71 results on '"Bialkowska AB"'
Search Results
2. TR-107, an Agonist of Caseinolytic Peptidase Proteolytic Subunit, Disrupts Mitochondrial Metabolism and Inhibits the Growth of Human Colorectal Cancer Cells.
- Author
-
Giarrizzo M, LaComb JF, Patel HR, Reddy RG, Haley JD, Graves LM, Iwanowicz EJ, and Bialkowska AB
- Subjects
- Humans, Cell Line, Tumor, Oxidative Phosphorylation drug effects, Mitochondria metabolism, Mitochondria drug effects, Colorectal Neoplasms drug therapy, Colorectal Neoplasms metabolism, Colorectal Neoplasms pathology, Cell Proliferation drug effects, Endopeptidase Clp metabolism
- Abstract
Oxidative phosphorylation is an essential metabolic process for cancer proliferation and therapy resistance. The ClpXP complex maintains mitochondrial proteostasis by degrading misfolded proteins. Madera Therapeutics has developed a class of highly potent and selective small-molecule activators (TR compounds) of the ClpXP component caseinolytic peptidase proteolytic subunit (ClpP). This approach to cancer therapy eliminates substrate recognition and activates nonspecific protease function within mitochondria, which has shown encouraging preclinical efficacy in multiple malignancies. The class-leading compound TR-107 has demonstrated significantly improved potency in ClpP affinity and activation and enhanced pharmacokinetic properties over the multitargeting clinical agent ONC201. In this study, we investigate the in vitro efficacy of TR-107 against human colorectal cancer cells. TR-107 inhibited colorectal cancer cell proliferation in a dose- and time-dependent manner and induced cell cycle arrest at low nanomolar concentrations. Mechanistically, TR-107 downregulated the expression of proteins involved in the mitochondrial unfolded protein response and mitochondrial DNA transcription and translation. TR-107 attenuated oxygen consumption rate and glycolytic compensation, confirming inactivation of oxidative phosphorylation and a reduction in total cellular respiration. Multiomics analysis of treated cells indicated a downregulation of respiratory chain complex subunits and an upregulation of mitophagy and ferroptosis pathways. Further evaluation of ferroptosis revealed a depletion of antioxidant and iron toxicity defenses that could potentiate sensitivity to combinatory chemotherapeutics. Together, this study provides evidence and insight into the subcellular mechanisms employed by colorectal cancer cells in response to potent ClpP agonism. Our findings demonstrate a productive approach to disrupting mitochondrial metabolism, supporting the translational potential of TR-107., (©2024 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
3. A graded neonatal mouse model of necrotizing enterocolitis demonstrates that mild enterocolitis is sufficient to activate microglia and increase cerebral cytokine expression.
- Author
-
Sha C, Van Brunt T, Kudria J, Schmidt D, Yurovsky A, Bandovic J, Giarrizzo M, Lin J, Tsirka SA, Bialkowska AB, Wollmuth L, Speer E, and Hsieh H
- Abstract
Background: Necrotizing enterocolitis (NEC) is an inflammatory gastrointestinal process that afflicts approximately 10% of preterm infants born in the United States each year, with a mortality rate of 30%. NEC severity is graded using Bell's classification system, from stage I mild NEC to stage III severe NEC. Over half of NEC survivors present with neurodevelopmental impairment during adolescence, a long-term complication that is poorly understood but can occur even after mild NEC. Although multiple animal models exist, none allow the experimenter to control nor represent the gradient of symptom severities seen in NEC patients. We bridge this knowledge gap by developing a graded murine model of NEC and studying its relationship with neuroinflammation across a range of NEC severities., Methods: Postnatal day 3 (P3) C57BL/6 mice were fed a formula containing different concentrations (0% control, 0.25%, 1%, 2%, and 3%) of dextran sodium sulfate (DSS). P3 mice were fed every 3 hours for 72-hours. We collected data on weight gain and behavior (activity, response, body color) during feeding. At the end of the experiment, we collected tissues (intestine, liver, plasma, brain) for immunohistochemistry, immunofluorescence, and cytokine and chemokine analysis., Results: Throughout NEC induction, mice fed higher concentrations of DSS died sooner, lost weight faster, and became sick or lethargic earlier. Intestinal characteristics (dilation, color, friability) were worse in mice fed with higher DSS concentrations. Histology revealed small intestinal disarray among mice fed all DSS concentrations, while higher DSS concentrations resulted in reduced small intestinal cellular proliferation and increased hepatic and systemic inflammation. In the brain, IL-2, G-CSF, and CXCL1 concentrations increased with higher DSS concentrations. Although the number of neurons and microglia in the CA1 hippocampal region did not differ, microglial branching was significantly reduced in DSS-fed mice., Conclusion: We characterize a novel graded model of NEC that recapitulates the full range of NEC severities. We show that mild NEC is sufficient to initiate neuroinflammation and microglia activation. This model will facilitate studies on the neurodevelopmental effects of NEC.
- Published
- 2024
- Full Text
- View/download PDF
4. Activin A signaling stimulates neutrophil activation and macrophage migration in pancreatitis.
- Author
-
Wiley MB, Bauer J, Alvarez V, Mehrotra K, Cheng W, Kolics Z, Giarrizzo M, Ingle K, Bialkowska AB, and Jung B
- Subjects
- Animals, Mice, Humans, Neutrophils metabolism, Neutrophils immunology, Disease Models, Animal, RAW 264.7 Cells, Macrophage Activation, HL-60 Cells, Pancreatitis, Chronic metabolism, Pancreatitis, Chronic pathology, Male, Activins metabolism, Macrophages metabolism, Macrophages immunology, Pancreatitis metabolism, Pancreatitis pathology, Signal Transduction, Cell Movement, Neutrophil Activation
- Abstract
Acute Pancreatitis (AP) is associated with high mortality and current treatment options are limited to supportive care. We found that blockade of activin A (activin) in mice improves outcomes in two murine models of AP. To test the hypothesis that activin is produced early in response to pancreatitis and is maintained throughout disease progression to stimulate immune cells, we first performed digital spatial profiling (DSP) of human chronic pancreatitis (CP) patient tissue. Then, transwell migration assays using RAW264.7 mouse macrophages and qPCR analysis of "neutrophil-like" HL-60 cells were used for functional correlation. Immunofluorescence and western blots on cerulein-induced pancreatitis samples from pancreatic acinar cell-specific Kras knock-in (Ptf1aCre
ER ™; LSL-KrasG12D ) and functional WT Ptf1aCreER ™ mouse lines mimicking AP and CP to allow for in vivo confirmation. Our data suggest activin promotes neutrophil and macrophage activation both in situ and in vitro, while pancreatic activin production is increased as early as 1 h in response to pancreatitis and is maintained throughout CP in vivo. Taken together, activin is produced early in response to pancreatitis and is maintained throughout disease progression to promote neutrophil and macrophage activation., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
5. FABP5 Inhibition against PTEN -Mutant Therapy Resistant Prostate Cancer.
- Author
-
Swamynathan MM, Mathew G, Aziz A, Gordon C, Hillowe A, Wang H, Jhaveri A, Kendall J, Cox H, Giarrizzo M, Azabdaftari G, Rizzo RC, Diermeier SD, Ojima I, Bialkowska AB, Kaczocha M, and Trotman LC
- Abstract
Resistance to standard of care taxane and androgen deprivation therapy (ADT) causes the vast majority of prostate cancer (PC) deaths worldwide. We have developed RapidCaP, an autochthonous genetically engineered mouse model of PC. It is driven by the loss of PTEN and p53, the most common driver events in PC patients with life-threatening diseases. As in human ADT, surgical castration of RapidCaP animals invariably results in disease relapse and death from the metastatic disease burden. Fatty Acid Binding Proteins (FABPs) are a large family of signaling lipid carriers. They have been suggested as drivers of multiple cancer types. Here we combine analysis of primary cancer cells from RapidCaP (RCaP cells) with large-scale patient datasets to show that among the 10 FABP paralogs, FABP5 is the PC-relevant target. Next, we show that RCaP cells are uniquely insensitive to both ADT and taxane treatment compared to a panel of human PC cell lines. Yet, they share an exquisite sensitivity to the small-molecule FABP5 inhibitor SBFI-103. We show that SBFI-103 is well tolerated and can strongly eliminate RCaP tumor cells in vivo. This provides a pre-clinical platform to fight incurable PC and suggests an important role for FABP5 in PTEN -deficient PC.
- Published
- 2023
- Full Text
- View/download PDF
6. Krüppel-like Factor 5 Plays an Important Role in the Pathogenesis of Chronic Pancreatitis.
- Author
-
Alavi M, Mejia-Bautista A, Tang M, Bandovic J, Rosenberg AZ, and Bialkowska AB
- Abstract
Chronic pancreatitis results in the formation of pancreatic intraepithelial neoplasia (PanIN) and poses a risk of developing pancreatic cancer. Our previous study demonstrated that Krüppel-like factor 5 (KLF5) is necessary for forming acinar-to-ductal metaplasia (ADM) in acute pancreatitis. Here, we investigated the role of KLF5 in response to chronic injury in the pancreas. Human tissues originating from chronic pancreatitis patients showed increased levels of epithelial KLF5. An inducible genetic model combining the deletion of Klf5 and the activation of Kras
G12D mutant expression in pancreatic acinar cells together with chemically induced chronic pancreatitis was used. The chronic injury resulted in increased levels of KLF5 in both control and KrasG12D mutant mice. Furthermore, it led to numerous ADM and PanIN lesions and extensive fibrosis in the KRAS mutant mice. In contrast, pancreata with Klf5 loss (with or without KrasG12D ) failed to develop ADM, PanIN, or significant fibrosis. Furthermore, the deletion of Klf5 reduced the expression level of cytokines and fibrotic components such as Il1b , Il6 , Tnf , Tgfb1 , Timp1 , and Mmp9 . Notably, using ChIP-PCR, we showed that KLF5 binds directly to the promoters of Il1b , Il6 , and Tgfb1 genes. In summary, the inactivation of Klf5 inhibits ADM and PanIN formation and the development of pancreatic fibrosis.- Published
- 2023
- Full Text
- View/download PDF
7. AUM302, a novel triple kinase PIM/PI3K/mTOR inhibitor, is a potent in vitro pancreatic cancer growth inhibitor.
- Author
-
Ingle K, LaComb JF, Graves LM, Baines AT, and Bialkowska AB
- Subjects
- Humans, Phosphatidylinositol 3-Kinases metabolism, Growth Inhibitors pharmacology, Gemcitabine, TOR Serine-Threonine Kinases, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Phosphoinositide-3 Kinase Inhibitors pharmacology, Cell Proliferation, Cell Line, Tumor, Pancreatic Neoplasms pathology, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Carcinoma, Pancreatic Ductal pathology
- Abstract
Pancreatic cancer is one of the leading causes of cancer deaths, with pancreatic ductal adenocarcinoma (PDAC) being the most common subtype. Advanced stage diagnosis of PDAC is common, causing limited treatment opportunities. Gemcitabine is a frequently used chemotherapeutic agent which can be used as a monotherapy or in combination. However, tumors often develop resistance to gemcitabine. Previous studies show that the proto-oncogene PIM kinases (PIM1 and PIM3) are upregulated in PDAC compared to matched normal tissue and are related to chemoresistance and PDAC cell growth. The PIM kinases are also involved in the PI3K/AKT/mTOR pathway to promote cell survival. In this study, we evaluate the effect of the novel multikinase PIM/PI3K/mTOR inhibitor, AUM302, and commercially available PIM inhibitor, TP-3654. Using five human PDAC cell lines, we found AUM302 to be a potent inhibitor of cell proliferation, cell viability, cell cycle progression, and phosphoprotein expression, while TP-3654 was less effective. Significantly, AUM302 had a strong impact on the viability of gemcitabine-resistant PDAC cells. Taken together, these results demonstrate that AUM302 exhibits antitumor activity in human PDAC cells and thus has the potential to be an effective drug for PDAC therapy., Competing Interests: The authors have declared that no competing interests exist., (Copyright: © 2023 Ingle et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2023
- Full Text
- View/download PDF
8. Computational immunohistochemical mapping adds immune context to histological phenotypes in mouse models of colitis.
- Author
-
Kobayashi S, Sullivan C, Bialkowska AB, Saltz JH, and Yang VW
- Subjects
- Animals, Mice, Phenotype, Inflammation, Disease Models, Animal, Colitis chemically induced, Inflammatory Bowel Diseases, Colitis, Ulcerative, Crohn Disease
- Abstract
Inflammatory bowel disease (IBD) is characterized by chronic, dysregulated inflammation in the gastrointestinal tract. The heterogeneity of IBD is reflected through two major subtypes, Crohn's Disease (CD) and Ulcerative Colitis (UC). CD and UC differ across symptomatic presentation, histology, immune responses, and treatment. While colitis mouse models have been influential in deciphering IBD pathogenesis, no single model captures the full heterogeneity of clinical disease. The translational capacity of mouse models may be augmented by shifting to multi-mouse model studies that aggregate analysis across various well-controlled phenotypes. Here, we evaluate the value of histology in multi-mouse model characterizations by building upon a previous pipeline that detects histological disease classes in hematoxylin and eosin (H&E)-stained murine colons. Specifically, we map immune marker positivity across serially-sectioned slides to H&E histological classes across the dextran sodium sulfate (DSS) chemical induction model and the intestinal epithelium-specific, inducible Villin-CreER
T2 ;Klf5fl/fl (Klf5ΔIND ) genetic model. In this study, we construct the beginning frameworks to define H&E-patch-based immunophenotypes based on IHC-H&E mappings., (© 2023. Springer Nature Limited.)- Published
- 2023
- Full Text
- View/download PDF
9. Acute Pancreatitis: Current Clinical Approaches, Molecular Pathophysiology, and Potential Therapeutics.
- Author
-
Wiley MB, Mehrotra K, Bauer J, Yazici C, Bialkowska AB, and Jung B
- Subjects
- Humans, Acute Disease, Inflammation therapy, Fluid Therapy, Cytokines, Pancreatitis genetics, Pancreatitis therapy
- Abstract
Objective: Severe acute pancreatitis (SAP), pancreatic inflammation leading to multiorgan failure, is associated with high morbidity and mortality. There is a critical need to identify novel therapeutic strategies to improve clinical outcomes for SAP patients., Materials and Methods: A comprehensive literature review was performed to identify current clinical strategies, known molecular pathophysiology, and potential therapeutic targets for SAP., Results: Current clinical approaches focus on determining which patients will likely develop SAP. However, therapeutic options are limited to supportive care and fluid resuscitation. The application of a novel 5-cytokine panel accurately predicting disease outcomes in SAP suggests that molecular approaches will improve impact of future clinical trials in AP., Conclusions: Inflammatory outcomes in acute pancreatitis are driven by several unique molecular signals, which compound to promote both local and systemic inflammation. The identification of master cytokine regulators is critical to developing therapeutics, which reduce inflammation through several mechanisms., Competing Interests: The authors declare no conflict of interest., (Copyright © 2023 The Author(s). Published by Wolters Kluwer Health, Inc.)
- Published
- 2023
- Full Text
- View/download PDF
10. The Role of Krüppel-like Factors in Pancreatic Physiology and Pathophysiology.
- Author
-
Giarrizzo M, LaComb JF, and Bialkowska AB
- Subjects
- Humans, Transcription Factors, Zinc Fingers, Kruppel-Like Transcription Factors metabolism, Neoplasms
- Abstract
Krüppel-like factors (KLFs) belong to the family of transcription factors with three highly conserved zinc finger domains in the C-terminus. They regulate homeostasis, development, and disease progression in many tissues. It has been shown that KLFs play an essential role in the endocrine and exocrine compartments of the pancreas. They are necessary to maintain glucose homeostasis and have been implicated in the development of diabetes. Furthermore, they can be a vital tool in enabling pancreas regeneration and disease modeling. Finally, the KLF family contains proteins that act as tumor suppressors and oncogenes. A subset of members has a biphasic function, being upregulated in the early stages of oncogenesis and stimulating its progression and downregulated in the late stages to allow for tumor dissemination. Here, we describe KLFs' function in pancreatic physiology and pathophysiology.
- Published
- 2023
- Full Text
- View/download PDF
11. Krüppel-like Factors 4 and 5 in Colorectal Tumorigenesis.
- Author
-
Lee E, Cheung J, and Bialkowska AB
- Abstract
Krüppel-like factors (KLFs) are transcription factors regulating various biological processes such as proliferation, differentiation, migration, invasion, and homeostasis. Importantly, they participate in disease development and progression. KLFs are expressed in multiple tissues, and their role is tissue- and context-dependent. KLF4 and KLF5 are two fascinating members of this family that regulate crucial stages of cellular identity from embryogenesis through differentiation and, finally, during tumorigenesis. They maintain homeostasis of various tissues and regulate inflammation, response to injury, regeneration, and development and progression of multiple cancers such as colorectal, breast, ovarian, pancreatic, lung, and prostate, to name a few. Recent studies broaden our understanding of their function and demonstrate their opposing roles in regulating gene expression, cellular function, and tumorigenesis. This review will focus on the roles KLF4 and KLF5 play in colorectal cancer. Understanding the context-dependent functions of KLF4 and KLF5 and the mechanisms through which they exert their effects will be extremely helpful in developing targeted cancer therapy.
- Published
- 2023
- Full Text
- View/download PDF
12. 5-FU-miR-15a Inhibits Activation of Pancreatic Stellate Cells by Reducing YAP1 and BCL-2 Levels In Vitro.
- Author
-
Almanzar VMD, Shah K, LaComb JF, Mojumdar A, Patel HR, Cheung J, Tang M, Ju J, and Bialkowska AB
- Subjects
- Animals, Mice, Cell Proliferation genetics, Fibrosis, Proto-Oncogene Proteins c-bcl-2 metabolism, YAP-Signaling Proteins metabolism, Fluorouracil pharmacology, MicroRNAs genetics, MicroRNAs metabolism, Pancreatic Stellate Cells drug effects, Pancreatic Stellate Cells pathology, Pancreatitis, Chronic pathology
- Abstract
Chronic pancreatitis is characterized by chronic inflammation and fibrosis, processes heightened by activated pancreatic stellate cells (PSCs). Recent publications have demonstrated that miR-15a, which targets YAP1 and BCL-2 , is significantly downregulated in patients with chronic pancreatitis compared to healthy controls. We have utilized a miRNA modification strategy to enhance the therapeutic efficacy of miR-15a by replacing uracil with 5-fluorouracil (5-FU). We demonstrated increased levels of YAP1 and BCL-2 (both targets of miR-15a) in pancreatic tissues obtained from Ptf1aCre
ERTM and Ptf1aCreERTM ;LSL-KrasG12D mice after chronic pancreatitis induction as compared to controls. In vitro studies showed that delivery of 5-FU-miR-15a significantly decreased viability, proliferation, and migration of PSCs over six days compared to 5-FU, TGFβ1, control miR, and miR-15a. In addition, treatment of PSCs with 5-FU-miR-15a in the context of TGFβ1 treatment exerted a more substantial effect than TGFβ1 alone or when combined with other miRs. Conditioned medium obtained from PSC cells treated with 5-FU-miR-15a significantly inhibits the invasion of pancreatic cancer cells compared to controls. Importantly, we demonstrated that treatment with 5-FU-miR-15a reduced the levels of YAP1 and BCL-2 observed in PSCs. Our results strongly suggest that ectopic delivery of miR mimetics is a promising therapeutic approach for pancreatic fibrosis and that 5-FU-miR-15a shows specific promise.- Published
- 2023
- Full Text
- View/download PDF
13. The Role of MicroRNAs in Pancreatitis Development and Progression.
- Author
-
Patel HR, Diaz Almanzar VM, LaComb JF, Ju J, and Bialkowska AB
- Subjects
- Animals, Pancreas metabolism, Acinar Cells metabolism, MicroRNAs genetics, MicroRNAs metabolism, Pancreatitis genetics, Pancreatitis metabolism, Pancreas, Exocrine metabolism
- Abstract
Pancreatitis (acute and chronic) is an inflammatory disease associated with significant morbidity, including a high rate of hospitalization and mortality. MicroRNAs (miRs) are essential post-transcriptional modulators of gene expression. They are crucial in many diseases' development and progression. Recent studies have demonstrated aberrant miRs expression patterns in pancreatic tissues obtained from patients experiencing acute and chronic pancreatitis compared to tissues from unaffected individuals. Increasing evidence showed that miRs regulate multiple aspects of pancreatic acinar biology, such as autophagy, mitophagy, and migration, impact local and systemic inflammation and, thus, are involved in the disease development and progression. Notably, multiple miRs act on pancreatic acinar cells and regulate the transduction of signals between pancreatic acinar cells, pancreatic stellate cells, and immune cells, and provide a complex interaction network between these cells. Importantly, recent studies from various animal models and patients' data combined with advanced detection techniques support their importance in diagnosing and treating pancreatitis. In this review, we plan to provide an up-to-date summary of the role of miRs in the development and progression of pancreatitis.
- Published
- 2023
- Full Text
- View/download PDF
14. Sonic Hedgehog and WNT Signaling Regulate a Positive Feedback Loop Between Intestinal Epithelial and Stromal Cells to Promote Epithelial Regeneration.
- Author
-
Orzechowska-Licari EJ, Bialkowska AB, and Yang VW
- Subjects
- Animals, Mice, Feedback, Ligands, Regeneration physiology, Stromal Cells metabolism, Hedgehog Proteins genetics, Hedgehog Proteins metabolism, Wnt Signaling Pathway
- Abstract
Background and Aims: Active intestinal stem cells are prone to injury by ionizing radiation. We previously showed that upon radiation-induced injury, normally quiescent reserve intestinal stem cells (rISCs) (marked by BMI1) are activated by Musashi-1 (MSI1) and exit from the quiescent state to regenerate the intestinal epithelium. This study aims to further establish the mechanism that regulates activation of Bmi1-Cre
ER ;Rosa26eYFP (Bmi1-CreER ) rISCs following γ radiation-induced injury., Methods: Bmi1-CreER mice were treated with tamoxifen to initiate lineage tracing of BMI1 (eYFP+ ) cells and exposed to 12 Gy of total body γ irradiation or sham. Intestinal tissues were collected and analyzed by immunofluorescence, Western blot, reverse-transcription quantitative polymerase chain reaction, enzyme-linked immunosorbent assay, and chromatin immunoprecipitation real-time polymerase chain reaction., Results: After irradiation, increased expression of Msi1 in eYFP+ cells was accompanied by increased expression of Axin2, a WNT marker. Promoter studies of the Msi1 gene indicated that Msi1 is a WNT target gene. Coculture of stromal cells isolated from irradiated mice stimulated Bmi1-CreER -derived organoid regeneration more effectively than those from sham mice. Expression of WNT ligands, including Wnt2b, Wnt4, Wnt5a, and Rspo3, was increased in irradiated stromal cells compared with sham-treated stromal cells. Moreover, expression of the Sonic hedgehog (SHH) effector Gli1 was increased in stromal cells from irradiated mice. This was correlated with an increased expression of SHH in epithelial cells postirradiation, indicating epithelial-stromal interaction. Finally, preinjury treatment with SHH inhibitor cyclopamine significantly reduced intestinal epithelial regeneration and Msi1 expression postirradiation., Conclusions: Upon ionizing radiation-induced injury, intestinal epithelial cells increase SHH secretion, stimulating stromal cells to secrete WNT ligands. WNT activators induce Msi1 expression in the Bmi1-CreER cells. This stromal-epithelial interaction leads to Bmi1-CreER rISCs induction and epithelial regeneration., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
15. Acute postoperative pain and dorsal root ganglia transcriptomic signatures following total knee arthroplasty (TKA) in rats: An experimental study.
- Author
-
Komatsu DE, Uddin SMZ, Gordon C, Kanjiya MP, Bogdan D, Achonu J, DiBua A, Iftikhar H, Ackermann A, Shah RJ, Shieh J, Bialkowska AB, and Kaczocha M
- Subjects
- Rats, Animals, Ganglia, Spinal, Pain, Postoperative drug therapy, Pain, Postoperative genetics, Cytokines genetics, Arthroplasty, Replacement, Knee adverse effects
- Abstract
Total knee arthroplasty (TKA) is the final treatment option for patients with advanced knee osteoarthritis (OA). Unfortunately, TKA surgery is accompanied by acute postoperative pain that is more severe than arthroplasty performed in other joints. Elucidating the molecular mechanisms specific to post-TKA pain necessitates an animal model that replicates clinical TKA procedures, induces acute postoperative pain, and leads to complete functional recovery. Here, we present a new preclinical TKA model in rats and report on functional and behavioral outcomes indicative of pain, analgesic efficacy, serum cytokine levels, and dorsal root ganglia (DRG) transcriptomes during the acute postoperative period. Following TKA, rats exhibited marked deficits in weight bearing that persisted for 28 days. Home cage locomotion, rearing, and gait were similarly impacted and recovered by day 14. Cytokine levels were elevated on postoperative days one and/or two. Treatment with morphine, ketorolac, or their combination improved weight bearing while gabapentin lacked efficacy. When TKA was performed in rats with OA, similar functional deficits and comparable recovery time courses were observed. Analysis of DRG transcriptomes revealed upregulation of transcripts linked to multiple molecular pathways including inflammation, MAPK signaling, and cytokine signaling and production. In summary, we developed a clinically relevant rat TKA model characterized by resolution of pain and functional recovery within five weeks and with pain-associated behavioral deficits that are partially alleviated by clinically administered analgesics, mirroring the postoperative experience of TKA patients., Competing Interests: The authors have declared that no competing interests exist., (Copyright: © 2022 Komatsu et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2022
- Full Text
- View/download PDF
16. SP and KLF Transcription Factors in Cancer Metabolism.
- Author
-
Orzechowska-Licari EJ, LaComb JF, Mojumdar A, and Bialkowska AB
- Subjects
- Cell Transformation, Neoplastic, Humans, Zinc Fingers, Kruppel-Like Transcription Factors metabolism, Neoplasms metabolism, Sp Transcription Factors metabolism
- Abstract
Tumor development and progression depend on reprogramming of signaling pathways that regulate cell metabolism. Alterations to various metabolic pathways such as glycolysis, oxidative phosphorylation, lipid metabolism, and hexosamine biosynthesis pathway are crucial to sustain increased redox, bioenergetic, and biosynthesis demands of a tumor cell. Transcription factors (oncogenes and tumor suppressors) play crucial roles in modulating these alterations, and their functions are tethered to major metabolic pathways under homeostatic conditions and disease initiation and advancement. Specificity proteins (SPs) and Krüppel-like factors (KLFs) are closely related transcription factors characterized by three highly conserved zinc fingers domains that interact with DNA. Studies have demonstrated that SP and KLF transcription factors are expressed in various tissues and regulate diverse processes such as proliferation, differentiation, apoptosis, inflammation, and tumorigenesis. This review highlights the role of SP and KLF transcription factors in the metabolism of various cancers and their impact on tumorigenesis. A better understanding of the role and underlying mechanisms governing the metabolic changes during tumorigenesis could provide new therapeutic opportunities for cancer treatment.
- Published
- 2022
- Full Text
- View/download PDF
17. Deep learning-based approach to the characterization and quantification of histopathology in mouse models of colitis.
- Author
-
Kobayashi S, Shieh J, Ruiz de Sabando A, Kim J, Liu Y, Zee SY, Prasanna P, Bialkowska AB, Saltz JH, and Yang VW
- Subjects
- Animals, Colon pathology, Dextran Sulfate toxicity, Disease Models, Animal, Humans, Mice, Mice, Inbred C57BL, Colitis, Deep Learning
- Abstract
Inflammatory bowel disease (IBD) is a chronic immune-mediated disease of the gastrointestinal tract. While therapies exist, response can be limited within the patient population. Researchers have thus studied mouse models of colitis to further understand pathogenesis and identify new treatment targets. Flow cytometry and RNA-sequencing can phenotype immune populations with single-cell resolution but provide no spatial context. Spatial context may be particularly important in colitis mouse models, due to the simultaneous presence of colonic regions that are involved or uninvolved with disease. These regions can be identified on hematoxylin and eosin (H&E)-stained colonic tissue slides based on the presence of abnormal or normal histology. However, detection of such regions requires expert interpretation by pathologists. This can be a tedious process that may be difficult to perform consistently across experiments. To this end, we trained a deep learning model to detect 'Involved' and 'Uninvolved' regions from H&E-stained colonic tissue slides. Our model was trained on specimens from controls and three mouse models of colitis-the dextran sodium sulfate (DSS) chemical induction model, the recently established intestinal epithelium-specific, inducible Klf5ΔIND (Villin-CreERT2;Klf5fl/fl) genetic model, and one that combines both induction methods. Image patches predicted to be 'Involved' and 'Uninvolved' were extracted across mice to cluster and identify histological classes. We quantified the proportion of 'Uninvolved' patches and 'Involved' patch classes in murine swiss-rolled colons. Furthermore, we trained linear determinant analysis classifiers on these patch proportions to predict mouse model and clinical score bins in a prospectively treated cohort of mice. Such a pipeline has the potential to reveal histological links and improve synergy between various colitis mouse model studies to identify new therapeutic targets and pathophysiological mechanisms., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2022
- Full Text
- View/download PDF
18. Intestinal Epithelial Regeneration in Response to Ionizing Irradiation.
- Author
-
Orzechowska-Licari EJ, LaComb JF, Giarrizzo M, Yang VW, and Bialkowska AB
- Subjects
- Animals, Cell Differentiation, Cell Division, Enterocytes, Mice, Intestinal Mucosa metabolism, Stem Cells
- Abstract
The intestinal epithelium consists of a single layer of cells yet contains multiple types of terminally differentiated cells, which are generated by the active proliferation of intestinal stem cells located at the bottom of intestinal crypts. However, during events of acute intestinal injury, these active intestinal stem cells undergo cell death. Gamma irradiation is a widely used colorectal cancer treatment, which, while therapeutically efficacious, has the side effect of depleting the active stem cell pool. Indeed, patients frequently experience gastrointestinal radiation syndrome while undergoing radiotherapy, in part due to active stem cell depletion. The loss of active intestinal stem cells in intestinal crypts activates a pool of typically quiescent reserve intestinal stem cells and induces dedifferentiation of secretory and enterocyte precursor cells. If not for these cells, the intestinal epithelium would lack the ability to recover from radiotherapy and other such major tissue insults. New advances in lineage-tracing technologies allow tracking of the activation, differentiation, and migration of cells during regeneration and have been successfully employed for studying this in the gut. This study aims to depict a method for the analysis of cells within the mouse intestinal epithelium following radiation injury.
- Published
- 2022
- Full Text
- View/download PDF
19. KLF5 protects the intestinal epithelium against Th17 immune response in a murine colitis model.
- Author
-
Shieh J, Chu TH, Liu Y, Kim J, Ruiz de Sabando A, Kobayashi S, Zee SY, Sheridan BS, Bialkowska AB, and Yang VW
- Subjects
- Adaptive Immunity, Animals, Intestinal Mucosa pathology, Mice, Mice, Inbred C57BL, Colitis, Inflammatory Bowel Diseases pathology, Kruppel-Like Transcription Factors genetics
- Abstract
Inflammatory bowel disease (IBD) is a chronic illness characterized by dysregulated immune cascades in the intestines, in which the Th17 immune response plays an important role. We demonstrated that mice with intestinal epithelium-specific deletion of Krüppel-like factor 5 (Klf5) developed Th17-dependent colonic inflammation. In the absence of KLF5, there was aberrant cellular localization of phosphorylated STAT3, an essential mediator of the Th17-associated cytokine, IL-22, which is required for epithelial tissue regeneration. In contrast, mitigation of IL-17A with anti-IL-17A neutralizing antibody attenuated colitis in Klf5-deficient mice. There was also a considerable shift in the colonic microbiota of Klf5-deficient mice that phenocopied human IBD. Notably, the inflammatory response due to Klf5 deletion was alleviated by antibiotic treatment, implicating the role of microbiota in pathogenesis. Finally, human colitic tissues had reduced KLF5 levels when compared with healthy tissues. Together, these findings demonstrated the importance of KLF5 in protecting the intestinal epithelium against Th17-mediated immune and inflammatory responses. The mice described herein may serve as a potential model for human IBD.
- Published
- 2022
- Full Text
- View/download PDF
20. IL-17RA-signaling in Lgr5 + intestinal stem cells induces expression of transcription factor ATOH1 to promote secretory cell lineage commitment.
- Author
-
Lin X, Gaudino SJ, Jang KK, Bahadur T, Singh A, Banerjee A, Beaupre M, Chu T, Wong HT, Kim CK, Kempen C, Axelrad J, Huang H, Khalid S, Shah V, Eskiocak O, Parks OB, Berisha A, McAleer JP, Good M, Hoshino M, Blumberg R, Bialkowska AB, Gaffen SL, Kolls JK, Yang VW, Beyaz S, Cadwell K, and Kumar P
- Subjects
- 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, Basic Helix-Loop-Helix Transcription Factors metabolism, Intestinal Mucosa cytology, Receptors, G-Protein-Coupled metabolism, Receptors, Interleukin-17 metabolism, Stem Cells metabolism
- 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., Competing Interests: Declaration of interests K.C. receives research funding from Pfizer, Takeda, and Abbvie; has consulted for or received an honorarium from Puretech Health, Genentech, and Abbvie; and holds U.S. patent 10,722,600 and provisional patent 62/935,035. S.L.G. consults for Aclaris Therapeutics and Eli Lilly., (Copyright © 2021 Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
21. Podocyte-specific KLF4 is required to maintain parietal epithelial cell quiescence in the kidney.
- Author
-
Pace JA, Bronstein R, Guo Y, Yang Y, Estrada CC, Gujarati N, Salant DJ, Haley J, Bialkowska AB, Yang VW, He JC, and Mallipattu SK
- Abstract
Podocyte loss triggering aberrant activation and proliferation of parietal epithelial cells (PECs) is a central pathogenic event in proliferative glomerulopathies. Podocyte-specific Krüppel-like factor 4 (KLF4), a zinc-finger transcription factor, is essential for maintaining podocyte homeostasis and PEC quiescence. Using mice with podocyte-specific knockdown of Klf4, we conducted glomerular RNA-sequencing, tandem mass spectrometry, and single-nucleus RNA-sequencing to identify cell-specific transcriptional changes that trigger PEC activation due to podocyte loss. Integration with in silico chromatin immunoprecipitation identified key ligand-receptor interactions, such as fibronectin 1 (FN1)–αVβ6, between podocytes and PECs dependent on KLF4 and downstream signal transducer and activator of transcription 3 (STAT3) signaling. Knockdown of Itgb6 in PECs attenuated PEC activation. Additionally, podocyte-specific induction of human KLF4 or pharmacological inhibition of downstream STAT3 activation reduced FN1 and integrin β 6 (ITGB6) expression and mitigated podocyte loss and PEC activation in mice. Targeting podocyte-PEC crosstalk might be a critical therapeutic strategy in proliferative glomerulopathies.
- Published
- 2021
- Full Text
- View/download PDF
22. Cardiomyocyte Krüppel-Like Factor 5 Promotes De Novo Ceramide Biosynthesis and Contributes to Eccentric Remodeling in Ischemic Cardiomyopathy.
- Author
-
Hoffman M, Palioura D, Kyriazis ID, Cimini M, Badolia R, Rajan S, Gao E, Nikolaidis N, Schulze PC, Goldberg IJ, Kishore R, Yang VW, Bannister TD, Bialkowska AB, Selzman CH, Drakos SG, and Drosatos K
- Subjects
- Animals, Disease Models, Animal, Humans, Male, Mice, Cardiomyopathies physiopathology, Ceramides metabolism, Kruppel-Like Transcription Factors metabolism, Myocytes, Cardiac metabolism, Ventricular Remodeling physiology
- 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.- Published
- 2021
- Full Text
- View/download PDF
23. IL-22 receptor signaling in Paneth cells is critical for their maturation, microbiota colonization, Th17-related immune responses, and anti-Salmonella immunity.
- Author
-
Gaudino SJ, Beaupre M, Lin X, Joshi P, Rathi S, McLaughlin PA, Kempen C, Mehta N, Eskiocak O, Yueh B, Blumberg RS, van der Velden AWM, Shroyer KR, Bialkowska AB, Beyaz S, and Kumar P
- Subjects
- Animals, Cell Differentiation, Immunity, Mucosal, Interleukins genetics, Lymphocyte Activation, Mice, Mice, Inbred C57BL, Mice, Knockout, Paneth Cells pathology, Receptors, Interleukin genetics, Signal Transduction, Interleukin-22, Interleukins metabolism, Microbiota physiology, Paneth Cells metabolism, Receptors, Interleukin metabolism, Salmonella typhi physiology, Th17 Cells immunology, Typhoid Fever immunology
- Abstract
Interleukin-22 (IL-22) signaling in the intestines is critical for promoting tissue-protective functions. However, since a diverse array of cell types (absorptive and secretory epithelium as well as stem cells) express IL-22Ra1, a receptor for IL-22, it has been difficult to determine what cell type(s) specifically respond to IL-22 to mediate intestinal mucosal host defense. Here, we report that IL-22 signaling in the small intestine is positively correlated with Paneth cell differentiation programs. Our Il22Ra1
fl/fl ;Lgr5-EGFP-creERT2 -specific knockout mice and, independently, our lineage-tracing findings rule out the involvement of Lgr5+ intestinal stem cell (ISC)-dependent IL-22Ra1 signaling in regulating the lineage commitment of epithelial cells, including Paneth cells. Using novel Paneth cell-specific IL-22Ra1 knockout mice (Il22Ra1fl/fl ;Defa6-cre), we show that IL-22 signaling in Paneth cells is required for small intestinal host defense. We show that Paneth cell maturation, antimicrobial effector function, expression of specific WNTs, and organoid morphogenesis are dependent on cell-intrinsic IL-22Ra1 signaling. Furthermore, IL-22 signaling in Paneth cells regulates the intestinal commensal bacteria and microbiota-dependent IL-17A immune responses. Finally, we show ISC and, independently, Paneth cell-specific IL-22Ra1 signaling are critical for providing immunity against Salmonella enterica serovar Typhimurium. Collectively, our findings illustrate a previously unknown role of IL-22 in Paneth cell-mediated small intestinal host defense.- Published
- 2021
- Full Text
- View/download PDF
24. KLF5 Is Induced by FOXO1 and Causes Oxidative Stress and Diabetic Cardiomyopathy.
- Author
-
Kyriazis ID, Hoffman M, Gaignebet L, Lucchese AM, Markopoulou E, Palioura D, Wang C, Bannister TD, Christofidou-Solomidou M, Oka SI, Sadoshima J, Koch WJ, Goldberg IJ, Yang VW, Bialkowska AB, Kararigas G, and Drosatos K
- Subjects
- 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, Mice, Diabetic Cardiomyopathies metabolism, Forkhead Box Protein O1 metabolism, Kruppel-Like Transcription Factors metabolism, Myocytes, Cardiac metabolism, Oxidative Stress, PPAR alpha metabolism
- 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.
- Published
- 2021
- Full Text
- View/download PDF
25. KLF4 Regulates Goblet Cell Differentiation in BMI1 + Reserve Intestinal Stem Cell Lineage during Homeostasis.
- Author
-
Katano T, Bialkowska AB, and Yang VW
- Abstract
Krüppel-like factor 4 (KLF4) is a zinc-finger transcription factor, expressed in villus cells of the intestinal epithelium, that promotes cellular differentiation and tissue homeostasis. Previous studies suggest that BMI1
+ cells represent secretory progenitors with reserve intestinal stem cell (rISC) activity. However, it has not been elucidated how KLF4 contributes to crypt regeneration originated from BMI1+ rISC lineage during homeostasis. In this study, Bmi1 -CreER ;Rosa26eYFP ( Bmi1 Ctrl) and Bmi1 -CreER ;Rosa26eYFP ; Klf4fl/fl ( Bmi1ΔKlf4 ) mice were injected with tamoxifen to label BMI1+ cells and their lineage and to delete Klf4 . During homeostasis, MUC2+ goblet cells appeared in the BMI1+ cell lineage 2, 3 and 7 days after tamoxifen administration. After Klf4 deletion in BMI1+ cells, the number of KLF4+ and MUC2+ cells ineYFP + cells decreased in Bmi1ΔKlf4 mice compared with Bmi1 Ctrl mice. Thus, KLF4 was positively correlated with goblet cell differentiation in BMI1+ cell derived lineage. In ex-vivo analysis, organoids derived from singleeYFP + cells of Bmi1 Ctrl mice contained MUC2-expressing cells that co-expressed KLF4. On the other hand, organoids derived from Klf4-deletedeYFP + cells from Bmi1ΔKlf4 mice showed reduced number of MUC2-expressing cells. In conclusion, these results suggest that KLF4 regulates goblet cell differentiation in BMI1+ ISC-derived lineage during homeostasis.- Published
- 2020
- Full Text
- View/download PDF
26. Interplay among p21 Waf1/Cip1 , MUSASHI-1 and Krüppel-like factor 4 in activation of Bmi1-Cre ER reserve intestinal stem cells after gamma radiation-induced injury.
- Author
-
Orzechowska EJ, Katano T, Bialkowska AB, and Yang VW
- Subjects
- Animals, HEK293 Cells, Humans, Intestinal Mucosa cytology, Intestinal Mucosa metabolism, Kruppel-Like Factor 4, Mice, Polymerase Chain Reaction, Stem Cells metabolism, Cyclin-Dependent Kinase Inhibitor p21 metabolism, Gamma Rays adverse effects, Intestinal Mucosa radiation effects, Kruppel-Like Transcription Factors metabolism, Nerve Tissue Proteins metabolism, Polycomb Repressive Complex 1 metabolism, Proto-Oncogene Proteins metabolism, RNA-Binding Proteins metabolism, Radiation Injuries, Experimental metabolism, Stem Cells radiation effects
- Abstract
Gamma radiation is a commonly used adjuvant treatment for abdominally localized cancer. Since its therapeutic potential is limited due to gastrointestinal (GI) syndrome, elucidation of the regenerative response following radiation-induced gut injury is needed to develop a preventive treatment. Previously, we showed that Krüppel-like factor 4 (KLF4) activates certain quiescent intestinal stem cells (ISCs) marked by Bmi1-Cre
ER to give rise to regenerating crypts following γ irradiation. In the current study, we showed that γ radiation-induced expression of p21Waf1/Cip1 in Bmi1-CreER cells is likely mitigated by MUSASHI-1 (MSI1) acting as a negative regulator of p21Waf1/Cip1 mRNA translation, which promotes exit of the Bmi1-CreER cells from a quiescent state. Additionally, Bmi1-specific Klf4 deletion resulted in decreased numbers of MSI1+ cells in regenerating crypts compared to those of control mice. We showed that KLF4 binds to the Msi1 promoter and activates its expression in vitro. Since MSI1 has been shown to be crucial for crypt regeneration, this finding elucidates a pro-proliferative role of KLF4 during the postirradiation regenerative response. Taken together, our data suggest that the interplay among p21Waf1/Cip1 , MSI1 and KLF4 regulates Bmi1-CreER cell survival, exit from quiescence and regenerative potential upon γ radiation-induced injury.- Published
- 2020
- Full Text
- View/download PDF
27. Krüppel-like Factor 5 Regulates Stemness, Lineage Specification, and Regeneration of Intestinal Epithelial Stem Cells.
- Author
-
Kim CK, Saxena M, Maharjan K, Song JJ, Shroyer KR, Bialkowska AB, Shivdasani RA, and Yang VW
- Subjects
- Adult Stem Cells radiation effects, Animals, Case-Control Studies, Cell Lineage genetics, Cell Self Renewal genetics, Cells, Cultured, Colitis etiology, Colitis pathology, Colorectal Neoplasms pathology, Colorectal Neoplasms therapy, Disease Models, Animal, Enteritis etiology, Enteritis pathology, Epigenesis, Genetic, Female, Humans, Intestinal Mucosa cytology, Intestinal Mucosa radiation effects, Kruppel-Like Transcription Factors analysis, Kruppel-Like Transcription Factors genetics, Male, Mice, Mice, Transgenic, Organoids, Primary Cell Culture, RNA-Seq, Receptors, G-Protein-Coupled genetics, Transcriptional Activation, Whole-Body Irradiation, Wnt Signaling Pathway genetics, Adult Stem Cells physiology, Intestinal Mucosa physiology, Kruppel-Like Transcription Factors metabolism, Radiation Injuries pathology, Regeneration genetics
- Abstract
Background & Aims: Self-renewal and multipotent differentiation are cardinal properties of intestinal stem cells (ISCs), mediated in part by WNT and NOTCH signaling. Although these pathways are well characterized, the molecular mechanisms that control the 'stemness' of ISCs are still not well defined. Here, we investigated the role of Krüppel-like factor 5 (KLF5) in regulating ISC functions., Methods: We performed studies in adult Lgr5
EGFP-IRES-creERT2 ;Rosa26LSLtdTomato (Lgr5Ctrl ) and Lgr5EGFP-IRES-creERT2 ;Klf5fl/fl ;Rosa26LSLtdTomato (Lgr5ΔKlf5 ) mice. Mice were injected with tamoxifen to activate Cre recombinase, which deletes Klf5 from the intestinal epithelium in Lgr5ΔKlf5 but not Lgr5Crtl mice. In experiments involving irradiation, mice were subjected to 12 Gy total body irradiation (TBI). Tissues were collected for immunofluorescence (IF) analysis and next generation sequencing. Oganoids were derived from fluoresecence activated cell sorted- (FACS-) single cells from tamoxifen-treated Lgr5ΔKlf5 or Lgr5Crtl mice and examined by immunofluorescence stain., Results: Lgr5+ ISCs lacking KLF5 proliferate faster than control ISCs but fail to self-renew, resulting in a depleted ISC compartment. Transcriptome analysis revealed that Klf5-null Lgr5+ cells lose ISC identity and prematurely differentiate. Following irradiation injury, which depletes Lgr5+ ISCs, reserve Klf5-null progenitor cells fail to dedifferentiate and regenerate the epithelium. Absence of KLF5 inactivates numerous selected enhancer elements and direct transcriptional targets including canonical WNT- and NOTCH-responsive genes. Analysis of human intestinal tissues showed increased levels of KLF5 in the regenerating epithelium as compared to those of healthy controls., Conclusion: We conclude that ISC self-renewal, lineage specification, and precursor dedifferentiation require KLF5, by its ability to regulate epigenetic and transcriptional activities of ISC-specific gene sets. These findings have the potential for modulating ISC functions by targeting KLF5 in the intestinal epithelium., (Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
28. Oncogenic KRAS Reduces Expression of FGF21 in Acinar Cells to Promote Pancreatic Tumorigenesis in Mice on a High-Fat Diet.
- Author
-
Luo Y, Yang Y, Liu M, Wang D, Wang F, Bi Y, Ji J, Li S, Liu Y, Chen R, Huang H, Wang X, Swidnicka-Siergiejko AK, Janowitz T, Beyaz S, Wang G, Xu S, Bialkowska AB, Luo CK, Pin CL, Liang G, Lu X, Wu M, Shroyer KR, Wolff RA, Plunkett W, Ji B, Li Z, Li E, Li X, Yang VW, Logsdon CD, Abbruzzese JL, and Lu W
- Subjects
- 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, 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
- 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 (Kras
G12D/+ 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., (Copyright © 2019 AGA Institute. Published by Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
29. The Novel Small-Molecule SR18662 Efficiently Inhibits the Growth of Colorectal Cancer In Vitro and In Vivo .
- Author
-
Kim J, Wang C, de Sabando AR, Cole HL, Huang TJ, Yang J, Bannister TD, Yang VW, and Bialkowska AB
- Subjects
- Acrylamides chemistry, Acrylamides pharmacology, Animals, Cell Line, Tumor, Cell Proliferation drug effects, Colorectal Neoplasms metabolism, Cyclic S-Oxides chemistry, Cyclic S-Oxides pharmacology, Cyclins metabolism, Dose-Response Relationship, Drug, Gene Expression Regulation, Neoplastic drug effects, HCT116 Cells, HT29 Cells, Humans, Mice, Small Molecule Libraries chemistry, Small Molecule Libraries pharmacology, Wnt Signaling Pathway drug effects, Xenograft Model Antitumor Assays, Acrylamides administration & dosage, Colorectal Neoplasms drug therapy, Cyclic S-Oxides administration & dosage, Kruppel-Like Transcription Factors metabolism, Small Molecule Libraries administration & dosage
- Abstract
Krüppel-like factor 5 (KLF5), a member of the SP/KLF family of zinc finger transcription factors, is overexpressed in human colorectal cancer specimens, and this overabundance is associated with aggressive cancer development and progression. We demonstrated that mice haploinsufficient for Klf5 had reduced intestinal tumor burden in the background of germline mutation in Apc , a gatekeeper of intestinal tumorigenesis. Based on a high-throughput screening strategy, we developed ML264, a small-molecule compound that inhibits KLF5, and showed that it inhibits growth of colorectal cancer in vitro and in vivo Through optimization efforts based on the structure of ML264, we have now identified a new lead compound, SR18662. We find that treatment with SR18662 significantly reduces growth and proliferation of colorectal cancer cells as compared with treatment with vehicle control, ML264, or SR15006 (a less optimized analogue from SAR efforts leading to SR18662). SR18662 showed improved efficacy in reducing the viability of multiple colorectal cancer cell lines. Flow cytometry analysis following SR18662 treatment showed an increase in cells captured in either S or G
2 -M phases of the cell cycle and a significant increase in the number of apoptotic cells, the latter a unique property compared with ML264 or SR15006. SR18662 treatment also reduces the expression of cyclins and components of the MAPK and WNT signaling pathways. Importantly, we observed a significant dose-dependent inhibition of xenograft growth in mice following SR18662 treatment that exceeded the effect of ML264 at equivalent doses. These findings support further development of SR18662 and its analogues for colorectal cancer therapy., (©2019 American Association for Cancer Research.)- Published
- 2019
- Full Text
- View/download PDF
30. Increased Genetic Instability and Accelerated Progression of Colitis-Associated Colorectal Cancer through Intestinal Epithelium-specific Deletion of Klf4 .
- Author
-
Yang VW, Liu Y, Kim J, Shroyer KR, and Bialkowska AB
- Subjects
- Animals, Carcinogenesis, Cell Proliferation physiology, Centrosome metabolism, Colitis pathology, Colorectal Neoplasms pathology, Gene Deletion, Intestinal Mucosa pathology, Kruppel-Like Factor 4, Kruppel-Like Transcription Factors genetics, Kruppel-Like Transcription Factors metabolism, MAP Kinase Signaling System, Mice, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Colitis genetics, Colitis metabolism, Colorectal Neoplasms genetics, Colorectal Neoplasms metabolism, Intestinal Mucosa metabolism, Kruppel-Like Transcription Factors deficiency
- Abstract
Krüppel-like factor 4 (KLF4), a zinc finger transcription factor, regulates homeostasis of the intestinal epithelium. Previously, it was reported that KLF4 functions as a tumor suppressor in colorectal cancer. Here, evidence demonstrates that KLF4 mitigates the development and progression of colitis-associated colorectal cancer (CAC) in a murine model. Mice with intestinal epithelium-specific deletion of Klf4 ( Klf4
ΔIS ) and control mice ( Klf4fl/fl ) were used to explore the role of KLF4 in the development of azoxymethane (AOM) and dextran sodium sulfate (DSS)-induced CAC. Upon AOM and DSS treatment, KLF4 expression was progressively lost in colonic tissues of Klf4fl/fl mice during tumor development. Klf4ΔIS mice treated with AOM/DSS developed significantly more adenomatous polyps and carcinomas in situ in comparison with treated Klf4fl/fl mice. Adenomatous polyps, but not normal-appearing mucosa, from colonic tissues of treated Klf4ΔIS mice contained a significantly increased number of mitotic cells with more than 2 centrosomes relative to treated control mice. KLF4 and p53 colocalize to the centrosomes in mouse embryonic fibroblasts (MEF). Absence of KLF4 in Klf4-/- MEFs inhibits and its overexpression restores p53 localization to the centrosomes in Klf4-/- MEFs. IMPLICATIONS: Taken together, these results indicate that KLF4 plays a protective role against progression of CAC by guarding against genetic instability., (©2018 American Association for Cancer Research.)- Published
- 2019
- Full Text
- View/download PDF
31. Loss of the Krüppel-like factor 4 tumor suppressor is associated with epithelial-mesenchymal transition in colorectal cancer.
- Author
-
Agbo KC, Huang JZ, Ghaleb AM, Williams JL, Shroyer KR, Bialkowska AB, and Yang VW
- Abstract
Aim: Colorectal cancer (CRC) is the third leading cancer-related cause of death due to its propensity to metastasize. Epithelial-mesenchymal transition (EMT) is a multistep process important for invasion and metastasis of CRC. Krüppel-like factor 4 (KLF4) is a zinc finger transcription factor highly expressed in differentiated cells of the intestinal epithelium. KLF4 has been shown to play a tumor suppressor role during CRC tumorigenesis - its loss accelerates development and progression of cancer. The present study examined the relationship between KLF4 and markers of EMT in CRC., Methods: Immunofluorescence staining for KLF4 and EMT markers was performed on archived patient samples after colorectal cancer resection and on colonic tissues of mice with colitis-associated cancer., Results: We found that KLF4 expression is lost in tumor sections obtained from CRC patients and in those of mouse colon following azoxymethane and dextran sodium sulfate (AOM/DSS) treatment when compared to their respective normal appearing mucosa. Importantly, in CRC patient tumor sections, we observed a negative correlation between KLF4 levels and mesenchymal markers including TWIST, β-catenin, claudin-1, N-cadherin, and vimentin. Similarly, in tumor tissues from AOM/DSS-treated mice, KLF4 levels were negatively correlated with mesenchymal markers including SNAI2, β-catenin, and vimentin and positively correlated with the epithelial marker E-cadherin., Conclusion: These findings suggest that the loss of KLF4 expression is a potentially significant indicator of EMT in CRC., Competing Interests: Conflicts of interest All authors declared that there are no conflicts of interest.
- Published
- 2019
- Full Text
- View/download PDF
32. Krüppel-like factor 4 is a negative regulator of STAT3-induced glomerular epithelial cell proliferation.
- Author
-
Estrada CC, Paladugu P, Guo Y, Pace J, Revelo MP, Salant DJ, Shankland SJ, D'Agati VD, Mehrotra A, Cardona S, Bialkowska AB, Yang VW, He JC, and Mallipattu SK
- Subjects
- Animals, Cell Cycle, Cell Proliferation, Disease Models, Animal, Epithelial Cells pathology, Glomerulosclerosis, Focal Segmental, Humans, Interleukin-6, Intracellular Signaling Peptides and Proteins metabolism, Kidney Glomerulus pathology, Kruppel-Like Factor 4, Kruppel-Like Transcription Factors genetics, Membrane Proteins metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Nephritis blood, Podocytes pathology, Transcription Factors, Epithelial Cells metabolism, Kidney Glomerulus metabolism, Kruppel-Like Transcription Factors metabolism, STAT3 Transcription Factor metabolism
- Abstract
Pathologic glomerular epithelial cell (GEC) hyperplasia is characteristic of both rapidly progressive glomerulonephritis (RPGN) and subtypes of focal segmental glomerulosclerosis (FSGS). Although initial podocyte injury resulting in activation of STAT3 signals GEC proliferation in both diseases, mechanisms regulating this are unknown. Here, we show that the loss of Krüppel-like factor 4 (KLF4), a zinc-finger transcription factor, enhances GEC proliferation in both RPGN and FSGS due to dysregulated STAT3 signaling. We observed that podocyte-specific knockdown of Klf4 (C57BL/6J) increased STAT3 signaling and exacerbated crescent formation after nephrotoxic serum treatment. Interestingly, podocyte-specific knockdown of Klf4 in the FVB/N background alone was sufficient to activate STAT3 signaling, resulting in FSGS with extracapillary proliferation, as well as renal failure and reduced survival. In cultured podocytes, loss of KLF4 resulted in STAT3 activation and cell-cycle reentry, leading to mitotic catastrophe. This triggered IL-6 release into the supernatant, which activated STAT3 signaling in parietal epithelial cells. Conversely, either restoration of KLF4 expression or inhibition of STAT3 signaling improved survival in KLF4-knockdown podocytes. Finally, human kidney biopsy specimens with RPGN exhibited reduced KLF4 expression with a concomitant increase in phospho-STAT3 expression as compared with controls. Collectively, these results suggest the essential role of KLF4/STAT3 signaling in podocyte injury and its regulation of aberrant GEC proliferation.
- Published
- 2018
- Full Text
- View/download PDF
33. Krüppel-like Factor 5, Increased in Pancreatic Ductal Adenocarcinoma, Promotes Proliferation, Acinar-to-Ductal Metaplasia, Pancreatic Intraepithelial Neoplasia, and Tumor Growth in Mice.
- Author
-
He P, Yang JW, Yang VW, and Bialkowska AB
- Subjects
- 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, Carcinoma in Situ metabolism, Carcinoma, Pancreatic Ductal metabolism, Cell Proliferation, Cell Transformation, Neoplastic metabolism, Kruppel-Like Transcription Factors metabolism, Pancreatic Neoplasms metabolism
- 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 (Klf5
fl/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., (Copyright © 2018 AGA Institute. Published by Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
34. KLF5 mediates the hyper-proliferative phenotype of the intestinal epithelium in mice with intestine-specific endogenous K-Ras G12D expression.
- Author
-
Nandan MO, Bialkowska AB, and Yang VW
- Abstract
Oncogenic K-Ras activation is a common mutational event in colorectal cancer. We previously showed that transcription factor, Krüppel-like factor 5 (KLF5), contributes to intestinal polyposis in mice with K-Ras activation. At 14 months of age, Villin-Cre/LSL-K-Ras
G12D mice developed small intestinal and colonic hyperplastic polyps while LSL-K-RasG12D had none. The intestinal crypts of Villin-Cre/LSL-K-RasG12D mice contained a higher number of mitotic figures and increased crypt heights compared to controls. The intestinal epithelium of Villin-Cre/LSL-K-RasG12D mice showed prolific KLF5 expression throughout and above the elongated crypts. In contrast, KLF5 expression was limited to the upper crypt region in the controls. The levels of K-Ras effectors were significantly increased in Villin-Cre/LSL-K-RasG12D as compared to controls. The Villin-Cre/LSL-K-RasG12D mice showed decreased survival upon treatment with azoxymethane (AOM) as compared to controls. Furthermore, loss of one of Klf5 alleles reduced levels of K-Ras effector proteins and prevented mortality of Villin-Cre/LSL-K-RasG12D mice upon AOM treatment. The Villin-Cre/LSL-K-RasG12D mice spontaneously develop hyperplastic intestinal polyps and display a hyper-proliferative intestinal phenotype with elongated crypts, increased numbers of mitotic figures, elevated expression of KLF5, and other pro-proliferative targets. Induction of colonic tumorigenesis with AOM is detrimental to Villin-Cre/LSL-K-RasG12D mice that is in part dependent of KLF5., Competing Interests: None.- Published
- 2018
35. Correction to: The Role of Intestinal Stem Cells in Epithelial Regeneration Following Radiation-Induced Gut Injury.
- Author
-
Kim CK, Yang VW, and Bialkowska AB
- Abstract
[This corrects the article DOI: 10.1007/s40778-017-0103-7.].
- Published
- 2018
- Full Text
- View/download PDF
36. The loss of Krüppel-like factor 15 in Foxd1 + stromal cells exacerbates kidney fibrosis.
- Author
-
Gu X, Mallipattu SK, Guo Y, Revelo MP, Pace J, Miller T, Gao X, Jain MK, Bialkowska AB, Yang VW, He JC, and Mei C
- Subjects
- 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, DNA-Binding Proteins metabolism, Forkhead Transcription Factors metabolism, Kidney pathology, Kidney Diseases pathology, Myofibroblasts pathology, Transcription Factors metabolism, Wnt Signaling Pathway
- 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 Klf15
fl/fl mice and accompanied by the activation of Wnt/β-catenin signaling. Furthermore, knockdown of Klf15 in cultured mouse embryonic fibroblasts activated canonical Wnt/β-catenin signaling, increased profibrotic transcripts, and increased proliferation after treatment with a Wnt1 ligand. Conversely, the overexpression of KLF15 inhibited phospho-β-catenin (Ser552) expression in Wnt1-treated cells. Thus, KLF15 has a critical role in attenuating kidney fibrosis by inhibiting the canonical Wnt/β-catenin pathway., (Copyright © 2017 International Society of Nephrology. Published by Elsevier Inc. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
37. Krüppel-like factor 5 is essential for maintenance of barrier function in mouse colon.
- Author
-
Liu Y, Chidgey M, Yang VW, and Bialkowska AB
- Subjects
- 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, Colon physiology, Kruppel-Like Transcription Factors physiology
- 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-CreER
T2 ;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., (Copyright © 2017 the American Physiological Society.)- Published
- 2017
- Full Text
- View/download PDF
38. SP and KLF Transcription Factors in Digestive Physiology and Diseases.
- Author
-
Kim CK, He P, Bialkowska AB, and Yang VW
- Subjects
- 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, Digestive System metabolism, Digestive System Diseases metabolism, Kruppel-Like Transcription Factors metabolism, Sp Transcription Factors metabolism
- 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., (Copyright © 2017 AGA Institute. Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
39. Novel sphingosine kinase-1 inhibitor, LCL351, reduces immune responses in murine DSS-induced colitis.
- Author
-
Pulkoski-Gross MJ, Uys JD, Orr-Gandy KA, Coant N, Bialkowska AB, Szulc ZM, Bai A, Bielawska A, Townsend DM, Hannun YA, Obeid LM, and Snider AJ
- Subjects
- A549 Cells, Chemokine CXCL1 genetics, Chemokine CXCL2 genetics, Colitis chemically induced, Colitis genetics, Gene Expression Regulation drug effects, Gene Expression Regulation immunology, Guanidines therapeutic use, Humans, Sphingosine therapeutic use, Tumor Necrosis Factor-alpha genetics, Colitis drug therapy, Colitis immunology, Dextran Sulfate adverse effects, Guanidines pharmacology, Phosphotransferases (Alcohol Group Acceptor) antagonists & inhibitors, Sphingosine pharmacology
- Abstract
Sphingosine-1-phosphate (S1P) is a biologically active sphingolipid metabolite which has been implicated in many diseases including cancer and inflammatory diseases. Recently, sphingosine kinase 1 (SK1), one of the isozymes which generates S1P, has been implicated in the development and progression of inflammatory bowel disease (IBD). Based on our previous work, we set out to determine the efficacy of a novel SK1 selective inhibitor, LCL351, in a murine model of IBD. LCL351 selectively inhibits SK1 both in vitro and in cells. LCL351, which accumulates in relevant tissues such as colon, did not have any adverse side effects in vivo. In mice challenged with dextran sodium sulfate (DSS), a murine model for IBD, LCL351 treatment protected from blood loss and splenomegaly. Additionally, LCL351 treatment reduced the expression of pro-inflammatory markers, and reduced neutrophil infiltration in colon tissue. Our results suggest inflammation associated with IBD can be targeted pharmacologically through the inhibition and degradation of SK1. Furthermore, our data also identifies desirable properties of SK1 inhibitors., (Published by Elsevier Inc.)
- Published
- 2017
- Full Text
- View/download PDF
40. Krüppel-like factors in mammalian stem cells and development.
- Author
-
Bialkowska AB, Yang VW, and Mallipattu SK
- Subjects
- Animals, Cell Differentiation, Cell Lineage, Female, Humans, Male, Mice, Mice, Transgenic, Phylogeny, Regeneration, Transcription Factors metabolism, Zinc Fingers, Embryonic Development, Kruppel-Like Transcription Factors metabolism, Lung embryology, Stem Cells cytology
- 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., (© 2017. Published by The Company of Biologists Ltd.)
- Published
- 2017
- Full Text
- View/download PDF
41. Epithelial derived-matrix metalloproteinase (MMP9) exhibits a novel defensive role of tumor suppressor in colitis associated cancer by activating MMP9-Notch1-ARF-p53 axis.
- Author
-
Walter L, Pujada A, Bhatnagar N, Bialkowska AB, Yang VW, Laroui H, and Garg P
- Subjects
- Animals, Apoptosis, Caspase 3 metabolism, Cell Proliferation, Colitis chemically induced, Colitis complications, Colon immunology, Colon metabolism, Colon pathology, Colonic Neoplasms etiology, Cyclin E metabolism, Cyclin-Dependent Kinase Inhibitor p16 metabolism, Cyclin-Dependent Kinase Inhibitor p21 metabolism, DNA Damage, Dextran Sulfate toxicity, Fibroblasts, HCT116 Cells, Humans, Intestinal Mucosa metabolism, Intestinal Mucosa pathology, Matrix Metalloproteinase 9 genetics, Mice, Mice, Inbred C57BL, Mice, Transgenic, Neoplasms, Experimental etiology, Neoplasms, Experimental pathology, Receptor, Notch1 metabolism, S Phase Cell Cycle Checkpoints, Signal Transduction, Transfection, Tumor Burden, Tumor Suppressor Protein p53 metabolism, Tumor Suppressor Proteins genetics, Colitis pathology, Colonic Neoplasms pathology, Intestinal Mucosa enzymology, Matrix Metalloproteinase 9 metabolism, Tumor Suppressor Proteins metabolism
- Abstract
Colitis associated cancer (CAC) is chronic inflammation driven colon cancer, prevalent among individuals with Inflammatory Bowel Disease. Matrix-metalloproteinase (MMP9) is one of the essential regulators of extra cellular matrix components. We have shown that MMP9 is protective in CAC contrary to its inflammatory role in acute-colitis. Aim of our study is to identify the mechanism of the protective role of epithelial derived-MMP9 in CAC. We used homozygous transgenic mice constitutively-expressing MMP9 in colonic-epithelium (TgM9) and wild-type (WT) littermates for in vivo experiments. Stably-transfected HCT116 with/without MMP9, and mouse embryonic-fibroblasts (WT and MMP9-/-, MEFs) were used for in vitro experiments. TgM9 mice exhibited less tumor burden, increased apoptosis, and increased expressions of active-Notch1, p53, p21WAF1/Cip1, caspase-3 and cyclin E in CAC compared to WTs. These results were supported by MEFs data. HCT116-cells overexpressing MMP9 indicated decreased cell proliferation, S-phase cell-cycle arrest and less DNA damage compared to vector. MMP9-/- mice showed attenuation of MMP9 was directly associated with p19ARF. Our study identifies the tumor suppressor role of epithelial derived-MMP9 in CAC via novel mechanistic pathway "MMP9-Notch1-ARF-p53 axis" regulating apoptosis, cell-cycle arrest and DNA damage implying, that MMP9 expression might be a natural/biological way to suppress colonic ulceration due to chronic inflammation.
- Published
- 2017
- Full Text
- View/download PDF
42. Krüppel-Like Factor 15 Mediates Glucocorticoid-Induced Restoration of Podocyte Differentiation Markers.
- Author
-
Mallipattu SK, Guo Y, Revelo MP, Roa-Peña L, Miller T, Ling J, Shankland SJ, Bialkowska AB, Ly V, Estrada C, Jain MK, Lu Y, Ma'ayan A, Mehrotra A, Yacoub R, Nord EP, Woroniecki RP, Yang VW, and He JC
- Subjects
- 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, Cell Differentiation drug effects, DNA-Binding Proteins physiology, Glucocorticoids pharmacology, Podocytes cytology, Podocytes drug effects, Transcription Factors physiology
- 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., (Copyright © 2016 by the American Society of Nephrology.)
- Published
- 2017
- Full Text
- View/download PDF
43. The Role of Intestinal Stem Cells in Epithelial Regeneration Following Radiation-Induced Gut Injury.
- Author
-
Kim CK, Yang VW, and Bialkowska AB
- Abstract
Purpose of Review: Intestinal epithelial cells show remarkable plasticity in regenerating the epithelium following radiation injury. In this review, we explore the regenerative capacity and mechanisms of various populations of intestinal stem cells (ISCs) in response to ionizing radiation., Recent Findings: Ionizing radiation targets mitotic cells that include "active" ISCs and progenitor cells. Lineage-tracing experiments showed that several different cell types identified by a single or combination of markers are capable of regenerating the epithelium, confirming that ISCs exhibit a high degree of plasticity. However, the identities of the contributing cells marked by various markers require further validation., Summary: Following radiation injury, quiescent and/or radioresistant cells become active stem cells to regenerate the epithelium. Looking forward, understanding the mechanisms by which ISCs govern tissue regeneration is crucial to determine therapeutic approaches to promote intestinal epithelial regeneration following injury., Competing Interests: Compliance with Ethical StandardsChang-Kyung Kim, Vincent W. Yang, and Agnieszka B. Bialkowska declare that they have no conflict of interest.This article does not contain any studies with human or animal subjects performed by any of the authors.
- Published
- 2017
- Full Text
- View/download PDF
44. Role of neutral ceramidase in colon cancer.
- Author
-
García-Barros M, Coant N, Kawamori T, Wada M, Snider AJ, Truman JP, Wu BX, Furuya H, Clarke CJ, Bialkowska AB, Ghaleb A, Yang VW, Obeid LM, and Hannun YA
- Subjects
- Animals, Colon metabolism, Humans, Male, Mice, Knockout, Sphingolipids metabolism, beta Catenin metabolism, Ceramides metabolism, Colonic Neoplasms enzymology, Lipid Metabolism physiology, Neutral Ceramidase metabolism
- 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 β-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., (© FASEB.)
- Published
- 2016
- Full Text
- View/download PDF
45. Intestinal stem cell resurgence by enterocyte precursors.
- Author
-
Kim CK, Bialkowska AB, and Yang VW
- Abstract
Competing Interests: The authors have no conflicts of interest to declare.
- Published
- 2016
- Full Text
- View/download PDF
46. Improved Swiss-rolling Technique for Intestinal Tissue Preparation for Immunohistochemical and Immunofluorescent Analyses.
- Author
-
Bialkowska AB, Ghaleb AM, Nandan MO, and Yang VW
- Subjects
- Animals, Colon, Mice, Fluorescent Antibody Technique methods, Histological Techniques methods, Immunohistochemistry methods, Intestines
- Abstract
Understanding the role of factors that regulate intestinal epithelial homeostasis and response to injury and regeneration is important. The current literature describes several different methodological approaches to obtain images of intestinal tissues for data validation. In this paper, we delineate a common protocol relating to the derivation and processing of mouse intestinal tissues. Proper fixation of intestinal tissues and Swiss-roll techniques that enhance intestinal epithelial morphology are discussed. Postresection processing and reorientation of embedded intestinal tissues are critical in obtaining paraffin-embedded blocks that display intact intestinal structural features after sectioning. The Swiss-rolling technique helps in histological assessment of the complete intestinal or colonic sections examined. An ability to differentiate intestinal structural features can be vital in quantitative measurements of intestinal inflammation and tumorigenesis along the entire length. Finally, paraffin-embedded sections are ideal for robust processing using both immunohistochemical and immunofluorescent detection methods. Nonfluorescent immunohistochemical sections provide a vibrant image of the tissue detailing different cellular structural features but do not provide flexibility for intracellular co-localization experiments. Multiple fluorescent channels can be appropriately utilized with immunofluorescent detection for co-localization experiments, lending support to mechanistic studies.
- Published
- 2016
- Full Text
- View/download PDF
47. Krüppel-like Factor 4 Modulates Development of BMI1(+) Intestinal Stem Cell-Derived Lineage Following γ-Radiation-Induced Gut Injury in Mice.
- Author
-
Kuruvilla JG, Kim CK, Ghaleb AM, Bialkowska AB, Kuo CJ, and Yang VW
- Subjects
- Animals, Apoptosis radiation effects, Bacterial Proteins genetics, Bacterial Proteins metabolism, Cell Count, Cell Proliferation radiation effects, Gamma Rays, Gene Deletion, Gene Expression Regulation, Genes, Reporter, Intestinal Mucosa metabolism, Intestines cytology, Kruppel-Like Factor 4, Kruppel-Like Transcription Factors deficiency, Luminescent Proteins genetics, Luminescent Proteins metabolism, Mice, Mice, Transgenic, Polycomb Repressive Complex 1 metabolism, Proto-Oncogene Proteins metabolism, Radiation Injuries genetics, Radiation Injuries metabolism, Radiation Injuries pathology, Regeneration genetics, Signal Transduction, Stem Cells cytology, Stem Cells metabolism, Intestines radiation effects, Kruppel-Like Transcription Factors genetics, Polycomb Repressive Complex 1 genetics, Proto-Oncogene Proteins genetics, Radiation Injuries rehabilitation, Stem Cells radiation effects
- Abstract
In response to ionizing radiation-induced injury, the normally quiescent intestinal stem cells marked by BMI1 participate in the regenerative response. Previously, we established a protective role for Krüppel-like factor 4 (KLF4) in the intestinal epithelium where it reduces senescence, apoptosis, and crypt atrophy following γ-radiation-induced gut injury. We also described a pro-proliferative function for KLF4 during the regenerative phase post irradiation. In the current study, using a mouse model in which Klf4 is deleted from quiescent BMI1(+) intestinal stem cells, we observed increased proliferation from the BMI1(+) lineage during homeostasis. In contrast, following irradiation, Bmi1-specific Klf4 deletion leads to decreased expansion of the BMI1(+) lineage due to a combination of reduced proliferation and increased apoptosis. Our results support a critical role for KLF4 in modulating BMI1(+) intestinal stem cell fate in both homeostasis and the regenerative response to radiation injury., (Copyright © 2016 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
48. KLF4 Suppresses Tumor Formation in Genetic and Pharmacological Mouse Models of Colonic Tumorigenesis.
- Author
-
Ghaleb AM, Elkarim EA, Bialkowska AB, and Yang VW
- Subjects
- Adenomatous Polyposis Coli Protein genetics, Animals, Cells, Cultured, Colonic Neoplasms chemically induced, Colonic Neoplasms genetics, Epigenesis, Genetic, Gene Deletion, Gene Expression Regulation, Neoplastic drug effects, HCT116 Cells, Humans, Intestinal Mucosa metabolism, Intestinal Mucosa pathology, Kruppel-Like Factor 4, Mice, Neoplasms, Experimental, TOR Serine-Threonine Kinases metabolism, Azoxymethane adverse effects, Colonic Neoplasms pathology, Kruppel-Like Transcription Factors genetics, Kruppel-Like Transcription Factors metabolism, Mutation
- Abstract
Unlabelled: The zinc finger transcription factor Krüppel-like factor 4 (KLF4) is frequently downregulated in colorectal cancer. Previous studies showed that KLF4 is a tumor suppressor in the intestinal tract and plays an important role in DNA damage-repair mechanisms. Here, the in vivo effects of Klf4 deletion were examined from the mouse intestinal epithelium (Klf4(ΔIS)) in a genetic or pharmacological setting of colonic tumorigenesis:Apc(Min/⁺) mutation or carcinogen treatment with azoxymethane (AOM), respectively.Klf4 (ΔIS)/Apc (Min/⁺) mice developed significantly more colonic adenomas with 100% penetrance as compared with Apc(Min/⁺) mice with intact Klf4 (Klf4(fl/fl)/Apc (Min/⁺)). The colonic epithelium of Klf4 (ΔIS)/Apc (Min/⁺)mice showed increased mTOR pathway activity, together with dysregulated epigenetic mechanism as indicated by altered expression of HDAC1 and p300. Colonic adenomas from both genotypes stained positive for γH2AX, indicating DNA double-strand breaks. InKlf4 (ΔIS)/Apc (Min/+) mice, this was associated with reduced nonhomologous end joining (NHEJ) repair and homologous recombination repair (HRR) mechanisms as indicated by reduced Ku70 and Rad51 staining, respectively. In a separate model, following treatment with AOM, Klf4 (ΔIS) mice developed significantly more colonic tumors than Klf4 (fl/fl) mice, with more Klf4 (ΔIS) mice harboring K-Rasmutations than Klf4 (fl/fl)mice. Compared with AOM-treated Klf4 (fl/fl)mice, adenomas of treated Klf4 (ΔIS) mice had suppressed NHEJ and HRR mechanisms, as indicated by reduced Ku70 and Rad51 staining. This study highlights the important role of KLF4 in suppressing the development of colonic neoplasia under different tumor-promoting conditions., Implications: The study demonstrates that KLF4 plays a significant role in the pathogenesis of colorectal neoplasia., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
49. Alkaline ceramidase 3 deficiency aggravates colitis and colitis-associated tumorigenesis in mice by hyperactivating the innate immune system.
- Author
-
Wang K, Xu R, Snider AJ, Schrandt J, Li Y, Bialkowska AB, Li M, Zhou J, Hannun YA, Obeid LM, Yang VW, and Mao C
- Subjects
- Alkaline Ceramidase deficiency, Animals, Cell Transformation, Neoplastic, Ceramides analysis, Ceramides metabolism, Colitis metabolism, Colitis pathology, Colon metabolism, Colon pathology, Cytokines metabolism, Dextran Sulfate toxicity, Disease Models, Animal, Female, Leukocytes, Mononuclear cytology, Leukocytes, Mononuclear immunology, Leukocytes, Mononuclear metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Substrate Specificity, Up-Regulation drug effects, Alkaline Ceramidase genetics, Colitis etiology, Immunity, Innate drug effects, Lipopolysaccharides toxicity
- Abstract
Increasing studies suggest that ceramides differing in acyl chain length and/or degree of unsaturation have distinct roles in mediating biological responses. However, still much remains unclear about regulation and role of distinct ceramide species in the immune response. Here, we demonstrate that alkaline ceramidase 3 (Acer3) mediates the immune response by regulating the levels of C18:1-ceramide in cells of the innate immune system and that Acer3 deficiency aggravates colitis in a murine model by augmenting the expression of pro-inflammatory cytokines in myeloid and colonic epithelial cells (CECs). According to the NCBI Gene Expression Omnibus (GEO) database, ACER3 is downregulated in immune cells in response to lipopolysaccharides (LPS), a potent inducer of the innate immune response. Consistent with these data, we demonstrated that LPS downregulated both Acer3 mRNA levels and its enzymatic activity while elevating C(18:1)-ceramide, a substrate of Acer3, in murine immune cells or CECs. Knocking out Acer3 enhanced the elevation of C(18:1)-ceramide and the expression of pro-inflammatory cytokines in immune cells and CECs in response to LPS challenge. Similar to Acer3 knockout, treatment with C(18:1)-ceramide, but not C18:0-ceramide, potentiated LPS-induced expression of pro-inflammatory cytokines in immune cells. In the mouse model of dextran sulfate sodium-induced colitis, Acer3 deficiency augmented colitis-associated elevation of colonic C(18:1)-ceramide and pro-inflammatory cytokines. Acer3 deficiency aggravated diarrhea, rectal bleeding, weight loss and mortality. Pathological analyses revealed that Acer3 deficiency augmented colonic shortening, immune cell infiltration, colonic epithelial damage and systemic inflammation. Acer3 deficiency also aggravated colonic dysplasia in a mouse model of colitis-associated colorectal cancer. Taken together, these results suggest that Acer3 has an important anti-inflammatory role by suppressing cellular or tissue C(18:1)-ceramide, a potent pro-inflammatory bioactive lipid and that dysregulation of ACER3 and C(18:1)-ceramide may contribute to the pathogenesis of inflammatory diseases including cancer.
- Published
- 2016
- Full Text
- View/download PDF
50. Murine Model for Colitis-Associated Cancer of the Colon.
- Author
-
Snider AJ, Bialkowska AB, Ghaleb AM, Yang VW, Obeid LM, and Hannun YA
- Subjects
- Animals, Colitis complications, Colonic Neoplasms pathology, Disease Models, Animal, Female, Humans, Male, Mice, Azoxymethane toxicity, Colitis chemically induced, Colonic Neoplasms chemically induced, Dextran Sulfate toxicity
- Abstract
Inflammatory bowel disease (IBD), including ulcerative colitis (UC) and Crohn's disease (CD), significantly increases the risk for development of colorectal cancer. Specifically, dysplasia and cancer associated with IBD (colitis-associated cancer or CAC) develop as a result of repeated cycles of injury and healing in the intestinal epithelium. Animal models are utilized to examine the mechanisms of CAC, the role of epithelial and immune cells in this process, as well as the development of novel therapeutic targets. These models typically begin with the administration of a carcinogenic compound, and inflammation is caused by repeated cycles of colitis-inducing agents. This review describes a common CAC model that utilizes the pro-carcinogenic compound azoxymethane (AOM) followed by dextran sulfate sodium (DSS) which induces the inflammatory insult.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.