factors/metabolism , Lung/embryology , Stem Cells/cytology , Animals , Cell Differentiation , Cell Lineage , Female , Humans , Male , Mice , Mice, Transgenic , Phylogeny , Regeneration , Transcription Factors/metabolism , Zinc Fingers
10.
Gastroenterology ; 154(5): 1494-1508.e13, 2018 04.
Article in English | MEDLINE | ID: mdl-29248441

ABSTRACT

BACKGROUND & AIMS: Activating mutations in KRAS are detected in most pancreatic ductal adenocarcinomas (PDACs). Expression of an activated form of KRAS (KrasG12D) in pancreata of mice is sufficient to induce formation of pancreatic intraepithelial neoplasia (PanINs)-a precursor of PDAC. Pancreatitis increases formation of PanINs in mice that express KrasG12D by promoting acinar-to-ductal metaplasia (ADM). We investigated the role of the transcription factor Krüppel-like factor 5 (KLF5) in ADM and KRAS-mediated formation of PanINs. METHODS: We performed studies in adult mice with conditional disruption of Klf5 (Klf5fl/fl) and/or expression of KrasG12D (LSL-KrasG12D) via CreERTM recombinase regulated by an acinar cell-specific promoter (Ptf1a). Activation of KrasG12D and loss of KLF5 was achieved by administration of tamoxifen. Pancreatitis was induced in mice by administration of cerulein; pancreatic tissues were collected, analyzed by histology and immunohistochemistry, and transcriptomes were compared between mice that did or did not express KLF5. We performed immunohistochemical analyses of human tissue microarrays, comparing levels of KLF5 among 96 human samples of PDAC. UN-KC-6141 cells (pancreatic cancer cells derived from Pdx1-Cre;LSL-KrasG12D mice) were incubated with inhibitors of different kinases and analyzed in proliferation assays and by immunoblots. Expression of KLF5 was knocked down with small hairpin RNAs or CRISPR/Cas9 strategies; cells were analyzed in proliferation and gene expression assays, and compared with cells expressing control vectors. Cells were subcutaneously injected into flanks of syngeneic mice and tumor growth was assessed. RESULTS: Of the 96 PDAC samples analyzed, 73% were positive for KLF5 (defined as nuclear staining in more than 5% of tumor cells). Pancreata from Ptf1a-CreERTM;LSL-KrasG12D mice contained ADM and PanIN lesions, which contained high levels of nuclear KLF5 within these structures. In contrast, Ptf1a-CreERTM;LSL-KrasG12D;Klf5fl/fl mice formed fewer PanINs. After cerulein administration, Ptf1a-CreERTM;LSL-KrasG12D mice formed more extensive ADM than Ptf1a-CreERTM;LSL-KrasG12D;Klf5fl/fl mice. Pancreata from Ptf1a-CreERTM;LSL-KrasG12D;Klf5fl/fl mice had increased expression of the tumor suppressor NDRG2 and reduced phosphorylation (activation) of STAT3, compared with Ptf1a-CreERTM;LSL-KrasG12D mice. In UN-KC-6141 cells, PI3K and MEK signaling increased expression of KLF5; a high level of KLF5 increased proliferation. Cells with knockdown of Klf5 had reduced proliferation, compared with control cells, had reduced expression of ductal markers, and formed smaller tumors (71.61 ± 30.79 mm3 vs 121.44 ± 34.90 mm3 from control cells) in flanks of mice. CONCLUSION: Levels of KLF5 are increased in human PDAC samples and in PanINs of Ptf1a-CreERTM;LSL-KrasG12D mice, compared with controls. KLF5 disruption increases expression of NDRG2 and reduces activation of STAT3 and reduces ADM and PanINs formation in mice. Strategies to reduce KLF5 activity might reduce progression of acinar cells from ADM to PanIN and pancreatic tumorigenesis.


Subject(s)
Carcinoma in Situ/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Kruppel-Like Transcription Factors/metabolism , Pancreatic Neoplasms/metabolism , Animals , Carcinoma in Situ/genetics , Carcinoma in Situ/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Ceruletide , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Genes, ras , Humans , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/genetics , Metaplasia , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatitis/chemically induced , Pancreatitis/genetics , Pancreatitis/metabolism , Pancreatitis/pathology , RNA Interference , Signal Transduction , Time Factors , Transfection , Tumor Burden
11.
Cell Commun Signal ; 17(1): 19, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30819189

ABSTRACT

Oncogenic KRAS plays a vital role in controlling tumor metabolism by enhancing aerobic glycolysis. Obesity driven by chronic consumption of high-fat diet (HFD) is a major risk factor for oncogenic KRAS-mediated pancreatic ductal adenocarcinoma (PDAC). However, the role of HFD in KRAS-mediated metabolic reprogramming has been obscure. Here, by using genetically engineered mouse models expressing an endogenous level of KRASG12D in pancreatic acinar cells, we demonstrate that hyperactivation of KRASG12D by obesogenic HFD, as compared to carbohydrate-rich diet, is responsible for enhanced aerobic glycolysis that associates with critical pathogenic responses in the path towards PDAC. Ablation of Cox-2 attenuates KRAS hyperactivation leading to the reversal of both aggravated aerobic glycolysis and high-grade dysplasia under HFD challenge. Our data highlight a pivotal role of the cooperative interaction between obesity-ensuing HFD and oncogenic KRAS in driving the heightened aerobic glycolysis during pancreatic tumorigenesis and suggest that in addition to directly targeting KRAS and aerobic glycolysis pathway, strategies to target the upstream of KRAS hyperactivation may bear important therapeutic value.


Subject(s)
Diet, High-Fat , Glycolysis , Obesity/metabolism , Oncogenes , Proto-Oncogene Proteins p21(ras)/metabolism , Aerobiosis , Animals , Cyclooxygenase 2/metabolism , Dietary Carbohydrates , Mice , Models, Biological , Obesity/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms
12.
J Am Soc Nephrol ; 29(10): 2529-2545, 2018 10.
Article in English | MEDLINE | ID: mdl-30143559

ABSTRACT

BACKGROUND: Podocyte injury is the hallmark of proteinuric kidney diseases, such as FSGS and minimal change disease, and destabilization of the podocyte's actin cytoskeleton contributes to podocyte dysfunction in many of these conditions. Although agents, such as glucocorticoids and cyclosporin, stabilize the actin cytoskeleton, systemic toxicity hinders chronic use. We previously showed that loss of the kidney-enriched zinc finger transcription factor Krüppel-like factor 15 (KLF15) increases susceptibility to proteinuric kidney disease and attenuates the salutary effects of retinoic acid and glucocorticoids in the podocyte. METHODS: We induced podocyte-specific KLF15 in two proteinuric murine models, HIV-1 transgenic (Tg26) mice and adriamycin (ADR)-induced nephropathy, and used RNA sequencing of isolated glomeruli and subsequent enrichment analysis to investigate pathways mediated by podocyte-specific KLF15 in Tg26 mice. We also explored in cultured human podocytes the potential mediating role of Wilms Tumor 1 (WT1), a transcription factor critical for podocyte differentiation. RESULTS: In Tg26 mice, inducing podocyte-specific KLF15 attenuated podocyte injury, glomerulosclerosis, tubulointerstitial fibrosis, and inflammation, while improving renal function and overall survival; it also attenuated podocyte injury in ADR-treated mice. Enrichment analysis of RNA sequencing from the Tg26 mouse model shows that KLF15 induction activates pathways involved in stabilization of actin cytoskeleton, focal adhesion, and podocyte differentiation. Transcription factor enrichment analysis, with further experimental validation, suggests that KLF15 activity is in part mediated by WT1. CONCLUSIONS: Inducing podocyte-specific KLF15 attenuates kidney injury by directly and indirectly upregulating genes critical for podocyte differentiation, suggesting that KLF15 induction might be a potential strategy for treating proteinuric kidney disease.


Subject(s)
DNA-Binding Proteins/biosynthesis , Kidney Diseases/metabolism , Podocytes/metabolism , Proteinuria/metabolism , Transcription Factors/biosynthesis , Actin Cytoskeleton/metabolism , Animals , Cell Differentiation , Cells, Cultured , DNA-Binding Proteins/genetics , Disease Models, Animal , Focal Adhesions , Gene Knockdown Techniques , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/metabolism , Glomerulosclerosis, Focal Segmental/pathology , Humans , Kidney Diseases/genetics , Kidney Diseases/pathology , Kruppel-Like Transcription Factors/antagonists & inhibitors , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Transgenic , Nephrosis, Lipoid/genetics , Nephrosis, Lipoid/metabolism , Nephrosis, Lipoid/pathology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Podocytes/pathology , Proteinuria/genetics , Proteinuria/pathology , Transcription Factors/genetics , Up-Regulation , WT1 Proteins/antagonists & inhibitors , WT1 Proteins/genetics , WT1 Proteins/metabolism
13.
Gastroenterology ; 152(8): 1845-1875, 2017 06.
Article in English | MEDLINE | ID: mdl-28366734

ABSTRACT

Specificity proteins (SPs) and Krüppel-like factors (KLFs) belong to the family of transcription factors that contain conserved zinc finger domains involved in binding to target DNA sequences. Many of these proteins are expressed in different tissues and have distinct tissue-specific activities and functions. Studies have shown that SPs and KLFs regulate not only physiological processes such as growth, development, differentiation, proliferation, and embryogenesis, but pathogenesis of many diseases, including cancer and inflammatory disorders. Consistently, these proteins have been shown to regulate normal functions and pathobiology in the digestive system. We review recent findings on the tissue- and organ-specific functions of SPs and KLFs in the digestive system including the oral cavity, esophagus, stomach, small and large intestines, pancreas, and liver. We provide a list of agents under development to target these proteins.


Subject(s)
Digestive System Diseases/metabolism , Digestive System/metabolism , Kruppel-Like Transcription Factors/metabolism , Sp Transcription Factors/metabolism , Animals , Digestive System/pathology , Digestive System/physiopathology , Digestive System Diseases/genetics , Digestive System Diseases/pathology , Digestive System Diseases/physiopathology , Gene Expression Regulation , Humans , Kruppel-Like Transcription Factors/genetics , Signal Transduction , Sp Transcription Factors/genetics
14.
J Am Soc Nephrol ; 28(1): 166-184, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27288011

ABSTRACT

Podocyte injury is the inciting event in primary glomerulopathies, such as minimal change disease and primary FSGS, and glucocorticoids remain the initial and often, the primary treatment of choice for these glomerulopathies. Because inflammation is not readily apparent in these diseases, understanding the direct effects of glucocorticoids on the podocyte, independent of the immunomodulatory effects, may lead to the identification of targets downstream of glucocorticoids that minimize toxicity without compromising efficacy. Several studies showed that treatment with glucocorticoids restores podocyte differentiation markers and normal ultrastructure and improves cell survival in murine podocytes. We previously determined that Krüppel-like factor 15 (KLF15), a kidney-enriched zinc finger transcription factor, is required for restoring podocyte differentiation markers in mice and human podocytes under cell stress. Here, we show that in vitro treatment with dexamethasone induced a rapid increase of KLF15 expression in human and murine podocytes and enhanced the affinity of glucocorticoid receptor binding to the promoter region of KLF15 In three independent proteinuric murine models, podocyte-specific loss of Klf15 abrogated dexamethasone-induced podocyte recovery. Furthermore, knockdown of KLF15 reduced cell survival and destabilized the actin cytoskeleton in differentiated human podocytes. Conversely, overexpression of KLF15 stabilized the actin cytoskeleton under cell stress in human podocytes. Finally, the level of KLF15 expression in the podocytes and glomeruli from human biopsy specimens correlated with glucocorticoid responsiveness in 35 patients with minimal change disease or primary FSGS. Thus, these studies identify the critical role of KLF15 in mediating the salutary effects of glucocorticoids in the podocyte.


Subject(s)
Cell Differentiation/drug effects , DNA-Binding Proteins/physiology , Glucocorticoids/pharmacology , Podocytes/cytology , Podocytes/drug effects , Transcription Factors/physiology , Adolescent , Adult , Animals , Antigens, Differentiation/drug effects , Child , Dexamethasone/pharmacology , Female , Glomerulosclerosis, Focal Segmental/immunology , Humans , Kruppel-Like Transcription Factors , Male , Mice , Middle Aged , Nephrosis, Lipoid/immunology , Young Adult
15.
Physiol Rev ; 90(4): 1337-81, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20959618

ABSTRACT

The Krüppel-like factor (KLF) family of transcription factors regulates diverse biological processes that include proliferation, differentiation, growth, development, survival, and responses to external stress. Seventeen mammalian KLFs have been identified, and numerous studies have been published that describe their basic biology and contribution to human diseases. KLF proteins have received much attention because of their involvement in the development and homeostasis of numerous organ systems. KLFs are critical regulators of physiological systems that include the cardiovascular, digestive, respiratory, hematological, and immune systems and are involved in disorders such as obesity, cardiovascular disease, cancer, and inflammatory conditions. Furthermore, KLFs play an important role in reprogramming somatic cells into induced pluripotent stem (iPS) cells and maintaining the pluripotent state of embryonic stem cells. As research on KLF proteins progresses, additional KLF functions and associations with disease are likely to be discovered. Here, we review the current knowledge of KLF proteins and describe common attributes of their biochemical and physiological functions and their pathophysiological roles.


Subject(s)
Kruppel-Like Transcription Factors/physiology , Amino Acid Sequence , Animals , Gene Expression Regulation/physiology , Humans , Kruppel-Like Transcription Factors/chemistry , Kruppel-Like Transcription Factors/classification , Phylogeny
16.
Kidney Int ; 92(5): 1178-1193, 2017 11.
Article in English | MEDLINE | ID: mdl-28651950

ABSTRACT

Large epidemiological studies clearly demonstrate that multiple episodes of acute kidney injury contribute to the development and progression of kidney fibrosis. Although our understanding of kidney fibrosis has improved in the past two decades, we have limited therapeutic strategies to halt its progression. Myofibroblast differentiation and proliferation remain critical to the progression of kidney fibrosis. Although canonical Wnt signaling can trigger the activation of myofibroblasts in the kidney, mediators of Wnt inhibition in the resident progenitor cells are unclear. Recent studies demonstrate that the loss of a Krüppel-like factor 15 (KLF15), a kidney-enriched zinc-finger transcription factor, exacerbates kidney fibrosis in murine models. Here, we tested whether Klf15 mRNA and protein expression are reduced in late stages of fibrosis in mice that underwent unilateral ureteric obstruction, a model of progressive renal fibrosis. Knockdown of Klf15 in Foxd1-expressing cells (Foxd1-Cre Klf15fl/fl) increased extracellular matrix deposition and myofibroblast proliferation as compared to wildtype (Foxd1-Cre Klf15+/+) mice after three and seven days of ureteral obstruction. This was validated in mice receiving angiotensin II treatment for six weeks. In both these murine models, the increase in renal fibrosis was found in Foxd1-Cre Klf15fl/fl mice and accompanied by the activation of Wnt/ß-catenin signaling. Furthermore, knockdown of Klf15 in cultured mouse embryonic fibroblasts activated canonical Wnt/ß-catenin signaling, increased profibrotic transcripts, and increased proliferation after treatment with a Wnt1 ligand. Conversely, the overexpression of KLF15 inhibited phospho-ß-catenin (Ser552) expression in Wnt1-treated cells. Thus, KLF15 has a critical role in attenuating kidney fibrosis by inhibiting the canonical Wnt/ß-catenin pathway.


Subject(s)
DNA-Binding Proteins/metabolism , Forkhead Transcription Factors/metabolism , Kidney Diseases/pathology , Kidney/pathology , Myofibroblasts/pathology , Transcription Factors/metabolism , Wnt Signaling Pathway , Angiotensin II/toxicity , Animals , Cell Proliferation , Cells, Cultured , DNA-Binding Proteins/genetics , Disease Models, Animal , Disease Progression , Fibrosis , Gene Expression Regulation , Gene Knockdown Techniques , Kidney/cytology , Kidney Diseases/etiology , Kruppel-Like Transcription Factors , Male , Mice , Mice, Inbred C57BL , Myofibroblasts/metabolism , Phosphorylation , RNA, Messenger/metabolism , Stromal Cells/metabolism , Stromal Cells/pathology , Transcription Factors/genetics , Wnt1 Protein/metabolism , beta Catenin/metabolism
17.
Am J Physiol Gastrointest Liver Physiol ; 313(5): G478-G491, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28864500

ABSTRACT

Krüppel-like factor 5 (KLF5) is a member of the zinc finger family of transcription factors that regulates homeostasis of the intestinal epithelium. Previous studies suggested an indispensable role of KLF5 in maintaining intestinal barrier function. In the current study, we investigated the mechanisms by which KLF5 regulates colonic barrier function in vivo and in vitro. We used an inducible and a constitutive intestine-specific Klf5 knockout mouse models (Villin-CreERT2;Klf5fl/fl designated as Klf5ΔIND and Villin-Cre;Klf5fl/fl as Klf5ΔIS ) and studied an inducible KLF5 knockdown in Caco-2 BBe cells using a lentiviral Tet-on system (Caco-2 BBe KLF5ΔIND). Specific knockout of Klf5 in colonic tissues, either inducible or constitutive, resulted in increased intestinal permeability. The phenotype was accompanied by a significant reduction in Dsg2, which encodes desmoglein-2, a desmosomal cadherin, at both mRNA and protein levels. Transmission electron microscopy showed alterations of desmosomal morphology in both KLF5 knockdown Caco-2 BBe cells and Klf5 knockout mouse colonic tissues. Inducible knockdown of KLF5 in Caco-2BBe cells grown on Transwell plates led to impaired barrier function as evidenced by decreased transepithelial electrical resistance and increased paracellular permeability to fluorescein isothiocyanate-4 kDa dextran. Furthermore, DSG2 was significantly decreased in KLF5 knockdown cells, and DSG2 overexpression partially rescued the impaired barrier function caused by KLF5 knockdown. Electron microscopy studies demonstrated altered desmosomal morphology after KLF5 knockdown. In combination with chromatin immunoprecipitation analysis and promoter study, our data show that KLF5 regulates intestinal barrier function by mediating the transcription of DSG2, a gene encoding a major component of desmosome structures.NEW & NOTEWORTHY The study is original research on the direct function of a Krüppel-like factor on intestinal barrier function, which is commonly exerted by cell junctions, including tight junctions, adherens junctions, and desmosomes. Numerous previous studies were focused on tight junctions and adherens junctions. However, this study provided a new perspective on how the intestinal barrier function is regulated by KLF5 through DSG2, a component of desmosome complexes.


Subject(s)
Colon/physiology , Kruppel-Like Transcription Factors/physiology , Animals , Caco-2 Cells , Desmocollins , Desmoglein 2/biosynthesis , Desmoglein 2/genetics , Desmosomes/ultrastructure , Electric Impedance , Gene Expression Regulation/genetics , Gene Knockdown Techniques , HEK293 Cells , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/ultrastructure , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Permeability , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
18.
Cell Tissue Res ; 370(3): 441-449, 2017 12.
Article in English | MEDLINE | ID: mdl-28856432

ABSTRACT

Kruppel-like factor 4 (KLF4) is a zinc finger transcription factor that plays crucial roles during the development and maintenance of multiple organs. We and others have previously shown that KLF4 is involved in bone modeling and remodeling but roles played by KLF4 during skeletogenesis are still not fully understood. Here, we show that KLF4 is expressed in the epiphyseal growth plate and articular chondrocytes. Most articular chondrocytes expressed KLF4 in embryos but it localized only in a subset of superficial zone cells in postnatal mice. When KLF4 was overexpressed in chondrocytes in vitro, it severely repressed chondrocytic gene expressions. Global gene expression profiling of KLF4-transduced chondrocytes revealed matrix degrading proteinases of the matrix metalloproteinase and disintegrin and metalloproteinase with thrombospondin-1 domain families within the group of upregulated genes. Proteinase induction by KLF4 was alleviated by Trichostatin A treatment suggesting the possible involvement of epigenetic mechanisms on proteinase induction by KLF4. These results indicate the possible involvement of KLF4 in physiological and pathological aspects during cartilage development and maintenance.


Subject(s)
Cartilage, Articular/metabolism , Chondrocytes/metabolism , Endopeptidases/biosynthesis , Kruppel-Like Transcription Factors/metabolism , Matrix Metalloproteinases/biosynthesis , Thrombospondin 1/biosynthesis , Animals , Cells, Cultured , Endopeptidases/genetics , Gene Expression Regulation, Developmental , Hydroxamic Acids/pharmacology , Kruppel-Like Factor 4 , Male , Matrix Metalloproteinases/genetics , Mice , Mice, Inbred ICR , Protein Synthesis Inhibitors/pharmacology , Thrombospondin 1/genetics
19.
FASEB J ; 30(12): 4159-4171, 2016 12.
Article in English | MEDLINE | ID: mdl-27609772

ABSTRACT

Alterations in sphingolipid metabolism, especially ceramide and sphingosine 1-phosphate, have been linked to colon cancer, suggesting that enzymes of sphingolipid metabolism may emerge as novel regulators and targets in colon cancer. Neutral ceramidase (nCDase), a key enzyme in sphingolipid metabolism that hydrolyzes ceramide into sphingosine, is highly expressed in the intestine; however, its role in colon cancer has not been defined. Here we show that molecular and pharmacological inhibition of nCDase in colon cancer cells increases ceramide, and this is accompanied by decreased cell survival and increased apoptosis and autophagy, with minimal effects on noncancerous cells. Inhibition of nCDase resulted in loss of ß-catenin and inhibition of ERK, components of pathways relevant for colon cancer development. Furthermore, inhibition of nCDase in a xenograft model delayed tumor growth and increased ceramide while decreasing proliferation. It is noteworthy that mice lacking nCDase treated with azoxymethane were protected from tumor formation. Taken together, these studies show that nCDase is pivotal for regulating initiation and development of colon cancer, and these data suggest that this enzyme is a suitable and novel target for colon cancer therapy.-García-Barros, M., Coant, N., Kawamori, T., Wada, M., Snider, A. J., Truman, J.-P., Wu, B. X., Furuya, H., Clarke, C. J., Bialkowska, A. B., Ghaleb, A., Yang, V. W., Obeid, L. M., Hannun, Y. A. Role of neutral ceramidase in colon cancer.


Subject(s)
Ceramides/metabolism , Colonic Neoplasms/enzymology , Lipid Metabolism/physiology , Neutral Ceramidase/metabolism , Animals , Colon/metabolism , Humans , Male , Mice, Knockout , Sphingolipids/metabolism , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL