Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Nat Immunol ; 25(5): 790-801, 2024 May.
Article in English | MEDLINE | ID: mdl-38664585

ABSTRACT

Innate immune cells generate a multifaceted antitumor immune response, including the conservation of essential nutrients such as iron. These cells can be modulated by commensal bacteria; however, identifying and understanding how this occurs is a challenge. Here we show that the food commensal Lactiplantibacillus plantarum IMB19 augments antitumor immunity in syngeneic and xenograft mouse tumor models. Its capsular heteropolysaccharide is the major effector molecule, functioning as a ligand for TLR2. In a two-pronged manner, it skews tumor-associated macrophages to a classically active phenotype, leading to generation of a sustained CD8+ T cell response, and triggers macrophage 'nutritional immunity' to deploy the high-affinity iron transporter lipocalin-2 for capturing and sequestering iron in the tumor microenvironment. This process induces a cycle of tumor cell death, epitope expansion and subsequent tumor clearance. Together these data indicate that food commensals might be identified and developed into 'oncobiotics' for a multi-layered approach to cancer therapy.


Subject(s)
Iron , Tumor Microenvironment , Animals , Iron/metabolism , Mice , Tumor Microenvironment/immunology , Humans , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 2/immunology , Mice, Inbred C57BL , Lipocalin-2/metabolism , Lipocalin-2/immunology , Female , Symbiosis/immunology , Macrophages/immunology , Macrophages/metabolism , Macrophage Activation/immunology , Mice, Knockout
2.
Nature ; 588(7839): 664-669, 2020 12.
Article in English | MEDLINE | ID: mdl-33328632

ABSTRACT

Current organoid models are limited by their inability to mimic mature organ architecture and associated tissue microenvironments1,2. Here we create multilayer bladder 'assembloids' by reconstituting tissue stem cells with stromal components to represent an organized architecture with an epithelium surrounding stroma and an outer muscle layer. These assembloids exhibit characteristics of mature adult bladders in cell composition and gene expression at the single-cell transcriptome level, and recapitulate in vivo tissue dynamics of regenerative responses to injury. We also develop malignant counterpart tumour assembloids to recapitulate the in vivo pathophysiological features of urothelial carcinoma. Using the genetically manipulated tumour-assembloid platform, we identify tumoural FOXA1, induced by stromal bone morphogenetic protein (BMP), as a master pioneer factor that drives enhancer reprogramming for the determination of tumour phenotype, suggesting the importance of the FOXA1-BMP-hedgehog signalling feedback axis between tumour and stroma in the control of tumour plasticity.


Subject(s)
Organoids/pathology , Organoids/physiology , Regeneration , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/physiopathology , Urinary Bladder/pathology , Urinary Bladder/physiology , Adult , Animals , Bone Morphogenetic Proteins/metabolism , Female , Hedgehogs/metabolism , Hepatocyte Nuclear Factor 3-alpha/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Organoids/physiopathology , Single-Cell Analysis , Stem Cells/cytology , Stem Cells/pathology , Stem Cells/physiology , Transcriptome , Urinary Bladder/cytology , Urinary Tract Infections/metabolism , Urinary Tract Infections/pathology
3.
Cell ; 141(4): 704-16, 2010 May 14.
Article in English | MEDLINE | ID: mdl-20478259

ABSTRACT

Mechanosensitive sensory hair cells are the linchpin of our senses of hearing and balance. The inability of the mammalian inner ear to regenerate lost hair cells is the major reason for the permanence of hearing loss and certain balance disorders. Here, we present a stepwise guidance protocol starting with mouse embryonic stem and induced pluripotent stem cells, which were directed toward becoming ectoderm capable of responding to otic-inducing growth factors. The resulting otic progenitor cells were subjected to varying differentiation conditions, one of which promoted the organization of the cells into epithelial clusters displaying hair cell-like cells with stereociliary bundles. Bundle-bearing cells in these clusters responded to mechanical stimulation with currents that were reminiscent of immature hair cell transduction currents.


Subject(s)
Embryonic Stem Cells/cytology , Hair Cells, Auditory/cytology , Hair Cells, Vestibular/cytology , Induced Pluripotent Stem Cells/cytology , Animals , Cell Culture Techniques , Cell Differentiation , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hair Cells, Auditory/physiology , Hair Cells, Auditory/ultrastructure , Hair Cells, Vestibular/physiology , Hair Cells, Vestibular/ultrastructure , Mechanotransduction, Cellular , Mice
4.
J Pineal Res ; 76(4): e12958, 2024 May.
Article in English | MEDLINE | ID: mdl-38747060

ABSTRACT

Endothelial-to-mesenchymal transition (EndMT) is a complex biological process of cellular transdifferentiation by which endothelial cells (ECs) lose their characteristics and acquire mesenchymal properties, leading to cardiovascular remodeling and complications in the adult cardiovascular diseases environment. Melatonin is involved in numerous physiological and pathological processes, including aging, and has anti-inflammatory and antioxidant activities. This molecule is an effective therapeutic candidate for preventing oxidative stress, regulating endothelial function, and maintaining the EndMT balance to provide cardiovascular protection. Although recent studies have documented improved cardiac function by melatonin, the mechanism of action of melatonin on EndMT remains unclear. The present study investigated the effects of melatonin on induced EndMT by transforming growth factor-ß2/interleukin-1ß in both in vivo and in vitro models. The results revealed that melatonin reduced the migratory ability and reactive oxygen species levels of the cells and ameliorated mitochondrial dysfunction in vitro. Our findings indicate that melatonin prevents endothelial dysfunction and inhibits EndMT by activating related pathways, including nuclear factor kappa B and Smad. We also demonstrated that this molecule plays a crucial role in restoring cardiac function by regulating the EndMT process in the ischemic myocardial condition, both in vessel organoids and myocardial infarction (MI) animal models. In conclusion, melatonin is a promising agent that attenuates EC dysfunction and ameliorates cardiac damage compromising the EndMT process after MI.


Subject(s)
Melatonin , NF-kappa B , Melatonin/pharmacology , Animals , NF-kappa B/metabolism , Epithelial-Mesenchymal Transition/drug effects , Humans , Signal Transduction/drug effects , Mice , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Male , Myocardial Infarction/metabolism , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , Reactive Oxygen Species/metabolism
5.
Proc Natl Acad Sci U S A ; 113(47): E7545-E7553, 2016 11 22.
Article in English | MEDLINE | ID: mdl-27815529

ABSTRACT

Inflammation disrupts tissue architecture and function, thereby contributing to the pathogenesis of diverse diseases; the signals that promote or restrict tissue inflammation thus represent potential targets for therapeutic intervention. Here, we report that genetic or pharmacologic Hedgehog pathway inhibition intensifies colon inflammation (colitis) in mice. Conversely, genetic augmentation of Hedgehog response and systemic small-molecule Hedgehog pathway activation potently ameliorate colitis and restrain initiation and progression of colitis-induced adenocarcinoma. Within the colon, the Hedgehog protein signal does not act directly on the epithelium itself, but on underlying stromal cells to induce expression of IL-10, an immune-modulatory cytokine long known to suppress inflammatory intestinal damage. IL-10 function is required for the full protective effect of small-molecule Hedgehog pathway activation in colitis; this pharmacologic augmentation of Hedgehog pathway activity and stromal IL-10 expression are associated with increased presence of CD4+Foxp3+ regulatory T cells. We thus identify stromal cells as cellular coordinators of colon inflammation and suggest their pharmacologic manipulation as a potential means to treat colitis.


Subject(s)
Colitis/metabolism , Dextran Sulfate/adverse effects , Hedgehog Proteins/metabolism , Interleukin-10/metabolism , Signal Transduction , Animals , CD4 Antigens/metabolism , Colitis/chemically induced , Colitis/drug therapy , Disease Models, Animal , Disease Progression , Forkhead Transcription Factors/metabolism , Hedgehog Proteins/drug effects , Humans , Mice , Mutation , Signal Transduction/drug effects , Small Molecule Libraries/administration & dosage , Small Molecule Libraries/pharmacology , T-Lymphocytes, Regulatory/metabolism , Zinc Finger Protein GLI1/genetics
6.
Nature ; 472(7341): 110-4, 2011 Apr 07.
Article in English | MEDLINE | ID: mdl-21389986

ABSTRACT

Epithelial integrity in metazoan organs is maintained through the regulated proliferation and differentiation of organ-specific stem and progenitor cells. Although the epithelia of organs such as the intestine regenerate constantly and thus remain continuously proliferative, other organs, such as the mammalian urinary bladder, shift from near-quiescence to a highly proliferative state in response to epithelial injury. The cellular and molecular mechanisms underlying this injury-induced mode of regenerative response are poorly defined. Here we show in mice that the proliferative response to bacterial infection or chemical injury within the bladder is regulated by signal feedback between basal cells of the urothelium and the stromal cells that underlie them. We demonstrate that these basal cells include stem cells capable of regenerating all cell types within the urothelium, and are marked by expression of the secreted protein signal Sonic hedgehog (Shh). On injury, Shh expression in these basal cells increases and elicits increased stromal expression of Wnt protein signals, which in turn stimulate the proliferation of both urothelial and stromal cells. The heightened activity of this signal feedback circuit and the associated increase in cell proliferation appear to be required for restoration of urothelial function and, in the case of bacterial injury, may help clear and prevent further spread of infection. Our findings provide a conceptual framework for injury-induced epithelial regeneration in endodermal organs, and may provide a basis for understanding the roles of signalling pathways in cancer growth and metastasis.


Subject(s)
Epithelial Cells/cytology , Hedgehog Proteins/metabolism , Regeneration/physiology , Stem Cells/cytology , Urinary Bladder/cytology , Wnt Proteins/metabolism , Animals , Cell Lineage , Cell Proliferation , Epithelial Cells/metabolism , Feedback, Physiological , Female , Fibroblast Growth Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Organoids/cytology , Signal Transduction , Stem Cells/metabolism , Stromal Cells/cytology , Stromal Cells/metabolism , Urinary Bladder/drug effects , Urinary Bladder/injuries , Urinary Bladder/metabolism , Urinary Bladder Diseases/chemically induced , Urinary Bladder Diseases/metabolism , Urinary Bladder Diseases/microbiology , Urinary Bladder Diseases/pathology , Uropathogenic Escherichia coli/physiology , Urothelium/cytology , Zinc Finger Protein GLI1
7.
Int J Stem Cells ; 17(2): 141-146, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38764433

ABSTRACT

Recent advancements in organoid technology have led to a vigorous movement towards utilizing it as a substitute for animal experiments. Organoid technology offers versatile applications, particularly in toxicity testing of pharmaceuticals or chemical substances. However, for the practical use in toxicity testing, minimal guidance is required to ensure reliability and relevance. This paper aims to provide minimal guidelines for practical uses of kidney organoids derived from human pluripotent stem cells as a toxicity evaluation model in vitro.

8.
BMB Rep ; 56(1): 1, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36698280

ABSTRACT

Studying human biology has been challenging with conventional animal models or two-dimensional (2D) cultured cell lines. Recent advances in stem cell biology have made it possible to culture stem cells in vitro, leading to the establishment of in vitro three-dimensional (3D) organ-like structures known as organoids. Organoids are self-organizing 3D miniature tissues that mimic the tissue architecture and functionality of in vivo counterparts. Currently, organoids can be established for multiple tissues such as the intestine, brain, kidney, prostate, pancreas, liver, bladder, heart, and retina, either from pluripotent stem cells (PSCs), including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), or adult stem cells (AdSCs). In addition to normal organoids, patient-derived tumor organoids have been established from various human tumors such as pancreatic, colorectal, breast, liver, prostate, and bladder tumors. Also, bioengineering technologies including biomaterial and scaffold fabrication, bioprinting, and microfluidics have been recently applied to create more mature and complex organoids and miniature tissues in vitro. Incorporating recently advanced computational analyses including multi-omics profiling and bioinformatics further facilitated the process of using human organoids as a novel platform for human disease modeling, drug screening to identify potential targets and novel therapeutics, and the development of precision medicine and regenerative therapies. [BMB Reports 2023; 56(1): 1].


Subject(s)
Induced Pluripotent Stem Cells , Neoplasms , Pluripotent Stem Cells , Animals , Humans , Kidney , Liver , Neoplasms/therapy , Neoplasms/metabolism , Organoids/metabolism
9.
Exp Mol Med ; 55(10): 2127-2137, 2023 10.
Article in English | MEDLINE | ID: mdl-37779144

ABSTRACT

Recent discoveries in stem cell and developmental biology have introduced a new era marked by the generation of in vitro models that recapitulate early mammalian development, providing unprecedented opportunities for extensive research in embryogenesis. Here, we present an overview of current techniques that model early mammalian embryogenesis, specifically noting models created from stem cells derived from two significant species: Homo sapiens, for its high relevance, and Mus musculus, a historically common and technically advanced model organism. We aim to provide a holistic understanding of these in vitro models by tracing the historical background of the progress made in stem cell biology and discussing the fundamental underlying principles. At each developmental stage, we present corresponding in vitro models that recapitulate the in vivo embryo and further discuss how these models may be used to model diseases. Through a discussion of these models as well as their potential applications and future challenges, we hope to demonstrate how these innovative advances in stem cell research may be further developed to actualize a model to be used in clinical practice.


Subject(s)
Embryo, Mammalian , Stem Cells , Humans , Animals , Mice , Embryonic Development , Developmental Biology , Mammals
10.
Front Plant Sci ; 13: 922694, 2022.
Article in English | MEDLINE | ID: mdl-35712604

ABSTRACT

The production of recombinant proteins in plant systems is receiving wider attention. Indeed, various plant-produced pharmaceuticals have been shown to be biologically active. However, the production of human growth factors and cytokines in heterologous systems is still challenging because they often act as complex forms, such as homo- or hetero-dimers, and their production is tightly regulated in vivo. In this study, we demonstrated that the mature form of human TGFß1 produced and purified from Nicotiana benthamiana shows biological activity in animal cells. To produce the mature form of TGFß1, various recombinant genes containing the mature form of TGFß1 were generated and produced in N. benthamiana. Of these, a recombinant construct, BiP:M:CBM3:LAP[C33S]:EK:TGFß1, was expressed at a high level in N. benthamiana. Recombinant proteins were one-step purified using cellulose-binding module 3 (CBM3) as an affinity tag and microcrystalline cellulose (MCC) beads as a matrix. The TGFß1 recombinant protein bound on MCC beads was proteolytically processed with enterokinase to separate mature TGFß1. The mature TGFß1 still associated with Latency Associated Protein, [LAP(C33S)] that had been immobilized on MCC beads was released by HCl treatment. Purified TGFß1 activated TGFß1-mediated signaling in the A549 cell line, thereby inducing phosphorylation of SMAD-2, the expression of ZEB-2 and SNAIL1, and the formation of a filopodia-like structure. Based on these results, we propose that active mature TGFß1, one of the most challenging growth factors to produce in heterologous systems, can be produced from plants at a high degree of purity via a few steps.

11.
Nat Commun ; 13(1): 3703, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35764641

ABSTRACT

Immune checkpoint inhibitors (ICIs) have substantially improved the survival of cancer patients over the past several years. However, only a minority of patients respond to ICI treatment (~30% in solid tumors), and current ICI-response-associated biomarkers often fail to predict the ICI treatment response. Here, we present a machine learning (ML) framework that leverages network-based analyses to identify ICI treatment biomarkers (NetBio) that can make robust predictions. We curate more than 700 ICI-treated patient samples with clinical outcomes and transcriptomic data, and observe that NetBio-based predictions accurately predict ICI treatment responses in three different cancer types-melanoma, gastric cancer, and bladder cancer. Moreover, the NetBio-based prediction is superior to predictions based on other conventional ICI treatment biomarkers, such as ICI targets or tumor microenvironment-associated markers. This work presents a network-based method to effectively select immunotherapy-response-associated biomarkers that can make robust ML-based predictions for precision oncology.


Subject(s)
Biomarkers, Tumor , Melanoma , Biomarkers, Tumor/genetics , Humans , Immunologic Factors , Immunotherapy/methods , Machine Learning , Melanoma/therapy , Precision Medicine , Tumor Microenvironment
12.
J Cell Biol ; 168(5): 705-11, 2005 Feb 28.
Article in English | MEDLINE | ID: mdl-15738264

ABSTRACT

Recent studies have revealed an important role for tight junction protein complexes in epithelial cell polarity. One of these complexes contains the apical transmembrane protein, Crumbs, and two PSD95/discs large/zonula occludens domain proteins, protein associated with Lin seven 1 (PALS1)/Stardust and PALS1-associated tight junction protein (PATJ). Although Crumbs and PALS1/Stardust are known to be important for cell polarization, recent studies have suggested that Drosophila PATJ is not essential and its function is unclear. Here, we find that PATJ is targeted to the apical region and tight junctions once cell polarization is initiated. We show using RNAi techniques that reduction in PATJ expression leads to delayed tight junction formation as well as defects in cell polarization. These effects are reversed by reintroduction of PATJ into these RNAi cells. This study provides new functional information on PATJ as a polarity protein and increases our understanding of the Crumbs-PALS1-PATJ complex function in epithelial polarity.


Subject(s)
Cell Polarity/physiology , Eye Proteins/metabolism , Membrane Proteins/metabolism , Tight Junctions/metabolism , Animals , Epithelium/metabolism , Genes, Reporter , Humans , RNA Interference , Tight Junction Proteins
13.
J Vis Exp ; (155)2020 01 31.
Article in English | MEDLINE | ID: mdl-32065155

ABSTRACT

The development of advanced tumor models has long been encouraged because current cancer models have shown limitations such as lack of three-dimensional (3D) tumor architecture and low relevance to human cancer. Researchers have recently developed a 3D in vitro cancer model referred to as tumor organoids that can mimic the characteristics of a native tumor in a culture dish. Here, experimental procedures are described in detail for the establishment of bladder tumor organoids from a carcinogen-induced murine bladder tumor, including culture, passage, and maintenance of the resulting 3D tumor organoids in vitro. In addition, protocols to manipulate the established bladder tumor organoid lines for genetic engineering using lentivirus-mediated transduction are described, including optimized conditions for the efficient introduction of new genetic elements into tumor organoids. Finally, the procedure for orthotopic transplantation of bladder tumor organoids into the wall of the murine bladder for further analysis is laid out. The methods described in this article can facilitate the establishment of an in vitro model for bladder cancer for the development of better therapeutic options.


Subject(s)
Organoids/transplantation , Urinary Bladder Neoplasms/therapy , Animals , Disease Models, Animal , Mice , Organoids/physiology
14.
Front Plant Sci ; 11: 440, 2020.
Article in English | MEDLINE | ID: mdl-32328082

ABSTRACT

Plants show great potential for producing recombinant proteins in a cost-effective manner. Many strategies have therefore been employed to express high levels of recombinant proteins in plants. Although foreign domains are fused to target proteins for high expression or as an affinity tag for purification, the retention of foreign domains on a target protein may be undesirable, especially for biomedical purposes. Thus, their removal is often crucial at a certain time point after translation. Here, we developed a new strategy to produce target proteins without foreign domains. This involved in vivo removal of foreign domains fused to the N-terminus by the small ubiquitin-related modifier (SUMO) domain/SUMO-specific protease system. This strategy was tested successfully by generating a recombinant gene, BiP:p38:bdSUMO : His:hLIF, that produced human leukemia inhibitory factor (hLIF) fused to p38, a coat protein of the Turnip crinkle virus; the inclusion of p38 increased levels of protein expression. The recombinant protein was expressed at high levels in the leaf tissue of Nicotiana benthamiana. Coexpression of bdSENP1, a SUMO-specific protease, proteolytically released His:hLIF from the full-length recombinant protein in the endoplasmic reticulum of N. benthamiana leaf cells. His:hLIF was purified from leaf extracts via Ni2+-NTA affinity purification resulting in a yield of 32.49 mg/kg, and the N-terminal 5-residues were verified by amino acid sequencing. Plant-produced His:hLIF was able to maintain the pluripotency of mouse embryonic stem cells. This technique thus provides a novel method of removing foreign domains from a target protein in planta.

15.
Nat Commun ; 11(1): 5485, 2020 10 30.
Article in English | MEDLINE | ID: mdl-33127883

ABSTRACT

Cancer patient classification using predictive biomarkers for anti-cancer drug responses is essential for improving therapeutic outcomes. However, current machine-learning-based predictions of drug response often fail to identify robust translational biomarkers from preclinical models. Here, we present a machine-learning framework to identify robust drug biomarkers by taking advantage of network-based analyses using pharmacogenomic data derived from three-dimensional organoid culture models. The biomarkers identified by our approach accurately predict the drug responses of 114 colorectal cancer patients treated with 5-fluorouracil and 77 bladder cancer patients treated with cisplatin. We further confirm our biomarkers using external transcriptomic datasets of drug-sensitive and -resistant isogenic cancer cell lines. Finally, concordance analysis between the transcriptomic biomarkers and independent somatic mutation-based biomarkers further validate our method. This work presents a method to predict cancer patient drug responses using pharmacogenomic data derived from organoid models by combining the application of gene modules and network-based approaches.


Subject(s)
Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , Machine Learning , Organoids/metabolism , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cisplatin/therapeutic use , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Drug Development/methods , Fluorouracil/therapeutic use , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks/drug effects , Humans , Organoids/drug effects , Protein Interaction Maps/drug effects , Transcriptome , Urinary Bladder/drug effects , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism
16.
Biofabrication ; 12(3): 035030, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32428886

ABSTRACT

Quantification of intratumoral heterogeneity is essential for designing effective therapeutic strategies in the age of personalized medicine. In this study, we used a piezoelectric inkjet printer to enable analysis of intratumoral heterogeneity in a bladder cancer for the first time. Patient-derived tumor organoids were dissociated into single cell suspension and used as a bioink. The individual cells were precisely allocated into a microwell plate by drop-on-demand inkjet printing without any additive or treatment, followed by culturing into organoids for further analysis. The sizes and morphologies of the organoids were observed, so as the expression of proliferation and apoptotic markers. The tumor organoids also showed heterogeneous responses against chemotherapeutic agent. Further, we quantified mRNA expression levels of representative luminal and basal genes in both type of tumor organoids. These results verify the heterogeneous expression of various genes among individual organoids. This study demonstrates that the fully automated inkjet printing technique can be used as an effective tool to sort cells for evaluating intratumoral heterogeneity.


Subject(s)
Bioprinting , Ink , Neoplasms/pathology , Organoids/pathology , Single-Cell Analysis , Biomarkers, Tumor/metabolism , Cell Shape/drug effects , Cisplatin/pharmacology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasms/genetics , Organoids/drug effects
17.
J Cell Biol ; 157(1): 161-72, 2002 Apr 01.
Article in English | MEDLINE | ID: mdl-11927608

ABSTRACT

Membrane-associated guanylate kinase (Maguk) proteins are scaffold proteins that contain PSD-95-Discs Large-zona occludens-1 (PDZ), Src homology 3, and guanylate kinase domains. A subset of Maguk proteins, such as mLin-2 and protein associated with Lin-7 (Pals)1, also contain two L27 domains: an L27C domain that binds mLin-7 and an L27N domain of unknown function. Here, we demonstrate that the L27N domain targets Pals1 to tight junctions by binding to a PDZ domain protein, Pals1-associated tight junction (PATJ) protein, via a unique Maguk recruitment domain. PATJ is a homologue of Drosophila Discs Lost, a protein that is crucial for epithelial polarity and that exists in a complex with the apical polarity determinant, Crumbs. PATJ and a human Crumbs homologue, CRB1, colocalize with Pals1 to tight junctions, and CRB1 interacts with PATJ albeit indirectly via binding the Pals1 PDZ domain. In agreement, we find that a Drosophila homologue of Pals1 participates in identical interactions with Drosophila Crumbs and Discs Lost. This Drosophila Pals1 homologue has been demonstrated recently to represent Stardust, a crucial polarity gene in Drosophila. Thus, our data identifies a new multiprotein complex that appears to be evolutionarily conserved and likely plays an important role in protein targeting and cell polarity.


Subject(s)
Carrier Proteins/metabolism , Eye Proteins/metabolism , Membrane Proteins/metabolism , Membrane Transport Proteins , Nerve Tissue Proteins , Nucleoside-Phosphate Kinase/metabolism , Tight Junctions/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cells, Cultured , Conserved Sequence , Drosophila , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Eye Proteins/genetics , Guanylate Kinases , Humans , Insect Proteins/genetics , Kidney/cytology , Membrane Proteins/chemistry , Membrane Proteins/genetics , Molecular Sequence Data , Mutagenesis/physiology , Nucleoside-Phosphate Kinase/genetics , Protein Structure, Tertiary , Tight Junction Proteins , Vesicular Transport Proteins
18.
Elife ; 82019 04 30.
Article in English | MEDLINE | ID: mdl-31036156

ABSTRACT

In bladder, loss of mammalian Sonic Hedgehog (Shh) accompanies progression to invasive urothelial carcinoma, but the molecular mechanisms underlying this cancer-initiating event are poorly defined. Here, we show that loss of Shh results from hypermethylation of the CpG shore of the Shh gene, and that inhibition of DNA methylation increases Shh expression to halt the initiation of murine urothelial carcinoma at the early stage of progression. In full-fledged tumors, pharmacologic augmentation of Hedgehog (Hh) pathway activity impedes tumor growth, and this cancer-restraining effect of Hh signaling is mediated by the stromal response to Shh signals, which stimulates subtype conversion of basal to luminal-like urothelial carcinoma. Our findings thus provide a basis to develop subtype-specific strategies for the management of human bladder cancer.


Subject(s)
Epigenesis, Genetic , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Hedgehogs/genetics , Signal Transduction/genetics , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , Aged , Aged, 80 and over , Animals , DNA Methylation , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Middle Aged , Stromal Cells/metabolism , Stromal Cells/pathology , Survival Analysis , Urinary Bladder/pathology , Urinary Bladder Neoplasms/pathology
19.
Mol Biol Cell ; 15(4): 1981-90, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14718565

ABSTRACT

Prior work in our laboratory established a connection between the PALS1/PATJ/CRB3 and Par6/Par3/aPKC protein complexes at the tight junction of mammalian epithelial cells. Utilizing a stable small interfering RNA expression system, we have markedly reduced expression of the tight junction-associated protein PALS1 in MDCKII cells. The loss of PALS1 resulted in a corresponding loss of expression of PATJ, a known binding partner of PALS1, but had no effect on the expression of CRB3. However, the absence of PALS1 and PATJ expression did result in the decreased association of CRB3 with members of the Par6/Par3/aPKC protein complex. The consequences of the loss of PALS1 and PATJ were exhibited by a delay in the polarization of MDCKII monolayers after calcium switch, a decrease in the transepithelial electrical resistance, and by the inability of these cells to form lumenal cysts when grown in a collagen gel matrix. These defects in polarity determination may be the result of the lack of recruitment of aPKC to the tight junction in PALS1-deficient cells, as observed by confocal microscopy, and subsequent alterations in downstream signaling events.


Subject(s)
Eye Proteins/physiology , Membrane Proteins/physiology , Tight Junctions , Animals , Base Sequence , Calcium/metabolism , Cell Line , Collagen/chemistry , Collagen/metabolism , DNA/metabolism , Dogs , Electrophysiology , Epithelial Cells/metabolism , Epithelium/metabolism , Immunoblotting , Membrane Glycoproteins/metabolism , Mice , Microscopy, Confocal , Molecular Sequence Data , Peptides/chemistry , Precipitin Tests , Protein Binding , RNA, Small Interfering/metabolism , Signal Transduction , Tight Junction Proteins , Time Factors , Transfection
20.
Science ; 356(6335)2017 04 21.
Article in English | MEDLINE | ID: mdl-28280246

ABSTRACT

The stem cell niche is a complex local signaling microenvironment that sustains stem cell activity during organ maintenance and regeneration. The mammary gland niche must support its associated stem cells while also responding to systemic hormonal regulation that triggers pubertal changes. We find that Gli2, the major Hedgehog pathway transcriptional effector, acts within mouse mammary stromal cells to direct a hormone-responsive niche signaling program by activating expression of factors that regulate epithelial stem cells as well as receptors for the mammatrophic hormones estrogen and growth hormone. Whereas prior studies implicate stem cell defects in human disease, this work shows that niche dysfunction may also cause disease, with possible relevance for human disorders and in particular the breast growth pathogenesis associated with combined pituitary hormone deficiency.


Subject(s)
Growth Hormone/metabolism , Hedgehog Proteins/metabolism , Mammary Glands, Animal/growth & development , Stem Cell Niche/genetics , Zinc Finger Protein Gli2/physiology , Animals , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/physiology , Estrogens/metabolism , Female , Gene Expression , Growth Hormone/blood , Growth Hormone/deficiency , Hedgehog Proteins/genetics , Insulin-Like Growth Factor II/genetics , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mice , Prolactin/metabolism , Sexual Maturation/genetics , Signal Transduction/genetics , Stromal Cells/metabolism , Wnt Proteins/genetics , Zinc Finger Protein Gli2/genetics
SELECTION OF CITATIONS
SEARCH DETAIL